Opsonin-Deficient Nucleoproteic Corona Endows UnPEGylated Liposomes with Stealth Properties In VivoClick to copy article linkArticle link copied!
- Francesca GiulimondiFrancesca GiulimondiDepartment of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, ItalyMore by Francesca Giulimondi
- Elisabetta VulpisElisabetta VulpisDepartment of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, ItalyMore by Elisabetta Vulpis
- Luca DigiacomoLuca DigiacomoDepartment of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, ItalyMore by Luca Digiacomo
- Maria Valeria GiuliMaria Valeria GiuliDepartment of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, ItalyMore by Maria Valeria Giuli
- Angelica MancusiAngelica MancusiDepartment of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, ItalyMore by Angelica Mancusi
- Anna Laura CapriottiAnna Laura CapriottiDepartment of Chemistry, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, ItalyMore by Anna Laura Capriotti
- Aldo LaganàAldo LaganàDepartment of Chemistry, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, ItalyMore by Aldo Laganà
- Andrea CerratoAndrea CerratoDepartment of Chemistry, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, ItalyMore by Andrea Cerrato
- Riccardo Zenezini ChiozziRiccardo Zenezini ChiozziBiomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The NetherlandsMore by Riccardo Zenezini Chiozzi
- Carmine NicolettiCarmine NicolettiUnit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Viale A. Scarpa 16, 00161 Rome, ItalyMore by Carmine Nicoletti
- Heinz AmenitschHeinz AmenitschInstitute of inorganic Chemistry, Graz University of Technology, Stremayerg 6/IV, 8010 Graz, AustriaMore by Heinz Amenitsch
- Francesco CardarelliFrancesco CardarelliNEST, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, ItalyMore by Francesco Cardarelli
- Laura MasuelliLaura MasuelliDepartment of Experimental Medicine, University of Rome “Sapienza”, Viale Regina Elena 324, 00161 Rome, ItalyMore by Laura Masuelli
- Roberto BeiRoberto BeiDepartment of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, ItalyMore by Roberto Bei
- Isabella ScrepantiIsabella ScrepantiDepartment of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, ItalyMore by Isabella Screpanti
- Daniela PozziDaniela PozziDepartment of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, ItalyMore by Daniela Pozzi
- Alessandra ZingoniAlessandra ZingoniDepartment of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, ItalyMore by Alessandra Zingoni
- Saula Checquolo*Saula Checquolo*Email: [email protected]Department of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, ItalyMore by Saula Checquolo
- Giulio Caracciolo*Giulio Caracciolo*Email: [email protected]Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, ItalyMore by Giulio Caracciolo
Abstract
For several decades, surface grafted polyethylene glycol (PEG) has been a go-to strategy for preserving the synthetic identity of liposomes in physiological milieu and preventing clearance by immune cells. However, the limited clinical translation of PEGylated liposomes is mainly due to the protein corona formation and the subsequent modification of liposomes’ synthetic identity, which affects their interactions with immune cells and blood residency. Here we exploit the electric charge of DNA to generate unPEGylated liposome/DNA complexes that, upon exposure to human plasma, gets covered with an opsonin-deficient protein corona. The final product of the synthetic process is a biomimetic nanoparticle type covered by a proteonucleotidic corona, or “proteoDNAsome”, which maintains its synthetic identity in vivo and is able to slip past the immune system more efficiently than PEGylated liposomes. Accumulation of proteoDNAsomes in the spleen and the liver was lower than that of PEGylated systems. Our work highlights the importance of generating stable biomolecular coronas in the development of stealth unPEGylated particles, thus providing a connection between the biological behavior of particles in vivo and their synthetic identity.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Results
DNA-Decorated Lipoplexes
Lipoplex-Protein Complexes
Composition of the Lipoplex-Protein Corona
Cellular Uptake by Monocytic THP-1 Cells
Particle Sequestration by Circulating Leukocytes
Capture by Immune Cells In Vivo
Conclusions
Material and Methods
Chemicals
Liposome and Lipoplex Preparation
Lipoplex-Protein Complexes
Size and Zeta Potential Experiments
Synchrotron Small-Angle X-ray Scattering Experiments
Transmission Electron Microscopy Experiments
1D SDS-PAGE Experiments
Proteomics
Cell Culture
Flow Cytometry
Particle Sequestration from Circulating Leukocytes
In Vivo Animal Experiments
NF-kB Luciferase Reporter Assay
Statistical Analysis
Minimum Information Reporting in Bio–Nano Experimental Literature (MIRIBEL)
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.1c07687.
Supporting Excel file, scheme of the in vitro and in vivo experiments (XLSX)
Figure S1, size, zeta potential, synchrotron SAXS patterns, and the corresponding computed electron density profiles of cationic liposomes CL1 and CL2; Table S1, DLS and synchrotron SAXS parameters for cationic liposomes CL1 and CL2; Figure S2, representative transmission electron microscopy (TEM) images of plain and DNA-decorated lipoplexes; Figure S3, protein corona analysis of plain and DNA-decorated lipoplexes at increasing amounts of human plasma (HP); Table S2, lists of plasma proteins identified in the coronas of DDLs and PLs by nano-LC-MS/MS; Figure S4, gating strategy of mouse phagocyte populations; Figure S5, characterization of CL2/oligonucleotide (ON) lipoplexes as a function of the cationic lipid/ON weight ratio, ρ; Figure S6, luciferase activity in HEK293/Luc reporter cells transfected or not with TLR9 and incubated for 5 h with PL1, PL2, DDL1, DDL2, or a specific TLR9 agonist; and Supplementary Table S3, companion checklist about Minimum Information Reporting in Bio–Nano Experimental Literature (MIRIBEL) (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.
References
This article references 56 other publications.
- 1Zhou, Z.; Liu, X.; Zhu, D.; Wang, Y.; Zhang, Z.; Zhou, X.; Qiu, N.; Chen, X.; Shen, Y. Nonviral cancer gene therapy: Delivery Cascade and Vector Nanoproperty Integration. Adv. Drug Deliver Rev. 2017, 115, 115– 154, DOI: 10.1016/j.addr.2017.07.021Google ScholarThere is no corresponding record for this reference.
- 2Caracciolo, G.; Palchetti, S.; Digiacomo, L.; Chiozzi, R. Z.; Capriotti, A. L.; Amenitsch, H.; Tentori, P. M.; Palmieri, V.; Papi, M.; Cardarelli, F.; Pozzi, D.; Lagana, A. Human Biomolecular Corona of Liposomal Doxorubicin: the Overlooked Factor in Anticancer Drug Delivery. ACS Appl. Mater. Interfaces 2018, 10, 22951– 22962, DOI: 10.1021/acsami.8b04962Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFeqs73J&md5=0e7ab53be0bdad05fdeb35ecd85808c4Human Biomolecular Corona of Liposomal Doxorubicin: The Overlooked Factor in Anticancer Drug DeliveryCaracciolo, Giulio; Palchetti, Sara; Digiacomo, Luca; Chiozzi, Riccardo Zene zini; Capriotti, Anna Laura; Amenitsch, Heinz; Tentori, Paolo Maria; Palmieri, Valentina; Papi, Massimiliano; Cardarelli, Francesco; Pozzi, Daniela; Lagana, AldoACS Applied Materials & Interfaces (2018), 10 (27), 22951-22962CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)More than 20 years after its approval by the Food and Drug Administration (FDA), liposomal doxorubicin (DOX) is still the drug of choice for the treatment of breast cancer and other conditions such as ovarian cancer and multiple myeloma. Yet, despite the efforts, liposomal DOX did not satisfy expectations at the clin. level. When liposomal drugs enter a physiol. environment, their surface gets coated by a dynamic biomol. corona (BC). The BC changes liposome's synthetic identity, providing it with a new one, referred to as "biol. identity" (size, aggregation state, and BC compn.). Today, the concept is emerging that specific BCs may det. either success (e.g., stealth effect and accumulation at the target site) or failure (e.g., rapid blood clearance and off-target interactions) of liposomal drugs. To get a comprehensive investigation of liposome synthetic identity, biol. identity, and cellular response as a function of human plasma (HP) concn., here we used a straightforward combination of quant. anal. and imaging tools, including dynamic light scattering, microelectrophoresis, synchrotron small-angle X-ray scattering, transmission electron microscopy (TEM), fluorescence lifetime imaging microscopy (FLIM), nano-liq. chromatog. tandem mass spectrometry/mass spectrometry (nano-LC-MS/MS), confocal microscopy, flow cytometry, and cell viability assays. Doxoves was selected as a ref. Following exposure to HP, Doxoves was surrounded by a complex BC that changed liposome's synthetic identity. Observations made with nano-LC-MS/MS revealed that the BC of Doxoves did not evolve as a function of HP concn. and was poorly enriched of typical "opsonins" (complement proteins, Igs, etc.). This provides a possible explanation for the prolonged blood circulation of liposomal DOX. On the other hand, flow cytometry showed that protein binding reduced the internalization of DOX in MCF7 and MDA-MB-435S human breast carcinoma. Combining FLIM and TEM expts., we clarified that redn. in DOX intracellular content was likely due to the frequent rupture of the liposome membrane and consequent leakage of the cargo. In light of reported results, we are prompted to speculate that a detailed understanding of BC formation, compn., and effects on liposome stability and uptake is an indispensable task of future research in the field, esp. along the way to clin. translation of liposomal drugs.
- 3Torchilin, V. P. Recent Advances With Liposomes As Pharmaceutical Carriers. Nat. Rev. Drug Discovery 2005, 4, 145– 160, DOI: 10.1038/nrd1632Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpt1WlsA%253D%253D&md5=f25804d328c08cbfadeafb90ca7e3554Recent advances with liposomes as pharmaceutical carriersTorchilin, Vladimir P.Nature Reviews Drug Discovery (2005), 4 (2), 145-160CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Liposomes - microscopic phospholipid bubbles with a bilayered membrane structure - have received a lot of attention during the past 30 years as pharmaceutical carriers of great potential. More recently, many new developments have been seen in the area of liposomal drugs - from clin. approved products to new exptl. applications, with gene delivery and cancer therapy still being the principal areas of interest. For further successful development of this field, promising trends must be identified and exploited, albeit with a clear understanding of the limitations of these approaches.
- 4Rejman, J.; Oberle, V.; Zuhorn, I. S.; Hoekstra, D. Size-Dependent Internalization Of Particles Via the Pathways of Clathrin-and Caveolae-Mediated Endocytosis. Biochemical journal 2004, 377, 159– 169, DOI: 10.1042/bj20031253Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXis1Wnsg%253D%253D&md5=e517e4c6cbf87ecba1a1c9537a0c67efSize-dependent internalization of particles via the pathways of clathrin- and caveolae-mediated endocytosisRejman, Joanna; Oberle, Volker; Zuhorn, Inge S.; Hoekstra, DickBiochemical Journal (2004), 377 (1), 159-169CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)Non-phagocytic eukaryotic cells can internalize particles <1 μm in size, encompassing pathogens, liposomes for drug delivery or lipoplexes applied in gene delivery. In the present study, we have investigated the effect of particle size on the pathway of entry and subsequent intracellular fate in non-phagocytic B16 cells, using a range of fluorescent latex beads of defined sizes (50-1000 nm). Our data reveal that particles as large as 500 nm were internalized by cells via an energy-dependent process. With an increase in size (50-500 nm), cholesterol depletion increased the efficiency of inhibition of uptake. The processing of the smaller particles was significantly perturbed upon microtubule disruption, while displaying a negligible effect on that of the 500 nm beads. Inhibitor and co-localization studies revealed that the mechanism by which the beads were internalized, and their subsequent intracellular routing, was strongly dependent on particle size. Internalization of microspheres with a diam. <200 nm involved clathrin-coated pits. With increasing size, a shift to a mechanism that relied on caveolae-mediated internalization became apparent, which became the predominant pathway of entry for particles of 500 nm in size. At these conditions, delivery to the lysosomes was no longer apparent. The data indicate that the size itself of (ligand-devoid) particles can det. the pathway of entry. The clathrin-mediated pathway of endocytosis shows an upper size limit for internalization of approx. 200 Nm, and kinetic parameters may det. the almost exclusive internalization of such particles along this pathway rather than via caveolae.
- 5Betker, J. L.; Jones, D.; Childs, C. R.; Helm, K. M.; Terrell, K.; Nagel, M. A.; Anchordoquy, T. J. Nanoparticle Uptake by Circulating Leukocytes: a Major Barrier to Tumor Delivery. J. Controlled Release 2018, 286, 85– 93, DOI: 10.1016/j.jconrel.2018.07.031Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlOit7rL&md5=83fef6f20fad50e4d3f20ef7f12bd841Nanoparticle uptake by circulating leukocytes: A major barrier to tumor deliveryBetker, Jamie L.; Jones, Dallas; Childs, Christine R.; Helm, Karen M.; Terrell, Kristina; Nagel, Maria A.; Anchordoquy, Thomas J.Journal of Controlled Release (2018), 286 (), 85-93CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Decades of research into improving drug delivery to tumors has documented uptake of particulate delivery systems by resident macrophages in the lung, liver, and spleen, and correlated short circulation times with reduced tumor accumulation. An implicit assumption in these studies is that nanoparticles present in the blood are available for distribution to the tumor. This study documents significant levels of lipoplex uptake by circulating leukocytes, and its effect on distribution to the tumor and other organs. In agreement with previous studies, PEGylation dramatically extends circulation times and enhances tumor delivery. However, our studies suggest that this relationship is not straightforward, and that particle sequestration by leukocytes can significantly alter biodistribution, esp. with non-PEGylated nanoparticle formulations. We conclude that leukocyte uptake should be considered in biodistribution studies, and that delivery to these circulating cells may present opportunities for treating viral infections and leukemia.
- 6Mislick, K. A.; Baldeschwieler, J. D. Evidence for the Role of Proteoglycans in Cation-Mediated Gene Transfer. Proc. Natl. Acad. Sci. U. S. A. 1996, 93, 12349– 12354, DOI: 10.1073/pnas.93.22.12349Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xms1GjtLc%253D&md5=82590756f2ed276c18b3d9cfdc89d778Evidence for the role of proteoglycans in cation-mediated gene transferMislick, Kimberly A.; Baldeschwieler, John D.Proceedings of the National Academy of Sciences of the United States of America (1996), 93 (22), 12349-12354CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We report evidence that gene complexes, consisting of polycations and plasmid DNA enter cells via binding to membrane-assocd. proteoglycans. Treatment of HeLa cells with sodium chlorate, a potent inhibitor of proteoglycan sulfation, reduced luciferase expression by 69%. Cellular treatment with heparinase and chondroitinase ABC inhibited expression by 78% and 20% with respect to control cells. Transfection was dramatically inhibited by heparin and heparan sulfate and to a smaller extent by chondroitin sulfate B. Transfection of mutant, proteoglycan deficient Chinese hamster ovary cells was 53× lower than of wild-type cells. For each of these assays, the intracellular uptake of DNA at 37°C and the binding of DNA to the cell membrane at 4°C was impaired. Preliminary transfection expts. conducted in mutant and wild-type Chinese hamster ovary cells suggest that transfection by some cationic lipids is also proteoglycan dependent. The variable distribution of proteoglycans among tissues may explain why some cell types are more susceptible to transfection than others.
- 7Pozzi, D.; Colapicchioni, V.; Caracciolo, G.; Piovesana, S.; Capriotti, A. L.; Palchetti, S.; De Grossi, S.; Riccioli, A.; Amenitsch, H.; Laganà, A. Effect of Polyethyleneglycol (PEG) Chain Length on the Bio–Nano-Interactions between PEGylated Lipid Nanoparticles and Biological Fluids: from Nanostructure to Uptake in Cancer Cells. Nanoscale 2014, 6, 2782– 2792, DOI: 10.1039/c3nr05559kGoogle Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXisVahtrw%253D&md5=e52e369b784c0d20d6f7f308197d5888Effect of polyethyleneglycol (PEG) chain length on the bio-nano-interactions between PEGylated lipid nanoparticles and biological fluids: from nanostructure to uptake in cancer cellsPozzi, Daniela; Colapicchioni, Valentina; Caracciolo, Giulio; Piovesana, Susy; Capriotti, Anna Laura; Palchetti, Sara; De Grossi, Stefania; Riccioli, Anna; Amenitsch, Heinz; Lagana, AldoNanoscale (2014), 6 (5), 2782-2792CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)When nanoparticles (NPs) enter a physiol. environment, medium components compete for binding to the NP surface leading to formation of a rich protein shell known as the "protein corona". Unfortunately, opsonins are also adsorbed. These proteins are immediately recognized by the phagocyte system with rapid clearance of the NPs from the bloodstream. Polyethyleneglycol (PEG) coating of NPs (PEGylation) is the most efficient anti-opsonization strategy. Linear chains of PEG, grafted onto the NP surface, are able to create steric hindrance, resulting in a significant inhibition of protein adsorption and less recognition by macrophages. However, excessive PEGylation can lead to a strong inhibition of cellular uptake and less efficient binding with protein targets, reducing the potential of the delivery system. To reach a compromise in this regard we employed a multi-component (MC) lipid system with uncommon properties of cell uptake and endosomal escape and increasing length of PEG chains. Nano liq. chromatog. coupled with tandem mass spectrometry (nanoLC-MS/MS) anal. allowed us to accurately det. the corona compn. showing that apolipoproteins are the most abundant class in the corona and that increasing the PEG length reduced the protein adsorption and the liposomal surface affinity for apolipoproteins. Due to the abundance of apolipoproteins, we exploited the "protein corona effect" to deliver cationic liposome-human plasma complexes to human prostate cancer PC3 cells that express a high level of scavenger receptor class B type 1 to evaluate the cellular uptake efficiency of the systems used. Combining laser scanning confocal microscopy with flow cytometry anal. in PC3 cells we demonstrated that MC-PEG2k is the best compromise between an anti-opsonization strategy and active targeting and could be a promising candidate to treat prostate cancer in vivo.
- 8Srinivasan, C.; Burgess, D. J. Optimization and Characterization of Anionic Lipoplexes for Gene Delivery. Journal of controlled release 2009, 136, 62– 70, DOI: 10.1016/j.jconrel.2009.01.022Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXlvFGjtLg%253D&md5=f59f8f868862bf9ebaa2c3eff935fddfOptimization and characterization of anionic lipoplexes for gene deliverySrinivasan, Charudharshini; Burgess, Diane J.Journal of Controlled Release (2009), 136 (1), 62-70CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Anionic lipoplexes, comprising divalent cations, DNA and anionic liposomes, were optimized for high transfection efficiency and low cytotoxicity. Different molar ratios of anionic to zwitterionic lipid, 1:9 to 1:1 (1,2-dioleoyl-sn-glycero-3-[phospho-rac-(1-glycerol)] (DOPG): 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), various cations (Ca2+, Mg2+ and Na+) and different anionic lipid/DNA ratios were investigated. The optimized formulation was composed of: anionic/zwitterionic lipid molar ratio 1:4 (DOPG:DOPE); 15-20 mM Ca2+; and 15-20 μg lipid for complexation with 0.8 μg plasmid DNA. Biophys. studies (particle size anal., gel electrophoresis, transmission electron microscopy (TEM), and confocal microscopy) were conducted to characterize the different formulations. TEM revealed structural differences between the complexed and uncomplexed lipoplexes. Gel electrophoresis confirmed the formation of anionic lipoplexes with the amt. of free DNA minimized for the optimized formulation. Confocal imaging showed cellular uptake of the anionic lipoplexes. Most significantly the anionic lipoplex formulation, optimized for the highest transfection efficiency (approx. 78% in the presence of serum) exhibited the highest cell viability approx. 93%, (3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyl tetrazolium bromide) MTT assay. This was compared to Lipofectamine2000 which had a transfection efficiency and cell viability of approx. 68% and 35%, resp. The anionic lipoplex formulation developed here shows promise as a non-viral vector with high transfection efficiency and low cytotoxicity.
- 9McManus, J. J.; Rädler, J. O.; Dawson, K. A. Does Calcium Turn a Zwitterionic Lipid Cationic?. J. Phys. Chem. B 2003, 107, 9869– 9875, DOI: 10.1021/jp034463dGoogle Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXmsVegtLo%253D&md5=9238f106a72aadd839604b2f0d8e28ddDoes Calcium Turn a Zwitterionic Lipid Cationic?McManus, Jennifer J.; Raedler, Joachim O.; Dawson, Kenneth A.Journal of Physical Chemistry B (2003), 107 (36), 9869-9875CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)The structure of a complex formed from DNA, calcium, and DPPC has been studied by small-angle X-ray scattering (SAXS). Previous DSC and temp. scanning ultrasound have shown that in excess lipid two DPPC phases exist, one corresponding to DNA-bound lipid and the other corresponding to lipid not directly involved in complex formation. We have investigated the effect of calcium on both phases within the complex structure. Contrary to the observations in the ternary DPPC/calcium/water system, the binding of calcium to the DPPC membrane in this quaternary system contg. DNA appears to be significantly enhanced. We have estd. the stoichiometry of calcium binding within the complex, and we concluded that the zwitterionic lipid is directly involved in binding to DNA.
- 10Palchetti, S.; Pozzi, D.; Marchini, C.; Amici, A.; Andreani, C.; Bartolacci, C.; Digiacomo, L.; Gambini, V.; Cardarelli, F.; Di Rienzo, C.; Peruzzi, G.; Amenitsch, H.; Palermo, R.; Screpanti, I.; Caracciolo, G. Manipulation of Lipoplex Concentration at the Cell Surface Boosts Transfection Efficiency in Hard-to-Transfect Cells. Nanomedicine: Nanotechnology, Biology and Medicine 2017, 13, 681– 691, DOI: 10.1016/j.nano.2016.08.019Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2szks1elug%253D%253D&md5=e9844674edc9429b5ba523ae3b3e86cdManipulation of lipoplex concentration at the cell surface boosts transfection efficiency in hard-to-transfect cellsPalchetti Sara; Pozzi Daniela; Marchini Cristina; Amici Augusto; Andreani Cristina; Bartolacci Caterina; Gambini Valentina; Digiacomo Luca; Cardarelli Francesco; Di Rienzo Carmine; Peruzzi Giovanna; Palermo Rocco; Amenitsch Heinz; Screpanti Isabella; Caracciolo GiulioNanomedicine : nanotechnology, biology, and medicine (2017), 13 (2), 681-691 ISSN:.To date, efficiency upon non-viral DNA delivery remains low and this implies the existence of unidentified transfection barriers. Here we explore the mechanisms of action of multicomponent (MC) cationic liposome/DNA complexes (lipoplexes) by a combination of reporter technologies, dynamic light scattering (DLS), synchrotron small angle X-ray scattering (SAXS), fluorescence activated cell sorting (FACS) analysis and laser scanning confocal microscopy (LSCM) in live cells. Lipofectamine - the gold standard among transfection reagents - was used as a reference. On the basis of our results, we suggest that an additional transfection barrier impairs transfection efficiency, that is: low lipoplex concentration at the cell surface. Based on the acquired knowledge we propose an optimized transfection protocol that allowed us to efficiently transfect DND41, JURKAT, MOLT3, P12-ICHIKAWA, ALL-SILL, TALL-1 human T-cell acute lymphoblastic leukemia (T-ALL) cell lines known to be difficult-to-transfect by using non-viral vectors and where LFN-based technologies fail to give satisfactory results.
- 11Pan, J.; Heberle, F. A.; Tristram-Nagle, S.; Szymanski, M.; Koepfinger, M.; Katsaras, J.; Kučerka, N. Molecular Structures of Fluid Phase Phosphatidylglycerol Bilayers As Determined by Small Angle Neutron and X-Ray Scattering. Biochimica et Biophysica Acta (BBA)-Biomembranes 2012, 1818, 2135– 2148, DOI: 10.1016/j.bbamem.2012.05.007Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XovVaju7g%253D&md5=957c783ae34f395d4cccfc473dfe18e9Molecular structures of fluid phase phosphatidylglycerol bilayers as determined by small angle neutron and X-ray scatteringPan, Jianjun; Heberle, Frederick A.; Tristram-Nagle, Stephanie; Szymanski, Michelle; Koepfinger, Mary; Katsaras, John; Kucerka, NorbertBiochimica et Biophysica Acta, Biomembranes (2012), 1818 (9), 2135-2148CODEN: BBBMBS; ISSN:0005-2736. (Elsevier B.V.)We have detd. the mol. structures of commonly used phosphatidylglycerols (PGs) in the commonly accepted biol. relevant fluid phase. This was done by simultaneously analyzing small angle neutron and X-ray scattering data, with the constraint of measured lipid vols. We report the temp. dependence of bilayer parameters obtained using the one-dimensional scattering d. profile model - which was derived from mol. dynamics simulations - including the area per lipid, the overall bilayer thickness, as well as other intrabilayer parameters (e.g., hydrocarbon thickness). Lipid areas are found to be larger than their phosphatidylcholine (PC) counterparts, a result likely due to repulsive electrostatic interactions taking place between the charged PG headgroups even in the presence of sodium counterions. In general, PG and PC bilayers show a similar response to changes in temp. and chain length, but differ in their response to chain unsatn. For example, compared to PC bilayers, the inclusion of a first double bond in PG lipids results in a smaller incremental change to the area per lipid and bilayer thickness. However, the extrapolated lipid area of satd. PG lipids to infinite chain length is found to be similar to that of PCs, an indication of the glycerol-carbonyl backbone's pivotal role in influencing the lipid-water interface.
- 12Caracciolo, G. Liposome-Protein Corona in a Physiological Environment: Challenges and Opportunities for Targeted Delivery of Nanomedicines. Nanomedicine: Nanotechnology, Biology, and Medicine 2015, 11, 543– 557, DOI: 10.1016/j.nano.2014.11.003Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2Mvjt1CnsA%253D%253D&md5=4ad22c113ca7fd1830353edcb8c5395eLiposome-protein corona in a physiological environment: challenges and opportunities for targeted delivery of nanomedicinesCaracciolo GiulioNanomedicine : nanotechnology, biology, and medicine (2015), 11 (3), 543-57 ISSN:.Active targeting that exploits the (over)expression of surface receptors in target cells by ligand incorporation is a central concept in nanomedicine research. Despite unprecedented efforts, no targeted liposome-based therapeutics is commercially available for clinical practice. What is inhibiting the efficient translation of targeted liposome technology from bench to bedside? After introduction in the bloodstream, the lipid surface is immediately modified by the adsorption of a "protein corona" and preserving the surface functionality appears to be challenging. On the other hand, a long-standing corona with receptor-binding sites could associate with the target cell long enough to activate the cell's uptake machinery, triggering liposome endocytosis and intracellular cargo delivery. This opens the intriguing possibility to manipulate the corona composition by liposome design. This review will focus on the emerging field of liposome-protein corona research from basic, descriptive research to readily applicable knowledge and technologies for implementation in drug improvement and development. From the clinical editor: This review is addressing the liposome protein corona research concerning the potential gains in drug improvement and for drug development.
- 13Pozzi, D.; Caracciolo, G.; Caminiti, R.; De Sanctis, S. C.; Amenitsch, H.; Marchini, C.; Montani, M.; Amici, A. 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. ACS Appl. Mater. Interfaces 2009, 1, 2237– 2249, DOI: 10.1021/am900406bGoogle Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFOiurjM&md5=e309feac0c73ff4b503c7c62e31c249bToward 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 releasePozzi, Daniela; Caracciolo, Giulio; Caminiti, Ruggero; De Sanctis, Sofia Candeloro; Amenitsch, Heinz; Marchini, Cristina; Montani, Maura; Amici, AugustoACS Applied Materials & Interfaces (2009), 1 (10), 2237-2249CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)A viewpoint now emerging is that a crit. factor in lipid-mediated transfection (lipofection) is the structural evolution of lipoplexes upon interaction with anionic cellular lipids, resulting in DNA release. At the early stages of interaction, the authors found a universal behavior of lipoplex/anionic lipid (AL) mixts.: the lipoplex structure is slightly perturbed, while the one-dimensional DNA lattice between cationic membranes is largely dild. by ALs. This finding is in excellent agreement with previous suggestions on the mechanism of DNA unbinding from lipoplexes by ALs. Upon further interaction, the propensity of a given lipoplex structure to be solubilized by anionic cellular lipids strongly depends on the shape coupling between lipoplex and ALs. Furthermore, the authors investigated the effect of the membrane charge d. and a general correlation resulted: the higher the membrane charge d. of anionic membranes, the higher their ability to solubilize the structure of lipoplexes and to promote DNA release. Lastly, the formation of nonlamellar phases in lipoplex/AL mixts. is regulated by the propensity of anionic cellular lipids to adopt nonlamellar phases. Remarkably, also phase transition rates and DNA release were found to be strongly affected by the shape coupling between lipoplex and ALs. It thus seems likely that the structural and phase evolution of lipoplexes may only be meaningful in the context of specific anionic cellular membranes. These results highlight the phase properties of the carrier lipid/cellular lipid mixts. as a decisive factor for optimal DNA release and suggest a potential strategy for the rational design of efficient cationic lipid carriers.
- 14Pozzi, D.; Marchini, C.; Cardarelli, F.; Rossetta, A.; Colapicchioni, V.; Amici, A.; Montani, M.; Motta, S.; Brocca, P.; Cantu, L.; Caracciolo, G. Mechanistic Understanding of Gene Delivery Mediated by Highly Efficient Multicomponent Envelope-Type Nanoparticle Systems. Mol. Pharmaceutics 2013, 10, 4654– 4665, DOI: 10.1021/mp400470pGoogle Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslWlsr3I&md5=25b7631ca1963e0eef925f241625110bMechanistic Understanding of Gene Delivery Mediated by Highly Efficient Multicomponent Envelope-Type Nanoparticle SystemsPozzi, D.; Marchini, C.; Cardarelli, F.; Rossetta, A.; Colapicchioni, V.; Amici, A.; Montani, M.; Motta, S.; Brocca, P.; Cantu, L.; Caracciolo, G.Molecular Pharmaceutics (2013), 10 (12), 4654-4665CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)We packaged condensed DNA/protamine particles in multicomponent envelope-type nanoparticle systems (MENS) combining different molar fractions of the cationic lipids 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and 3β-[N-(N,N-dimethylaminoethane)-carbamoyl] cholesterol (DC-Chol) and the zwitterionic lipids dioleoylphosphocholine (DOPC) and dioleoylphosphatidylethanolamine (DOPE). Dynamic light scattering (DLS) and microelectrophoresis allowed us to identify the cationic lipid/DNA charge ratio at which MENS are small sized and pos. charged, while synchrotron small-angle X-ray scattering (SAXS) and at. force microscopy (AFM) revealed that MENS are well-shaped DNA/protamine particles covered by a lipid monobilayer. Transfection efficiency (TE) expts. indicate that a nanoparticle formulation, termed MENS-3, was not cytotoxic and highly efficient to transfect Chinese hamster ovary (CHO) cells. To rationalize TE, we performed a quant. investigation of cell uptake, intracellular trafficking,endosomal escape, and final fate by laser scanning confocal microscopy (LSCM). We found that fluid-phase macropinocytosis is the only endocytosis pathway used by MENS-3. Once taken up by the cell, complexes that are actively transported by microtubules frequently fuse with lysosomes, while purely diffusing systems do not. Indeed, spatiotemporal image correlation spectroscopy (STICS) clarified that MENS-3 mostly exploit diffusion to move in the cytosol of CHO cells, thus explaining the high TE levels obsd. Also, MENS-3 exhibited a marked endosomal rupture ability resulting in extraordinary DNA release. The lipid-dependent and structure-dependent TE boost suggests that ecient transfection requires both the membrane fusogenic activity of the nanocarrier envelope and the employment of lipid species with intrinsic endosomal rupture ability.
- 15Koltover, I.; Salditt, T.; Safinya, C. Phase Diagram, Stability, and Overcharging of Lamellar Cationic Lipid–DNA Self-Assembled Complexes. Biophys. J. 1999, 77, 915– 924, DOI: 10.1016/S0006-3495(99)76942-0Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXltVyju7w%253D&md5=ef8851417cf746d60ee8bef076e9d8fcPhase diagram, stability, and overcharging of lamellar cationic lipid-DNA self-assembled complexesKoltover, I.; Salditt, T.; Safinya, C. R.Biophysical Journal (1999), 77 (2), 915-924CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)Cationic lipid-DNA (CL-DNA) complexes comprise a promising new class of synthetic nonviral gene delivery systems. When pos. charged, they attach to the anionic cell surface and transfer DNA into the cell cytoplasm. A comprehensive x-ray diffraction study is reported of the lamellar CL-DNA self-assemblies as a function of lipid compn. and lipid/DNA ratio, aimed at elucidating the interactions detg. their structure, charge, and thermodn. stability. The driving force for the formation of charge-neutral complexes is the release of DNA and lipid counterions. Neg. charged complexes have a higher DNA packing d. than isoelec. complexes, whereas pos. charged ones have a lower packing d. This indicates that the overcharging of the complex away from its isoelec. point is caused by changes of the bulk structure with absorption of excess DNA or cationic lipid. The degree of overcharging is dependent on the membrane charge d., which is controlled by the ration of neutral to cationic lipid in the bilayers. Importantly, overcharged complexes are obsd. to move toward their isoelec. charge-neutral point at higher concn. of salt co-ions, with pos. overcharged complexes expelling cationic lipid and neg. overcharged complexes expelling DNA. These observations should apply universally to the formation and structure of self-assemblies between oppositely charged macromols.
- 16Cukalevski, R.; Ferreira, S. A.; Dunning, C. J.; Berggård, T.; Cedervall, T. IgG and Fibrinogen Driven Nanoparticle Aggregation. Nano Research 2015, 8, 2733– 2743, DOI: 10.1007/s12274-015-0780-4Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVKqurnK&md5=65e384125977dcef8e779f96b42633fcIgG and fibrinogen driven nanoparticle aggregationCukalevski, Risto; Ferreira, Silvia A.; Dunning, Christopher J.; Berggaard, Tord; Cedervall, TommyNano Research (2015), 8 (8), 2733-2743CODEN: NRAEB5; ISSN:1998-0000. (Springer GmbH)A thorough understanding of how proteins induce nanoparticle (NP) aggregation is crucial when designing in vitro and in vivo assays and interpreting exptl. results. This knowledge is also crucial when developing nano-applications and formulation for drug delivery systems. In this study, we found that extn. of IgG (IgG) from cow serum results in lower polystyrene NPs aggregation. Moreover, addn. of isolated IgG or fibrinogen to fetal cow serum enhanced this aggregation, thus demonstrating that these factors are major drivers of NP aggregation in serum. Counter-intuitively, NP aggregation was inversely dependent on protein concn.; i.e., low protein concns. induced large aggregates, whereas high protein concns. induced small aggregates. Protein-induced NP aggregation and aggregate size were monitored by absorbance at 400 nm and dynamic light scattering, resp. Here, we propose a mechanism behind the protein concn. dependent aggregation; this mechanism involves the effects of multiple protein interactions on the NP surface, surface area limitations, aggregation kinetics, and the influence of other serum proteins. [Figure not available: see fulltext.].
- 17Walkey, C. D.; Chan, W. C. Understanding and Controlling the Interaction of Nanomaterials with Proteins in a Physiological Environment. Chem. Soc. Rev. 2012, 41, 2780– 2799, DOI: 10.1039/C1CS15233EGoogle Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xjs1Cksb8%253D&md5=cc7bfcf4dd59ced93710ef642e64a044Understanding and controlling the interaction of nanomaterials with proteins in a physiological environmentWalkey, Carl D.; Chan, Warren C. W.Chemical Society Reviews (2012), 41 (7), 2780-2799CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. Nanomaterials hold promise as multifunctional diagnostic and therapeutic agents. However, the effective application of nanomaterials is hampered by limited understanding and control over their interactions with complex biol. systems. When a nanomaterial enters a physiol. environment, it rapidly adsorbs proteins forming what is known as the protein corona'. The protein corona alters the size and interfacial compn. of a nanomaterial, giving it a biol. identity that is distinct from its synthetic identity. The biol. identity dets. the physiol. response including signalling, kinetics, transport, accumulation, and toxicity. The structure and compn. of the protein corona depends on the synthetic identity of the nanomaterial (size, shape, and compn.), the nature of the physiol. environment (blood, interstitial fluid, cell cytoplasm, etc.), and the duration of exposure. In this crit. review, we discuss the formation of the protein corona, its structure and compn., and its influence on the physiol. response. We also present an adsorbome' of 125 plasma proteins that are known to assoc. with nanomaterials. We further describe how the protein corona is related to the synthetic identity of a nanomaterial, and highlight efforts to control protein-nanomaterial interactions. We conclude by discussing gaps in the understanding of protein-nanomaterial interactions along with strategies to fill them (167 refs.).
- 18Caracciolo, G.; Pozzi, D.; Capriotti, A. L.; Cavaliere, C.; Foglia, P.; Amenitsch, H.; Laganà, A. Evolution of the Protein Corona of Lipid Gene Vectors as a Function of Plasma Concentration. Langmuir 2011, 27, 15048– 15053, DOI: 10.1021/la202912fGoogle Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtl2kt7rO&md5=2d5172301cfaceda4abedbfd6ee2c526Evolution of the protein corona of lipid gene vectors as a function of plasma concentrationCaracciolo, Giulio; Pozzi, Daniela; Capriotti, Anna Laura; Cavaliere, Chiara; Foglia, Patrizia; Amenitsch, Heinz; Lagana, AldoLangmuir (2011), 27 (24), 15048-15053CODEN: LANGD5; ISSN:0743-7463. (American Chemical Society)The concept that the effective unit of interest in the cell-nanomaterial interaction is the particle and its corona of assocd. proteins is emerging. Here we investigate the compositional evolution of the protein corona of 1,2-dioleoyl-3-trimethylammonium propane (DOTAP) cationic liposomes (CLs) and DOTAP/DNA lipoplexes over a wide range of plasma concns. (2.5-80%). The compn. of the hard corona of lipoplexes is quite stable, but that of CLs does evolve considerably. We show that the protein corona of CLs is made of both low-affinity and competitive-binding proteins whose relative abundance changes with the plasma concn. This result may have deep biol. implications for the application of lipid-based gene vectors both in vitro and in vivo.
- 19Monopoli, M. P.; Walczyk, D.; Campbell, A.; Elia, G.; Lynch, I.; Baldelli Bombelli, F.; Dawson, K. A. Physical–Chemical Aspects of Protein Corona: Relevance to In Vitro and In Vivo Biological Impacts of Nanoparticles. J. Am. Chem. Soc. 2011, 133, 2525– 2534, DOI: 10.1021/ja107583hGoogle Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsVOnt7w%253D&md5=a93981208087f4c615ac8c7df33a20ddPhysical-Chemical Aspects of Protein Corona: Relevance to in Vitro and in Vivo Biological Impacts of NanoparticlesMonopoli, Marco P.; Walczyk, Dorota; Campbell, Abigail; Elia, Giuliano; Lynch, Iseult; Baldelli Bombelli, Francesca; Dawson, Kenneth A.Journal of the American Chemical Society (2011), 133 (8), 2525-2534CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)It is now clearly emerging that besides size and shape, the other primary defining element of nanoscale objects in biol. media is their long-lived protein ("hard") corona. This corona may be expressed as a durable, stabilizing coating of the bare surface of nanoparticle (NP) monomers, or it may be reflected in different subpopulations of particle assemblies, each presenting a durable protein coating. Using the approach and concepts of phys. chem., we relate studies on the compn. of the protein corona at different plasma concns. with structural data on the complexes both in situ and free from excess plasma. This enables a high degree of confidence in the meaning of the hard protein corona in a biol. context. Here, we present the protein adsorption for two compositionally different NPs, namely sulfonated polystyrene and silica NPs. NP-protein complexes are characterized by differential centrifugal sedimentation, dynamic light scattering, and zeta-potential both in situ and once isolated from plasma as a function of the protein/NP surface area ratio. We then introduce a semiquant. detn. of their hard corona compn. using one-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis and electrospray liq. chromatog. mass spectrometry, which allows us to follow the total binding isotherms for the particles, identifying simultaneously the nature and amt. of the most relevant proteins as a function of the plasma concn. We find that the hard corona can evolve quite significantly as one passes from protein concns. appropriate to in vitro cell studies to those present in in vivo studies, which has deep implications for in vitro-in vivo extrapolations and will require some consideration in the future.
- 20Caracciolo, G.; Pozzi, D.; Capriotti, A.; Cavaliere, C.; Piovesana, S.; Amenitsch, H.; Laganà, A. Lipid Composition: a “Key Factor” for the Rational Manipulation of the Liposome–Protein Corona by Liposome Design. Rsc Adv. 2015, 5, 5967– 5975, DOI: 10.1039/C4RA13335HGoogle Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFaltb%252FN&md5=d1a9c991f2a5a3b33d9c0d42c54a2e97Lipid composition: a "key factor" for the rational manipulation of the liposome-protein corona by liposome designCaracciolo, G.; Pozzi, D.; Capriotti, A. L.; Cavaliere, C.; Piovesana, S.; Amenitsch, H.; Lagana, A.RSC Advances (2015), 5 (8), 5967-5975CODEN: RSCACL; ISSN:2046-2069. (Royal Society of Chemistry)When liposomes are exposed to biol. fluids, a dynamic coating of proteins immediately covers them. Similarly to the aura of plasma surrounding the Sun, plasma proteins are thought of as establishing an aura that surrounds each liposome, hence the phenomenon was dubbed 'protein corona'. This natural functionalization includes proteins engaged from the blood that can interact with receptors (over)expressed on the plasma membrane of target cells, thus targeting the liposomes to their final destination. Exploiting the liposome-protein corona for targeting has the potential to revolutionize the treatment of many disorders and requires a deep understanding of the factors shaping the corona. Following incubation with human plasma (HP), here we manipulated this corona by using six liposomal formulations with systematic changes in lipid compn. The lipids we employed are among the most frequently used lipid species for drug and gene delivery applications in vitro and in vivo. The six liposome-protein coronas were thoroughly characterized by synchrotron small angle X-ray scattering, dynamic light scattering, zeta-potential and nanoliquid-chromatog. tandem mass spectrometry expts. We identified general principles shaping the liposome-protein corona and established clear-cut relationships between lipid species and classes of plasma proteins. This knowledge sets the basis for a rational manipulation of the protein corona for targeted drug delivery by liposome design.
- 21Martínez-Negro, M.; González-Rubio, G.; Aicart, E.; Landfester, K.; Guerrero-Martínez, A.; Junquera, E. Insights into Colloidal Nanoparticle-Protein Corona Interactions for Nanomedicine Applications. Adv. Colloid Interface Sci. 2021, 289, 102366, DOI: 10.1016/j.cis.2021.102366Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXivValsrs%253D&md5=5bb72bf9c5caa188e23a832283cc2abfInsights into colloidal nanoparticle-protein corona interactions for nanomedicine applicationsMartinez-Negro, Maria; Gonzalez-Rubio, Guillermo; Aicart, Emilio; Landfester, Katharina; Guerrero-Martinez, Andres; Junquera, ElenaAdvances in Colloid and Interface Science (2021), 289 (), 102366CODEN: ACISB9; ISSN:0001-8686. (Elsevier B.V.)A review. Nevertheless, the successful implementation of NPs in medicine demands a proper understanding of their interactions with the different proteins found in biol. fluids. Once introduced into the body, NPs are covered by a protein corona (PC) that dets. the biol. behavior of the NPs. The formation of the PC can eventually favor the rapid clearance of the NPs from the body before fulfilling the desired objective or lead to increased cytotoxicity. The PC nature varies as a function of the different repulsive and attractive forces that govern the NP-protein interaction and their colloidal stability. This review focuses on the phenomenon of PC formation on NPs from a physicochem. perspective, aiming to provide a general overview of this crit. process. Main issues related to NP toxicity and clearance from the body as a result of protein adsorption are covered, including the most promising strategies to control PC formation and, thereby, ensure the successful application of NPs in nanomedicine. Colloidal nanoparticles (NPs) have attracted significant attention due to their unique physicochem. properties suitable for diagnosing and treating different human diseases.
- 22Danhier, F.; Feron, O.; Préat, V. To Exploit the Tumor Microenvironment: Passive and Active Tumor Targeting of Nanocarriers for Anti-Cancer Drug Delivery. Journal of controlled release 2010, 148, 135– 146, DOI: 10.1016/j.jconrel.2010.08.027Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsVCmsrzF&md5=04d79d93a1828d379bfde830da737608To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug deliveryDanhier, Fabienne; Feron, Olivier; Preat, VeroniqueJournal of Controlled Release (2010), 148 (2), 135-146CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)A review. Because of the particular characteristics of the tumor microenvironment and tumor angiogenesis, it is possible to design drug delivery systems that specifically target anti-cancer drugs to tumors. Most of the conventional chemotherapeutic agents have poor pharmacokinetics profiles and are distributed non-specifically in the body leading to systemic toxicity assocd. with serious side effects. Therefore, the development of drug delivery systems able to target the tumor site is becoming a real challenge that is currently addressed. Nanomedicine can reach tumor passively through the leaky vasculature surrounding the tumors by the enhanced permeability and retention effect whereas ligands grafted at the surface of nanocarriers allow active targeting by binding to the receptors overexpressed by cancer cells or angiogenic endothelial cells. This review is divided into 2 parts: the first one describes the tumor microenvironment and the second one focuses on the exploitation and the understanding of these characteristics to design new drug delivery systems targeting the tumor. Delivery of conventional chemotherapeutic anti-cancer drugs is mainly discussed.
- 23Sawant, R. R.; Torchilin, V. P. Challenges in Development of Targeted Liposomal Therapeutics. AAPS journal 2012, 14, 303– 315, DOI: 10.1208/s12248-012-9330-0Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xlslant74%253D&md5=e017520ac851c05a4ab607645b15a263Challenges in Development of Targeted Liposomal TherapeuticsSawant, Rupa R.; Torchilin, Vladimir P.AAPS Journal (2012), 14 (2), 303-315CODEN: AJAOB6; ISSN:1550-7416. (Springer)A review. Liposomes, phospholipid vesicles with a bilayered membrane structure, have been widely used as pharmaceutical carriers for drugs and genes, in particular for treatment of cancer. To enhance the efficacy of the liposomal drugs, drug-loaded liposomes are targeted to the tumors by means of passive (enhanced permeability and retention mediated) targeting, based on the longevity of liposomes in blood and its accumulation in pathol. sites with compromised vasculature, and active targeting, based on the attachment of specific ligands to the liposomal surface to bind certain antigens on the target cells. Antibody-targeted liposomes loaded with anticancer drugs demonstrate high potential for clin. applications. This review highlights evolution of liposomes for both passive and active targeting and challenges in development of targeted liposomal therapeutics specifically antibody-targeted liposomes.
- 24Tenzer, S.; Docter, D.; Kuharev, J.; Musyanovych, A.; Fetz, V.; Hecht, R.; Schlenk, F.; Fischer, D.; Kiouptsi, K.; Reinhardt, C.; Landfester, K.; Schild, H.; Maskos, M.; Knauer, S. K.; Stauber, R. H. Rapid Formation of Plasma Protein Corona Critically Affects Nanoparticle Pathophysiology. Nat. Nano 2013, 8, 772– 781, DOI: 10.1038/nnano.2013.181Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVyqurrL&md5=927a38bd98a5850c71525a7707dc5073Rapid formation of plasma protein corona critically affects nanoparticle pathophysiologyTenzer, Stefan; Docter, Dominic; Kuharev, Joerg; Musyanovych, Anna; Fetz, Verena; Hecht, Rouven; Schlenk, Florian; Fischer, Dagmar; Kiouptsi, Klytaimnistra; Reinhardt, Christoph; Landfester, Katharina; Schild, Hansjoerg; Maskos, Michael; Knauer, Shirley K.; Stauber, Roland H.Nature Nanotechnology (2013), 8 (10), 772-781CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)In biol. fluids, proteins bind to the surface of nanoparticles to form a coating known as the protein corona, which can critically affect the interaction of the nanoparticles with living systems. As physiol. systems are highly dynamic, it is important to obtain a time-resolved knowledge of protein-corona formation, development and biol. relevancy. Here we show that label-free snapshot proteomics can be used to obtain quant. time-resolved profiles of human plasma coronas formed on silica and polystyrene nanoparticles of various size and surface functionalization. Complex time- and nanoparticle-specific coronas, which comprise almost 300 different proteins, were found to form rapidly (<0.5 min) and, over time, to change significantly in terms of the amt. of bound protein, but not in compn. Rapid corona formation is found to affect hemolysis, thrombocyte activation, nanoparticle uptake and endothelial cell death at an early exposure time.
- 25Kelly, P. M.; Åberg, C.; Polo, E.; O’Connell, A.; Cookman, J.; Fallon, J.; Krpetić, Ž.; Dawson, K. A. Mapping Protein Binding Sites on the Biomolecular Corona of Nanoparticles. Nature Nanotechnol. 2015, 10, 472– 479, DOI: 10.1038/nnano.2015.47Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXlsVOnsrw%253D&md5=7accf5d96e7e11cc3b5513bf2d0ed118Mapping protein binding sites on the biomolecular corona of nanoparticlesKelly, Philip M.; Aberg, Christoffer; Polo, Ester; O'Connell, Ann; Cookman, Jennifer; Fallon, Jonathan; Krpetic, Zeljka; Dawson, Kenneth A.Nature Nanotechnology (2015), 10 (5), 472-479CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)Nanoparticles in a biol. milieu are known to form a sufficiently long-lived and well-organized 'corona' of biomols. to confer a biol. identity to the particle. Because this nanoparticle-biomol. complex interacts with cells and biol. barriers, potentially engaging with different biol. pathways, it is important to clarify the presentation of functional biomol. motifs at its interface. Here, we demonstrate that by using antibody-labeled gold nanoparticles, differential centrifugal sedimentation and various imaging techniques it is possible to identify the spatial location of proteins, their functional motifs and their binding sites. We show that for transferrin-coated polystyrene nanoparticles only a minority of adsorbed proteins exhibit functional motifs and the spatial organization appears random, which is consistent, overall, with a stochastic and irreversible adsorption process. Our methods are applicable to a wide array of nanoparticles and can offer a microscopic mol. description of the biol. identity of nanoparticles.
- 26Herda, L. M.; Hristov, D. R.; Lo Giudice, M. C.; Polo, E.; Dawson, K. A. Mapping of Molecular Structure of the Nanoscale Surface in Bionanoparticles. J. Am. Chem. Soc. 2017, 139, 111– 114, DOI: 10.1021/jacs.6b12297Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFCgs7vL&md5=e168198d8ae5f0ebacabe3ffde2bb533Mapping of Molecular Structure of the Nanoscale Surface in BionanoparticlesHerda, Luciana M.; Hristov, Delyan R.; Lo Giudice, Maria Cristina; Polo, Ester; Dawson, Kenneth A.Journal of the American Chemical Society (2017), 139 (1), 111-114CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Characterizing the orientation of covalently conjugated proteins on nanoparticles, produced for in vitro and in vivo targeting, though an important feature of such a system, has proved challenging. Although extensive physicochem. characterization of targeting nanoparticles can be addressed in detail, relevant biol. characterization of the nanointerface is crucial to select suitable nanomaterials for further in vitro or in vivo expts. The authors adopt a methodol. using antibody fragments (Fab) conjugated to gold nanoparticles (immunogold) to map the available epitopes on a transferrin grafted silica particle (SiO2-PEG8-Tf) as a proxy methodol. to predict nanoparticle biol. function, and therefore cellular receptor engagement. Data from the adopted method suggest that, on av., only ∼3.5% of proteins grafted on the SiO2-PEG8-Tf nanoparticle surface have a favorable orientation for recognition by the cellular receptor.
- 27Lara, S.; Alnasser, F.; Polo, E.; Garry, D.; Lo Giudice, M. C.; Hristov, D. R.; Rocks, L.; Salvati, A.; Yan, Y.; Dawson, K. A. Identification of Receptor Binding to the Biomolecular Corona of Nanoparticles. ACS Nano 2017, 11, 1884– 1893, DOI: 10.1021/acsnano.6b07933Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVahu7Y%253D&md5=05f69ea88f149d67968bd9a8b7a69943Identification of Receptor Binding to the Biomolecular Corona of NanoparticlesLara, Sandra; Alnasser, Fatima; Polo, Ester; Garry, David; Lo Giudice, Maria Cristina; Hristov, Delyan R.; Rocks, Louise; Salvati, Anna; Yan, Yan; Dawson, Kenneth A.ACS Nano (2017), 11 (2), 1884-1893CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Biomols. adsorbed on nanoparticles are known to confer a biol. identity to nanoparticles, mediating the interactions with cells and biol. barriers. However, how these mols. are presented on the particle surface in biol. milieu remains unclear. The central aim of this study is to identify key protein recognition motifs and link them to specific cell-receptor interactions. Here, the authors employed an immuno-mapping technique to quantify epitope presentations of two major proteins in the serum corona, low-d. lipoprotein and IgG. Combined with a purpose-built receptor expression system, both proteins present functional motifs to allow simultaneous recognition by low-d. lipoprotein receptor and Fc-gamma receptor I of the corona. The authors' results suggest that the "labeling" of nanoparticles by biomol. adsorption processes allows for multiple pathways in biol. processes in which they may be "mistaken" for endogenous objects, such as lipoproteins, and exogenous ones, such as viral infections.
- 28Gianneli, M.; Polo, E.; Lopez, H.; Castagnola, V.; Aastrup, T.; Dawson, K. Label-Free in-Flow Detection of Receptor Recognition Motifs on the Biomolecular Corona of Nanoparticles. Nanoscale 2018, 10, 5474– 5481, DOI: 10.1039/C7NR07887KGoogle Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjvFWqur4%253D&md5=114cf2f49f7766a1a712d46db373cf44Label-free in-flow detection of receptor recognition motifs on the biomolecular corona of nanoparticlesGianneli, M.; Polo, E.; Lopez, H.; Castagnola, V.; Aastrup, T.; Dawson, K. A.Nanoscale (2018), 10 (12), 5474-5481CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)Nanomedicine, nanotargeting and nanotherapeutics have in the last few years faced several difficulties in translating the promising results obtained in vitro to an in vivo scenario. The origin of this discrepancy might be found in the lack of a detailed and realistic characterization of the biol. surface of nanoparticles. Despite the capability to engineer nanomaterials with a great variety and a precise control of the surface functionalization, the targeting capability is lost when the nanoparticles are embedded in complex biol. media, due to the formation of a biol. layer (biomol. corona). This biol. layer represents the ultimate nanoparticle surface, likely to interact with the cell machinery. Therefore, in addn. to traditional nanoparticle characterization techniques, a more insightful investigation of the biomol. corona is needed, including the capability to assess the orientation and functionality of specific key mol. features. Here we present a method for the rapid screening of exposed protein recognition motifs on the surface of nanoparticles exploiting quartz crystal microbalance (QCM). We quantify accessible functional epitopes of transferrin-coated nanoparticles and correlate them to differences in nanoparticle size and functionalization. The target recognition occurs label free in flow, thereby pushing our investigation into a more in vivo-like scenario. Our method is applicable to a wide array of nanoparticles and therefore holds the potential to become an advanced technique for the classification of all kinds of nanobioconstructs based on their biol. external functionality.
- 29Moein Moghimi, S.; Patel, H. M. Opsonophagocytosis of Liposomes by Peritoneal Macrophages and Bone Marrow Reticuloendothelial Cells. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research 1992, 1135, 269– 274, DOI: 10.1016/0167-4889(92)90230-9Google ScholarThere is no corresponding record for this reference.
- 30Papini, E.; Tavano, R.; Mancin, F. Opsonins and Dysopsonins of Nanoparticles: Facts, Concepts, and Methodological Guidelines. Frontiers in Immunology 2020, 11, 2343, DOI: 10.3389/fimmu.2020.567365Google ScholarThere is no corresponding record for this reference.
- 31Languino, L. R.; Duperray, A.; Joganic, K. J.; Fornaro, M.; Thornton, G. B.; Altieri, D. C. Regulation of Leukocyte-Endothelium Interaction and Leukocyte Transendothelial Migration by Intercellular Adhesion Molecule 1-Fibrinogen Recognition. Proc. Natl. Acad. Sci. U. S. A. 1995, 92, 1505– 1509, DOI: 10.1073/pnas.92.5.1505Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXktFWktbs%253D&md5=dbc15273eacdcfa66d31356f6ee8c030Regulation of leukocyte-endothelium interaction and leukocyte transendothelial migration by intercellular adhesion molecule 1-fibrinogen recognitionLanguino, Lucia R.; Duperray, Alain; Joganic, Karen J.; Fornaro, Mara; Thornton, George B.; Altieri, Dario C.Proceedings of the National Academy of Sciences of the United States of America (1995), 92 (5), 1505-9CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Although primarily recognized for its role in hemostasis, fibrinogen is also required for competent inflammatory reactions in vivo. It is now shown that fibrinogen promotes adhesions to and migration across an endothelial monolayer of terminally differentiated myelomonocytic cells. This process does not require chemotactic/haptotactic gradients or cytokine stimulation of the endothelium and is specific for the assocn. of fibrinogen with intercellular adhesion mol. 1 (ICAM-1) on endothelium. Among other adhesive plasma proteins, fibronectin fails to increase the binding of leukocytes to endothelium, or transendothelial migration, whereas vitronectin promotes the binding but not the migration. The fibrinogen-mediated leukocyte adhesion and transendothelial migration could be inhibited by a peptide from the fibrinogen γ-chain sequence N117NQKIVNLKEKVAQLEA133, which blocks the binding of fibrinogen to ICAM-1. This interaction could also be inhibited by new anti-ICAM-1 monoclonal antibodies that did not affect the ICAM-1-CD11a/CD18 recognition, thus suggesting that the fibrinogen binding site on ICAM-1 may be structurally distinct from regions previously implicated in leukocyte-endothelium interaction. Therefore, binding of fibrinogen to vascular cell receptors is sufficient to initiate (i) increased leukocyte adhesion to endothelium and (ii) leukocyte transendothelial migration. These two processes are the earliest events of immune inflammatory responses and may also contribute to atherosclerosis.
- 32Deng, Z. J.; Liang, M.; Monteiro, M.; Toth, I.; Minchin, R. F. Nanoparticle-Induced Unfolding of Fibrinogen Promotes Mac-1 Receptor Activation and Inflammation. Nat. Nanotechnol 2011, 6, 39– 44, DOI: 10.1038/nnano.2010.250Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhs1Wju73P&md5=de9f322b14e197e47e6b1fe034525498Nanoparticle-induced unfolding of fibrinogen promotes Mac-1 receptor activation and inflammationDeng, Zhou J.; Liang, Mingtao; Monteiro, Michael; Toth, Istvan; Minchin, Rodney F.Nature Nanotechnology (2011), 6 (1), 39-44CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)The chem. compn., size, shape and surface characteristics of nanoparticles affect the way proteins bind to these particles, and this in turn influences the way in which nanoparticles interact with cells and tissues. Nanomaterials bound with proteins can result in physiol. and pathol. changes, including macrophage uptake, blood coagulation, protein aggregation and complement activation, but the mechanisms that lead to these changes remain poorly understood. Here, we show that neg. charged poly(acrylic acid)-conjugated gold nanoparticles bind to and induce unfolding of fibrinogen, which promotes interaction with the integrin receptor, Mac-1. Activation of this receptor increases the NF-κB signalling pathway, resulting in the release of inflammatory cytokines. However, not all nanoparticles that bind to fibrinogen demonstrated this effect. Our results show that the binding of certain nanoparticles to fibrinogen in plasma offers an alternative mechanism to the more commonly described role of oxidative stress in the inflammatory response to nanomaterials.
- 33Deng, Z. J.; Liang, M.; Toth, I.; Monteiro, M. J.; Minchin, R. F. Molecular Interaction of Poly (Acrylic Acid) Gold Nanoparticles with Human Fibrinogen. ACS Nano 2012, 6, 8962– 8969, DOI: 10.1021/nn3029953Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhtl2iu7bI&md5=45a7894ab9195c99c6e9cbb5f7e6f758Molecular interaction of poly(acrylic acid) gold nanoparticles with human fibrinogenDeng, Zhou J.; Liang, Mingtao; Toth, Istvan; Monteiro, Michael J.; Minchin, Rodney F.ACS Nano (2012), 6 (10), 8962-8969CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)The binding of fibrinogen to various nanoparticles can result in protein unfolding and exposure of cryptic epitopes that subsequently interact with cell surface receptors. This response is dependent on the size, charge, and concn. of the nanoparticle. Here, the authors examd. the binding kinetics of human fibrinogen to neg. charged poly(acrylic acid)-coated Au-nanoparticles ranging in size from 7 to 22 nm. These particles have previously been shown to elicit an inflammatory response in human cells. The larger nanoparticles bound fibrinogen with increasing affinity and a slower dissocn. rate. Each fibrinogen mol. could accommodate 2 7-nm nanoparticles, but only one when the diam. increased to 10 nm. Nanoparticles of size >12 nm bound multiple fibrinogen mols. in a pos. cooperative manner. However, in the presence of excess nanoparticle, fibrinogen induced the aggregation of the larger particles that could bind more than one protein mol. This was consistent with interparticle bridging by the fibrinogen. Taken together, these results demonstrate that subtle changes in nanoparticle size can influence protein binding both with the surface of the nanoparticle and within the protein corona.
- 34Tenzer, S.; Docter, D.; Kuharev, J.; Musyanovych, A.; Fetz, V.; Hecht, R.; Schlenk, F.; Fischer, D.; Kiouptsi, K.; Reinhardt, C.; Landfester, K.; Schild, H.; Maskos, M.; Knauer, S. K.; Stauber, R. H. Rapid Formation of Plasma Protein Corona Critically Affects Nanoparticle Pathophysiology. Nature Nanotechnol. 2013, 8, 772– 781, DOI: 10.1038/nnano.2013.181Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVyqurrL&md5=927a38bd98a5850c71525a7707dc5073Rapid formation of plasma protein corona critically affects nanoparticle pathophysiologyTenzer, Stefan; Docter, Dominic; Kuharev, Joerg; Musyanovych, Anna; Fetz, Verena; Hecht, Rouven; Schlenk, Florian; Fischer, Dagmar; Kiouptsi, Klytaimnistra; Reinhardt, Christoph; Landfester, Katharina; Schild, Hansjoerg; Maskos, Michael; Knauer, Shirley K.; Stauber, Roland H.Nature Nanotechnology (2013), 8 (10), 772-781CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)In biol. fluids, proteins bind to the surface of nanoparticles to form a coating known as the protein corona, which can critically affect the interaction of the nanoparticles with living systems. As physiol. systems are highly dynamic, it is important to obtain a time-resolved knowledge of protein-corona formation, development and biol. relevancy. Here we show that label-free snapshot proteomics can be used to obtain quant. time-resolved profiles of human plasma coronas formed on silica and polystyrene nanoparticles of various size and surface functionalization. Complex time- and nanoparticle-specific coronas, which comprise almost 300 different proteins, were found to form rapidly (<0.5 min) and, over time, to change significantly in terms of the amt. of bound protein, but not in compn. Rapid corona formation is found to affect hemolysis, thrombocyte activation, nanoparticle uptake and endothelial cell death at an early exposure time.
- 35Tsuchiya, S.; Yamabe, M.; Yamaguchi, Y.; Kobayashi, Y.; Konno, T.; Tada, K. Establishment and Characterization of a Human Acute Monocytic Leukemia Cell Line (THP-1). International journal of cancer 1980, 26, 171– 176, DOI: 10.1002/ijc.2910260208Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL3M7ktFSisw%253D%253D&md5=ef2b5e1157ca18009c33fc80f183d5f9Establishment and characterization of a human acute monocytic leukemia cell line (THP-1)Tsuchiya S; Yamabe M; Yamaguchi Y; Kobayashi Y; Konno T; Tada KInternational journal of cancer (1980), 26 (2), 171-6 ISSN:0020-7136.A human leukemic cell line (THP-1) cultured from the blood of a boy with acute monocytic leukemia is described. This cell line had Fc and C3b receptors, but no surface or cytoplasmic immunoglobulins. HLA haplotypes of THP-1 were HLA-A2, -A9, -B5, -DRW1 and -DRW2. The monocytic nature of the cell line was characterized by: (1) the presence of alpha-naphthyl butyrate esterase activities which could be inhibited by NaF; (2) lysozyme production; (3) the phagocytosis of latex particles and sensitized sheep erythrocytes; and (4) the ability to restore T-lymphocyte response to Con A. The cells did not possess Epstein-Barr virus-associated nuclear antigen. These results indicate that THP-1 is a leukemia cell line with distinct monocytic markers. During culture, THP-1 maintained these monocytic characteristics for over 14 months.
- 36Kleiveland, C.; Verhoeckx, K.; Cotter, P.; López-Expósito, I.; Kleiveland, C.; Lea, T.; Mackie, A. Peripheral Blood Mononuclear Cells. In The Impact of Food Bioactives on Health: In Vitro and Ex Vivo Models; Springer: Cham, Switzerland, 2015; pp 161– 167, DOI: 10.1007/978-3-319-16104-4_15 .Google ScholarThere is no corresponding record for this reference.
- 37Giulimondi, F.; Digiacomo, L.; Pozzi, D.; Palchetti, S.; Vulpis, E.; Capriotti, A. L.; Chiozzi, R. Z.; Lagana, A.; Amenitsch, H.; Masuelli, L.; Peruzzi, G.; Mahmoudi, M.; Screpanti, I.; Zingoni, A.; Caracciolo, G. Interplay of Protein Corona and Immune Cells Controls Blood Residency of Liposomes. Nat. Commun. 2019, 10, 3686, DOI: 10.1038/s41467-019-11642-7Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1Sit7jJ&md5=dfcfc6cb1ec61aa332021ef39373243fInterplay of protein corona and immune cells controls blood residency of liposomesGiulimondi, Francesca; Digiacomo, Luca; Pozzi, Daniela; Palchetti, Sara; Vulpis, Elisabetta; Capriotti, Anna Laura; Chiozzi, Riccardo Zenezini; Lagana, Aldo; Amenitsch, Heinz; Masuelli, Laura; Mahmoudi, Morteza; Screpanti, Isabella; Zingoni, Alessandra; Caracciolo, GiulioNature Communications (2019), 10 (1), 3686CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)In vivo liposomes, like other types of nanoparticles, acquire a totally new 'biol. identity' due to the formation of a biomol. coating known as the protein corona that depends on and modifies the liposomes' synthetic identity. The liposome-protein corona is a dynamic interface that regulates the interaction of liposomes with the physiol. environment. Here we show that the biol. identity of liposomes is clearly linked to their sequestration from peripheral blood mononuclear cells (PBMCs) of healthy donors that ultimately leads to removal from the bloodstream. Pre-coating liposomes with an artificial corona made of human plasma proteins drastically reduces capture by circulating leukocytes in whole blood and may be an effective strategy to enable prolonged circulation in vivo. We conclude with a crit. assessment of the key concepts of liposome technol. that need to be reviewed for its definitive clin. translation.
- 38Chung, A.; Gao, Q.; Kao, W. J. Macrophage Matrix Metalloproteinase-2/-9 Gene and Protein Expression Following Adhesion to ECM-Derived Multifunctional Matrices Via Integrin Complexation. Biomaterials 2007, 28, 285– 298, DOI: 10.1016/j.biomaterials.2006.08.038Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD28nht1Gkuw%253D%253D&md5=69e547f4029caf5cc4806ef0e8384036Macrophage matrix metalloproteinase-2/-9 gene and protein expression following adhesion to ECM-derived multifunctional matrices via integrin complexationChung Amy; Gao Qiang; Kao Weiyuan JohnBiomaterials (2007), 28 (2), 285-98 ISSN:0142-9612.Macrophages are commonly observed at the biomaterial-tissue interface and interact with the extracellular matrix (ECM) mainly by integrin receptors to play a critical role in ECM turnover by secreting matrix metalloproteinases (MMPs). To investigate beta1 and beta3 containing integrin-mediated adhesion and subsequent MMP-2/-9 protein and gene expression in human blood-derived monocytes, biofunctional peptides immobilized onto flexible polyethylene glycol (PEG) arms were grafted onto a gelatin-based interpenetrating network (IPN). Adherent monocyte density was dramatically greater in the presence of RGD immobilized onto flexible PEG arms of the gelatin-based IPN. Pretreatment of monocytes with either anti-integrin beta1 or beta3 led to a significant decrease in adherent cell density on RGD-PEG-grafted IPNs. MMP-2 and MMP-9 protein and MMP-9 mRNA expression increased in the presence of IPNs initially, independent of ligand identity. Anti-integrin beta1 or beta3 antibody pretreatment of monocytes led to a general decrease in MMP-2/-9 protein expression. These results demonstrate the importance of beta1 and beta3 containing integrins in mediating monocyte adhesion onto RGD immobilized onto flexible PEG arms of the IPN. The results also reveal that MMP-2/-9 protein and gene expression is influenced by the presence of gelatin and not the ligands immobilized on the PEG arms of the IPN.
- 39Simon, J.; Müller, L. K.; Kokkinopoulou, M.; Lieberwirth, I.; Morsbach, S.; Landfester, K.; Mailänder, V. Exploiting the Biomolecular Corona: Pre-Coating of Nanoparticles Enables Controlled Cellular Interactions. Nanoscale 2018, 10, 10731– 10739, DOI: 10.1039/C8NR03331EGoogle Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVSgtbnL&md5=e5342d5993c7d20829636788e9ba7907Exploiting the biomolecular corona: pre-coating of nanoparticles enables controlled cellular interactionsSimon, Johanna; Mueller, Laura K.; Kokkinopoulou, Maria; Lieberwirth, Ingo; Morsbach, Svenja; Landfester, Katharina; Mailaender, VolkerNanoscale (2018), 10 (22), 10731-10739CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)Formation of the biomol. corona ultimately dets. the successful application of nanoparticles in vivo. Adsorption of biomols. such as proteins is an inevitable process that takes place instantaneously upon contact with physiol. fluid (e.g. blood). Therefore, strategies are needed to control this process in order to improve the properties of the nanoparticles and to allow targeted drug delivery. Here, we show that the design of the protein corona by a pre-formed protein corona with tailored properties enables targeted cellular interactions. Nanoparticles were pre-coated with Ig depleted plasma to create and design a protein corona that reduces cellular uptake by immune cells. It was proven that a pre-formed protein corona remains stable even after nanoparticles were re-introduced to plasma. This opens up the great potential to exploit protein corona formation, which will significantly influence the development of novel nanomaterials.
- 40Suk, J. S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L. M. PEGylation As a Strategy for Improving Nanoparticle-Based Drug and Gene Delivery. Adv. Drug Delivery Rev. 2016, 99, 28– 51, DOI: 10.1016/j.addr.2015.09.012Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhs1CqurrP&md5=fecfbdedd72ee97571c913a9ac3cd93fPEGylation as a strategy for improving nanoparticle-based drug and gene deliverySuk, Jung Soo; Xu, Qingguo; Kim, Namho; Hanes, Justin; Ensign, Laura M.Advanced Drug Delivery Reviews (2016), 99 (Part_A), 28-51CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)Coating the surface of nanoparticles with polyethylene glycol (PEG), or "PEGylation", is a commonly used approach for improving the efficiency of drug and gene delivery to target cells and tissues. Building from the success of PEGylating proteins to improve systemic circulation time and decrease immunogenicity, the impact of PEG coatings on the fate of systemically administered nanoparticle formulations has, and continues to be, widely studied. PEG coatings on nanoparticles shield the surface from aggregation, opsonization, and phagocytosis, prolonging systemic circulation time. Here, we briefly describe the history of the development of PEGylated nanoparticle formulations for systemic administration, including how factors such as PEG mol. wt., PEG surface d., nanoparticle core properties, and repeated administration impact circulation time. A less frequently discussed topic, we then describe how PEG coatings on nanoparticles have also been utilized for overcoming various biol. barriers to efficient drug and gene delivery assocd. with other modes of administration, ranging from gastrointestinal to ocular. Finally, we describe both methods for PEGylating nanoparticles and methods for characterizing PEG surface d., a key factor in the effectiveness of the PEG surface coating for improving drug and gene delivery.
- 41Ishida, T.; Ichihara, M.; Wang, X.; Kiwada, H. Spleen Plays an Important Role in the Induction of Accelerated Blood Clearance of PEGylated Liposomes. J. Controlled Release 2006, 115, 243– 250, DOI: 10.1016/j.jconrel.2006.08.001Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFeitr3K&md5=d8f3548771bd68147d1e85f7cf36af52Spleen plays an important role in the induction of accelerated blood clearance of PEGylated liposomesIshida, Tatsuhiro; Ichihara, Masako; Wang, XinYu; Kiwada, HiroshiJournal of Controlled Release (2006), 115 (3), 243-250CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)It is well known that steric stabilization of the surface of liposomes by a polyethylene glycol (PEG) conjugated lipid results in reduced recognition of the liposomes by the cells of the mononuclear phagocyte system and consequently extended circulation times of the liposomes (t1/2 ≈ 20 h in rat). Recently, we reported on the "accelerated blood clearance (ABC) phenomenon", causing PEGylated liposomes to be cleared very rapidly from the circulation upon repeated injection. We also reported that abundant binding of IgM, secreted into the blood stream after the first dose and, to PEGylated liposomes, plays an essential role in the induction of the ABC phenomenon. Spleen is well known to play a central role in the immune reaction and to produce IgM following a bacterial infection. The aim of the present study was to det. whether spleen contributes to the induction of the ABC phenomenon and to unravel its role in the phenomenon. In rats that were splenectomized (surgical removal of spleen) prior to the first injection of liposomes (0.001 μmol phospholipids/kg), the ABC phenomenon was totally abolished. In these rats serum IgM concns. as well as the amts. of IgM bound to PEGylated liposomes were substantially reduced. Splenectomy attenuated the ABC phenomenon when performed until 3 days post-first injection. Removal of the spleen 4 days post-first injection left the ABC phenomenon unchanged. This finding indicates that the immune reaction in the spleen against the PEGylated liposomes occurs during at least 2-3 days following the first administration and then IgM reactive to PEGylated liposomes is produced. The present study proves that the spleen plays a crit. role in the induction phase of the ABC phenomenon. For effective clin. application, many liposomal drug formulations will require multiple injections. The ABC phenomenon described in this and several preceding papers therefore has important implications for the development and evaluation of therapeutically useful liposomal formulations requiring multiple-dose administration.
- 42Hadjidemetriou, M.; Al-Ahmady, Z.; Mazza, M.; Collins, R. F.; Dawson, K.; Kostarelos, K. In Vivo Biomolecule Corona Around Blood-Circulating, Clinically Used and Antibody-Targeted Lipid Bilayer Nanoscale Vesicles. ACS Nano 2015, 9, 8142– 8156, DOI: 10.1021/acsnano.5b03300Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFWgsrjN&md5=c8402b491c8438400f0fea698c6824cbIn Vivo Biomolecule Corona around Blood-Circulating, Clinically Used and Antibody-Targeted Lipid Bilayer Nanoscale VesiclesHadjidemetriou, Marilena; Al-Ahmady, Zahraa; Mazza, Mariarosa; Collins, Richard F.; Dawson, Kenneth; Kostarelos, KostasACS Nano (2015), 9 (8), 8142-8156CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)The adsorption of proteins and their layering onto nanoparticle surfaces has been called the "protein corona". This dynamic process of protein adsorption has been extensively studied following in vitro incubation of many different nanoparticles with plasma proteins. However, the formation of protein corona under dynamic, in vivo conditions remains largely unexplored. Extrapolation of in vitro formed protein coronas to predict the fate and possible toxicol. burden from nanoparticles in vivo is of great interest. However, complete lack of such direct comparisons for clin. used nanoparticles makes the study of in vitro and in vivo formed protein coronas of great importance. Our aim was to study the in vivo protein corona formed onto i.v. injected, clin. used liposomes, based on the compn. of the PEGylated liposomal formulation that constitutes the anticancer agent Doxil. The formation of in vivo protein corona was detd. after the recovery of the liposomes from the blood circulation of CD-1 mice 10 min postinjection. In comparison, in vitro protein corona was formed by the incubation of liposomes in CD-1 mouse plasma. In vivo and in vitro formed protein coronas were compared in terms of morphol., compn. and cellular internalization. The protein coronas on bare (non-PEGylated) and monoclonal antibody (IgG) targeted liposomes of the same lipid compn. were also comparatively investigated. A network of linear fibrillary structures constituted the in vitro formed protein corona, whereas the in vivo corona had a different morphol. but did not appear to coat the liposome surface entirely. Even though the total amt. of protein attached on circulating liposomes correlated with that obsd. from in vitro incubations, the variety of mol. species in the in vivo corona were considerably wider. Both in vitro and in vivo formed protein coronas were found to significantly reduce receptor binding and cellular internalization of antibody-conjugated liposomes; however, the in vivo corona formation did not lead to complete ablation of their targeting capability.
- 43Hadjidemetriou, M.; Al-Ahmady, Z.; Kostarelos, K. Time-Evolution of In Vivo Protein Corona onto Blood-Circulating PEGylated Liposomal Doxorubicin (DOXIL) Nanoparticles. Nanoscale 2016, 8, 6948– 6957, DOI: 10.1039/C5NR09158FGoogle Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xjs1Gisb0%253D&md5=ea5ffb6e06758c99fce6a74f22e47f94Time-evolution of in vivo protein corona onto blood-circulating PEGylated liposomal doxorubicin (DOXIL) nanoparticlesHadjidemetriou, Marilena; Al-Ahmady, Zahraa; Kostarelos, KostasNanoscale (2016), 8 (13), 6948-6957CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)Nanoparticles (NPs) are instantly modified once injected in the bloodstream because of their interaction with the blood components. The spontaneous coating of NPs by proteins, once in contact with biol. fluids, has been termed the 'protein corona' and it is considered to be a determinant factor for the pharmacol., toxicol. and therapeutic profile of NPs. Protein exposure time is thought to greatly influence the compn. of protein corona, however the dynamics of protein interactions under realistic, in vivo conditions remain unexplored. The aim of this study was to quant. and qual. investigate the time evolution of in vivo protein corona, formed onto blood circulating, clin. used, PEGylated liposomal doxorubicin. Protein adsorption profiles were detd. 10 min, 1 h and 3 h post-injection of liposomes into CD-1 mice. The results demonstrated that a complex protein corona was formed as early as 10 min post-injection. Even though the total amt. of protein adsorbed did not significantly change over time, the fluctuation of protein abundances obsd. indicated highly dynamic protein binding kinetics.
- 44Hadjidemetriou, M.; McAdam, S.; Garner, G.; Thackeray, C.; Knight, D.; Smith, D.; Al-Ahmady, Z.; Mazza, M.; Rogan, J.; Clamp, A.; Kostarelos, K. The Human In Vivo Biomolecule Corona onto PEGylated liposomes: a Proof-of-Concept Clinical Study. Adv. Mater. 2019, 31, 1803335, DOI: 10.1002/adma.201803335Google ScholarThere is no corresponding record for this reference.
- 45Cerullo, V.; Seiler, M. P.; Mane, V.; Brunetti-Pierri, N.; Clarke, C.; Bertin, T. K.; Rodgers, J. R.; Lee, B. Toll-Like Receptor 9 Triggers an Innate Immune Response to Helper-Dependent Adenoviral Vectors. Molecular Therapy 2007, 15, 378– 385, DOI: 10.1038/sj.mt.6300031Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtVejtbfJ&md5=229648334245ab0ae325af09963fd70eToll-like Receptor 9 Triggers an Innate Immune Response to Helper-dependent Adenoviral VectorsCerullo, Vincenzo; Seiler, Michael P.; Mane, Viraj; Brunetti-Pierri, Nicola; Clarke, Christian; Bertin, Terry K.; Rodgers, John R.; Lee, BrendanMolecular Therapy (2007), 15 (2), 378-385CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)A major obstacle to the clin. application of systemic adenoviral gene replacement therapy is the host innate immune response. Although recent studies have attempted to characterize the cellular basis for this response to systemically administered helper-dependent adenoviral vector (HD-Ad), the underlying mol. components of the innate immune repertoire required to recognize the viral vector have yet to be identified. Here, the authors show that primary macrophages can sense HD-Ad vectors via the Toll-like Receptor 9 (TLR9) and respond by increasing pro-inflammatory cytokine secretion. Moreover, TLR9 sensing is involved in the rapid innate immune response to HD-Ad in vivo. TLR9 deficiency attenuates the innate immune response to HD-Ad, whereas TLR9 blockade reduces the acute inflammatory response after i.v. injection of the vector. Moreover, HD-Ad upregulates TLR9 gene expression independent of TLR9 function, suggesting that addnl. innate signaling pathways work cooperatively with TLR9. The identification of the components of the innate immune response to adenovirus will facilitate the development of combinatorial therapy directed at increasing the maximal tolerated dose of systemically delivered adenoviral vectors.
- 46Moghimi, S. M.; Patel, H. M. Tissue Specific Opsonins for Phagocytic Cells and Their Different Affinity for Cholesterol-Rich Liposomes. FEBS letters 1988, 233, 143– 147, DOI: 10.1016/0014-5793(88)81372-3Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1cXktlKntLs%253D&md5=a8ce77eac7904341196b88e9a102888aTissue specific opsonins for phagocytic cells and their different affinity for cholesterol-rich liposomesMoghimi, S. Moein; Patel, Harish M.FEBS Letters (1988), 233 (1), 143-7CODEN: FEBLAL; ISSN:0014-5793.In vivo expts. show that Kupffer cells avidly take up cholesterol-poor but not cholesterol-rich liposomes, whereas splenic phagocytic cells take up preferentially cholesterol-rich rather than cholesterol-poor liposomes in the presence of serum. Apparently, serum contains opsonins specific for hepatic and splenic phagocytic cells and these opsonins have different affinities for cholesterol-rich and cholesterol-poor liposomes.
- 47Moghimi, S. M.; Simberg, D.; Skotland, T.; Yaghmur, A.; Hunter, C. The Interplay between Blood Proteins, Complement, and Macrophages on Nanomedicine Performance and Responses. Journal of Pharmacology and Experimental Therapeutics 2019, 370, 581– 592, DOI: 10.1124/jpet.119.258012Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisVCnsr7P&md5=b1abe6a7abcbf81f7b089d56a90f69e0The interplay between blood proteins, complement, and macrophages on nanomedicine performance and responsesMoghimi, S. Moein; Simberg, Dmitri; Skotland, Tore; Yaghmur, Anan; Hunter, A. ChristyJournal of Pharmacology and Experimental Therapeutics (2019), 370 (3), 581-592CODEN: JPETAB; ISSN:1521-0103. (American Society for Pharmacology and Experimental Therapeutics)A review. In the blood, depending on their physicochem. characteristics, nanoparticles attract a wide range of plasma biomols. The majority of blood biomols. bind nonspecifically to nanoparticles. On the other hand, biomols. such as pattern-recognition complement-sensing proteins may recognize some structural determinants of the pristine surface, causing complement activation. Adsorption of nonspecific blood proteins could also recruit natural antibodies and initiate complement activation, and this seems to be a global process with many preclin. and clin. nanomedicines. We discuss these issues, since complement activation has ramifications in nanomedicine stability and pharmacokinetics, as well as in inflammation and disease progression. Some studies have also predicted a role for complement systems in infusion-related reactions, whereas others show a direct role for macrophages and other immune cells independent of complement activation. We comment on these discrepancies and suggest directions for exploring the underlying mechanisms.
- 48Verhoef, J.; Anchordoquy, T. Drug Delivery Transl. Res. 2013, 3, 499– 503, DOI: 10.1007/s13346-013-0176-5Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslCmtLrF&md5=6ec259f3c3bd2d3b4dda73c3d75d1c9aQuestioning the use of PEGylation for drug deliveryVerhoef, Johan J. F.; Anchordoquy, Thomas J.Drug Delivery and Translational Research (2013), 3 (6), 499-503CODEN: DDTRCY; ISSN:2190-3948. (Springer)A review. Polyethylene glycol (PEG) is widely utilized in drug delivery and nanotechnol. due to its reported "stealth" properties and biocompatibility. It is generally thought that PEGylation allows particulate delivery systems and biomaterials to evade the immune system and thereby prolong circulation lifetimes. However, numerous studies over the past decade have demonstrated that PEGylation causes significant redns. in drug delivery, including enhanced serum protein binding, reduced uptake by target cells, and the elicitation of an immune response that facilitates clearance in vivo. This report reviews some of the extensive literature documenting the detrimental effects of PEGylation, and thereby questions the wisdom behind employing this strategy in drug development.
- 49Song, J.; Ju, Y.; Amarasena, T. H.; Lin, Z.; Mettu, S.; Zhou, J.; Rahim, M. A.; Ang, C.-S.; Cortez-Jugo, C.; Kent, S. J.; Caruso, F. Influence of Poly (Ethylene Glycol) Molecular Architecture on Particle Assembly and Ex Vivo Particle–Immune Cell Interactions in Human Blood. ACS Nano 2021, 15, 10025, DOI: 10.1021/acsnano.1c01642Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2c%252FmsVygsQ%253D%253D&md5=684f7871c88d26995c23227cddc1b8fdInfluence of Poly(ethylene glycol) Molecular Architecture on Particle Assembly and Ex Vivo Particle-Immune Cell Interactions in Human BloodSong Jiaying; Ju Yi; Lin Zhixing; Zhou Jiajing; Rahim Md Arifur; Cortez-Jugo Christina; Caruso Frank; Amarasena Thakshila H; Kent Stephen J; Mettu Srinivas; Ang Ching-SengACS nano (2021), 15 (6), 10025-10038 ISSN:.Poly(ethylene glycol) (PEG) is widely used in particle assembly to impart biocompatibility and stealth-like properties in vivo for diverse biomedical applications. Previous studies have examined the effect of PEG molecular weight and PEG coating density on the biological fate of various particles; however, there are few studies that detail the fundamental role of PEG molecular architecture in particle engineering and bio-nano interactions. Herein, we engineered PEG particles using a mesoporous silica (MS) templating method and investigated how the PEG building block architecture impacted the physicochemical properties (e.g., surface chemistry and mechanical characteristics) of the PEG particles and subsequently modulated particle-immune cell interactions in human blood. Varying the PEG architecture from 3-arm to 4-arm, 6-arm, and 8-arm generated PEG particles with a denser, stiffer structure, with increasing elastic modulus from 1.5 to 14.9 kPa, inducing an increasing level of immune cell association (from 15% for 3-arm to 45% for 8-arm) with monocytes. In contrast, the precursor PEG particles with the template intact (MS@PEG) were stiffer and generally displayed higher levels of immune cell association but showed the opposite trend-immune cell association decreased with increasing PEG arm numbers. Proteomics analysis demonstrated that the biomolecular corona that formed on the PEG particles minimally influenced particle-immune cell interactions, whereas the MS@PEG particle-cell interactions correlated with the composition of the corona that was abundant in histidine-rich glycoproteins. Our work highlights the role of PEG architecture in the design of stealth PEG-based particles, thus providing a link between the synthetic nature of particles and their biological behavior in blood.
- 50Kristensen, K.; Urquhart, A. J.; Thormann, E.; Andresen, T. L. Binding of Human Serum Albumin to PEGylated Liposomes: Insights into Binding Numbers and Dynamics by Fluorescence Correlation Spectroscopy. Nanoscale 2016, 8, 19726– 19736, DOI: 10.1039/C6NR05455BGoogle Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xhs1yku7zO&md5=791626aae556dd6a559a79373b16e80dBinding of human serum albumin to PEGylated liposomes: insights into binding numbers and dynamics by fluorescence correlation spectroscopyKristensen, Kasper; Urquhart, Andrew J.; Thormann, Esben; Andresen, Thomas L.Nanoscale (2016), 8 (47), 19726-19736CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)Liposomes for medical applications are often administered by i.v. injection. Once in the bloodstream, the liposomes are covered with a "protein corona", which impacts the behavior and eventual fate of the liposomes. Currently, many aspects of the liposomal protein corona are not well understood. For example, there is generally a lack of knowledge about the liposome binding affinities and dynamics of common types of blood plasma proteins. Fluorescence correlation spectroscopy (FCS) is a powerful exptl. technique that potentially can provide such knowledge. In this study, we have used FCS to investigate the binding of human serum albumin (HSA) to std. types of PEGylated fluid-phase liposomes (consisting of DOPC and DOPE-PEG2k) and PEGylated gel-phase liposomes (consisting of DSPC and DSPE-PEG2k) with various PEG chain surface densities. We detected no significant binding of HSA to the PEGylated fluid-phase liposomes. In contrast, we found that HSA bound tightly to the PEGylated gel-phase liposomes, although only a low no. of HSA mols. could be accommodated per liposome. Overall, we believe that our data provides a useful benchmark for other researchers interested in studying the liposomal protein corona.
- 51Jhaveri, A.; Deshpande, P.; Pattni, B.; Torchilin, V. Transferrin-Targeted, Resveratrol-Loaded Liposomes for the Treatment of Glioblastoma. Journal of controlled release 2018, 277, 89– 101, DOI: 10.1016/j.jconrel.2018.03.006Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXlt1yjurw%253D&md5=bd9a0331761b9dcc86c394ea852e3875Transferrin-targeted, resveratrol-loaded liposomes for the treatment of glioblastomaJhaveri, Aditi; Deshpande, Pranali; Pattni, Bhushan; Torchilin, VladimirJournal of Controlled Release (2018), 277 (), 89-101CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Glioblastomas (GBMs) are highly aggressive brain tumors with a very grim prognosis even after multi-modal therapeutic regimens. Conventional chemotherapeutic agents frequently lead to drug resistance and result in severe toxicities to non-cancerous tissues. Resveratrol (RES), a natural polyphenol with pleiotropic health benefits, has proven chemopreventive effects in all the stages of cancer including initiation, promotion and progression. However, the poor physico-chem. properties of RES severely limit its use as a free drug. In this study, RES was loaded into PEGylated liposomes (RES-L) to counter its drawbacks as a free drug. Since transferrin receptors (TfRs) are up-regulated in GBM, the liposome surface was modified with transferrin moieties (Tf-RES-L) to make them cancer cell-specific. The liposomal nanomedicines developed in this project were aimed at enhancing the physico-chem. properties of RES and exploiting the passive and active targeting capabilities of liposomes to effectively treat GBM. The RES-L were stable, had a good drug-loading capacity, prolonged drug-release in vitro and were easily scalable. Flow cytometry and confocal microscopy were used to study the assocn. with, and internalization of, Tf-L into U-87 MG cells. The Tf-RES-Ls were significantly more cytotoxic and induced higher levels of apoptosis accompanied by activation of caspases 3/7 in GBM cells when compared to free RES or RES-L. The ability of RES to arrest cells in the S-phase of the cell cycle, and selectively induce prodn. of reactive oxygen species in cancer cells were probably responsible for its cytotoxic effects. The therapeutic efficacy of RES formulations was evaluated in a s.c. xenograft mouse model of GBM. A tumor growth inhibition study and a modified survival study showed that Tf-RES-Ls were more effective than other treatments in their ability to inhibit tumor growth and improve survival in mice. Overall, the liposomal nanomedicines of RES developed in this project exhibited favorable in vitro and in vivo efficacies, which warrant their further investigation for the treatment of GBMs.
- 52Moyer, L. S.; Gorin, M. H. Eletectrokinetic Aspects of Surface Chemistry: IX. The Electric Mobilitites of Quartz and Collodion Particles in Mixtures of Horse Serum and Serum Proteins in Relation to the Mechanisms of Film Formation. J. Biol. Chem. 1940, 133, 605– 619, DOI: 10.1016/S0021-9258(18)73343-7Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaH3cXis1Knsg%253D%253D&md5=697ea21300760c5b8cd4b6c49c76a888Electrokinetic aspects of surface chemistry. IX. Electric mobilities of quartz and collodion particles in mixtures of horse serum and serum proteins in relation to the mechanism of film formationMoyer, Laurence S.; Gorin, Manuel H.Journal of Biological Chemistry (1940), 133 (), 605-19CODEN: JBCHA3; ISSN:0021-9258.cf. C. A. 34, 1696.3. The electrophoretic mobilities of quartz and collodion particles were detd. after exposure to mixts. of serum proteins or after they had been coated with one protein and then exposed to another or to serum. There is little tendency for the various constituents used to adsorb on each other, but one protein may replace another at a surface; thus a film of one of the protein components is obtained rather than a mosaic. The nature of the underlying surface influences the adsorption, the more hydrophilic proteins being adsorbed more readily by the more hydrophilic surfaces, and vice versa. Globulin when coated on quartz appears to be either irreversibly adsorbed or the most strongly adsorbed of the three proteins, replacement by albumin not occurring. The tendency to be adsorbed on quartz is roughly in the order albumin A > albumin B = globulin, with the addnl. assumption that globulin can replace the albumins whenever the free energy differences are favorable. On collodion, globulin is more strongly adsorbed than albumin A or B. The biol. significance of these findings is discussed.
- 53Caracciolo, G.; Cardarelli, F.; Pozzi, D.; Salomone, F.; Maccari, G.; Bardi, G.; Capriotti, A. L.; Cavaliere, C.; Papi, M.; Laganà, A. Selective Targeting Capability Acquired with a Protein Corona Adsorbed on the Surface of 1,2-Dioleoyl-3-Trimethylammonium Propane/DNA Nanoparticles. ACS Appl. Mater. Interfaces 2013, 5, 13171– 13179, DOI: 10.1021/am404171hGoogle Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsl2nsbjI&md5=0a989575d1ad35338a3689251a70dcb4Selective Targeting Capability Acquired with a Protein Corona Adsorbed on the Surface of 1,2-Dioleoyl-3-trimethylammonium Propane/DNA NanoparticlesCaracciolo, Giulio; Cardarelli, Francesco; Pozzi, Daniela; Salomone, Fabrizio; Maccari, Giuseppe; Bardi, Giuseppe; Capriotti, Anna Laura; Cavaliere, Chiara; Papi, Massimiliano; Lagana, AldoACS Applied Materials & Interfaces (2013), 5 (24), 13171-13179CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)A possible turning point in drug delivery has been recently reached: the protein shell, which covers nanocarriers in vivo, can be used for targeting. Here, we show that nanoparticles can acquire a selective targeting capability with a protein corona adsorbed on the surface. We demonstrate that lipid particles made of 1,2-dioleoyl-3-trimethylammonium propane (DOTAP) and DNA, upon interaction with human plasma components, spontaneously become coated with vitronectin that promotes efficient uptake in cancer cells expressing high levels of the vitronectin ανβ3 integrin receptor.
- 54Digiacomo, L.; Cardarelli, F.; Pozzi, D.; Palchetti, S.; Digman, M.; Gratton, E.; Capriotti, A.; Mahmoudi, M.; Caracciolo, G. An Apolipoprotein-Enriched Biomolecular Corona Switches the Cellular Uptake Mechanism and Trafficking Pathway of Lipid Nanoparticles. Nanoscale 2017, 9, 17254– 17262, DOI: 10.1039/C7NR06437CGoogle Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1yjt7zN&md5=d533413573a757ffaeea4b52dc2bf837An apolipoprotein-enriched biomolecular corona switches the cellular uptake mechanism and trafficking pathway of lipid nanoparticlesDigiacomo, L.; Cardarelli, F.; Pozzi, D.; Palchetti, S.; Digman, M. A.; Gratton, E.; Capriotti, A. L.; Mahmoudi, M.; Caracciolo, G.Nanoscale (2017), 9 (44), 17254-17262CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)Following exposure to biol. milieus (e.g. after systemic administration), nanoparticles (NPs) get covered by an outer biomol. corona (BC) that defines many of their biol. outcomes, such as the elicited immune response, biodistribution, and targeting abilities. In spite of this, the role of BC in regulating the cellular uptake and the subcellular trafficking properties of NPs has remained elusive. Here, we tackle this issue by employing multicomponent (MC) lipid NPs, human plasma (HP) and HeLa cells as models for nanoformulations, biol. fluids, and target cells, resp. By conducting confocal fluorescence microscopy expts. and image correlation analyses, we quant. demonstrate that the BC promotes a neat switch of the cell entry mechanism and subsequent intracellular trafficking, from macropinocytosis to clathrin-dependent endocytosis. Nano-liq. chromatog. tandem mass spectrometry identifies apolipoproteins as the most abundant components of the BC tested here. Interestingly, this class of proteins target the LDL receptors, which are overexpressed in clathrin-enriched membrane domains. Our results highlight the crucial role of BC as an intrinsic trigger of specific NP-cell interactions and biol. responses and set the basis for a rational exploitation of the BC for targeted delivery.
- 55Digiacomo, L.; Giulimondi, F.; Capriotti, A. L.; Piovesana, S.; Montone, C. M.; Chiozzi, R. Z.; Laganà, A.; Mahmoudi, M.; Pozzi, D.; Caracciolo, G. Optimal Centrifugal Isolating of Liposome–Protein Complexes From Human Plasma. Nanoscale Advances 2021, 3, 3824, DOI: 10.1039/D1NA00211BGoogle Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtV2ksb3O&md5=b8e3d0d0602076e429d625e96bd89d2bOptimal centrifugal isolating of liposome-protein complexes from human plasmaDigiacomo, Luca; Giulimondi, Francesca; Capriotti, Anna Laura; Piovesana, Susy; Montone, Carmela Maria; Chiozzi, Riccardo Zenezini; Lagana, Aldo; Mahmoudi, Morteza; Pozzi, Daniela; Caracciolo, GiulioNanoscale Advances (2021), 3 (13), 3824-3834CODEN: NAADAI; ISSN:2516-0230. (Royal Society of Chemistry)In the past few years, characterization of the protein corona (PC) that forms around liposomal systems has gained increasing interest for the development of novel therapeutic and diagnostic technologies. At the crossroads of fast-moving research fields, the interdisciplinarity of protein corona investigations poses challenges for exptl. design and reporting. Isolation of liposome-protein complexes from biol. fluids has been identified as a fundamental step of the entire workflow of PC characterization but exact specifications for conditions to optimize pelleting remain elusive. In the present work, key factors affecting pptn. of liposome-protein complexes by centrifugation, including time of centrifugation, total sample vol., lipid : protein ratio and contamination from biol. NPs were comprehensively evaluated. Here we show that the total amt. of isolated liposome-protein complexes and the extent of contamination from biol. NPs may vary with influence factors. Our results provide protein corona researchers with precise indications to sep. liposome-protein complexes from protein-rich fluids and include proper controls, thus they are anticipated to catalyze improved consistency of data mining and computational modeling of protein corona compn.
- 56Franciosa, G; Diluvio, G; Gaudio, F D.; Giuli, M V; Palermo, R; Grazioli, P; Campese, A F; Talora, C; Bellavia, D; D'Amati, G; Besharat, Z M; Nicoletti, C; Siebel, C W; Choy, L; Rustighi, A; Sal, G D.; Screpanti, I; Checquolo, S Prolyl-Isomerase Pin1 Controls Notch3 Protein Expression and Regulates T-ALL Progression. Oncogene 2016, 35, 4741– 4751, DOI: 10.1038/onc.2016.5Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XisFWqsL4%253D&md5=956448aa6da4918fc70930ce436fca72Prolyl-isomerase Pin1 controls Notch3 protein expression and regulates T-ALL progressionFranciosa, G.; Diluvio, G.; Del Gaudio, F.; Giuli, M. V.; Palermo, R.; Grazioli, P.; Campese, A. F.; Talora, C.; Bellavia, D.; D'Amati, G.; Besharat, Z. M.; Nicoletti, C.; Siebel, C. W.; Choy, L.; Rustighi, A.; Sal, G. Del; Screpanti, I.; Checquolo, S.Oncogene (2016), 35 (36), 4741-4751CODEN: ONCNES; ISSN:0950-9232. (Nature Publishing Group)Deregulated Notch signaling is assocd. with T-cell Acute Lymphoblastic Leukemia (T-ALL) development and progression. Increasing evidence reveals that Notch pathway has an important role in the invasion ability of tumor cells, including leukemia, although the underlying mol. mechanisms remain mostly unclear. Here, we show that Notch3 is a novel target protein of the prolyl-isomerase Pin1, which is able to regulate Notch3 protein processing and to stabilize the cleaved product, leading to the increased expression of the intracellular domain (N3IC), finally enhancing Notch3-dependent invasiveness properties. We demonstrate that the combined inhibition of Notch3 and Pin1 in the Notch3-overexpressing human leukemic TALL-1 cells reduces their high invasive potential, by decreasing the expression of the matrix metalloprotease MMP9. Consistently, Pin1 depletion in a mouse model of Notch3-induced T-ALL, by reducing N3IC expression and signaling, impairs the expansion/invasiveness of CD4+CD8+ DP cells in peripheral lymphoid and non-lymphoid organs. Notably, in in silico gene expression anal. of human T-ALL samples we obsd. a significant correlation between Pin1 and Notch3 expression levels, which may further suggest a key role of the newly identified Notch3-Pin1 axis in T-ALL aggressiveness and progression. Thus, combined suppression of Pin1 and Notch3 proteins may be exploited as an addnl. target therapy for T-ALL.
Cited By
This article is cited by 40 publications.
- Luca Digiacomo, Serena Renzi, Andrea Pirrottina, Heinz Amenitsch, Valentina De Lorenzi, Daniela Pozzi, Francesco Cardarelli, Giulio Caracciolo. PEGylation-Dependent Cell Uptake of Lipid Nanoparticles Revealed by Spatiotemporal Correlation Spectroscopy. ACS Pharmacology & Translational Science 2024, 7
(10)
, 3004-3010. https://doi.org/10.1021/acsptsci.4c00419
- Yan Zhao, Junjun Hou, Linjie Guo, Shitai Zhu, Xiaoling Hou, Shuting Cao, Mo Zhou, Jiye Shi, Jiang Li, Kai Liu, Hongjie Zhang, Lihua Wang, Chunhai Fan, Ying Zhu. DNA-Engineered Degradable Invisibility Cloaking for Tumor-Targeting Nanoparticles. Journal of the American Chemical Society 2024, 146
(36)
, 25253-25262. https://doi.org/10.1021/jacs.4c09479
- Jingyi Zhu, Limei Xu, Wenxin Wang, Min Xiao, Jian Li, Lushan Wang, Xukai Jiang. Molecular Dynamics Simulations Reveal Octanoylated Hyaluronic Acid Enhances Liposome Stability, Stealth and Targeting. ACS Omega 2024, 9
(31)
, 33833-33844. https://doi.org/10.1021/acsomega.4c03526
- Francesca Giulimondi, Luca Digiacomo, Serena Renzi, Chiara Cassone, Andrea Pirrottina, Rosa Molfetta, Ilaria Elena Palamà, Gabriele Maiorano, Giuseppe Gigli, Heinz Amenitsch, Daniela Pozzi, Alessandra Zingoni, Giulio Caracciolo. Optimizing Transfection Efficiency in CAR-T Cell Manufacturing through Multiple Administrations of Lipid-Based Nanoparticles. ACS Applied Bio Materials 2024, 7
(6)
, 3746-3757. https://doi.org/10.1021/acsabm.4c00103
- Zhenhua He, Shaohua Qu, Li Shang. Perspectives on Protein–Nanoparticle Interactions at the In Vivo Level. Langmuir 2024, 40
(15)
, 7781-7790. https://doi.org/10.1021/acs.langmuir.4c00181
- Daniela Pozzi, Giulio Caracciolo. Looking Back, Moving Forward: Lipid Nanoparticles as a Promising Frontier in Gene Delivery. ACS Pharmacology & Translational Science 2023, 6
(11)
, 1561-1573. https://doi.org/10.1021/acsptsci.3c00185
- Cintia Marques, Mohammad Javad Hajipour, Célia Marets, Alexandra Oudot, Reihaneh Safavi-sohi, Mélanie Guillemin, Gerrit Borchard, Olivier Jordan, Lucien Saviot, Lionel Maurizi. Identification of the Proteins Determining the Blood Circulation Time of Nanoparticles. ACS Nano 2023, 17
(13)
, 12458-12470. https://doi.org/10.1021/acsnano.3c02041
- Jiayu Ren, Nicholas Andrikopoulos, Kelly Velonia, Huayuan Tang, Rong Cai, Feng Ding, Pu Chun Ke, Chunying Chen. Chemical and Biophysical Signatures of the Protein Corona in Nanomedicine. Journal of the American Chemical Society 2022, 144
(21)
, 9184-9205. https://doi.org/10.1021/jacs.2c02277
- Serena Renzi, Luca Digiacomo, Daniela Pozzi, Erica Quagliarini, Elisabetta Vulpis, Maria Valeria Giuli, Angelica Mancusi, Bianca Natiello, Maria Gemma Pignataro, Gianluca Canettieri, Laura Di Magno, Luca Pesce, Valentina De Lorenzi, Samuele Ghignoli, Luisa Loconte, Carmela Maria Montone, Anna Laura Capriotti, Aldo Laganà, Carmine Nicoletti, Heinz Amenitsch, Marco Rossi, Francesco Mura, Giacomo Parisi, Francesco Cardarelli, Alessandra Zingoni, Saula Checquolo, Giulio Caracciolo. Structuring lipid nanoparticles, DNA, and protein corona into stealth bionanoarchitectures for in vivo gene delivery. Nature Communications 2024, 15
(1)
https://doi.org/10.1038/s41467-024-53569-8
- Chantalle Moulton, Anna Baroni, Erica Quagliarini, Lucia Leone, Luca Digiacomo, Marta Morotti, Giulio Caracciolo, Maria Vittoria Podda, Ennio Tasciotti. Navigating the nano-bio immune interface: advancements and challenges in CNS nanotherapeutics. Frontiers in Immunology 2024, 15 https://doi.org/10.3389/fimmu.2024.1447567
- Siyao Xiao, Junbiao Wang, Luca Digiacomo, Augusto Amici, Valentina De Lorenzi, Licia Anna Pugliese, Francesco Cardarelli, Andrea Cerrato, Aldo Laganà, Lishan Cui, Massimiliano Papi, Giulio Caracciolo, Cristina Marchini, Daniela Pozzi. Protein corona alleviates adverse biological effects of nanoplastics in breast cancer cells. Nanoscale 2024, 16
(35)
, 16671-16683. https://doi.org/10.1039/D4NR01850H
- Tao Li, Yupeng Wang, Dongfang Zhou. Manipulation of protein corona for nanomedicines. WIREs Nanomedicine and Nanobiotechnology 2024, 16
(4)
https://doi.org/10.1002/wnan.1982
- Giulio Caracciolo. Artificial protein coronas: directing nanoparticles to targets. Trends in Pharmacological Sciences 2024, 45
(7)
, 602-613. https://doi.org/10.1016/j.tips.2024.05.003
- Xinlin Ma, Tianyi Zhao, Xiaoyue Ren, Hui Lin, Pan He. Recent Progress in Polyion Complex Nanoparticles with Enhanced Stability for Drug Delivery. Polymers 2024, 16
(13)
, 1871. https://doi.org/10.3390/polym16131871
- Jianjie Xie, Zongyu Huang, Jinpeng Gao, Huicong Feng, Chang Liu. Liposome-loaded Prussian blue nanoparticles accelerate wound healing by promoting anti-inflammatory effects. New Journal of Chemistry 2024, 48
(21)
, 9542-9548. https://doi.org/10.1039/D4NJ00403E
- Mingdi Hu, Xiaoyan Li, Zhen You, Rong Cai, Chunying Chen. Physiological Barriers and Strategies of Lipid‐Based Nanoparticles for Nucleic Acid Drug Delivery. Advanced Materials 2024, 36
(22)
https://doi.org/10.1002/adma.202303266
- Cintia Marques, Gerrit Borchard, Olivier Jordan. Unveiling the challenges of engineered protein corona from the proteins’ perspective. International Journal of Pharmaceutics 2024, 654 , 123987. https://doi.org/10.1016/j.ijpharm.2024.123987
- Seigo Kimura, Hideyoshi Harashima. Nano–Bio Interactions: Exploring the Biological Behavior and the Fate of Lipid-Based Gene Delivery Systems. BioDrugs 2024, 38
(2)
, 259-273. https://doi.org/10.1007/s40259-024-00647-4
- Raffaele Carrano, Martina Grande, Eleonora Leti Maggio, Carlotta Zucca, Riccardo Bei, Camilla Palumbo, Chiara Focaccetti, Daniela Nardozi, Valeria Lucarini, Valentina Angiolini, Patrizia Mancini, Francesca Barberini, Giovanni Barillari, Loredana Cifaldi, Laura Masuelli, Monica Benvenuto, Roberto Bei. Dietary Polyphenols Effects on Focal Adhesion Plaques and Metalloproteinases in Cancer Invasiveness. Biomedicines 2024, 12
(3)
, 482. https://doi.org/10.3390/biomedicines12030482
- Xiaoding Shen, Dayi Pan, Qiyong Gong, Zhongwei Gu, Kui Luo. Enhancing drug penetration in solid tumors via nanomedicine: Evaluation models, strategies and perspectives. Bioactive Materials 2024, 32 , 445-472. https://doi.org/10.1016/j.bioactmat.2023.10.017
- Trieu TM Tran, Steve R Roffler. Interactions between nanoparticle corona proteins and the immune system. Current Opinion in Biotechnology 2023, 84 , 103010. https://doi.org/10.1016/j.copbio.2023.103010
- Wenting Zhang, Mingdi Hu, Rong Cai, Chunying Chen. Chemical and biophysical characteristics of protein corona in nanomedicine and its regulatory strategies. Chinese Science Bulletin 2023, 68
(32)
, 4328-4345. https://doi.org/10.1360/TB-2023-0580
- Rowshan Ara Islam, Nabilah Ibnat, Maeirah Afzal Ashaie, Syafiq Asnawi Zainal Abidin, Ezharul Hoque Chowdhury. Carbonate apatite: effect of serum and impact on the cellular proteome. Journal of Nanoparticle Research 2023, 25
(10)
https://doi.org/10.1007/s11051-023-05838-8
- Liangjia Qiu, Ying Zhang, Genxia Wei, Chen Wang, Yinhua Zhu, Tong Yang, Zheng Chu, Peng Gao, Guangqing Cheng, Ang Ma, Yin Kwan Wong, Junzhe Zhang, Chengchao Xu, Jigang Wang, Huan Tang. How eluents define proteomic fingerprinting of protein corona on nanoparticles. Journal of Colloid and Interface Science 2023, 648 , 497-510. https://doi.org/10.1016/j.jcis.2023.05.045
- Mareike F. S. Deuker, Volker Mailänder, Svenja Morsbach, Katharina Landfester. Anti-PEG antibodies enriched in the protein corona of PEGylated nanocarriers impact the cell uptake. Nanoscale Horizons 2023, 8
(10)
, 1377-1385. https://doi.org/10.1039/D3NH00198A
- Luca Digiacomo, Serena Renzi, Erica Quagliarini, Daniela Pozzi, Heinz Amenitsch, Gianmarco Ferri, Luca Pesce, Valentina De Lorenzi, Giulia Matteoli, Francesco Cardarelli, Giulio Caracciolo. Investigating the mechanism of action of DNA-loaded PEGylated lipid nanoparticles. Nanomedicine: Nanotechnology, Biology and Medicine 2023, 53 , 102697. https://doi.org/10.1016/j.nano.2023.102697
- Ina Mishra, Meredith Garrett, Stephen Curry, Jeffrey Jameson, Michail Kastellorizios. Effect of Composition and Size on Surface Properties of Anti-Cancer Nanoparticles. International Journal of Molecular Sciences 2023, 24
(17)
, 13417. https://doi.org/10.3390/ijms241713417
- Junjie Lu, Xiao Gao, Siyao Wang, Yuan He, Xiaowei Ma, Tingbin Zhang, Xiaoli Liu. Advanced strategies to evade the mononuclear phagocyte system clearance of nanomaterials. Exploration 2023, 3
(1)
https://doi.org/10.1002/EXP.20220045
- Francesca Giulimondi, Erica Quagliarini, Luca Digiacomo, Serena Renzi, Valentina Palmieri, Massimiliano Papi, Daniela Pozzi, Giulio Caracciolo. Reproducibility of Biomolecular Corona Experiments: A Primer for Reliable Results. Particle & Particle Systems Characterization 2023, 40
(2)
https://doi.org/10.1002/ppsc.202200169
- Shile Wang, Yanyu Chen, Jiancheng Guo, Qinqin Huang. Liposomes for Tumor Targeted Therapy: A Review. International Journal of Molecular Sciences 2023, 24
(3)
, 2643. https://doi.org/10.3390/ijms24032643
- Woojun Kim, Nhu Ky Ly, Yanying He, Yongzhe Li, Zhongyue Yuan, Yoon Yeo. Protein corona: Friend or foe? Co-opting serum proteins for nanoparticle delivery. Advanced Drug Delivery Reviews 2023, 192 , 114635. https://doi.org/10.1016/j.addr.2022.114635
- Francesco Millozzi, Andrea Papait, Marina Bouché, Ornella Parolini, Daniela Palacios. Nano-Immunomodulation: A New Strategy for Skeletal Muscle Diseases and Aging?. International Journal of Molecular Sciences 2023, 24
(2)
, 1175. https://doi.org/10.3390/ijms24021175
- Vitaly P. Pozharov, Tamara Minko. Nanotechnology-Based RNA Vaccines: Fundamentals, Advantages and Challenges. Pharmaceutics 2023, 15
(1)
, 194. https://doi.org/10.3390/pharmaceutics15010194
- Yalin Cong, Didar Baimanov, Yunlong Zhou, Chunying Chen, Liming Wang. Penetration and translocation of functional inorganic nanomaterials into biological barriers. Advanced Drug Delivery Reviews 2022, 191 , 114615. https://doi.org/10.1016/j.addr.2022.114615
- Erica Quagliarini, Luca Digiacomo, Serena Renzi, Daniela Pozzi, Giulio Caracciolo. A decade of the liposome-protein corona: Lessons learned and future breakthroughs in theranostics. Nano Today 2022, 47 , 101657. https://doi.org/10.1016/j.nantod.2022.101657
- Lishan Cui, Erica Quagliarini, Siyao Xiao, Francesca Giulimondi, Serena Renzi, Luca Digiacomo, Giulio Caracciolo, Junbiao Wang, Augusto Amici, Cristina Marchini, Daniela Pozzi. The protein corona reduces the anticancer effect of graphene oxide in HER-2-positive cancer cells. Nanoscale Advances 2022, 4
(18)
, 4009-4015. https://doi.org/10.1039/D2NA00308B
- Valentina Palmieri, Giulio Caracciolo. Tuning the immune system by nanoparticle–biomolecular corona. Nanoscale Advances 2022, 4
(16)
, 3300-3308. https://doi.org/10.1039/D2NA00290F
- Chan Wang, Mengdie Xue, Xiao Liu, Jingjing Chen, Mengdie Jiang, Liuting Zheng, Ruxuan Ma, Chengjin Ding, Yaping Tao, Hao Zhang, Qin Liu, Da Huo. Versatile Protein Coronation Approach with Multiple Depleted Serum for Creating Biocompatible, Precision Nanomedicine. Small 2022, 18
(30)
https://doi.org/10.1002/smll.202202002
- Roberto Bei. Meet the Co-Editor. Anti-Cancer Agents in Medicinal Chemistry 2022, 22
(12)
, 2203-2203. https://doi.org/10.2174/187152062212220415110750
- Raquel Taléns-Visconti, Octavio Díez-Sales, Jesus Vicente de Julián-Ortiz, Amparo Nácher. Nanoliposomes in Cancer Therapy: Marketed Products and Current Clinical Trials. International Journal of Molecular Sciences 2022, 23
(8)
, 4249. https://doi.org/10.3390/ijms23084249
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
References
This article references 56 other publications.
- 1Zhou, Z.; Liu, X.; Zhu, D.; Wang, Y.; Zhang, Z.; Zhou, X.; Qiu, N.; Chen, X.; Shen, Y. Nonviral cancer gene therapy: Delivery Cascade and Vector Nanoproperty Integration. Adv. Drug Deliver Rev. 2017, 115, 115– 154, DOI: 10.1016/j.addr.2017.07.021There is no corresponding record for this reference.
- 2Caracciolo, G.; Palchetti, S.; Digiacomo, L.; Chiozzi, R. Z.; Capriotti, A. L.; Amenitsch, H.; Tentori, P. M.; Palmieri, V.; Papi, M.; Cardarelli, F.; Pozzi, D.; Lagana, A. Human Biomolecular Corona of Liposomal Doxorubicin: the Overlooked Factor in Anticancer Drug Delivery. ACS Appl. Mater. Interfaces 2018, 10, 22951– 22962, DOI: 10.1021/acsami.8b049622https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFeqs73J&md5=0e7ab53be0bdad05fdeb35ecd85808c4Human Biomolecular Corona of Liposomal Doxorubicin: The Overlooked Factor in Anticancer Drug DeliveryCaracciolo, Giulio; Palchetti, Sara; Digiacomo, Luca; Chiozzi, Riccardo Zene zini; Capriotti, Anna Laura; Amenitsch, Heinz; Tentori, Paolo Maria; Palmieri, Valentina; Papi, Massimiliano; Cardarelli, Francesco; Pozzi, Daniela; Lagana, AldoACS Applied Materials & Interfaces (2018), 10 (27), 22951-22962CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)More than 20 years after its approval by the Food and Drug Administration (FDA), liposomal doxorubicin (DOX) is still the drug of choice for the treatment of breast cancer and other conditions such as ovarian cancer and multiple myeloma. Yet, despite the efforts, liposomal DOX did not satisfy expectations at the clin. level. When liposomal drugs enter a physiol. environment, their surface gets coated by a dynamic biomol. corona (BC). The BC changes liposome's synthetic identity, providing it with a new one, referred to as "biol. identity" (size, aggregation state, and BC compn.). Today, the concept is emerging that specific BCs may det. either success (e.g., stealth effect and accumulation at the target site) or failure (e.g., rapid blood clearance and off-target interactions) of liposomal drugs. To get a comprehensive investigation of liposome synthetic identity, biol. identity, and cellular response as a function of human plasma (HP) concn., here we used a straightforward combination of quant. anal. and imaging tools, including dynamic light scattering, microelectrophoresis, synchrotron small-angle X-ray scattering, transmission electron microscopy (TEM), fluorescence lifetime imaging microscopy (FLIM), nano-liq. chromatog. tandem mass spectrometry/mass spectrometry (nano-LC-MS/MS), confocal microscopy, flow cytometry, and cell viability assays. Doxoves was selected as a ref. Following exposure to HP, Doxoves was surrounded by a complex BC that changed liposome's synthetic identity. Observations made with nano-LC-MS/MS revealed that the BC of Doxoves did not evolve as a function of HP concn. and was poorly enriched of typical "opsonins" (complement proteins, Igs, etc.). This provides a possible explanation for the prolonged blood circulation of liposomal DOX. On the other hand, flow cytometry showed that protein binding reduced the internalization of DOX in MCF7 and MDA-MB-435S human breast carcinoma. Combining FLIM and TEM expts., we clarified that redn. in DOX intracellular content was likely due to the frequent rupture of the liposome membrane and consequent leakage of the cargo. In light of reported results, we are prompted to speculate that a detailed understanding of BC formation, compn., and effects on liposome stability and uptake is an indispensable task of future research in the field, esp. along the way to clin. translation of liposomal drugs.
- 3Torchilin, V. P. Recent Advances With Liposomes As Pharmaceutical Carriers. Nat. Rev. Drug Discovery 2005, 4, 145– 160, DOI: 10.1038/nrd16323https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpt1WlsA%253D%253D&md5=f25804d328c08cbfadeafb90ca7e3554Recent advances with liposomes as pharmaceutical carriersTorchilin, Vladimir P.Nature Reviews Drug Discovery (2005), 4 (2), 145-160CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Liposomes - microscopic phospholipid bubbles with a bilayered membrane structure - have received a lot of attention during the past 30 years as pharmaceutical carriers of great potential. More recently, many new developments have been seen in the area of liposomal drugs - from clin. approved products to new exptl. applications, with gene delivery and cancer therapy still being the principal areas of interest. For further successful development of this field, promising trends must be identified and exploited, albeit with a clear understanding of the limitations of these approaches.
- 4Rejman, J.; Oberle, V.; Zuhorn, I. S.; Hoekstra, D. Size-Dependent Internalization Of Particles Via the Pathways of Clathrin-and Caveolae-Mediated Endocytosis. Biochemical journal 2004, 377, 159– 169, DOI: 10.1042/bj200312534https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXis1Wnsg%253D%253D&md5=e517e4c6cbf87ecba1a1c9537a0c67efSize-dependent internalization of particles via the pathways of clathrin- and caveolae-mediated endocytosisRejman, Joanna; Oberle, Volker; Zuhorn, Inge S.; Hoekstra, DickBiochemical Journal (2004), 377 (1), 159-169CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)Non-phagocytic eukaryotic cells can internalize particles <1 μm in size, encompassing pathogens, liposomes for drug delivery or lipoplexes applied in gene delivery. In the present study, we have investigated the effect of particle size on the pathway of entry and subsequent intracellular fate in non-phagocytic B16 cells, using a range of fluorescent latex beads of defined sizes (50-1000 nm). Our data reveal that particles as large as 500 nm were internalized by cells via an energy-dependent process. With an increase in size (50-500 nm), cholesterol depletion increased the efficiency of inhibition of uptake. The processing of the smaller particles was significantly perturbed upon microtubule disruption, while displaying a negligible effect on that of the 500 nm beads. Inhibitor and co-localization studies revealed that the mechanism by which the beads were internalized, and their subsequent intracellular routing, was strongly dependent on particle size. Internalization of microspheres with a diam. <200 nm involved clathrin-coated pits. With increasing size, a shift to a mechanism that relied on caveolae-mediated internalization became apparent, which became the predominant pathway of entry for particles of 500 nm in size. At these conditions, delivery to the lysosomes was no longer apparent. The data indicate that the size itself of (ligand-devoid) particles can det. the pathway of entry. The clathrin-mediated pathway of endocytosis shows an upper size limit for internalization of approx. 200 Nm, and kinetic parameters may det. the almost exclusive internalization of such particles along this pathway rather than via caveolae.
- 5Betker, J. L.; Jones, D.; Childs, C. R.; Helm, K. M.; Terrell, K.; Nagel, M. A.; Anchordoquy, T. J. Nanoparticle Uptake by Circulating Leukocytes: a Major Barrier to Tumor Delivery. J. Controlled Release 2018, 286, 85– 93, DOI: 10.1016/j.jconrel.2018.07.0315https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlOit7rL&md5=83fef6f20fad50e4d3f20ef7f12bd841Nanoparticle uptake by circulating leukocytes: A major barrier to tumor deliveryBetker, Jamie L.; Jones, Dallas; Childs, Christine R.; Helm, Karen M.; Terrell, Kristina; Nagel, Maria A.; Anchordoquy, Thomas J.Journal of Controlled Release (2018), 286 (), 85-93CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Decades of research into improving drug delivery to tumors has documented uptake of particulate delivery systems by resident macrophages in the lung, liver, and spleen, and correlated short circulation times with reduced tumor accumulation. An implicit assumption in these studies is that nanoparticles present in the blood are available for distribution to the tumor. This study documents significant levels of lipoplex uptake by circulating leukocytes, and its effect on distribution to the tumor and other organs. In agreement with previous studies, PEGylation dramatically extends circulation times and enhances tumor delivery. However, our studies suggest that this relationship is not straightforward, and that particle sequestration by leukocytes can significantly alter biodistribution, esp. with non-PEGylated nanoparticle formulations. We conclude that leukocyte uptake should be considered in biodistribution studies, and that delivery to these circulating cells may present opportunities for treating viral infections and leukemia.
- 6Mislick, K. A.; Baldeschwieler, J. D. Evidence for the Role of Proteoglycans in Cation-Mediated Gene Transfer. Proc. Natl. Acad. Sci. U. S. A. 1996, 93, 12349– 12354, DOI: 10.1073/pnas.93.22.123496https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xms1GjtLc%253D&md5=82590756f2ed276c18b3d9cfdc89d778Evidence for the role of proteoglycans in cation-mediated gene transferMislick, Kimberly A.; Baldeschwieler, John D.Proceedings of the National Academy of Sciences of the United States of America (1996), 93 (22), 12349-12354CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We report evidence that gene complexes, consisting of polycations and plasmid DNA enter cells via binding to membrane-assocd. proteoglycans. Treatment of HeLa cells with sodium chlorate, a potent inhibitor of proteoglycan sulfation, reduced luciferase expression by 69%. Cellular treatment with heparinase and chondroitinase ABC inhibited expression by 78% and 20% with respect to control cells. Transfection was dramatically inhibited by heparin and heparan sulfate and to a smaller extent by chondroitin sulfate B. Transfection of mutant, proteoglycan deficient Chinese hamster ovary cells was 53× lower than of wild-type cells. For each of these assays, the intracellular uptake of DNA at 37°C and the binding of DNA to the cell membrane at 4°C was impaired. Preliminary transfection expts. conducted in mutant and wild-type Chinese hamster ovary cells suggest that transfection by some cationic lipids is also proteoglycan dependent. The variable distribution of proteoglycans among tissues may explain why some cell types are more susceptible to transfection than others.
- 7Pozzi, D.; Colapicchioni, V.; Caracciolo, G.; Piovesana, S.; Capriotti, A. L.; Palchetti, S.; De Grossi, S.; Riccioli, A.; Amenitsch, H.; Laganà, A. Effect of Polyethyleneglycol (PEG) Chain Length on the Bio–Nano-Interactions between PEGylated Lipid Nanoparticles and Biological Fluids: from Nanostructure to Uptake in Cancer Cells. Nanoscale 2014, 6, 2782– 2792, DOI: 10.1039/c3nr05559k7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXisVahtrw%253D&md5=e52e369b784c0d20d6f7f308197d5888Effect of polyethyleneglycol (PEG) chain length on the bio-nano-interactions between PEGylated lipid nanoparticles and biological fluids: from nanostructure to uptake in cancer cellsPozzi, Daniela; Colapicchioni, Valentina; Caracciolo, Giulio; Piovesana, Susy; Capriotti, Anna Laura; Palchetti, Sara; De Grossi, Stefania; Riccioli, Anna; Amenitsch, Heinz; Lagana, AldoNanoscale (2014), 6 (5), 2782-2792CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)When nanoparticles (NPs) enter a physiol. environment, medium components compete for binding to the NP surface leading to formation of a rich protein shell known as the "protein corona". Unfortunately, opsonins are also adsorbed. These proteins are immediately recognized by the phagocyte system with rapid clearance of the NPs from the bloodstream. Polyethyleneglycol (PEG) coating of NPs (PEGylation) is the most efficient anti-opsonization strategy. Linear chains of PEG, grafted onto the NP surface, are able to create steric hindrance, resulting in a significant inhibition of protein adsorption and less recognition by macrophages. However, excessive PEGylation can lead to a strong inhibition of cellular uptake and less efficient binding with protein targets, reducing the potential of the delivery system. To reach a compromise in this regard we employed a multi-component (MC) lipid system with uncommon properties of cell uptake and endosomal escape and increasing length of PEG chains. Nano liq. chromatog. coupled with tandem mass spectrometry (nanoLC-MS/MS) anal. allowed us to accurately det. the corona compn. showing that apolipoproteins are the most abundant class in the corona and that increasing the PEG length reduced the protein adsorption and the liposomal surface affinity for apolipoproteins. Due to the abundance of apolipoproteins, we exploited the "protein corona effect" to deliver cationic liposome-human plasma complexes to human prostate cancer PC3 cells that express a high level of scavenger receptor class B type 1 to evaluate the cellular uptake efficiency of the systems used. Combining laser scanning confocal microscopy with flow cytometry anal. in PC3 cells we demonstrated that MC-PEG2k is the best compromise between an anti-opsonization strategy and active targeting and could be a promising candidate to treat prostate cancer in vivo.
- 8Srinivasan, C.; Burgess, D. J. Optimization and Characterization of Anionic Lipoplexes for Gene Delivery. Journal of controlled release 2009, 136, 62– 70, DOI: 10.1016/j.jconrel.2009.01.0228https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXlvFGjtLg%253D&md5=f59f8f868862bf9ebaa2c3eff935fddfOptimization and characterization of anionic lipoplexes for gene deliverySrinivasan, Charudharshini; Burgess, Diane J.Journal of Controlled Release (2009), 136 (1), 62-70CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Anionic lipoplexes, comprising divalent cations, DNA and anionic liposomes, were optimized for high transfection efficiency and low cytotoxicity. Different molar ratios of anionic to zwitterionic lipid, 1:9 to 1:1 (1,2-dioleoyl-sn-glycero-3-[phospho-rac-(1-glycerol)] (DOPG): 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), various cations (Ca2+, Mg2+ and Na+) and different anionic lipid/DNA ratios were investigated. The optimized formulation was composed of: anionic/zwitterionic lipid molar ratio 1:4 (DOPG:DOPE); 15-20 mM Ca2+; and 15-20 μg lipid for complexation with 0.8 μg plasmid DNA. Biophys. studies (particle size anal., gel electrophoresis, transmission electron microscopy (TEM), and confocal microscopy) were conducted to characterize the different formulations. TEM revealed structural differences between the complexed and uncomplexed lipoplexes. Gel electrophoresis confirmed the formation of anionic lipoplexes with the amt. of free DNA minimized for the optimized formulation. Confocal imaging showed cellular uptake of the anionic lipoplexes. Most significantly the anionic lipoplex formulation, optimized for the highest transfection efficiency (approx. 78% in the presence of serum) exhibited the highest cell viability approx. 93%, (3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyl tetrazolium bromide) MTT assay. This was compared to Lipofectamine2000 which had a transfection efficiency and cell viability of approx. 68% and 35%, resp. The anionic lipoplex formulation developed here shows promise as a non-viral vector with high transfection efficiency and low cytotoxicity.
- 9McManus, J. J.; Rädler, J. O.; Dawson, K. A. Does Calcium Turn a Zwitterionic Lipid Cationic?. J. Phys. Chem. B 2003, 107, 9869– 9875, DOI: 10.1021/jp034463d9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXmsVegtLo%253D&md5=9238f106a72aadd839604b2f0d8e28ddDoes Calcium Turn a Zwitterionic Lipid Cationic?McManus, Jennifer J.; Raedler, Joachim O.; Dawson, Kenneth A.Journal of Physical Chemistry B (2003), 107 (36), 9869-9875CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)The structure of a complex formed from DNA, calcium, and DPPC has been studied by small-angle X-ray scattering (SAXS). Previous DSC and temp. scanning ultrasound have shown that in excess lipid two DPPC phases exist, one corresponding to DNA-bound lipid and the other corresponding to lipid not directly involved in complex formation. We have investigated the effect of calcium on both phases within the complex structure. Contrary to the observations in the ternary DPPC/calcium/water system, the binding of calcium to the DPPC membrane in this quaternary system contg. DNA appears to be significantly enhanced. We have estd. the stoichiometry of calcium binding within the complex, and we concluded that the zwitterionic lipid is directly involved in binding to DNA.
- 10Palchetti, S.; Pozzi, D.; Marchini, C.; Amici, A.; Andreani, C.; Bartolacci, C.; Digiacomo, L.; Gambini, V.; Cardarelli, F.; Di Rienzo, C.; Peruzzi, G.; Amenitsch, H.; Palermo, R.; Screpanti, I.; Caracciolo, G. Manipulation of Lipoplex Concentration at the Cell Surface Boosts Transfection Efficiency in Hard-to-Transfect Cells. Nanomedicine: Nanotechnology, Biology and Medicine 2017, 13, 681– 691, DOI: 10.1016/j.nano.2016.08.01910https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2szks1elug%253D%253D&md5=e9844674edc9429b5ba523ae3b3e86cdManipulation of lipoplex concentration at the cell surface boosts transfection efficiency in hard-to-transfect cellsPalchetti Sara; Pozzi Daniela; Marchini Cristina; Amici Augusto; Andreani Cristina; Bartolacci Caterina; Gambini Valentina; Digiacomo Luca; Cardarelli Francesco; Di Rienzo Carmine; Peruzzi Giovanna; Palermo Rocco; Amenitsch Heinz; Screpanti Isabella; Caracciolo GiulioNanomedicine : nanotechnology, biology, and medicine (2017), 13 (2), 681-691 ISSN:.To date, efficiency upon non-viral DNA delivery remains low and this implies the existence of unidentified transfection barriers. Here we explore the mechanisms of action of multicomponent (MC) cationic liposome/DNA complexes (lipoplexes) by a combination of reporter technologies, dynamic light scattering (DLS), synchrotron small angle X-ray scattering (SAXS), fluorescence activated cell sorting (FACS) analysis and laser scanning confocal microscopy (LSCM) in live cells. Lipofectamine - the gold standard among transfection reagents - was used as a reference. On the basis of our results, we suggest that an additional transfection barrier impairs transfection efficiency, that is: low lipoplex concentration at the cell surface. Based on the acquired knowledge we propose an optimized transfection protocol that allowed us to efficiently transfect DND41, JURKAT, MOLT3, P12-ICHIKAWA, ALL-SILL, TALL-1 human T-cell acute lymphoblastic leukemia (T-ALL) cell lines known to be difficult-to-transfect by using non-viral vectors and where LFN-based technologies fail to give satisfactory results.
- 11Pan, J.; Heberle, F. A.; Tristram-Nagle, S.; Szymanski, M.; Koepfinger, M.; Katsaras, J.; Kučerka, N. Molecular Structures of Fluid Phase Phosphatidylglycerol Bilayers As Determined by Small Angle Neutron and X-Ray Scattering. Biochimica et Biophysica Acta (BBA)-Biomembranes 2012, 1818, 2135– 2148, DOI: 10.1016/j.bbamem.2012.05.00711https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XovVaju7g%253D&md5=957c783ae34f395d4cccfc473dfe18e9Molecular structures of fluid phase phosphatidylglycerol bilayers as determined by small angle neutron and X-ray scatteringPan, Jianjun; Heberle, Frederick A.; Tristram-Nagle, Stephanie; Szymanski, Michelle; Koepfinger, Mary; Katsaras, John; Kucerka, NorbertBiochimica et Biophysica Acta, Biomembranes (2012), 1818 (9), 2135-2148CODEN: BBBMBS; ISSN:0005-2736. (Elsevier B.V.)We have detd. the mol. structures of commonly used phosphatidylglycerols (PGs) in the commonly accepted biol. relevant fluid phase. This was done by simultaneously analyzing small angle neutron and X-ray scattering data, with the constraint of measured lipid vols. We report the temp. dependence of bilayer parameters obtained using the one-dimensional scattering d. profile model - which was derived from mol. dynamics simulations - including the area per lipid, the overall bilayer thickness, as well as other intrabilayer parameters (e.g., hydrocarbon thickness). Lipid areas are found to be larger than their phosphatidylcholine (PC) counterparts, a result likely due to repulsive electrostatic interactions taking place between the charged PG headgroups even in the presence of sodium counterions. In general, PG and PC bilayers show a similar response to changes in temp. and chain length, but differ in their response to chain unsatn. For example, compared to PC bilayers, the inclusion of a first double bond in PG lipids results in a smaller incremental change to the area per lipid and bilayer thickness. However, the extrapolated lipid area of satd. PG lipids to infinite chain length is found to be similar to that of PCs, an indication of the glycerol-carbonyl backbone's pivotal role in influencing the lipid-water interface.
- 12Caracciolo, G. Liposome-Protein Corona in a Physiological Environment: Challenges and Opportunities for Targeted Delivery of Nanomedicines. Nanomedicine: Nanotechnology, Biology, and Medicine 2015, 11, 543– 557, DOI: 10.1016/j.nano.2014.11.00312https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2Mvjt1CnsA%253D%253D&md5=4ad22c113ca7fd1830353edcb8c5395eLiposome-protein corona in a physiological environment: challenges and opportunities for targeted delivery of nanomedicinesCaracciolo GiulioNanomedicine : nanotechnology, biology, and medicine (2015), 11 (3), 543-57 ISSN:.Active targeting that exploits the (over)expression of surface receptors in target cells by ligand incorporation is a central concept in nanomedicine research. Despite unprecedented efforts, no targeted liposome-based therapeutics is commercially available for clinical practice. What is inhibiting the efficient translation of targeted liposome technology from bench to bedside? After introduction in the bloodstream, the lipid surface is immediately modified by the adsorption of a "protein corona" and preserving the surface functionality appears to be challenging. On the other hand, a long-standing corona with receptor-binding sites could associate with the target cell long enough to activate the cell's uptake machinery, triggering liposome endocytosis and intracellular cargo delivery. This opens the intriguing possibility to manipulate the corona composition by liposome design. This review will focus on the emerging field of liposome-protein corona research from basic, descriptive research to readily applicable knowledge and technologies for implementation in drug improvement and development. From the clinical editor: This review is addressing the liposome protein corona research concerning the potential gains in drug improvement and for drug development.
- 13Pozzi, D.; Caracciolo, G.; Caminiti, R.; De Sanctis, S. C.; Amenitsch, H.; Marchini, C.; Montani, M.; Amici, A. 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. ACS Appl. Mater. Interfaces 2009, 1, 2237– 2249, DOI: 10.1021/am900406b13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFOiurjM&md5=e309feac0c73ff4b503c7c62e31c249bToward 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 releasePozzi, Daniela; Caracciolo, Giulio; Caminiti, Ruggero; De Sanctis, Sofia Candeloro; Amenitsch, Heinz; Marchini, Cristina; Montani, Maura; Amici, AugustoACS Applied Materials & Interfaces (2009), 1 (10), 2237-2249CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)A viewpoint now emerging is that a crit. factor in lipid-mediated transfection (lipofection) is the structural evolution of lipoplexes upon interaction with anionic cellular lipids, resulting in DNA release. At the early stages of interaction, the authors found a universal behavior of lipoplex/anionic lipid (AL) mixts.: the lipoplex structure is slightly perturbed, while the one-dimensional DNA lattice between cationic membranes is largely dild. by ALs. This finding is in excellent agreement with previous suggestions on the mechanism of DNA unbinding from lipoplexes by ALs. Upon further interaction, the propensity of a given lipoplex structure to be solubilized by anionic cellular lipids strongly depends on the shape coupling between lipoplex and ALs. Furthermore, the authors investigated the effect of the membrane charge d. and a general correlation resulted: the higher the membrane charge d. of anionic membranes, the higher their ability to solubilize the structure of lipoplexes and to promote DNA release. Lastly, the formation of nonlamellar phases in lipoplex/AL mixts. is regulated by the propensity of anionic cellular lipids to adopt nonlamellar phases. Remarkably, also phase transition rates and DNA release were found to be strongly affected by the shape coupling between lipoplex and ALs. It thus seems likely that the structural and phase evolution of lipoplexes may only be meaningful in the context of specific anionic cellular membranes. These results highlight the phase properties of the carrier lipid/cellular lipid mixts. as a decisive factor for optimal DNA release and suggest a potential strategy for the rational design of efficient cationic lipid carriers.
- 14Pozzi, D.; Marchini, C.; Cardarelli, F.; Rossetta, A.; Colapicchioni, V.; Amici, A.; Montani, M.; Motta, S.; Brocca, P.; Cantu, L.; Caracciolo, G. Mechanistic Understanding of Gene Delivery Mediated by Highly Efficient Multicomponent Envelope-Type Nanoparticle Systems. Mol. Pharmaceutics 2013, 10, 4654– 4665, DOI: 10.1021/mp400470p14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslWlsr3I&md5=25b7631ca1963e0eef925f241625110bMechanistic Understanding of Gene Delivery Mediated by Highly Efficient Multicomponent Envelope-Type Nanoparticle SystemsPozzi, D.; Marchini, C.; Cardarelli, F.; Rossetta, A.; Colapicchioni, V.; Amici, A.; Montani, M.; Motta, S.; Brocca, P.; Cantu, L.; Caracciolo, G.Molecular Pharmaceutics (2013), 10 (12), 4654-4665CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)We packaged condensed DNA/protamine particles in multicomponent envelope-type nanoparticle systems (MENS) combining different molar fractions of the cationic lipids 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and 3β-[N-(N,N-dimethylaminoethane)-carbamoyl] cholesterol (DC-Chol) and the zwitterionic lipids dioleoylphosphocholine (DOPC) and dioleoylphosphatidylethanolamine (DOPE). Dynamic light scattering (DLS) and microelectrophoresis allowed us to identify the cationic lipid/DNA charge ratio at which MENS are small sized and pos. charged, while synchrotron small-angle X-ray scattering (SAXS) and at. force microscopy (AFM) revealed that MENS are well-shaped DNA/protamine particles covered by a lipid monobilayer. Transfection efficiency (TE) expts. indicate that a nanoparticle formulation, termed MENS-3, was not cytotoxic and highly efficient to transfect Chinese hamster ovary (CHO) cells. To rationalize TE, we performed a quant. investigation of cell uptake, intracellular trafficking,endosomal escape, and final fate by laser scanning confocal microscopy (LSCM). We found that fluid-phase macropinocytosis is the only endocytosis pathway used by MENS-3. Once taken up by the cell, complexes that are actively transported by microtubules frequently fuse with lysosomes, while purely diffusing systems do not. Indeed, spatiotemporal image correlation spectroscopy (STICS) clarified that MENS-3 mostly exploit diffusion to move in the cytosol of CHO cells, thus explaining the high TE levels obsd. Also, MENS-3 exhibited a marked endosomal rupture ability resulting in extraordinary DNA release. The lipid-dependent and structure-dependent TE boost suggests that ecient transfection requires both the membrane fusogenic activity of the nanocarrier envelope and the employment of lipid species with intrinsic endosomal rupture ability.
- 15Koltover, I.; Salditt, T.; Safinya, C. Phase Diagram, Stability, and Overcharging of Lamellar Cationic Lipid–DNA Self-Assembled Complexes. Biophys. J. 1999, 77, 915– 924, DOI: 10.1016/S0006-3495(99)76942-015https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXltVyju7w%253D&md5=ef8851417cf746d60ee8bef076e9d8fcPhase diagram, stability, and overcharging of lamellar cationic lipid-DNA self-assembled complexesKoltover, I.; Salditt, T.; Safinya, C. R.Biophysical Journal (1999), 77 (2), 915-924CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)Cationic lipid-DNA (CL-DNA) complexes comprise a promising new class of synthetic nonviral gene delivery systems. When pos. charged, they attach to the anionic cell surface and transfer DNA into the cell cytoplasm. A comprehensive x-ray diffraction study is reported of the lamellar CL-DNA self-assemblies as a function of lipid compn. and lipid/DNA ratio, aimed at elucidating the interactions detg. their structure, charge, and thermodn. stability. The driving force for the formation of charge-neutral complexes is the release of DNA and lipid counterions. Neg. charged complexes have a higher DNA packing d. than isoelec. complexes, whereas pos. charged ones have a lower packing d. This indicates that the overcharging of the complex away from its isoelec. point is caused by changes of the bulk structure with absorption of excess DNA or cationic lipid. The degree of overcharging is dependent on the membrane charge d., which is controlled by the ration of neutral to cationic lipid in the bilayers. Importantly, overcharged complexes are obsd. to move toward their isoelec. charge-neutral point at higher concn. of salt co-ions, with pos. overcharged complexes expelling cationic lipid and neg. overcharged complexes expelling DNA. These observations should apply universally to the formation and structure of self-assemblies between oppositely charged macromols.
- 16Cukalevski, R.; Ferreira, S. A.; Dunning, C. J.; Berggård, T.; Cedervall, T. IgG and Fibrinogen Driven Nanoparticle Aggregation. Nano Research 2015, 8, 2733– 2743, DOI: 10.1007/s12274-015-0780-416https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVKqurnK&md5=65e384125977dcef8e779f96b42633fcIgG and fibrinogen driven nanoparticle aggregationCukalevski, Risto; Ferreira, Silvia A.; Dunning, Christopher J.; Berggaard, Tord; Cedervall, TommyNano Research (2015), 8 (8), 2733-2743CODEN: NRAEB5; ISSN:1998-0000. (Springer GmbH)A thorough understanding of how proteins induce nanoparticle (NP) aggregation is crucial when designing in vitro and in vivo assays and interpreting exptl. results. This knowledge is also crucial when developing nano-applications and formulation for drug delivery systems. In this study, we found that extn. of IgG (IgG) from cow serum results in lower polystyrene NPs aggregation. Moreover, addn. of isolated IgG or fibrinogen to fetal cow serum enhanced this aggregation, thus demonstrating that these factors are major drivers of NP aggregation in serum. Counter-intuitively, NP aggregation was inversely dependent on protein concn.; i.e., low protein concns. induced large aggregates, whereas high protein concns. induced small aggregates. Protein-induced NP aggregation and aggregate size were monitored by absorbance at 400 nm and dynamic light scattering, resp. Here, we propose a mechanism behind the protein concn. dependent aggregation; this mechanism involves the effects of multiple protein interactions on the NP surface, surface area limitations, aggregation kinetics, and the influence of other serum proteins. [Figure not available: see fulltext.].
- 17Walkey, C. D.; Chan, W. C. Understanding and Controlling the Interaction of Nanomaterials with Proteins in a Physiological Environment. Chem. Soc. Rev. 2012, 41, 2780– 2799, DOI: 10.1039/C1CS15233E17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xjs1Cksb8%253D&md5=cc7bfcf4dd59ced93710ef642e64a044Understanding and controlling the interaction of nanomaterials with proteins in a physiological environmentWalkey, Carl D.; Chan, Warren C. W.Chemical Society Reviews (2012), 41 (7), 2780-2799CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. Nanomaterials hold promise as multifunctional diagnostic and therapeutic agents. However, the effective application of nanomaterials is hampered by limited understanding and control over their interactions with complex biol. systems. When a nanomaterial enters a physiol. environment, it rapidly adsorbs proteins forming what is known as the protein corona'. The protein corona alters the size and interfacial compn. of a nanomaterial, giving it a biol. identity that is distinct from its synthetic identity. The biol. identity dets. the physiol. response including signalling, kinetics, transport, accumulation, and toxicity. The structure and compn. of the protein corona depends on the synthetic identity of the nanomaterial (size, shape, and compn.), the nature of the physiol. environment (blood, interstitial fluid, cell cytoplasm, etc.), and the duration of exposure. In this crit. review, we discuss the formation of the protein corona, its structure and compn., and its influence on the physiol. response. We also present an adsorbome' of 125 plasma proteins that are known to assoc. with nanomaterials. We further describe how the protein corona is related to the synthetic identity of a nanomaterial, and highlight efforts to control protein-nanomaterial interactions. We conclude by discussing gaps in the understanding of protein-nanomaterial interactions along with strategies to fill them (167 refs.).
- 18Caracciolo, G.; Pozzi, D.; Capriotti, A. L.; Cavaliere, C.; Foglia, P.; Amenitsch, H.; Laganà, A. Evolution of the Protein Corona of Lipid Gene Vectors as a Function of Plasma Concentration. Langmuir 2011, 27, 15048– 15053, DOI: 10.1021/la202912f18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtl2kt7rO&md5=2d5172301cfaceda4abedbfd6ee2c526Evolution of the protein corona of lipid gene vectors as a function of plasma concentrationCaracciolo, Giulio; Pozzi, Daniela; Capriotti, Anna Laura; Cavaliere, Chiara; Foglia, Patrizia; Amenitsch, Heinz; Lagana, AldoLangmuir (2011), 27 (24), 15048-15053CODEN: LANGD5; ISSN:0743-7463. (American Chemical Society)The concept that the effective unit of interest in the cell-nanomaterial interaction is the particle and its corona of assocd. proteins is emerging. Here we investigate the compositional evolution of the protein corona of 1,2-dioleoyl-3-trimethylammonium propane (DOTAP) cationic liposomes (CLs) and DOTAP/DNA lipoplexes over a wide range of plasma concns. (2.5-80%). The compn. of the hard corona of lipoplexes is quite stable, but that of CLs does evolve considerably. We show that the protein corona of CLs is made of both low-affinity and competitive-binding proteins whose relative abundance changes with the plasma concn. This result may have deep biol. implications for the application of lipid-based gene vectors both in vitro and in vivo.
- 19Monopoli, M. P.; Walczyk, D.; Campbell, A.; Elia, G.; Lynch, I.; Baldelli Bombelli, F.; Dawson, K. A. Physical–Chemical Aspects of Protein Corona: Relevance to In Vitro and In Vivo Biological Impacts of Nanoparticles. J. Am. Chem. Soc. 2011, 133, 2525– 2534, DOI: 10.1021/ja107583h19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsVOnt7w%253D&md5=a93981208087f4c615ac8c7df33a20ddPhysical-Chemical Aspects of Protein Corona: Relevance to in Vitro and in Vivo Biological Impacts of NanoparticlesMonopoli, Marco P.; Walczyk, Dorota; Campbell, Abigail; Elia, Giuliano; Lynch, Iseult; Baldelli Bombelli, Francesca; Dawson, Kenneth A.Journal of the American Chemical Society (2011), 133 (8), 2525-2534CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)It is now clearly emerging that besides size and shape, the other primary defining element of nanoscale objects in biol. media is their long-lived protein ("hard") corona. This corona may be expressed as a durable, stabilizing coating of the bare surface of nanoparticle (NP) monomers, or it may be reflected in different subpopulations of particle assemblies, each presenting a durable protein coating. Using the approach and concepts of phys. chem., we relate studies on the compn. of the protein corona at different plasma concns. with structural data on the complexes both in situ and free from excess plasma. This enables a high degree of confidence in the meaning of the hard protein corona in a biol. context. Here, we present the protein adsorption for two compositionally different NPs, namely sulfonated polystyrene and silica NPs. NP-protein complexes are characterized by differential centrifugal sedimentation, dynamic light scattering, and zeta-potential both in situ and once isolated from plasma as a function of the protein/NP surface area ratio. We then introduce a semiquant. detn. of their hard corona compn. using one-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis and electrospray liq. chromatog. mass spectrometry, which allows us to follow the total binding isotherms for the particles, identifying simultaneously the nature and amt. of the most relevant proteins as a function of the plasma concn. We find that the hard corona can evolve quite significantly as one passes from protein concns. appropriate to in vitro cell studies to those present in in vivo studies, which has deep implications for in vitro-in vivo extrapolations and will require some consideration in the future.
- 20Caracciolo, G.; Pozzi, D.; Capriotti, A.; Cavaliere, C.; Piovesana, S.; Amenitsch, H.; Laganà, A. Lipid Composition: a “Key Factor” for the Rational Manipulation of the Liposome–Protein Corona by Liposome Design. Rsc Adv. 2015, 5, 5967– 5975, DOI: 10.1039/C4RA13335H20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFaltb%252FN&md5=d1a9c991f2a5a3b33d9c0d42c54a2e97Lipid composition: a "key factor" for the rational manipulation of the liposome-protein corona by liposome designCaracciolo, G.; Pozzi, D.; Capriotti, A. L.; Cavaliere, C.; Piovesana, S.; Amenitsch, H.; Lagana, A.RSC Advances (2015), 5 (8), 5967-5975CODEN: RSCACL; ISSN:2046-2069. (Royal Society of Chemistry)When liposomes are exposed to biol. fluids, a dynamic coating of proteins immediately covers them. Similarly to the aura of plasma surrounding the Sun, plasma proteins are thought of as establishing an aura that surrounds each liposome, hence the phenomenon was dubbed 'protein corona'. This natural functionalization includes proteins engaged from the blood that can interact with receptors (over)expressed on the plasma membrane of target cells, thus targeting the liposomes to their final destination. Exploiting the liposome-protein corona for targeting has the potential to revolutionize the treatment of many disorders and requires a deep understanding of the factors shaping the corona. Following incubation with human plasma (HP), here we manipulated this corona by using six liposomal formulations with systematic changes in lipid compn. The lipids we employed are among the most frequently used lipid species for drug and gene delivery applications in vitro and in vivo. The six liposome-protein coronas were thoroughly characterized by synchrotron small angle X-ray scattering, dynamic light scattering, zeta-potential and nanoliquid-chromatog. tandem mass spectrometry expts. We identified general principles shaping the liposome-protein corona and established clear-cut relationships between lipid species and classes of plasma proteins. This knowledge sets the basis for a rational manipulation of the protein corona for targeted drug delivery by liposome design.
- 21Martínez-Negro, M.; González-Rubio, G.; Aicart, E.; Landfester, K.; Guerrero-Martínez, A.; Junquera, E. Insights into Colloidal Nanoparticle-Protein Corona Interactions for Nanomedicine Applications. Adv. Colloid Interface Sci. 2021, 289, 102366, DOI: 10.1016/j.cis.2021.10236621https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXivValsrs%253D&md5=5bb72bf9c5caa188e23a832283cc2abfInsights into colloidal nanoparticle-protein corona interactions for nanomedicine applicationsMartinez-Negro, Maria; Gonzalez-Rubio, Guillermo; Aicart, Emilio; Landfester, Katharina; Guerrero-Martinez, Andres; Junquera, ElenaAdvances in Colloid and Interface Science (2021), 289 (), 102366CODEN: ACISB9; ISSN:0001-8686. (Elsevier B.V.)A review. Nevertheless, the successful implementation of NPs in medicine demands a proper understanding of their interactions with the different proteins found in biol. fluids. Once introduced into the body, NPs are covered by a protein corona (PC) that dets. the biol. behavior of the NPs. The formation of the PC can eventually favor the rapid clearance of the NPs from the body before fulfilling the desired objective or lead to increased cytotoxicity. The PC nature varies as a function of the different repulsive and attractive forces that govern the NP-protein interaction and their colloidal stability. This review focuses on the phenomenon of PC formation on NPs from a physicochem. perspective, aiming to provide a general overview of this crit. process. Main issues related to NP toxicity and clearance from the body as a result of protein adsorption are covered, including the most promising strategies to control PC formation and, thereby, ensure the successful application of NPs in nanomedicine. Colloidal nanoparticles (NPs) have attracted significant attention due to their unique physicochem. properties suitable for diagnosing and treating different human diseases.
- 22Danhier, F.; Feron, O.; Préat, V. To Exploit the Tumor Microenvironment: Passive and Active Tumor Targeting of Nanocarriers for Anti-Cancer Drug Delivery. Journal of controlled release 2010, 148, 135– 146, DOI: 10.1016/j.jconrel.2010.08.02722https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsVCmsrzF&md5=04d79d93a1828d379bfde830da737608To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug deliveryDanhier, Fabienne; Feron, Olivier; Preat, VeroniqueJournal of Controlled Release (2010), 148 (2), 135-146CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)A review. Because of the particular characteristics of the tumor microenvironment and tumor angiogenesis, it is possible to design drug delivery systems that specifically target anti-cancer drugs to tumors. Most of the conventional chemotherapeutic agents have poor pharmacokinetics profiles and are distributed non-specifically in the body leading to systemic toxicity assocd. with serious side effects. Therefore, the development of drug delivery systems able to target the tumor site is becoming a real challenge that is currently addressed. Nanomedicine can reach tumor passively through the leaky vasculature surrounding the tumors by the enhanced permeability and retention effect whereas ligands grafted at the surface of nanocarriers allow active targeting by binding to the receptors overexpressed by cancer cells or angiogenic endothelial cells. This review is divided into 2 parts: the first one describes the tumor microenvironment and the second one focuses on the exploitation and the understanding of these characteristics to design new drug delivery systems targeting the tumor. Delivery of conventional chemotherapeutic anti-cancer drugs is mainly discussed.
- 23Sawant, R. R.; Torchilin, V. P. Challenges in Development of Targeted Liposomal Therapeutics. AAPS journal 2012, 14, 303– 315, DOI: 10.1208/s12248-012-9330-023https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xlslant74%253D&md5=e017520ac851c05a4ab607645b15a263Challenges in Development of Targeted Liposomal TherapeuticsSawant, Rupa R.; Torchilin, Vladimir P.AAPS Journal (2012), 14 (2), 303-315CODEN: AJAOB6; ISSN:1550-7416. (Springer)A review. Liposomes, phospholipid vesicles with a bilayered membrane structure, have been widely used as pharmaceutical carriers for drugs and genes, in particular for treatment of cancer. To enhance the efficacy of the liposomal drugs, drug-loaded liposomes are targeted to the tumors by means of passive (enhanced permeability and retention mediated) targeting, based on the longevity of liposomes in blood and its accumulation in pathol. sites with compromised vasculature, and active targeting, based on the attachment of specific ligands to the liposomal surface to bind certain antigens on the target cells. Antibody-targeted liposomes loaded with anticancer drugs demonstrate high potential for clin. applications. This review highlights evolution of liposomes for both passive and active targeting and challenges in development of targeted liposomal therapeutics specifically antibody-targeted liposomes.
- 24Tenzer, S.; Docter, D.; Kuharev, J.; Musyanovych, A.; Fetz, V.; Hecht, R.; Schlenk, F.; Fischer, D.; Kiouptsi, K.; Reinhardt, C.; Landfester, K.; Schild, H.; Maskos, M.; Knauer, S. K.; Stauber, R. H. Rapid Formation of Plasma Protein Corona Critically Affects Nanoparticle Pathophysiology. Nat. Nano 2013, 8, 772– 781, DOI: 10.1038/nnano.2013.18124https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVyqurrL&md5=927a38bd98a5850c71525a7707dc5073Rapid formation of plasma protein corona critically affects nanoparticle pathophysiologyTenzer, Stefan; Docter, Dominic; Kuharev, Joerg; Musyanovych, Anna; Fetz, Verena; Hecht, Rouven; Schlenk, Florian; Fischer, Dagmar; Kiouptsi, Klytaimnistra; Reinhardt, Christoph; Landfester, Katharina; Schild, Hansjoerg; Maskos, Michael; Knauer, Shirley K.; Stauber, Roland H.Nature Nanotechnology (2013), 8 (10), 772-781CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)In biol. fluids, proteins bind to the surface of nanoparticles to form a coating known as the protein corona, which can critically affect the interaction of the nanoparticles with living systems. As physiol. systems are highly dynamic, it is important to obtain a time-resolved knowledge of protein-corona formation, development and biol. relevancy. Here we show that label-free snapshot proteomics can be used to obtain quant. time-resolved profiles of human plasma coronas formed on silica and polystyrene nanoparticles of various size and surface functionalization. Complex time- and nanoparticle-specific coronas, which comprise almost 300 different proteins, were found to form rapidly (<0.5 min) and, over time, to change significantly in terms of the amt. of bound protein, but not in compn. Rapid corona formation is found to affect hemolysis, thrombocyte activation, nanoparticle uptake and endothelial cell death at an early exposure time.
- 25Kelly, P. M.; Åberg, C.; Polo, E.; O’Connell, A.; Cookman, J.; Fallon, J.; Krpetić, Ž.; Dawson, K. A. Mapping Protein Binding Sites on the Biomolecular Corona of Nanoparticles. Nature Nanotechnol. 2015, 10, 472– 479, DOI: 10.1038/nnano.2015.4725https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXlsVOnsrw%253D&md5=7accf5d96e7e11cc3b5513bf2d0ed118Mapping protein binding sites on the biomolecular corona of nanoparticlesKelly, Philip M.; Aberg, Christoffer; Polo, Ester; O'Connell, Ann; Cookman, Jennifer; Fallon, Jonathan; Krpetic, Zeljka; Dawson, Kenneth A.Nature Nanotechnology (2015), 10 (5), 472-479CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)Nanoparticles in a biol. milieu are known to form a sufficiently long-lived and well-organized 'corona' of biomols. to confer a biol. identity to the particle. Because this nanoparticle-biomol. complex interacts with cells and biol. barriers, potentially engaging with different biol. pathways, it is important to clarify the presentation of functional biomol. motifs at its interface. Here, we demonstrate that by using antibody-labeled gold nanoparticles, differential centrifugal sedimentation and various imaging techniques it is possible to identify the spatial location of proteins, their functional motifs and their binding sites. We show that for transferrin-coated polystyrene nanoparticles only a minority of adsorbed proteins exhibit functional motifs and the spatial organization appears random, which is consistent, overall, with a stochastic and irreversible adsorption process. Our methods are applicable to a wide array of nanoparticles and can offer a microscopic mol. description of the biol. identity of nanoparticles.
- 26Herda, L. M.; Hristov, D. R.; Lo Giudice, M. C.; Polo, E.; Dawson, K. A. Mapping of Molecular Structure of the Nanoscale Surface in Bionanoparticles. J. Am. Chem. Soc. 2017, 139, 111– 114, DOI: 10.1021/jacs.6b1229726https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFCgs7vL&md5=e168198d8ae5f0ebacabe3ffde2bb533Mapping of Molecular Structure of the Nanoscale Surface in BionanoparticlesHerda, Luciana M.; Hristov, Delyan R.; Lo Giudice, Maria Cristina; Polo, Ester; Dawson, Kenneth A.Journal of the American Chemical Society (2017), 139 (1), 111-114CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Characterizing the orientation of covalently conjugated proteins on nanoparticles, produced for in vitro and in vivo targeting, though an important feature of such a system, has proved challenging. Although extensive physicochem. characterization of targeting nanoparticles can be addressed in detail, relevant biol. characterization of the nanointerface is crucial to select suitable nanomaterials for further in vitro or in vivo expts. The authors adopt a methodol. using antibody fragments (Fab) conjugated to gold nanoparticles (immunogold) to map the available epitopes on a transferrin grafted silica particle (SiO2-PEG8-Tf) as a proxy methodol. to predict nanoparticle biol. function, and therefore cellular receptor engagement. Data from the adopted method suggest that, on av., only ∼3.5% of proteins grafted on the SiO2-PEG8-Tf nanoparticle surface have a favorable orientation for recognition by the cellular receptor.
- 27Lara, S.; Alnasser, F.; Polo, E.; Garry, D.; Lo Giudice, M. C.; Hristov, D. R.; Rocks, L.; Salvati, A.; Yan, Y.; Dawson, K. A. Identification of Receptor Binding to the Biomolecular Corona of Nanoparticles. ACS Nano 2017, 11, 1884– 1893, DOI: 10.1021/acsnano.6b0793327https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVahu7Y%253D&md5=05f69ea88f149d67968bd9a8b7a69943Identification of Receptor Binding to the Biomolecular Corona of NanoparticlesLara, Sandra; Alnasser, Fatima; Polo, Ester; Garry, David; Lo Giudice, Maria Cristina; Hristov, Delyan R.; Rocks, Louise; Salvati, Anna; Yan, Yan; Dawson, Kenneth A.ACS Nano (2017), 11 (2), 1884-1893CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Biomols. adsorbed on nanoparticles are known to confer a biol. identity to nanoparticles, mediating the interactions with cells and biol. barriers. However, how these mols. are presented on the particle surface in biol. milieu remains unclear. The central aim of this study is to identify key protein recognition motifs and link them to specific cell-receptor interactions. Here, the authors employed an immuno-mapping technique to quantify epitope presentations of two major proteins in the serum corona, low-d. lipoprotein and IgG. Combined with a purpose-built receptor expression system, both proteins present functional motifs to allow simultaneous recognition by low-d. lipoprotein receptor and Fc-gamma receptor I of the corona. The authors' results suggest that the "labeling" of nanoparticles by biomol. adsorption processes allows for multiple pathways in biol. processes in which they may be "mistaken" for endogenous objects, such as lipoproteins, and exogenous ones, such as viral infections.
- 28Gianneli, M.; Polo, E.; Lopez, H.; Castagnola, V.; Aastrup, T.; Dawson, K. Label-Free in-Flow Detection of Receptor Recognition Motifs on the Biomolecular Corona of Nanoparticles. Nanoscale 2018, 10, 5474– 5481, DOI: 10.1039/C7NR07887K28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjvFWqur4%253D&md5=114cf2f49f7766a1a712d46db373cf44Label-free in-flow detection of receptor recognition motifs on the biomolecular corona of nanoparticlesGianneli, M.; Polo, E.; Lopez, H.; Castagnola, V.; Aastrup, T.; Dawson, K. A.Nanoscale (2018), 10 (12), 5474-5481CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)Nanomedicine, nanotargeting and nanotherapeutics have in the last few years faced several difficulties in translating the promising results obtained in vitro to an in vivo scenario. The origin of this discrepancy might be found in the lack of a detailed and realistic characterization of the biol. surface of nanoparticles. Despite the capability to engineer nanomaterials with a great variety and a precise control of the surface functionalization, the targeting capability is lost when the nanoparticles are embedded in complex biol. media, due to the formation of a biol. layer (biomol. corona). This biol. layer represents the ultimate nanoparticle surface, likely to interact with the cell machinery. Therefore, in addn. to traditional nanoparticle characterization techniques, a more insightful investigation of the biomol. corona is needed, including the capability to assess the orientation and functionality of specific key mol. features. Here we present a method for the rapid screening of exposed protein recognition motifs on the surface of nanoparticles exploiting quartz crystal microbalance (QCM). We quantify accessible functional epitopes of transferrin-coated nanoparticles and correlate them to differences in nanoparticle size and functionalization. The target recognition occurs label free in flow, thereby pushing our investigation into a more in vivo-like scenario. Our method is applicable to a wide array of nanoparticles and therefore holds the potential to become an advanced technique for the classification of all kinds of nanobioconstructs based on their biol. external functionality.
- 29Moein Moghimi, S.; Patel, H. M. Opsonophagocytosis of Liposomes by Peritoneal Macrophages and Bone Marrow Reticuloendothelial Cells. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research 1992, 1135, 269– 274, DOI: 10.1016/0167-4889(92)90230-9There is no corresponding record for this reference.
- 30Papini, E.; Tavano, R.; Mancin, F. Opsonins and Dysopsonins of Nanoparticles: Facts, Concepts, and Methodological Guidelines. Frontiers in Immunology 2020, 11, 2343, DOI: 10.3389/fimmu.2020.567365There is no corresponding record for this reference.
- 31Languino, L. R.; Duperray, A.; Joganic, K. J.; Fornaro, M.; Thornton, G. B.; Altieri, D. C. Regulation of Leukocyte-Endothelium Interaction and Leukocyte Transendothelial Migration by Intercellular Adhesion Molecule 1-Fibrinogen Recognition. Proc. Natl. Acad. Sci. U. S. A. 1995, 92, 1505– 1509, DOI: 10.1073/pnas.92.5.150531https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXktFWktbs%253D&md5=dbc15273eacdcfa66d31356f6ee8c030Regulation of leukocyte-endothelium interaction and leukocyte transendothelial migration by intercellular adhesion molecule 1-fibrinogen recognitionLanguino, Lucia R.; Duperray, Alain; Joganic, Karen J.; Fornaro, Mara; Thornton, George B.; Altieri, Dario C.Proceedings of the National Academy of Sciences of the United States of America (1995), 92 (5), 1505-9CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Although primarily recognized for its role in hemostasis, fibrinogen is also required for competent inflammatory reactions in vivo. It is now shown that fibrinogen promotes adhesions to and migration across an endothelial monolayer of terminally differentiated myelomonocytic cells. This process does not require chemotactic/haptotactic gradients or cytokine stimulation of the endothelium and is specific for the assocn. of fibrinogen with intercellular adhesion mol. 1 (ICAM-1) on endothelium. Among other adhesive plasma proteins, fibronectin fails to increase the binding of leukocytes to endothelium, or transendothelial migration, whereas vitronectin promotes the binding but not the migration. The fibrinogen-mediated leukocyte adhesion and transendothelial migration could be inhibited by a peptide from the fibrinogen γ-chain sequence N117NQKIVNLKEKVAQLEA133, which blocks the binding of fibrinogen to ICAM-1. This interaction could also be inhibited by new anti-ICAM-1 monoclonal antibodies that did not affect the ICAM-1-CD11a/CD18 recognition, thus suggesting that the fibrinogen binding site on ICAM-1 may be structurally distinct from regions previously implicated in leukocyte-endothelium interaction. Therefore, binding of fibrinogen to vascular cell receptors is sufficient to initiate (i) increased leukocyte adhesion to endothelium and (ii) leukocyte transendothelial migration. These two processes are the earliest events of immune inflammatory responses and may also contribute to atherosclerosis.
- 32Deng, Z. J.; Liang, M.; Monteiro, M.; Toth, I.; Minchin, R. F. Nanoparticle-Induced Unfolding of Fibrinogen Promotes Mac-1 Receptor Activation and Inflammation. Nat. Nanotechnol 2011, 6, 39– 44, DOI: 10.1038/nnano.2010.25032https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhs1Wju73P&md5=de9f322b14e197e47e6b1fe034525498Nanoparticle-induced unfolding of fibrinogen promotes Mac-1 receptor activation and inflammationDeng, Zhou J.; Liang, Mingtao; Monteiro, Michael; Toth, Istvan; Minchin, Rodney F.Nature Nanotechnology (2011), 6 (1), 39-44CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)The chem. compn., size, shape and surface characteristics of nanoparticles affect the way proteins bind to these particles, and this in turn influences the way in which nanoparticles interact with cells and tissues. Nanomaterials bound with proteins can result in physiol. and pathol. changes, including macrophage uptake, blood coagulation, protein aggregation and complement activation, but the mechanisms that lead to these changes remain poorly understood. Here, we show that neg. charged poly(acrylic acid)-conjugated gold nanoparticles bind to and induce unfolding of fibrinogen, which promotes interaction with the integrin receptor, Mac-1. Activation of this receptor increases the NF-κB signalling pathway, resulting in the release of inflammatory cytokines. However, not all nanoparticles that bind to fibrinogen demonstrated this effect. Our results show that the binding of certain nanoparticles to fibrinogen in plasma offers an alternative mechanism to the more commonly described role of oxidative stress in the inflammatory response to nanomaterials.
- 33Deng, Z. J.; Liang, M.; Toth, I.; Monteiro, M. J.; Minchin, R. F. Molecular Interaction of Poly (Acrylic Acid) Gold Nanoparticles with Human Fibrinogen. ACS Nano 2012, 6, 8962– 8969, DOI: 10.1021/nn302995333https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhtl2iu7bI&md5=45a7894ab9195c99c6e9cbb5f7e6f758Molecular interaction of poly(acrylic acid) gold nanoparticles with human fibrinogenDeng, Zhou J.; Liang, Mingtao; Toth, Istvan; Monteiro, Michael J.; Minchin, Rodney F.ACS Nano (2012), 6 (10), 8962-8969CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)The binding of fibrinogen to various nanoparticles can result in protein unfolding and exposure of cryptic epitopes that subsequently interact with cell surface receptors. This response is dependent on the size, charge, and concn. of the nanoparticle. Here, the authors examd. the binding kinetics of human fibrinogen to neg. charged poly(acrylic acid)-coated Au-nanoparticles ranging in size from 7 to 22 nm. These particles have previously been shown to elicit an inflammatory response in human cells. The larger nanoparticles bound fibrinogen with increasing affinity and a slower dissocn. rate. Each fibrinogen mol. could accommodate 2 7-nm nanoparticles, but only one when the diam. increased to 10 nm. Nanoparticles of size >12 nm bound multiple fibrinogen mols. in a pos. cooperative manner. However, in the presence of excess nanoparticle, fibrinogen induced the aggregation of the larger particles that could bind more than one protein mol. This was consistent with interparticle bridging by the fibrinogen. Taken together, these results demonstrate that subtle changes in nanoparticle size can influence protein binding both with the surface of the nanoparticle and within the protein corona.
- 34Tenzer, S.; Docter, D.; Kuharev, J.; Musyanovych, A.; Fetz, V.; Hecht, R.; Schlenk, F.; Fischer, D.; Kiouptsi, K.; Reinhardt, C.; Landfester, K.; Schild, H.; Maskos, M.; Knauer, S. K.; Stauber, R. H. Rapid Formation of Plasma Protein Corona Critically Affects Nanoparticle Pathophysiology. Nature Nanotechnol. 2013, 8, 772– 781, DOI: 10.1038/nnano.2013.18134https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVyqurrL&md5=927a38bd98a5850c71525a7707dc5073Rapid formation of plasma protein corona critically affects nanoparticle pathophysiologyTenzer, Stefan; Docter, Dominic; Kuharev, Joerg; Musyanovych, Anna; Fetz, Verena; Hecht, Rouven; Schlenk, Florian; Fischer, Dagmar; Kiouptsi, Klytaimnistra; Reinhardt, Christoph; Landfester, Katharina; Schild, Hansjoerg; Maskos, Michael; Knauer, Shirley K.; Stauber, Roland H.Nature Nanotechnology (2013), 8 (10), 772-781CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)In biol. fluids, proteins bind to the surface of nanoparticles to form a coating known as the protein corona, which can critically affect the interaction of the nanoparticles with living systems. As physiol. systems are highly dynamic, it is important to obtain a time-resolved knowledge of protein-corona formation, development and biol. relevancy. Here we show that label-free snapshot proteomics can be used to obtain quant. time-resolved profiles of human plasma coronas formed on silica and polystyrene nanoparticles of various size and surface functionalization. Complex time- and nanoparticle-specific coronas, which comprise almost 300 different proteins, were found to form rapidly (<0.5 min) and, over time, to change significantly in terms of the amt. of bound protein, but not in compn. Rapid corona formation is found to affect hemolysis, thrombocyte activation, nanoparticle uptake and endothelial cell death at an early exposure time.
- 35Tsuchiya, S.; Yamabe, M.; Yamaguchi, Y.; Kobayashi, Y.; Konno, T.; Tada, K. Establishment and Characterization of a Human Acute Monocytic Leukemia Cell Line (THP-1). International journal of cancer 1980, 26, 171– 176, DOI: 10.1002/ijc.291026020835https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL3M7ktFSisw%253D%253D&md5=ef2b5e1157ca18009c33fc80f183d5f9Establishment and characterization of a human acute monocytic leukemia cell line (THP-1)Tsuchiya S; Yamabe M; Yamaguchi Y; Kobayashi Y; Konno T; Tada KInternational journal of cancer (1980), 26 (2), 171-6 ISSN:0020-7136.A human leukemic cell line (THP-1) cultured from the blood of a boy with acute monocytic leukemia is described. This cell line had Fc and C3b receptors, but no surface or cytoplasmic immunoglobulins. HLA haplotypes of THP-1 were HLA-A2, -A9, -B5, -DRW1 and -DRW2. The monocytic nature of the cell line was characterized by: (1) the presence of alpha-naphthyl butyrate esterase activities which could be inhibited by NaF; (2) lysozyme production; (3) the phagocytosis of latex particles and sensitized sheep erythrocytes; and (4) the ability to restore T-lymphocyte response to Con A. The cells did not possess Epstein-Barr virus-associated nuclear antigen. These results indicate that THP-1 is a leukemia cell line with distinct monocytic markers. During culture, THP-1 maintained these monocytic characteristics for over 14 months.
- 36Kleiveland, C.; Verhoeckx, K.; Cotter, P.; López-Expósito, I.; Kleiveland, C.; Lea, T.; Mackie, A. Peripheral Blood Mononuclear Cells. In The Impact of Food Bioactives on Health: In Vitro and Ex Vivo Models; Springer: Cham, Switzerland, 2015; pp 161– 167, DOI: 10.1007/978-3-319-16104-4_15 .There is no corresponding record for this reference.
- 37Giulimondi, F.; Digiacomo, L.; Pozzi, D.; Palchetti, S.; Vulpis, E.; Capriotti, A. L.; Chiozzi, R. Z.; Lagana, A.; Amenitsch, H.; Masuelli, L.; Peruzzi, G.; Mahmoudi, M.; Screpanti, I.; Zingoni, A.; Caracciolo, G. Interplay of Protein Corona and Immune Cells Controls Blood Residency of Liposomes. Nat. Commun. 2019, 10, 3686, DOI: 10.1038/s41467-019-11642-737https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1Sit7jJ&md5=dfcfc6cb1ec61aa332021ef39373243fInterplay of protein corona and immune cells controls blood residency of liposomesGiulimondi, Francesca; Digiacomo, Luca; Pozzi, Daniela; Palchetti, Sara; Vulpis, Elisabetta; Capriotti, Anna Laura; Chiozzi, Riccardo Zenezini; Lagana, Aldo; Amenitsch, Heinz; Masuelli, Laura; Mahmoudi, Morteza; Screpanti, Isabella; Zingoni, Alessandra; Caracciolo, GiulioNature Communications (2019), 10 (1), 3686CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)In vivo liposomes, like other types of nanoparticles, acquire a totally new 'biol. identity' due to the formation of a biomol. coating known as the protein corona that depends on and modifies the liposomes' synthetic identity. The liposome-protein corona is a dynamic interface that regulates the interaction of liposomes with the physiol. environment. Here we show that the biol. identity of liposomes is clearly linked to their sequestration from peripheral blood mononuclear cells (PBMCs) of healthy donors that ultimately leads to removal from the bloodstream. Pre-coating liposomes with an artificial corona made of human plasma proteins drastically reduces capture by circulating leukocytes in whole blood and may be an effective strategy to enable prolonged circulation in vivo. We conclude with a crit. assessment of the key concepts of liposome technol. that need to be reviewed for its definitive clin. translation.
- 38Chung, A.; Gao, Q.; Kao, W. J. Macrophage Matrix Metalloproteinase-2/-9 Gene and Protein Expression Following Adhesion to ECM-Derived Multifunctional Matrices Via Integrin Complexation. Biomaterials 2007, 28, 285– 298, DOI: 10.1016/j.biomaterials.2006.08.03838https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD28nht1Gkuw%253D%253D&md5=69e547f4029caf5cc4806ef0e8384036Macrophage matrix metalloproteinase-2/-9 gene and protein expression following adhesion to ECM-derived multifunctional matrices via integrin complexationChung Amy; Gao Qiang; Kao Weiyuan JohnBiomaterials (2007), 28 (2), 285-98 ISSN:0142-9612.Macrophages are commonly observed at the biomaterial-tissue interface and interact with the extracellular matrix (ECM) mainly by integrin receptors to play a critical role in ECM turnover by secreting matrix metalloproteinases (MMPs). To investigate beta1 and beta3 containing integrin-mediated adhesion and subsequent MMP-2/-9 protein and gene expression in human blood-derived monocytes, biofunctional peptides immobilized onto flexible polyethylene glycol (PEG) arms were grafted onto a gelatin-based interpenetrating network (IPN). Adherent monocyte density was dramatically greater in the presence of RGD immobilized onto flexible PEG arms of the gelatin-based IPN. Pretreatment of monocytes with either anti-integrin beta1 or beta3 led to a significant decrease in adherent cell density on RGD-PEG-grafted IPNs. MMP-2 and MMP-9 protein and MMP-9 mRNA expression increased in the presence of IPNs initially, independent of ligand identity. Anti-integrin beta1 or beta3 antibody pretreatment of monocytes led to a general decrease in MMP-2/-9 protein expression. These results demonstrate the importance of beta1 and beta3 containing integrins in mediating monocyte adhesion onto RGD immobilized onto flexible PEG arms of the IPN. The results also reveal that MMP-2/-9 protein and gene expression is influenced by the presence of gelatin and not the ligands immobilized on the PEG arms of the IPN.
- 39Simon, J.; Müller, L. K.; Kokkinopoulou, M.; Lieberwirth, I.; Morsbach, S.; Landfester, K.; Mailänder, V. Exploiting the Biomolecular Corona: Pre-Coating of Nanoparticles Enables Controlled Cellular Interactions. Nanoscale 2018, 10, 10731– 10739, DOI: 10.1039/C8NR03331E39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVSgtbnL&md5=e5342d5993c7d20829636788e9ba7907Exploiting the biomolecular corona: pre-coating of nanoparticles enables controlled cellular interactionsSimon, Johanna; Mueller, Laura K.; Kokkinopoulou, Maria; Lieberwirth, Ingo; Morsbach, Svenja; Landfester, Katharina; Mailaender, VolkerNanoscale (2018), 10 (22), 10731-10739CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)Formation of the biomol. corona ultimately dets. the successful application of nanoparticles in vivo. Adsorption of biomols. such as proteins is an inevitable process that takes place instantaneously upon contact with physiol. fluid (e.g. blood). Therefore, strategies are needed to control this process in order to improve the properties of the nanoparticles and to allow targeted drug delivery. Here, we show that the design of the protein corona by a pre-formed protein corona with tailored properties enables targeted cellular interactions. Nanoparticles were pre-coated with Ig depleted plasma to create and design a protein corona that reduces cellular uptake by immune cells. It was proven that a pre-formed protein corona remains stable even after nanoparticles were re-introduced to plasma. This opens up the great potential to exploit protein corona formation, which will significantly influence the development of novel nanomaterials.
- 40Suk, J. S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L. M. PEGylation As a Strategy for Improving Nanoparticle-Based Drug and Gene Delivery. Adv. Drug Delivery Rev. 2016, 99, 28– 51, DOI: 10.1016/j.addr.2015.09.01240https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhs1CqurrP&md5=fecfbdedd72ee97571c913a9ac3cd93fPEGylation as a strategy for improving nanoparticle-based drug and gene deliverySuk, Jung Soo; Xu, Qingguo; Kim, Namho; Hanes, Justin; Ensign, Laura M.Advanced Drug Delivery Reviews (2016), 99 (Part_A), 28-51CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)Coating the surface of nanoparticles with polyethylene glycol (PEG), or "PEGylation", is a commonly used approach for improving the efficiency of drug and gene delivery to target cells and tissues. Building from the success of PEGylating proteins to improve systemic circulation time and decrease immunogenicity, the impact of PEG coatings on the fate of systemically administered nanoparticle formulations has, and continues to be, widely studied. PEG coatings on nanoparticles shield the surface from aggregation, opsonization, and phagocytosis, prolonging systemic circulation time. Here, we briefly describe the history of the development of PEGylated nanoparticle formulations for systemic administration, including how factors such as PEG mol. wt., PEG surface d., nanoparticle core properties, and repeated administration impact circulation time. A less frequently discussed topic, we then describe how PEG coatings on nanoparticles have also been utilized for overcoming various biol. barriers to efficient drug and gene delivery assocd. with other modes of administration, ranging from gastrointestinal to ocular. Finally, we describe both methods for PEGylating nanoparticles and methods for characterizing PEG surface d., a key factor in the effectiveness of the PEG surface coating for improving drug and gene delivery.
- 41Ishida, T.; Ichihara, M.; Wang, X.; Kiwada, H. Spleen Plays an Important Role in the Induction of Accelerated Blood Clearance of PEGylated Liposomes. J. Controlled Release 2006, 115, 243– 250, DOI: 10.1016/j.jconrel.2006.08.00141https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFeitr3K&md5=d8f3548771bd68147d1e85f7cf36af52Spleen plays an important role in the induction of accelerated blood clearance of PEGylated liposomesIshida, Tatsuhiro; Ichihara, Masako; Wang, XinYu; Kiwada, HiroshiJournal of Controlled Release (2006), 115 (3), 243-250CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)It is well known that steric stabilization of the surface of liposomes by a polyethylene glycol (PEG) conjugated lipid results in reduced recognition of the liposomes by the cells of the mononuclear phagocyte system and consequently extended circulation times of the liposomes (t1/2 ≈ 20 h in rat). Recently, we reported on the "accelerated blood clearance (ABC) phenomenon", causing PEGylated liposomes to be cleared very rapidly from the circulation upon repeated injection. We also reported that abundant binding of IgM, secreted into the blood stream after the first dose and, to PEGylated liposomes, plays an essential role in the induction of the ABC phenomenon. Spleen is well known to play a central role in the immune reaction and to produce IgM following a bacterial infection. The aim of the present study was to det. whether spleen contributes to the induction of the ABC phenomenon and to unravel its role in the phenomenon. In rats that were splenectomized (surgical removal of spleen) prior to the first injection of liposomes (0.001 μmol phospholipids/kg), the ABC phenomenon was totally abolished. In these rats serum IgM concns. as well as the amts. of IgM bound to PEGylated liposomes were substantially reduced. Splenectomy attenuated the ABC phenomenon when performed until 3 days post-first injection. Removal of the spleen 4 days post-first injection left the ABC phenomenon unchanged. This finding indicates that the immune reaction in the spleen against the PEGylated liposomes occurs during at least 2-3 days following the first administration and then IgM reactive to PEGylated liposomes is produced. The present study proves that the spleen plays a crit. role in the induction phase of the ABC phenomenon. For effective clin. application, many liposomal drug formulations will require multiple injections. The ABC phenomenon described in this and several preceding papers therefore has important implications for the development and evaluation of therapeutically useful liposomal formulations requiring multiple-dose administration.
- 42Hadjidemetriou, M.; Al-Ahmady, Z.; Mazza, M.; Collins, R. F.; Dawson, K.; Kostarelos, K. In Vivo Biomolecule Corona Around Blood-Circulating, Clinically Used and Antibody-Targeted Lipid Bilayer Nanoscale Vesicles. ACS Nano 2015, 9, 8142– 8156, DOI: 10.1021/acsnano.5b0330042https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFWgsrjN&md5=c8402b491c8438400f0fea698c6824cbIn Vivo Biomolecule Corona around Blood-Circulating, Clinically Used and Antibody-Targeted Lipid Bilayer Nanoscale VesiclesHadjidemetriou, Marilena; Al-Ahmady, Zahraa; Mazza, Mariarosa; Collins, Richard F.; Dawson, Kenneth; Kostarelos, KostasACS Nano (2015), 9 (8), 8142-8156CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)The adsorption of proteins and their layering onto nanoparticle surfaces has been called the "protein corona". This dynamic process of protein adsorption has been extensively studied following in vitro incubation of many different nanoparticles with plasma proteins. However, the formation of protein corona under dynamic, in vivo conditions remains largely unexplored. Extrapolation of in vitro formed protein coronas to predict the fate and possible toxicol. burden from nanoparticles in vivo is of great interest. However, complete lack of such direct comparisons for clin. used nanoparticles makes the study of in vitro and in vivo formed protein coronas of great importance. Our aim was to study the in vivo protein corona formed onto i.v. injected, clin. used liposomes, based on the compn. of the PEGylated liposomal formulation that constitutes the anticancer agent Doxil. The formation of in vivo protein corona was detd. after the recovery of the liposomes from the blood circulation of CD-1 mice 10 min postinjection. In comparison, in vitro protein corona was formed by the incubation of liposomes in CD-1 mouse plasma. In vivo and in vitro formed protein coronas were compared in terms of morphol., compn. and cellular internalization. The protein coronas on bare (non-PEGylated) and monoclonal antibody (IgG) targeted liposomes of the same lipid compn. were also comparatively investigated. A network of linear fibrillary structures constituted the in vitro formed protein corona, whereas the in vivo corona had a different morphol. but did not appear to coat the liposome surface entirely. Even though the total amt. of protein attached on circulating liposomes correlated with that obsd. from in vitro incubations, the variety of mol. species in the in vivo corona were considerably wider. Both in vitro and in vivo formed protein coronas were found to significantly reduce receptor binding and cellular internalization of antibody-conjugated liposomes; however, the in vivo corona formation did not lead to complete ablation of their targeting capability.
- 43Hadjidemetriou, M.; Al-Ahmady, Z.; Kostarelos, K. Time-Evolution of In Vivo Protein Corona onto Blood-Circulating PEGylated Liposomal Doxorubicin (DOXIL) Nanoparticles. Nanoscale 2016, 8, 6948– 6957, DOI: 10.1039/C5NR09158F43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xjs1Gisb0%253D&md5=ea5ffb6e06758c99fce6a74f22e47f94Time-evolution of in vivo protein corona onto blood-circulating PEGylated liposomal doxorubicin (DOXIL) nanoparticlesHadjidemetriou, Marilena; Al-Ahmady, Zahraa; Kostarelos, KostasNanoscale (2016), 8 (13), 6948-6957CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)Nanoparticles (NPs) are instantly modified once injected in the bloodstream because of their interaction with the blood components. The spontaneous coating of NPs by proteins, once in contact with biol. fluids, has been termed the 'protein corona' and it is considered to be a determinant factor for the pharmacol., toxicol. and therapeutic profile of NPs. Protein exposure time is thought to greatly influence the compn. of protein corona, however the dynamics of protein interactions under realistic, in vivo conditions remain unexplored. The aim of this study was to quant. and qual. investigate the time evolution of in vivo protein corona, formed onto blood circulating, clin. used, PEGylated liposomal doxorubicin. Protein adsorption profiles were detd. 10 min, 1 h and 3 h post-injection of liposomes into CD-1 mice. The results demonstrated that a complex protein corona was formed as early as 10 min post-injection. Even though the total amt. of protein adsorbed did not significantly change over time, the fluctuation of protein abundances obsd. indicated highly dynamic protein binding kinetics.
- 44Hadjidemetriou, M.; McAdam, S.; Garner, G.; Thackeray, C.; Knight, D.; Smith, D.; Al-Ahmady, Z.; Mazza, M.; Rogan, J.; Clamp, A.; Kostarelos, K. The Human In Vivo Biomolecule Corona onto PEGylated liposomes: a Proof-of-Concept Clinical Study. Adv. Mater. 2019, 31, 1803335, DOI: 10.1002/adma.201803335There is no corresponding record for this reference.
- 45Cerullo, V.; Seiler, M. P.; Mane, V.; Brunetti-Pierri, N.; Clarke, C.; Bertin, T. K.; Rodgers, J. R.; Lee, B. Toll-Like Receptor 9 Triggers an Innate Immune Response to Helper-Dependent Adenoviral Vectors. Molecular Therapy 2007, 15, 378– 385, DOI: 10.1038/sj.mt.630003145https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtVejtbfJ&md5=229648334245ab0ae325af09963fd70eToll-like Receptor 9 Triggers an Innate Immune Response to Helper-dependent Adenoviral VectorsCerullo, Vincenzo; Seiler, Michael P.; Mane, Viraj; Brunetti-Pierri, Nicola; Clarke, Christian; Bertin, Terry K.; Rodgers, John R.; Lee, BrendanMolecular Therapy (2007), 15 (2), 378-385CODEN: MTOHCK; ISSN:1525-0016. (Nature Publishing Group)A major obstacle to the clin. application of systemic adenoviral gene replacement therapy is the host innate immune response. Although recent studies have attempted to characterize the cellular basis for this response to systemically administered helper-dependent adenoviral vector (HD-Ad), the underlying mol. components of the innate immune repertoire required to recognize the viral vector have yet to be identified. Here, the authors show that primary macrophages can sense HD-Ad vectors via the Toll-like Receptor 9 (TLR9) and respond by increasing pro-inflammatory cytokine secretion. Moreover, TLR9 sensing is involved in the rapid innate immune response to HD-Ad in vivo. TLR9 deficiency attenuates the innate immune response to HD-Ad, whereas TLR9 blockade reduces the acute inflammatory response after i.v. injection of the vector. Moreover, HD-Ad upregulates TLR9 gene expression independent of TLR9 function, suggesting that addnl. innate signaling pathways work cooperatively with TLR9. The identification of the components of the innate immune response to adenovirus will facilitate the development of combinatorial therapy directed at increasing the maximal tolerated dose of systemically delivered adenoviral vectors.
- 46Moghimi, S. M.; Patel, H. M. Tissue Specific Opsonins for Phagocytic Cells and Their Different Affinity for Cholesterol-Rich Liposomes. FEBS letters 1988, 233, 143– 147, DOI: 10.1016/0014-5793(88)81372-346https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1cXktlKntLs%253D&md5=a8ce77eac7904341196b88e9a102888aTissue specific opsonins for phagocytic cells and their different affinity for cholesterol-rich liposomesMoghimi, S. Moein; Patel, Harish M.FEBS Letters (1988), 233 (1), 143-7CODEN: FEBLAL; ISSN:0014-5793.In vivo expts. show that Kupffer cells avidly take up cholesterol-poor but not cholesterol-rich liposomes, whereas splenic phagocytic cells take up preferentially cholesterol-rich rather than cholesterol-poor liposomes in the presence of serum. Apparently, serum contains opsonins specific for hepatic and splenic phagocytic cells and these opsonins have different affinities for cholesterol-rich and cholesterol-poor liposomes.
- 47Moghimi, S. M.; Simberg, D.; Skotland, T.; Yaghmur, A.; Hunter, C. The Interplay between Blood Proteins, Complement, and Macrophages on Nanomedicine Performance and Responses. Journal of Pharmacology and Experimental Therapeutics 2019, 370, 581– 592, DOI: 10.1124/jpet.119.25801247https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisVCnsr7P&md5=b1abe6a7abcbf81f7b089d56a90f69e0The interplay between blood proteins, complement, and macrophages on nanomedicine performance and responsesMoghimi, S. Moein; Simberg, Dmitri; Skotland, Tore; Yaghmur, Anan; Hunter, A. ChristyJournal of Pharmacology and Experimental Therapeutics (2019), 370 (3), 581-592CODEN: JPETAB; ISSN:1521-0103. (American Society for Pharmacology and Experimental Therapeutics)A review. In the blood, depending on their physicochem. characteristics, nanoparticles attract a wide range of plasma biomols. The majority of blood biomols. bind nonspecifically to nanoparticles. On the other hand, biomols. such as pattern-recognition complement-sensing proteins may recognize some structural determinants of the pristine surface, causing complement activation. Adsorption of nonspecific blood proteins could also recruit natural antibodies and initiate complement activation, and this seems to be a global process with many preclin. and clin. nanomedicines. We discuss these issues, since complement activation has ramifications in nanomedicine stability and pharmacokinetics, as well as in inflammation and disease progression. Some studies have also predicted a role for complement systems in infusion-related reactions, whereas others show a direct role for macrophages and other immune cells independent of complement activation. We comment on these discrepancies and suggest directions for exploring the underlying mechanisms.
- 48Verhoef, J.; Anchordoquy, T. Drug Delivery Transl. Res. 2013, 3, 499– 503, DOI: 10.1007/s13346-013-0176-548https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslCmtLrF&md5=6ec259f3c3bd2d3b4dda73c3d75d1c9aQuestioning the use of PEGylation for drug deliveryVerhoef, Johan J. F.; Anchordoquy, Thomas J.Drug Delivery and Translational Research (2013), 3 (6), 499-503CODEN: DDTRCY; ISSN:2190-3948. (Springer)A review. Polyethylene glycol (PEG) is widely utilized in drug delivery and nanotechnol. due to its reported "stealth" properties and biocompatibility. It is generally thought that PEGylation allows particulate delivery systems and biomaterials to evade the immune system and thereby prolong circulation lifetimes. However, numerous studies over the past decade have demonstrated that PEGylation causes significant redns. in drug delivery, including enhanced serum protein binding, reduced uptake by target cells, and the elicitation of an immune response that facilitates clearance in vivo. This report reviews some of the extensive literature documenting the detrimental effects of PEGylation, and thereby questions the wisdom behind employing this strategy in drug development.
- 49Song, J.; Ju, Y.; Amarasena, T. H.; Lin, Z.; Mettu, S.; Zhou, J.; Rahim, M. A.; Ang, C.-S.; Cortez-Jugo, C.; Kent, S. J.; Caruso, F. Influence of Poly (Ethylene Glycol) Molecular Architecture on Particle Assembly and Ex Vivo Particle–Immune Cell Interactions in Human Blood. ACS Nano 2021, 15, 10025, DOI: 10.1021/acsnano.1c0164249https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2c%252FmsVygsQ%253D%253D&md5=684f7871c88d26995c23227cddc1b8fdInfluence of Poly(ethylene glycol) Molecular Architecture on Particle Assembly and Ex Vivo Particle-Immune Cell Interactions in Human BloodSong Jiaying; Ju Yi; Lin Zhixing; Zhou Jiajing; Rahim Md Arifur; Cortez-Jugo Christina; Caruso Frank; Amarasena Thakshila H; Kent Stephen J; Mettu Srinivas; Ang Ching-SengACS nano (2021), 15 (6), 10025-10038 ISSN:.Poly(ethylene glycol) (PEG) is widely used in particle assembly to impart biocompatibility and stealth-like properties in vivo for diverse biomedical applications. Previous studies have examined the effect of PEG molecular weight and PEG coating density on the biological fate of various particles; however, there are few studies that detail the fundamental role of PEG molecular architecture in particle engineering and bio-nano interactions. Herein, we engineered PEG particles using a mesoporous silica (MS) templating method and investigated how the PEG building block architecture impacted the physicochemical properties (e.g., surface chemistry and mechanical characteristics) of the PEG particles and subsequently modulated particle-immune cell interactions in human blood. Varying the PEG architecture from 3-arm to 4-arm, 6-arm, and 8-arm generated PEG particles with a denser, stiffer structure, with increasing elastic modulus from 1.5 to 14.9 kPa, inducing an increasing level of immune cell association (from 15% for 3-arm to 45% for 8-arm) with monocytes. In contrast, the precursor PEG particles with the template intact (MS@PEG) were stiffer and generally displayed higher levels of immune cell association but showed the opposite trend-immune cell association decreased with increasing PEG arm numbers. Proteomics analysis demonstrated that the biomolecular corona that formed on the PEG particles minimally influenced particle-immune cell interactions, whereas the MS@PEG particle-cell interactions correlated with the composition of the corona that was abundant in histidine-rich glycoproteins. Our work highlights the role of PEG architecture in the design of stealth PEG-based particles, thus providing a link between the synthetic nature of particles and their biological behavior in blood.
- 50Kristensen, K.; Urquhart, A. J.; Thormann, E.; Andresen, T. L. Binding of Human Serum Albumin to PEGylated Liposomes: Insights into Binding Numbers and Dynamics by Fluorescence Correlation Spectroscopy. Nanoscale 2016, 8, 19726– 19736, DOI: 10.1039/C6NR05455B50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xhs1yku7zO&md5=791626aae556dd6a559a79373b16e80dBinding of human serum albumin to PEGylated liposomes: insights into binding numbers and dynamics by fluorescence correlation spectroscopyKristensen, Kasper; Urquhart, Andrew J.; Thormann, Esben; Andresen, Thomas L.Nanoscale (2016), 8 (47), 19726-19736CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)Liposomes for medical applications are often administered by i.v. injection. Once in the bloodstream, the liposomes are covered with a "protein corona", which impacts the behavior and eventual fate of the liposomes. Currently, many aspects of the liposomal protein corona are not well understood. For example, there is generally a lack of knowledge about the liposome binding affinities and dynamics of common types of blood plasma proteins. Fluorescence correlation spectroscopy (FCS) is a powerful exptl. technique that potentially can provide such knowledge. In this study, we have used FCS to investigate the binding of human serum albumin (HSA) to std. types of PEGylated fluid-phase liposomes (consisting of DOPC and DOPE-PEG2k) and PEGylated gel-phase liposomes (consisting of DSPC and DSPE-PEG2k) with various PEG chain surface densities. We detected no significant binding of HSA to the PEGylated fluid-phase liposomes. In contrast, we found that HSA bound tightly to the PEGylated gel-phase liposomes, although only a low no. of HSA mols. could be accommodated per liposome. Overall, we believe that our data provides a useful benchmark for other researchers interested in studying the liposomal protein corona.
- 51Jhaveri, A.; Deshpande, P.; Pattni, B.; Torchilin, V. Transferrin-Targeted, Resveratrol-Loaded Liposomes for the Treatment of Glioblastoma. Journal of controlled release 2018, 277, 89– 101, DOI: 10.1016/j.jconrel.2018.03.00651https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXlt1yjurw%253D&md5=bd9a0331761b9dcc86c394ea852e3875Transferrin-targeted, resveratrol-loaded liposomes for the treatment of glioblastomaJhaveri, Aditi; Deshpande, Pranali; Pattni, Bhushan; Torchilin, VladimirJournal of Controlled Release (2018), 277 (), 89-101CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Glioblastomas (GBMs) are highly aggressive brain tumors with a very grim prognosis even after multi-modal therapeutic regimens. Conventional chemotherapeutic agents frequently lead to drug resistance and result in severe toxicities to non-cancerous tissues. Resveratrol (RES), a natural polyphenol with pleiotropic health benefits, has proven chemopreventive effects in all the stages of cancer including initiation, promotion and progression. However, the poor physico-chem. properties of RES severely limit its use as a free drug. In this study, RES was loaded into PEGylated liposomes (RES-L) to counter its drawbacks as a free drug. Since transferrin receptors (TfRs) are up-regulated in GBM, the liposome surface was modified with transferrin moieties (Tf-RES-L) to make them cancer cell-specific. The liposomal nanomedicines developed in this project were aimed at enhancing the physico-chem. properties of RES and exploiting the passive and active targeting capabilities of liposomes to effectively treat GBM. The RES-L were stable, had a good drug-loading capacity, prolonged drug-release in vitro and were easily scalable. Flow cytometry and confocal microscopy were used to study the assocn. with, and internalization of, Tf-L into U-87 MG cells. The Tf-RES-Ls were significantly more cytotoxic and induced higher levels of apoptosis accompanied by activation of caspases 3/7 in GBM cells when compared to free RES or RES-L. The ability of RES to arrest cells in the S-phase of the cell cycle, and selectively induce prodn. of reactive oxygen species in cancer cells were probably responsible for its cytotoxic effects. The therapeutic efficacy of RES formulations was evaluated in a s.c. xenograft mouse model of GBM. A tumor growth inhibition study and a modified survival study showed that Tf-RES-Ls were more effective than other treatments in their ability to inhibit tumor growth and improve survival in mice. Overall, the liposomal nanomedicines of RES developed in this project exhibited favorable in vitro and in vivo efficacies, which warrant their further investigation for the treatment of GBMs.
- 52Moyer, L. S.; Gorin, M. H. Eletectrokinetic Aspects of Surface Chemistry: IX. The Electric Mobilitites of Quartz and Collodion Particles in Mixtures of Horse Serum and Serum Proteins in Relation to the Mechanisms of Film Formation. J. Biol. Chem. 1940, 133, 605– 619, DOI: 10.1016/S0021-9258(18)73343-752https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaH3cXis1Knsg%253D%253D&md5=697ea21300760c5b8cd4b6c49c76a888Electrokinetic aspects of surface chemistry. IX. Electric mobilities of quartz and collodion particles in mixtures of horse serum and serum proteins in relation to the mechanism of film formationMoyer, Laurence S.; Gorin, Manuel H.Journal of Biological Chemistry (1940), 133 (), 605-19CODEN: JBCHA3; ISSN:0021-9258.cf. C. A. 34, 1696.3. The electrophoretic mobilities of quartz and collodion particles were detd. after exposure to mixts. of serum proteins or after they had been coated with one protein and then exposed to another or to serum. There is little tendency for the various constituents used to adsorb on each other, but one protein may replace another at a surface; thus a film of one of the protein components is obtained rather than a mosaic. The nature of the underlying surface influences the adsorption, the more hydrophilic proteins being adsorbed more readily by the more hydrophilic surfaces, and vice versa. Globulin when coated on quartz appears to be either irreversibly adsorbed or the most strongly adsorbed of the three proteins, replacement by albumin not occurring. The tendency to be adsorbed on quartz is roughly in the order albumin A > albumin B = globulin, with the addnl. assumption that globulin can replace the albumins whenever the free energy differences are favorable. On collodion, globulin is more strongly adsorbed than albumin A or B. The biol. significance of these findings is discussed.
- 53Caracciolo, G.; Cardarelli, F.; Pozzi, D.; Salomone, F.; Maccari, G.; Bardi, G.; Capriotti, A. L.; Cavaliere, C.; Papi, M.; Laganà, A. Selective Targeting Capability Acquired with a Protein Corona Adsorbed on the Surface of 1,2-Dioleoyl-3-Trimethylammonium Propane/DNA Nanoparticles. ACS Appl. Mater. Interfaces 2013, 5, 13171– 13179, DOI: 10.1021/am404171h53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsl2nsbjI&md5=0a989575d1ad35338a3689251a70dcb4Selective Targeting Capability Acquired with a Protein Corona Adsorbed on the Surface of 1,2-Dioleoyl-3-trimethylammonium Propane/DNA NanoparticlesCaracciolo, Giulio; Cardarelli, Francesco; Pozzi, Daniela; Salomone, Fabrizio; Maccari, Giuseppe; Bardi, Giuseppe; Capriotti, Anna Laura; Cavaliere, Chiara; Papi, Massimiliano; Lagana, AldoACS Applied Materials & Interfaces (2013), 5 (24), 13171-13179CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)A possible turning point in drug delivery has been recently reached: the protein shell, which covers nanocarriers in vivo, can be used for targeting. Here, we show that nanoparticles can acquire a selective targeting capability with a protein corona adsorbed on the surface. We demonstrate that lipid particles made of 1,2-dioleoyl-3-trimethylammonium propane (DOTAP) and DNA, upon interaction with human plasma components, spontaneously become coated with vitronectin that promotes efficient uptake in cancer cells expressing high levels of the vitronectin ανβ3 integrin receptor.
- 54Digiacomo, L.; Cardarelli, F.; Pozzi, D.; Palchetti, S.; Digman, M.; Gratton, E.; Capriotti, A.; Mahmoudi, M.; Caracciolo, G. An Apolipoprotein-Enriched Biomolecular Corona Switches the Cellular Uptake Mechanism and Trafficking Pathway of Lipid Nanoparticles. Nanoscale 2017, 9, 17254– 17262, DOI: 10.1039/C7NR06437C54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1yjt7zN&md5=d533413573a757ffaeea4b52dc2bf837An apolipoprotein-enriched biomolecular corona switches the cellular uptake mechanism and trafficking pathway of lipid nanoparticlesDigiacomo, L.; Cardarelli, F.; Pozzi, D.; Palchetti, S.; Digman, M. A.; Gratton, E.; Capriotti, A. L.; Mahmoudi, M.; Caracciolo, G.Nanoscale (2017), 9 (44), 17254-17262CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)Following exposure to biol. milieus (e.g. after systemic administration), nanoparticles (NPs) get covered by an outer biomol. corona (BC) that defines many of their biol. outcomes, such as the elicited immune response, biodistribution, and targeting abilities. In spite of this, the role of BC in regulating the cellular uptake and the subcellular trafficking properties of NPs has remained elusive. Here, we tackle this issue by employing multicomponent (MC) lipid NPs, human plasma (HP) and HeLa cells as models for nanoformulations, biol. fluids, and target cells, resp. By conducting confocal fluorescence microscopy expts. and image correlation analyses, we quant. demonstrate that the BC promotes a neat switch of the cell entry mechanism and subsequent intracellular trafficking, from macropinocytosis to clathrin-dependent endocytosis. Nano-liq. chromatog. tandem mass spectrometry identifies apolipoproteins as the most abundant components of the BC tested here. Interestingly, this class of proteins target the LDL receptors, which are overexpressed in clathrin-enriched membrane domains. Our results highlight the crucial role of BC as an intrinsic trigger of specific NP-cell interactions and biol. responses and set the basis for a rational exploitation of the BC for targeted delivery.
- 55Digiacomo, L.; Giulimondi, F.; Capriotti, A. L.; Piovesana, S.; Montone, C. M.; Chiozzi, R. Z.; Laganà, A.; Mahmoudi, M.; Pozzi, D.; Caracciolo, G. Optimal Centrifugal Isolating of Liposome–Protein Complexes From Human Plasma. Nanoscale Advances 2021, 3, 3824, DOI: 10.1039/D1NA00211B55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtV2ksb3O&md5=b8e3d0d0602076e429d625e96bd89d2bOptimal centrifugal isolating of liposome-protein complexes from human plasmaDigiacomo, Luca; Giulimondi, Francesca; Capriotti, Anna Laura; Piovesana, Susy; Montone, Carmela Maria; Chiozzi, Riccardo Zenezini; Lagana, Aldo; Mahmoudi, Morteza; Pozzi, Daniela; Caracciolo, GiulioNanoscale Advances (2021), 3 (13), 3824-3834CODEN: NAADAI; ISSN:2516-0230. (Royal Society of Chemistry)In the past few years, characterization of the protein corona (PC) that forms around liposomal systems has gained increasing interest for the development of novel therapeutic and diagnostic technologies. At the crossroads of fast-moving research fields, the interdisciplinarity of protein corona investigations poses challenges for exptl. design and reporting. Isolation of liposome-protein complexes from biol. fluids has been identified as a fundamental step of the entire workflow of PC characterization but exact specifications for conditions to optimize pelleting remain elusive. In the present work, key factors affecting pptn. of liposome-protein complexes by centrifugation, including time of centrifugation, total sample vol., lipid : protein ratio and contamination from biol. NPs were comprehensively evaluated. Here we show that the total amt. of isolated liposome-protein complexes and the extent of contamination from biol. NPs may vary with influence factors. Our results provide protein corona researchers with precise indications to sep. liposome-protein complexes from protein-rich fluids and include proper controls, thus they are anticipated to catalyze improved consistency of data mining and computational modeling of protein corona compn.
- 56Franciosa, G; Diluvio, G; Gaudio, F D.; Giuli, M V; Palermo, R; Grazioli, P; Campese, A F; Talora, C; Bellavia, D; D'Amati, G; Besharat, Z M; Nicoletti, C; Siebel, C W; Choy, L; Rustighi, A; Sal, G D.; Screpanti, I; Checquolo, S Prolyl-Isomerase Pin1 Controls Notch3 Protein Expression and Regulates T-ALL Progression. Oncogene 2016, 35, 4741– 4751, DOI: 10.1038/onc.2016.556https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XisFWqsL4%253D&md5=956448aa6da4918fc70930ce436fca72Prolyl-isomerase Pin1 controls Notch3 protein expression and regulates T-ALL progressionFranciosa, G.; Diluvio, G.; Del Gaudio, F.; Giuli, M. V.; Palermo, R.; Grazioli, P.; Campese, A. F.; Talora, C.; Bellavia, D.; D'Amati, G.; Besharat, Z. M.; Nicoletti, C.; Siebel, C. W.; Choy, L.; Rustighi, A.; Sal, G. Del; Screpanti, I.; Checquolo, S.Oncogene (2016), 35 (36), 4741-4751CODEN: ONCNES; ISSN:0950-9232. (Nature Publishing Group)Deregulated Notch signaling is assocd. with T-cell Acute Lymphoblastic Leukemia (T-ALL) development and progression. Increasing evidence reveals that Notch pathway has an important role in the invasion ability of tumor cells, including leukemia, although the underlying mol. mechanisms remain mostly unclear. Here, we show that Notch3 is a novel target protein of the prolyl-isomerase Pin1, which is able to regulate Notch3 protein processing and to stabilize the cleaved product, leading to the increased expression of the intracellular domain (N3IC), finally enhancing Notch3-dependent invasiveness properties. We demonstrate that the combined inhibition of Notch3 and Pin1 in the Notch3-overexpressing human leukemic TALL-1 cells reduces their high invasive potential, by decreasing the expression of the matrix metalloprotease MMP9. Consistently, Pin1 depletion in a mouse model of Notch3-induced T-ALL, by reducing N3IC expression and signaling, impairs the expansion/invasiveness of CD4+CD8+ DP cells in peripheral lymphoid and non-lymphoid organs. Notably, in in silico gene expression anal. of human T-ALL samples we obsd. a significant correlation between Pin1 and Notch3 expression levels, which may further suggest a key role of the newly identified Notch3-Pin1 axis in T-ALL aggressiveness and progression. Thus, combined suppression of Pin1 and Notch3 proteins may be exploited as an addnl. target therapy for T-ALL.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.1c07687.
Supporting Excel file, scheme of the in vitro and in vivo experiments (XLSX)
Figure S1, size, zeta potential, synchrotron SAXS patterns, and the corresponding computed electron density profiles of cationic liposomes CL1 and CL2; Table S1, DLS and synchrotron SAXS parameters for cationic liposomes CL1 and CL2; Figure S2, representative transmission electron microscopy (TEM) images of plain and DNA-decorated lipoplexes; Figure S3, protein corona analysis of plain and DNA-decorated lipoplexes at increasing amounts of human plasma (HP); Table S2, lists of plasma proteins identified in the coronas of DDLs and PLs by nano-LC-MS/MS; Figure S4, gating strategy of mouse phagocyte populations; Figure S5, characterization of CL2/oligonucleotide (ON) lipoplexes as a function of the cationic lipid/ON weight ratio, ρ; Figure S6, luciferase activity in HEK293/Luc reporter cells transfected or not with TLR9 and incubated for 5 h with PL1, PL2, DDL1, DDL2, or a specific TLR9 agonist; and Supplementary Table S3, companion checklist about Minimum Information Reporting in Bio–Nano Experimental Literature (MIRIBEL) (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.