Nanoparticle size and surface chemistry determine serum protein adsorption and macrophage uptake.

Delivery and toxicity are critical issues facing nanomedicine research. Currently, there is limited understanding and connection between the physicochemical properties of a nanomaterial and its interactions with a physiological system. As a result, it remains unclear how to optimally synthesize and chemically modify nanomaterials for in vivo applications. It has been suggested that the physicochemical properties of a nanomaterial after synthesis, known as its "synthetic identity", are not what a cell encounters in vivo. Adsorption of blood components and interactions with phagocytes can modify the size, aggregation state, and interfacial composition of a nanomaterial, giving it a distinct "biological identity". Here, we investigate the role of size and surface chemistry in mediating serum protein adsorption to gold nanoparticles and their subsequent uptake by macrophages. Using label-free liquid chromatography tandem mass spectrometry, we find that over 70 different serum proteins are heterogeneously adsorbed to the surface of gold nanoparticles. The relative density of each of these adsorbed proteins depends on nanoparticle size and poly(ethylene glycol) grafting density. Variations in serum protein adsorption correlate with differences in the mechanism and efficiency of nanoparticle uptake by a macrophage cell line. Macrophages contribute to the poor efficiency of nanomaterial delivery into diseased tissues, redistribution of nanomaterials within the body, and potential toxicity. This study establishes principles for the rational design of clinically useful nanomaterials.

[1]  George M. Whitesides,et al.  Molecular Conformation in Oligo(ethylene glycol)-Terminated Self-Assembled Monolayers on Gold and Silver Surfaces Determines Their Ability To Resist Protein Adsorption , 1998 .

[2]  J. Koziol,et al.  Label-free, normalized quantification of complex mass spectrometry data for proteomics analysis , 2009, Nature Biotechnology.

[3]  Warren C W Chan,et al.  Nanoparticle-mediated cellular response is size-dependent. , 2008, Nature nanotechnology.

[4]  B. Bunker,et al.  Grafting of High-Density Poly(Ethylene Glycol) Monolayers on Si(111) , 2001 .

[5]  Jürgen Groll,et al.  Rapid uptake of gold nanorods by primary human blood phagocytes and immunomodulatory effects of surface chemistry. , 2010, ACS nano.

[6]  Byron Ballou,et al.  Noninvasive imaging of quantum dots in mice. , 2004, Bioconjugate chemistry.

[7]  Chad A Mirkin,et al.  The role radius of curvature plays in thiolated oligonucleotide loading on gold nanoparticles. , 2009, ACS nano.

[8]  Z. Ku,et al.  Nanostructures and Functional Materials Fabricated by Interferometric Lithography , 2011, Advanced materials.

[9]  Mauro Ferrari,et al.  Seven challenges for nanomedicine. , 2008, Nature nanotechnology.

[10]  T. Veenstra,et al.  The Human Plasma Proteome , 2004, Molecular & Cellular Proteomics.

[11]  Ralph Weissleder,et al.  A light-activated theranostic nanoagent for targeted macrophage ablation in inflammatory atherosclerosis. , 2010, Small.

[12]  Younan Xia,et al.  The effect of sedimentation and diffusion on cellular uptake of gold nanoparticles. , 2011, Nature nanotechnology.

[13]  Kinam Park To PEGylate or not to PEGylate, that is not the question. , 2010, Journal of controlled release : official journal of the Controlled Release Society.

[14]  C. Salesse,et al.  On the Nature of Conformational Transition in Poly(ethylene glycol) Chains Grafted onto Phospholipid Monolayers , 2004 .

[15]  Christian Mühlfeld,et al.  Quantitative evaluation of cellular uptake and trafficking of plain and polyethylene glycol-coated gold nanoparticles. , 2010, Small.

[16]  Warren C W Chan,et al.  Mediating tumor targeting efficiency of nanoparticles through design. , 2009, Nano letters.

[17]  James S Murday,et al.  Translational nanomedicine: status assessment and opportunities. , 2009, Nanomedicine : nanotechnology, biology, and medicine.

[18]  Helmut Thissen,et al.  Effects of cloud-point grafting, chain length, and density of PEG layers on competitive adsorption of ocular proteins. , 2002, Biomaterials.

[19]  W. Chan,et al.  Synthesis and surface modification of highly monodispersed, spherical gold nanoparticles of 50-200 nm. , 2009, Journal of the American Chemical Society.

[20]  Tanya S. Hauck,et al.  Exploring Primary Liver Macrophages for Studying Quantum Dot Interactions with Biological Systems , 2010, Advanced materials.

[21]  R. Langer,et al.  Emerging nanotechnology approaches for HIV/AIDS treatment and prevention. , 2010, Nanomedicine.

[22]  Chung-Yuan Mou,et al.  Size effect on cell uptake in well-suspended, uniform mesoporous silica nanoparticles. , 2009, Small.

[23]  Susan Newbigging,et al.  In vivo quantum-dot toxicity assessment. , 2010, Small.

[24]  Marcus Textor,et al.  Poly(l-lysine)-graft-poly(ethylene glycol) Assembled Monolayers on Niobium Oxide Surfaces: A Quantitative Study of the Influence of Polymer Interfacial Architecture on Resistance to Protein Adsorption by ToF-SIMS and in Situ OWLS , 2003 .

[25]  L. Unsworth,et al.  Protein-resistant poly(ethylene oxide)-grafted surfaces: chain density-dependent multiple mechanisms of action. , 2008, Langmuir : the ACS journal of surfaces and colloids.

[26]  S. Bhatia,et al.  Probing the Cytotoxicity Of Semiconductor Quantum Dots. , 2004, Nano letters.

[27]  Warren C W Chan,et al.  Effect of gold nanoparticle aggregation on cell uptake and toxicity. , 2011, ACS nano.

[28]  Ming-Hsien Tsai,et al.  Persistent Tissue Kinetics and Redistribution of Nanoparticles, Quantum Dot 705, in Mice: ICP-MS Quantitative Assessment , 2007, Environmental health perspectives.

[29]  Hans C. Fischer,et al.  Pharmacokinetics of Nanoscale Quantum Dots: In Vivo Distribution, Sequestration, and Clearance in the Rat , 2006 .

[30]  A Paul Alivisatos,et al.  From artificial atoms to nanocrystal molecules: preparation and properties of more complex nanostructures. , 2009, Annual review of physical chemistry.

[31]  Sara Linse,et al.  Understanding the nanoparticle–protein corona using methods to quantify exchange rates and affinities of proteins for nanoparticles , 2007, Proceedings of the National Academy of Sciences.

[32]  P Couvreur,et al.  Investigation of the role of macrophages on the cytotoxicity of doxorubicin and doxorubicin-loaded nanoparticles on M5076 cells in vitro. , 2000, Journal of controlled release : official journal of the Controlled Release Society.

[33]  Joseph D. Andrade,et al.  Protein—surface interactions in the presence of polyethylene oxide: II. Effect of protein size , 1991 .

[34]  L. Unsworth,et al.  Protein resistance of surfaces prepared by sorption of end-thiolated poly(ethylene glycol) to gold: effect of surface chain density. , 2005, Langmuir : the ACS journal of surfaces and colloids.

[35]  Seungpyo Hong,et al.  The Binding Avidity of a Nanoparticle-based Multivalent Targeted Drug Delivery Platform , 2022 .

[36]  Leaf Huang,et al.  Nanoparticles evading the reticuloendothelial system: role of the supported bilayer. , 2009, Biochimica et biophysica acta.

[37]  Steven P Gygi,et al.  Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry , 2007, Nature Methods.

[38]  Zahi A. Fayad,et al.  Perspectives and opportunities for nanomedicine in the management of atherosclerosis , 2011, Nature Reviews Drug Discovery.

[39]  Nicholas A Peppas,et al.  Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles. , 2006, International journal of pharmaceutics.

[40]  J. Satulovsky,et al.  Kinetic and thermodynamic control of protein adsorption. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[41]  Iseult Lynch,et al.  Quantitative assessment of the comparative nanoparticle-uptake efficiency of a range of cell lines. , 2011, Small.

[42]  R. M. Owen,et al.  Selective tumor cell targeting using low-affinity, multivalent interactions. , 2007, ACS chemical biology.

[43]  Andrew Emili,et al.  PRISM, a Generic Large Scale Proteomic Investigation Strategy for Mammals*S , 2003, Molecular & Cellular Proteomics.

[44]  Thomas Kislinger,et al.  Large-scale characterization and analysis of the murine cardiac proteome. , 2009, Journal of proteome research.

[45]  Lawrence Tamarkin,et al.  Colloidal Gold: A Novel Nanoparticle Vector for Tumor Directed Drug Delivery , 2004, Drug delivery.

[46]  Arezou A Ghazani,et al.  Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells. , 2006, Nano letters.

[47]  T. Allen Toxicity of drug carriers to the mononuclear phagocyte system , 1988 .

[48]  H. Kruth,et al.  Fluorescent pegylated nanoparticles demonstrate fluid-phase pinocytosis by macrophages in mouse atherosclerotic lesions. , 2009, The Journal of clinical investigation.

[49]  Istvan Toth,et al.  Nanoparticle-induced unfolding of fibrinogen promotes Mac-1 receptor activation and inflammation. , 2011, Nature nanotechnology.

[50]  Mark E. Davis,et al.  Nanoparticle therapeutics: an emerging treatment modality for cancer , 2008, Nature Reviews Drug Discovery.

[51]  Shuming Nie,et al.  Understanding and overcoming major barriers in cancer nanomedicine. , 2010, Nanomedicine.

[52]  Jean-Pierre Benoit,et al.  Parameters influencing the stealthiness of colloidal drug delivery systems. , 2006, Biomaterials.

[53]  J Szebeni,et al.  Complement activation cascade triggered by PEG-PL engineered nanomedicines and carbon nanotubes: the challenges ahead. , 2010, Journal of controlled release : official journal of the Controlled Release Society.

[54]  Kenneth A. Dawson,et al.  Nanoparticle size and surface properties determine the protein corona with possible implications for biological impacts , 2008, Proceedings of the National Academy of Sciences.

[55]  I. Szleifer,et al.  Protein adsorption on surfaces with grafted polymers: a theoretical approach. , 1997, Biophysical journal.

[56]  Parag Aggarwal,et al.  Interaction of colloidal gold nanoparticles with human blood: effects on particle size and analysis of plasma protein binding profiles. , 2009, Nanomedicine : nanotechnology, biology, and medicine.

[57]  S M Moghimi,et al.  Long-circulating and target-specific nanoparticles: theory to practice. , 2001, Pharmacological reviews.

[58]  Leaf Huang,et al.  Pharmacokinetics and biodistribution of nanoparticles. , 2008, Molecular pharmaceutics.

[59]  M. van Lookeren Campagne,et al.  Macrophage complement receptors and pathogen clearance , 2007, Cellular microbiology.

[60]  Younan Xia,et al.  Understanding the role of surface charges in cellular adsorption versus internalization by selectively removing gold nanoparticles on the cell surface with a I2/KI etchant. , 2009, Nano letters.

[61]  Warren C W Chan,et al.  Understanding and controlling the interaction of nanomaterials with proteins in a physiological environment. , 2012, Chemical Society reviews.

[62]  Subra Suresh,et al.  Size‐Dependent Endocytosis of Nanoparticles , 2009, Advanced materials.

[63]  O. Bourdon,et al.  Relationship between complement activation, cellular uptake and surface physicochemical aspects of novel PEG-modified nanocapsules. , 2001, Biomaterials.

[64]  Joseph D. Andrade,et al.  Protein—surface interactions in the presence of polyethylene oxide , 1991 .

[65]  T. Xia,et al.  Understanding biophysicochemical interactions at the nano-bio interface. , 2009, Nature materials.

[66]  S. Nie,et al.  Nanotechnology applications in cancer. , 2007, Annual review of biomedical engineering.

[67]  Takuro Niidome,et al.  PEG-modified gold nanorods with a stealth character for in vivo applications. , 2006, Journal of controlled release : official journal of the Controlled Release Society.