You are on page 1of 16

Biomedicine & Pharmacotherapy 139 (2021) 111541

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

Research progress and application opportunities of nanoparticle–protein


corona complexes
Wei Huang a, b, Gao Xiao c, Yujuan Zhang b, *, Weiping Min b
a
Department of Pharmacy, The First People’s Hospital of Jiande, Jiande 311600, China
b
Department of immunology, School of Basic Medical Sciences and School of Pharmacy, Nanchang University, Nanchang 330006, China
c
College of Environment and Resources, Fuzhou University, Fuzhou 350108, China

A R T I C L E I N F O A B S T R A C T

Keywords: Nanoparticles (NPs) can be used to design for nanomedicines with different chemical surface properties owing to
Nanoparticles their size advantages and the capacity of specific delivery to targeted sites in organisms. The discovery of the
Protein corona presence of protein corona (PC) has changed our classical view of NPs, stimulating researchers to investigate the
Targeted drug delivery
in vivo fate of NPs as they enter biological systems. Both NPs and PC have their specificity but complement each
Immune
other, so they should be considered as a whole. The formation and characterization of NP-PC complexes provide
new insights into the design, functionalization, and application of nanocarriers. Based on progress of recent
researches, we reviewed the formation, characterization, and composition of the PC, and introduced those
critical factors influencing PC, simultaneously expound the effect of PC on the biological function of NPs.
Especially we put forward the opportunities and challenges when NP-PC as a novel nano-drug carrier for targeted
applications. Furthermore, we discussed the pros versus cons of the PC, as well as how to make better PC in the
future application of NPs.

1. Introduction nanomaterial, whether active or not, has an average dose of only 0.7%
that reaches the tumors [1,2]. This reminds us that, even if the active
In recent decades, with the development of nanotechnology and the targeting strategy exists, the actual concentration of the material
invention of various nanomaterials, an increasing number of nano­ reaching the target site may not be as good as expected. We need to take
materials has been applied in the fields of biology and medicine. NPs it into consideration about the fate of nanomaterials as they enter living
have been designed as nanomedicine delivery systems with different systems.
surface chemistries for precise delivery to the diseased sites. Solid tu­ Either passive targeting or active targeting strategy including the
mors exhibit enhanced permeability and retention effects, while NPs are design, synthesis and production of NPs can be achieved through in vitro
small enough to leak through loose porous blood vessels and enter the experimental manipulations. However, the fate and function of NPs
tumor tissue without penetrating into the normal tissue through the entering the biological system are unknown. According to the relevant
dense normal blood vessels. This passively tumor-targeted strategy literatures, NPs have a large specific surface area and high surface free
prevents NPs from accumulating in the heart and other non-targeting energy. After entering the biological system, NPs are more likely to
sites, thereby reducing the toxic side effects on normal tissues. Howev­ adsorb various biological molecules, such as nucleic acids, cytokines,
er, this strategy still cannot prevent the NPs from accumulating in amino acids, and proteins, to form a biomolecular layer on the surface,
certain organs such as spleen and kidney. Another strategy is to modify which is mainly composed of proteins, so called a “protein corona”. PC
NPs with the targeting molecules, making NPs possessing an active can affect the surface physicochemical properties of NPs, specific tar­
targeting function; however, this is difficult to be implemented. Warren geting, toxicity, in vivo clearance, cellular uptake, signal transduction,
Chan and his colleagues have reviewed more than 100 nanomedical and immune response [3–6]. This review will begin with an explanation
papers published over the past 10 years and found that any of how the PC is produced and list the factors that influence its

Abbreviations: NP, nanoparticles; PC, protein corona; HSA, human serum albumin; AuNR, gold nanorods; MNPs, magnetic nanoparticles; BSA, bovine serum
albumin; FBS, fetal bovine serum; PEG, polyethylene glycol.
* Corresponding author.
E-mail address: yujuanzhang@ncu.edu.cn (Y. Zhang).

https://doi.org/10.1016/j.biopha.2021.111541
Received 27 November 2020; Received in revised form 22 February 2021; Accepted 23 March 2021
Available online 10 April 2021
0753-3322/© 2021 The Author(s). Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

formation. Then we will discuss whether a PC changes the properties of sufficient, the surface of the NPs will be completely occupied by hard
NPs. Finally, we will summarize the interaction between NPs and cells, corona proteins, and the soft corona protein will adhere to the surface of
to show how the NP-PC complex presents a challenge for the application the hard corona protein. However, when the affinity of the protein is
and development of new nano-drug delivery systems. low, it may cause the unstable NPs to aggregate or still adsorb some of
the low affinity, reversible soft corona proteins. The soft corona protein
2. Formation, characterization, and composition of the PC changes with the biological environment, while the hard corona protein
adsorbed on the surface of the NPs maintains the initial binding status. A
2.1. Formation and development schematic diagram of the dynamic adsorption of NPs and proteins is
shown in Fig. 1. Unlike most of the studies that separated NPs from
Owing to the high percentage of surface atoms and colloidal nature protein-containing fluids, Shang et al. used fluorescence correlation
of NPs, upon contacting with physiological media, the NPs will adopt spectroscopy (FCS) as an in situ technique to detect NP-protein in­
more stable thermodynamic states [7], which includes aggregation, teractions while the NPs were suspended in biological fluids [14]. The
corrosion, dissolution and interaction with mediator proteins. Vorman authors observed that the protein layer thickness always matched the
[8] found that at least 5 proteins were substituted with each other on one known molecular dimensions of the proteins binding in certain orien­
surface in plasma within 1 min. This indicated that the surface contact of tations, such as human serum albumin (HSA). Notably, they pointed out
inorganic NPs and proteins occurs promptly when NPs enter a physio­ that while the corona formed from a single type of serum protein was
logical environment, such as plasma, and the protein adsorbed on the reversible, protein adsorption from a complex biological media, such as
surface of the NP changes with the exposure time. At first, the proteins blood serum, was entirely irreversible.
transiently adsorbed are mostly those with a high-concentration and The rearrangement process that proteins undergo on the surface of
low-affinity. At this stage, the adsorption is reversible and the proteins NPs is called the conformational change. Direct contact of hard corona
can be desorbed. As time progresses, these proteins are gradually proteins with NPs typically causes changes in the secondary structure
replaced by high-affinity proteins. At the end, the protein adsorption (such as loss of α-helical and increase of β-sheets or β-turns), which can
becomes irreversible. The continuous adsorption and desorption process result in a partial loss of biochemical activity [15–17]. The thickness of
of this plasma protein on the surface involves factors such as incubation the PC is dependent on many parameters such as protein concentration,
time and surface properties, which is called the “Vorman effect” [8–10]. particle size, and surface properties of the particle [17].
Like gas adsorption, proteins will randomly attach to the surface of NPs
at the beginning, then they will recombine on the surface of NPs through
a rearrangement reaction and crowding effect to form a domain [11], 2.2. Identification and analysis methods
which eventually exists in an irreversible minimum energy state.
The protein layer adsorbed on the surface of the NP is determined by Protein-bound NPs possess new roles, which need certain methods to
the surface properties of NPs, exposure time, protein concentration and identify and analyze them. Some methods can qualitatively describe the
affinity of the protein. A protein layer, which has a high affinity, high changes in the physicochemical properties of nanoparticles before and
binding rate and irreversible adsorption on the surface of the NP, is after the coating of proteins. Generally, scanning electron microscopy
called a "hard corona". In contrast, a protein layer with low affinity and (SEM) and transmission electron microscopy (TEM) are the most com­
reversible protein adsorption layer is called a "soft corona" [12,13]. If mon, direct, clear and persuasive methods to identify a new NP. TEM
the affinity of the protein is high enough and the exposure time is can directly observe the morphology of nanomaterials, especially the
finer and smaller nanostructures [18,19]. Wang et al. [20] observed a

Fig. 1. Hypothetical dynamic adsorption diagram of proteins on the surface of a NP. Proteins on the NP are always in the dynamic process of adsorption and
desorption. With the prolongation of exposure time, the low affinity and high concentration of proteins close to the NP would be replaced by proteins with high
affinity and low concentration, which rearrange at the surface of the particle and eventually form an irreversible stably hard corona composed of tightly bound
proteins, while the replaced proteins absorbed on the outside form a reversible unsteadily soft corona consisting of loosely bound proteins.

2
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

thicker organic layer of gold nanorods (AuNRs) after protein adsorption tool can be used to monitor the physical status of protein-coated AuNPs
and measured the thickness of the PC through TEM images, and in the two media. Moreover, surface-enhanced Raman scattering (SERS)
concluded that the serum PC not only increased the stability of AuNRs, was also often used in the study of NP-PC complexes. Winuprasith et al.
but also prevented their directly destructive effects on membrane [37] observed protein-coated NPs after bile salt addition contained
structures by SEM and TEM images of cytoplasmic membrane structures. bands from both protein and bile salts by SERS spectra, indicating that
Cryogenic transmission electron microscopy (Cryo-TEM) is more suit­ the protein was not fully displaced by the bile salts. The SERS spectra of
able for the observation of proteins, biological slices and other bovine serum (BS) incubated particles showed peak broadening,
temperature-sensitive samples, but has strict requirements on the tech­ consistent with significant conformational changes to proteins in the PC
nical operation of sample preparation [21]. Therefore, Cryo-TEM is [38]. Fluorescence correlation spectroscopy (FCS) and fluorescence
suitable to visualize the soft corona (weakly bound proteins) but little cross-correlation spectroscopy (FCCS) are sensitive spectroscopy tech­
studied for its poor stability. Kokkinopoulou et al. [22] studied the niques that measure fluorescence intensity fluctuations of single mole­
evolution of soft plus hard coronas (in situ analysis) towards hard corona cules inside a femtoliter confocal volume, and they are suitable for
(after three washing steps) on polystyrene nanoparticles by using studying the formation of PC around NPs and for examining corona
Cryo-TEM. Franqui et al. [23] observed a higher electronic density stability in situ in biological matrixes [39]. Fourier transform infrared
distributed throughout the hard corona coated single-layer graphene spectroscopy (FTIR) is used to probe the absorption of infrared light on
oxide (HC@SLGO) surface than bare SLGO by Cryo-TEM images, and NP surfaces and to provide information on protein adsorption, including
Cryo-TEM allowed the in-situ observation of GO sheets in DMEM me­ the evaluation of protein coatings on the surfaces of NPs and identifi­
dium, once the sample was instantly frozen keeping its integrity in the cation of a protein’s secondary structure [40]. By comparing the FTIR of
solution media (i.e., avoiding the drying effect and the structural human serum (HS), magnetic nanoparticles (MNPs) and HS@MNPs, the
deformation of the sample by the microscope vacuum). In Franqui’s characteristic peaks at 1651 cm-1 and 1548 cm-1 indicated the presence
study, atomic force microscopy (AFM) allowed a visualization of the of human serum protein on MNPs, and excessive weight loss of
protein coating and its distribution on the GO surface, the measurement HS@MNPs specifies human serum protein degradation in thermograms
of surface roughness and thickness of GO-protein complexes. It is an [41].
important analysis method which can be used to evaluate the sizes of The most commonly used methods for the identification and quan­
particles and the thickness of PCs, as well as characterize the interaction tification of PC components are sodium dodecyl sulfate polyacrylamide
of proteins with NPs [24]. gel electrophoresis (SDS-PAGE) and mass spectrometry (MS) or liquid
In addition to the methods mentioned above that can directly chromatography–tandem mass spectrometry (LC-MS/MS) [32,42].
observe or image the morphology of NPs, there are also some indirect In addition, techniques have been developed to determine the
methods that can provide more information including charge, size, conformation of the binding protein using circular dichroism spectros­
structure, and so on. For charge analysis, zeta-potential analysis is the copy (CD), infrared (FTIR), SERS NMR, fluorescence quenching [43],
most commonly used and indispensable method. As for size analysis, the and computational simulations [17,44]. CD is sensitive to detect the NP
most conventional methods are dynamic light scattering (DLS) and interaction-induced conformational changes of secondary structures and
differential centrifugal sedimentation (DCS). DLS can measure the hy­ the folding and binding properties of proteins according to chiral
drodynamic size distribution of the NPs before and after protein properties [16,45,46]. SERS can provide information on the structure,
adsorption [25,26], while DCS is capable of separating the components composition, and interaction of molecules in the proximity of gold NPs,
of a mixture based on their densities and sizes, which can be applied in thereby enabling studies of adsorbed biomolecules in vivo [47]. It can
measuring the sizes of NP-PC complexes with a broad size range [27,28]. achieve in situ and real-time monitoring of ligand exchange reactions on
Moreover, small-angle X-ray scattering (SAXS) can be used to measure the gold surface [48]. By using SERS, Szekeres found bovine serum al­
the particles with the sizes in the range of 1–100 nm, especially ranging bumin (BSA) and HSA interacted with the citrate ligands at the NP
from 5 to 25 nm [29]. Although SAXS applied to biological macromol­ surface via lysine residues [49]. Circu et al. [50] investigated the surface
ecules is a low-resolution structural technique, it is particularly accurate interaction between the obtained nanosystems and BSA was by FTIR,
in distinguishing between different structural models proposed from specially emphasized on the conformational changes of BSA induced by
higher resolution techniques. Spinozzi et al. [30] propose a novel vinylimidazole-rare earth complexes. The results obtained from FTIR
method for determining the structural and thermodynamic properties of analysis showed the potential of these nanosystems to contribute to the
nanoparticle-protein complexes under physiological conditions by SAXS process of alpha/beta conversion of protein which highlights their ap­
and neutron-scattering measurements. Perez et al. [31] used plications in study of aspects regarding protein misfolding and aggre­
size-exclusion chromatography (SEC) and SAXS to model the detergent gation. Ban et al. [51] provides a method to accurately and
corona around aquaporin-0, a membrane protein of known structure. quantitatively predict the functional composition of the PC by inte­
There are also some spectroscopic methods used to analyze the in­ grating a machine learning model and meta-analysis. Their study found
teractions between NP and PC. UV–visible (UV–vis) spectroscopy is the functional protein compositions (e.g., immune proteins, complement
usually used to record the absorption peak spectra generated by the proteins, and apolipoproteins) in complex coronas were precisely pre­
surface plasmon resonance of metal NPs, whose position and intensity dicted with good R-2 (most over 0.80). Moreover, the method success­
depend on NP size, shape, and dielectric constant of the medium in close fully predicted the cellular recognition (e.g., cellular uptake by
proximity to the surface of the NPs [32]. In UV-Vis absorption spectrum, macrophages and cytokine release) mediated by functional corona
we can directly observe the red shift of the plasmonic resonance ab­ proteins. Some methods can be used to study the formation of PC in
sorption peaks caused by the changes of size and surface of NPs after complex environments. Carril et al. [52] mentioned a methodology for
proteins coated successfully [33], and the change of waveform caused determining PC formation in complex media by using nuclear magnetic
by the failure of coating [34]. In particular, the surface plasmon reso­ resonance (NMR) spectroscopy: NPs are labeled with (19)F and their
nance (SPR) mentioned in Cedervall’s study worked on the same prin­ diffusion coefficient measured using (19)F diffusion-ordered NMR. (19)
ciple. By using gold surfaces with thiol-conjugated NPs, they studied the F diffusion NMR measurements of hydrodynamic radii allow for in situ
kinetics of association and dissociation of plasma, HSA and fibrinogen characterization of NPs in complex environments by quantification of
with NPs. Then they found the hydrophobicity of the particles depended protein adsorption to the surface of NPs, as determined by increase in
the association and dissociation rates [35]. In Maiorano’s study, based hydrodynamic radius. Note that the methodology is not optics based,
on an enhanced plasmon resonance light-scattering peak at about and thus can be used in turbid environments, as in the presence of cells.
550 nm for the interparticle interactions of gold nanoparticles (AuNPs) Moreover, the use of site-specific labeling with spin probes and fluo­
[36], so plasmon resonance light scattering (PRLS) as a very sensitive rophores combined with electron paramagnetic resonance (EPR)

3
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

spectroscopy and fluorescence resonance energy transfer (FRET) can proteins, and each adsorbent protein mass was observed to exceed 10%
reveal the structural and dynamic changes of NPs interacting with blood of the total adsorbent protein mass on at least one of the nanomaterials.
proteins and in vivo [15]. The plasma PC appears to follow a general structure, with 2–6 proteins
Size exclusion chromatography (SEC), surface plasmon resonance adsorbed at high abundance, and many more adsorbed at low abun­
(SPR), and isothermal titration calorimetry (ITC) have also been re­ dance. The most abundant proteins accounted for average 29% of the
ported to estimate the affinity of binding proteins to the surface of total adsorbed protein, and the top 3 most abundant proteins accounted
nanomaterials [13,14]. Cedervall et al. [35] determined the rates of for average 56%. Some studies have also shown that among the
protein association and dissociation using SPR technology with nano­ approximately 3700 proteins in human plasma, only a few tens, at the
particles that are thiol-linked to gold, and through SEC of most, are ever abundant in the hard corona. In addition, these proteins
protein-nanoparticle mixtures. SEC can be used to isolate NP-associated rarely correspond to the most abundant proteins in plasma and are not
proteins as a systematic methodology. They also found ITC is suitable for necessarily proteins with the highest individual affinity for the surface of
studying the affinity and stoichiometry of protein binding to NPs. By NPs [58,59]. When NPs such as iron oxide, silica, AuNPs, liposomes and
using ITC, Danner et al. [53] found the protein affinity depends on the carbon nanotubes directly enter the blood environment, the following
number of phosphonate moieties on the particle surface, because initial proteins are mainly adsorbed: albumin, apolipoprotein, immunoglobu­
slope of the binding isotherm of ITC can present the binding affinity of lins, fibrinogen, complement [57,60–63]. The physiological functions
plasma proteins. It can also show the influence of the negative surface corresponding to these adsorbed proteins are mainly lipid transport,
charge of NPs toward the thermodynamic properties of the protein complement activation, coagulation, pathogen recognition and ion
adsorption. It is reported that FRET can also be used to study the affinity transport. Although each nanomaterial can adsorb large amounts of
and adsorption orientation of proteins in PC [46]. certain proteins, these abundant proteins are not always the same.
In general, one method is not enough to provide a comprehensive Additionally, even for a fixed material type, the size of the particle and
and accurate description and characterization of the PC, and it is usually its surface modification are able to entirely change the nature of the
combined with several different methods to analyze [54,55]. The above biologically active proteins in the corona, and thereby possibly also the
methods for studying PC are summarized in Fig. 2. biological impacts [64].
The composition of the PC formed by a single protein is relatively
2.3. Main protein composition simple, and the hard protein of the PC is the determinate protein. To
better understand the process of PC formation, some scientists start by
The composition and amount of proteins in the corona depends on modeling single protein, such as common albumin, immunoglobulin,
the physical and chemical characteristics of the NPs, the type of bio­ and uncommon soluble yeast protein extracts [65] and so on. By un­
logical medium and the exposure time [56], so that each nanomaterial’s derstanding the time and process of crowning of a single protein, we can
PC are unique. In addition to the physical and chemical properties of the also infer the status of the protein in serum or plasma environment
NPs, the PCs determine part of the biological functions of the NP-PC where is more complex. Some single protein interacts with specific
conjugates. NP-adsorbed proteins are identified to be important pro­ materials, such as magnetic nanomaterials and transferrin. That iron
teins involved in key biological processes. Walkey and Chan [57] sum­ content governs the PC formation and induces a strong effect on the
marized the composition and abundance of PCs formed by 63 thermal stability of the bound protein [66]. The study of a single protein
nanomaterials in 26 studies. They found that a total of 125 unique is beneficial for understanding its properties, but ultimately it will come
plasma proteins had been identified, of which 21 were high-abundance back to the study of NP and protein, protein and protein, protein and

Fig. 2. Graphic classification and summary of common methods for analyzing PC.

4
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

environment in the more complex environment of serum or plasma or NPs and HSA. They found that the size of the NPs directly influenced the
other biological fluid. nature of the PC, with larger particles favoring the formation of a soft
Serum stripped of fibrin contains 60–80 mg/mL protein, whose corona, owing to the decreased PS-HSA attraction.
57–71% is albumin, and followed by 8–26% are γ-globulins, especially
immunoglobulins G (IgG) [67]. Vitali et al. [63] confirmed that albumin 3.2. Concentrations of NPs in medium
and serum can develop a PC evolving from unstable (soft) to stable
(hard) with AuNPs, while IgG incubation inevitably failed to form The concentration of NPs also greatly affects the formation of PC and
corona, resulting in NP aggregation. Moreover, albumin proved to be the the properties of their surfaces. Zhang et al. [79] constructed four bio­
most abundant protein in the hard PC formed by human serum and polymers (poly-(hydroxybutyrate-co-hydroxyhexanoate, PHBHHx) that
AuNPs. This result has also been confirmed by other gold NPs in spite of were composed of the same substance with similar particle sizes
their size and shape [42]. Some studies have found that the most (230 ± 20 nm) but different charges (− 30 to +40 mV), and studied the
abundant protein in certain silicon-based materials is apolipoprotein protein adsorption of the four biopolymers at different concentrations
[68]. Das et al. [69] found that both in the culture media Dulbecco’s (125 μg/mL, 500 μg/mL, 1000 μg/mL) after incubation with 5% FBS.
Modified Eagle Medium (DMEM) and Roswell Park Memorial Institute The results indicated that the concentration of NPs led to a substantial
Medium (RPMI 1640) could find some cell matrix adhesion proteins on alteration in both the protein adsorption capacity and adsorbed protein
AuNRs, but these proteins were missing on silica modified gold nanorods type. NPs with a lower concentration had a higher protein adsorption
(SiO2-AuNRs), which is a lateral reflection of the greater toxicity of the capacity. When the concentration of the four biopolymers was less than
naked AuNRs. In general, Cellular matrix adhesion proteins such as 500 μg/mL, which was incubated in the serum protein at a concentra­
immunoglobulins, complement proteins and other opsonins like plasma tion of 5% FBS, the zeta potential of all NPs-FBS was ~− 20 mV, which
fibrinogen are involved in the recognition and phagocytosis of NPs entry indicated that the number of proteins in 5% FBS were sufficient to cover
into cells. Their preferential adsorption enhances phagocytosis and almost all of the surface of the NPs. In contrast, when the concentration
trigger inflammation, determining NP toxic or non-toxic to cells, and of NPs was increased to 1000 μg/mL, the zeta potential of the four
leading to NP removal from blood [70–72]. Conversely, serum albumin polymers-FBS was altered, which resulted from the combination of the
and apolipoproteins are known to stabilize colloidal NPs and extend associated proteins and uncovered part of the original NPs. This sug­
their blood circulation [73–75]. A kinetic analysis of the PC of solid NPs gested that the number of proteins in 5% FBS was insufficient to
showed that the albumin was initially bound to the NPs and then completely cover the surface of the NPs.
replaced by fibrinogen, after which fibrinogen was replaced by apoli­
poproteins [56,63]. An earlier study came to similar conclusions: when 3.3. Nanoparticle surface properties
proteins with different affinity competing for the available nanoparticle
surface area in a typical biological environment, HSA and fibrinogen 3.3.1. Zeta potential and electrostatic force
may dominate on the particle surface at short times, but will subse­ It has been demonstrated that the zeta potential is a key factor
quently be displaced by lower abundance proteins with higher affinity affecting NP-protein interactions [79]. The zeta potential not only plays
and slower kinetics, for example apolipoprotein A-I [35]. High plasma important roles in stabilizing NPs-based colloidal systems through the
concentrations of apolipoproteins have been shown to be able to replace electrostatic repulsive force, but is also a regulator for the particle size of
fibrinogen within seconds [76]. NPs made of the same material, where the larger the zeta potential, the
larger the electrostatic repulsion, and the smaller the particle size.
3. Critical factors influencing PC Moreover, in terms of protein adsorption, the composite charge of those
adsorbed serum proteins was negative, so that the larger the absolute
3.1. Size of NPs value of the zeta potential of the negatively charged NPs, the greater the
repulsive force and the less protein was absorbed [79]. The larger the
There are many other factors that influence the formation of PC. The absolute value of the zeta potential of the positively charged NPs, the
thickness and density of the PC formed on the surface of the NP are greater the attraction and the more proteins were absorbed. However,
directly related to the size of the NP. Piella et al. [33] studied the Shang et al. [14] proposed that local charge distributions of the binding
interaction of citrate-stabilized AuNPs with proteins in the size at a interfaces mediate electrostatic interactions rather than the net charge
range of 3.5–150 nm, and found that the thickness and density of the PC of the proteins and NPs, and proteins adsorbed onto the NP surfaces in
were strongly dependent on the size of the NPs. The particle size can be certain orientations so as to allow efficient electrostatic interactions
determined by different modes of transition: (i) formation of an between the charged patches on the proteins and the counter-charged
incomplete corona, (ii) formation of a nearly single-density PC layer, surfaces of the NPs.
and (iii) formation of a multilayer corona. At first the authors chose an Electrostatic force is one of the most important forces in the inter­
appropriate exposure condition on the premise of stable NPs exist: for action between NPs and proteins. Kubiak and Mulheran [80] used
the same citrate stabilized 10 nm AuNPs, when exposure experiments atomic molecular dynamics to study the adsorption of chicken protein
performing in serum-free DMEM media, the immediate aggregation of lysozyme (HEWL) on a negatively charged hydrophilic surface, and
NPs can be clearly observed by UV–vis spectroscopy; while in the concluded that the main force controlling the protein adsorption was the
complete cell culture medium (cCCM) composed of DMEM supple­ electrostatic attraction of the protein fractions and surfaces. Ghosh et al.
mented with 10% of fetal bovine serum (FBS), the main driver of [81] proposed the protein-nanoparticle-RCP (reverse charge
nanoparticle aggregation is particle concentration. The presence of parity)-counterion model and demonstrated the electrostatic interaction
proteins is an important factor for the stabilization of NPs in high between negative surface NPs and positive surface HEWL. Additionally,
strength electrolyte CCM. When AuNPs with different sizes were com­ the irreversible denaturation of the protein conformation, owing to the
bined with the CCM, the gold NP-PC, with a particle size ≤ 12 nm, was binding of counterions, was reported. They further used this model,
smaller than the expected single-layer PC, while a single, full dense layer which involved reverse charge parity, to show the selective binding of
was formed on the 12–80 nm AuNPs. The AuNPs with a particle proteins on a charged surface iron oxide nanoparticles (IONPs) [82].
size > 80 nm formed a 2–3 layer multilayer PC, similar to that reported Dyawanapelly et al. [83] studied the interaction of lysozyme with
in flat surfaces [77]. The protein adsorption kinetics showed that the counterion-associated iron oxide nanoparticles (IONPs) using three
smaller the particle size, the faster the kinetic evolution and the thinner different types of NPs, namely, bare IONPs, low molecular weight chi­
(incomplete) the protein layer. Kihara et al. [78] structurally evaluated tosan modified IONPs (LMWC-IONPs) and the counterion
the soft and hard components of the PC formed from polystyrene (PS) (Na+)-associated sodium tripolyphosphate IONPs (STP-LMWC-IONPs).

5
W. Huang et al.
Table 1
Overview of serum/plasma proteins adsorbed on the surface of different types of nanomaterials with varied surface chemistries.
Materials Type Size (TEM) Linker Medium Methods Main proteins brief description Refs

Polystyrene Ch-linPG21 170–280 nm Polyglycerol Plasma LC-MS/


[53]
MS
Ch-linP(G15-co- SDS-
GPE6) PAGE
Ch-linP(G12-co-
GPE9)
Polystyrene Galactose 33 nm,669 nm Galactopyranoside Normal human serum (incubated 1 h, MS Complement protein C3, Apolipoprotein A-I, Apolipoprotein E [101]
Glucose Glucopyranoside 37 ℃) Complement protein C3, Apolipoprotein A-I, Apolipoprotein E
SiO2 SiO2 22.4 ± 2.2 nm Fetal bovine serum LC-MS- Except for albumin, major proteins were apolipoproteins, [91]
MS
SiO2-NH2 –NH2 Alpha-2-HS-glycoprotein,complement C3 and hemoglobin.
SiO2-COO- –COO– PCs of three NPs changed dynamically during 1–48 h.
6

SiO2 UC-NPs 93.1 ± 14.1 nm Carboxylic acid Human plasma (incubated 1 h, 37 ℃) LC-MS/ Apolipoprotein E, Apolipoprotein E chain A, Alpha fibrinogen [94]
MS
HSA-NPs 98.0 ± 10.5 nm HSA pe-coated SDS- Apolipoprotein E, Apolipoprotein E4 chain A, Kininogen-1
PAGE
GG-NPs 101.9 ± 12.8 nm γ-Globulins pe-coated Apolipoprotein B100, Apolipoprotein E, Ig gmma-1 heavy chain
AuNPs BPEI-AuNP 40 nm, 80 nm Branched polyethyleneimine LFQ-LC-MS 40-BPEI-AuNP: albumin, Fibrinogen β chain, Fibrinogen α chain [102]
LA-AuNP Lipoic acid 55% human plasma (incubated SDS-PAGE 80-BPEI-AuNP: albumin, Fibrinogen α chain, Fibrinogen β chain
1 h)
PEG-AuNP Polyethylene 40-LA-AuNP: albumin, Talin-1, Myosin-9
glycol
80-LA-AuNP: albumin, Myosin-9, Talin-1
40/80-PEG-AuNP: albumin, Talin-1, Myosin-9
SPIONs CA-MF 10 nm Citric acid 10%/55% human plasma (incubated SDS- 10% plasma: CA-MF: Apolipoprotein B100, and Fibronectin [103]
1 h, 25 ℃) PAGE

Biomedicine & Pharmacotherapy 139 (2021) 111541


PAA-MF Poly(acrylic acid) MS OAOA-MF: Apolipoprotein A1
OAOA-MF Oleic acid 50% plasma: CA-MF: complement factor proteins and immunoglobulin
(CA and PAA SPIONs showed a very similar protein pattern) OAOA-MF: lipoproteins (Lipoprotein B100, ApoE, and ApoA)
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

The experimental results showed that as a result of the binding of Na+ proteins such as fibronectin, required for cell adhesion. Sulfonate holds
counter ions to proteins, the lysozyme fluorescence spectrum showed a on to albumin more strongly, producing a persistent hard corona likely
quenching effect after HEWL interacted with STP-LMWC-IONP (pH 9.0), to remain biocompatible. The mechanism is thought to be related to the
which indicated HEWL denaturation. The CD spectrum revealed a higher energy of interaction between sulfonate and amine than between
conformational change in the HEWL secondary structure. In addition, a carboxylate and amine. Danner et al. [53] studied polystyrenes with
counterion-induced lysozyme inactivation was confirmed by an enzyme gradual increase of negative charges by modifying polyglycerol (PG),
activity assay involving Micrococcus lysodeikticus. A pH of 9.0 was and finally they concluded that high amount of phosphoric acid group is
observed to be a more favorable condition compared with pH 7.4 not conducive to the stealth effect of nanocarriers. When the phosphate
because of the strongest electrostatic interaction between lysozyme and content is low, clusterin is the dominant protein. As the phosphate
NP. Moreover, selective binding of LMWC-IONPs was observed with content increases, the proportion of clusterin drops to about 20%, which
protein at pH 9.0, which is in agreement with the earlier results using the seems to reach saturation. In addition, the fibrinogen and especially
reverse charge parity model. Lysozyme, as an amyloid-forming model immunoglobulin components were simultaneously increasing. The
protein, is also widely used as a model protein for studying folding and introduction of phosphate not only resulted in a much higher absolute
aggregation mechanisms. This study assumed that counterions in the adsorption capacity of the protein, but also a much lower specificity of
surface coating of NPs can improve protein misfolding or unfolding, and the protein. Fibrinogen and immunoglobulin were enriched with the
also prevent their aggregation, and thus can be considered as a powerful increase of phosphate groups; however, they are known to facilitate
and potential therapeutic strategy for the treatment of incurable uptake of phagocytic cells that remove the nanocarriers from the
neurodegenerative diseases. bloodstream. These proteins identified may also be involved in the ag­
gregation process. Mortensen et al. [91] demonstrated that the PC
3.3.2. Hydrophobicity/hydrophilicity continues to change for over 48 hrs, being heavily influenced by the NP
The main forces that stabilize the structure of proteins are electro­ surface chemistry. They found both native SiO2 NP and SiO2-COO- NP
static interactions, which include hydrophobic, hydrogen bonding, co­ had been identified with significantly higher levels of complement
valent bonds, and weak van der Waals forces that play an important role pathway proteins in the PC. And serum albumin is believed to be a
in stabilizing biopolymers. Shemetov et al. [84] reported that at the dysopsonin, and both SiO2 and SiO2-NH2 NP had the highest count for
atomic level, hydrophilicity is determined by the presence of polar this protein in contrast to SiO2-COO- NP, which is consistent with Del­
functional groups (such as Si2OH and Ti2OH) or under coordinating gado’s conclusion. But it has been reported that apolipoproteins were
metal ions (such as Fe2+ and Zr4+) in the outer surface layer. The less frequently detected for PS-COO- NP than for the native PS NPs,
number of these sites determines the degree of hydrophilicity and hy­ which is in contrast with their study [92].
drophobicity of the NPs. When a protein molecule approaches an NP, the Most functional groups designed for nanoparticles are intended to
interaction of the protein with NP depends on the hydrophilicity and enhance targeting and prolong blood circulation. However, in order to
hydrophobicity of the NP surface. In the case of a hydrophilic (polar) better understand the interaction between the functional groups of
surface, the interaction occurs directly through hydrogen bonds with nanomaterials and proteins, cells and biological environment, we need
structures exposed on the surface of the protein. Whereas when the to enlarge the role of functional groups, and the proteins adsorbed and
surface is hydrophobic, the non-polar portion hidden in the dissolved the PC formed are also functional surfaces. Martens et al. [93] showed
protein molecule may be exposed to the surface without making contact that citrate and dextran are easily displaced by fibrinogen (Fb), which
with water, in which case, the interaction depends only on the degree of upon binding to the surface stabilize the magnetic NPs from aggregation
hydrophobicity of the surface. Studies have shown that proteins adsor­ over time. The PEG coating increased the stability of the magnetic NPs
bed on hydrophobic surfaces have less natural structure than proteins by acting as a barrier for Fb adsorption on the surface of the NPs, and
adsorbed on hydrophilic surfaces, which leads to severe protein dena­ make the Fb conjugates present a much higher binding to the integrin
turation [85]. containing membrane. It’s worth noting that their study provides an
Albumin and IgG tend to preferentially be absorbed by hydrophilic evidence that the coating of NPs with Fb together with other substances
nanomaterials, and the hydrophobic forces played a predominant role e.g., drugs might be applicable to enhance the targeted interaction with
[86]. While hydrophobic nanomaterials tend to preferentially adsorb membrane proteins. Moreover, Mirshafiee et al. [94] used γ-globulins
apolipoproteins, which may result from interactions between the lipid pre-coated SiO2 NPs to recruit opsonins and tested the ability of
binding domains of the apolipoproteins and the hydrophobic surface of phagocytes uptaking them. In the PCs, compared to the corona-UC-NPs
the nanomaterial. But not all apolipoproteins conform to this trend, for (carboxylic acid-functionalized silica NPs), pre-coating with HSA
example, ApoA-IV, ApoB, ApoC-III, and ApoE adsorb preferentially to increased the abundance of lipoproteins and reduced the abundance of
hydrophobic cholesterol or atheronal-B quantum dots, while ApoA-I, coagulation proteins, such as fibrinogen. Pre-coating with γ-globulins
ApoA-II, and ApoC-I adsorb preferentially to more hydrophilic had the opposite effect, reducing the wt% of lipoproteins and drastically
NH2-modified quantum dots [87]. increasing the wt% of complement factors (e.g., complement C3, C4A,
C5, C1s, C8 subunits, C7, factor B, and C6) and immunoglobulins in the
3.3.3. Functional groups corona as compared to UC-NPs. Their experiment proved that the
The material and the functional groups on the surface of the NPs also γ-globulins precoating promotes the recruitment of opsonins, namely
affect the formation of the PC. Park et al. [88] observed that FBS in cell immunoglobulins and complement components, in the plasma to the PC,
culture medium was preferentially adsorbed on aromatic thiol-modified but they did not enhance NP uptake by macrophages. It suggested that
gold NPs relative to thiol-modified gold NPs with amino, carboxyl and other plasma proteins, such as apolipoproteins, covered the active sites
hydroxyl groups. Matczuk et al. [89] showed that the proteination of the on the immunoglobulins in the PC and prevented them from binding to
carboxyl-modified AuNP in a real serum environment occurred without receptors on immune cells.
the participation of albumin, which abolished the PC under equilibrium The surface chemistry of NPs can easily be tuned using different
conditions. In contrast, AuNR with a surface amino group first formed an coating materials such as surfactants or polyelectrolytes. Polyelectrolyte
albumin conjugate, but the albumin in this "soft" corona was gradually coatings can stabilize NP dispersions due to high charge densities and
replaced by other proteins that showed a higher affinity for the aminated can also be used for target, stealth and so on. In general, polyzwitterions
surface. The ease with which albumin can be lost depends on the surface often show anti-fouling properties in terms of reduced unspecific protein
functional group-carboxylate or sulfonate, in particular aromatic sulfo­ adsorption. Typical examples for polyzwitterions showing antifouling
nate. Delgado et al. [90] found that carboxylate permits easier loss of behavior are poly(betaines) like poly(carboxybetaine acrylamide)
albumin, which presumably allows the subsequent adsorption of (pCBMA) [95]. There were exceptions, Gräfe et al. [96] detected

7
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

Table 2 solutions in a wide pH range. It had been found that these bioconjugates
Overview of the degree of HSA conformational change induced by several gold were stable in neutral and alkaline solutions and, at pH lower than the
NPs modified with different functional group at different pH values. protein isoelectric point, aggregation took place.
Ligands PH

3.8 7.4 9.3


3.5. Biological environment
Change
The PC is formed in a biological fluid medium, so the biological fluid
Citrate Larger Larger Unaltered
PEG-OMe Smaller Unaltered Unaltered environment also affects the formation and composition of the PC. The
PEG-NH2 Larger Larger Moderate formation of the PC has been observed in pure protein solution, cell
PEG-COOH Larger Unaltered Larger culture medium, full serum and also human body fluids [58,112–115].
Glycan Larger Unaltered Larger The cell culture media DMEM and RPMI 1640, which mimic the in vivo
biological environment, were found to differ in the formation of protein
increasing relative amounts of albumin (67 kDa) via specific Western caps on the surface of NPs, and these differences also affected the
blot assays on polydehydroalanine-coated multicore iron oxide NPs. And cellular responses. DMEM is more likely to form a large number of
some block copolymer-stabilized soft NPs, even with fairly short hy­ time-dependent, stable and abundant PC, while RPMI shows different
drophilic chains, exhibit prominent protein repellent properties [97]. protein adsorption kinetics and a reduced protein coating [32]. The
There have been many studies demonstrating that polyethylene glycol protein concentration in the biological fluid also affects the formation of
(PEG) can reduce but not eliminate protein adsorption [16,57,98]. the PC, which includes the stability of the NPs [116,117]. Lundqvist
Specially, the better stealth property of the micelles with et al. [114] showed the evolution of the PC as a result of the transfer of
alpha-glutamyl-terminated PEG shells was presumably attributed to the NPs from one biological fluid (plasma) into another (cytosolic fluid), a
zwitterionic property of the alpha-glutamyl groups [99]. The relevant simple illustrative model for the uptake of NPs into cells. Their results
content is further explained in Section 5.1. supported that the “hard” PC formed around NPs evolved and changed
In conclusion, it can be found that the physical and chemical prop­ its identity by integrating new proteins when the protein-particle com­
erties of nanomaterials and their surface modified functional groups or plex was introduced into the second biological solution, which sug­
coatings have a great influence on the formation of PCs (Table 1). Not gested that the final corona contained a “fingerprint” of its history.
only that, the duration of incubation and serum or plasma concentration Therefore the corona composition could potentially depend not only on
also affect the composition of the PC. Most of the time, even with the current environment of the nanoparticle, but also on all environ­
different functional groups, the most abundant protein in the PC formed ments it has moved through [59].
for the same material during a short incubation period or at a low plasma In summary, the formation of PC has the following characteristics:
concentration is different in content but the same kind. The difference formation time, NP concentration, nanoparticle size, surface charge of
mainly exists in some low abundance proteins [100]. With the prolon­ NPs, hydrophilic/hydrophobic properties, and biological fluid
gation of incubation time, the composition of the PC changed obviously, environment.
and the influence of specific functional groups on the PC was more
obvious [91]. 4. Effect of PC on the biological function of NPs

It can be seen from the above that the formed PC is significantly


3.4. pH influenced by the physical and chemical properties of the NP in the
biological fluid such as particle size, morphology, surface charge, sta­
NPs may absorb proteins within a fairly narrow pH depending on bility and biocompatibility. Simultaneously, the corona affects the bio­
their isoelectric point. The closer the pH is to the isoelectric point, the logical effects of the NP and determines the result of the interaction
more protein is absorbed [104,105]. Most serum proteins have an iso­ between NPs and cells. The adsorbed proteins, rather than the NP itself,
electric point between pH 6 and 7, and therefore have a net negative determine the cellular receptors used for binding, the internalization
charge at pH 7.4. After an ion serum protein is adsorbed, it produces a mechanism, the intracellular transport pathway and the subsequent
net negative charge to the NPs [106]. pH can affect the structure of immune response [16].
proteins. For example, the reduction of the α-helical structure of
AuNP-BSA bioconjugates is strongly dependent on pH [107]. Similarly, 4.1. Effect on cells uptake and toxicity
Meesaragandla et al. [108] mentioned that HSA, the most abundant
protein in blood plasma, which is known to undergo pH-dependent It is well known that a series of reactive oxygen species (ROS) are
conformational changes [109]. The conformational changes of second­ involved in aerobic cells during metabolism. Medium and high con­
ary structure of HSA in AuNP-HSA conjugates formed by different li­ centrations of ROS induce apoptosis and even necrosis through cellular
gands coating AuNPs and HSA in different pH solutions were studied by oxidative stress. Choi et al. [118] showed that the interaction of a
circular dichromatography. Compared with the original conformation of positively charged branched polyethylenimine-capped gold nano­
HSA in different pH solutions, the general changes were shown in the particle (BPEI-AuNP) with hepatocytes adversely affected cell uptake
table below (Table 2). The ellipticity of HSA varied with the surface and other cellular responses, but the presence of a PC played a protective
ligand type of NPs and the pH value of the medium. Whether in physi­ role. All naked BPEI-AuNP (40 nm and 80 nm) caused an increase in
ological pH 7.4, or in acidic or alkaline pH, there were always some oxidative stress in hepatocytes over time by sustained ROS/reactive
AuNP-HSA conjugates that showed a decrease in the percentage of nitrogen species (RNS) production, but human plasma (HP) protein
α-helix and an increase in the percentage of β-sheet content. The results significantly reduced ROS, RNS and SO levels in a size-independent
showed that both electrostatic and hydrogen bonding interactions had manner. Additionally, the naked BPEI-AuNP showed the highest
an influence on the protein secondary structure which was further cellular uptake in human cells, but HSA and HP corona reduced uptake.
influenced by the pH of the medium. Moreover, pH influences PC sta­ At cytotoxic levels, naked BPEI-AuNP induces antioxidant and
bility (via salt bridges and hydrogen bonding) and PC protein folding pro-apoptotic genes, but HP reduces other antioxidant genes. The results
[110]. Del Cano et al. [111] focused on the behavior of the bio­ showed that both HSA and HP can reduce the toxicity of BPEI-AuNP,
conjugates made of the protein hemoglobin (Hb) and AuNP capped with although the toxicological characteristics caused by HP are more
three different molecular layers (citrate anions (c), 6-mercaptopurine complicated. Li and Monteiro-Riviere [119] studied the uptake of
(MP) and ω-mercaptoundecanoic acid (MUA)) in aqueous buffered different sizes (40 nm and 80 nm) of gold nanorods by human epidermal

8
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

keratinocytes with lipoic acid, PEG and BPEI surface modifications, and 4.3. Effect on nanoparticle targeting
found that PC reduced uptake. Moreover, in addition to 40-nm lipoic
acid-AuNP, AuNP uptake is energy dependent. Wang et al. [120] The surface of the NPs can modify various active moieties and
demonstrated that when gold nanorod particles entered a functional groups to selectively deliver the drug to a targeted site in the
serum-containing medium or in vivo environment, a stable serum PC specific cells and tissues. Several targeted functional biomolecules have
could effectively prevent direct contact between the gold nanorod and been reported, which include transferrin, insulin, folic acid [123],
the cell membrane, which reduced the damage to the cell membrane, mannose [124], apo A-1 and apo E. PC on the surface of NPs can reduce
thereby effectively reducing the toxicity of the gold nanorods. When the targeting of NPs. For example, the targeting ability of transfer pro­
CTAB/AuNRs (cetyltrimethylammonium bromide-capped gold nano­ teins was impaired when exposed to a biological environment (FBS),
rods) penetrate into the cell membrane, the plasma membrane structure owing to the shielding of plasma proteins in transferrin-conjugated silica
becomes thinner then subsequently is destroyed, followed by an increase NPs (SiO2-PEG8-Tf), which reduced the specificity for transferrin re­
in the number of bubbles and loss of microvilli. For FBS/AuNRs, the ceptors [4]. However, even if the surface of the NPs is coated with a
membrane structure remains intact, and some gold nanorods can be protein layer, it cannot completely cover the surface-modified func­
found around the plasma membrane, while the gold nanorods are tional groups or targeting molecules [17]. Su et al. constructed twenty
mainly located in the bubble-like lysosome. types of cRGD-functionalized PEGylation Au-NPs by a combinatorial
strategy [125]. The surface chemistry of these NPs was diverse, which
4.2. Effect on drug release of drug-loaded NPs included four different PEG lengths and five different targeting ligand
densities. When exposed to a serum-rich medium, a PC was formed on
The drug release profiles of nanocarriers depend on their interaction these NPs. By incubation with targeted cells in a complete or serum-free
with the PC, not only related to the type and amount of the adsorbed medium, the targeting abilities of cRGD were maintained even with a
proteins, but also to the size and type of the nanocarriers. It has been density of 25%. However, the active targeting efficiency of
reported that the PC forms a protein buffer layer on the surface of the protein-bound NPs was remarkably reduced compared with the
NPs in vivo, which shields and blocks drug release [121]. The PC non-protein bound NPs. When macrophages were incubated in complete
significantly affects the nanocarrier drug release profile, which is also or serum-free medium, the PC provided a stealth effect on macrophage
associated with the surrounding medium. Al-Ahmady et al. [122] cell recognition, which is beneficial for the passive targeting caused by
demonstrated that temperature-sensitive liposomes coated with a the EPR effect. Moreover, NPs conjugated with short PEG molecules and
plasma PC release doxorubicin slowly but not completely; but liposomes moderate targeting ligand density might take on a lower macrophage
recovered rapidly release doxorubicin without plasma protein media­ cellular uptake and a higher targeted cell internalization, thus exhibiting
tors. This suggests that the plasma protein medium around the NP-PC a higher targeting efficiency. These surface chemistry results provide
can adsorb some released drugs. guidelines for designing targeted NPs with improved targeting
efficiency.

Fig. 3. Effects of PC on immune system and immunotoxicity. When NPs enter the biological system, they are quickly surrounded by various proteins, some of which
help them to be recognized by phagocytes, which belong to innate immunity. They are then cleared primarily by phagocytic cells in the lungs, liver, and spleen,
producing immunotoxicity.

9
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

4.4. Effect on immune system and immunotoxicity increased or decreased the secretion of specific cytokines.
The amount and identity of proteins adsorbed on the surface of the
NPs as foreign substances entering a living body can induce complex different materials were strongly influenced by surface chemistry and
immune responses, including immunosuppression and immune activa­ wettability, which led to a distinct immune response from macrophages
tion [117]. These responses primarily begin in the innate immune sys­ [132]. It was investigated how the surface chemistry-induced specific PC
tem. PC plays a critical role in making the particles easily recognized by affects the phagocytosis and immune responses of macrophages exposed
the innate immune system, especially by phagocytes, causing their quick to the corona-nanoparticle complexes [133]. The PC alters the inter­
clearance by phagocytic cells located in organs such as the lungs, liver, nalization pathways of gold nanorods by macrophages via the in­
and spleen [126] (Fig. 3). This defines the immunogenicity and immu­ teractions of the predominant coronal proteins with specific receptors
notoxicity of NPs. By using quantitative imaging based on laser ablation on the cell membrane. Serum proteins perturb the recognition of AuNPs
inductively-coupled plasma mass spectrometry (LA-ICP-MS), Elci et al. by scavenger receptors while promote the recognition by clathrin re­
[127] found that neutral AuNPs are more likely to interact with the ceptors in phagocytes. Therefore, clathrin-mediated endocytosis path­
immune system, as evidenced by their greater relative accumulation in ways dominated the internalization by phagocytic cells cultured in
the marginal zone and white pulp regions of the spleen and with the serum-containing medium [134]. The cytokine secretion profile of
Kupffer cells of the liver. Positively-charged NPs accumulated exten­ macrophages is also highly dependent on the adsorption pattern of the
sively in the glomeruli, liver hepatocytes, and red pulp of the spleen. The PC. Therefore, proper design of NPs and control of PC components may
negatively charged NPs accumulated extensively in the red pulp of the help to design drug carriers with prolonged circulation, enhanced
spleen but had a broader distribution in the liver. biocompatibility and reduced toxicity [135].
The complement system is a critical fluid component of innate im­
munity and is the first line of defense against foreign invaders including 5. Opportunities and challenges: NP-PC as a novel nano-drug
nanomaterials, thus the complement-induced the immune response is carrier for targeted applications
the most common reaction during defense immunity. Saha et al. showed
that macrophages uptake NPs by recognizing specific complement pro­ In previous studies, some scientists have been aware of the existence
teins in the PC, and the higher the content of complement component of the PC, and some targeted designs have been made for the hindrance
C4BPA, the greater the uptake of macrophages [128]. The complement and influence of the PC (Fig. 4). These designs could reduce the surface
pathway activation is translated into an exacerbate inflammation adsorption of proteins by NP particles, either to expose targeted mole­
response [129]. It has been demonstrated that different types of gold cules to targeted cells and tissues, or to reduce some unnecessary bio­
nanomaterials (AuNMs), such as spherical gold NPs (AuNP), gold logical reactions, such as complement reactions, or to reduce the
nanostars (AuNS) and gold nanorods (AuNR), can simultaneously acti­ recognition and phagocytosis by phagocytes. These designs are effective
vate the complement system through all three complement pathways and play a significant role in providing enlightenment and guidance for
[130]. In the initial phase, non-specific adsorption of complement pro­ the subsequent application of NPs.
teins on the surface of NPs can determine the subsequent immune Even so, we should know that the adsorption of the PC is inevitable.
cascade and can stimulate or attenuate the immune response, which In recent years, researchers have identified that a PC can redefine and
activates the three complement pathways downstream by recognition improve the targeting of NPs known as "targeted drug corona effects",
and binding of the complement protein. Dai et al. [131] observed the which allows a nanoparticle-protein complex to target specific cell types
differences in proinflammatory cytokine secretion and immune cell in a controlled manner [136,137]. It is more noteworthy that PC has
apoptosis when NPs interacted with immune cells in various biological significant advantages in drug delivery due to their huge surface area
environments, where the PC formed determined whether the NPs and excellent biocompatibility.

Fig. 4. NPs versus PC. NPs attempt to break through the barrier of the PC in different forms and have some potentially beneficial results.

10
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

5.1. Challenge to design protein-free corona 5.2. Protein-cell receptor-mediated targeting

PC is often the prime reason for loss of NP stability, quick clearance, Casals et al. [159] demonstrated the main presence of albumin (BSA)
and potentially harmful immunologic reactions [138], which has been on NPs using mass spectrometry. When NPs were mixed with a
easily considered a “fluid biological barrier” blocking the delivery of NPs BSA-specific antibody, the particle size increased, and this effect was not
to the tumor’s target site [126]. To design protein-free NPs has long been obtained by the action of the control antibody IgG. This indicated that
a challenge for researchers. Although protein adsorption is inevitable, the protein on the surface of the PC maintained a natural epitope. This
the reduction of protein adsorption will maximize the surface chemical finding also suggested that proteins on the surface of the PC can serve as
biological effects of NPs. PEG modification is a classic method for specific design targets. The study by Barran-Berdon et al. [160]
nanoparticle anti-biomolecule adsorption [139,140]. The PC of PEGy­ demonstrated that the positively charged lipid nanocarrier
lated nanocarriers is enriched with certain so-called dysopsonins that DC-Chol-DOPE (the cationic lipid 3β-[N-(N’,N’-dimethylaminoetha­
are responsible for the reduced uptake in immune cells [53,141,142]. In ne)-carbamoyl] combined with the zwitterionic lipid dio­
some cases, PEG coating has a shielding effect, which possibly eliminates leoylphosphatidylethanolamine) was coated with proteins in HP for
the toxicity and immunogenicity of NPs in the body [93]. It has been 1–60 min to form a PC, which was mainly enriched in apolipoprotein
reported in the literature that gold NPs can resist the adsorption of (Apo AI, Apo C-II, Apo D and Apo E). It was demonstrated that
proteins by reducing the affinity for serum proteins through glutathione DC-Chol-DOPE/HP complex enters PC3 cells through receptor-mediated
coating [143] or functionalizing with lysine and cysteine [144]. Deco­ endocytosis. With only weak endocytosis, even PC can inhibit endocy­
rating a branched or cyclic end group on the NP surface typically can tosis mediated by acupoint-like invagination when NPs enter cell
also adsorb less protein [128]. Moyano et al. [145] designed a series of membranes [119]. Fleischer et al. [16] used a set of polystyrene NPs to
zwitterionic NPs with variable hydrophobicity. The zwitterionic func­ adsorb protein of bovine serum albumin, which resulted in the binding
tional group can replace the PEG effect, which produces strong elec­ of two different cell surface receptors: the native albumin receptor and
trostatic binding with water, and makes the zwitterionic system stable the scavenger receptor. The BSA-NP complex formed by anionic NP
and has the effect of a corona-less adsorption. These NPs do not adsorb bound to the native albumin receptor, whereas the BSA-NP complex
proteins on moderate levels of serum proteins and do not form protein formed by the cationic NPs bound to the scavenger receptor. It was also
hard corona at physiological serum concentrations. Klepac et al. [146] demonstrated that the secondary structure of the adsorbed bovine serum
used EPR, ITC, and DLS methods to demonstrate that a hard corona is albumin protein controlled the cellular receptors used by the protein-NP
not formed around the NP of HPMA amphiphilic polymer conjugates. complex. Therefore, PC-cell receptor-mediated cell targeting should
HPMA copolymers provide perfect "invisible" properties to NPs, which monitor the structure of proteins, especially secondary structures.
prevent them from interacting with human blood plasma proteins and,
thus, keeps their functionality unchanged. The authors suggested that all 5.3. NP-PC as a novel nano-drug delivery system
previous publications reporting a "hard corona-soft corona" should now
re-evaluate the types of NPs that had been used to study protein Serum proteins are multifaceted and can be used not only to stabilize
adsorption. Knittel et al. [147] showed that ultrasmall AuNPs encom­ NPs, but also to make hydrophobic drugs or charged molecules soluble
passing a zwitterionic glutathione monoethyl ester surface coating [161–163]. Kah et al. [164] reported that the PC around the NPs has a
(AuGSH(zwt)) were highly resistant to aggregation and serum protein high load capacity like a sponge. It was also found that the ability of the
interactions. And the strep-tagged AuGSH(zwt) particles were not only PC on the surface of cetyltrimethylammonium bromide-capped gold
highly resistant to nonspecific protein interactions but also retained nanorods (CTAB-GNRs) to load small molecules is 5–10 times greater
their targeting capability in biological fluid, displaying efficient binding than covalent binding, either by loading a negatively charged DNA
to streptactin receptors in nearly undiluted serum. Moreover, it was oligonucleotide or a positively charged doxorubicin (DOX). The optical
found that both zwitterion attachment and PEGylation could reduce PC properties of GNR resonance in near-infrared light can remotely control
formation on AuNPs, and zwitterion attachment significantly prevented and assist in the release of the PC payload. Ling Yeo et al. [58] used a
PC formation, even more effectively than PEGylation [148]. In fact, complex of mouse serum and gold nanorods to load and trigger the
zwitterions or some specially coated nanomaterials have proposed new release of the photosensitizer Chlorin e6 (Ce6), which was used to treat
strategies for the modification and application of NPs in biological NCr tumor mice by photodynamic therapy (PDT) and photothermal
fluids, which deserves more attention. Reducing complement activation therapy (PTT). The results showed that the temperature of the tumor
by nanomaterials can also reduce protein adsorption, and recently some increased by 16.85 ◦ C under the irradiation of a local laser for 20 min,
scientists use membrane coatings to camouflage NPs to avoid immune and the tumor regression was completed within 19 days, which indi­
clearance, thus reducing complement activation by NPs [149,150]. cated that the PC could successfully load and transport the drug for in
On the one hand, the reduction of protein adsorption is to avoid the vivo treatment.
function designed by NPs being concealed; on the other hand, it is also
conducive to the reduction of phagocytic cell recognition and clearance. 6. Conclusions and outlook
To design protein-free NPs is an important strategy for NPs to overcome
the clearance of the reticuloendothelial system (RES) or mononuclear Nanoparticle-protein complexes are what biosystems ’see’ and
phagocytic system (MPS) [151,152]. Some scientists have proposed a ’respond to’ [112,165], which determines the fate of NPs in biosystems
method of temporary blocking MPS to reduce consumption of phago­ rather than the bare NPs [59]. This raises new questions for the appli­
cytes to NPs, such as modification of NPs with PEG [153]; grafting NPs cation of NPs from ideal states to complex biological fluid environments
with polyglycerol [154]; injecting specifically modified liposomes to in vitro and in vivo: is a PC good or bad for NP applications? How can
enclose the cell membranes of phagocytes and thus reducing in­ one treat and fully use a NP-PC? The way in which NPs interact with
teractions between phagocytes and subsequently injected NPs [155]; or biological fluids greatly affects the properties and physiological func­
using antibody-mediated red blood cells to promote the absorption of tions of the NPs. In the past, the successful application and failure
phagocytes and make the immune system saturated firstly and then experience of nanomaterials in the biomedical field requires a
injecting NPs [156–158], etc. These measures could improve the rethinking of the impact of the biological environment, and the perfor­
bioavailability of nano particles in the blood, extend biologic half-life of mance of the NPs needs to be re-evaluated. PC formed on the surface of
the drug, and lower toxicity, is an effective strategy to improve the nano different nanomaterials are specific and cannot be generalized. There
drug delivery to target site. are many kinds of proteins in the body of organisms, but they are limited
after all. The commonness and specificity of PC formed by different NPs

11
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

remain to be studied. In the future, scientists should establish a [9] L. Vroman, Effect of adsorbed proteins on the wettability of hydrophilic and
hydrophobic solids, Nature 196 (4853) (1962) 476–477, https://doi.org/
nanoparticle-PC database to better serve the research on the application
10.1038/196476a0.
of NPs in biological systems. [10] X. Kai, K. Huating, L. Xiaoxia, S. Yanhong, H. Qing, Z. Ying, Nanoparticle-protein
There have been several reviews to summarize PC-NP complexes interactions and their biological effects, Sci. Sin. Chim. 45 (7) (2015), https://doi.
from different aspects, including methods, composition, interactions, etc org/10.1360/n032014-00264.
[11] E. Casals, V.F. Puntes, Inorganic nanoparticle biomolecular corona: formation,
[54,55,166,167]. This review redescribes the relevant content and evolution and biological impact, Nanomedicine 7 (12) (2012) 1917–1930,
highlights the influence of PCs on the immune system and phagocytes. In https://doi.org/10.2217/nnm.12.169.
particular, the strategies for NPs to break through the barrier of PC and [12] I. Capjak, S.S. Goreta, D.D. Jurasin, I.V. Vrcek, How protein coronas determine
the fate of engineered nanoparticles in biological environment, Arh. Hig. Rada
the targeting application of PC-NP complexes as drug carriers are pro­ Toksiko 68 (4) (2017) 245–253, https://doi.org/10.1515/aiht-2017-68-3054.
spected. The PC is a double-edged sword, seen as a biological barrier for [13] Q. Saquib, Cellular and Molecular Toxicology of Nanoparticles, Springer Berlin
NPs, and could also play a key role in drug delivery. Weighing the pros Heidelberg, New York, NY, 2018.
[14] L. Shang, G.U. Nienhaus, In situ characterization of protein adsorption onto
and cons and designing experiments properly is the key. When NPs are nanoparticles by fluorescence correlation spectroscopy, Acc. Chem. Res. 50 (2)
designed as a drug delivery system, they should be viewed not only from (2017) 387–395, https://doi.org/10.1021/acs.accounts.6b00579.
a chemical and engineering standpoint, but also from a biological [15] M. Rahman, S. Laurent, N. Tawil, H. Yahia, M. Mahmoudi, Protein-Nanoparticle
Interactions: the Bio-nano Interface, Springer, Heidelberg; New York, 2013.
standpoint. The whole protein-covered nanoparticle system should be [16] C.C. Fleischer, C.K. Payne, Secondary structure of corona proteins determines the
viewed and studied as a biological delivery system, which requires us to cell surface receptors used by nanoparticles, J. Phys. Chem. B 118 (49) (2014)
more accurately simulate the biological microenvironment. The com­ 14017–14026, https://doi.org/10.1021/jp502624n.
[17] B. Kharazian, N.L. Hadipour, M.R. Ejtehadi, Understanding the nanoparticle-
bination of data from biological experiments and computer analysis to
protein corona complexes using computational and experimental methods, Int J.
construct experimental frameworks will be one of the ways to build safe Biochem. Cell B 75 (2016) 162–174, https://doi.org/10.1016/j.
and efficient nano-drug delivery systems in the future [168,169]. biocel.2016.02.008.
The development of new technologies and new instruments will [18] J.M. Howe, H. Mori, Z.L. Wang, In situ high-resolution transmission electron
microscopy in the study of nanomaterials and properties, MRS Bull. 33 (2) (2008)
gradually unveil the mechanism of the interaction between PC and NPs. 115–121, https://doi.org/DOI 10.1557/mrs2008.24.
Understanding the principles and influencing factors of the interaction [19] R.M. Imamov, V.V. Klechkovskaya, E.I. Suvorova, High-resolution transmission
between PC and NPs facilitates the design of targeted nanomaterials, electron microscopy for crystallographic study of nanomaterials, Crystallogr. Rep.
56 (4) (2011) 650–661, https://doi.org/10.1134/S1063774511040079.
which will make the PC controllable and functionalized, to construct [20] L. Wang, J. Li, J. Pan, X. Jiang, Y. Ji, Y. Li, Y. Qu, Y. Zhao, X. Wu, C. Chen,
new drug delivery systems. These systems would be beneficial to Revealing the binding structure of the protein corona on gold nanorods using
improve the biotoxicity and immunotoxicity, improve the targeting and synchrotron radiation-based techniques: understanding the reduced damage in
cell membranes, J. Am. Chem. Soc. 135 (46) (2013) 17359–17368, https://doi.
effectiveness, prolong the action time in vivo and improve the clearance org/10.1021/ja406924v.
rate, so as to realize the accurate and safe use of nanomedicine. [21] J. Watt, D.L. Huber, P.L. Stewart, Soft matter and nanomaterials characterization
by cryogenic transmission electron microscopy, MRS Bull. 44 (12) (2019)
942–948, https://doi.org/10.1557/mrs.2019.285.
[22] M. Kokkinopoulou, J. Simon, K. Landfester, V. Mailander, I. Lieberwirth,
Declaration of conflicting interests Visualization of the protein corona: towards a biomolecular understanding of
nanoparticle-cell-interactions, Nanoscale 9 (25) (2017) 8858–8870, https://doi.
org/10.1039/c7nr02977b.
The authors declare no conflict of interest.
[23] L.S. Franqui, M.A. De Farias, R.V. Portugal, C.A.R. Costa, R.R. Domingues, A.
G. Souza Filho, V.R. Coluci, A.F.P. Leme, D.S.T. Martinez, Interaction of graphene
Acknowledgments oxide with cell culture medium: evaluating the fetal bovine serum protein corona
formation towards in vitro nanotoxicity assessment and nanobiointeractions,
Mater. Sci. Eng. C Mater. Biol. Appl. 100 (2019) 363–377, https://doi.org/
The current study was supported by grants from the National Natural 10.1016/j.msec.2019.02.066.
Science Foundation of China (Grant Nos. 81803064, 81673009). [24] J. Schaefer, C. Schulze, E.E. Marxer, U.F. Schaefer, W. Wohlleben, U. Bakowsky,
C.M. Lehr, Atomic force microscopy and analytical ultracentrifugation for
probing nanomaterial protein interactions, ACS Nano 6 (6) (2012) 4603–4614,
References https://doi.org/10.1021/nn202657q.
[25] A.R. Karow, J. Gotzl, P. Garidel, Resolving power of dynamic light scattering for
protein and polystyrene nanoparticles, Pharm. Dev. Technol. 20 (1) (2015)
[1] K. Bourzac, News feature: cancer nanomedicine, reengineered, Proc. Natl. Acad.
84–89, https://doi.org/10.3109/10837450.2014.910808.
Sci. U. S. A. 113 (45) (2016) 12600–12603, https://doi.org/10.1073/
[26] T.Y. Zheng, P. Cherubin, L. Cilenti, K. Teter, Q. Huo, A simple and fast method to
pnas.1616895113.
study the hydrodynamic size difference of protein disulfide isomerase in oxidized
[2] S. Wilhelm, A.J. Tavares, Q. Dai, S. Ohta, J. Audet, H.F. Dvorak, W.C.W. Chan,
and reduced form using gold nanoparticles and dynamic light scattering, Analyst
Analysis of nanoparticle delivery to tumours, Nat. Rev. Mater. 1 (5) (2016)
141 (3) (2016) 934–938, https://doi.org/10.1039/c5an02248g.
16014, https://doi.org/10.1038/natrevmats.2016.14.
[27] P.M. Kelly, C. Aberg, E. Polo, A. O’Connell, J. Cookman, J. Fallon, Z. Krpetic, K.
[3] C. Corbo, R. Molinaro, A. Parodi, N.E.T. Furman, F. Salvatore, E. Tasciotti, The
A. Dawson, Mapping protein binding sites on the biomolecular corona of
impact of nanoparticle protein corona on cytotoxicity, immunotoxicity and target
nanoparticles, Nat. Nanotechnol. 10 (5) (2015) 472–479, https://doi.org/
drug delivery, Nanomedicine 11 (1) (2016) 81–100, https://doi.org/10.2217/
10.1038/nnano.2015.47.
nnm.15.188.
[28] A.M. Davidson, M. Brust, D.L. Cooper, M. Volk, Sensitive analysis of protein
[4] A. Salvati, A.S. Pitek, M.P. Monopoli, K. Prapainop, F.B. Bombelli, D.R. Hristov, P.
adsorption to colloidal gold by differential centrifugal sedimentation, Anal.
M. Kelly, C. Aberg, E. Mahon, K.A. Dawson, Transferrin-functionalized
Chem. 89 (12) (2017) 6807–6814, https://doi.org/10.1021/acs.
nanoparticles lose their targeting capabilities when a biomolecule corona adsorbs
analchem.7b01229.
on the surface, Nat. Nanotechnol. 8 (2) (2013) 137–143, https://doi.org/
[29] J. Mukherjee, M.N. Gupta, Protein aggregates: forms, functions and applications,
10.1038/Nnano.2012.237.
Int. J. Biol. Macromol. 97 (2017) 778–789, https://doi.org/10.1016/j.
[5] H. Nejadnik, S.M. Taghavi-Garmestani, S.J. Madsen, K. Li, S. Zanganeh, P. Yang,
ijbiomac.2016.11.014.
M. Mahmoudi, H.E. Daldrup-Link, The protein corona around nanoparticles
[30] F. Spinozzi, G. Ceccone, P. Moretti, G. Campanella, C. Ferrero, S. Combet,
facilitates stem cell labeling for clinical MR imaging, Radiology 286 (3) (2018)
I. Ojeajimenez, P. Ghigna, Structural and thermodynamic properties of
938–947, https://doi.org/10.1148/radiol.2017170130.
nanoparticle protein complexes: a combined SAXS and SANS study, Langmuir 33
[6] N.V. Konduru, R.M. Molina, A. Swami, F. Damiani, G. Pyrgiotakis, P. Lin,
(9) (2017) 2248–2256, https://doi.org/10.1021/acs.langmuir.6b04072.
P. Andreozzi, T.C. Donaghey, P. Demokritou, S. Krol, W. Kreyling, J.D. Brain,
[31] J. Perez, A. Koutsioubas, Memprot: a program to model the detergent corona
Protein corona: implications for nanoparticle interactions with pulmonary cells,
around a membrane protein based on SEC-SAXS data, Acta Crystallogr. D Biol.
Part Fibre Toxicol. 14 (1) (2017) 42, https://doi.org/10.1186/s12989-017-0223-
Crystallogr. 71 (Pt 1) (2015) 86–93, https://doi.org/10.1107/
3.
S1399004714016678.
[7] I. Ardao, J. Comenge, M.D. Benaiges, G. Alvaro, V.F. Puntes, Rational
[32] G. Maiorano, S. Sabella, B. Sorce, V. Brunetti, M.A. Malvindi, R. Cingolani, P.
nanoconjugation improves biocatalytic performance of enzymes: aldol addition
P. Pompa, Effects of cell culture media on the dynamic formation of protein-
catalyzed by immobilized rhamnulose-1-phosphate aldolase, Langmuir 28 (15)
nanoparticle complexes and influence on the cellular response, ACS Nano 4 (12)
(2012) 6461–6467, https://doi.org/10.1021/la3003993.
(2010) 7481–7491, https://doi.org/10.1021/nn101557e.
[8] L. Vroman, Finding seconds count after contact with blood (and that is all I did),
[33] J. Piella, N.G. Bastus, V. Puntes, Size-dependent protein-nanoparticle interactions
Colloids Surf. B Biointerfaces 62 (1) (2008) 1–4, https://doi.org/10.1016/j.
in citrate-stabilized gold nanoparticles: the emergence of the protein corona,
colsurfb.2007.11.017.

12
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

Bioconjug. Chem. 28 (1) (2017) 88–97, https://doi.org/10.1021/acs. [55] Y. Li, J.S. Lee, Insights into characterization methods and biomedical applications
bioconjchem.6b00575. of nanoparticle-protein corona, Materials 13 (14) (2020) 3093, https://doi.org/
[34] S. Dominguez-Medina, L. Kisley, L.J. Tauzin, A. Hoggard, B. Shuang, A. 10.3390/ma13143093.
S. Indrasekara, S. Chen, L.Y. Wang, P.J. Derry, A. Liopo, E.R. Zubarev, C. [56] P. Breznica, R. Koliqi, A. Daka, A review of the current understanding of
F. Landes, S. Link, Adsorption and unfolding of a single protein triggers nanoparticles protein corona composition, Med. Pharm. Rep. 93 (4) (2020)
nanoparticle aggregation, ACS Nano 10 (2) (2016) 2103–2112, https://doi.org/ 342–350, https://doi.org/10.15386/mpr-1756.
10.1021/acsnano.5b06439. [57] C.D. Walkey, W.C. Chan, Understanding and controlling the interaction of
[35] T. Cedervall, I. Lynch, S. Lindman, T. Berggard, E. Thulin, H. Nilsson, K. nanomaterials with proteins in a physiological environment, Chem. Soc. Rev. 41
A. Dawson, S. Linse, Understanding the nanoparticle-protein corona using (7) (2012) 2780–2799, https://doi.org/10.1039/c1cs15233e.
methods to quantify exchange rates and affinities of proteins for nanoparticles, [58] J. Martel, D. Young, A. Young, C.Y. Wu, C.D. Chen, J.S. Yu, J.D. Young,
Proc. Natl. Acad. Sci. U. S. A. 104 (7) (2007) 2050–2055, https://doi.org/ Comprehensive proteomic analysis of mineral nanoparticles derived from human
10.1073/pnas.0608582104. body fluids and analyzed by liquid chromatography-tandem mass spectrometry,
[36] K. Aslan, J.R. Lakowicz, C.D. Geddes, Plasmon light scattering in biology and Anal. Biochem. 418 (1) (2011) 111–125, https://doi.org/10.1016/j.
medicine: new sensing approaches, visions and perspectives, Curr. Opin. Chem. ab.2011.06.018.
Biol. 9 (5) (2005) 538–544, https://doi.org/10.1016/j.cbpa.2005.08.021. [59] M.P. Monopoli, C. Aberg, A. Salvati, K.A. Dawson, Biomolecular coronas provide
[37] T. Winuprasith, S. Chantarak, M. Suphantharika, L. He, D.J. McClements, the biological identity of nanosized materials, Nat. Nanotechnol. 7 (12) (2012)
Alterations in nanoparticle protein corona by biological surfactants: impact of 779–786, https://doi.org/10.1038/nnano.2012.207.
bile salts on beta-lactoglobulin-coated gold nanoparticles, J. Colloid Interface Sci. [60] M.A. Dobrovolskaia, A.K. Patri, J.W. Zheng, J.D. Clogston, N. Ayub, P. Aggarwal,
426 (2014) 333–340, https://doi.org/10.1016/j.jcis.2014.04.018. B.W. Neun, J.B. Hall, S.E. McNeil, Interaction of colloidal gold nanoparticles with
[38] I. Kuschnerus, M. Lau, K. Giri, N. Bedford, J. Biazik, J. Ruan, A. Garcia-Bennett, human blood: effects on particle size and analysis of plasma protein binding
Effect of a protein corona on the fibrinogen induced cellular oxidative stress of profiles, Nanomed. Nanotechnol. 5 (2) (2009) 106–117, https://doi.org/
gold nanoparticles, Nanoscale 12 (10) (2020) 5898–5905, https://doi.org/ 10.1016/j.nano.2008.08.001.
10.1039/d0nr00371a. [61] S. Tenzer, D. Docter, S. Rosfa, A. Wlodarski, J. Kuharev, A. Rekik, S.K. Knauer,
[39] M. Martinez-Moro, D. Di Silvio, S.E. Moya, Fluorescence correlation spectroscopy C. Bantz, T. Nawroth, C. Bier, J. Sirirattanapan, W. Mann, L. Treuel, R. Zellner,
as a tool for the study of the intracellular dynamics and biological fate of protein M. Maskos, H. Schild, R.H. Stauber, Nanoparticle size is a critical
corona, Biophys. Chem. 253 (2019), 106218, https://doi.org/10.1016/j. physicochemical determinant of the human blood plasma corona: a
bpc.2019.106218. comprehensive quantitative proteomic analysis, ACS Nano 5 (9) (2011)
[40] I.A. Mudunkotuwa, A.A. Minshid, V.H. Grassian, ATR-FTIR spectroscopy as a tool 7155–7167, https://doi.org/10.1021/nn201950e.
to probe surface adsorption on nanoparticles at the liquid-solid interface in [62] M. Hadjidemetriou, S. McAdam, G. Garner, C. Thackeray, D. Knight, D. Smith,
environmentally and biologically relevant media, Analyst 139 (5) (2014) Z. Al-Ahmady, M. Mazza, J. Rogan, A. Clamp, K. Kostarelos, The human in vivo
870–881, https://doi.org/10.1039/c3an01684f. biomolecule corona onto PEGylated liposomes: a proof-of-concept clinical study,
[41] M.M. Yallapu, N. Chauhan, S.F. Othman, V. Khalilzad-Sharghi, M.C. Ebeling, Adv. Mater. 31 (4) (2019), 1803335, https://doi.org/10.1002/adma.201803335.
S. Khan, M. Jaggi, S.C. Chauhan, Implications of protein corona on physico- [63] M. Vitali, E. Casals, F. Canals, N. Colome, V. Puntes, Simple spectroscopic
chemical and biological properties of magnetic nanoparticles, Biomaterials 46 determination of the hard protein corona composition in AuNPs: albumin at 75,
(2015) 1–12, https://doi.org/10.1016/j.biomaterials.2014.12.045. Nanoscale 12 (29) (2020) 15832–15844, https://doi.org/10.1039/d0nr02379e.
[42] R. Garcia-Alvarez, M. Hadjidemetriou, A. Sanchez-Iglesias, L.M. Liz-Marzan, [64] M. Lundqvist, J. Stigler, G. Elia, I. Lynch, T. Cedervall, K.A. Dawson, Nanoparticle
K. Kostarelos, In vivo formation of protein corona on gold nanoparticles. The size and surface properties determine the protein corona with possible
effect of their size and shape, Nanoscale 10 (3) (2018) 1256–1264, https://doi. implications for biological impacts, Proc. Natl. Acad. Sci. U. S. A. 105 (38) (2008)
org/10.1039/c7nr08322j. 14265–14270, https://doi.org/10.1073/pnas.0805135105.
[43] L.U. Khan, R. Petry, A.J. Paula, M. Knobel, D.S.T. Martinez, Protein corona [65] L. Marichal, G. Klein, J. Armengaud, Y. Boulard, S. Chedin, J. Labarre, S. Pin, J.
formation on magnetic nanoparticles conjugated with luminescent Europium P. Renault, J.C. Aude, Protein corona composition of silica nanoparticles in
complexes, ChemNanoMat 4 (12) (2018) 1202–1208, https://doi.org/10.1002/ complex media: nanoparticle size does not matter, Nanomaterials 10 (2) (2020)
cnma.201800358. 240, https://doi.org/10.3390/nano10020240.
[44] E. Papa, J.P. Doucet, A. Sangion, A. Doucet-Panaye, Investigation of the influence [66] U. Martens, D. Bottcher, D. Talbot, U. Bornscheuer, A. Abou-Hassan, M. Delcea,
of protein corona composition on gold nanoparticle bioactivity using machine Maghemite nanoparticles stabilize the protein corona formed with transferrin
learning approaches, SAR QSAR Environ. Res. 27 (7) (2016) 521–538, https:// presenting different iron-saturation levels, Nanoscale 11 (34) (2019)
doi.org/10.1080/1062936x.2016.1197310. 16063–16070, https://doi.org/10.1039/c9nr04967c.
[45] R. Huang, R.P. Carney, F. Stellacci, B.L. Lau, Protein-nanoparticle interactions: [67] K. Merrell, K. Southwick, S.W. Graves, M.S. Esplin, N.E. Lewis, C.D. Thulin,
the effects of surface compositional and structural heterogeneity are scale Analysis of low-abundance, low-molecular-weight serum proteins using mass
dependent, Nanoscale 5 (15) (2013) 6928–6935, https://doi.org/10.1039/ spectrometry, J. Biomol. Tech. 15 (4) (2004) 238–248.
c3nr02117c. [68] F.S.M. Tekie, M. Hajiramezanali, P. Geramifar, M. Raoufi, R. Dinarvand,
[46] S. Qu, F. Sun, Z. Qiao, J. Li, L. Shang, In situ investigation on the protein corona M. Soleimani, F. Atyabi, Controlling evolution of protein corona: a prosperous
formation of quantum dots by using fluorescence resonance energy transfer, approach to improve chitosan-based nanoparticle biodistribution and half-life,
Small 16 (21) (2020), 1907633, https://doi.org/10.1002/smll.201907633. Sci. Rep. 10 (1) (2020), https://doi.org/10.1038/s41598-020-66572-y.
[47] G.P. Szekeres, M. Montes-Bayon, J. Bettmer, J. Kneipp, Fragmentation of proteins [69] M. Das, D.K. Yi, S.S. An, Analyses of protein corona on bare and silica-coated gold
in the corona of gold nanoparticles as observed in live cell surface-enhanced nanorods against four mammalian cells, Int. J. Nanomed. 10 (2015) 1521–1545,
raman scattering, Anal. Chem. 92 (12) (2020) 8553–8560, https://doi.org/ https://doi.org/10.2147/IJN.S76187.
10.1021/acs.analchem.0c01404. [70] Z.J. Deng, M. Liang, M. Monteiro, I. Toth, R.F. Minchin, Nanoparticle-induced
[48] H.R. Wei, W.N. Leng, J. Song, C. Liu, M.R. Willner, Q.S. Huang, W. Zhou, P. unfolding of fibrinogen promotes Mac-1 receptor activation and inflammation,
J. Vikesland, Real-time monitoring of ligand exchange kinetics on gold Nat. Nanotechnol. 6 (1) (2011) 39–44, https://doi.org/10.1038/
nanoparticle surfaces enabled by hot spot-normalized surface-enhanced Raman nnano.2010.250.
scattering, Environ. Sci. Technol. 53 (2) (2019) 575–585, https://doi.org/ [71] D.E. Owens 3rd, N.A. Peppas, Opsonization, biodistribution, and
10.1021/acs.est.8b03144. pharmacokinetics of polymeric nanoparticles, Int. J. Pharm. 307 (1) (2006)
[49] G.P. Szekeres, J. Kneipp, Different binding sites of serum albumins in the protein 93–102, https://doi.org/10.1016/j.ijpharm.2005.10.010.
corona of gold nanoparticles, Analyst 143 (24) (2018) 6061–6068, https://doi. [72] R. Tavano, L. Gabrielli, E. Lubian, C. Fedeli, S. Visentin, P. Polverino De Laureto,
org/10.1039/c8an01321g. G. Arrigoni, A. Geffner-Smith, F. Chen, D. Simberg, G. Morgese, E.M. Benetti,
[50] M. Circu, T. Radu, A.S. Porav, R. Turcu, Surface functionalization of Fe3O4@SiO2 L. Wu, S.M. Moghimi, F. Mancin, E. Papini, C1q-mediated complement activation
core-shell nanoparticles with vinylimidazole-rare earth complexes: synthesis, and C3 opsonization trigger recognition of stealth poly(2-methyl-2-oxazoline)-
physico-chemical properties and protein interaction effects, Appl. Surf. Sci. 453 coated silica nanoparticles by human phagocytes, ACS Nano 12 (6) (2018)
(2018) 457–463, https://doi.org/10.1016/j.apsusc.2018.05.096. 5834–5847, https://doi.org/10.1021/acsnano.8b01806.
[51] Z. Ban, P. Yuan, F.B. Yu, T. Peng, Q.X. Zhou, X.G. Hu, Machine learning predicts [73] M.A. Wells, A. Abid, I.M. Kennedy, A.I. Barakat, Serum proteins prevent
the functional composition of the protein corona and the cellular recognition of aggregation of Fe2O3 and ZnO nanoparticles, Nanotoxicology 6 (2012) 837–846,
nanoparticles, Proc. Natl. Acad. Sci. U. S. A. 117 (19) (2020) 10492–10499, https://doi.org/10.3109/17435390.2011.625131.
https://doi.org/10.1073/pnas.1919755117. [74] C.B. Anders, J.J. Chess, D.G. Wingett, A. Punnoose, Serum proteins enhance
[52] M. Carril, D. Padro, P. Del Pino, C. Carrillo-Carrion, M. Gallego, W.J. Parak, In dispersion stability and influence the cytotoxicity and dosimetry of ZnO
situ detection of the protein corona in complex environments, Nat. Commun. 8 nanoparticles in suspension and adherent cancer cell models, Nanoscale Res. Lett.
(1) (2017) 1542, https://doi.org/10.1038/s41467-017-01826-4. 10 (1) (2015) 448, https://doi.org/10.1186/s11671-015-1158-y.
[53] A.K. Danner, S. Schottler, E. Alexandrino, S. Hammer, K. Landfester, [75] D. Chen, S. Ganesh, W. Wang, M. Amiji, Protein corona-enabled systemic delivery
V. Mailander, S. Morsbach, H. Frey, F.R. Wurm, Phosphonylation controls the and targeting of nanoparticles, AAPS J. 22 (4) (2020) 83, https://doi.org/
protein corona of multifunctional polyglycerol-modified nanocarriers, Macromol. 10.1208/s12248-020-00464-x.
Biosci. 19 (5) (2019), 1800468, https://doi.org/10.1002/mabi.201800468. [76] L. Treuel, D. Docter, M. Maskos, R.H. Stauber, Protein corona - from molecular
[54] C. Carrillo-Carrion, M. Carril, W.J. Parak, Techniques for the experimental adsorption to physiological complexity, Beilstein J. Nanotechnol. 6 (2015)
investigation of the protein corona, Curr. Opin. Biotechnol. 46 (2017) 106–113, 857–873, https://doi.org/10.3762/bjnano.6.88.
https://doi.org/10.1016/j.copbio.2017.02.009. [77] S. Goy-Lopez, J. Juarez, M. Alatorre-Meda, E. Casals, V.F. Puntes, P. Taboada,
V. Mosquera, Physicochemical characteristics of protein-NP bioconjugates: the
role of particle curvature and solution conditions on human serum albumin

13
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

conformation and fibrillogenesis inhibition, Langmuir 28 (24) (2012) 9113–9126, Colloid Surf. B 18 (3–4) (2000) 301–313, https://doi.org/10.1016/S0927-7765
https://doi.org/10.1021/la300402w. (99)00156-3.
[78] S. Kihara, N.J. van der Heijden, C.K. Seal, J.P. Mata, A.E. Whitten, I. Koper, D. [99] X.J. Wang, C.P. Yang, C.H. Wang, L.J. Quo, T.H. Zhang, Z.Q. Zhang, H.S. Yan, K.
J. McGillivray, Soft and hard interactions between polystyrene nanoplastics and L. Liu, Polymeric micelles with alpha-glutamyl-terminated PEG shells show low
human serum albumin protein corona, Bioconjug. Chem. 30 (2019) 1067–1076, non-specific protein adsorption and a prolonged in vivo circulation time, Mat. Sci.
https://doi.org/10.1021/acs.bioconjchem.9b00015. Eng. C Mater. 59 (2016) 766–772, https://doi.org/10.1016/j.msec.2015.10.084.
[79] T.X. Zhang, G.Y. Zhu, B.Y. Lu, C.L. Zhang, Q. Peng, Concentration-dependent [100] U. Sakulkhu, M. Mahmoudi, L. Maurizi, J. Salaklang, H. Hofmann, Protein corona
protein adsorption at the nano-bio interfaces of polymeric nanoparticles and composition of superparamagnetic iron oxide nanoparticles with various physico-
serum proteins, Nanomedicine 12 (22) (2017) 2757–2769, https://doi.org/ chemical properties and coatings, Sci. Rep. 4 (2014) 5020, https://doi.org/
10.2217/nnm-2017-0238. 10.1038/srep05020.
[80] K. Kubiak, P.A. Mulheran, Molecular dynamics simulations of hen egg white [101] K. Yu, B.F. Lai, J.H. Foley, M.J. Krisinger, E.M. Conway, J.N. Kizhakkedathu,
lysozyme adsorption at a charged solid surface, J. Phys. Chem. B 113 (36) (2009) Modulation of complement activation and amplification on nanoparticle surfaces
12189–12200, https://doi.org/10.1021/jp901521x. by glycopolymer conformation and chemistry, ACS Nano 8 (8) (2014)
[81] G. Ghosh, L. Panicker, R.S. Ningthoujam, K.C. Barick, R. Tewari, Counter ion 7687–7703, https://doi.org/10.1021/nn504186b.
induced irreversible denaturation of hen egg white lysozyme upon electrostatic [102] P. Chandran, J.E. Riviere, N.A. Monteiro-Riviere, Surface chemistry of gold
interaction with iron oxide nanoparticles: a predicted model, Colloids Surf. B nanoparticles determines the biocorona composition impacting cellular uptake,
Biointerfaces 103 (2013) 267–274, https://doi.org/10.1016/j. toxicity and gene expression profiles in human endothelial cells, Nanotoxicology
colsurfb.2012.10.034. 11 (4) (2017) 507–519, https://doi.org/10.1080/17435390.2017.1314036.
[82] G. Ghosh, L. Panicker, K.C. Barick, Selective binding of proteins on functional [103] A. Jedlovszky-Hajdu, F.B. Bombelli, M.P. Monopoli, E. Tombacz, K.A. Dawson,
nanoparticles via reverse charge parity model: anin vitrostudy, Mater. Res. Surface coatings shape the protein corona of SPIONs with relevance to their
Express 1 (1) (2014), 015017, https://doi.org/10.1088/2053-1591/1/1/015017. application in vivo, Langmuir 28 (42) (2012) 14983–14991, https://doi.org/
[83] S. Dyawanapelly, P. Mehrotra, G. Ghosh, D.D. Jagtap, P. Dandekar, R. Jain, How 10.1021/la302446h.
the surface functionalized nanoparticles affect conformation and activity of [104] C.H. Yu, A. Al-Saadi, S.J. Shih, L. Qiu, K.Y. Tam, S.C. Tsang, Immobilization of
proteins: exploring through protein-nanoparticle interactions, Bioorg. Chem. 82 BSA on silica-coated magnetic iron oxide nanoparticle, J. Phys. Chem. C 113 (2)
(2019) 17–25, https://doi.org/10.1016/j.bioorg.2018.09.020. (2009) 537–543, https://doi.org/10.1021/jp809662a.
[84] A.A. Shemetov, I. Nabiev, A. Sukhanova, Molecular interaction of proteins and [105] D. Sternik, P. Staszczuk, J. Pekalska, G. Grodzicka, B. Gawdzik, J. Osypiuk-
peptides with nanoparticles, ACS Nano 6 (6) (2012) 4585–4602, https://doi.org/ Tomasik, P. Witer, Surface properties of silica gel samples modified by selected
10.1021/nn300415x. proteins, J. Therm. Anal. Calorim. 86 (1) (2006) 85–91, https://doi.org/10.1007/
[85] M. Mahmoudi, I. Lynch, M.R. Ejtehadi, M.P. Monopoli, F.B. Bombelli, S. Laurent, s10973-006-7581-7.
Protein-nanoparticle interactions: opportunities and challenges, Chem. Rev. 111 [106] C.D. Walkey, J.B. Olsen, F. Song, R. Liu, H. Guo, D.W. Olsen, Y. Cohen, A. Emili,
(9) (2011) 5610–5637, https://doi.org/10.1021/cr100440g. W.C. Chan, Protein corona fingerprinting predicts the cellular interaction of gold
[86] M.M. Yin, P. Dong, W.Q. Chen, S.P. Xu, L.Y. Yang, F.L. Jiang, Y. Liu, and silver nanoparticles, ACS Nano 8 (3) (2014) 2439–2455, https://doi.org/
Thermodynamics and mechanisms of the interactions between ultrasmall 10.1021/nn406018q.
fluorescent gold nanoclusters and human serum albumin, gamma-globulins, and [107] Y. Kim, S.M. Ko, J.M. Nam, Protein-nanoparticle interaction-induced changes in
transferrin: a spectroscopic approach, Langmuir 33 (21) (2017) 5108–5116, protein structure and aggregation, Chem. Asian J. 11 (13) (2016) 1869–1877,
https://doi.org/10.1021/acs.langmuir.7b00196. https://doi.org/10.1002/asia.201600236.
[87] K. Prapainop, P. Wentworth Jr., A shotgun proteomic study of the protein corona [108] B. Meesaragandla, I. Garcia, D. Biedenweg, J. Toro-Mendoza, I. Coluzza, L.M. Liz-
associated with cholesterol and atheronal-B surface-modified quantum dots, Eur. Marzan, M. Delcea, H-bonding-mediated binding and charge reorganization of
J. Pharm. Biopharm. 77 (3) (2011) 353–359, https://doi.org/10.1016/j. proteins on gold nanoparticles, Phys. Chem. Chem. Phys. 22 (8) (2020)
ejpb.2010.12.026. 4490–4500, https://doi.org/10.1039/c9cp06371d.
[88] J. Park, J.H. Park, K.S. Ock, E.O. Ganbold, N.W. Song, K. Cho, S.Y. Lee, S.W. Joo, [109] K. Baler, O.A. Martin, M.A. Carignano, G.A. Ameer, J.A. Vila, I. Szleifer,
Preferential adsorption of fetal bovine serum on bare and aromatic thiol- Electrostatic unfolding and interactions of albumin driven by pH changes: a
functionalized gold surfaces in cell culture media, J. Colloid Interface Sci. 363 (1) molecular dynamics study, J. Phys. Chem. B 118 (4) (2014) 921–930, https://doi.
(2011) 105–113, https://doi.org/10.1016/j.jcis.2011.07.006. org/10.1021/jp409936v.
[89] M. Matczuk, J. Legat, F. Scaletti, L. Messori, A.R. Timerbaev, M. Jarosz, The fate [110] M. Raoufi, M.J. Hajipour, S.M. Kamali Shahri, I. Schoen, U. Linn, M. Mahmoudi,
of differently functionalized gold nanorods in human serum: a response from Probing fibronectin conformation on a protein corona layer around nanoparticles,
capillary electrophoresis-inductively coupled plasma mass spectrometry, Nanoscale 10 (3) (2018) 1228–1233, https://doi.org/10.1039/c7nr06970g.
J. Chromatogr. A 1499 (2017) 222–225, https://doi.org/10.1016/j. [111] R. del Cano, L. Mateus, G. Sanchez-Obrero, J.M. Sevilla, R. Madueno,
chroma.2017.03.081. M. Blazquez, T. Pineda, Hemoglobin bioconjugates with surface-protected gold
[90] J.D. Delgado, R.L. Surmaitis, C.J. Arias, J.B. Schlenoff, Surface sulfonates lock nanoparticles in aqueous media: the stability depends on solution pH and protein
serum albumin into a “hard” corona, Biomater. Sci. 7 (8) (2019) 3213–3225, properties, J. Colloid Interface Sci. 505 (2017) 1165–1171, https://doi.org/
https://doi.org/10.1039/c9bm00475k. 10.1016/j.jcis.2017.07.011.
[91] N.P. Mortensen, G.B. Hurst, W. Wang, C.M. Foster, P.D. Nallathamby, S. [112] S.T. Yang, Y. Liu, Y.W. Wang, A. Cao, Biosafety and bioapplication of
T. Retterer, Dynamic development of the protein corona on silica nanoparticles: nanomaterials by designing protein-nanoparticle interactions, Small 9 (9–10)
composition and role in toxicity, Nanoscale 5 (14) (2013) 6372–6380, https:// (2013) 1635–1653, https://doi.org/10.1002/smll.201201492.
doi.org/10.1039/c3nr33280b. [113] E. Casals, T. Pfaller, A. Duschl, G.J. Oostingh, V.F. Puntes, Hardening of the
[92] M. Lundqvist, J. Stigler, G. Elia, I. Lynch, T. Cedervall, K.A. Dawson, Nanoparticle nanoparticle-protein corona in metal (Au, Ag) and oxide (Fe3O4, CoO, and CeO2)
size and surface properties determine the protein corona with possible nanoparticles, Small 7 (24) (2011) 3479–3486, https://doi.org/10.1002/
implications for biological impacts, Proc. Natl. Acad. Sci. U. S. A. 105 (38) (2008) smll.201101511.
14265–14270, https://doi.org/10.1073/pnas.0805135105. [114] M. Lundqvist, J. Stigler, T. Cedervall, T. Berggard, M.B. Flanagan, I. Lynch,
[93] U. Martens, U. Janke, S. Moller, D. Talbot, A. Abou-Hassan, M. Delcea, Interaction G. Elia, K. Dawson, The evolution of the protein corona around nanoparticles: a
of fibrinogen-magnetic nanoparticle bioconjugates with integrin reconstituted test study, ACS Nano 5 (9) (2011) 7503–7509, https://doi.org/10.1021/
into artificial membranes, Nanoscale 12 (38) (2020) 19918–19930, https://doi. nn202458g.
org/10.1039/d0nr04181e. [115] S. Milani, F.B. Bombelli, A.S. Pitek, K.A. Dawson, J. Radler, Reversible versus
[94] V. Mirshafiee, R. Kim, S. Park, M. Mahmoudi, M.L. Kraft, Impact of protein pre- irreversible binding of transferrin to polystyrene nanoparticles: soft and hard
coating on the protein corona composition and nanoparticle cellular uptake, corona, ACS Nano 6 (3) (2012) 2532–2541, https://doi.org/10.1021/nn204951s.
Biomaterials 75 (2016) 295–304, https://doi.org/10.1016/j. [116] S. Dominguez-Medina, L. Kisley, L.J. Tauzin, A. Hoggard, B. Shuang, A.S.D.
biomaterials.2015.10.019. S. Indrasekara, S.S. Chen, L.Y. Wang, P.J. Derry, A. Liopo, E.R. Zubarev, C.
[95] H. Keshavarz, A. Khavandi, S. Alamolhoda, M.R. Naimi-Jamal, Magnetite F. Landes, S. Link, Adsorption and unfolding of a single protein triggers
mesoporous silica nanoparticles embedded in carboxybetaine methacrylate for nanoparticle aggregation, ACS Nano 10 (2) (2016) 2103–2112, https://doi.org/
application in hyperthermia and drug delivery, New J. Chem. 44 (20) (2020) 10.1021/acsnano.5b06439.
8232–8240, https://doi.org/10.1039/d0nj00939c. [117] F. Barbero, L. Russo, M. Vitali, J. Piella, I. Salvo, M.L. Borrajo, M. Busquets-Fite,
[96] C. Grafe, M. von der Luhe, A. Weidner, P. Globig, J.H. Clement, S. Dutz, F. R. Grandori, N.G. Bastus, E. Casals, V. Puntes, Formation of the protein corona:
H. Schacher, Protein corona formation and its constitutional changes on magnetic the interface between nanoparticles and the immune system, Semin. Immunol. 34
nanoparticles in serum featuring a polydehydroalanine coating: effects of charge (2017) 52–60, https://doi.org/10.1016/j.smim.2017.10.001.
and incubation conditions, Nanotechnology 30 (2019), 265707, https://doi.org/ [118] K. Choi, J.E. Riviere, N.A. Monteiro-Riviere, Protein corona modulation of
10.1088/1361-6528/ab0ed0. hepatocyte uptake and molecular mechanisms of gold nanoparticle toxicity,
[97] F.A. de Oliveira, L.J.C. Albuquerque, K.A. Riske, E. Jager, F.C. Giacomelli, Nanotoxicology 11 (1) (2017) 64–75, https://doi.org/10.1080/
Outstanding protein-repellent feature of soft nanoparticles based on poly(N-(2- 17435390.2016.1264638.
hydroxypropyl) methacrylamide) outer shells, J. Colloid Interface Sci. 574 (2020) [119] Y. Li, N.A. Monteiro-Riviere, Mechanisms of cell uptake, inflammatory potential
260–271, https://doi.org/10.1016/j.jcis.2020.04.048. and protein corona effects with gold nanoparticles, Nanomedicine 11 (24) (2016)
[98] R. Gref, M. Luck, P. Quellec, M. Marchand, E. Dellacherie, S. Harnisch, T. Blunk, 3185–3203, https://doi.org/10.2217/nnm-2016-0303.
R.H. Muller, ‘Stealth’ corona-core nanoparticles surface modified by polyethylene [120] L.M. Wang, J.Y. Li, J. Pan, X.M. Jiang, Y.L. Ji, Y.F. Li, Y. Qu, Y.L. Zhao, X.C. Wu,
glycol (PEG): influences of the corona (PEG chain length and surface density) and C.Y. Chen, Revealing the binding structure of the protein corona on gold nanorods
of the core composition on phagocytic uptake and plasma protein adsorption, using synchrotron radiation-based techniques: understanding the reduced

14
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

damage in cell membranes, J. Am. Chem. Soc. 135 (46) (2013) 17359–17368, [142] B. Kang, P. Okwieka, S. Schottler, S. Winzen, J. Langhanki, K. Mohr, T. Opatz,
https://doi.org/10.1021/ja406924v. V. Mailander, K. Landfester, F.R. Wurm, Carbohydrate-based nanocarriers
[121] S. Behzadi, V. Serpooshan, R. Sakhtianchi, B. Muller, K. Landfester, D. Crespy, exhibiting specific cell targeting with minimum influence from the protein
M. Mahmoudi, Protein corona change the drug release profile of nanocarriers: corona, Angew. Chem. Int Ed. Engl. 54 (25) (2015) 7436–7440, https://doi.org/
The “overlooked” factor at the nanobio interface, Colloid Surf. B 123 (2014) 10.1002/anie.201502398.
143–149, https://doi.org/10.1016/j.colsurfb.2014.09.009. [143] M. Yu, J. Xu, J. Zheng, Renal clearable luminescent gold nanoparticles: from the
[122] Z.S. Al-Ahmady, M. Hadjidemetriou, J. Gubbins, K. Kostarelos, Formation of bench to the clinic, Angew. Chem. Int. Ed. 0 (0) (2018), https://doi.org/10.1002/
protein corona in vivo affects drug release from temperature-sensitive liposomes, anie.201807847.
J. Control. Release 276 (2018) 157–167, https://doi.org/10.1016/j. [144] A.K. Murthy, R.J. Stover, W.G. Hardin, R. Schramm, G.D. Nie, S. Gourisankar, T.
jconrel.2018.02.038. M. Truskett, K.V. Sokolov, K.P. Johnston, Charged gold nanoparticles with
[123] Y. Zhang, X. Zhan, S. Peng, Y. Cai, Y.S. Zhang, Y. Liu, Z. Wang, Y. Yu, Y. Wang, essentially zero serum protein adsorption in undiluted fetal bovine serum, J. Am.
Q. Shi, X. Zeng, K. Yuan, N. Zhou, R. Joshi, M. Zhang, Z. Zhang, W. Min, Targeted- Chem. Soc. 135 (21) (2013) 7799–7802, https://doi.org/10.1021/ja400701c.
gene silencing of BRAF to interrupt BRAF/MEK/ERK pathway synergized [145] D.F. Moyano, K. Saha, G. Prakash, B. Yan, H. Kong, M. Yazdani, V.M. Rotello,
photothermal therapeutics for melanoma using a novel FA-GNR-siBRAF Fabrication of corona-free nanoparticles with tunable hydrophobicity, ACS nano
nanosystem, Nanomedicine 14 (5) (2018) 1679–1693, https://doi.org/10.1016/j. 8 (7) (2014) 6748–6755, https://doi.org/10.1021/nn5006478.
nano.2018.04.010. [146] D. Klepac, H. Kostkova, S. Petrova, P. Chytil, T. Etrych, S. Kereiche, I. Raska, D.
[124] Y. Zhang, J. Fu, Y. Shi, S. Peng, Y. Cai, X. Zhan, N. Song, Y. Liu, Z. Wang, Y. Yu, A. Weitz, S.K. Filippov, Interaction of spin-labeled HPMA-based nanoparticles
Y. Wang, Q. Shi, Y. Fu, K. Yuan, N. Zhou, R. Joshi, T.E. Ichim, W. Min, A new with human blood plasma proteins - the introduction of protein-corona-free
cancer immunotherapy via simultaneous DC-mobilization and DC-targeted IDO polymer nanomedicine, Nanoscale 10 (13) (2018) 6194–6204, https://doi.org/
gene silencing using an immune-stimulatory nanosystem, Int. J. Cancer 143 (8) 10.1039/C7NR09355A.
(2018) 2039–2052, https://doi.org/10.1002/ijc.31588. [147] L.L. Knittel, H.Y. Zhao, A. Nguyen, A. Miranda, P. Schuck, A.A. Sousa, Ultrasmall
[125] G. Su, H. Jiang, B. Xu, Y. Yu, X. Chen, Effects of protein corona on active and gold nanoparticles coated with zwitterionic glutathione monoethyl ester: a model
passive targeting of cyclic RGD peptide-functionalized PEGylation nanoparticles, platform for the incorporation of functional peptides, J. Phys. Chem. B 124 (19)
Mol. Pharm. 15 (11) (2018) 5019–5030, https://doi.org/10.1021/acs. (2020) 3892–3902, https://doi.org/10.1021/acs.jpcb.0c01444.
molpharmaceut.8b00612. [148] A. Gupta, D.F. Moyano, A. Parnsubsakul, A. Papadopoulos, L.S. Wang, R.
[126] R. Rampado, S. Crotti, P. Caliceti, S. Pucciarelli, M. Agostini, Recent advances in F. Landis, R. Das, V.M. Rotello, Ultrastable and biofunctionalizable gold
understanding the protein corona of nanoparticles and in the formulation of nanoparticles, ACS Appl. Mater. Interfaces 8 (22) (2016) 14096–14101, https://
“stealthy” nanomaterials, Front. Bioeng. Biotechnol. 8 (2020) 166, https://doi. doi.org/10.1021/acsami.6b02548.
org/10.3389/fbioe.2020.00166. [149] P.Y. Li, Z. Fan, H. Cheng, Cell membrane bioconjugation and membrane-derived
[127] S.G. Elci, Y. Jiang, B. Yan, S.T. Kim, K. Saha, D.F. Moyano, G. Yesilbag Tonga, L. nanomaterials for immunotherapy, Bioconjug. Chem. 29 (3) (2018) 624–634,
C. Jackson, V.M. Rotello, R.W. Vachet, Surface charge controls the suborgan https://doi.org/10.1021/acs.bioconjchem.7b00669.
biodistributions of gold nanoparticles, ACS Nano 10 (5) (2016) 5536–5542, [150] Z. Fan, P.Y. Li, J. Deng, S.C. Bady, H. Cheng, Cell membrane coating for reducing
https://doi.org/10.1021/acsnano.6b02086. nanoparticle-induced inflammatory responses to scaffold constructs, Nano Res. 11
[128] K. Saha, M. Rahimi, M. Yazdani, S.T. Kim, D.F. Moyano, S. Hou, R. Das, R. Mout, (10) (2018) 5573–5583, https://doi.org/10.1007/s12274-018-2084-y.
F. Rezaee, M. Mahmoudi, V.M. Rotello, Regulation of macrophage recognition [151] V. Schafer, H. von Briesen, H. Rubsamen-Waigmann, A.M. Steffan, C. Royer,
through the interplay of nanoparticle surface functionality and protein corona, J. Kreuter, Phagocytosis and degradation of human serum albumin microspheres
ACS Nano 10 (4) (2016) 4421–4430, https://doi.org/10.1021/acsnano.6b00053. and nanoparticles in human macrophages, J. Microencapsul. 11 (3) (1994)
[129] I. Mikelez-Alonso, A. Aires, A.L. Cortajarena, Cancer nano-immunotherapy from 261–269, https://doi.org/10.3109/02652049409040455.
the injection to the target: the role of protein corona, Int. J. Mol. Sci. 21 (2) [152] Y.G. Roh, S.W. Shin, S.Y. Kim, S. Kim, Y.T. Lim, B.K. Oh, S.H. Um, Protein
(2020) 519, https://doi.org/10.3390/ijms21020519. nanoparticle fabrication for optimized reticuloendothelial system evasion and
[130] Q.H. Quach, J.C.Y. Kah, Non-specific adsorption of complement proteins affects tumor accumulation, Langmuir 35 (11) (2019) 3992–3998, https://doi.org/
complement activation pathways of gold nanomaterials, Nanotoxicology 11 (3) 10.1021/acs.langmuir.8b03776.
(2017) 382–394, https://doi.org/10.1080/17435390.2017.1306131. [153] V. Gomez-Vallejo, M. Puigivila, S. Plaza-Garcia, B. Szczupak, R. Pinol, J.
[131] Q. Dai, J.L. Guo, Y. Yan, C.S. Ang, N. Bertleff-Zieschang, F. Caruso, Cell- L. Murillo, V. Sorribas, G. Lou, S. Veintemillas, P. Ramos-Cabrer, J. Llop,
conditioned protein coronas on engineered particles influence immune responses, A. Millan, PEG-copolymer-coated iron oxide nanoparticles that avoid the
Biomacromolecules 18 (2) (2017) 431–439, https://doi.org/10.1021/acs. reticuloendothelial system and act as kidney MRI contrast agents, Nanoscale 10
biomac.6b01545. (29) (2018) 14153–14164, https://doi.org/10.1039/c8nr03084g.
[132] R.M. Visalakshan, M.N. MacGregor, S. Sasidharan, A. Ghazaryan, A. [154] Y. Zou, S. Ito, F. Yoshino, Y. Suzuki, L. Zhao, N. Komatsu, Polyglycerol grafting
M. Mierczynska-Vasilev, S. Morsbach, V. Mailander, K. Landfester, J.D. Hayball, shields nanoparticles from protein corona formation to avoid macrophage uptake,
K. Vasilev, Biomaterial surface hydrophobicity-mediated serum protein ACS Nano 14 (6) (2020) 7216–7226, https://doi.org/10.1021/acsnano.0c02289.
adsorption and immune responses, ACS Appl. Mater. Interfaces 11 (31) (2019) [155] Y. Tang, X. Wang, J. Li, Y. Nie, G. Liao, Y. Yu, C. Li, Overcoming the
27615–27623, https://doi.org/10.1021/acsami.9b09900. reticuloendothelial system barrier to drug delivery with a “Don’t-Eat-Us” strategy,
[133] R. Cai, J. Ren, Y. Ji, Y. Wang, Y. Liu, Z. Chen, Z. Farhadi Sabet, X. Wu, I. Lynch, ACS Nano 13 (11) (2019) 13015–13026, https://doi.org/10.1021/
C. Chen, Corona of thorns: the surface chemistry-mediated protein corona acsnano.9b05679.
perturbs the recognition and immune response of macrophages, ACS Appl. Mater. [156] M.P. Nikitin, I.V. Zelepukin, V.O. Shipunova, I.L. Sokolov, S.M. Deyev, P.
Interfaces 12 (2) (2020) 1997–2008, https://doi.org/10.1021/acsami.9b15910. I. Nikitin, Enhancement of the blood-circulation time and performance of
[134] F. Charbgoo, M. Nejabat, K. Abnous, F. Soltani, S.M. Taghdisi, M. Alibolandi, nanomedicines via the forced clearance of erythrocytes, Nat. Biomed. Eng. 4 (7)
W. Thomas Shier, T.W.J. Steele, M. Ramezani, Gold nanoparticle should (2020) 717–731, https://doi.org/10.1038/s41551-020-0581-2.
understand protein corona for being a clinical nanomaterial, J. Control. Release [157] H. Cabral, K. Kataoka, Erythrocyte depletion lifts nanoparticle half-lives, Nat.
272 (2018) 39–53, https://doi.org/10.1016/j.jconrel.2018.01.002. Biomed. Eng. 4 (7) (2020) 670–671, https://doi.org/10.1038/s41551-020-0586-
[135] X. Lu, P. Xu, H.M. Ding, Y.S. Yu, D. Huo, Y.Q. Ma, Tailoring the component of x.
protein corona via simple chemistry, Nat. Commun. 10 (1) (2019) 4520, https:// [158] L. Rao, J.H. Xu, B. Cai, H. Liu, M. Li, Y. Jia, L. Xiao, S.S. Guo, W. Liu, X.Z. Zhao,
doi.org/10.1038/s41467-019-12470-5. Synthetic nanoparticles camouflaged with biomimetic erythrocyte membranes for
[136] D. Pozzi, G. Caracciolo, A.L. Capriotti, C. Cavaliere, S. Piovesana, reduced reticuloendothelial system uptake, Nanotechnology 27 (8) (2016),
V. Colapicchioni, S. Palchetti, A. Riccioli, A. Lagana, A proteomics-based 085106, https://doi.org/10.1088/0957-4484/27/8/085106.
methodology to investigate the protein corona effect for targeted drug delivery, [159] E. Casals, T. Pfaller, A. Duschl, G.J. Oostingh, V. Puntes, Time evolution of the
Mol. Biosyst. 10 (11) (2014) 2815–2819, https://doi.org/10.1039/c4mb00292j. nanoparticle protein corona, ACS Nano 4 (7) (2010) 3623–3632, https://doi.org/
[137] G. Caracciolo, The protein corona effect for targeted drug delivery, Bioinspired 10.1021/nn901372t.
Biomim. Nanobiomater. 2 (1) (2013) 54–57, https://doi.org/10.1680/ [160] A.L. Barran-Berdon, D. Pozzi, G. Caracciolo, A.L. Capriotti, G. Caruso,
bbn.12.00027. C. Cavaliere, A. Riccioli, S. Palchetti, A. Lagana, Time evolution of nanoparticle-
[138] D. Westmeier, R.H. Stauber, D. Docter, The concept of bio-corona in modulating protein corona in human plasma: relevance for targeted drug delivery, Langmuir
the toxicity of engineered nanomaterials (ENM), Toxicol. Appl. Pharm. 299 29 (21) (2013) 6485–6494, https://doi.org/10.1021/la401192x.
(2016) 53–57, https://doi.org/10.1016/j.taap.2015.11.008. [161] S. Dreis, F. Rothweller, A. Michaelis, J. Cinatl, J. Kreuter, K. Langer, Preparation,
[139] A. Solorio-Rodriguez, V. Montesinos-Cruz, V. Escamilla-Rivera, M. Uribe- characterisation and maintenance of drug efficacy of doxorubicin-loaded human
Ramirez, C.M. Garcia-Cuellar, Characterization of protein corona around of SiO2- serum albumin (HSA) nanoparticles, Int J. Pharm. 341 (1–2) (2007) 207–214,
PEG-Tf NP in human plasma and its influence in active targeting, Toxicol. Lett. https://doi.org/10.1016/j.ijpharm.2007.03.036.
259 (2016) S185, https://doi.org/10.1016/j.toxlet.2016.07.442. [162] H. Jeong, M. Huh, S.J. Lee, H. Koo, I.C. Kwon, S.Y. Jeong, K. Kim, Photosensitizer-
[140] Y.C. Li, Y.L. Xu, C.C. Fleischer, J. Huang, R. Lin, L. Yang, H. Mao, Impact of anti- conjugated human serum albumin nanoparticles for effective photodynamic
biofouling surface coatings on the properties of nanomaterials and their therapy, Theranostics 1 (2011) 230–239.
biomedical applications, J. Mater. Chem. B 6 (1) (2018) 9–24, https://doi.org/ [163] J. Kufleitner, S. Wagner, F. Worek, H. von Briesen, J. Kreuter, Adsorption of
10.1039/c7tb01695f. obidoxime onto human serum albumin nanoparticles: drug loading, particle size
[141] B. Kang, P. Okwieka, S. Schottler, O. Seifert, R.E. Kontermann, K. Pfizenmaier, and drug release, J. Microencapsul. 27 (6) (2010) 506–513, https://doi.org/
A. Musyanovych, R. Meyer, M. Diken, U. Sahin, V. Mailander, F.R. Wurm, 10.3109/02652041003681406.
K. Landfester, Tailoring the stealth properties of biocompatible polysaccharide [164] J.C.Y. Kah, J. Chen, A. Zubieta, K. Hamad-Schifferli, Exploiting the protein corona
nanocontainers, Biomaterials 49 (2015) 125–134, https://doi.org/10.1016/j. around gold nanorods for loading and triggered release, ACS Nano 6 (8) (2012)
biomaterials.2015.01.042. 6730–6740, https://doi.org/10.1021/nn301389c.

15
W. Huang et al. Biomedicine & Pharmacotherapy 139 (2021) 111541

[165] D. Walczyk, F.B. Bombelli, M.P. Monopoli, I. Lynch, K.A. Dawson, What the cell [168] W. Poon, B.R. Kingston, B. Ouyang, W. Ngo, W.C.W. Chan, A framework for
“Sees” in bionanoscience, J. Am. Chem. Soc. 132 (16) (2010) 5761–5768, https:// designing delivery systems, Nat. Nanotechnol. 15 (10) (2020) 819–829, https://
doi.org/10.1021/ja910675v. doi.org/10.1038/s41565-020-0759-5.
[166] S.R. Saptarshi, A. Duschl, A.L. Lopata, Interaction of nanoparticles with proteins: [169] Z. Ban, P. Yuan, F. Yu, T. Peng, Q. Zhou, X. Hu, Machine learning predicts the
relation to bio-reactivity of the nanoparticle, J. Nanobiotechnol. 11 (2013) 26, functional composition of the protein corona and the cellular recognition of
https://doi.org/10.1186/1477-3155-11-26. nanoparticles, Proc. Natl. Acad. Sci. U. S. A. 117 (19) (2020) 10492–10499,
[167] V.H. Nguyen, B.J. Lee, Protein corona: a new approach for nanomedicine design, https://doi.org/10.1073/pnas.1919755117.
Int. J. Nanomed. 12 (2017) 3137–3151, https://doi.org/10.2147/IJN.S129300.

16

You might also like