You are on page 1of 24

Nanoparticle-Protein Interaction:

The Significance and Role 11
of Protein Corona

Saad Mohammad Ahsan, Chintalagiri Mohan Rao,
and Md. Faiz Ahmad

Abstract
The physico-chemical properties of nanoparticles, as characterized under
idealized laboratory conditions, have been suggested to differ significantly
when studied under complex physiological environments. A major reason
for this variation has been the adsorption of biomolecules (mainly pro-
teins) on the nanoparticle surface, constituting the so-called “biomolecular
corona”. The formation of biomolecular corona on the nanoparticle sur-
face has been reported to influence various nanoparticle properties viz.
cellular targeting, cellular interaction, in vivo clearance, toxicity, etc.
Understanding the interaction of nanoparticles with proteins upon admin-
istration in vivo thus becomes important for the development of effective
nanotechnology-based platforms for biomedical applications. In this chap-
ter, we describe the formation of protein corona on nanoparticles and the
differences arising in its composition due to variations in nanoparticle
properties. Also discussed is the influence of protein corona on various
nanoparticle activities.

Keywords
Protein corona · Nanoparticles · Biomolecular corona · Nanoparticles-
protein interaction

11.1 Introduction
S.M. Ahsan (*) · C.M. Rao After entering into a biological system, foreign
Centre for Cellular and Molecular Biology (CSIR-­
CCMB), Hyderabad, Telangana State, India particles interact with various biomolecules. As
e-mail: saadmahsan@gmail.com compared to bulk matter, such interactions are
Md.F. Ahmad (*) more prominent in case of nanomaterials due to
Department of Pharmacology, School of Medicine, their large surface area to volume ratio and a high
Case Western Reserve University, surface free energy. With rapid developments in
Cleveland, OH, USA the field of nanomedicine and the realisation of
e-mail: fam37@case.edu

© The Author(s) 2018 175


Q. Saquib et al. (eds.), Cellular and Molecular Toxicology of Nanoparticles, Advances
in Experimental Medicine and Biology 1048, https://doi.org/10.1007/978-3-319-72041-8_11
176 S.M. Ahsan et al.

regulatory/safety features in nanoparticle based and high abundance in the plasma. With time,
therapeutics, it has become important to study proteins with higher affinities and longer resi-
the interaction of components from biological dence times replace proteins with lower affinities
fluids with such nanoscale materials. The interac- and short residence times.
tion of nanomaterials with the surrounding bio- Proteins which bind to the nanoparticle sur-
logical environment results in the formation of a face with high affinities and display high associa-
layer of adsorbed biomolecules on the nanomate- tion rates constitute the “hard corona”. The
rial surface (Fig. 11.1a–c), leading to the modifi- proteins constituting the hard corona directly
cation of its properties and imparting it with a interact with the nanoparticle surface. On the
new identity [1]. This layer of biomolecules is other hand, proteins that are loosely bound to the
termed as the “biomolecular corona” [2]. The nanoparticle surface with low binding affinities
lipid profile of the biomolecular corona has been and display low association rates constitute the
mainly studied in context of the pulmonary envi- “soft corona” (Fig. 11.1e).
ronment and is shown to be relatively conserved The interaction of proteins present in the soft
across different nanoparticle types and depends corona with the nanoparticle surface is indirect
on the relative abundance of the lipids [3, 4]. The and meditated via the hard corona [1, 5].
protein composition of the biomolecular corona The proteins constituting the soft corona rap-
however, has been a subject of extensive study idly exchange from the surrounding environment
due to its variations and significance in dictating on short time scales. The hard corona is more
nanoparticle properties [3, 5]. stable and tightly associated with the nanoparti-
cles and is considered to play an important role in
nanoparticle interaction with its surrounding
11.2 Formation of Protein Corona cells [11]. As a nanoparticle passes through a
variety of biological environments, the soft
The process of continuous adsorption and desorp- corona is altered. However, the hard corona is
tion of plasma proteins on a surface with regards altered to a lesser extent and retains the proteins
to time of incubation, surface properties, etc. is adsorbed during initial exposures.
termed as the “Vroman effect” [6–8]. The adsorp- A variety of techniques are used to study the
tion of plasma proteins and the subsequent for- structure and composition of the protein corona.
mation of protein corona on nanoparticle surface These techniques include dynamic light scatter-
is a rapid event that occurs within a few seconds ing (DLS), differential centrifugal sedimentation
of nanoparticle exposure to a biological environ- (DCS), size exclusion chromatography (SEC)
ment. The proteins adsorbed onto a surface stay and transmission electron microscopy (TEM) for
in a continuous state of flux and the composition determining the thickness of the corona layer;
of these proteins may vary with time (Fig. 11.1d) poly(acrylamide) gel electrophoresis (PAGE)
[9]. On a nanoparticle surface, the initial corona and liquid chromatography tandem mass spec-
consists of proteins with high association rates trometry (LC-MS/MS) for identification and

Fig. 11.1  (continued) adsorb with low affinity. These adsorbed on the nanoparticles through protein-protein
proteins are subsequently displaced by proteins with a interactions constitute the soft corona. The dynamic
higher affinity (blue). The adsorption of certain proteins nature of protein corona arises due to competitive parti-
(blue and green) may aid in the binding of other proteins cle-protein interactions, continuous particle-protein asso-
(yellow) with an initial low affinity for the particle sur- ciation/dissociation and a characteristic protein profile
face. Other proteins (red) do not adsorb at all (Reprinted specific to a particular biological fluid. A continuous asso-
by permission from Macmillan Publishers Ltd.: [Nature ciation and dissociation of proteins alters the protein
Nanotechnology] (Biomolecular coronas provide the bio- corona composition prior to equilibrium after which it is
logical identity of nanosized materials), Copyright (2012) stabilized (Reproduced from “Nanoparticle–protein
[2]). (e) Schematic representation depicting the hard/soft corona complexes govern the biological fates and func-
corona and the dynamic nature of protein corona: Proteins tions of nanoparticles” with permission of The Royal
bound to a nanoparticle surface via protein-nanoparticle Society of Chemistry [10])
interactions constitute the hard corona, while proteins
11  Nanoparticle-Protein Interaction: The Significance and Role of Protein Corona 177

100
a b
BARE
PLASMA
c
80

A(intensity)
60

40

20

0
4 5 6 7 89 2 3 4 5 6 7 89
100 DH[nm] 1000

Time
e
Dynamic nature of protein corona ‘Hard’ and ‘soft’ protein corona
Changes in the secondary
structure of the bound proteins
due to release of surface entropy
Strong affinity
protein Depleted protein
Continuous
protein exchange
association

dissociation

Enriched protein
Hard corona Soft corona

Low affinity protein


‘Free’ proteins,
each with unique concentration
and binding affinity for a specific
type of partilcle surface
Unchanged protein composition
Weak binding affinity
at equilibrium despite the continuous Strong binding affinity
Shorter residence time
protein exchange Longer residence time
Lower abundance in the corona
Higher abundance in the corona

Fig. 11.1 TEM image of (a) bare 100 nm surface-­ (white bars). Protein-PSCOOH nanoparticles show an
carboxylated polystyrene (PSCOOH) nanoparticles and increase in size (Reprinted with permission from “What
(b) protein-nanoparticle complex. (c) DLS (intensity-­ the Cell sees in Bionanoscience”. Copyright (2010)
weighted size distribution) for 100 nm surface-­ American Chemical Society [1]) (d) Time based evolution
carboxylated polystyrene (PSCOOH) nanoparticles (bare) of the biomolecular corona (left to right): Initial corona
(gray bars) and protein-PSCOOH nanoparticle complex consists of highly abundant proteins (green) that
178 S.M. Ahsan et al.

q­ uantification of the proteins; circular dichroism and human carbonic anhydrase II, no change in
spectroscopy (CD), fluorescence quenching and enthalpy is observed. The binding in these cases
computational simulations for determining the is suggested to be entropy driven and is accom-
conformation of the bound proteins; size exclu- panied by a release of bound water molecule
sion chromatography (SEC), surface plasmon from the nanoparticle surface. Such an entropy
resonance (SPR) and isothermal titration calo- driven mechanism of protein binding with the
rimetry (ITC) for estimating the affinities of the release of water molecule from the nanoparticle
bound proteins with the nanomaterial surface [5]. surface does not alter the protein conformation
Various factors dictate the composition of the [15]. In other examples, protein conformation
protein corona on the nanoparticle surface and may be altered if it is thermodynamically
are discussed in detail later. favourable and allows for hydrophobic patches
An important parameter that determines the or charged sequences within proteins to interact
adsorption of proteins on a nanomaterial surface with a complementary region on the nanomate-
is the thermodynamic feasibility of the interac- rial surface [16]. In some cases, specific domains
tion. It can be defined in terms of the Gibbs free within proteins may enhance interaction with
energy of adsorption (ΔGADS) [5, 12]. nanoparticles. As an example, the interaction of
high molecular weight kininogen (HMWK) to
∆GADS = ∆ H ADS − T ∆SADS < 0
iron oxide nanoparticles occurs via the histi-
where, ΔHADS and ΔSADS are the enthalpy and dine-rich domain, with the imidazole side chains
entropy during adsorption and T is the temper- of histidines interacting with iron oxide
ature. The stability of a protein-nanoparticle nanoparticles [17]. Apart from directly interact-
interaction is determined by the net free energy ing with a nanoparticle surface, proteins may
of adsorption (ΔGADS). Proteins adsorbed with indirectly interact with nanoparticles via pro-
a large ΔGADS have a low probability of desorp- tein-protein interactions. These interactions
tion and hence a prolonged association with may be specific where a conformation depen-
the nanoparticles. On the other hand, proteins dent protein-protein interaction takes place.
adsorbed with a low ΔGADS are easily desorbed Such interactions and adsorption of proteins on
and return to solution. In general, nanoparti- to a nanoparticle surface is observed for com-
cles with either charged surface groups or plement factors and coagulation proteins, and is
hydrophobic surfaces form more stable inter- shown to play an important role in mediating a
actions with proteins as compared to nanopar- humoral response [18]. Whereas, in non-spe-
ticles with neutral surface charges and cific interactions an initial conformational
hydrophilic surfaces. change in a protein, post adsorption on a
A number of parameters viz. formation of nanoparticle surface, may lead to an exposure of
bonds, rearrangement of interfacial water mole- a hydrophobic or charged patch which subse-
cules, or conformational changes in either the quently mediates the secondary interactions. As
protein or the nanomaterial surface contribute to an example, the interaction of α-chymotrypsin
a favourable change in enthalpy or entropy. (ChT) and lysozyme (Lyz) with gold nanoparti-
Hydrogen bonding, solvation forces, Van der cles (AuNP) (either nanospheres;AuNS or
Waals interactions, etc. between the nanoparti- nanorods;AuNR), has been shown to result in a
cle surface and the adsorbed proteins have been loss of protein structure and enzyme activity.
reported [13]. In the case of human serum albu- The effect of nanoparticle morphology is found
min adsorption on polymeric nanoparticles, the to be primarily protein-­specific in this case. The
interaction has been shown to be driven with a binding of proteins to gold nanoparticles fol-
release of heat suggesting an enthalpy change lows three steps (Fig. 11.2) viz. (i) an initial
favouring the interaction [14]. For other pro- protein-surface interactions (Region I), (ii) an
teins such as fibrinogen, lysozyme, ovalbumin intermediate stage characterized by protein-sur-
11  Nanoparticle-Protein Interaction: The Significance and Role of Protein Corona 179

Fig. 11.2 Schematic representation of Chymotrypsin face. ChT-AuNS stay in Region I as stable conjugates
(ChT) and Lysozyme (Lyz) adsorption on gold nanopar- with ChT in a native conformation. ChT-AuNR form sta-
ticles (AuNP). Initially (region I) both proteins get ble conjugates and proceed to region III with multiple lay-
adsorbed onto AuNP with minimal structural perturba- ers of ChT adsorbed on the nanoparticle surface.
tion. Later, as depicted in region II, protein-surface inter- Lyz-AuNP conjugates progress to region II, where a mix-
actions (as in the case of ChT-AuNR, Lyz-AuNS and ture of protein-surface and protein-protein interactions
Lyz-AuNR) lead to changes in protein conformation. This result in the aggregation of the conjugates (Reprinted
stage is characterized by protein-surface and protein-­ from “Effect of gold nanoparticle morphology on
protein interactions. The final stage (region III) is domi- adsorbed protein structure and function”, 32(29), Gagner
nated by protein-protein interactions resulting in the JE, Lopez MD, Dordick JS & Siegel RW, 7241–52,
stacking of multiple layers of protein on the particle sur- Copyright (2011), with permission from Elsevier [19])

face and protein-protein interactions (Region 11.3 C


 omposition of Protein
II), and (iii) a final stage dominated by protein- Corona
protein interactions (Region III). The interac-
tion of ChT with gold nanospheres exists in The biomolecular corona formed on nanoparti-
Region I, and does not lead to structural altera- cles is constituted by a variety of plasma proteins
tion of ChT. ChT-AuNR conjugates, on the which fall into the following major categories viz.
other hand progress to Region III, with altera- apolipoproteins, immunoglobulins, complement
tions in ChT conformation and formation of a factors, coagulation factors, acute phase proteins,
protein multilayer. The Lyz adsorption on tissue leakage components and other miscella-
AuNPs proceeds to Region II, characterized by neous proteins [20–22]. As an example, the
Lyz adsorption and denaturation leading to the adsorption of various plasma proteins on silica
exposure of hydrophobic patches in the protein. nanoparticles (SiNPs) of different sizes is pro-
This phenomenon is observed regardless of the vided in Fig. 11.3 [21]. The physiological func-
AuNP (AuNS or AuNR) morphology. The Lyz-­ tion of the major components of the adsorbed
surface and Lyz-Lyz interactions thereby lead to proteins is mainly lipid transport, blood coagula-
aggregation of the conjugates (Fig. 11.2) [19]. tion, complement activation, pathogen recogni-
tion and ion transport. The relative concentrations
Fig. 11.3  Bioinformatic classification (according to their tein groups viz. immunoglobulins (e), acute phase
functions) of corona proteins identified on the surface of response proteins (f), and other components (mainly
silica nanoparticles (SiNPs) of 125 nm (SiNP-125), 20 nm serum albumin) (h) although present in high amounts in
(SiNP-20) and 8 nm (SiNP-8). (a) Proteins identified in the plasma, displayed a lower affinity for the SiNPs
the respective SiNPs corona are classified according to (Reprinted with permission from “Nanoparticle Size Is a
their biological functions. (Relative percentages of the Critical Physicochemical Determinant of the Human
proteins compared to crude plasma are shown). Significant Blood Plasma Corona: A Comprehensive Quantitative
enrichment of plasma proteins involved in complement Proteomic Analysis”. Copyright (2011) American
activation (b), lipoproteins (c), coagulation (d) and tissue Chemical Society [21])
leakage proteins (g) was evident in the corona. Other pro-
11  Nanoparticle-Protein Interaction: The Significance and Role of Protein Corona 181

of different proteins on the nanoparticle surface plasma abundance. The interaction is mediated
constituting the corona does not correspond to by various nanoparticle properties as discussed in
their plasma concentrations, suggesting a selec- the following section.
tive enrichment of some proteins. The selective
adsorption of proteins on the surface of nanopar-
ticles has been shown to be a complex interplay 11.4 F
 actors Influencing Protein
of a variety of nanoparticle features (discussed in Corona
detail later). A detailed account of various pro-
teins adsorbed on different nanoparticles, termed 11.4.1 Nanoparticle Size
as the “adsorbome”, has been provided by
Walkey and Chan [5]. The size of nanomaterials determines its surface
Some of the abundant proteins identified on curvature and also influences its surface area. It is
the corona formed on most nanoparticles are thus an important factor in determining the quali-
albumin, α-2-macroglobulin, apolipoprotein A-I, tative and quantitative composition of the
apolipoprotein A-III, apolipoprotein C-III, adsorbed proteins on a nanoparticle surface.
immunoglobulin kappa chain, different forms of While some reports suggest variations only in the
light- and heavy-chains of immunoglobulins, amount of bound protein with particle size and
complement C3, complement C4, haptoglobin, curvature [22], other reports have shown both
alpha-1-antitrypsin, kininogen, plasminogen, quantitative and qualitative size dependent
keratin, vitronectin, etc. [21, 22]. While the bind- changes in the protein corona [20, 21, 24, 25]. A
ing of albumin and apolipoproteins helps in the difference of as low as 10 nm has been found to
transport of nanoparticles, the binding of comple- significantly affect the nanoparticle corona com-
ment proteins and immunoglobulins leads to par- position. In a study on gold nanoparticles, an
ticle opsonization thereby promoting nanoparticle increase in size leads to an increase in the binding
clearance from circulation [23]. constant of proteins along with an increase in the
Walkey and Chan have reported around two to degree of cooperativity of nanoparticle-protein
six proteins to be adsorbed at high abundance on binding [26]. In a separate study on adsorption of
most nanomaterials, with the most abundant plasma proteins on silica nanoparticles of differ-
identified protein (across all nanomaterials) rep- ent sizes, it was revealed that the binding of
resenting approximately 29% of the total proteins approximately 37% of all corona proteins was
adsorbed. Collectively the three most abundant significantly affected by particle size [21].
proteins represent an average 56% of the total Lundqvist et al. have reported a comparison of
adsorbed proteins. By defining a threshold of protein corona formed on 50 and 100 nm polysty-
10% of the total adsorbed protein mass as “high rene nanoparticles with different surface func-
abundance”, Walkey and Chan have identified 21 tionalities. While the plain polystyrene
out of 125 proteins above this threshold on at nanoparticles of the two sizes showed almost
least one type of nanomaterial. These proteins are 80% similarity in the composition of protein
albumin, transferrin, fibrinogen, haptoglobin, corona, the size effect was more pronounced for
hemoglobin, histidine-rich glycoproteins, Ig the carboxyl modified nanoparticles of the two
gamma chain, Ig light chain, Ig mu chain, inter sizes which showed only 50% similarity [20].
alpha trypsin inhibitor H1, mannose-binding pro-
tein C, paraoxonase-1, antithrombin-III, apolipo-
protein A-1, apolipoprotein A-IV, apolipoprotein 11.4.2 Nanoparticle Surface
B-100, apolipoprotein C-II, apolipoprotein C-III, Properties
apolipoprotein E, clusterin and complement C3
[5]. The interaction of these proteins with the Surface features of nanoparticles viz. charge and
nanoparticle surface, as mentioned earlier, is not hydrophobicity/hydrophilicity are important fac-
related to their physiological roles or relative tors in determining the protein corona composi-
182 S.M. Ahsan et al.

tion. As a general concept, negatively charged ference in opsonization rates of the nanoparticles.
nanoparticles attract positively charged proteins Neutrally charged nanoparticles show slow opso-
and vice versa. Proteins with pI >5.5 show a pref- nization rates as compared to charged nanoparti-
erence for negatively charged (nanoparticles with cles, suggesting a direct correlation between the
acidic functional groups) nanoparticles, while corona composition and nanoparticle uptake [23,
those with pI <5.5 show a preference for posi- 31]. As an example, a charged based cellular
tively charged (nanoparticles with basic func- uptake for albumin nanoparticles has been
tional groups) nanoparticles. In such cases, the reported by Roser et al. Albumin nanoparticles
protein adsorption on nanoparticle surfaces is with neutral surface charge showed a reduced
expected to be mainly driven by coulombic inter- phagocytic uptake in comparison with albumin
actions [27]. However, as most plasma proteins nanoparticles which were modified to carry a net
possess a negative charge at physiological pH positive or negative surface charge [31]. The
(most plasma proteins have a pI in the range of serum proteins identified in the opsonization pro-
6–8), charge based interaction of plasma proteins cess were C3b-complement, IgG and fibronectin,
with negatively charged nanoparticles is a far whereas albumin was shown to reduce phago-
more complex interaction. In such cases, cou- cytic uptake of the particles.
lombic interactions take place between specific Apart from surface charge, the hydrophilicity/
surface domains of the proteins that possess a hydrophobicity of nanoparticles has also been
complementary charge to that of the nanoparticle reported to influence the protein corona forma-
surface. As the coulombic interactions are tion both quantitatively and qualitatively [22,
achieved between molecules that are in close 32]. The major proteins affected by surface
vicinity of each other, such charged based inter- hydrophobicity are albumin, fibrinogen and apo-
actions are expected to involve individual/spe- lipoproteins [33]. As an example, a comparison
cific domains on the protein and nanoparticle of proteins adsorbed on a less hydrophobic 85:15
surface rather than the entire surface [28]. In a NIPAM/BAM copolymer particle and a more
study on the adsorption of three human serum hydrophobic 50:50 copolymer particle revealed
proteins viz. serum albumin, apolipoprotein A-I clear differences in the amount and type of pro-
and apolipoprotein E4 on negatively charged teins bound. While the less hydrophobic 85:15
(carboxylated) FePt nanoparticles, an increase in NIPAM/BAM copolymer particle bound virtu-
the hydrodynamic radius of the nanoparticles ally no protein (trace amounts of albumin), the
with protein concentration was observed suggest- more hydrophobic 50:50 copolymer particles
ing corona formation. Equilibrium dissociation preferentially bound apolipoproteins (AI, AII,
coefficient studies suggested electrostatic inter- AIV, and E), human serum albumin, fibrinogen
actions between protein and nanoparticles. and various other proteins [22]. As mentioned
Structural studies revealed the presence of posi- earlier, the differences in the amount and type of
tively charged patches on the protein surface, plasma proteins adsorbed on nanoparticles leads
through which the protein molecules were shown to variation in their opsonization rates.
to interact electrostatically with the nanoparticle Nanoparticles with hydrophobic surfaces thus
surface [29]. Studies on the effect of nanoparticle show high opsonization rates due to an enhanced
surface charge density have revealed a direct adsorption of plasma proteins [23, 34].
relation between the surface charge and protein
adsorption profiles. A higher surface charge leads
to an increased plasma protein adsorption, with- 11.4.3 Time of Exposure
out much difference in the qualitative profile of
the of the detected proteins [7, 30]. The Vroman effect exhibited by protein adsorp-
In general, charged nanoparticles adsorb more tion on to a nanoparticle surface can be divided
protein than nanoparticles with a neutral surface. into “early” and “late” stages. The early stage is
This differential adsorption further leads to a dif- characterized by the adsorption of proteins with
11  Nanoparticle-Protein Interaction: The Significance and Role of Protein Corona 183

high mobility although low binding affinity. different manner [9]. While the protein quantity
Thereafter, the low binding affinity proteins are on commercial 30 nm silica nanoparticles
progressively displaced by the ones with higher (AmSil30) decreased with time, it was shown to
binding affinities. The initial interaction of low increase for negatively and positively charged
binding affinity proteins is an attempt on the part polystyrene nanoparticles (nPsNP and PPsNP)
of the nanoparticles to lower its surface energy (Fig. 11.4a, b).
followed by exchange of proteins on shorter time Apart from quantitative variations, time based
scales from the available set of proteins that may differences in composition of the proteins con-
diffuse to the surface [35]. Various studies have stituting the corona were also observed for dif-
revealed the importance of plasma exposure time ferent nanoparticles (Fig. 11.4c). In a report on
on the quantity and composition of the corona the characterization of adsorbed proteins on the
proteins. Although the corona deposition is dic- surface of gold nanoparticles, it has been
tated by the NP composition and surface func- reported that at initial time points (1 h), high
tionalization, exposure time is also an important molecular weight (HMW) and low molecular
factor. As demonstrated by Tenzer et al. the weight (LMW) proteins are adsorbed while the
corona on nanoparticles with different types of medium molecular weight (MMW) proteins are
surface functionalization, evolves in a completely down represented. However, after prolonged

Fig. 11.4 (a) SDS–PAGE of plasma proteins bound to 140 nm laboratory synthesized silica nanoparticles
different nanoparticles. (b) Quantification of proteins in (SiNP140); Amine functionalized SiNP140
the corona (protein (fg) per nanoparticle) at different (SiNP140NH2); Carboxylate functionalized SiNP140
time points. (c) Identification and classification of (SiNP140-COOH); negatively or positively charged
corona proteins identified on different nanoparticles. polystyrene nanoparticles (nPsNP and pPsNP)
(30 nm commercial silica nanoparticles (AmSil30); (Reprinted by permission from Macmillan Publishers
30 nm laboratory synthesized silica nanoparticles Ltd.: [Nature Nanotechnology] (Rapid formation of
(SiNP30); Amine functionalized SiNP30 (SiNP30-NH2); plasma protein corona critically affects nanoparticle
carboxylate functionalized SiNP30 (SiNP30-COOH); pathophysiology), Copyright (2013) [9])
184 S.M. Ahsan et al.

incubation (48 h) the concentration of the MMW While the initial phase of corona formation
proteins was found to increase while that of may be a rapid event occurring on a time scale of
LMW was found to decrease. The major proteins few seconds to minutes, the late stage may take a
identified were MMW proteins and some LMW few hours to days [5, 40]. However, depending on
proteins, involved in important biological pro- the evolution stage and the protein corona at that
cesses such as transport and trafficking (apolipo- particular moment, the physiological response to
protein A1, transferrin, vitamin D-binding the nanoparticle may vary.
protein, etc.), blood coagulation (protein C
inhibitor, antithrombin III, coagulation factor V,
etc.) and tissue development (fibulin 1, periostin, 11.4.4 Temperature
thrombospondin-1, galectin 3 binding protein,
etc.). The concentration of the HMW proteins Physiological temperatures are known to fluctuate
was however not effected significantly with time and vary between 35 and 41 °C under different
[36]. A sequential adsorption of plasma proteins conditions. Exploring the effect of these subtle
hasalsobeendemonstratedonN-iso-­propylacrylamide/N- temperature changes on the surface adsorption of
tert-butylacrylamide (NIPAM/BAM) nanoparti- proteins thus becomes essential. Mahmoudi et al.
cles through mathematical modelling. For these studied the effect of temperature on the adsorp-
nanoparticles, the initial phase is characterized tion of albumin and apo-transferrin (apo-Tf) on
by the deposition of high abundance and fast dis- polymer coated iron-platinum (FePt) nanoparti-
sociation proteins viz. albumin and fibrinogen cles. At lower temperatures (13 and 23 °C) the
followed by higher affinity and slow-exchanging deposition of both albumin and apo-Tf took place
apolipoproteins AI, AII, AIV and E [22, 37]. The till the formation of a monolayer, after which
relative abundance of proteins in the serum also increasing the concentration of the respective pro-
plays an important role in determining the initial tein had no effect on the corona thickness.
binding partners. The first proteins to be adsorbed However, at a higher temperature (43 °C) a
on nanoparticles are albumin, IgG and fibrino- decrease in the thickness of the corona shell was
gen which may later be replaced by apolipopro- observed for both HSA and apo-Tf, which was
teins and coagulation factors. In a surface proposed due to (i) conformation changes in the
plasmon resonance (SPR) based study on silver protein molecules, (ii) decrease in the number of
coated slides, it was observed that initially the protein molecules adsorbed on the nanoparticle
smallest and most abundant protein viz. albumin surface and (iii) an increase in the flexibility of the
adsorbs to the surface followed by IgG and polymer coating on the nanoparticle surface lead-
fibrinogen [38]. ing to the insertion of the protein molecules in the
As the quantitative and qualitative profile of polymer shell [41]. Mahmoudi et al. have also
protein corona changes with incubation time, it studied the protein corona composition on gold
also influences the cellular uptake of nanoparti- nanorods (AuNR) after plasmonic heating and
cles. As an example, the hepatic uptake of thermal heating under two different serum condi-
lecithin-­coated polystyrene nanospheres has been tions mimicking an in vitro (10% serum) environ-
found to increase significantly with an increase in ment and an in vivo (100% serum) environment.
incubation time. This is mainly due to an increase Subtle changes in zeta-potential of AuNRs were
in the opsonin concentration in the adsorbed observed following both plasmonic and thermal
layer and an opsonin-mediated uptake by Kupfer heating suggesting some compositional differ-
cells. The major opsonins identified were com- ences arising due to the change in temperature.
plement C3 (C3) and immunoglobulin G (IgG). The decrease in surface charge was slightly more
Their concentrations were found to increase with in AuNRs incubated in 100% serum as compared
time, thereby accounting for an increase in cel- to those incubated in 10% serum. Analysis of the
lular uptake [39]. protein composition revealed an increase in the
amount of several important proteins viz. serum
11  Nanoparticle-Protein Interaction: The Significance and Role of Protein Corona 185

albumin, α-2-HS-­ glycoprotein precursor, apoli- may be due to the heat induced depletion of com-
poprotein A-II p­ recursor and apolipoprotein C− plement factors and the unresponsiveness of the
III precursor in the plasmonic heating 10% serum cell line (A549) to complement based particle
model as compared to the sample incubated at uptake.
37 °C. The levels of apolipoprotein A-I precursor
protein was found to be decreased. However,
there were significant differences in the in vitro 11.4.5 Biological Environment
(10% serum) and in vivo (100% serum) models.
In terms of the molecular weight of the proteins The biomolecular corona formed on nanoparti-
adsorbed, it was observed that for the 10% serum cles may also vary depending on the variations in
model a heat treatment whether thermal or plas- the physiological environments. Differences have
monic did not have a significant effect on the been reported in the corona composition of
adsorption of the low molecular weight proteins nanoparticles when incubated with normal serum
(<50 kDa). However, the amount of high molecu- and hyperlipidemic serum, used as a model for
lar weight proteins (50–100 kDa) were observed cardiovascular diseases and obesity. Incubation
to increase. On the contrary, in the in vivo model with hyperlipidemic serum results in an increased
the amount of low molecular weight proteins amount of cholesterol adsorption on Fe3O4
(<30 kDa) were observed to be significantly nanoparticles. In this study, apart from differ-
decreased upon plasmonic heating but not thermal ences in lipid composition, differences in protein
heating. No major change in the high molecular composition were also reported. A total of 29
weight fraction (>30 kDa) was observed in the unique proteins were found to associate with
100% serum model with either plasmonic or ther- nanoparticles incubated in the lipid-rich media.
mal heating [42]. The corona formed on nanoparticles upon incu-
Elevated temperatures have been reported to bation in a lipid rich media resulted in an upregu-
bring variations in the composition of serum, lation of genes associated with inflammation and
most notable being aggregation and depletion of cell adhesion in rat aortic endothelial cells
complement proteins and immunoglobulins. The (RAECs) to a larger extent as compared to
binding of heat inactivated serum on polystyrene nanoparticles with biocorona formed after incu-
nanoparticles of 100 nm reveals an increase in bation with normal serum [44].
nanoparticle size at physiological temperature The corona formation and nanoparticle uptake
suggesting a thicker corona layer. However, no studies in vitro are usually performed in the com-
such differences were observed for smaller monly used cell culture media viz. RPMI and
nanoparticles (40 nm). These differences in the DMEM. Interestingly, in a study with gold
amount of proteins adsorbed on the nanoparticles nanoparticles, it was found that the corona for-
effect their uptake in A549 cells, with nanoparti- mation is dependent on the nanoparticle dimen-
cles of smaller sizes (20 and 40 nm) showing a sions for RPMI media. However, in case of
higher cellular uptake in non-heat inactivated DMEM, no such correlation on nanoparticle
serum as compared to heat inactivated serum. On dimensions was observed. The total amount of
the other hand, nanoparticles of larger sizes (100 hard protein corona was found to be higher in
and 200 nm), showed a complex behaviour, with nanoparticles incubated in DMEM as compared
nanoparticle to serum ratios playing an important to those incubated in RPMI media resulting in an
role. A higher cellular uptake was observed in increased toxicity of the nanoparticles in RPMI
heat inactivated serum at low nanoparticle con- media [36].
centrations and non-heat inactivated serum at Another variation in media arises due to the
high nanoparticle concentrations. Interestingly, use of plasma or serum as a protein source. The
in all the conditions tested in this study, forma- use of plasma or serum has also been shown to
tion of a protein rich corona layer was found to determine the corona composition and eventual
inhibit cellular uptake of nanoparticles [43]. This fate of nanoparticles. In a study on silver and
186 S.M. Ahsan et al.

silica nanoparticles incubated with plasma and hard protein corona from different sources viz.
serum it was shown that a similar amount of pro- human serum, human plasma, cell conditioned
tein with drastic variations in composition media from HeLa, LoVo and THP-1 cells also
adsorbed on these nanoparticles. The variation in effect cytokine production from target cells.
the corona composition led to a difference in the While the protein corona from FBS, HP and
nanoparticle uptake and cell viability with LoVo conditioned media increased the IL-1β pro-
nanoparticles incubated in plasma showing a duction and decreased MCP-1 secretion, the
higher viability and lower cellular uptake as com- presence of protein corona from HS and condi-
pared to nanoparticles incubated in serum. This tioned media from HeLa and THP-1 cells resulted
difference was attributed to the concentration of in a decreased production of IL-1β and an
apolipoprotein J (clusterin) adsorbed on the increased secretion of MCP-1 (Fig. 11.5e). These
nanoparticles upon incubation with the two fluids results suggest the complexities of the corona
[45]. composition and its consequences on nanoparti-
A comparative study on the effect of static cle activity [47].
(unconditioned) media viz. human plasma (HP),
human serum (HS), free albumin, free fibrinogen,
etc. and dynamic (conditioned) media viz. media 11.4.6 Plasma Concentration
obtained after exposure to cells, on the quantita-
tive and qualitative profile of the protein corona As a nanoparticle passes through a variety of
formed on polymeric nanoparticles has also been microenvironments, it is expected to encounter
studied (Fig. 11.5a). Since cells secrete out a plasma proteins in different concentrations. To
variety of molecules, the composition of cell-­ mimic this situation, the effect of gradient plasma
conditioned media is drastically different from on the corona composition of nanoparticles has
that obtained from a defined source [46]. been studied and compared to the corona formed
Incubation in a protein rich static media results in after incubation in plasma of defined concentra-
the adsorption of higher amount of proteins on tions. A clear difference in terms of the amount
the nanoparticle surface and leads to a larger and composition of the corona was observed
reduction in surface charge (neutralization) as [48]. A major difference in the corona composi-
compared to incubation in a dynamic in vitro tion of nanoparticles exposed to a plasma gradi-
cell-conditioned media (Fig. 11.5b, c). In general ent was the depletion of low molecular weight
the hard protein corona composition, nanoparti- proteins including apolipoprotein precursor A-1,
cle composition and the cell phenotype deter- A-II, C-I, C-II, C-III, plasma retinol-binding pro-
mines the interaction of nanoparticles with cells. tein precursor, transthyretin precursor, Isoform 1
As an example, cell conditioned media obtained of haptoglobin-related protein precursor, beta-2
from LoVo cell culture was found to inhibit the microglobulin precursor and serum amyloid A
nanoparticle association with undifferentiated protein precursor [48]. A similar observation of
and differentiated THP-1 cells to a larger extent selective enrichment of low molecular weight
than conditioned media obtained from HeLa or proteins at low plasma concentrations has been
THP-1 cell cultures, suggesting the importance made by Monopoli et al. for hydrophilic silica
of the conditioned media in dictating nanoparti- and hydrophobic sulphonated polystyrene
cle fate in vivo (Fig. 11.5d). Nanoparticles with a (PSOSO3) nanoparticles [35].

Fig. 11.5 (continued) by arrow). Numbers above the tive pro-inflammatory cytokine secretion levels by THP-1
image indicate the source from which the hard protein cells upon treatment by CAPs (green) or CSPs (blue)
coronas (hPCs) were derived. (c) Total intensity of each coated with hPCs derived from various environments (as
lane, indicating the total amount of protein in the gel. (d) indicated by the numbers above the maps) (Reprinted with
Association of PMA CAPs (green bars) or CSPs (blue permission from “Cell Conditioned Protein Coronas on
bars) coated with different hPCs with THP-1 cells, as mea- Engineered Particles Influence Immune Responses”.
sured by flow cytometry. (e) Heat maps showing the rela- Copyright (2017) American Chemical Society [47])
11  Nanoparticle-Protein Interaction: The Significance and Role of Protein Corona 187

Fig. 11.5 (a) Schematic illustration of the formation of SDS-PAGE image of corona proteins derived from vari-
protein corona on poly (methacrylic acid) (PMA) particles ous environments on core-shell particles (CSPs) and hol-
in unconditioned and cell-conditioned environments. (b) low capsules (CAPs). (Molecular weight marker is shown
188 S.M. Ahsan et al.

In general, an increase in the plasma protein the secondary binders comprise the soft corona.
concentration leads to an increased adsorption of The contribution of the hard and soft corona to
proteins on nanoparticles till the formation of a the overall thickness of the protein corona is not
monolayer [41, 49]. While the amount of protein known. However, a study by Monopoli et al. [35],
adsorbed is dependent on the plasma concentra- on sulphonated polystyrene nanoparticles
tion, increasing the plasma concentration (PSOSO3) suggests that the soft corona is less
decreases the number of proteins present in the thick in comparison to the hard corona.
corona of the nanoparticles. As an example, for
zeolite nanoparticles, an increased selective
adsorption of apolipoprotein C-III (APOC-III) 11.5.2 Effect on Zeta-Potential
and fibrinogen (FIBA, FIBB and FIBG) was
observed at 100% plasma concentration, while Apart from increasing the size of the nanoparti-
exposure to low plasma concentration (10%) cles, the adsorption of proteins on nanoparticle
resulted in the selective enrichment of surface also leads to an alteration in its zeta-­
­immunoglobulin gamma (IGHG1, IGHG2, and potential. As most plasma proteins exhibit a neg-
IGHG4) [50]. ative zeta-potential at neutral physiological pH
(7.4), the formation of protein corona on nanopar-
ticle surface imparts a zeta-potential in the range
11.5 C
 onsequences of Protein of −10 to −20 mV. This negative zeta-potential is
Corona observed irrespective of the nanoparticle physi-
cochemical properties [5].
The formation of protein corona has been
reported to play an important role in defining the
biological properties and physiological responses 11.5.3 Cellular Targeting
viz. toxicity and immunogenicity of nanoparti- of Nanoparticles
cles [51–53]. The various effects and conse-
quences of protein corona formation on the The formation of corona has been reported to
nanoparticle surface are discussed below. reduce the targeting abilities of nanoparticles. In
a study on transferrin conjugated silica nanopar-
ticles (SiO2-PEG8-Tf), it was observed that the
11.5.1 Effect on Nanoparticle Size targeting ability of transferrin, when exposed to a
biological environment (FBS), was compromised
The adsorption of plasma proteins results in the due to the shielding effect of the plasma proteins.
formation of a thick layer on the nanoparticle sur- Although, the nanoparticles were still found to
face. Various studies have reported the layer to be enter cells, their specificity for transferrin recep-
around 20–40 nm for different types of nanopar- tors was found to be decreased (Fig. 11.6) [54].
ticles. As the hydrodynamic diameters of most Apart from a loss of targeting ability, the
plasma proteins are in the range of 3–15 nm, the deposition of corona on nanoparticle surface may
thickness of protein corona formed on nanopar- also lead to undesirable localization of nanopar-
ticles suggests the existence of multiple layers of ticles resulting in unintended toxicities. Such a
adsorbed proteins. The primary binders may be case has been reported for superparamagnetic
the ones interacting with the nanoparticle surface iron oxide nanoparticles (SPIONs), where the
directly. The secondary binders then interact with deposition of protein corona leads to the crossing
the primary binders through protein-protein of the nanoparticles through the blood brain bar-
interactions. It could thus be assumed that the rier (BBB) [55]. The BBB crossing ability was
primary binders constitute the hard corona and attributed to the presence of apoliprotein,
11  Nanoparticle-Protein Interaction: The Significance and Role of Protein Corona 189

a b
100 100

Relative Mw
100 100

Relative Mw
98 98
80 96 80
96
Relative Mw

Relative Mw
0.05 0.06 0.07
60 Dapp (µm) 60 0.05 0.06 0.07
−1
0 µg ml TfR Dapp (µm)
−1
2.5 µg ml TfR
40 −1
5.0 µg ml TfR
40 No receptor (in PBS)
−1
10 µg ml TfR With TfR in PBS
−1
20 µg ml TfR
20 −1
30 µg ml TfR 20 With TfR in 10% serum
−1
40 µg ml TfR With TfR in 55% serum

0 0

0.06
0.07
0.08
0.19
0

0.2

0.4
0.5
0.6
0.3
3

4
5
0.06
0.07
0.08
0.19
0

0.2

0.4
0.5
0.6
0.3

0.0

0.0

0.0
0.0

0.0
0.0
0.0

Dapp (µm) Dapp (µm)

TfR

SiO2-PEG8-Tf
Different serum proteins

c e

d f

Fig. 11.6 (a) Apparent diameter (Dapp) as obtained by shielding effect of protein corona. Shown in the box is a
differential centrifugal sedimentation (DCS) for cartoon representation of SiO2-PEG8-Tf nanoparticles,
Tf-decorated silica nanoparticles with PEG8 spacers soluble TfR and different serum proteins. (c) Schematic
(SiO2-PEG8-Tf) incubated with increasing amounts of representation of nanoparticle binding to soluble TfR and
transferrin receptor (TfR). A shift in the Dapp with (d) cell surface TfR. (e) Schematic representation of inhi-
increasing concentrations of TfR suggests interaction of bition of nanoparticle binding to soluble TfR and (f) cell
SiO2-PEG8-Tf nanoparticles with TfR. (b) DCS assess- surface TfR in the presence of FBS proteins (Reprinted by
ment of TfR binding to SiO2-PEG8-Tf nanoparticles in permission from Macmillan Publishers Ltd.: [Nature
the presence of fetal bovine serum (FBS) proteins. With Nanotechnology] (Transferrin-functionalized nanoparti-
increasing concentration of FBS a reduction in Dapp is cles lose their targeting capabilities when a biomolecule
observed, suggesting a loss of TfR binding due to the corona adsorbs on the surface), Copyright (2013) [54])
190 S.M. Ahsan et al.

ApoA-I. The nanoparticle corona of most proteins as well as membrane lipids. Also the
nanoparticles has been shown to consist of apoli- number of proteins identified in the corona of
poproteins including ApoA-I. ApoA-I has been these nanoparticles was higher (approximately
reported to cross the BBB and hence its 800) as compared to the total proteins identified
­association with nanoparticles may lead to an in the corona of nanoparticles incubated in serum
unintended accumulation of nanoparticles in the (approximately 300). This was attributed to cel-
brain tissue. lular damage observed in the case of corona free
nanoparticles. The cellular uptake for the corona
free nanoparticles was found to be higher and the
11.5.4 Cellular Uptake nanoparticles were observed to be localized both
of Nanoparticles in the cytosol and lysosome of cells as compared
to nanoparticles with a preformed corona, which
Generally a protein corona enriched with opso- localized exclusively in the lysosomal compart-
nins and coagulation proteins is believed to acti- ment of the cells. The results clearly demon-
vate immune cells promoting phagocytosis strated differences in the nanoparticle uptake
thereby leading to the clearance of the NPs from efficiency and routes in the presence and absence
the bloodstream. Conversely, enrichment of the of protein corona on nanoparticles [58].
protein corona with dysopsonins such as human The use of polyethylene glycol (PEG) as a
serum albumin (HSA), apolipoproteins, and so stealth molecule for preventing nonspecific
forth promotes blood circulation [56]. The nanoparticle uptake has been well established.
dependence of nanoparticle uptake by macro- However, Schottler et al. reported this phenome-
phages on the adsorbed complement factors has non to be not only dependent on the PEG poly-
been reported for gold nanoparticles with differ- mer, but a consequence of the protein corona
ent hydrophobicities. Immunoglobulins were formed due to the PEGylation or polyphophoester
shown to have a negative effect on macrophage (PEEP) conjugation. The corona on PEGylated
uptake [57]. and PEEP conjugated silica nanoparticles was
It is worth mentioning that different cell types found to be enriched in apolipoprotein J (clus-
interact with nanoparticles in completely differ- terin). The binding of clusterin (Apo J) on PEG
ent ways. As an example, in a study on the cel- and PEEP was reported to be responsible for
lular uptake of silica nanoparticles by A549 cells, decreasing the macrophage uptake and providing
in the presence and absence of corona, the mem- a stealth effect (Fig. 11.7) [59].
brane association and internalization of silica The role of clusterin in preventing the macro-
nanoparticles was reported to be much higher in phage uptake of non-PEGylated silver and silica
serum free media as compared to the ones with a nanoparticles has been demonstrated by Aoyama
preformed corona. Nanoparticles recovered from et al. [45]. Nanoparticles incubated with plasma
the cell surface also developed a corona layer and serum showed variations in their corona
consisting mainly of membrane and cytoskeletal compositions and macrophage uptake. Clusterin,

Fig. 11.7 (continued) (a) Quantitative analysis of human CellMask Orange (red) and nanocarriers are shown in
plasma proteins adsorbed on different nanocarriers green; scale bars, 10 μm). (d) Classification of protein
(Polystyrene nanoparticles (PS-NH2) functionalized with corona components according to their function. (e) Heat
polyethylene glycol with degree of polymerization 44 map of the most abundant proteins in the protein corona
(PS-PEG44), and 110 (PS-PEG110), polystyrene of different nanocarriers. (f) Flow cytometry analysis of
nanoparticles functionalized with poly(ethyl ethylene cellular uptake of PS-PEEP92 by RAW264.7 cells after
phosphate) with degree of polymerization 49 (PS-PEEP49) incubation in water, human plasma, clusterin or human
and 92 (PS-PEEP92)). (b) Flow cytometry analysis of serum albumin (Reprinted by permission from Macmillan
nanoparticle uptake by RAW264.7 cells. (c) Laser scan- Publishers Ltd.: [Nature Nanotechnology] (Protein
ning microscopy images of nanoparticle uptake by adsorption is required for stealth effect of poly(ethylene
RAW264.7 cells incubated in 100% human plasma or glycol)- and poly(phosphoester)-coated nanocarriers),
DMEM without plasma (Cell membrane is stained with Copyright (2016) [59])
11  Nanoparticle-Protein Interaction: The Significance and Role of Protein Corona 191
192 S.M. Ahsan et al.

although present in the corona of nanoparticles release suggesting that the protein layer is respon-
incubated in serum and plasma, was found to be sible for the impediment of the burst release. The
more in the nanoparticles incubated in plasma. release profile of paclitaxel from Abraxane also
Nanoparticles incubated in plasma therefore revealed a similar trend with both hard corona and
showed reduction in cellular uptake. Nanoparticle soft corona playing an important role in decreas-
uptake based on PEG density has also been stud- ing the burst release from these nanoparticles [62].
ied [60]. It was found that increasing the PEG
density on gold nanoparticles led to decrease in
the adsorption of a few proteins. These proteins 11.5.6 Nanoparticle Biocompatibility
were thus speculated to play an important role in and Toxicity
inhibition of macrophage uptake. They included
complement C3, transferrin, clusterin, alpha-­2-­ The interaction of plasma proteins with nanomate-
macroglobulin, etc. [60]. rials in general increases their biocompatibility
The conformation of proteins bound to the [63]. The amount of protein corona deposited on
nanoparticle surface is also important to mediate nanomaterials directly governs their toxicity, with a
their interactions with cells. In a study, a preven- thicker layer making the nanomaterial more bio-
tion of macrophage uptake of protein-coated NPs compatible [36]. For instance the interaction of
was demonstrated for lipid and silica nanoparti- plasma proteins with graphene oxide (GO)
cles. While the corona formed on lipid nanopar- nanosheets has been shown to mitigate its cytotoxic
ticles was enriched in complement proteins, effects. The cytotoxicity of GO nanosheets arises
lipoproteins and immunoglobulins, the protein due to its direct interaction and subsequent rupture
corona of silica NPs was found to be enriched in of cellular membranes. However, the presence of
coagulation proteins and acute phase proteins. protein corona reduced this cytotoxic effect drasti-
Although the proteins identified in this study are cally in a concentration dependent manner with a
known to promote macrophage uptake through 10% serum having a more pronounced effect than
scavenger receptors, it was speculated that the 1% serum. Although GO nanosheets display a dif-
functional motifs of these proteins may not be ferential affinity towards serum proteins (in the
presented appropriately for recognition through order fibrinogen > Igs > Tf > BSA), the major role
these receptors. There is also a possibility that the of the protein corona is to prevent a direct interac-
opsonins present on these particles are shielded tion between the cell membranes and GO
by the binding of the more abundant proteins nanosheets and the composition of the corona had
such as albumin and lipoproteins, which are no major role in imparting biocompatibility [64,
known to inhibit macrophage uptake [61]. 65]. Interestingly, the cytotoxicity of silica nanopar-
ticles has been shown to depend on the time of
exposure of nanoparticles to plasma, with nanopar-
11.5.5 Effect on Drug Release ticles subjected to a prolonged plasma exposure
being relatively less toxic as compared to nanopar-
As the formation of protein corona adds a layer on ticles subjected to a brief exposure. The rapid
the nanoparticle surface it is expected to retard the corona formation on these nanoparticles also
drug release rates from nanoparticles. In case of reduced thrombocyte activation, erythrocyte aggre-
nanocapsules, the thickness and the rigidity of the gation and prevented haemolysis [9].
capsule shell is considered to be an important fac- Formation of a corona layer may also have an
tor in determining the drug release rates rather effect on the inflammatory response. As an exam-
than the protein corona, which is comparatively ple, the fibrinogen binding affinity of zeolite
less thick than the capsule shell. However, in case nanoparticles has been implicated in a potential
of polymer coated SPIONs, the burst release of the pro-inflammatory response by these particles.
drug from the polymer layer has been shown to be Although, zeolite nanoparticles were shown to
impeded by the corona layer. An increase of tem- have no negative effect on major cellular pro-
perature was however found to increase the burst cesses viz. cell cycle, oxidative stress and cellular
11  Nanoparticle-Protein Interaction: The Significance and Role of Protein Corona 193

toxicity their high affinity towards fibrinogen (an an example, the interaction of transferrin with
acute phase protein) was speculated to induce a SPIONs leads to an irreversible change in trans-
pro-inflammatory effect [66]. The immune acti- ferrin structure with a release of Fe. The transfer-
vation and cytokine expression of nanoparticles rin protein does not regain its original structure
has been reported to be dependent on the hydro- upon desorption [78]. The degree of denaturation
phobicity of the nanoparticles with a more hydro- in proteins is dependent on the nanomaterial
phobic surface eliciting a higher immune properties and the protein itself, with some pro-
response [67]. teins being more susceptible to conformation
Apart from eliciting a toxic response, nanopar- change than others [79]. Also, the surface func-
ticles may have certain indirect toxic effects by tionalization on nanoparticles may have an effect
inducing fibrillation, denaturation and conforma- on proteins, for example CdSe nanoparticles
tional changes in the adsorbed proteins, leading functionalized with oligoethylene glycol (OEG)
to an exposure of new antigenic epitopes or a loss terminated with hydroxyl groups do not interact
of function, etc. on the adsorbed proteins. It has with chymotrypsin, however carboxylate- termi-
been reported that various nanoparticles such as nated thioalkyl ligands and carboxylate-­
cerium oxide NPs, CNTs and copolymer NPs terminated OEG leads to denaturation and
induce fibrillation in human beta-2 microglobu- inhibition of enzyme activity [80]. A similar
lin. Although the mechanism of nucleation and PEGylated gold nanoparticle mediated unfolding
fibril formation is not clear, it is speculated that and aggregation has also been reported for lyso-
the interaction with nanoparticle surface leads to zyme (Fig. 11.8c) [81]. In this case, the
appropriate conformational changes that may PEGylation density was shown to play an impor-
promote fibrillation and may also increase the tant role, with a low PEGylation density resulting
local concentration of human beta-2 microglobu- in a protein aggregation and protein-Au nanopar-
lin monomers which may promote oligomer for- ticle assemblies. A high PEGylation density was
mation (Fig. 11.8a, b) [68]. Although the exact shown to inhibit protein adsorption and aggrega-
mechanism of protein unfolding at nanoparticle tion even after prolonged exposures.
surface is not known, it is expected to involve
contact forces such as the release of surface free
energy through structural reorganization [69]. 11.6 A
 ttempts on the Fabrication
Conformational changes and unfolding of of Corona Free Nanoparticles
proteins upon interaction with nanoparticle sur-
face has been a well reported phenomenon [16, The fabrication of corona free nanoparticles has
26, 70, 71]. It has been shown that nanomaterials been a long standing challenge for researchers. A
with a high surface charge density or hydropho- classical approach has been the use of polyethyl-
bicity alter the conformation of the adsorbed pro- ene glycol (PEG), that is known to prevent the
teins to a larger extent than the nanomaterials adsorption of proteins on to surfaces [82].
with hydrophilic and neutral surfaces [72–74]. However, as mentioned earlier, recent studies
The effect of nanoparticles surface charge and have revealed the adsorption of plasma proteins
hydrophobicity on protein structure has been on PEG functionalized nanoparticles [59]. An
studied for albumin, fibrinogen and cytochrome c alternative to PEG has been the utilization of
with nanoparticles having a net positive or nega- zwitterionic molecules such as amino acids and
tive surface charge or high hydrophobicity show- poly(carboxybetadiene) [83–87]. Surface func-
ing an enhanced denaturation of proteins as tionalization of silica nanoparticles with cysteine
compared to nanoparticles with a neutral surface has been reported to increase their stability in pro-
charge or high hydrophilicity [75, 76]. The con- tein (lysozyme and albumin) solutions [85]. Also,
formation changes in protein structures are usu- the functionalization of gold nanospheres with
ally irreversible even after the desorption of cysteine and lysine has been reported to reduce
proteins from the nanoparticle surface [77]. As the adsorption of proteins upon incubation with
194 S.M. Ahsan et al.

Fig. 11.8 (a) Thioflavin T fluorescence to study β2m describing the various steps of protein unfolding and
fibrillation without (black) or with 85:15 NIPAM/BAM aggregation in the presence of gold nanoparticle (AuNPs).
copolymer nanoparticles (blue) or 50:50 copolymer The first step (i) is characterized by a conformational
NIPAM/BAM nanoparticles (red). (Smaller symbols are change in the protein molecules upon adsorption onto
used for 70 nm particles and larger symbols are used for AuNPs. In the intermediate step (ii) partially unfolded
200 nm particles). (b) Transmission electron microscopy proteins on the AuNP surfaces act as a seed for further
(TEM) image of β2m fibres grown in the presence of protein aggregation. In the final step (iii), coalescence of
70 nm 85:15 NIPAM/BAM copolymer nanoparticles protein-coated AuNPs takes place (Reprinted with per-
(Reprinted with permission from “Nucleation of protein mission from “Gold Nanoparticles Can Induce the
fibrillation by nanoparticles”. Copyright (2007) National Formation of Protein based Aggregates at Physiological
Academy of Sciences [68]). c Schematic representation pH”. Copyright (2009) American Chemical Society [81])
11  Nanoparticle-Protein Interaction: The Significance and Role of Protein Corona 195

fetal bovine serum. The low interaction observed References


for zwitterion-functionalized n­anoparticles with
proteins is due to the lack of a net charge and 1. Walczyk D, Bombelli FB, Monopoli MP et al (2010)
What the cell “sees” in bionanoscience. JACS
strong hydration of these ­ molecules [86]. 132:5761–5768
Functionalization with amino acids and other 2. Monopoli MP, Åberg C, Salvati A et al (2012)
zwitterions provide an efficient mode of nanopar- Biomolecular coronas provide the biological
ticle fabrication, as most of these zwitterions pos- identity of nanosized materials. Nat Nanotechnol
7:779–786
sess functional groups for surface modifications 3. Raesch SS, Tenzer S, Storck W et al (2015) Proteomic
and prevent non specific adsorption and shielding and lipidomic analysis of nanoparticle corona upon
effect from surrounding proteins. contact with lung surfactant reveals differences
in protein, but not lipid composition. ACS Nano
9:11872–11885
4. Kapralov AA, Feng WH, Amoscato AA et al (2012)
11.7 Conclusion Adsorption of surfactant lipids by single-walled car-
bon nanotubes in mouse lung upon pharyngeal aspira-
It has become clear that the mechanism of nano- tion. ACS Nano 6:4147–4156
5. Walkey CD, Chan WC (2012) Understanding and
material interaction with biological fluids is sig- controlling the interaction of nanomaterials with pro-
nificantly different from the way in which bulk teins in a physiological environment. Chem Soc Rev
matter interacts. Such interactions greatly influ- 41:2780–2799
ence nanoparticle properties and their physiolog- 6. Vroman L, Adams A, Fischer G et al (1980)
Interaction of high molecular weight kininogen, fac-
ical functions. With the rapid advancement in tor XII, and fibrinogen in plasma at interfaces. Blood
nanoparticle mediated drug delivery in pharma- 55:156–159
ceutical sciences, it becomes essential to study 7. Aggarwal P, Hall JB, McLeland CB et al (2009)
the interaction of such nanomaterials with their Nanoparticle interaction with plasma proteins as
it relates to particle biodistribution, biocompat-
surrounding environment. A variety of nanoma- ibility and therapeutic efficacy. Adv Drug Deliv Rev
terials have been studied in this context, and it 61:428–437
could be stated that the interaction and influence 8. Vroman L (1962) Effect of adsorbed proteins on the
of biomolecules in general and proteins in par- wettability of hydrophilic and hydrophobic solids.
Nature 196:476–477
ticular is unique to the nanoparticle type and can- 9. Tenzer S, Docter D, Kuharev J et al (2013) Rapid
not be generalized. However, with the large formation of plasma protein corona critically affects
volume of data available, the biological identity nanoparticle pathophysiology. Nat Nanotechnol
and hence physiological response of nanomateri- 8:772–781
10. Gunawan C, Lim M, Marquis CP et al (2014)

als can be predicted based on the synthetic com- Nanoparticle–protein corona complexes govern
positions of the nanoparticles. Also, the design of the biological fates and functions of nanoparticles.
novel nanomaterials may be done accordingly J Mater Chem B 2:2060–2083
depending on the desired physiological response. 11. Maiolo D, Bergese P, Mahon E et al (2014) Surfactant
titration of nanoparticle–protein corona. Anal Chem
86:12055–12063
Acknowledgements SMA acknowledges Council for 12. Norde W (1994) Protein adsorption at solid sur-

Scientific and Industrial Research (CSIR) for the research faces: a thermodynamic approach. Pure Appl Chem
associate fellowship. CMR acknowledges the Department 66:491–496
of Science and Technology, India for Sir J.C. Bose 13. Saptarshi SR, Duschl A, Lopata AL (2013) Interaction
Fellowship. MFA acknowledges Dr. Chris Dealwis and of nanoparticles with proteins: relation to bio-­
NIH for supporting Postdoctoral/Senior Research associ- reactivity of the nanoparticle. J Nanobiotechnol 11:26
ate fellowship. Also, we would like to thank Uzma Nisar 14. Cedervall T, Lynch I, Lindman S et al (2007)

for proofreading. Understanding the nanoparticle–protein corona using
methods to quantify exchange rates and affinities of
proteins for nanoparticles. Proc Natl Acad Sci U S A
104:2050–2055
15. Lynch I, Dawson KA (2008) Protein-nanoparticle

interactions. Nano Today 3:40–47
196 S.M. Ahsan et al.

16. Chakraborty S, Joshi P, Shanker V et al (2011)


31. Roser M, Fischer D, Kissel T (1998) Surface-modified
Contrasting effect of gold nanoparticles and nanorods biodegradable albumin nano-and microspheres. II:
with different surface modifications on the structure effect of surface charges on in vitro phagocytosis
and activity of bovine serum albumin. Langmuir and biodistribution in rats. Eur J Pharm Biopharm
27:7722–7731 46:255–263
17. Simberg D, Park J-H, Karmali PP et al (2009)
32. Lück M, Paulke BR, Schröder W et al (1998) Analysis
Differential proteomics analysis of the surface het- of plasma protein adsorption on polymeric nanopar-
erogeneity of dextran iron oxide nanoparticles and the ticles with different surface characteristics. J Biomed
implications for their in vivo clearance. Biomaterials Mater Res A 39:478–485
30:3926–3933 33. Gessner A, Waicz R, Lieske A et al (2000)

18. Andersson J, Ekdahl KN, Larsson R et al (2002)
Nanoparticles with decreasing surface hydropho-
C3 adsorbed to a polymer surface can form an ini- bicities: influence on plasma protein adsorption. Int
tiating alternative pathway convertase. J Immunol J Pharm 196:245–249
168:5786–5791 34. Müller RH, Wallis KH, Troester SD et al (1992) In
19. Gagner JE, Lopez MD, Dordick JS et al (2011) Effect vitro characterization of poly (methyl-methaerylate)
of gold nanoparticle morphology on adsorbed protein nanoparticles and correlation to their in vivo fate.
structure and function. Biomaterials 32:7241–7252 J Control Release 20:237–246
20. Lundqvist M, Stigler J, Elia G et al (2008) Nanoparticle 35. Monopoli MP, Walczyk D, Campbell A et al (2011)
size and surface properties determine the protein Physical− chemical aspects of protein corona: rel-
corona with possible implications for biological evance to in vitro and in vivo biological impacts of
impacts. Proc Natl Acad Sci U S A 105:14265–14270 nanoparticles. JACS 133:2525–2534
21. Tenzer S, Docter D, Rosfa S et al (2011) Nanoparticle 36. Maiorano G, Sabella S, Sorce B et al (2010) Effects
size is a critical physicochemical determinant of the of cell culture media on the dynamic formation of
human blood plasma corona: a comprehensive quanti- protein− nanoparticle complexes and influence on the
tative proteomic analysis. ACS Nano 5:7155–7167 cellular response. ACS Nano 4:7481–7491
22. Cedervall T, Lynch I, Foy M et al (2007) Detailed 37. Dell'Orco D, Lundqvist M, Oslakovic C et al (2010)
identification of plasma proteins adsorbed on copoly- Modeling the time evolution of the nanoparticle-­
mer nanoparticles. Angew Chem Int Ed 46:5754–5756 protein corona in a body fluid. PLoS One 5:e10949
23. Owens DE, Peppas NA (2006) Opsonization, bio- 38. Green R, Davies M, Roberts C et al (1999) Competitive
distribution, and pharmacokinetics of polymeric protein adsorption as observed by surface plasmon
nanoparticles. Int J Pharm 307:93–102 resonance. Biomaterials 20:385–391
24. Dobrovolskaia MA, Patri AK, Zheng J et al (2009) 39. Nagayama S, Ogawara K, Fukuoka Y et al (2007)
Interaction of colloidal gold nanoparticles with human Time-dependent changes in opsonin amount associ-
blood: effects on particle size and analysis of plasma ated on nanoparticles alter their hepatic uptake char-
protein binding profiles. Nanomed Nanotechnol Biol acteristics. Int J Pharm 342:215–221
Med 5:106–117 40. Casals E, Pfaller T, Duschl A et al (2010) Time evo-
25. Deng ZJ, Mortimer G, Schiller T et al (2009)
lution of the nanoparticle protein corona. ACS Nano
Differential plasma protein binding to metal oxide 4:3623–3632
nanoparticles. Nanotechnology 20:455101 41. Mahmoudi M, Abdelmonem AM, Behzadi S et al
26. Lacerda SHDP, Park JJ, Meuse C et al (2009)
(2013) Temperature: the “ignored” factor at the nano-
Interaction of gold nanoparticles with common bio interface. ACS Nano 7:6555–6562
human blood proteins. ACS Nano 4:365–379 42. Mahmoudi M, Lohse SE, Murphy CJ et al (2013)
27. Gessner A, Lieske A, Paulke BR et al (2003)
Variation of protein corona composition of gold
Functional groups on polystyrene model nanoparti- nanoparticles following plasmonic heating. Nano Lett
cles: influence on protein adsorption. J Biomed Mater 14:6–12
Res A 65:319–326 43. Lesniak A, Campbell A, Monopoli MP et al (2010)
28. Treuel L, Brandholt S, Maffre P et al (2014) Impact Serum heat inactivation affects protein corona com-
of protein modification on the protein corona on position and nanoparticle uptake. Biomaterials
nanoparticles and nanoparticle–cell interactions. ACS 31:9511–9518
Nano 8:503–513 44. Shannahan JH, Fritz KS, Raghavendra AJ et al (2016)
29. Maffre P, Nienhaus K, Amin F et al (2011)
From the cover: disease-induced disparities in forma-
Characterization of protein adsorption onto FePt tion of the nanoparticle-biocorona and the toxicologi-
nanoparticles using dual-focus fluorescence correla- cal consequences. Toxicol Sci 152:406–416
tion spectroscopy. Beilstein J Nanotechnol 2:374–383 45. Aoyama M, Hata K, Higashisaka K et al (2016)

30. Gessner A, Lieske A, Paulke BR et al (2002)
Clusterin in the protein corona plays a key role in
Influence of surface charge density on protein adsorp- the stealth effect of nanoparticles against phagocytes.
tion on polymeric nanoparticles: analysis by two- Biochem Biophys Res Commun 480:690–695
dimensional electrophoresis. Eur J Pharm Biopharm 46. Albanese A, Walkey CD, Olsen JB et al (2014)

54:165–170 Secreted biomolecules alter the biological identity
11  Nanoparticle-Protein Interaction: The Significance and Role of Protein Corona 197

and cellular interactions of nanoparticles. ACS Nano nanocarriers: the “overlooked” factor at the nanobio
8:5515–5526 interface. Colloids Surf B Biointerfaces 123:143–149
47. Dai Q, Guo J, Yan Y et al (2017) Cell-conditioned 63. Landgraf L, Christner C, Storck W et al (2015) A
protein coronas on engineered particles influence plasma protein corona enhances the biocompatibility
immune responses. Biomacromolecules 18:431–439 of Au@ Fe3O4 Janus particles. Biomaterials 68:77–88
48. Ghavami M, Saffar S, Emamy BA et al (2013) Plasma 64. Hu W, Peng C, Lv M et al (2011) Protein corona-­
concentration gradient influences the protein corona mediated mitigation of cytotoxicity of graphene
decoration on nanoparticles. RSC Adv 3:1119–1126 oxide. ACS Nano 5:3693–3700
49. Röcker C, Pötzl M, Zhang F et al (2009) A quanti- 65. Chong Y, Ge C, Yang Z et al (2015) Reduced cyto-
tative fluorescence study of protein monolayer for- toxicity of graphene nanosheets mediated by blood-­
mation on colloidal nanoparticles. Nat Nanotechnol protein coating. ACS Nano 9:5713–5724
4:577–580 66. Laurent S, Ng E-P, Thirifays C et al (2013) Corona
50. Rahimi M, Ng E, Bakhtiari K et al (2014) Zeolite protein composition and cytotoxicity evaluation of
nanoparticles for selective sorption of plasma pro- ultra-small zeolites synthesized from template free
teins. Sci Rep 5:17259–17259 precursor suspensions. Toxicol Res 2:270–279
51. Corbo C, Molinaro R, Parodi A et al (2016) The impact 67. Moyano DF, Goldsmith M, Solfiell DJ et al (2012)
of nanoparticle protein corona on cytotoxicity, immu- Nanoparticle hydrophobicity dictates immune
notoxicity and target drug delivery. Nanomedicine response. JACS 134:3965–3967
11:81–100 68. Linse S, Cabaleiro-Lago C, Xue W-F et al (2007)
52. Miclăuş T, Beer C, Chevallier J et al (2016) Dynamic Nucleation of protein fibrillation by nanoparticles.
protein coronas revealed as a modulator of silver Proc Natl Acad Sci U S A 104:8691–8696
nanoparticle sulphidation in vitro. Nat Commun 69.
Nel AE, Mädler L, Velegol D et al (2009)
7:11770 Understanding biophysicochemical interactions at the
53. Lee YK, Choi E-J, Webster TJ et al (2015) Effect of nano–bio interface. Nat Mater 8:543–557
the protein corona on nanoparticles for modulating 70. Smith JR, Cicerone MT, Meuse CW (2009) Tertiary
cytotoxicity and immunotoxicity. Int J Nanomedicine structure changes in albumin upon surface adsorption
10:97–113 observed via fourier transform infrared spectroscopy.
54. Salvati A, Pitek AS, Monopoli MP et al (2013)
Langmuir 25:4571–4578
Transferrin-functionalized nanoparticles lose their 71. Tsai DH, DelRio FW, Keene AM et al (2011)

targeting capabilities when a biomolecule corona Adsorption and conformation of serum albumin
adsorbs on the surface. Nat Nanotechnol 8:137–143 protein on gold nanoparticles investigated using
55. Mahmoudi M, Sheibani S, Milani AS et al (2015) dimensional measurements and in situ spectroscopic
Crucial role of the protein corona for the specific tar- methods. Langmuir 27:2464–2477
geting of nanoparticles. Nanomedicine 10:215–226 72. Buijs J, Vera CC, Ayala E et al (1999) Conformational
56. Tabata Y, Ikada Y (1990) Phagocytosis of polymer stability of adsorbed insulin studied with mass
microspheres by macrophages. In: New polymer spectrometry and hydrogen exchange. Anal Chem
materials, advances in polymer science, vol 94. 71:3219–3225
Springer, Berlin, pp 107–141 73. Buijs J, Norde W, Lichtenbelt JWT (1996) Changes
57. Saha K, Rahimi M, Yazdani M et al (2016) Regulation in the secondary structure of adsorbed IgG and F
of macrophage recognition through the interplay of (ab ‘) 2 studied by FTIR spectroscopy. Langmuir
nanoparticle surface functionality and protein corona. 12:1605–1613
ACS Nano 10:4421–4430 74. Buijs J, James WT, Norde W et al (1995) Adsorption
58. Lesniak A, Fenaroli F, Monopoli MP et al (2012) of monoclonal IgGs and their F (ab′) 2 fragments onto
Effects of the presence or absence of a protein corona polymeric surfaces. Colloids Surf B Biointerfaces
on silica nanoparticle uptake and impact on cells. 5:11–23
ACS Nano 6:5845–5857 75. Roach P, Farrar D, Perry CC (2005) Interpretation of
59. Schöttler S, Becker G, Winzen S et al (2016) Protein protein adsorption: surface-induced conformational
adsorption is required for stealth effect of poly (ethyl- changes. JACS 127:8168–8173
ene glycol)-and poly (phosphoester)-coated nanocar- 76. Aubin-Tam ME, Hamad-Schifferli K (2005) Gold

riers. Nat Nanotechnol 11:372–377 nanoparticle− cytochrome C complexes: the effect
60. Walkey CD, Olsen JB, Guo H et al (2012)
of nanoparticle ligand charge on protein structure.
Nanoparticle size and surface chemistry determine Langmuir 21:12080–12084
serum protein adsorption and macrophage uptake. 77. Norde W, Giacomelli CE (2000) BSA structural

JACS 134:2139–2147 changes during homomolecular exchange between
61. Caracciolo G, Palchetti S, Colapicchioni V et al (2015) the adsorbed and the dissolved states. J Biotechnol
Stealth effect of biomolecular corona on nanoparticle 79:259–268
uptake by immune cells. Langmuir 31:10764–10773 78. Mahmoudi M, Shokrgozar MA, Sardari S et al (2011)
62. Behzadi S, Serpooshan V, Sakhtianchi R et al (2014) Irreversible changes in protein conformation due
Protein corona change the drug release profile of
198 S.M. Ahsan et al.

to interaction with superparamagnetic iron oxide poly (ethylene glycol) for stealth nanoparticles. Nano
nanoparticles. Nanoscale 3:1127–1138 Today 7:404–413
79. Kondo A, Murakami F, Higashitani K (1992) Circular 84. Yang W, Zhang L, Wang S et al (2009) Functionalizable
dichroism studies on conformational changes in pro- and ultra stable nanoparticles coated with zwitter-
tein molecules upon adsorption on ultrafine polysty- ionic poly (carboxybetaine) in undiluted blood serum.
rene particles. Biotechnol Bioeng 40:889–894 Biomaterials 30:5617–5621
80. Hong R, Fischer NO, Verma A et al (2004) Control 85. Rosen JE, Gu FX (2011) Surface functionalization of
of protein structure and function through surface silica nanoparticles with cysteine: a low-fouling zwit-
recognition by tailored nanoparticle scaffolds. JACS terionic surface. Langmuir 27:10507–10513
126:739–743 86. Murthy AK, Stover RJ, Hardin WG et al (2013)

81. Zhang D, Neumann O, Wang H et al (2009) Gold Charged gold nanoparticles with essentially zero
nanoparticles can induce the formation of protein-­based serum protein adsorption in undiluted fetal bovine
aggregates at physiological pH. Nano Lett 9:666–671 serum. JACS 135:7799–7802
82. Prime KL, Whitesides GM (1991) Self-assembled
87. Moyano DF, Saha K, Prakash G et al (2014)

organic monolayers: model systems for studying Fabrication of corona-free nanoparticles with tunable
adsorption of proteins at surfaces. Science 252:1164 hydrophobicity. ACS Nano 8:6748–6755
83. Cao Z, Jiang S (2012) Super-hydrophilic zwitterionic
poly (carboxybetaine) and amphiphilic non-ionic

You might also like