ArticlePDF AvailableLiterature Review

Nano-bio interactions: a neutrophil-centric view

Authors:

Abstract and Figures

Neutrophils are key components of the innate arm of the immune system and represent the frontline of host defense against intruding pathogens. However, neutrophils can also cause damage to the host. Nanomaterials are being developed for a multitude of different purposes and these minute materials may find their way into the body through deliberate or inadvertent exposure; understanding nanomaterial interactions with the immune system is therefore of critical importance. However, whereas numerous studies have focused on macrophages, less attention is devoted to nanomaterial interactions with neutrophils, the most abundant leukocytes in the blood. We discuss the impact of engineered nanomaterials on neutrophils and how neutrophils, in turn, may digest certain carbon-based materials such as carbon nanotubes and graphene oxide. We also discuss the role of the corona of proteins adsorbed onto the surface of nanomaterials and whether nanomaterials are sensed as pathogens by cells of the immune system.
Content may be subject to copyright.
Keshavan et al. Cell Death and Disease (2019) 10:569
https://doi.org/10.1038/s41419-019-1806-8 Cell Death & Disease
REVIEW ARTICLE Open Access
Nano-bio interactions: a neutrophil-centric
view
Sandeep Keshavan
1
, Paolo Calligari
2
, Lorenzo Stella
2
,LauraFusco
3
,LuciaGemmaDelogu
4,5
and Bengt Fadeel
1
Abstract
Neutrophils are key components of the innate arm of the immune system and represent the frontline of host defense
against intruding pathogens. However, neutrophils can also cause damage to the host. Nanomaterials are being
developed for a multitude of different purposes and these minute materials may nd their way into the body through
deliberate or inadvertent exposure; understanding nanomaterial interactions with the immune system is therefore of
critical importance. However, whereas numerous studies have focused on macrophages, less attention is devoted to
nanomaterial interactions with neutrophils, the most abundant leukocytes in the blood. We discuss the impact of
engineered nanomaterials on neutrophils and how neutrophils, in turn, may digest certain carbon-based materials
such as carbon nanotubes and graphene oxide. We also discuss the role of the corona of proteins adsorbed onto the
surface of nanomaterials and whether nanomaterials are sensed as pathogens by cells of the immune system.
Known facts
Nanomaterials are inevitably cloaked with proteins
giving rise to a bio-corona.
Nanomaterials can trigger inammation with
activation of the inammasome.
Carbon-based materials may undergo digestion by
macrophages or neutrophils.
Open questions
Does the innate immune system sense engineered
nanomaterials as pathogens?
Are nanomaterial-induced neutrophil extracellular
traps or NETs good or bad?
Can nanomaterials elicit exosome-mediated pro- or
anti-inammatory signals?
Introduction
Inammation is a complex biological response involving
soluble factors and cells that arises in a tissue in response
to harmful stimuli including pathogens, toxicants, or dead
cells. The process normally leads to recovery and healing.
However, inammation can also lead to persistent tissue
damage and may even promote neoplastic transforma-
tion
1,2
. Indeed, the distinction between acute and chronic
inammation is important, not least in toxicology.
Inammation is fundamentally a normal, protective phy-
siological response to injury or infection. However, if
inammatory responses are persistent due to an exag-
gerated or dysregulated response, including failure of
resolution of inammation, a pathological response
occurs
3
.
Understanding interactions of engineered nanomater-
ials with the immune system is of considerable relevance
both from a toxicological and biomedical perspective
4
.
However, whereas numerous publications have focused
on nanomaterial interactions with macrophages, less
attention is devoted to neutrophils, despite the fact that
neutrophils are key factors in inammation. In fact,
research in recent years has revealed that these cells may
also inform and shape adaptive immune response, in
© The Author(s) 2019
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, whi ch permits use, sharing, adaptation, distribution and reproduction
in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a linktotheCreativeCommons license, and indicate if
changes were made. The images or other third party material in this article are included in the articles Creative Commons license, unless indicated other wise in a credit line to the material. If
material is not included in the articles Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
Correspondence: Bengt Fadeel (bengt.fadeel@ki.se)
1
Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
2
Department of Chemical Sciences and Technologies, University of Rome Tor
Vergata, Rome, Italy
Full list of author information is available at the end of the article.
Edited by H.-U. Simon
Ofcial journal of the Cell Death Differentiation Association
1234567890():,;
1234567890():,;
1234567890():,;
1234567890():,;
addition to their traditional roles as hunters and killers of
microbes
5
. Furthermore, although this has previously
been overlooked, neutrophils also express a rich reper-
toire of so-called pattern recognition receptors or PRRs
6
.
We will focus here on the interactions of engineered
nanomaterials with neutrophils, the most abundant of the
white blood cells.
Nanomaterial effects on neutrophils
Neutrophils are key factors in inammation and
numerous studies have shown that engineered nanoma-
terials may elicit acute and/or chronic inammation in
different animal models
7
. However, despite numerous
studies showing tissue inltration of neutrophils upon
exposure to nanoparticles, it can be argued that neu-
trophils are a somewhat neglected cell in nanotoxicology,
as there are relatively few studies on direct interactions of
nanomaterials with these cells. Nevertheless, neutrophils
are normally the rst responders in an inammatory
reaction while macrophages arrive in the second wave of
inammation and serve mainly to remove cell debris and
to promote tissue healing
8
. Similarly, it is worth noting
that macrophages are not the only cells that are involved
in the clearance of nanoparticles from the blood; in fact, a
recent study showed that neutrophils also play a major
role in nanoparticle clearance, at least in some mouse
strains
9
. Notably, although neutrophils are cleared from
the circulation via the liver and spleen, evidence has been
put forward that the bone marrow is a major site of
neutrophil clearance
10
. It follows that nanoparticles that
are cleared from the circulation by neutrophils could end
up in the bone marrow and yet the bone marrow is fre-
quently overlooked as a possible site for the sequestration
of nanoparticles, as particle uptake by macrophages in the
liver or spleen is usually in focus
11
.
Girard and colleagues
1215
have published a series of
papers in which various metal and metal oxide nano-
particles including nanoparticles of titanium dioxide, zinc
oxide, and silver were shown to activate neutrophils and/
or to inhibit neutrophil apoptosis. In contrast, gold
nanoparticles were found to activate or accelerate neu-
trophil apoptosis
16
. Needless to say, careful attention to
endotoxin contamination of the tested particles is
required
17
. Other authors have shown that silver nano-
particles differently affect distinct subpopulations of
neutrophils
18
. Fromen et al.
19
documented interactions
between injected nanoparticles and circulating neu-
trophils, which could drive particle clearance but could
also alter neutrophil responses in a mouse model of acute
lung injury. The attachment of poly(ethylene glycol)
(PEG) onto the surface of nanoparticles is commonly
thought to prevent particle opsonization and macrophage
uptake. However, a recent study suggested, instead, that
neutrophils preferentially internalized PEGylated particles
(i.e., polystyrene microspheres) in the presence of human
plasma
20
. Notably, when the authors used model cell lines
such as HL-60 or THP-1 cultured in standard cell med-
ium supplemented with 10% fetal bovine serum, PEGy-
lation reduced uptake of the particles. However, when
these cells were cultured in human plasma, the PEGylated
particles were more avidly taken up, in line with the
results obtained with primary cells
20
. This suggests that
nanomedicine approaches based on PEGylation of nano-
particles need to be reconsideredperhaps the adminis-
tered particles with their shieldof surface-attached
polymers are being sequestered by neutrophils in the
blood? Bisso et al.
21
conducted an in-depth study of
nanomaterial interactions with human neutrophils
focusing on polymeric and liposomal particles ranging in
size from 20 nm to 5 µm. The authors found that nano-
particles were readily internalized by neutrophils ex vivo
in the absence of serum proteins, and that the inter-
nalization was size-dependent insofar as a signicant
increase in uptake of the 200 nm particles was observed
over particles < 100 nm in diameter. The inclusion of
albumin in the cell culture medium prevented uptake of
polystyrene particles and reduced the uptake of liposomal
nanoparticles, but enhanced neutrophil uptake of poly
(lactic-co-glycolic acid) (PLGA) particles. Notably,
particle-laden neutrophils (i.e., 1 µg/mL of polystyrene
particles or 5 µg/mL of liposomes) were found to undergo
normal degranulation upon stimulation with conventional
agonists
21
.
Carbon-based nanomaterials, including carbon nano-
tubes (CNTs), are widely studied in nanotoxicology
22
and
these materials were shown to trigger apoptosis and/or
autophagic cell death in macrophages (Box 1). Less is
known in regards to neutrophils. In a recent study con-
ducted in the frame of the Horizon2020 project BIOR-
IMA, the toxicity of three multi-walled CNTs
(MWCNTs) with varying physicochemical properties was
evaluated in neutrophils vs. macrophages. Macrophages
were susceptible only to the ber-like MWCNTs, but
neutrophil cell viability was signicantly affected by all
three CNTs, both long and tangled (Keshavan et al.,
manuscript in preparation). Thus, although macrophages
are capable of ingesting nanomaterials and are widely
used as a model in nanotoxicology, neutrophils should
not be ignored.
Bio-corona formation on nanoparticles
Nanomaterials promptly adsorb biomolecules leading to
the formation of a so-called bio-corona
23
. The binding of
proteins or other biomolecules to nanoparticle surfaces
may thereby afford a new identityto the nanoparticle
24
.
Deng et al.
25
showed that poly(acrylic acid)-coated gold
nanoparticles bind brinogen, a protein involved in blood
clot formation, in a charge-dependent manner, inducing
Keshavan et al. Cell Death and Disease (2019) 10:569 Page 2 of 11
Ofcial journal of the Cell Death Differentiation Association
unfolding of the protein, and that binding to integrin
receptors on the surface of the monocytic cell line, THP-1
leads to activation of the nuclear factor-κB pathway and
secretion of the pro-inammatory cytokine tumor
necrosis factor-α. In a comprehensive study using a panel
of silica and polystyrene nanoparticles of various sizes and
surface modications, Tenzer et al.
26
could show that
plasma protein adsorption occurs very rapidly, and that it
affects hemolysis, thrombocyte activation, cellular uptake,
and endothelial cell death. Vlasova et al.
27
reported that
adsorbed plasma proteins inuenced neutrophil responses
caused by polymer-coated, single-walled CNTs
(SWCNTs). Specically, the adsorption of IgG resulted in
neutrophil activation, as determined by degranulation and
release of myeloperoxidase (MPO). Similarly, protein
adsorption modulated neutrophil responses toward car-
boxylated, non-PEGylated SWCNTs
28
. Lara et al.
29
employed an immuno-mapping technique to study epi-
tope presentation of two major proteins in the serum
corona, low-density lipoprotein (LDL) and
immunoglobulin G. The authors could show that both
proteins displayed functional motifs allowing for recog-
nition of the bio-corona on silica nanoparticles by LDL
receptor and Fc-γreceptor I, respectively. On the other
hand, others have pointed out that the bio-corona could
shield targeting ligands on nanoparticles
30
. However,
recent studies suggested clever ways in which to cir-
cumvent this problem
31,32
. Furthermore, purposeful sur-
face modication of CdSe/ZnS quantum dots to induce a
protein misfolding event in the bio-corona enabled
receptor-mediated endocytosis of the particles
33
. Bisso
et al.
21
reported that the presence of serum reduced the
ex vivo uptake of poly(styrene) nanoparticles and lipo-
somes by neutrophils and enhanced the uptake of micro-
and nanosized PLGA particles. However, the composition
of the bio-corona and the role of specic proteins for
neutrophil uptake, or lack thereof, was not examined.
Furthermore, neutrophils were found to preferentially
internalize PEGylated particles
20
. The authors noted that
this is linked to factor(s) in human plasma and provided
some evidence for a role of complement. Complement
factors also facilitate phagocytosis of apoptotic cells and
microbes
4
.
Viruses are natural nanoparticles and it is not surprising
that a bio-corona of proteins may form on viruses in
various biological uids, or that the bio-corona may affect
immune responses to viruses
34
. Indeed, the adsorbed bio-
corona could be considered as part of the motifs that are
sensed by immune cells. Hence, the boundaries between
pathogen-associated molecular patterns (PAMPs) and
damage-associated molecular patterns (DAMPs) appar-
ently begin to dissolve at the nano-bio interface, as both
engineered and natural nanoparticles (i.e., viruses) adsorb
host proteins on their surface. This topic is discussed in
more detail below.
Exosomes: message in a bottle
Exosomes are nanosized extracellular vesicles that are
naturally secreted by cells and they may play a particularly
important role in conveying information between
immune cells
35
. Exosomes were initially thought to fulll
a janitorial function by providing the cell with a means of
getting rid of non-functional proteins and other mole-
cules. Subsequent studies suggested important roles of
exosomes in intercellular communication and the interest
in exosomes and other microvesicles has escalated in the
past two decades due to their emerging roles in health and
disease
36
. Exosomes thus harbor specic proteins and
nucleic acids, mostly small RNAs, such as ribosomal RNA,
transfer RNA, microRNA, and mRNA molecules
36
. Exo-
somes from activated neutrophils were recently reported
to acquire surface-bound neutrophil elastase (NE) and the
exosomes were shown to degrade extracellular matrix
components causing the hallmarks of chronic obstructive
Box 1 Cell death: implications for nanotoxicology
The Nomenclature Committee on Cell Death recently provided
guidelines for molecular denitions of various cell death
modalities
128
, although this is hardly the rst nor the last attempt
at dening cell death
129
. It is important to bear in mind that
nanomaterials can elicit different modes of cell death depending
on the material as well as on the cell type, and that the mitigation
of the adverse effects of such materials requires the correct
diagnosis of cell death
130
.
Apoptosis: Regulated cell death precipitated by caspases, either
through plasma membrane receptor ligation (extrinsic apoptosis)
or via perturbation of mitochondria (intrinsic apoptosis). Nano-
materials such as CNTs trigger apoptosis, for instance in lung
cells
131
, but chronic, low-dose exposure may result in apoptosis
resistance and oncogenic transformation
132
.
Pyroptosis: Regulated cell death associated with the formation of
pores in the plasma membrane by gasdermin proteins, usually
provoked by caspase-1 activation
133
. Rare earth metal nanopar-
ticles were found to trigger pyroptosis in macrophages and
apoptosis in hepatocytes
134
.
Necroptosis: Regulated cell death that transpires with RIP1/RIP3
activation and subsequent plasma membrane permeabilization
by MLKL (mixed-lineage kinase domain-like pseudokinase)
135
.
Few, if any, studies have reported evidence of nanoparticle-
induced necroptosis.
Ferroptosis: Novel, iron-dependent cell death characterized by
lipid peroxidation that is subject to regulation by GPX4
(glutathione peroxidase 4) and tightly linked to glutathione
synthesis
136
. Importantly, a recent study revealed that nanopar-
ticles trigger different forms of cell death (i.e., apoptosis or
ferroptosis) depending on subtle variations in nanoparticle
surface properties
137
.
Autophagic cell death: Cell death that requires components of the
autophagic machinery (note that autophagic cell death should
not be confused with autophagy, a cell survival mechanism).
Nanoparticles with a high propensity to release toxic ions may
harness autophagy to trigger cell death
138
.
Keshavan et al. Cell Death and Disease (2019) 10:569 Page 3 of 11
Ofcial journal of the Cell Death Differentiation Association
pulmonary disease
37
. Exosomal NE was much more
potent than free NE. In the latter study, neutrophils were
activated with the bacterial peptide, formyl-methionine-
leucine-phenylalanine (fMLP), a known neutrophil ago-
nist. Whether or not such exosome-mediated pathological
responses occur following nanomaterial exposure merits
close attention; several studies have shown that CNTs
cause pulmonary inammation as well as airway remo-
deling
38
. In a seminal study, Zhu et al.
39
reported that
exosomes were generated in signicant numbers in the
lungs of mice exposed to iron oxide nanoparticles, and
noted that the exosomes were quickly transferred to the
systemic circulation, thereby conveying immune activa-
tion in extrapulmonary organs. The authors inferred that
the exosomes were of macrophage origin, although fur-
ther studies are warranted to discern the source of
nanoparticle-induced exosomes and whether neutrophils
are also involved. Studies have shown that different metal
or metal oxide nanoparticles may elicit varying cellular
patterns of inammation and one cannot a priori assume
that macrophages are the only cell type at play
40
.In
another recent study, ZnO nanoparticles were found to
trigger neutrophilic inammation in rats and numerous
microRNAs were shown to be selectively up- or down-
regulated in serum exosomes from ZnO-exposed animals
when compared with controls
41
. Using single-particle
inductively coupled plasma-mass spectrometry and other
techniques, Logozzi et al.
42
reported that primary human
macrophages are capable of endocytosis of gold nano-
particles (20 nm) with subsequent discharge of the
nanoparticles via exosomes. Further studies are required
to determine whether this is a general phenomenon and
to what extent the exosomal content of nanoparticles
correlates with the delivered dose. Nevertheless, it is
conceivable that exosomes could be exploited as bio-
markers of exposure to nanoparticles
42
.
Neutrophil traps: a necessary nuisance?
Brinkmann et al.
43
reported 15 years ago that neu-
trophils kill pathogens extracellularly by releasing so-
called neutrophil extracellular traps or NETs. NETs are
comprised a backbone of nuclear chromatin decorated
with antimicrobial proteins such as MPO and NE. In
addition to the classical or most commonly studied form
of NADPH oxidase-dependent NETs, which contain
nuclear chromatin, some studies have shown that neu-
trophils under certain conditions release NETs compris-
ing mitochondrial DNA
44,45
.
NET formation is frequently viewed as a specialized
form of neutrophil cell death that is distinct from apop-
tosis and necrosis
46
, and this cell death has been dubbed
NETosis. This has led to some confusion in the literature,
as the term NETosis is commonly equated with NET
formation, and to further compound the situation, some
authors refer to vital NETosis
47,48
. Indeed, as the terms
suicidal and vital NETosis are controversial, it is advisable
to simply refer to neutrophil formation of NETs with or
without attendant cell death. On the other hand, it is well
established that the stimulation of neutrophils with
phorbol 12-myristate 13-acetate (PMA) leads to a cas-
pase-independent, non-apoptotic form of cell death
49,50
.
Recent studies suggest some commonalities between NET
formation and other forms of programmed cell death
(Box 1). Hence, gasdermin D, a pore-forming protein and
a key executor of pyroptosis, is required for NET forma-
tion in neutrophils stimulated with PMA
51,52
. Further-
more, anti-neutrophil cytoplasmic antibodies trigger
NETs via receptor-interacting protein kinase 1/3 and
MLKL, key factors in necroptosis
53
, although it is
important to note in this context that different stimuli
may trigger different pathways of NET formation
54,55
.
NETs are thought to play a role during infection by
allowing neutrophils to capture and kill pathogens
extracellularly
43,56
. However, mounting evidence suggests
that uncontrolled or excessive production of NETs, or
defective degradation or removal of NETs, is related to the
exacerbation of inammation and the development of
several diseases
57
. Hakkim et al.
58
reported that impair-
ment of DNaseI-mediated NET degradation is associated
with systemic lupus erythematosus. Excessive formation
of NETs, on the other hand, could clog blood vessels and
provide a scaffold for thrombus formation
59
, whereas a
recent study has shown that both DNase1 and DNase1-
like 3 are capable of degrading NETs in circulation
60
.We
found that NETs are handled differently by macrophages
and dendritic cells with LL-37-dependent uptake followed
by intracellular degradation in the former case, and
extracellular, DNase1L3-mediated degradation of NETs in
the latter case (Lazzaretto et al., manuscript in prepara-
tion). NETs were shown to prime T cells and reduce the
activation threshold to specic antigens
61
. TLR9, an
intracellular sensor that functions to alert the immune
system of viral and bacterial infections by binding to
DNA, was not involved. Nevertheless, this suggests that
NETs may serve as a link between the innate and adaptive
immune system. Furthermore, and in support of this
notion, a recent study showed that deposition of cell-free
DNA through neutrophil formation and ejection of NETs
occurs at the site of immunization and drives the activity
of aluminum adjuvant (alum), thereby enhancing
adjuvant-induced adaptive immune responses
62
.
Can nanomaterials trigger NETs? Early work suggested
that rod-shaped gold nanoparticles, as well as cationic lipid
nanoparticles, are capable of triggering NETs, but compel-
ling evidence was not presented as it is difcult to distin-
guish between neutrophil cell death with (passive) release of
intracellular contents vs. the production of NETs
63,64
.
Naturally, endotoxin contamination also needs to be
Keshavan et al. Cell Death and Disease (2019) 10:569 Page 4 of 11
Ofcial journal of the Cell Death Differentiation Association
excluded. More recently, agglomerates of endotoxin-free
superparamagnetic iron oxide nanoparticles (SPIONs) were
shown to elicit NETs, albeit at a very high concentration
(200 µg/mL)
65
. Importantly, stabilization of the SPIONs
with human serum albumin prevented NET formation. The
authors also found that agglomerates of SPIONs triggered
NET formation in vivo in an animal model, and that the
particles were gluedtogether by the NETs, and they
suggested that such SPION-NET co-aggregates might
occlude blood vessels
65
. The study highlights the need for
careful particle design and passivation strategies to make
nanoparticles safe for intravenous use. Muñoz et al.
66
reported that nanodiamonds (10nm)causeplasmamem-
brane damage and signs of lysosomal instability in neu-
trophils, and found that these nanoparticles triggered the
formation of NETs at high concentrations of nanoparticles
(200 µg/mL). In contrast, larger particles (1001000 nm)
were relatively inert. The smaller particles also triggered
inammation following subcutaneous injection of a high
dose (1 mg) into the foot pads of mice, but the swelling was
resolved after 12weeks
66
.Therelevanceofthesendings
is difcult to judge due to the high doses that were applied.
It is worth noting that nanodiamonds were recently shown
to be well-tolerated in sub-acute and chronic duration
studies in rodents and non-human primates following
intravenous injections
67
.
We recently provided rst evidence that graphene oxide
(GO) can trigger NETs in primary human neutrophils,
indicating that the immune system can senseeven a
two-dimensional material (Fig. 1). Hence, we could show
that low doses of micrometer-sized GO sheets triggered
NETs in primary human neutrophils more effectively than
small GO with nanosized lateral dimensions
68
. Both
materials were produced under sterile conditions and
were proven to be endotoxin-free. Interestingly, the large
GO sheets initiated the oxidation of cholesterol in the
plasma membrane of neutrophils as evidenced by time-of-
ight secondary ion mass spectrometry (ToF-SIMS) fur-
thermore, the release of NETs was reduced by Trolox, a
potent lipid antioxidant
68
. Neumann et al.
69
previously
reported that methyl-β-cyclodextrin (MβCD), a
cholesterol-depleting agent, triggered the formation of
NETs in a manner that was independent of the NADPH
oxidase (i.e., insensitive to pharmacological inhibition
using diphenylene iodonium [DPI]). To study the signal-
ing pathway underlying the formation of NETs in GO-
exposed cells, we explored the effect of DPI on NET
formation in neutrophils exposed to GO, PMA, or MβCD.
DPI was found to block PMA-induced production of
NETs, as expected, and blocked the production of NETs
in neutrophils exposed to the small GO sheets
68
. How-
ever, NET formation in MβCD-treated cells and in cells
incubated with large GO was unaffected by DPI, indicat-
ing that NADPH oxidase activation is not required.
Interestingly, the mitochondria-targeted antioxidant
MitoTEMPO signicantly reduced the production of
NETs by GO. Mitochondrial reactive oxygen species
(ROS) are also required for calcium ionophore-induced
NETs
70
. In a companion paper, we showed that small and
large GO are degraded in an MPO-dependent manner in
NETs puried from activated neutrophils
71
. Neutrophils
can also enzymatically digest CNTs (discussed below).
Thus, it appears that neutrophils are capable of handling
at least some carbon-based nanomaterials as pathogens,
leading to the destruction of the offending agents
72
.
Neutrophil degradation of nanomaterials
The membrane-bound NADPH oxidase generates ROS
that are instrumental for the killing of ingested pathogens.
Degranulation with the release of MPO is also an
important feature of the microbicidal actions of neu-
trophils
73
. In addition to its role in antimicrobial defense,
MPO is also reported to be involved in the degradation of
CNTs and the clearance of CNTs from the lungs was
markedly less efcient in MPO-decient mice when
GO
NET
GO
A
B
C
D
E
Fig. 1 Neutrophils capture graphene oxide sheets in extracellular
traps. acConfocal images of neutrophils incubated in the presence
of large GO sheets (12.5 μg/mL). Cells were stained with antibodies to
neutrophil elastase (NE) followed by a secondary FITC-labeled
antibody (green) and counterstained with DAPI (blue) for visualization
of cell nuclei. dLight and uorescence microscopy images
superimposed to show the presence of GO. eScanning electron
microscopy (SEM) image of neutrophils exposed to GO (12.5 μg/mL).
The arrow points to a large GO sheet that has been capturedin a
network of chromatin bers (i.e., NETs). Reproduced from Mukherjee
et al.
68
, with permission from Elsevier
Keshavan et al. Cell Death and Disease (2019) 10:569 Page 5 of 11
Ofcial journal of the Cell Death Differentiation Association
compared with wild-type mice
74
. Neutrophil-mediated
destruction of CNTs was rst described by Kagan et al.
75
.
Subsequently, MPO-dependent degradation of PEGylated
CNTs was reported and this was suggested to occur in a
two-step process whereby the polymers were rst
removed from the CNTs by NE followed by the degra-
dation of the CNTs themselves by MPO
76
. Eosinophils
can also digest CNTs
77
. Furthermore, CNTs and GO were
shown to undergo acellular degradation in NETs puried
from activated neutrophils
71,78
(Fig. 2). In a recent study,
GO functionalized with fMLP was shown to stimulate
neutrophil degranulation leading to degradation (Martin
et al., manuscript in preparation). Even graphene can
undergo neutrophil-mediated degradation, although the
process is considerably slower when compared with GO
79
.
The latter studies were conducted ex vivo using human
neutrophils. Notably, a recent in vivo study has shown
that GO (20 mg/kg) administered subcutaneously elicits
an inammatory reaction in mice in response to
implantation consistent with a foreign body reaction
80
.
The latter study did not evaluate biodegradation of the
implanted materials. Girish et al.
81
, on the other hand,
explored biodegradation after intravenously injecting
graphene (20 mg/kg) into mice by using a Raman confocal
imaging approach. The authors noted graphene engulf-
ment by tissue-bound macrophages and found that
degradation was prominent after 90 days. Tuning the
properties of graphene-based materials to achieve optimal
performance while maintaining an acceptable degree of
biocompatibility and biodegradability remains an impor-
tant challenge in the eld
82
.
Inammasomes: double-edged swords
How do nanomaterials and other exogenous substances
trigger inammation? Numerous studies have shown that
the inammasome, originally described by Tschopp and
colleagues
83
, is a key signaling hub that regulates innate
immunity and inammation
84
. The inammasomes are
multiprotein complexes that are activated in response to
diverse pathogen- and host-derived danger signals leading
to the activation of caspase-1 with processing of cytosolic
pro-IL-1βand secretion of pro-inammatory IL-1β
85
.
NLRP3, in particular, responds to a diverse array of dif-
ferent stimuli including crystalline and particulate matter
such as uric acid crystals, silica, asbestos, and alum, as well
as to pathogens
8689
. NLRP3 is also a sensor of various
nanomaterials
90
. Indeed, not only long and rigid CNTs
but also ultrathin GO sheets and spherical carbon parti-
cles are able to trigger NRLP3-dependent IL-1βsecretion
in human macrophages
9193
(Fig. 2). However, less is
known regarding inammasome activation in neutrophils.
Neutrophils are capable of activating the NLRC4 inam-
masome in response to bacterial challenge, but this occurs
without the induction of pyroptosis
94
. On the other hand,
NE cleaves gasdermin D in neutrophils
95
, thus providing
an alternative route to pyroptosis in these cells. Indeed, it
should be noted that the production of IL-1βis not
exclusively dependent on caspase-1
96
. Thus, neutrophil-
derived serine proteases are also involved in the proces-
sing of IL-1 family cytokines, and serine proteases and/or
caspases may be involved in neutrophils depending upon
the stimulus
97
.
How big is a speck? ASC (apoptosis-associated speck-like
protein containing a CARD) is a CARD (caspase
cytokines
exosomes
PEG
cytokines
ASC
NLRP3
pro-casp-1
pro-IL-1β IL-1β
NETs
MPO
PC
NE
O2O2·-
NADPH NADP+ + H+
e-
H2O2HOCl
MPO
NADPH oxidase
inflammasome
NOX
NOX
iNOS
NO ONOO-
iNOS
Fig. 2 Nano-bio interactions: from coronation to degradation.
This schematic diagram depicts nanoparticles with or without a
protein corona (PC) and/or a polymer coating (i.e., poly(ethylene
glycol) or PEG) interacting with neutrophils (left) vs. macrophages
(right). Neutrophils release neutrophil extracellular traps (NETs)
consisting of nuclear chromatin decorated with granule proteins such
as myeloperoxidase (MPO), and recent studies have shown that
carbon nanotubes (CNTs) and graphene oxide (GO) are captured and
digested in NETs in an MPO-dependent manner
71,78
. The NADPH
oxidase (commonly abbreviated as NOX) is a multiprotein complex
expressed in phagocytes that catalyzes the generation of superoxide.
Superoxide, in turn, dismutates to form hydrogen peroxide and MPO
catalyzes the formation of hypochlorous acid, a freely diffusible
oxidant that is microbicidal and also is responsible for the degradation
of carbon-based nanomaterials
124
. In addition, superoxide and nitric
oxide, produced by inducible nitric oxide synthase (iNOS), react to
form peroxynitrite, which was shown to digest nanomaterials in
macrophages
125
. Macrophages emit pro-inammatory IL-1βthrough
an inammasome-dependent mechanism (a cytosolic protein
complex shown outside the cell for clarity). Neutrophils and
macrophages release exosomes, thus providing a further means of
cell-to-cell communication and propagation of inammation. Recent
work has shown that neutrophil-derived exosomes express neutrophil
elastase (NE) on the surface; these exosomes degrade extracellular
matrix more readily when compared with free NE
37
Keshavan et al. Cell Death and Disease (2019) 10:569 Page 6 of 11
Ofcial journal of the Cell Death Differentiation Association
recruitment domain) carrying protein of 22 kDa that is
involved in the recruitment of pro-caspase-1 to the
inammasome. ASC was described 20 years ago as a protein
that could be visualized as a small spot or speck in the
cytosol of apoptotic cells
98
. Intriguingly, although inam-
masome activation was originally believed to take place in
the cytosol of cells, subsequent studies have shown that cells
may transmit inammation in a prion-like manner via
extracellular ASC. These micrometer-sized clumps of ASC
proteins continued to stimulate caspase-1 activation extra-
cellularly and stimulated further inammasome activation
in neighboring macrophages that had ingested the ASC
oligomers
99,100
. Extracellular ASC was found in tissues of
patients with inammatory diseases and autoantibodies to
ASC developed in some patients with autoimmune
pathologies. Thus, as pointed out previously, danger signals
come in many shapes and sizes
101
.Itshouldthereforenot
come as a surprise that the immune system is capable of
responding to synthetic (nano)particles.
Decoding danger at the nanoscale
Cells of the immune system are equipped with PRRs
that monitor the extracellular or intracellular environ-
ment for signs of infection or danger. Toll-like receptors
(TLRs) are present both on cell surfaces and in endosomal
compartments, whereas retinoid acid-inducible gene-I-
like receptors and nucleotide-binding and oligomerization
domain-like receptors (NLRs) are present in the cyto-
sol
102
. Furthermore, soluble scavenging receptors have
been described
103,104
. PRRs presumably evolved to dis-
criminate between foreign intruders and self,but they
also recognize DAMPs released from stressed or damaged
cells
105
. It has been estimated that a single cell may
express as many as 50 distinct PRRs, thus testifying to the
importance of sensing danger
106
. Nanomaterialswith
or without a corona of proteins or other biomolecules
may be considered as a particular case of danger signals
that are able to trigger sterile inammatory responses.
Indeed, we have previously postulated that engineered
nanomaterials may present nanomaterial-associated
molecular patterns or NAMPs to cells of the immune
system
107
. Thus, in analogy with microorganisms (bac-
teria, viruses) displaying PAMPs and damaged or stressed
cells releasing DAMPs, we postulated that engineered
nanomaterials coated with a corona of biomolecules may
act as nanoparticle-associated molecular patterns or
NAMPs
107
. The notion of nanomaterial-associated
molecular patterns has captured the attention of several
other authors
108111
. The fact that nanoparticles with an
adsorbed corona of proteins may display epitopes that are
sensed by the immune system is of considerable interest,
as this may point toward a systematic understanding of
nano-bio interactions
112
. It may also be worthwhile to
explore whether nanoparticles per se present molecular
patterns that are decoded by the immune system. Indeed,
emerging studies suggest that some nanoparticles could
act as protein mimics capable of engaging with intra- or
extracellular receptors
113
, and the combination of
experimental and theoretical studies promises to shed
light on this exciting topic
114,115
. Using a proteomics
approach, He et al.
116
found that carbon-based nanoma-
terials (i.e., single-walled carbon nanohorns, SWCNTs,
and MWCNTs) bound to glycoprotein nonmetastatic
melanoma protein B (GPNMB, also known as osteoacti-
vin) in macrophages. The authors suggested that GPNMB
serves as an intracellular PRR for these nanomaterials. We
have recently demonstrated, by using a transcriptomics
approach, that SWCNTs prompted the upregulation and
secretion of chemokines in primary human macrophages,
and we provided evidence for direct binding of CNTs to
TLR2/4
117
. The nanomaterials used were endotoxin-free.
Taken together, these results give credence to the idea
that nanomaterials may act as NAMPs
107
. Interestingly,
computational studies predicted that the binding of
carbon-based nanostructures to proteins is guided mainly
by hydrophobic interactions
114
. More specically, we and
others have analyzed the association of CNTs and other
carbon nanostructures to TLRs
117,118
. Turabekova et al.
118
predicted that the hydrophobic pockets of some TLRs
might be capable of binding pristine SWCNTs and C
60
fullerenes (Fig. 3). Furthermore, we suggested that ion-
pair interactions with positively charged residues might
strengthen the binding of carboxylated CNTs to TLRs
117
.
It will certainly be important to study whether engineered
nanomaterials also engage with neutrophil PRRs.
It is worth noting that nanoparticles can also be
exploited for the removal of DAMPs such as cell-free
DNA that is expelled from dying cells to ameliorate
inammatory diseases initiated by the inappropriate
activation of TLR signaling. Hence, Liang et al.
119
pre-
pared cationic nanoparticles composed of the block
copolymer of PLGA and poly(2-(diethylamino)ethyl
methacrylate), and found that these particles had a high
DNA-binding capacity. Furthermore, when injected
intravenously the cationic nanoparticles could alleviate
symptoms in animal models of arthritis. These results,
along with previous work by other investigators, suggest
that cationic nanoparticles may act as nucleic acid sca-
vengers
120,121
. Further studies are needed to formally
address whether the acquisition of a protein corona on
scavenger particles navigating the blood stream would
interfere with or promote nucleic acid binding.
Concluding remarks
Neutrophils are key effector cells of the innate arm of
the immune system and play important roles in host
defense against pathogens, and yet, paradoxically, they are
also involved in numerous pathological conditions
Keshavan et al. Cell Death and Disease (2019) 10:569 Page 7 of 11
Ofcial journal of the Cell Death Differentiation Association
characterized by chronic inammation. Studies in recent
years have shown that nanomaterials can modulate and
activate neutrophils and other immune cells. Moreover,
activated neutrophils may capture and digest certain
carbon-based nanomaterials. Neutrophils also play a role
in particle clearance in the systemic circulation (at least in
mice). Understanding the interactions between nanoma-
terials and neutrophils is important for the development
of safe and effective nanomaterials for biomedical
applications.
The nanotoxicology literature is replete with publica-
tions on the negative impact of nanomaterials, often
referencing the pro-inammatory effects of the materials,
even when studies are performed in cell culture where
coordinated immune reactions cannot occur. However, it
is important to note that inammation as such is not a
detrimental response. Therefore, one should not seek to
prevent (acute) inammation at every cost. Instead,
careful design of nanomaterials is required in order to
avoid chronic, adverse reactions. Furthermore, nanoma-
terials may be exploited to harness immune responses to
ameliorate chronic inammation and/or autoimmune
diseases, and leverage immune responses toward cancer
cells
122,123
.
Acknowledgements
The work is supported by the European Commissions H2020 program through
BIORIMA (grant agreement number 760928) and the Graphene Flagship (grant
agreement number 785219), and the Swedish Research Council (grant
agreement number 2016-02040). L.D. acknowledges the support of the Marie
Skłodowska-Curie Actions Individual Fellowship IMM-GNR (grant agreement
number 797914).
Author details
1
Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
2
Department of Chemical Sciences and Technologies, University of Rome Tor
Vergata, Rome, Italy.
3
Department of Chemical and Pharmaceutical Sciences,
University of Trieste, Trieste, Italy.
4
Department of Biomedical Sciences,
University of Padua, Padua, Italy.
5
Fondazione Istituto di Ricerca Pediatrica Città
della Speranza, Padua, Italy
Author contributions
B.F. wrote the paper with input from S.K., P.C., L.S., L.F., and L.D. P.C. and L.S.
performed docking studies. L.F. prepared the schematic gure. All co-authors
approved the nal version of the paper.
Conict of interest
The authors declare that they have no conict of interest.
TLR3
RNA
CNT
TLR4TLR4
BA
DC
TLR1
TLR2
Fig. 3 Macrophage sensing of single-walled carbon nanotubes as pathogens. Computational modeling of a SWCNT bound to the extracellular
domains of TLR1/TLR2 (a). The nanotube is surrounded by amino acids of mostly hydrophobic nature giving rise to strong van der Waals interactions
(b). The results shown in cdepict one of the best binding poses obtained by molecular docking of carboxylated SWCNTs with TLR4
117
. Interestingly,
the highest scoring binding mode of SWCNT and TLR4 shared several similarities with the experimentally resolved structure of TLR3 in complex with
double-stranded RNA (d)
126
. These ndings suggest that TLR4 homodimers may engage with SWCNTs through a tweezer-like mechanism. It is noted
that these modeling results were derived in the absence of a protein corona in order to elucidate direct binding to TLRs. The efciency of protein
adsorption is well-known to be proportional to the diameter of the nanotubes
127
. Therefore, the small diameter of these SWCNTs may limit protein
adsorption, thus leaving a sufcient surface for the direct interaction with TLRs
117
. Panel aand bare from Turabekova et al.
118
with permission from
The Royal Society of Chemistry. Results shown in panel care from ref.
117
while results in panel d were generated based on ref.
126
Keshavan et al. Cell Death and Disease (2019) 10:569 Page 8 of 11
Ofcial journal of the Cell Death Differentiation Association
Publishers note
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional afliations.
Received: 7 June 2019 Revised: 4 July 2019 Accepted: 9 July 2019
References
1. Nathan, C. Points of control in inammation. Nature 420,846852 (2002).
2. Medzhitov, R. Origin and physiological roles of inammation. Nature 454,
428435 (2008).
3. Bhattacharya,K.,Andón,F.T.,El-Sayed,R.&Fadeel,B.Mechanismsofcarbon
nanotube-induced toxicity: focus on pulmonary inammation. Adv. Drug
Deliv. Rev. 65,20872097 (2013).
4. Boraschi, D. et al. Nanoparticles and innate immunity: new perspectives on
host defence. Semin. Immunol. 34,3451 (2017).
5. Nathan,C.Neutrophilsandimmunity: challenges and opportunities. Nat. Rev.
Immunol. 6,173182 (2006).
6. Thomas,C.J.&Schroder,K.Patternrecognitionreceptorfunctioninneu-
trophils. Trends Immunol. 34,317328 (2013).
7. Leso, V., Fontana, L. & Iavicoli, I. Nanomaterial exposure and sterile inam-
matory reactions. Toxicol. Appl. Pharmacol. 355,8092 (2018).
8. Underhill, D. M. & Goodridge, H. S. Information processing during phago-
cytosis. Nat. Rev. Immunol. 12,492502 (2012).
9. Jones, S. W. et al. Nanoparticle clearance is governed by Th1 / Th2 immunity
and strain background. J. Clin. Invest. 123, 30613073 (2013).
10. Furze,R.C.&Rankin,S.M.Theroleofthebonemarrowinneutrophil
clearance under homeostatic conditions in the mouse. FASEB J. 22,
31113119 (2008).
11. Tavares,A.J.etal.Effectofremoving Kupffer cells on nanoparticle tumor
delivery. Proc. Natl Acad. Sci. USA 114, E10871E10880 (2017).
12. Gonçalves, D. M., Chiasson, S. & Girard, D. Activation of human neu-
trophils by titanium dioxide (TiO
2
) nanoparticles. Toxicol In Vitro.24,
10021008 (2010).
13. Babin,K.,Antoine,F.,Goncalves,D.M.&Girard,D.TiO
2
,CeO
2
and ZnO
nanoparticles and modulation of the degranulation process in human
neutrophils. Toxicol. Lett. 221,5763 (2013).
14. Goncalves, D. M. & Girard, D. Zinc oxide nanoparticles delay human
neutrophil apoptosis by a de novo protein synthesis-dependent and
reactive oxygen species-independent mechanism. Toxicol In Vitro. 28,
926931 (2014).
15. Babin, K., Goncalves, D. M. & Girard, D. Nanoparticles enhance the ability of
human neutrophils to exert phagocytosis by a Syk-dependent mechanism.
Biochim. Biophys. Acta 1850, 22762282 (2015).
16. Noël, C., Simard, J. C. & Girard, D. Gold nanoparticles induce apoptosis,
endoplasmic reticulum stress events and cleavage of cytoskeletal proteins in
human neutrophils. Toxicol In Vitro. 31,1222 (2016).
17. Gonçalves, D. M. & Girard, D. Evidence that polyhydroxylated C
60
full-
erenes (fullerenols) amplify the effect of lipopolysaccharides to induce
rapid leukocyte inltration in vivo. Chem. Res. Toxicol. 26, 18841892
(2013).
18. Fraser, J. A. et al. Silver nanoparticles promote the emergence of hetero-
geneic human neutrophil sub-populations. Sci. Rep. 8,114 (2018).
19. Fromen, C. A. et al. Neutrophil-particle interactions in blood circulation drive
particle clearance and alter neutrophil responses in acute inammation. ACS
Nano. 11, 1079710807 (2017).
20. Kelley,W.J.,Fromen,C.A.,Lopez-Cazares,G.&Eniola-Adefeso,O.PEGylation
of model drug carriers enhances phagocytosis by primary human neu-
trophils. Acta Biomater. 79, 283293 (2018).
21. Bisso, P. W., Gaglione, S., Guimarães, P. P. G., Mitchell, M. J. & Langer, R.
Nanomaterial interactions with human neutrophils. ACS Biomater. Sci. Eng. 4,
42554265 (2018).
22. Bhattacharya, K. et al. Biological interactions of carbon-based nanomaterials:
from coronation to degradation. Nanomedicine 12,333351 (2016).
23. Barbero, F. et al. Formation of the protein corona: the interface between
nanoparticles and the immune system. Semin. Immunol. 34,5260
(2017).
24. Fadeel, B. Hide and seek: nanomaterial interactions with the immune system.
Front. Immunol. 10, 133 (2019).
25. Deng,Z.J.,Liang,M.,Monteiro,M.,Toth,I.&Minchin,R.F.Nanoparticle-
induced unfolding of brinogen promotes Mac-1 receptor activation and
inammation. Nat. Nanotechnol. 6,3944 (2011).
26. Tenzer, S. et al. Rapid formation of plasma protein corona critically affects
nanoparticle pathophysiology. Nat. Nanotechnol. 8,772781 (2013).
27. Vlasova, I. I. et al. Adsorbed plasma proteins modulate the effects of single-
walled carbon nanotubes on neutrophils in blood. Nanomedicine 12,
16151625 (2016).
28. Lu,N.,Sui,Y.,Tian,R.&Peng,Y.Y.Adsorptionofplasmaproteinsonsingle-
walled carbon nanotubes reduced cytotoxicity and modulated neutrophil
activation. Chem. Res. Toxicol. 31, 10611068 (2018).
29. Lara, S. et al. Identication of receptor binding to the biomolecular corona of
nanoparticles. ACS Nano. 11,18841893 (2017).
30. Salvati, A. et al. Transferrin-functionalized nanoparticles lose their targeting
capabilities when a biomolecule corona adsorbs on the surface. Nat.
Nanotechnol. 8,137143 (2013).
31. Tonigold, M. et al. Pre-adsorption of antibodies enables targeting of
nanocarriers despite a biomolecular corona. Nat. Nanotechnol. 13,862869
(2018).
32. Oh, J. Y. et al. Cloaking nanoparticles with protein corona shield for targeted
drug delivery. Nat. Commun. 9,19(2018).
33. Prapainop,K.,Witter,D.P.&Wentworth,P.Achemicalapproachforcell-
specic targeting of nanomaterials: small-molecule-initiated misfolding of
nanoparticle corona proteins. J. Am. Chem. Soc. 134,41004103 (2012).
34. Ezzat, K. et al. The viral protein corona directs viral pathogenesis and amyloid
aggregation. Nat. Commun. 10, 2331 (2019).
35. Théry, C., Ostrowski, M. & Segura, E. Membrane vesicles as conveyors of
immune responses. Nat. Rev. Immunol. 9,581593 (2009).
36. van Niel, G., DAngelo,G.&Raposo,G.Shedding light on the cell biology of
extracellular vesicles. Nat. Rev. Mol. Cell Biol. 19,213228 (2018).
37. Genschmer, K. R. et al. Activated PMN exosomes: pathogenic entities causing
matrix destruction and disease in the lung. Cell 176,113126 (2019).
38. Shvedova, A. A., Pietroiusti, A., Fadeel, B. & Kagan, V. E. Mechanisms of carbon
nanotube-induced toxicity: focus on oxidative stress. Toxicol. Appl. Pharmacol.
261,121133 (2012).
39. Zhu, M. et al. Exosomes as extrapulmonary signaling conveyors for
nanoparticle-induced systemic immune activation. Small 8,404412 (2012).
40. Cho,W.S.etal.Metaloxidenanoparticlesinduceuniqueinfammatory
footprints in the lung: important implications for nanoparticle testing.
Environ. Health Perspect. 118, 16991706 (2010).
41. Qiao, Y. et al. Identication of exosomal miRNAs in rats with pulmonary
neutrophilic inammation induced by zinc oxide nanoparticles. Front. Physiol.
9,111 (2018).
42. Logozzi, M. et al. Human primary macrophages scavenge AuNPs and
eliminate it through exosomes. A natural shuttling for nanomaterials. Eur. J.
Pharm. Biopharm. 137,2336 (2019).
43. Brinkmann,V.etal.Neutrophilextracellular traps kill bacteria. Science 303,
15321535 (2004).
44. Youse, S., Mihalache, C., Kozlowski, E., Schmid, I. & Simon, H. U. Viable
neutrophils release mitochondrial DNA to form neutrophil extracellular traps.
Cell Death Differ. 16, 14381444 (2009).
45. Lood, C. et al. Neutrophil extracellular traps enriched in oxidized mito-
chondrial DNA are interferogenic and contribute to lupus-like disease. Nat.
Med. 22,146153 (2016).
46. Fuchs,T.A.etal.Novelcelldeathprogram leads to neutrophil extracellular
traps. J. Cell Biol. 176, 231241 (2007).
47. Clark,S.R.etal.PlateletTLR4activates neutrophil extracellular traps to
ensnare bacteria in septic blood. Nat. Med. 13,463469 (2007).
48. Yipp, B. G. et al. Infection-induced NETosis is a dynamic process involving
neutrophil multitasking in vivo.Nat. Med. 18,13861393 (2012).
49. Takei, H., Araki, A., Watanabe, H., Ichinose, A. & Sendo, F. Rapid killing of
human neutrophils by the potent activator phorbol 12-myristate 13-acetate
(PMA) accompanied by changes different from typical apoptosis or necrosis.
J. Leukoc. Biol. 59,229240 (1996).
50. Fadeel,B.,Åhlin,A.,Henter,J.I.,Orrenius,S.&Hampton,M.B.Involvementof
caspases in neutrophil apoptosis: regulation by reactive oxygen species.
Blood 92,48084818 (1998).
51. Chen, K. W. et al. Noncanonical inammasome signaling elicits gasdermin D-
dependent neutrophil extracellular traps. Sci. Immunol. 3, eaar6676 (2018).
52. Sollberger, G. et al. Gasdermin D plays a vital role in the generation of
neutrophil extracellular traps. Sci. Immunol. 3, eaar6689 (2018).
Keshavan et al. Cell Death and Disease (2019) 10:569 Page 9 of 11
Ofcial journal of the Cell Death Differentiation Association
53. Schreiber, A. et al. Necroptosis controls NET generation and mediates
complement activation, endothelial damage, and autoimmune vasculitis.
Proc. Natl Acad. Sci. USA 114, E9618E9625 (2017).
54. Kenny,E.F.etal.Diversestimuliengagedifferent neutrophil extracellular trap
pathways. Elife 6, e24437 (2017).
55. VanDerLinden,M.,Westerlaken,G.H.A.,VanDerVlist,M.,VanMontfrans,J.&
Meyaard, L. Differential signalling and kinetics of neutrophil extracellular trap
release revealed by quantitative live imaging. Sci. Rep. 7, 6529 (2017).
56. Urban, C. F., Reichard, U., Brinkmann, V. & Zychlinsky, A. Neutrophil extra-
cellular traps capture and kill Candida albicans and hyphal forms. Cell.
Microbiol. 8,668676 (2006).
57. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease.
Nat. Rev. Immunol. 18,134147 (2018).
58. Hakkim, A. et al. Impairment of neutrophil extracellular trap degradation is
associated with lupus nephritis. Proc. Natl Acad. Sci. USA 107, 98139818
(2010).
59. Fuchs, T. A. et al. Extracellular DNA traps promote thrombosis. Proc. Natl Acad.
Sci. USA 107, 1588015885 (2010).
60. Jiménez-Alcázar, M. et al. Host DNases prevent vascular occlusion by neu-
trophil extracellular traps. Science 358,12021206 (2017).
61. Tillack, K., Breiden, P., Martin, R. & Sospedra, M. T lymphocyte priming by
neutrophil extracellular traps links innate and adaptive immune responses. J.
Immunol. 188, 31503159 (2012).
62. Stephen, J. et al. Neutrophil swarming and extracellular trap formation play a
signicant role in alum adjuvant activity. npj Vaccines 2, 1 (2017).
63. Bartneck, M., Keul, H. A., Gabriele, Z. K. & Groll, J. Phagocytosis independent
extracellular nanoparticle clearance by human immune cells. Nano Lett. 10,
5964 (2010).
64. Hwang, T. L., Aljuffali, I. A., Hung, C. F., Chen, C. H. & Fang, J. Y. The impact of
cationic solid lipid nanoparticles on human neutrophil activation and for-
mation of neutrophil extracellular traps (NETs). Chem. Biol. Interact. 235,
106114 (2015).
65. Bilyy, R. et al. Inert coats of magnetic nanoparticles prevent formation of
occlusive intravascular co-aggregates with neutrophil extracellular traps.
Front. Immunol. 9, 2266 (2018).
66. Muñoz, L. E. et al. Nanoparticles size-dependently initiate self-limiting
NETosis-driven inammation. Proc. Natl Acad. Sci. USA 113, E5856E5865
(2016).
67. Moore, L. et al. Biocompatibility assessment of detonation nanodiamond in
non-human primates and rats using histological, hematologic, and urine
analysis. ACS Nano. 10, 73857400 (2016).
68. Mukherjee, S. P. et al. Graphene oxide elicits membrane lipid changes and
neutrophil extracellular trap formation. Chem 4, 334358 (2018).
69. Neumann, A. et al. Lipid alterations in human blood-derived neutrophils lead
to formation of neutrophil extracellular traps. Eur. J. Cell Biol. 93,347354
(2014).
70. Douda,D.N.,Khan,M.A.,Grasemann,H.&Palaniyar,N.SK3channeland
mitochondrial ROS mediate NADPH oxidase-independent NETosis induced
by calcium inux. Proc. Natl Acad. Sci. USA 112, 28172822 (2015).
71. Mukherjee, S. P. et al. Graphene oxide is degraded by neutrophils and the
degradation products are non-genotoxic. Nanoscale 10, 11801188 (2018).
72. Mukherjee,S.P.,Bottini,M.&Fadeel,B.Grapheneandtheimmunesystem:a
romance of many dimensions. Front. Immunol. 8,111 (2017).
73. Klebanoff,S.J.,Kettle,A.J.,Rosen,H.,Winterbourn,C.C.&Nauseef,W.M.
Myeloperoxidase: a front-line defender against phagocytosed microorgan-
isms. J. Leukoc. Biol. 93, 185198 (2013).
74. Shvedova, A. A. et al. Impaired clearance and enhanced pulmonary
inammatory/brotic response to carbon nanotubes in myeloperoxidase-
decient mice. PLoS ONE 7, e30923 (2012).
75. Kagan,V.E.etal.Carbonnanotubesdegradedbyneutrophilmyeloperox-
idase induce less pulmonary inammation. Nat. Nanotechnol. 5,354359
(2010).
76. Bhattacharya, K. et al. Enzymatic strippingand degradation of PEGylated
carbon nanotubes. Nanoscale 6, 1468614690 (2014).
77. Andón, F. T. et al. Biodegradation of single-walled carbon nanotubes by
eosinophil peroxidase. Small 9, 27202729 (2013).
78. Farrera, C. et al. Extracellular entrapment and degradation of single-walled
carbon nanotubes. Nanoscale 6,69746983 (2014).
79. Kurapati, R., et al. Degradation of single-layer and few-layer graphene by
neutrophil myeloperoxidase. Angew. Chemie Int. Ed. Engl.57, 1172211727
(2018).
80. Sydlik, S. A., Jhunjhunwala, S., Webber, M. J., Anderson, D. G. & Langer, R. In
vivo compatibility of graphene oxide with differing oxidation states. ACS
Nano. 9,38663874 (2015).
81. Girish, C. M., Sasidharan, A., Gowd, G. S., Nair, S. & Koyakutty, M. Confocal
Raman imaging study showing macrophage mediated biodegradation of
graphene in vivo.Adv. Healthc. Mater. 2,14891500 (2013).
82. Fadeel, B. et al. Safety assessment of graphene-based materials: focus on
human health and the environment. ACS Nano. 12,1058210620 (2018).
83. Martinon, F., Burns, K. & Tschopp, J. The inammasome: a molecular platform
triggering activation of inammatory caspases and processing of proIL-β.
Mol. Cell. 10,417426 (2002).
84. Broz,P.&Dixit,V.M.Inammasomes: mechanism of assembly, regulation
and signalling. Nat. Rev. Immunol. 16,407420 (2016).
85. Mangan, M. S. J. et al. Targeting the NLRP3 inammasome in inammatory
diseases. Nat. Rev. Drug Discov. 17,588606 (2018).
86. Martinon, F., Pétrilli, V., Mayor, A., Tardivel, A. & Tschopp, J. Gout-associated
uric acid crystals activate the NALP3 inammasome. Nature 440,237241
(2006).
87. Dostert, C. et al. Innate immune activation through Nalp3 inammasome
sensing of asbestos and silica. Science 320,674677 (2008).
88. Cassel, S. L. et al. The Nalp3 inammasome is essential for the development
of silicosis. Proc. Natl Acad. Sci. USA 105,90359040 (2008).
89. Hornung, V. et al. Silica crystals and aluminum salts activate the NALP3
inammasome through phagosomal destabilization. Nat. Immunol. 9,
847856 (2008).
90. Sun,B.,Wang,X.,Ji,Z.,Li,R.&Xia,T.NLRP3inammasome activation induced
by engineered nanomaterials. Small 9,15951607 (2013).
91. Palomäki, J. et al. Long, needle-like carbon nanotubes and asbestos activate
the NLRP3 inammasome through a similar mechanism. ACS Nano. 5,
68616870 (2011).
92. Mukherjee,S.P.,Kostarelos,K.&Fadeel,B.Cytokineproling of primary
human macrophages exposed to endotoxin-free graphene oxide: size-
independent NLRP3 inammasome activation. Adv. Healthc. Mater.7,
1700815 (2018).
93. Andón, F. T. et al. Hollow carbon spheres trigger inammasome-dependent
IL-1βsecretion in macrophages. Carbon 113, 243251 (2017).
94. Chen, K. W. et al. The neutrophil NLRC4 inammasome selectively promotes
IL-1βmaturation without pyroptosis during acute Salmonella challenge. Cell
Rep. 8,570582 (2014).
95. Kambara,H.etal.GasderminDexertsanti-inammatory effects by pro-
moting neutrophil death. Cell Rep. 22,29242936 (2018).
96. Netea,M.G.,vandeVeerdonk,F.L.,vanderMeer,J.W.M.,Dinarello,C.A.&
Joosten, L. A. B. Inammasome-independent regulation of IL-1-family cyto-
kines. Annu. Rev. Immunol. 33,4977 (2015).
97. Bakele, M. et al. Localization and functionality of the inammasome in
neutrophils. J. Biol. Chem. 289,53205329 (2014).
98. Masumoto, J. et al. ASC, a novel 22-kDa protein, aggregates during apoptosis
of human promyelocytic leukemia HL-60 cells. J. Biol. Chem. 274,
3383533838 (1999).
99. Franklin, B. S. et al. The adaptor ASC has extracellular and prionoidactivities
that propagate inammation. Nat. Immunol. 15, 727737 (2014).
100. Baroja-Mazo, A. et al. The NLRP3 inammasome is released as a particulate
danger signal that amplies the inammatory response. Nat. Immunol. 15,
738748 (2014).
101. Rettig, L. et al. Particle size and activation threshold: a new dimension of
danger signaling. Blood 115,45334541 (2010).
102. Miyake, K. Innate immune sensing of pathogens and danger signals by cell
surface Toll-like receptors. Semin. Immunol. 19,310 (2007).
103. Litvack, M. L. & Palaniyar, N. Soluble innate immune pattern-recognition
proteins for clearing dying cells and cellular components: implications on
exacerbating or resolving inammation. Innate Immun. 16,191200 (2010).
104. Arai, S. & Miyazaki, T. A scavenging system against internal pathogens pro-
moted by the circulating protein apoptosis inhibitor of macrophage (AIM).
Semin. Immunopathol. 40, 567575 (2018).
105. Farrera,C.&Fadeel,B.Ittakestwototango:understandingtheinteractions
between engineered nanomaterials and the immune system. Eur. J. Pharm.
Biopharm. 95,312 (2015).
106. Silva, A. L. et al. Nanoparticle impact on innate immune cell pattern-
recognition receptors and inammasomes activation. Semin. Immunol. 34,
324 (2017).
Keshavan et al. Cell Death and Disease (2019) 10:569 Page 10 of 11
Ofcial journal of the Cell Death Differentiation Association
107. Fadeel, B. Clear and present danger? Engineered nanoparticles and the
immune system. Swiss Med. Wkly. 142, w13609 (2012).
108. Pradeu, T. & Cooper, E. L. The danger theory: 20 years later. Front. Immunol. 3,
287287 (2011).
109. Gallo,P.M.&Gallucci,S.Thedendriticcell response to classic, emerging, and
homeostatic danger signals. Implications for autoimmunity. Front. Immunol.
4, 138 (2013).
110. Shirasuna, K., Karasawa, T. & Takahashi, M. Exogenous nanoparticles and
endogenous crystalline molecules as danger signals for the NLRP3 inam-
masomes. J. Cell. Physiol. 234, 54365450 (2019).
111. Vita, A. A., Royse, E. A. & Pullen, N. A. Nanoparticles and danger signals: oral
delivery vehicles as potential disruptors of intestinal barrier homeostasis. J.
Leukoc. Biol.106,95103 (2019).
112. Nel, A. E. et al. Understanding biophysicochemical interactions at the nano-
bio interface. Nat. Mater. 8,543557 (2009).
113. Gallud, A. & Fadeel, B. Keeping it small: towards a molecular denition of
nanotoxicology. Eur. J. Nanomed. 7,143151 (2015).
114. Xiu, P., Zhou, R., Zhao, Y., Zuo, G. & Kang, S. Interactions between proteins and
carbon-based nanoparticles: exploring the origin of nanotoxicity at the
molecular level. Small 9,15461556 (2012).
115. Yanamala, N., Kagan, V. E. & Shvedova, A. A. Molecular modeling in structural
nano-toxicology: interactions of nano-particles with nano-machinery of cells.
Adv. Drug Deliv. Rev. 65, 20702077 (2013).
116. He, B. et al. Single-walled carbon-nanohorns improve biocompatibility over
nanotubes by triggering less protein-initiated pyroptosis and apoptosis in
macrophages. Nat. Commun. 9, 2393 (2018).
117. Mukherjee, S. P. et al. Macrophage sensing of single-walled carbon nano-
tubes via Toll-like receptors. Sci. Rep. 8,117 (2018).
118. Turabekova, M. et al. Immunotoxicity of nanoparticles: a computational study
suggests that CNTs and C
60
fullerenes might be recognized as pathogens by
Toll-like receptors. Nanoscale 6,34883495 (2014).
119. Liang, H. et al. Cationic nanoparticle as an inhibitor of cell-free DNA-induced
inammation. Nat. Commun. 9, 4291 (2018).
120. Lee, J. et al. Nucleic acid-binding polymers as anti-inammatory agents. Proc.
Natl Acad. Sci. USA 108, 1405514060 (2011).
121. Holl, E. K. et al. Scavenging nucleic acid debris to combat autoimmunity and
infectious disease. Proc. Natl Acad. Sci. USA 113,97289733 (2016).
122. Hubbell, J. A., Thomas, S. N. & Swartz, M. A. Materials engineering for
immunomodulation. Nature 462,449460 (2009).
123. Getts, D. R., Shea, L. D., Miller, S. D. & King, N. J. C. Harnessing nanoparticles for
immune modulation. Trends Immunol. 36,419427 (2015).
124. Vlasova, I. I. et al. Enzymatic oxidative biodegradation of nanoparticles:
mechanisms, signicance and applications. Toxicol. Appl. Pharmacol. 299,
5869 (2016).
125. Kagan, V. E. et al. Lung macrophages digestcarbon nanotubes using a
superoxide/peroxynitrite oxidative pathway. ACS Nano. 8,56105621 (2014).
126. Liu, L. et al. Structural basis of Toll-like receptor 3 signaling with double-
stranded RNA. Science 320,379381 (2008).
127. Mu, Q. et al. Protein binding by functionalized multiwalled carbon nanotubes
is governed by the surface chemistry of both parties and the nanotube
diameter. J. Phys. Chem. C 112,33003307 (2008).
128. Galluzzi, L. et al. Molecular mechanisms of cell death: recommendations of
the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 25,
486541 (2018).
129. Clarke,P.G.&Clarke,S.Nineteenthcenturyresearchoncelldeath.Exp. Oncol.
3, 139145 (2012).
130. Andón, F. T. & Fadeel, B. Programmed cell death: molecular mechanisms and
implications for safety assessment of nanomaterials. Acc. Chem. Res. 46,
733142 (2013).
131. Hussain, S. et al. Carbon black and titanium dioxide nanoparticles elicit distinct
apoptotic pathways in bronchial epithelial cells. Part. Fibre Toxicol. 7, 10 (2010).
132. Luanpitpong, S., Wang, L., Castranova, V. & Rojanasakul, Y. Induction of stem-like
cells with malignant properties by chronic exposure of human lung epithelial
cells to single-walled carbon nanotubes. Part. Fibre Toxicol. 11, 22 (2014).
133. Shi,J.,Gao,W.&Shao,F.Pyroptosis:gasdermin-mediatedprogrammed
necrotic cell death. Trends Biochem. Sci. 42,245254 (2017).
134. Mirshaee,V.etal.Toxicologicalproling of metal oxide nanoparticles in liver
context reveals pyroptosis in Kupffer cells and macrophages versus apoptosis
in hepatocytes. ACS Nano. 12,38363852 (2018).
135. Wang, X., Youse, S. & Simon, H. U. Necroptosis and neutrophil-associated
disorders. Cell Death Dis. 9, 111 (2018).
136. Stockwell, B. R. et al. Ferroptosis: a regulated cell death nexus linking
metabolism, redox biology, and disease. Cell 171,273285 (2017).
137. Szwed, M., et al. Small variations in nanoparticle structure dictate differential
cellular stress responses and mode of cell death. Nanotoxicology 13, 761782
(2019).
138. Zhang, J. et al. Zinc oxide nanoparticles harness autophagy to induce cell
death in lung epithelial cells. Cell Death Dis. 8, e2954 (2017).
Keshavan et al. Cell Death and Disease (2019) 10:569 Page 11 of 11
Ofcial journal of the Cell Death Differentiation Association
... Nevertheless, nerve injured regions typically include several other cell types playing a role in the regeneration pathways 4,[14][15][16][17] . An example is constituted by macrophages and other immune system cells like neutrophils 18 . Indeed, almost immediately after the damage, Schwann cells dissociate from axons, dedifferentiate, and secrete cytokines and chemokines 2 , prompting the recruitment of neutrophils, the first immune cell type that arrives in the site of inflammation from the bloodstream. ...
... Therefore, their modulation is a key factor to improve tissue-material integration 104,105 . Keshavan et al. discussed the impact of engineered nanomaterials on neutrophils and how neutrophils, in turn, may digest carbon nanotubes and graphene oxide 18 . They also observed that micrometric GO sheets disrupt the lipid rafts in neutrophils and induce the formation of NETs, associated with calcium influx and the production of ROS 72 . ...
Article
Full-text available
Graphene and bidimensional (2D) materials have been widely used in nerve conduits to boost peripheral nerve regeneration. Nevertheless, the experimental and commercial variability in graphene-based materials generates graphene forms with different structures and properties that can trigger entirely diverse biological responses from all the players involved in nerve repair. Herein, we focus on graphene and tungsten disulfide (WS2) interaction with non-neuronal cell types involved in nerve tissue regeneration. We synthesize highly-crystalline graphene and WS2 with scalable techniques such as thermal decomposition and chemical vapor deposition. Materials were able to trigger the activation of a neutrophils human model promoting Neutrophils Extracellular Traps (NETs) production, particularly in basal conditions, although neutrophils were not able to degrade graphene. Of note, pristine graphene acts as a repellent for the NETs adhesion, a beneficial property for nerve conduit long-term applications. Mesenchymal stem cells (MSCs) have been proposed as a promising strategy for nerve regeneration in combination with conduit. Thus, graphene interaction with MSCs was also investigated, and reduced viability was observed only on specific graphene substrates. Overall, the results confirm the possibility of regulating the cell response by varying graphene properties and selecting the most suitable graphene forms.
... An important phenomenon observed after the administration of nanoparticles to blood is an interaction with plasma proteins often described as a formation of "protein corona" [18,21,57]. (Figure 16) Albumin is one of the prominent components of the mammalian blood plasma (55-69% of plasma proteins) and significantly contributes to the protein corona. ...
... The summary of this process is illustrated Figure 17 with the final characteristics (size distribution, pH and electrical conductivi representing an interplay between adsorption-driven GO nanosizing [31], hydrolysis acidic precursor functional groups and desulfonation. An important phenomenon observed after the administration of nanoparticles blood is an interaction with plasma proteins often described as a formation of "prote corona" [18,21,57]. (Figure 16) Albumin is one of the prominent components of the ma Figure 17. ...
Article
Full-text available
Unlabelled: Graphene oxide's (GO) intravascular applications and biocompatibility are not fully explored yet, although it has been proposed as an anticancer drug transporter, antibacterial factor or component of wearable devices. Bivalent cations and the number of particles' atom layers, as well as their structural oxygen content and pH of the dispersion, all affect the GO size, shape, dispersibility and biological effects. Bovine serum albumin (BSA), an important blood plasma protein, is expected to improve GO dispersion stability in physiological concentrations of the precipitating calcium and magnesium cations to enable effective and safe tissue perfusion. Methods: Four types of GO commercially available aqueous dispersions (with different particle structures) were diluted, sonicated and studied in the presence of BSA and physiological cation concentrations. Nanoparticle populations sizes, electrical conductivity, zeta potential (Zetasizer NanoZS), structure (TEM and CryoTEM), functional groups content (micro titration) and dispersion pH were analyzed in consecutive preparation stages. Results: BSA effectively prevented the aggregation of GO in precipitating concentrations of physiological bivalent cations. The final polydispersity indexes were reduced from 0.66-0.91 to 0.36-0.43. The GO-containing isotonic dispersions were stable with the following Z-ave results: GO1 421.1 nm, GO2 382.6 nm, GO3 440.2 nm and GO4 490.1 nm. The GO behavior was structure-dependent. Conclusion: BSA effectively stabilized four types of GO dispersions in an isotonic dispersion containing aggregating bivalent physiological cations.
... As essential first-line innate immune defense cells, neutrophils play an invaluable protective role against invading pathogens. However, as potent initiators of inflammation neutrophils, they can also impart damaging effects to healthy tissues [73]. Although potentially the least well-studied immune cells in relation to nanotoxicology, neutrophils have been observed to preferentially phagocytose PEG-based NPs in the blood [74]. ...
Article
Full-text available
Aim: To investigate the influence of fluorine in reducing the adsorption of immune-reactive proteins onto PEGylated gold nanoparticles. Methods: Reversible addition fragmentation chain transfer polymerization, the Turkevich method and ligand exchange were used to prepare polymer-coated gold nanoparticles. Subsequent in vitro physicochemical and biological characterizations and proteomic analysis were performed. Results: Fluorine-modified polymers reduced the adsorption of complement and other immune-reactive proteins while potentially improving circulatory times and modulating liver toxicity by reducing apolipoprotein E adsorption. Fluorine actively discouraged phagocytosis while encouraging the adsorption of therapeutic targets, CD209 and signaling molecule calreticulin. Conclusion: This study suggests that the addition of fluorine in the surface coating of nanoparticles could lead to improved performance in nanomedicine designed for the intravenous delivery of cargos.
... The effect of NPs on neutrophils is very important for understanding the physiological and pathophysiological consequences of NPs' application [41][42][43]. The NPs studied were shown to induce a 2-fold increase in neutrophil ROS production and CD11b expression after their systemic administration. ...
Article
Full-text available
Insufficient drug accumulation in tumors is still a major concern for using cancer nanotherapeutics. Here, the neutrophil-based delivery of three nanoparticle types—liposomes, PLGA, and magnetite nanoparticles—was assessed both in vitro and in vivo. Confocal microscopy and a flow cytometry analysis demonstrated that all the studied nanoparticles interacted with neutrophils from the peripheral blood of mice with 4T1 mammary adenocarcinoma without a significant impact on neutrophil viability or activation state. Intravital microscopy of the tumor microenvironment showed that the neutrophils did not engulf the liposomes after intravenous administration, but facilitated nanoparticle extravasation in tumors through micro- and macroleakages. PLGA accumulated along the vessel walls in the form of local clusters. Later, PLGA nanoparticle-loaded neutrophils were found to cross the vascular barrier and migrate towards the tumor core. The magnetite nanoparticles extravasated in tumors both via spontaneous macroleakages and on neutrophils. Overall, the specific type of nanoparticles largely determined their behavior in blood vessels and their neutrophil-mediated delivery to the tumor. Since neutrophils are the first to migrate to the site of inflammation, they can increase nanodrug delivery effectiveness for nanomedicine application.
... Mesoporous silica nanoparticles have been widely investigated in contact with RBCs and their interactions with other blood components constitutes still an unexplored area. MSNs have recently received attention as a potential multiplatform for enhancing cancer immunotherapy because of their distinctive properties, including high porosity, good biocompatibility, ease of surface modification, self-adjuvanticity and their capacity to trigger cellular and tumor-mediated immune responses [57,58]. Currently, it is commonly accepted that regardless of the route of administration, after being concentrated in major organs such as liver, spleen, and lungs, they are being excreted from the body by the kidneys and hepatobiliary system [59,60], condition that satisfies FDA requirements for clinical application [61]. ...
Article
Full-text available
Emerging nanotechnologies offer numerous opportunities in the field of regenerative medicine and have been widely explored to design novel scaffolds for the regeneration and stimulation of nerve tissue. In this review, we focus on peripheral nerve regeneration. First, we introduce the biomedical problem and the present status of nerve conduits that can be used to guide, fasten and enhance regeneration. Then, we thoroughly discuss graphene as an emerging candidate in nerve tissue engineering, in light of its chemical, tribological and electrical properties. We introduce the graphene forms commonly used as neural interfaces, briefly review their applications, and discuss their potential toxicity. We then focus on the adoption of graphene in peripheral nervous system applications, a research field that has gained in the last years ever-increasing attention. We discuss the potential integration of graphene in guidance conduits, and critically review graphene interaction not only with peripheral neurons, but also with non-neural cells involved in nerve regeneration; indeed, the latter have recently emerged as central players in modulating the immune and inflammatory response and accelerating the growth of new tissue.
Article
Immune recognition and uptake of nanoparticles remain the hot topic in nanomedicine research. Complement is the central player in the immune recognition of engineered nanoparticles. Here, we summarize the accumulated knowledge on the role of complement in the interactions of nanomaterials with blood phagocytes. We describe the interplay between surface properties, complement opsonization, and immune uptake, primarily of iron oxide nanoparticles. We discuss the rigor of the published research and further identify the following knowledge gaps: 1) the role of complement in the variability of uptake of nanomaterials in healthy and diseased subjects, and 2) modulation of complement interactions to improve the performance of nanomaterials. Addressing these gaps is critical to improving translational chances of nanomaterials for drug delivery and imaging applications.
Article
Full-text available
Neutrophil extracellular traps (NETs) are large DNA reticular structures secreted by neutrophils and decorated with histones and antimicrobial proteins. As a key mechanism for neutrophils to resist microbial invasion, NETs play an important role in the killing of microorganisms (bacteria, fungi, and viruses). Although NETs are mostly known for mediating microbial killing, increasing evidence suggests that excessive NETs induced by stimulation of physical and chemical components, microorganisms, and pathological factors can exacerbate inflammation and organ damage. This review summarizes the induction and role of NETs in inflammation and focuses on the strategies of inhibiting NETosis and the mechanisms involved in pathogen evasion of NETs. Furthermore, herbal medicine inhibitors and nanodelivery strategies improve the efficiency of inhibition of excessive levels of NETs.
Article
Full-text available
Artificial nanoparticles accumulate a protein corona layer in biological fluids, which significantly influences their bioactivity. As nanosized obligate intracellular parasites, viruses share many biophysical properties with artificial nanoparticles in extracellular environments and here we show that respiratory syncytial virus (RSV) and herpes simplex virus type 1 (HSV-1) accumulate a rich and distinctive protein corona in different biological fluids. Moreover, we show that corona pre-coating differentially affects viral infectivity and immune cell activation. In addition, we demonstrate that viruses bind amyloidogenic peptides in their corona and catalyze amyloid formation via surface-assisted heterogeneous nucleation. Importantly, we show that HSV-1 catalyzes the aggregation of the amyloid β-peptide (Aβ 42), a major constituent of amyloid plaques in Alzheimer's disease, in vitro and in animal models. Our results highlight the viral protein corona as an acquired structural layer that is critical for viral-host interactions and illustrate a mechanistic convergence between viral and amyloid pathologies.
Article
Full-text available
Engineered nanomaterials hold promise for a wide range of applications in medicine. However, safe use of nanomaterials requires that interactions with biological systems, not least with the immune system, are understood. Do nanomaterials elicit novel or unexpected effects, or is it possible to predict immune responses to nanomaterials based on how the immune system handles pathogens? How does the bio-corona of adsorbed biomolecules influence subsequent immune interactions of nanomaterials? How does the grafting of polymers such as poly(ethylene glycol) onto nanomaterial surfaces impact on these interactions? Can ancient immune evasion or “stealth” strategies of pathogens inform the design of nanomaterials for biomedical applications? Can nanoparticles co-opt immune cells to target diseased tissues? The answers to these questions may prove useful for the development of nanomedicines.
Article
Full-text available
Targeted drug delivery using nanoparticles can minimize the side effects of conventional pharmaceutical agents and enhance their efficacy. However, translating nanoparticle-based agents into clinical applications still remains a challenge due to the difficulty in regulating interactions on the interfaces between nanoparticles and biological systems. Here, we present a targeting strategy for nanoparticles incorporated with a supramolecularly pre-coated recombinant fusion protein in which HER2-binding affibody combines with glutathione-S-transferase. Once thermodynamically stabilized in preferred orientations on the nanoparticles, the adsorbed fusion proteins as a corona minimize interactions with serum proteins to prevent the clearance of nanoparticles by macrophages, while ensuring systematic targeting functions in vitro and in vivo. This study provides insight into the use of the supramolecularly built protein corona shield as a targeting agent through regulating the interfaces between nanoparticles and biological systems.
Article
Human neutrophils have a short half-life and are believed to die by apoptosis or programmed cell death both in vivo and in vitro. We found that caspases are activated in a time-dependent manner in neutrophils undergoing spontaneous apoptosis, concomitant with other characteristic features of apoptotic cell death such as morphologic changes, phosphatidylserine (PS) exposure, and DNA fragmentation. The treatment of neutrophils with agonistic anti-Fas monoclonal antibodies (MoAbs) significantly accelerated this process. However, in cells treated with the potent neutrophil activator phorbol 12-myristate 13-acetate (PMA), caspase activity was only evident after pharmacologic inhibition of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Similarily, inhibition of the NADPH oxidase in constitutive and Fas/APO-1–triggered apoptosis resulted in increased rather than suppressed levels of caspase activity, suggesting that reactive oxygen species may prevent caspases from functioning optimally in these cells. Moreover, oxidants generated via the NADPH oxidase were essential for PS exposure during PMA-induced cell death, but not for neutrophils undergoing spontaneous apoptosis. We conclude that caspases are an important component of constitutive and Fas/APO-1–triggered neutrophil apoptosis. However, these redox sensitive enzymes are suppressed in activated neutrophils, and an alternate oxidant-dependent pathway is used to mediate PS exposure and neutrophil clearance under these conditions.
Article
Gut immune system homeostasis involves diverse structural interactions among resident microbiota, the protective mucus layer, and a variety of cells (intestinal epithelial, lymphoid, and myeloid). Due to the substantial surface area in direct contact with an “external” environment and the diversity of xenobiotic, abiotic, and self‐interactions coordinating to maintain gut homeostasis, there is enhanced potential for the generation of endogenous danger signals when this balance is lost. Here, we focus on the potential generation and reception of damage in the gut resulting from exposure to nanoparticles (NPs), common food and drug additives. Specifically, we describe recent evidence in the literature showing that certain NPs are potential generators of damage‐associated molecular patterns, as well as potential immune‐stimulating molecular patterns themselves.
Article
The use of nanomaterials is increasing but the real risk associated with their use in humans has to be defined. In fact, nanomaterials tend to accumulate in organs over a long period of time and are slowly degraded or eliminated by the body. Exosomes are nanovesicles actively shuttle molecules, including chemical products and metals, through the body. Macrophages scavenge the body from both organic and inorganic substances, and they use to release high amounts of exosomes. We hypothesized that macrophages may have a role in eliminating nanomaterials through their exosomes. We treated human primary macrophages with 20 nm gold nanoparticles (AuNPs), analyzing the presence of AuNPs in both cells and the released exosomes by the implementation of different techniques, including SP-ICP-MS and NTA. We showed that macrophages endocytosed AuNPs and released them through exosomes. Our study on one hand provide the evidence for a new methodology in the early identification of the nanomaterials levels in exposed subjects. On the other hand we depict a way our body shuttle virtually intact nanoparticles through macrophage-released exosomes.
Article
For optimal exploitation of nanoparticles (NPs) in biomedicine, and to predict nanotoxicity, detailed knowledge of the cellular responses to cell-bound or internalized NPs is imperative. The final outcome of NP-cell interaction is dictated by the type and magnitude of the NP insult and the cellular response. Here, this has been systematically studied by using poly(alkylcyanoacrylate) (PACA) particles differing only in their alkyl side chains; butyl (PBCA), ethylbutyl (PEBCA), or octyl (POCA), respectively. Surprisingly, these highly similar NPs induced different stress responses and modes of cell death in human cell lines. The POCA particles generally induced endoplasmic reticulum stress and apoptosis. In contrast, PBCA and PEBCA particles induced oxidative stress and lipid peroxidation depending on the level of the glutathione precursor cystine and transcription of the cystine transporter SLC7A11. The latter was induced as a protective response by the transcription factors ATF4 and Nrf2. PBCA particles strongly activated ATF4 downstream of the eIF2α kinase HRI, whereas PEBCA particles more potently induced Nrf2 antioxidant responses. Intriguingly, PBCA particles activated the cell death mechanism ferroptosis; a promising option for targeting multidrug-resistant cancers. Our findings highlight that even minor differences in NP composition can severely impact the cellular response to NPs. This may have important implications in therapeutic settings.
Article
Here, we describe a novel pathogenic entity, the activated PMN (polymorphonuclear leukocyte, i.e., neutrophil)-derived exosome. These CD63+/CD66b+ nanovesicles acquire surface-bound neutrophil elastase (NE) during PMN degranulation, NE being oriented in a configuration resistant to α1-antitrypsin (α1AT). These exosomes bind and degrade extracellular matrix (ECM) via the integrin Mac-1 and NE, respectively, causing the hallmarks of chronic obstructive pulmonary disease (COPD). Due to both ECM targeting and α1AT resistance, exosomal NE is far more potent than free NE. Importantly, such PMN-derived exosomes exist in clinical specimens from subjects with COPD but not healthy controls and are capable of transferring a COPD-like phenotype from humans to mice in an NE-driven manner. Similar findings were observed for another neutrophil-driven disease of ECM remodeling (bronchopulmonary dysplasia [BPD]). These findings reveal an unappreciated role for exosomes in the pathogenesis of disorders of ECM homeostasis such as COPD and BPD, providing a critical mechanism for proteolytic damage.
Article
Neutrophils are the most abundant circulating leukocyte, and the first point of contact between many drug delivery formulations and human cells. Despite their prevalence and implication in a range of immune functions, little is known about how human neutrophils respond to synthetic particulates. Here, we describe how ex vivo human neutrophils respond to particles which vary in both size (5 nm – 2 microns) and chemistry (lipids, poly(styrene), poly(lactic-co-glycolic acid), and gold). In particular, we show that (i) particle uptake is rapid, typically plateauing within 15 min, (ii) for a given particle chemistry, neutrophils preferentially take up larger particles at the nanoscale, up to 200 nm in size, (iii) uptake of nanoscale poly(styrene) and liposomal particles at concentrations of up to 5 μg/mL does not enhance apoptosis, activation or cell death, (iv) particle-laden neutrophils retain the ability to degranulate normally in response to chemical stimulation and (v) ingested particles reside in intracellular compartments that are retained during activation and degranulation. Aside from the implications for design of intravenously delivered particulate formulations in general, we expect these observations to be of particular use for targeting nanoparticles to circulating neutrophils, their clearance site (bone marrow), or distal sites of active inflammation.
Article
Graphene and its derivatives are heralded as ‘miracle’ materials with manifold applications in different sectors of society from electronics to energy storage to medicine. The increasing exploitation of graphene-based materials (GBMs) necessitates a comprehensive evaluation of the potential impact of these materials on human health and the environment. Here we discuss synthesis and characterization of GBMs as well as human and environmental hazard assessment of GBMs using in vitro and in vivo model systems with the aim to understand the properties that underlie the biological effects of these materials; not all GBMs are alike, and it is essential that we disentangle the structure-activity relationships for this class of materials.