ArticlePDF Available

Delivery of Mixed-Lineage Kinase Domain-Like Protein by Vapor Nanobubble Photoporation Induces Necroptotic-Like Cell Death in Tumor Cells

MDPI
International Journal of Molecular Sciences (IJMS)
Authors:

Abstract and Figures

Modern molecular medicine demands techniques to efficiently deliver molecules directly into mammalian cells. As proteins are the final mediators of most cellular pathways, efficient intracellular protein delivery techniques are highly desired. In this respect, photoporation is a promising recent technique for the delivery of proteins directly into living cells. Here, we show the possibility to deliver a model saccharide (FD70) and a model protein (FITC-BSA) into murine B16 melanoma cells by using the vapor nanobubble photoporation technique with an efficiency of 62% and 38%, respectively. Next, we delivered the mixed-lineage kinase domain-like (MLKL) protein, the most terminal mediator of necroptosis currently known, and caspase-8 and -3 protein, which are important proteins in the initiation and execution of apoptosis. A significant drop in cell viability with 62%, 71% and 64% cell survival for MLKL, caspase-8 and caspase-3, respectively, was observed. Remarkably, maximal cell death induction was already observed within 1 h after protein delivery. Transduction of purified recombinant MLKL by photoporation resulted in rapid cell death characterized by cell swelling and cell membrane rupture, both hallmarks of necroptosis. As necroptosis has been identified as a type of cell death with immunogenic properties, this is of interest to anti-cancer immunotherapy. On the other hand, transduction of purified recombinant active caspase-3 or -8 into the tumor cells resulted in rapid cell death preceded by membrane blebbing, which is typical for apoptosis. Our results suggest that the type of cell death of tumor cells can be controlled by direct transduction of effector proteins that are involved in the executioner phase of apoptosis or necroptosis.
This content is subject to copyright.
International Journal of
Molecular Sciences
Article
Delivery of Mixed-Lineage Kinase Domain-Like
Protein by Vapor Nanobubble Photoporation Induces
Necroptotic-Like Cell Death in Tumor Cells
Lien Van Hoecke 1, 2, , Laurens Raes 3, , Stephan Stremersch 3, Toon Brans 3,
Juan C. Fraire 3, Ria Roelandt 2,4, Wim Declercq 2,4 , Peter Vandenabeele 2,5,
Koen Raemdonck 3, Kevin Braeckmans 3and Xavier Saelens 1, 5, *
1VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, Belgium
2Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
3Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, 9000 Ghent, Belgium
4VIB-UGent Center for Inflammation Research, VIB, 9052 Ghent, Belgium
5Department of Biochemistry and Microbiology, Ghent University, 9000 Ghent, Belgium
*Correspondence: Xavier.saelens@vib-ugent.be
These authors contributed equally to this work.
Received: 6 August 2019; Accepted: 29 August 2019; Published: 30 August 2019


Abstract:
Modern molecular medicine demands techniques to eciently deliver molecules directly
into mammalian cells. As proteins are the final mediators of most cellular pathways, ecient
intracellular protein delivery techniques are highly desired. In this respect, photoporation is a
promising recent technique for the delivery of proteins directly into living cells. Here, we show
the possibility to deliver a model saccharide (FD70) and a model protein (FITC-BSA) into murine
B16 melanoma cells by using the vapor nanobubble photoporation technique with an eciency
of 62% and 38%, respectively. Next, we delivered the mixed-lineage kinase domain-like (MLKL)
protein, the most terminal mediator of necroptosis currently known, and caspase-8 and -3 protein,
which are important proteins in the initiation and execution of apoptosis. A significant drop in
cell viability with 62%, 71% and 64% cell survival for MLKL, caspase-8 and caspase-3, respectively,
was observed. Remarkably, maximal cell death induction was already observed within 1 h after
protein delivery. Transduction of purified recombinant MLKL by photoporation resulted in rapid
cell death characterized by cell swelling and cell membrane rupture, both hallmarks of necroptosis.
As necroptosis has been identified as a type of cell death with immunogenic properties, this is of
interest to anti-cancer immunotherapy. On the other hand, transduction of purified recombinant
active caspase-3 or -8 into the tumor cells resulted in rapid cell death preceded by membrane blebbing,
which is typical for apoptosis. Our results suggest that the type of cell death of tumor cells can be
controlled by direct transduction of eector proteins that are involved in the executioner phase of
apoptosis or necroptosis.
Keywords:
protein delivery; vapor nanobubble photoporation; necroptotic cell death; mixed-lineage
kinase domain-like; apoptotic cell death; caspases
1. Introduction
Cell death is an evolutionary conserved process in multicellular organisms with far reaching
implications in health and disease [
1
]. Regulated cell death is ultimately controlled by protein activities.
Apoptosis, a well-known form of cell death, is the consequence of a caspase signaling cascade involving
caspase-8 and -3 [
2
]. In recent years, it has become increasingly clear that the type of cell death strongly
influences the response of other cells in the vicinity of the dying cells. Cell death modalities may
Int. J. Mol. Sci. 2019,20, 4254; doi:10.3390/ijms20174254 www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2019,20, 4254 2 of 14
range from immunologically silent to a profound immune-stimulating outcome. The latter condition
can be highly beneficial in the control of tumors by the patient’s immune system. Indeed, it has
been well documented that an immunogenic tumor environment is essential for the clinical success
of immunotherapies. To reshape the tumor microenvironment, immune stimulatory molecules [
3
],
blockade of inhibitory cytokines [
4
] as well as an immune checkpoint blockade [
5
7
] can be introduced
at the tumor site to restore immunological fitness. The direct intratumoral injection of mRNA encoding
such immune modulatory proteins is an attractive approach. Hewitt and colleagues, for example,
delivered an IL-23/IL-36
γ
/OX40L triplet mRNA mixture combined with checkpoint blockade to
successfully reshape the tumor microenvironment [
3
]. One other way to induce such an immunogenic
environment is by eliciting immunogenic cell death that results in the release of damage-associated
molecular patterns (DAMPs) and other immune-stimulatory components that can recruit and activate
dendritic cells, which are professional antigen-presenting cells. For example, immunogenic apoptosis
of neoplastic cells has been reported as a beneficial outcome in response to irradiation, chemotherapies
and hypericin-based photodynamic therapy [812].
Necroptosis, a regulated form of necrosis, can also have immunogenic properties [
13
,
14
]. The
necroptotic pathway involves two core proteins, namely receptor interacting protein kinase 3 (RIPK3)
and its substrate, mixed-lineage domain-like protein (MLKL) [
15
20
]. Upon its phosphorylation
by RIPK3, phosphorylated MLKL oligomerizes and translocates to the plasma membrane, where it
induces membrane permeabilization and necroptosis. It is remarkable that many tumor types show
genetic and epigenetic changes in the necroptotic pathway. For example, there are cases reported of
colon carcinoma, acute myeloid and chronic lymphatic leukemia that show strongly reduced RIPK3
expression levels [
21
]. Moreover, in pancreatic cancers, reduced MLKL expression is associated with
decreased survival of the patient [
22
,
23
]. A potential way to restore these aberrations in the necroptotic
pathway is by delivery of the executioner protein MLKL, e.g., encoded by
in vitro
transcribed mRNA,
to the tumor cells [24].
An increasing number of mammalian gene products, including intracellular proteins, are now
available as purified recombinant proteins [
25
]. To study the function of such proteins in living
cells, the intracellular cytosolic proteins need to be delivered across the plasma membrane into the
cytoplasm. There are only a limited number of protein delivery techniques available to date, such as
electroporation, microinjection, lipofection or techniques based on cell-penetrating peptides that are
fused to the protein of interest [
26
29
]. Because of the relatively large size and structural complexity
of proteins, the current protein delivery methods often suer from limited ecacy, which strongly
depends on the protein of interest and cell type, or are associated with high cell toxicity. Therefore,
eorts are continuously ongoing in search of straightforward broadly applicable approaches for
intracellular delivery of macromolecules, including proteins [
30
32
]. One of the newest and most
promising transfection technologies is laser-induced vapor nanobubble (VNB) photoporation. It
requires cells to be incubated first for a short time with plasmonic gold nanoparticles (AuNPs) that
will adsorb onto the cellular plasma membrane. Next, the cells are illuminated with an intense short
laser pulse with a wavelength in close proximity to the AuNP plasmon resonance wavelength. Upon
light absorption, the AuNPs heat up rapidly, leading to the brisk evaporation of the water surrounding
the AuNP surface. The resulting water vapor nanobubble around each AuNP will quickly expand
until all thermal energy is consumed, after which, the bubble collapses [
33
,
34
]. This causes local shock
waves and transient permeabilization of the cell membrane at the location where AuNPs were present.
Extracellular molecules in the surrounding cell medium can then diuse into the cell cytoplasm.
The VNB photoporation technology was already shown to allow the ecient delivery of dierent
macromolecules, such as siRNA [
35
37
] and antibodies [
38
] in a wide variety of cell types, including
hard-to-transfect cells such as primary neurons [39] and lymphocytes [36,40].
In this study, our aim was to assess if exogenously delivered recombinant proteins associated
with two types of cell death, apoptosis and necroptosis, would be sucient to induce the respective
cell death types in B16 melanoma tumor cells. We show here that MLKL protein delivery by VNB
Int. J. Mol. Sci. 2019,20, 4254 3 of 14
photoporation into B16 cells induced cell death with necroptotic hallmarks (swelling of the cells and
no caspase activity) (Figure 1). In addition, we demonstrate that VNB photoporation of purified
recombinant caspase-3 and -8 into the B16 cells results in rapid blebbing and apoptotic cell death.
Int. J. Mol. Sci. 2019, 20, x FOR PEER REVIEW 3 of 14
no caspase activity) (Figure 1). In addition, we demonstrate that VNB photoporation of purified
recombinant caspase-3 and -8 into the B16 cells results in rapid blebbing and apoptotic cell death.
Figure 1. Induction of cell death with necroptotic or apoptotic hallmarks after mixed-lineage kinase
domain-like (MLKL) protein or caspase-3/-8 delivery by vapor nanobubble (VNB) photoporation,
respectively. B16 cells are first incubated with gold nanoparticles (AuNPs) that adsorb onto the
plasma membrane, followed by a nanosecond pulsed laser treatment that results in the formation of
vapor nanobubbles (VNBs). Collapse of the expanding VNBs leads to membrane pore formation and
allows the extracellular MLKL or caspase -3/-8 protein to diffuse into the cell cytoplasm. After a short
period of incubation, the MLKL protein has evoked cell death with necroptotic hallmarks, whereas
transduced caspase-3/-8 protein results in apoptosis.
2. Results
2.1. VNB Photoporation Enables Efficient Delivery of 70 kDa FITC-Dextrans into B16 Tumor Cells
To assess the possibility to use VNB photoporation for efficient macromolecular delivery into
mammalian tumor cells (B16 cells), we first applied fluorescently labeled dextrans (FITC-dextran) of
70 kDa (FD70), which are only slightly larger than the proteins of interest (murine MLKL: 56.3 kDa,
active murine caspase-3: 32 kDa and active murine caspase-8: 55 kDa). The first step in the
photoporation procedure is to incubate cells with AuNPs, which will locally permeabilize the cell
membrane upon pulsed laser irradiation. As the concentration of AuNPs that yields optimal
transduction results depends on the specific cell type, this parameter was first optimized for the B16
cells. Using confocal reflection microscopy, the number of cell-bound AuNPs after 30 min of
incubation was determined for different AuNP concentrations (Figure 2A). As expected, the AuNP
concentration correlates positively with the number of cell-adsorbed AuNPs, which ranged from <1
to about 6 AuNPs/cell depending on the concentration used. Next, we performed photoporation
experiments for these various AuNP concentrations and confirmed intracellular FD70 delivery by
confocal microscopy and, quantitatively, by flow cytometry. From the representative images in
Figure 2B, it can be appreciated that the delivery efficiency increases with higher concentrations of
AuNPs, which was further confirmed by flow cytometry analysis (Figure 2C). When the highest
concentration of AuNPs (16 × 10
7
AuNPs/mL, ~6 AuNPs/cell) was used, a transduction efficacy of up
Figure 1.
Induction of cell death with necroptotic or apoptotic hallmarks after mixed-lineage kinase
domain-like (MLKL) protein or caspase-3/-8 delivery by vapor nanobubble (VNB) photoporation,
respectively. B16 cells are first incubated with gold nanoparticles (AuNPs) that adsorb onto the plasma
membrane, followed by a nanosecond pulsed laser treatment that results in the formation of vapor
nanobubbles (VNBs). Collapse of the expanding VNBs leads to membrane pore formation and allows
the extracellular MLKL or caspase -3/-8 protein to diuse into the cell cytoplasm. After a short period
of incubation, the MLKL protein has evoked cell death with necroptotic hallmarks, whereas transduced
caspase-3/-8 protein results in apoptosis.
2. Results
2.1. VNB Photoporation Enables Ecient Delivery of 70 kDa FITC-Dextrans into B16 Tumor Cells
To assess the possibility to use VNB photoporation for ecient macromolecular delivery into
mammalian tumor cells (B16 cells), we first applied fluorescently labeled dextrans (FITC-dextran) of
70 kDa (FD70), which are only slightly larger than the proteins of interest (murine MLKL: 56.3 kDa, active
murine caspase-3: 32 kDa and active murine caspase-8: 55 kDa). The first step in the photoporation
procedure is to incubate cells with AuNPs, which will locally permeabilize the cell membrane upon
pulsed laser irradiation. As the concentration of AuNPs that yields optimal transduction results
depends on the specific cell type, this parameter was first optimized for the B16 cells. Using confocal
reflection microscopy, the number of cell-bound AuNPs after 30 min of incubation was determined for
dierent AuNP concentrations (Figure 2A). As expected, the AuNP concentration correlates positively
with the number of cell-adsorbed AuNPs, which ranged from <1 to about 6 AuNPs/cell depending
on the concentration used. Next, we performed photoporation experiments for these various AuNP
concentrations and confirmed intracellular FD70 delivery by confocal microscopy and, quantitatively,
by flow cytometry. From the representative images in Figure 2B, it can be appreciated that the delivery
eciency increases with higher concentrations of AuNPs, which was further confirmed by flow
cytometry analysis (Figure 2C). When the highest concentration of AuNPs (16
×
10
7
AuNPs/mL,
~6 AuNPs/cell) was used, a transduction ecacy of up to 62% positive cells was observed. Higher
Int. J. Mol. Sci. 2019,20, 4254 4 of 14
AuNP concentrations resulted in higher FD70 transduction eciency, as evident from the gradually
increasing mean fluorescence per cell relative to the negative control (rMFI), which is likely related to
the formation of more pores in the plasma membrane. However, a higher degree of permeabilization
also resulted in increased cell toxicity, as evidenced from the 40% reduction in cell viability when an
AuNP concentration of 16 ×107AuNPs/mL was applied.
Int. J. Mol. Sci. 2019, 20, x FOR PEER REVIEW 4 of 14
to 62% positive cells was observed. Higher AuNP concentrations resulted in higher FD70
transduction efficiency, as evident from the gradually increasing mean fluorescence per cell relative
to the negative control (rMFI), which is likely related to the formation of more pores in the plasma
membrane. However, a higher degree of permeabilization also resulted in increased cell toxicity, as
evidenced from the 40% reduction in cell viability when an AuNP concentration of 16 × 107
AuNPs/mL was applied.
Figure 2. Delivery of FITC-dextran 70 kDa to B16 tumor cells by VNB photoporation. (A) B16 cells
were incubated with different AuNP concentrations, after which, the excess of AuNPs was washed
away. Left: Attachment of AuNPs (concentration: 8 × 107 AuNPs/mL) to the cells visualized by
confocal reflection microscopy and overlayed with a light transmission image of the cells (scale bar =
20 µm). Right: The average number of cell-attached AuNPs was quantified for each of the AuNP
concentrations using image processing (quantified from >100 cells for each condition). B16 cells were
transfected with FITC-dextran 70 kDa (at 2 mg/mL) after incubation with different concentrations of
gold nanoparticles (AuNPs). Untreated cells, cells incubated with AuNPs and FITC-dextran, and cells
treated only with laser pulses (without AuNPs) were included as controls. (B) Confocal images of B16
cells after FITC-dextran delivery (scale bar = 100 µm). (C) FITC-dextran transfection efficiency as
Figure 2.
Delivery of FITC-dextran 70 kDa to B16 tumor cells by VNB photoporation. (
A
) B16 cells
were incubated with dierent AuNP concentrations, after which, the excess of AuNPs was washed
away. Left: Attachment of AuNPs (concentration: 8
×
10
7
AuNPs/mL) to the cells visualized by
confocal reflection microscopy and overlayed with a light transmission image of the cells (scale bar
=20
µ
m). Right: The average number of cell-attached AuNPs was quantified for each of the AuNP
concentrations using image processing (quantified from >100 cells for each condition). B16 cells were
transfected with FITC-dextran 70 kDa (at 2 mg/mL) after incubation with dierent concentrations of
gold nanoparticles (AuNPs). Untreated cells, cells incubated with AuNPs and FITC-dextran, and cells
treated only with laser pulses (without AuNPs) were included as controls. (
B
) Confocal images of
B16 cells after FITC-dextran delivery (scale bar =100
µ
m). (
C
) FITC-dextran transfection eciency as
determined by flow cytometry (n=4, independent experiments). (
D
) Cell viability after photoporation
treatment (n=3, independent experiments).
Int. J. Mol. Sci. 2019,20, 4254 5 of 14
2.2. Ecient Protein Delivery in B16 Tumor Cells by VNB Photoporation
In the next step, we assessed whether a model protein could be delivered into B16 cells by
photoporation. For this purpose, we selected FITC-conjugated bovine serum albumin (FITC-BSA),
which has a molecular weight of 66.5 kDa. Delivery eciency again increased with increasing AuNP
concentrations, reaching up to 38% FITC-BSA positive cells for 16
×
10
7
AuNP/mL (Figure 3A). On
the other hand, the protein transduction appears less ecient compared to FD70 at equal mass
concentrations, despite the similar molecular weight. In addition, the relative mean fluorescence
intensities (rMFI) of the FITC-BSA transfected cells was lower than that of FD70 transduced cells. This
can likely be explained by the relative dierence in fluorescence intensity of both compounds. Indeed,
measurement of the fluorescent intensity of solutions of FITC-BSA and FITC-dextran 70 kDa at equal
mass concentration by fluorimetry shows a >10-fold dierence in fluorescent signal (Figure 3B). Based
on these results, we can conclude that VNB photoporation enables ecient protein delivery into B16
tumor cells. These data, together with the FD70 transfection results, show that an AuNP concentration
of 4
×
10
7
AuNPs/mL (i.e., approximately 1 AuNP/cell) represents a good balance between optimal
transduction eciency and cell viability and was, therefore, used in all further experiments.
Int. J. Mol. Sci. 2019, 20, x FOR PEER REVIEW 5 of 14
determined by flow cytometry (n = 4, independent experiments). (D) Cell viability after photoporation
treatment (n = 3, independent experiments).
2.2. Efficient Protein Delivery in B16 Tumor Cells by VNB Photoporation
In the next step, we assessed whether a model protein could be delivered into B16 cells by
photoporation. For this purpose, we selected FITC-conjugated bovine serum albumin (FITC-BSA),
which has a molecular weight of 66.5 kDa. Delivery efficiency again increased with increasing AuNP
concentrations, reaching up to 38% FITC-BSA positive cells for 16 × 10
7
AuNP/mL (Figure 3A). On
the other hand, the protein transduction appears less efficient compared to FD70 at equal mass
concentrations, despite the similar molecular weight. In addition, the relative mean fluorescence
intensities (rMFI) of the FITC-BSA transfected cells was lower than that of FD70 transduced cells.
This can likely be explained by the relative difference in fluorescence intensity of both compounds.
Indeed, measurement of the fluorescent intensity of solutions of FITC-BSA and FITC-dextran 70 kDa
at equal mass concentration by fluorimetry shows a >10-fold difference in fluorescent signal (Figure
3B). Based on these results, we can conclude that VNB photoporation enables efficient protein
delivery into B16 tumor cells. These data, together with the FD70 transfection results, show that an
AuNP concentration of 4 × 10
7
AuNPs/mL (i.e., approximately 1 AuNP/cell) represents a good
balance between optimal transduction efficiency and cell viability and was, therefore, used in all
further experiments.
Figure 3. Delivery of FITC-BSA to B16 tumor cells by VNB photoporation. B16 cells were transfected
with FITC-BSA (at 2 mg/mL) after incubation with different concentrations of AuNPs. Untreated cells,
cells incubated with AuNPs and FITC-BSA, and cells treated only with laser pulses (without AuNPs)
were included as controls. (A) FITC-BSA transfection efficiency, as determined by flow cytometry (n
= 3, independent experiments). (B) Relative FITC fluorescence of solutions of FITC-BSA (66.5 kDa)
and FITC-dextran 70 kDa, measured by fluorimetry at an equal mass concentration of 1 mg/mL (n =
3, independent experiments).
2.3. Delivery of Caspase-3/-8 or MLKL by VNB Photoporation Induces Cell Death
We next investigated the functional delivery by photoporation of the necroptotic cell death
mediator MLKL and of purified recombinant caspase-3 and caspase-8, well-known executioners and
initiators of the apoptotic cell death pathway, respectively.
All three proteins were added at a concentration of 150 µg/mL to the photoporation cell medium.
After completing the photoporation procedure, the B16 melanoma cells were supplemented with
culture medium and placed back in the cell incubator. Six hours after photoporation, a significant
decline in viability was detected in the MLKL, caspase-3 and caspase-8 protein groups, as compared
to control cells that were photoporated in the absence of any of the three proteins (green bar, Figure
4). This observation was consistent with confocal microscopy images of the cells (Figure 4A) and
quantitative CellTiter-Glo
®
cell viability data (Figure 4B). As cell viability was not affected in the
Figure 3.
Delivery of FITC-BSA to B16 tumor cells by VNB photoporation. B16 cells were transfected
with FITC-BSA (at 2 mg/mL) after incubation with dierent concentrations of AuNPs. Untreated cells,
cells incubated with AuNPs and FITC-BSA, and cells treated only with laser pulses (without AuNPs)
were included as controls. (
A
) FITC-BSA transfection eciency, as determined by flow cytometry
(n=3,
independent experiments). (
B
) Relative FITC fluorescence of solutions of FITC-BSA (66.5 kDa)
and FITC-dextran 70 kDa, measured by fluorimetry at an equal mass concentration of 1 mg/mL (n=3,
independent experiments).
2.3. Delivery of Caspase-3/-8 or MLKL by VNB Photoporation Induces Cell Death
We next investigated the functional delivery by photoporation of the necroptotic cell death
mediator MLKL and of purified recombinant caspase-3 and caspase-8, well-known executioners and
initiators of the apoptotic cell death pathway, respectively.
All three proteins were added at a concentration of 150
µ
g/mL to the photoporation cell medium.
After completing the photoporation procedure, the B16 melanoma cells were supplemented with
culture medium and placed back in the cell incubator. Six hours after photoporation, a significant
decline in viability was detected in the MLKL, caspase-3 and caspase-8 protein groups, as compared to
control cells that were photoporated in the absence of any of the three proteins (green bar, Figure 4). This
observation was consistent with confocal microscopy images of the cells (Figure 4A) and quantitative
CellTiter-Glo
®
cell viability data (Figure 4B). As cell viability was not aected in the MLKL setting
without VNB photoporation (MLKL ctrl, Figure 4A), the detected increased cell death in the MLKL
setting was caused by the delivery of the protein via VNB photoporation and not by a possible
perturbation of the cell membrane integrity by exogenous MLKL in the cell culture medium. Relative
cell viabilities of the protein sample groups, as compared to the photoporation control, show that
Int. J. Mol. Sci. 2019,20, 4254 6 of 14
functional protein delivery resulted in a significant drop in cell viability with 62%, 71% and 64% cell
survival for MLKL, caspase-8 and caspase-3, respectively (Figure 4C). These results indicate that VNB
photoporation can be used to directly and functionally deliver the protein MLKL, as well as caspases-3
and -8 and that this delivery induces cell death.
Int. J. Mol. Sci. 2019, 20, x FOR PEER REVIEW 6 of 14
MLKL setting without VNB photoporation (MLKL ctrl, Figure 4A), the detected increased cell death
in the MLKL setting was caused by the delivery of the protein via VNB photoporation and not by a
possible perturbation of the cell membrane integrity by exogenous MLKL in the cell culture medium.
Relative cell viabilities of the protein sample groups, as compared to the photoporation control, show
that functional protein delivery resulted in a significant drop in cell viability with 62%, 71% and 64%
cell survival for MLKL, caspase-8 and caspase-3, respectively (Figure 4C). These results indicate that
VNB photoporation can be used to directly and functionally deliver the protein MLKL, as well as
caspases-3 and -8 and that this delivery induces cell death.
We previously reported that transfection of mRNA encoding MLKL results in B16 melanoma
cell death within 16 h after transfection [24]. To probe the time responses of the induction of cell death
after photoporation with the MLKL protein itself, we analyzed the kinetics of cell death after MLKL
delivery. Cell viability of the B16 cells was determined 1 h, 2 h, 4 h and 6 h after MLKL transfection
(Figure 4D). Remarkably, maximal cell death induction was already observed within 1 h after protein
delivery and the cell viability remained virtually constant within the following hours. This
observation is not surprising considering that VNB photoporation enables direct cytoplasmic
delivery of MLKL, which serves as the key player in the final stage for execution of necroptotic cell
death [41].
Figure 4. Induction of cell death after caspase-8, caspase-3 and MLKL delivery. B16 cells were
transduced with MLKL, caspase-3 or caspase-8 (150 µg/mL) proteins by VNB photoporation (4 ×
10
7
AuNPs/mL). (A) Confocal images of B16 cells 6 h after protein delivery (scale bar = 100 µm). (B)
The graph represents the relative cell viability, as compared to the untreated control (Untreated), 6 h
Figure 4.
Induction of cell death after caspase-8, caspase-3 and MLKL delivery. B16 cells were
transduced with MLKL, caspase-3 or caspase-8 (150
µ
g/mL) proteins by VNB photoporation (4
×
10
7
AuNPs/mL). (
A
) Confocal images of B16 cells 6 h after protein delivery (scale bar =100
µ
m). (
B
)
The graph represents the relative cell viability, as compared to the untreated control (Untreated), 6 h
after protein delivery (n=3, independent experiments). B16 cells only illuminated with the pulsed laser
(Laser ctrl), cells incubated with the protein storage buer (Buer ctrl), cells incubated with the MLKL
protein solution (MLKL ctrl) and cells treated by VNB photoporation in absence of proteins (VNB ctrl)
were included as controls. (
C
) Relative cell viability, compared to VNB photoporation control (VNB ctrl,
green bar), 6 h after protein delivery (n=3, independent experiments). (
D
) Time response analysis of
cell death after MLKL delivery. Cell viability was determined 1 h, 2 h, 4 h and 6 h after MLKL delivery,
as compared to the untreated control (n=2, independent experiments). * p<0.05; ** p<0.01; *** p<
0.001; **** p<0.0001; ns =non-significant (one-way ANOVA with Dunett’s multiple comparison test).
We previously reported that transfection of mRNA encoding MLKL results in B16 melanoma cell
death within 16 h after transfection [
24
]. To probe the time responses of the induction of cell death
after photoporation with the MLKL protein itself, we analyzed the kinetics of cell death after MLKL
Int. J. Mol. Sci. 2019,20, 4254 7 of 14
delivery. Cell viability of the B16 cells was determined 1 h, 2 h, 4 h and 6 h after MLKL transfection
(Figure 4D). Remarkably, maximal cell death induction was already observed within 1 h after protein
delivery and the cell viability remained virtually constant within the following hours. This observation
is not surprising considering that VNB photoporation enables direct cytoplasmic delivery of MLKL,
which serves as the key player in the final stage for execution of necroptotic cell death [41].
2.4. Cell Death Induced by MLKL Delivery Has Necroptotic Hallmarks while Caspase-3/-8 Delivery Induces
Apoptotic-Like Cell Death
We next evaluated the mode of cell death of B16 cells after MLKL, caspase-3 and caspase-8 protein
delivery by photoporation. Transduction of B16 cells with caspase-3 protein resulted in cell death
with clear apoptotic features as the formation of apoptotic bodies and membrane blebbing of B16
cells was observed (Figure 5A). In contrast, transduction of the cells with MLKL elicited hallmarks of
necroptotic cell death, with clear cell swelling (oncosis) followed by plasma membrane permeabilization
(Figure 5A) [42].
Int. J. Mol. Sci. 2019, 20, x FOR PEER REVIEW 7 of 14
after protein delivery (n = 3, independent experiments). B16 cells only illuminated with the pulsed
laser (Laser ctrl), cells incubated with the protein storage buffer (Buffer ctrl), cells incubated with the
MLKL protein solution (MLKL ctrl) and cells treated by VNB photoporation in absence of proteins
(VNB ctrl) were included as controls. (C) Relative cell viability, compared to VNB photoporation
control (VNB ctrl, green bar), 6 h after protein delivery (n = 3, independent experiments). (D) Time
response analysis of cell death after MLKL delivery. Cell viability was determined 1 h, 2 h, 4 h and 6
h after MLKL delivery, as compared to the untreated control (n = 2, independent experiments). * p <
0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001; ns = non-significant (one-way ANOVA with Dunett’s
multiple comparison test).
2.4. Cell Death Induced by MLKL Delivery Has Necroptotic Hallmarks while Caspase-3/-8 Delivery Induces
Apoptotic-Like Cell Death
We next evaluated the mode of cell death of B16 cells after MLKL, caspase-3 and caspase-8
protein delivery by photoporation. Transduction of B16 cells with caspase-3 protein resulted in cell
death with clear apoptotic features as the formation of apoptotic bodies and membrane blebbing of
B16 cells was observed (Figure 5A). In contrast, transduction of the cells with MLKL elicited
hallmarks of necroptotic cell death, with clear cell swelling (oncosis) followed by plasma membrane
permeabilization (Figure 5A) [42].
Western blot analysis of the MLKL, caspase-3 and caspase-8 in cell lysates prepared 1 h after
protein delivery confirmed that all proteins were efficiently delivered to the cells and retained their
functional structure after VNB photoporation treatment (Figure 5B). In the incubation controls, in
which the protein was added to the cell medium but without photoporation, no protein was detected
in the lysates.
To exclude that the apoptotic pathway is involved in the observed cell death after MLKL
delivery via photoporation, a caspase activity assay was performed (Figure 5C). In line with the
necroptotic cell death phenotype, the caspase activity in B16 cells transduced with the MLKL protein
was comparable to the untreated condition. Instead, caspase activity was enhanced compared to
untreated cells when caspase-3 or -8 were photoporated into the B16 cells.
Figure 5. Characterization of cell death modality after protein transfection. (A) Differential
interference contrast light microscopy visualization of the cell morphology 30 min after caspase-3 and
Figure 5.
Characterization of cell death modality after protein transfection. (
A
) Dierential interference
contrast light microscopy visualization of the cell morphology 30 min after caspase-3 and MLKL
protein transduction (scale bars =20
µ
m). (
B
) Western blot analysis of caspase-3, caspase-8 and MLKL
protein in B16 cell lysates with or without (ctrl) VNB photoporation. MLKL, caspase-3, caspase-8 were
probed simultaneously using an anti-His detection antibody (exposure 30 min). Actin was visualized
using an anti-actin antibody (exposure time 5 min). The cropped blots are presented here, and their
full-length blots are included in Supplementary Figure S1. (
C
) Caspase activity in lysates of cells that
were transduced with MLKL, caspase-3 or caspase-8 by VNB photoporation. Statistical dierences
between relative caspase activity in the dierent samples were determined relative to the untreated
control. **** p<0.0001; ns =non-significant (one-way ANOVA with Dunett’s multiple comparison
test).
Western blot analysis of the MLKL, caspase-3 and caspase-8 in cell lysates prepared 1 h after
protein delivery confirmed that all proteins were eciently delivered to the cells and retained their
functional structure after VNB photoporation treatment (Figure 5B). In the incubation controls, in
which the protein was added to the cell medium but without photoporation, no protein was detected
in the lysates.
Int. J. Mol. Sci. 2019,20, 4254 8 of 14
To exclude that the apoptotic pathway is involved in the observed cell death after MLKL delivery
via photoporation, a caspase activity assay was performed (Figure 5C). In line with the necroptotic cell
death phenotype, the caspase activity in B16 cells transduced with the MLKL protein was comparable
to the untreated condition. Instead, caspase activity was enhanced compared to untreated cells when
caspase-3 or -8 were photoporated into the B16 cells.
3. Discussion
Tumors often comprise a network of both malignant and non-malignant cells. Although various
immune eector cells are recruited to the tumor site, in many cases, their anti-tumor functions are
downregulated, largely due to tumor-derived immunosuppressive signals. As a consequence, immune
cells in the tumor microenvironment fail to exert antitumor eector functions and the tumor escapes
from an attack by the host immune system. The immunosuppressive tumor microenvironment is a
major obstacle in cancer immunotherapy. Thanks to growing insights into the suppressive mechanism
of the tumor microenvironment, possible ways to block tumor escape are currently under investigation,
including the delivery at the site of the tumor of immune stimulatory molecules (e.g., OX40 ligand and
IL-23 [
3
]), blockade of inhibitory cytokines (e.g., TGF-
β
receptor II to capture TGF-
β
[
4
]), as well as
immune checkpoint blockade (e.g., PD-1 or CTLA-4 [
5
7
]). Recently, it became clear that by eliciting
immunogenic cell death (ICD), an immunogenic tumor environment can be created. ICD is a common
denominator for dierent cell death modalities that result in the release of damage-associated molecular
patterns (DAMPs). These DAMPs can, together with other immune-stimulatory components, recruit
and activate DCs in the tumor microenvironment [
43
45
]. It is shown that tumor cells succumbing from
irradiation, chemotherapeutics and hypericin-based photodynamic therapy undergo immunogenic
apoptosis [
8
12
]. In addition, necroptosis is considered as a type of cell death with immunogenic
properties [13,14].
In this study, we show that caspase-3 or -8, or MLKL protein delivery by VNB photoporation
induces, respectively, apoptotic and necroptotic cell death in murine B16 melanoma tumor cells. The
B16 tumor cell line is frequently used as a model for human melanoma. VNB photoporation represents
a very promising physical transfection technology, already shown to enable gentle intracellular delivery
of dierent types of macromolecules (e.g., siRNA) in a wide variety cell types, such as cancer cell
lines [
35
,
46
], primary lymphocytes [
36
,
40
] and neuronal cells [
39
]. By combination of absorbing gold
nanoparticles and a weakly focused laser beam, the technology also allows simultaneous intracellular
delivery of macromolecules in a large number of cells, while maintaining a high level of spatial
control [
35
,
47
]. First, we showed that VNB photoporation enables ecient delivery of FITC-dextrans
and FITC-BSA into B16 tumor cells. For this, B16 cells were incubated with dierent concentrations of
gold nanoparticles (AuNPs), followed by VNB generation by pulsed laser irradiation in the presence
of FITC-dextrans or FITC-BSA molecules. Considering that only a few gold nanoparticles adsorb
to each cell and the arising VNBs only aect neighboring structures very locally, damage to nearby
molecules is possible but negligible on the total molecule ensemble level. In general, we found
that the higher the used AuNP concentration, the higher the transduction ecacy and, accordingly,
the lower the cell viability. Delivery of caspase-3 by gold-nanoparticle-mediated laser transfection
(GNOME) has been reported in ZMTH3 cells (originating from a pleomorphic mammary adenoma),
by
Heinemann et al. [37].
The authors were able to show that the transduction of ZMTH3 cells with
caspase-3 resulted in cell death. Here, we confirmed that transduction of the caspase-3 protein in B16
tumor cells results in the induction of cell death. In addition, we have shown that the evoked cell
death has apoptotic features, i.e., cell blebbing and intracellular caspase activity. Furthermore, by
transducing B16 tumor cells with caspase-8 protein, another member of the cysteine protease family
which is implicated in apoptosis, cell death with apoptotic features was induced.
Next to apoptosis, there are other kinds of regulated cell death modalities, each with their own
specific features. For example, necroptosis is an interesting type of cell death as it has immunogenic
properties [
13
,
14
,
24
]. Necroptotic cells release damage-associated molecular patterns (DAMPs) and
Int. J. Mol. Sci. 2019,20, 4254 9 of 14
other immune-stimulatory components that, in an organism, can recruit immune cells to the dying cell.
This has very interesting implications in the context of the induction of an immune response against
tumor cells. Nonetheless, induction of necroptosis in tumor cells represents a major challenge, as many
tumor types display genetic and epigenetic alterations in the pathway, leading to necroptosis [
48
]. For
this reason, we opted to deliver the MLKL protein as a downstream executioner of the necroptotic
pathway. By performing protein transduction, manipulation of cells at the genetic level is circumvented.
We showed that it is possible to induce cell death with necroptotic features, such as cell swelling and
absence of caspase activity, via MLKL protein delivery by means of VNB photoporation in B16 cells.
As MLKL protein is the final mediator of necroptosis, direct intracellular delivery of the protein by
VNB photoporation very rapidly resulted in cell death. In conclusion, we demonstrated that direct
transduction of proteins involved in the executioner phase of either apoptosis or necroptosis allows
accurate and rapid control of the cell death modality.
4. Materials and Methods
4.1. Materials
Cationic gold nanoparticles (AuNPs) with a core size of 60 nm were synthetized in-house using
the Turkevich method, as reported before [
40
,
49
]. The AuNPs had a zeta potential of at least +30 mV,
as verified by dynamic light scattering (Malvern Instruments, Worcestershire, UK). FITC-dextran
70 kDa and FITC-BSA were purchased from Sigma-Aldrich (Bornem, Belgium), dissolved in DPBS-
(without MgCl
2
and CaCl
2
) at concentrations of, respectively, 50 mg/mL and 25 mg/mL and stored at
4
C until further use. The recombinant mouse MLKL protein (recombinant 6His, N-terminus) (full
length) was purchased from LifeSpan BioSciences (Seattle, Washington, USA) (catalog number/lot
number: LS-G23454/132989) and dissolved in a Tris, 50% glycerol buer. The recombinant mouse
caspase-3 (Casp-3, recombinant 6His, N-terminus) (full length) and caspase-8 (Casp-8, recombinant
6His, N-terminus) (full length) proteins were made in-house.
4.2. Cell Lines and Culture Conditions
Cells were cultured in Dulbecco’s modified Eagle’s medium supplemented with 10% of fetal calf
serum, 2 mM of L-glutamine, 0.4 mM of Na-pyruvate, non-essential amino acids, 100 U:mL of penicillin
and 0.1 mg/mL of streptomycin at 37
C in a humidified atmosphere containing 5% CO
2
. Murine
tumor cells used were melanoma cell lines (B16). These cells were purchased from the American Type
Culture collection (ATCC, LGC Standards Sarl, Molsheim Cedex, France) and cultured in conditions
specified by the manufacturer.
4.3. Measurement of FITC Fluorescence
FITC-dextrans and FITC-BSA were diluted in DPBS- to a final concentration of 1 mg/mL, and 100
µ
L
of these dilutions was transferred to a black 96-well plate (Greiner Bio-One). FITC fluorescence was
measured using a Victor3 multilabel reader (Perkin Elmer, Boston, MA, USA) with an excitation/emission
wavelength of 485/535 nm and measurement time of 0.1 sec.
4.4. Visualization and Quantification of AuNP Attachment
B16 cells (30
×
10
3
cells/well) were seeded in a glass-bottom 96-well plate (Greiner Bio-One,
Frickenhausen, Germany) and allowed to attach overnight at 37
C in a humidified atmosphere
containing 5% CO
2
. The cells were incubated for 30 min with dierent concentrations of AuNPs (37
C
and 5% CO
2
), followed by a wash step with DPBS- to remove unbound AuNPs. Visualization of
cell-attached AuNPs was performed by confocal reflection microscopy (C2, Nikon Benelux, Brussels,
Belgium) with a 60
×
water immersion lens at room temperature. Image processing was carried out
using the ImageJ software package (FIJI), and involved masking the AuNP scattering and dilation of
Int. J. Mol. Sci. 2019,20, 4254 10 of 14
the obtained mask. By counting the number of AuNPs and cells in each image, the average number of
AuNPs per cell was quantified.
4.5. Transfection by Vapor Nanobubble (VNB) Photoporation
For FITC-dextran or protein delivery by VNB photoporation, B16 cells (30 ×103cells/well) were
seeded in a 96-well plate and allowed to attach overnight at 37
C in a humidified atmosphere
containing 5% CO
2
. First, the cells were incubated for 30 min with AuNPs at a concentration dependent
on the specific experiment, followed by a washing step with DPBS- to remove unbound AuNPs.
Prior to the pulsed laser treatment, the specific cargo was diluted in culture medium (FITC-dextran,
FITC-BSA) or Opti-MEM
(MLKL, Casp-8, Casp-3) to the appropriate concentration and added to the
cells. For generation of VNBs, a homemade set-up was used [
35
,
49
]. This set-up consists of a pulsed
laser (~7ns) tuned at a wavelength of 561 nm (Opolette HE 355 LD, OPOTEK Inc) that was applied for
illumination of the cell-bound AuNPs and subsequent generation of VNBs. A laser fluence of 1.9 J/cm
2
,
i.e., above the VNB generation threshold [
35
,
36
], was used to assure the ecient formation of VNBs.
After treatment at room temperature, the cells were washed twice with DPBS- and supplied with fresh
culture medium (FITC-dextran, FITC-BSA), or immediately supplemented with fresh culture medium
(MLKL, Casp-8, Casp-3).
4.6. Viability Assay
B16 cells were treated using dierent concentrations of AuNPs, as described previously. After 2 h
of incubation, cell viability was determined using a CellTiter-Glo
®
luminescence cell viability assay
(Promega, Leiden, The Netherlands). For this, an equal volume of CellTiter-Glo®reagent was added
to the culture medium on the cells (at room temperature). The plate was shaken for 10 min using an
orbital shaker (100 rpm) and the content of each well was transferred to an opaque 96-well plate. The
luminescent signal was allowed to stabilize for 10 min, after which, the luminescence in each well was
recorded by a GloMaxLuminometer (Promega, Leiden, The Netherlands).
4.7. Confocal Microcopy
Visualization of B16 cells transfected with FITC-dextran 70 kDa, caspase-8, caspase-3 or MLKL was
performed using confocal laser scanning microscopy (C2, Nikon Benelux, Brussels, Belgium) and a 10
×
objective lens. Uptake of FITC dextran 70 kDa was detected using a 488 nm laser as excitation source.
4.8. Flow Cytometry
To evaluate the transfection eciency after photoporation by flow cytometry, the B16 cells were
detached by trypsin/EDTA (0.25%), centrifuged (500
×
g, 5 min) and resuspended in flow buer (DPBS-,
0.1% Sodium Azide, 1% Bovine Serum Albumine). Flow cytometry was performed using a CytoFLEX
(Beckman Coulter, Suarl
é
e, Belgium) flow cytometer and FlowJo
software (Treestar Inc.) was used
for data analysis.
4.9. DIC Visualization of Cell Death
For enhanced contrast visualization of cell death, B16 cells (100
×
10
3
cells/dish) were seeded in
50 mm
γ
-irradiated glass-bottom dishes (MatTek) and allowed to attach overnight. The cells were
transfected with MLKL, caspase-8 or caspase-3, as described above. After 30 min of incubation, cell
death was visualized by dierential interference contrast imaging using a 60
×
oil immersion objective
lens (NA =1.4, Nikon). Background correction was applied to the resulting images.
4.10. Western Blot
B16 cells (30
×
10
3
cells/well) were seeded in a 96-well plate and transfected with MLKL,
caspase-8 and caspase-3 24 h later, as described above. After photoporation of B16 cells, cells were
Int. J. Mol. Sci. 2019,20, 4254 11 of 14
washed to remove recombinant proteins that were not taken up by the cells via photoporation.
One hour after photoporation, cells were lysed and lysates were separated using sodium dodecyl
sulfate-polyacrylamide gel electrophoresis (10% acrylamide). MLKL, caspase-8 and caspase-3 were
visualized by Western blotting with anti-His antibody (1000
×
dilution) (Bio-rad AbD Serotec, Kidlington,
United Kingdom catalog number: MCA1396).
4.11. Caspase Activity
To analyze caspase activity, a FLUOstar OMEGA (BMG Labtech, Ortenberg, Germany) assay was
performed. Therefore, B16 cells (30
×
10
3
cells/well) were seeded in a 96-well plate and photoporated
24 h later. After photoporation, cells were washed and lysed in CFS-buer (pH 7.5) with 10 mM
DTT to sustain caspase activity. CFS-buer contained 10 mM Hepes, 220 mM mannitol, 68 mM
sucrose, 2 mM NaCl, 2 mM MgCl
2
, and 2.5 mM KH
2
PO
4
. Two hours after photoporation, 33 mM
of Ac-DEVD-AMC and Ac-IETD-AMC were added to the cells. Caspase activity was measured by
cleavage of Av-DEVD-AMC and Ac-IETD-AMC into fluorescent 7-amino-4methylcoumarin (AMC)
(excitation 355 nm, emission 460 nm).
4.12. Statistical Analysis
All statistical analyses are performed using the GraphPad Prism 8 software (La Jolla). Data sets
are represented as mean ±standard deviation.
5. Conclusions
In conclusion, we show that by using VNB photoporation it is feasible to deliver,
in vitro
,
caspase-3/-8 or MLKL protein to murine B16 tumor cells. Such an intervention induces cell death with
respectively apoptotic or necroptotic characteristics.
Supplementary Materials:
Supplementary materials can be found at http://www.mdpi.com/1422-0067/20/17/
4254/s1. Figure S1. Full-length blots corresponding to Figure 5B.
Author Contributions:
L.V.H., L.R., S.S., T.B., J.C.F., R.R., W.D., P.V., K.R., K.B. and X.S. planned the study. L.V.H.
and L.R. equally contributed to this work and performed the experiments. L.V.H., L.R., K.B. and X.S. were involved
in data analysis and interpretation, and wrote the manuscript. All authors reviewed and approved the manuscript
before submission.
Funding:
Funding by the European Research Council (ERC) under the European Union’s Horizon 2020 research
and innovation program (Grant No. 648124) is acknowledged with gratitude. S.S. acknowledges the support of
the VLAIO grant (HBC.2017.0542.)
Acknowledgments: L.V.H. is a junior assistant of the Department of Biomedical Molecular Biology.
Conflicts of Interest:
L.V.H. and X.S. are named as inventors on a patent application entitled “Mixed lineage
kinase domain like protein in immunotherapeutic cancer control”.
Abbreviations
MLKL Mixed-lineage kinase domain-like
RIPK receptor interacting protein kinase
VNB vapor nanobubble
AuNPs gold nanoparticles
FD70 FITC-dextran of 70 kDa
GNOME gold-nanoparticle-mediated laser transfection
References
1. Labi, V.; Erlacher, M. How cell death shapes cancer. Cell Death Dis. 2015,6, e1675. [CrossRef] [PubMed]
2.
Tang, D.; Kang, R.; Berghe, T.V.; Vandenabeele, P.; Kroemer, G. The molecular machinery of regulated cell
death. Cell Res. 2019,29, 347–364. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2019,20, 4254 12 of 14
3.
Hewitt, S.L.; Bai, A.; Bailey, D.; Ichikawa, K.; Zielinski, J.; Karp, R.; Apte, A.; Arnold, K.; Zacharek, S.J.;
Iliou, M.S.; et al. Durable anticancer immunity from intratumoral administration of IL-23, IL-36gamma, and
OX40L mRNAs. Sci. Transl. Med. 2019,11, eaat9143. [CrossRef] [PubMed]
4.
Van der Jeught, K.; Joe, P.T.; Bialkowski, L.; Heirman, C.; Daszkiewicz, L.; Liechtenstein, T.; Escors, D.;
Thielemans, K.; Breckpot, K. Intratumoral administration of mRNA encoding a fusokine consisting of
IFN-beta and the ectodomain of the TGF-beta receptor II potentiates antitumor immunity. Oncotarget
2014
,5,
10100–10113. [CrossRef] [PubMed]
5.
Hugo, W.; Zaretsky, J.M.; Sun, L.; Song, C.; Moreno, B.H.; Hu-Lieskovan, S.; Berent-Maoz, B.; Pang, J.;
Chmielowski, B.; Cherry, G.; et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy
in Metastatic Melanoma. Cell 2016,165, 35–44. [CrossRef] [PubMed]
6.
Ribas, A.; Dummer, R.; Puzanov, I.; VanderWalde, A.; Andtbacka, R.H.I.; Michielin, O.; Olszanski, A.J.;
Malvehy, J.; Cebon, J.; Fernandez, E.; et al. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration
and Improves Anti-PD-1 Immunotherapy. Cell 2017,170, 1109–1119.e10. [CrossRef] [PubMed]
7.
Tumeh, P.C.; Harview, C.L.; Yearley, J.H.; Shintaku, I.P.; Taylor, E.J.; Robert, L.; Chmielowski, B.; Spasic, M.;
Henry, G.; Ciobanu, V.; et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance.
Nature 2014,515, 568–571. [CrossRef] [PubMed]
8.
Obeid, M.; Tesniere, A.; Ghiringhelli, F.; Fimia, G.M.; Apetoh, L.; Perfettini, J.L.; Castedo, M.; Mignot, G.;
Panaretakis, T.; Casares, N.; et al. Calreticulin exposure dictates the immunogenicity of cancer cell death.
Nat. Med. 2007,13, 54–61. [CrossRef] [PubMed]
9.
Garg, A.D.; Krysko, D.V.; Verfaillie, T.; Kaczmarek, A.; Ferreira, G.B.; Marysael, T.; Rubio, N.; Firczuk, M.;
Mathieu, C.; Roebroek, A.J.; et al. A novel pathway combining calreticulin exposure and ATP secretion in
immunogenic cancer cell death. EMBO J. 2012,31, 1062–1079. [CrossRef]
10.
Galluzzi, L.; Kepp, O.; Kroemer, G. Enlightening the impact of immunogenic cell death in photodynamic
cancer therapy. EMBO J. 2012,31, 1055–1057. [CrossRef]
11.
Galluzzi, L.; Kepp, O.; Kroemer, G. Immunogenic cell death in radiation therapy. Oncoimmunology
2013
,2,
e26536. [CrossRef] [PubMed]
12.
Kroemer, G.; Galluzzi, L.; Kepp, O.; Zitvogel, L. Immunogenic cell death in cancer therapy. Annu. Rev.
Immunol. 2013,31, 51–72. [CrossRef] [PubMed]
13.
Yatim, N.; Jusforgues-Saklani, H.; Orozco, S.; Schulz, O.; Barreira da Silva, R.; Reis e Sousa, C.; Green, D.R.;
Oberst, A.; Albert, M.L. RIPK1 and NF-kappaB signaling in dying cells determines cross-priming of CD8(+)
T cells. Science 2015,350, 328–334. [CrossRef] [PubMed]
14.
Aaes, T.L.; Kaczmarek, A.; Delvaeye, T.; De Craene, B.; De Koker, S.; Heyndrickx, L.; Delrue, I.; Taminau, J.;
Wiernicki, B.; De Groote, P.; et al. Vaccination with Necroptotic Cancer Cells Induces Ecient Anti-tumor
Immunity. Cell Rep. 2016,15, 274–287. [CrossRef] [PubMed]
15.
Sun, L.; Wang, H.; Wang, Z.; He, S.; Chen, S.; Liao, D.; Wang, L.; Yan, J.; Liu, W.; Lei, X.; et al. Mixed lineage
kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell
2012
,148, 213–227.
[CrossRef] [PubMed]
16.
Zhao, J.; Jitkaew, S.; Cai, Z.; Choksi, S.; Li, Q.; Luo, J.; Liu, Z.G. Mixed lineage kinase domain-like is a key
receptor interacting protein 3 downstream component of TNF-induced necrosis. Proc. Natl. Acad. Sci. USA
2012,109, 5322–5327. [CrossRef] [PubMed]
17.
Murphy, J.M.; Czabotar, P.E.; Hildebrand, J.M.; Lucet, I.S.; Zhang, J.G.; Alvarez-Diaz, S.; Lewis, R.; Lalaoui, N.;
Metcalf, D.; Webb, A.I.; et al. The pseudokinase MLKL mediates necroptosis via a molecular switch
mechanism. Immunity 2013,39, 443–453. [CrossRef] [PubMed]
18.
Wang, H.; Sun, L.; Su, L.; Rizo, J.; Liu, L.; Wang, L.F.; Wang, F.S.; Wang, X. Mixed lineage kinase domain-like
protein MLKL causes necrotic membrane disruption upon phosphorylation by RIP3. Mol. Cell
2014
,54,
133–146. [CrossRef] [PubMed]
19.
Dondelinger, Y.; Declercq, W.; Montessuit, S.; Roelandt, R.; Goncalves, A.; Bruggeman, I.; Hulpiau, P.;
Weber, K.; Sehon, C.A.; Marquis, R.W.; et al. MLKL compromises plasma membrane integrity by binding to
phosphatidylinositol phosphates. Cell Rep. 2014,7, 971–981. [CrossRef]
20.
Cai, Z.; Jitkaew, S.; Zhao, J.; Chiang, H.C.; Choksi, S.; Liu, J.; Ward, Y.; Wu, L.G.; Liu, Z.G. Plasma membrane
translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat. Cell Biol.
2014
,16,
55–65. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2019,20, 4254 13 of 14
21.
Moriwaki, K.; Bertin, J.; Gough, P.J.; Orlowski, G.M.; Chan, F.K. Dierential roles of RIPK1 and RIPK3 in
TNF-induced necroptosis and chemotherapeutic agent-induced cell death. Cell Death Dis.
2015
,6, e1636.
[CrossRef] [PubMed]
22.
Colbert, L.E.; Fisher, S.B.; Hardy, C.W.; Hall, W.A.; Saka, B.; Shelton, J.W.; Petrova, A.V.; Warren, M.D.;
Pantazides, B.G.; Gandhi, K.; et al. Pronecrotic mixed lineage kinase domain-like protein expression is a
prognostic biomarker in patients with early-stage resected pancreatic adenocarcinoma. Cancer
2013
,119,
3148–3155. [CrossRef]
23.
He, L.; Peng, K.; Liu, Y.; Xiong, J.; Zhu, F.F. Low expression of mixed lineage kinase domain-like protein is
associated with poor prognosis in ovarian cancer patients. OncoTargets Ther. 2013,6, 1539–1543.
24.
Van Hoecke, L.; Van Lint, S.; Roose, K.; Van Parys, A.; Vandenabeele, P.; Grooten, J.; Tavernier, J.; De Koker, S.;
Saelens, X. Treatment with mRNA coding for the necroptosis mediator MLKL induces antitumor immunity
directed against neo-epitopes. Nat. Commun. 2018,9, 3417. [CrossRef] [PubMed]
25.
McKenzie, E.A.; Abbott, W.M. Expression of recombinant proteins in insect and mammalian cells. Methods
2018,147, 40–49. [CrossRef] [PubMed]
26.
Ford, K.G.; Souberbielle, B.E.; Darling, D.; Farzaneh, F. Protein transduction: An alternative to genetic
intervention? Gene Ther. 2001,8, 1–4. [CrossRef] [PubMed]
27.
Morris, M.C.; Depollier, J.; Mery, J.; Heitz, F.; Divita, G. A peptide carrier for the delivery of biologically
active proteins into mammalian cells. Nat. Biotechnol. 2001,19, 1173–1176. [CrossRef]
28.
Todorova, R. Estimation of methods of protein delivery into mammalian cells—A comparative study by
electroporation and bioporter assay. Prikl. Biokhim. Mikrobiol. 2009,45, 493–496. [CrossRef]
29.
Zassler, B.; Blasig, I.E.; Humpel, C. Protein delivery of caspase-3 induces cell death in malignant C6 glioma,
primary astrocytes and immortalized and primary brain capillary endothelial cells. J. Neurooncol.
2005
,71,
127–134. [CrossRef]
30.
Stewart, M.P.; Langer, R.; Jensen, K.F. Intracellular Delivery by Membrane Disruption: Mechanisms, Strategies,
and Concepts. Chem. Rev. 2018,118, 7409–7531. [CrossRef]
31.
Zhang, Y.; Roise, J.J.; Lee, K.; Li, J.; Murthy, N. Recent developments in intracellular protein delivery. Curr.
Opin. Biotechnol. 2018,52, 25–31. [CrossRef] [PubMed]
32.
Bruce, V.J.; McNaughton, B.R. Inside Job: Methods for Delivering Proteins to the Interior of Mammalian
Cells. Cell Chem. Biol. 2017,24, 924–934. [CrossRef] [PubMed]
33.
Xiong, W.; Liu, Y.; Jiang, L.J.; Zhou, Y.S.; Li, D.W.; Jiang, L.; Silvain, J.F.; Lu, Y.F. Laser-Directed Assembly of
Aligned Carbon Nanotubes in Three Dimensions for Multifunctional Device Fabrication. Adv. Mater.
2016
,
28, 2002–2009. [CrossRef] [PubMed]
34.
Lapotko, D. Optical excitation and detection of vapor bubbles around plasmonic nanoparticles. Opt. Express
2009,17, 2538–2556. [CrossRef] [PubMed]
35.
Xiong, R.; Raemdonck, K.; Peynshaert, K.; Lentacker, I.; De Cock, I.; Demeester, J.; De Smedt, S.C.; Skirtach, A.G.;
Braeckmans, K. Comparison of gold nanoparticle mediated photoporation: Vapor nanobubbles outperform
direct heating for delivering macromolecules in live cells. ACS Nano 2014,8, 6288–6296. [CrossRef] [PubMed]
36.
Wayteck, L.; Xiong, R.; Braeckmans, K.; De Smedt, S.C.; Raemdonck, K. Comparing photoporation and
nucleofection for delivery of small interfering RNA to cytotoxic T cells. J. Control. Release
2017
,267, 154–162.
[CrossRef] [PubMed]
37.
Heinemann, D.; Kalies, S.; Schomaker, M.; Ertmer, W.; Murua Escobar, H.; Meyer, H.; Ripken, T. Delivery
of proteins to mammalian cells via gold nanoparticle mediated laser transfection. Nanotechnology
2014
,25,
245101. [CrossRef] [PubMed]
38.
Liu, J.; Xiong, R.; Brans, T.; Lippens, S.; Parthoens, E.; Zanacchi, F.C.; Magrassi, R.; Singh, S.K.; Kurungot, S.;
Szunerits, S.; et al. Repeated photoporation with graphene quantum dots enables homogeneous labeling of live
cells with extrinsic markers for fluorescence microscopy. Light Sci. Appl. 2018,7, 47. [CrossRef] [PubMed]
39.
Xiong, R.; Verstraelen, P.; Demeester, J.; Skirtach, A.G.; Timmermans, J.P.; De Smedt, S.C.; De
Vos, W.H.; Braeckmans, K. Selective Labeling of Individual Neurons in Dense Cultured Networks with
Nanoparticle-Enhanced Photoporation. Front. Cell. Neurosci. 2018,12, 80. [CrossRef] [PubMed]
40.
Raes, L.; Hecke, C.V.; Michiels, J.; Stremersch, S.; Fraire, J.C.; Brans, T.; Xiong, R.; De Smedt, S.;
Vandekerckhove, L.; Raemdonck, K.; et al. Gold Nanoparticle-Mediated Photoporation Enables Delivery of
Macromolecules over a Wide Range of Molecular Weights in Human CD4+T Cells. Crystals
2019
,9, 411.
[CrossRef]
Int. J. Mol. Sci. 2019,20, 4254 14 of 14
41.
Chen, D.; Yu, J.; Zhang, L. Necroptosis: An alternative cell death program defending against cancer. Biochim.
Biophys. Acta 2016,1865, 228–236. [CrossRef] [PubMed]
42.
Majno, G.; Joris, I. Apoptosis, oncosis, and necrosis. An overview of cell death. Am. J. Pathol.
1995
,146, 3–15.
[PubMed]
43.
Hackl, H.; Charoentong, P.; Finotello, F.; Trajanoski, Z. Computational genomics tools for dissecting
tumour-immune cell interactions. Nat. Rev. Genet. 2016,17, 441–458. [CrossRef] [PubMed]
44.
Krysko, O.; Love Aaes, T.; Bachert, C.; Vandenabeele, P.; Krysko, D.V. Many faces of DAMPs in cancer
therapy. Cell Death Dis. 2013,4, e631. [CrossRef] [PubMed]
45.
Krysko, D.V.; Garg, A.D.; Kaczmarek, A.; Krysko, O.; Agostinis, P.; Vandenabeele, P. Immunogenic cell death
and DAMPs in cancer therapy. Nat. Rev. Cancer 2012,12, 860–875. [CrossRef] [PubMed]
46.
Xiong, R.; Joris, F.; Liang, S.; De Rycke, R.; Lippens, S.; Demeester, J.; Skirtach, A.; Raemdonck, K.;
Himmelreich, U.; De Smedt, S.C.; et al. Cytosolic Delivery of Nanolabels Prevents Their Asymmetric
Inheritance and Enables Extended Quantitative in Vivo Cell Imaging. Nano Lett.
2016
,16, 5975–5986.
[CrossRef] [PubMed]
47.
Xiong, R.; Drullion, C.; Verstraelen, P.; Demeester, J.; Skirtach, A.G.; Abbadie, C.; De Vos, W.H.; De Smedt, S.C.;
Braeckmans, K. Fast spatial-selective delivery into live cells. J. Control. Release
2017
,266, 198–204. [CrossRef]
48.
Krysko, O.; Aaes, T.L.; Kagan, V.E.; D’Herde, K.; Bachert, C.; Leybaert, L.; Vandenabeele, P.; Krysko, D.V.
Necroptotic cell death in anti-cancer therapy. Immunol. Rev. 2017,280, 207–219. [CrossRef] [PubMed]
49.
Vermeulen, L.M.P.; Fraire, J.C.; Raes, L.; De Meester, E.; De Keulenaer, S.; Van Nieuwerburgh, F.; De Smedt, S.;
Remaut, K.; Braeckmans, K. Photothermally Triggered Endosomal Escape and Its Influence on Transfection
Eciency of Gold-Functionalized JetPEI/pDNA Nanoparticles. Int. J. Mol. Sci.
2018
,19, 2400. [CrossRef]
[PubMed]
©
2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... laser source is an important factor affecting photoporation. Generally, three modes of laser, continuous-wave (cw) laser, femtosecond pulsed (FS) laser, and NS laser, are used for photoporation (Gu et al., 2014;Minai et al., 2016;van Hoecke et al., 2019;Hosseinpour & walsh, 2021). NS laser is widely used for intracellular delivery in recent years. ...
... Meaningfully, they disrupted biofilms by vNBs mediated photoporation to improve diffusion in biofilms of antibiotics and antimicrobial agents, which can be used to resolve antibiotic resistance and wound care (teirlinck et al., 2018; 2019). Further, they developed various nanoparticles, including layer-to-layer nanoparticles, nanostars layers, graphene, black phosphorus, and biodegradable polydopamine nanosensitizers, to mediate photoporation (liu et al., 2018;Pylaev et al., 2019;liu et al., 2020;Shaabani et al., 2021;wang et al., 2021;Harizaj et al., 2021c). in addition, Kevin Braeckmans's team has reported some excellent photoporation techniques to address biomedical issues (Sauvage et al., 2019;van Hoecke et al., 2019;Xu et al., 2020;Raes et al., 2021;Harizaj et al., 2021a;2021b). in their recent work, NS laser induced vNBs was employed to disrupt vitreous opacities. ...
... Some studies have shown that delivery efficiency increases with the increase of nanoparticles concentration, but the cell viability starts to decline beyond a certain threshold of nanoparticle concentration (Harizaj et al., 2021a;2021b;2021c). therefore, some methods, such as scanning electron microscope, confocal reflection microscopy, reflected light microscopy, and fluorescence-lifetime imaging, were used to quantify the number of nanoparticle attached on cell membrane (Patskovsky et al., 2015;Yao et al., 2017;Raes et al., 2019;van Hoecke et al., 2019;Patskovsky et al., 2020). the number of nanoparticles attached on cell membrane can affect threshold of vNBs generation, which decreases with the increase of nanoparticle concentration (wang et al., 2018). ...
Article
Full-text available
Nanosecond pulsed laser induced photoporation has gained increasing attention from scholars as an effective method for delivering the membrane-impermeable extracellular materials into living cells. Compared with femtosecond laser, nanosecond laser has the advantage of high throughput and low costs. It also has a higher delivery efficiency than continuous wave laser. Here, we provide an extensive overview of current status of nanosecond pulsed laser induced photoporation, covering the photoporation mechanism as well as various factors that impact the delivery efficiency of photoporation. Additionally, we discuss various techniques for achieving photoporation, such as direct photoporation, nanoparticles-mediated photoporation and plasmonic substrates mediated photoporation. Among these techniques, nanoparticles-mediated photoporation is the most promising approach for potential clinical application. Studies have already been reported to safely destruct the vitreous opacities in vivo by nanosecond laser induced vapor nanobubble. Finally, we discuss the potential of nanosecond laser induced phototoporation for future clinical applications, particularly in the areas of skin and ophthalmic pathologies. We hope this review can inspire scientists to further improve nanosecond laser induced photoporation and facilitate its eventual clinical application.
... This results in the release of cellular contents, such as DAMPs, which trigger inflammation and immune response. Eventually, cell death occurs [33]. In addition, mitochondria dysfunction, lack of energy, and DNA damage can also lead to necroptosis [34]. ...
... In many cases, apoptosis occurs simultaneously with non-apoptotic cell death, and various drugs can induce the coexistence of multiple cell death pathways [31,33,34,61]. PCD is a tightly regulated process, and the interplay between different types of cell death pathways highly relies on caspase. ...
Article
Full-text available
Melanoma is a highly malignant skin cancer that is known for its resistance to treatments. In recent years, there has been significant progress in the study of non-apoptotic cell death, such as pyroptosis, ferroptosis, necroptosis, and cuproptosis. This review provides an overview of the mechanisms and signaling pathways involved in non-apoptotic cell death in melanoma. This article explores the interplay between various forms of cell death, including pyroptosis, necroptosis, ferroptosis, and cuproptosis, as well as apoptosis and autophagy. Importantly, we discuss how these non-apoptotic cell deaths could be targeted as a promising therapeutic strategy for the treatment of drug-resistant melanoma. This review provides a comprehensive overview of non-apoptotic processes and gathers recent experimental evidence that will guide future research and eventually the creation of treatment strategies to combat drug resistance in melanoma.
... The instant expansion and collapse of vapor nanobubbles induce damage of adjacent plasma membranes by highpressure shock waves and fluid shear stress, facilitating the passive diffusion of cargoes. This technique achieves a transfection efficiency of 45% and a 5-fold increase in the number of transfected viable cells compared to electroporation in Jurkat T cells [82][83][84]. In recent years, ionizable lipid NP formulations have been refined to reduce cytotoxicity [85,86]. ...
Article
Full-text available
Hematological malignancies (HMs) encompass a diverse group of blood neoplasms with significant morbidity and mortality. Immunotherapy has emerged as a validated and crucial treatment modality for patients with HMs. Despite notable advancements having been made in understanding and implementing immunotherapy for HMs over the past decade, several challenges persist. These challenges include immune-related adverse effects, the precise biodistribution and elimination of therapeutic antigens in vivo, immune tolerance of tumors, and immune evasion by tumor cells within the tumor microenvironment (TME). Nanotechnology, with its capacity to manipulate material properties at the nanometer scale, has the potential to tackle these obstacles and revolutionize treatment outcomes by improving various aspects such as drug targeting and stability. The convergence of nanotechnology and immunotherapy has given rise to nano-immunotherapy, a specialized branch of anti-tumor therapy. Nanotechnology has found applications in chimeric antigen receptor T cell (CAR-T) therapy, cancer vaccines, immune checkpoint inhibitors, and other immunotherapeutic strategies for HMs. In this review, we delineate recent developments and discuss current challenges in the field of nano-immunotherapy for HMs, offering novel insights into the potential of nanotechnology-based therapeutic approaches for these diseases.
... Although much research has been conducted on the disruption of cell membranes to mediate the direct intracellular delivery of drugs, these studies have focused on achieving high delivery efficiencies while minimizing acute cytotoxicity [33][34][35][36]. Studies that have researched the properties of the PM have focused on only the increase or decrease of markers and proteins in the cytoplasm during the destruction of the cell membrane by strong physical stimuli. ...
... [9][10][11] Among different types of nanoparticles, gold nanoparticle (AuNP)-mediated photoporation has been extensively studied, [12][13][14][15][16][17] and the results have shown that the vapor nanobubbles (VNBs) induced by pulsed laser activated AuNPs are more efficient for intracellular delivery of extracellular materials, also called loading efficiency, compared to that due to the direct heating generated by the photothermal effect. 4,12,13,15,[18][19][20][21][22][23] Recently, VNBs have been applied to destroy vitreous opacities aimed for the treatment of ophthalmologic diseases 24 and have even been used in vivo to perforate the retinal ganglion cells for therapies of retinal degenerative diseases. 25 In these applications, a critical parameter is the transient membrane pore opening time after irradiation, during which the extracellular molecules can enter into cells before membrane is self-repaired. ...
Article
Full-text available
Significance: Resealing time based loading efficiency of optoporation is the key parameter for drug or gene delivery. This work describes a comparatively simple optical approach to directly measure the cell membrane resealing time of the gold nanoparticle mediated photoporation. Aim: To establish a membrane potential detection optical system, which can provide a direct measurement of resealing time of the optoporated cells. Approach: Voltage sensitive dye has been used to label the gold nanoparticle covered cell before laser activation and the resealing time was estimated from the voltage change due to the fluorescence light intensity change before and after laser activation. The approach has been validated by the simulated data based on diffusion model and Monte Carlo simulation and the experimental data obtained from a flow cytometry analysis. Results: The measured resealing time after perforation varied from 28.6 to 163.8 s on Hela cells when the irradiation fluence was increased, with a correlation coefficient (R2) of 0.9938. This result is in agreement with the resealing time (1-2 min) of photothermal porated Hela cells measured by electrical impedance method. The intracellular delivery efficiency of extracellular macromolecular under the same irradiation fluence depends mainly on diffusion velocity rather than pore size. Conclusion: The method described here can be used to directly measure resealing time of optoporated cells for accurately estimating the loading efficiency on discovering the mechanism of optoporation.
Article
Colorectal cancer is a common malignant tumor with high morbidity and mortality rates, imposing a huge burden on both patients and the healthcare system. Traditional treatments such as surgery, chemotherapy and radiotherapy have limitations, so finding more effective diagnostic and therapeutic tools is critical to improving the survival and quality of life of colorectal cancer patients. While current tumor targeting research mainly focuses on exploring the function and mechanism of molecular targets and screening for excellent drug targets, it is crucial to test the efficacy and mechanism of tumor cell therapy that targets these molecular targets. Selecting the appropriate drug carrier is a key step in effectively targeting tumor cells. In recent years, nanoparticles have gained significant interest as gene carriers in the field of colorectal cancer diagnosis and treatment due to their low toxicity and high protective properties. Nanoparticles, synthesized from natural or polymeric materials, are NM-sized particles that offer advantages such as low toxicity, slow release, and protection of target genes during delivery. By modifying nanoparticles, they can be targeted towards specific cells for efficient and safe targeting of tumor cells. Numerous studies have demonstrated the safety, efficiency, and specificity of nanoparticles in targeting tumor cells, making them a promising gene carrier for experimental and clinical studies. This paper aims to review the current application of nanoparticles in colorectal cancer diagnosis and treatment to provide insights for targeted therapy for colorectal cancer while also highlighting future prospects for nanoparticle development.
Article
Full-text available
Cancer immunotherapy has emerged as a promising approach for the induction of an antitumor response. While immunotherapy response rates are very high in some cancers, the efficacy against solid tumors remains limited caused by the presence of an immunosuppressive tumor microenvironment. Induction of immunogenic cell death (ICD) in the tumor can be used to boost immunotherapy response in solid cancers by eliciting the release of immune‐stimulatory components. However, the delivery of components inducing ICD to tumor sites remains a challenge. Here, a novel delivery method is described for antitumor therapy based on MLKL (Mixed Lineage Kinase Domain‐Like), a key mediator of necroptosis and inducer of ICD. A novel highly branched poly (β‐amino ester)s (HPAEs) system is designed to efficiently deliver MLKL plasmid DNA to the tumor with consequent enhancement of immune antigen presentation for T cell responses in vitro, and improved antitumor response and prolonged survival in tumor‐bearing mice. Combination of the therapy with anti‐PD‐1 treatment revealed significant changes in the composition of the tumor microenvironment, including increased infiltration of CD8⁺ T cells and tumor‐associated lymphocytes. Overall, the HPAEs delivery system can enhance MLKL‐based cancer immunotherapy and promote antitumor immune responses, providing a potential treatment to boost cancer immunotherapies.
Article
Full-text available
The modification of CD4+ T cells with exogenous nucleic acids or proteins is a critical step in several research and therapeutic applications, such as HIV studies and cancer immunotherapies. However, efficient cell transfections are not always easily achieved when working with these primary hard-to-transfect cells. While the modification of T cells is typically performed by viral transduction or electroporation, their use is associated with safety issues or cytotoxicity. Vapor nanobubble (VNB) photoporation with sensitizing gold nanoparticles (AuNPs) has recently emerged as a new technology for safe and flexible cell transfections. In this work, we evaluated the potential of VNB photoporation as a novel technique for the intracellular delivery of macromolecules in primary human CD4+ T cells using fluorescent dextrans as model molecules. Our results show that VNB photoporation enables efficient delivery of fluorescent dextrans of 10 kDa in Jurkat (>60% FD10+ cells) as well as in primary human CD4+ T cells (±40% FD10+ cells), with limited cell toxicity (>70% cell viability). We also demonstrated that the technique allows the delivery of dextrans that are up to 500 kDa in Jurkat cells, suggesting its applicability for the delivery of biological macromolecules with a wide range of molecular weights. Altogether, VNB photoporation represents a promising technique for the universal delivery of macromolecules in view of engineering CD4+ T cells for use in a wide variety of research and therapeutic applications.
Article
Full-text available
Cells may die from accidental cell death (ACD) or regulated cell death (RCD). ACD is a biologically uncontrolled process, whereas RCD involves tightly structured signaling cascades and molecularly defined effector mechanisms. A growing number of novel non-apoptotic forms of RCD have been identified and are increasingly being implicated in various human pathologies. Here, we critically review the current state of the art regarding non-apoptotic types of RCD, including necroptosis, pyroptosis, ferroptosis, entotic cell death, netotic cell death, parthanatos, lysosome-dependent cell death, autophagy-dependent cell death, alkaliptosis and oxeiptosis. The in-depth comprehension of each of these lethal subroutines and their intercellular consequences may uncover novel therapeutic targets for the avoidance of pathogenic cell loss.
Article
Full-text available
Many solid cancers contain dysfunctional immune microenvironments. Immune system modulators that initiate responses to foreign pathogens could be promising candidates for reigniting productive responses toward tumors. Interleukin-1 (IL-1) and IL-12 cytokine family members cooperate at barrier tissues after microbial invasion, in human inflammatory diseases, and in antitumoral immunity. IL-36γ, in classic alarmin fashion, acts in damaged tissues, whereas IL-23 centrally coordinates immune responses to danger signals. In this study, direct intratumoral delivery of messenger RNAs (mRNAs) encoding these cytokines produced robust anticancer responses in a broad range of tumor microenvironments. The addition of mRNA encoding the T cell costimulator OX40L increased complete response rates in treated and untreated distal tumors compared to the cytokine mRNAs alone. Mice exhibiting complete responses were subsequently protected from tumor rechallenge. Treatments with these mRNA mixtures induced downstream cytokine and chemokine expression, and also activated multiple dendritic cell (DC) and T cell types. Consistent with this, efficacy was dependent on Batf3-dependent cross-presenting DCs and cytotoxic CD8 ⁺ T cells. IL-23/IL-36γ/OX40L triplet mRNA mixture triggered substantial immune cell recruitment into tumors, enabling effective tumor destruction irrespective of previous tumoral immune infiltrates. Last, combining triplet mRNA with checkpoint blockade led to efficacy in models otherwise resistant to systemic immune checkpoint inhibition. Human cell studies showed similar cytokine responses to the individual components of this mRNA mixture, suggesting translatability of immunomodulatory activity to human patients.
Article
Full-text available
Cancer immunotherapy can induce durable antitumor responses. However, many patients poorly respond to such therapies. Here we describe a generic antitumor therapy that is based on the intratumor delivery of mRNA that codes for the necroptosis executioner mixed lineage kinase domain-like (MLKL) protein. This intervention stalls primary tumor growth and protects against distal and disseminated tumor formation in syngeneic mouse melanoma and colon carcinoma models. Moreover, MLKL-mRNA treatment combined with immune checkpoint blockade further improves the antitumor activity. MLKL-mRNA treatment rapidly induces T cell responses directed against tumor neo-antigens and requires CD4+ and CD8+ T cells to prevent tumor growth. Type I interferon signaling and Batf3-dependent dendritic cells are essential for this mRNA treatment to elicit tumor antigen-specific T cell responses. Moreover, MLKL-mRNA treatment blunts the growth of human lymphoma in mice with a reconstituted human adaptive immune system. MLKL-based treatment can thus be exploited as an effective antitumor immunotherapy.
Article
Full-text available
Plasmonic nanoparticles for drug delivery have attracted increasing interest over the last few years. Their localized surface plasmon resonance causes photothermal effects on laser irradiation, which allows for delivering drugs in a spatio-temporally controlled manner. Here, we explore the use of gold nanoparticles (AuNP) as carriers for pDNA in combination with pulsed laser irradiation to induce endosomal escape, which is currently considered to be one of the major bottlenecks in macromolecular drug delivery on the intracellular level. In particular, we evaluate nanocomplexes composed of JetPEI (polyethylenimine)pDNA and 10 nm AuNP, which do not exhibit endosomal escape by themselves. After incubating HeLa cells with these complexes, we evaluated endosomal escape and transfection efficiency using low- and high-energy laser pulses. At low laser energy heat is produced by the nanocomplexes, while, at higher laser energy, explosive vapour nanobubbles (VNB) are formed. We investigated the ability of heat transfer and VNB formation to induce endosomal escape and we examine the integrity of pDNA cargo after inducing both photothermal effects. We conclude that JetPEI/pDNA/AuNP complexes are unable to induce meaningful transfection efficiencies because laser treatment causes either dysfunctionality of the cargo when VNB are formed or forms too small pores in the endosomal membrane to allow pDNA to escape in case of heating. We conclude that laser-induced VNB is the most suitable to induce effective pDNA endosomal escape, but a different nanocomplex structure will be required to keep the pDNA intact.
Article
Full-text available
In the replacement of genetic probes, there is increasing interest in labeling living cells with high-quality extrinsic labels, which avoid over-expression artifacts and are available in a wide spectral range. This calls for a broadly applicable technology that can deliver such labels unambiguously to the cytosol of living cells. Here, we demonstrate that nanoparticle-sensitized photoporation can be used to this end as an emerging intracellular delivery technique. We replace the traditionally used gold nanoparticles with graphene nanoparticles as photothermal sensitizers to permeabilize the cell membrane upon laser irradiation. We demonstrate that the enhanced thermal stability of graphene quantum dots allows the formation of multiple vapor nanobubbles upon irradiation with short laser pulses, allowing the delivery of a variety of extrinsic cell labels efficiently and homogeneously into live cells. We demonstrate high-quality time-lapse imaging with confocal, total internal reflection fluorescence (TIRF), and Airyscan super-resolution microscopy. As the entire procedure is readily compatible with fluorescence (super resolution) microscopy, photoporation with graphene quantum dots has the potential to become the long-awaited generic platform for controlled intracellular delivery of fluorescent labels for live-cell imaging.
Article
Full-text available
Neurodevelopmental and neurodegenerative disorders are characterized by subtle alterations in synaptic connections and perturbed neuronal network functionality. A hallmark of neuronal connectivity is the presence of dendritic spines, micron-sized protrusions of the dendritic shaft that compartmentalize single synapses to fine-tune synaptic strength. However, accurate quantification of spine density and morphology in mature neuronal networks is hampered by the lack of targeted labeling strategies. To resolve this, we have optimized a method to deliver cell-impermeable compounds into selected cells based on Spatially resolved NAnoparticle-enhanced Photoporation (SNAP). We show that SNAP enables efficient labeling of selected individual neurons and their spines in dense cultured networks without causing cytotoxicity. We compare SNAP with widely used spine labeling techniques such as the application of lipophilic dyes and genetically encoded fluorescent markers. Using SNAP, we demonstrate a time-dependent increase in spine density in healthy cultures as well as a reduction in spine density after chemical mimicry of hypoxia. Since, the sparse labeling procedure can be automated using an intelligent acquisition scheme, SNAP holds promise for high-content screening campaigns of neuronal connectivity in the context of neurodevelopmental and neurodegenerative disorders.
Article
Intracellular delivery is a key step in biological research and has enabled decades of biomedical discoveries. It is also becoming increasingly important in industrial and medical applications ranging from biomanufacture to cell-based therapies. Here, we review techniques for membrane disruption-based intracellular delivery from 1911 until the present. These methods achieve rapid, direct, and universal delivery of almost any cargo molecule or material that can be dispersed in solution. We start by covering the motivations for intracellular delivery and the challenges associated with the different cargo types—small molecules, proteins/peptides, nucleic acids, synthetic nanomaterials, and large cargo. The review then presents a broad comparison of delivery strategies followed by an analysis of membrane disruption mechanisms and the biology of the cell response. We cover mechanical, electrical, thermal, optical, and chemical strategies of membrane disruption with a particular emphasis on their applications and challenges to implementation. Throughout, we highlight specific mechanisms of membrane disruption and suggest areas in need of further experimentation. We hope the concepts discussed in our review inspire scientists and engineers with further ideas to improve intracellular delivery.
Article
Purified recombinant proteins are key reagents in academic and industrial research. The ability to make these proteins quickly often relies on the availability of higher eukaryotic cell hosts such as insect and mammalian cells where there is a very wide range of post-translational modifications, protein folding and trafficking pathways. This enables the generation of many proteins that cannot be made in microbial hosts. In this article we outline some of the most commonly used methods to express recombinant proteins in insect and mammalian cells.