1, rico f. tabor2 michael e. d’angelo1, stefania 2 1, 3 ... · perforin binding and...

20
Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance Reveals a Role for Cholesterol and Calcium Independent Membrane Binding* Sarah E. Stewart 1 , Catherina H. Bird 1 , Rico F. Tabor 2 , Michael E. D’Angelo 1 , Stefania Piantavigna 2 , James C. Whisstock 1, 3 , Joseph A. Trapani 4 , Lisandra L. Martin 2 , Phillip I. Bird 1 1 The Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia 2 School of Chemistry, Monash University, Clayton, Australia 3 The ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia 4 Cancer Cell Death Laboratory, Cancer Immunology Program, Peter MacCallum Cancer Centre, St Andrew’s Place, East Melbourne, Victoria 3002, Australia *Running title: Perforin Binding and Rearrangements Assessed by QCM-D To whom correspondence should be addressed: Phillip I. Bird, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia, Tel: +61 3 9902 9365, fax: +61 3 9902 9500, mail: [email protected] Keywords: Perforin, pore, MACPF, Streptolysin O, calcium, cholesterol, Quartz crystal microbalance, membrane bilayer, protein self-assembly, cellular immune response ABSTRACT Perforin is an essential component in the cytotoxic lymphocyte mediated cell death pathway. The traditional view holds that perforin monomers assemble into pores in the target cell membrane via a calciumdependent process, and facilitate translocation of cytotoxic proteases into the cytoplasm to induce apoptosis. While many studies have examined the structure and role of perforin, the mechanics of pore assembly and granzyme delivery remain unclear. Here we have employed quartz crystal microbalance with dissipation monitoring (QCM-D) to investigate binding and assembly of perforin on lipid membranes, and show that perforin monomers bind to the membrane in a cooperative manner. We also found that cholesterol influences perforin binding and activity on intact cells and model membranes. Finally, contrary to current thinking, perforin efficiently binds membranes in the absence of calcium. When calcium is added to perforin already on the membrane, the QCM-D response changes significantly, indicating that perforin becomes membranolytic only after calcium binding. Cytotoxic lymphocytes have the capacity to eliminate virally infected or otherwise compromised cells by releasing stored cytotoxins from granules and/or by engaging death receptors. One stored cytotoxin is the pore- forming protein, perforin, which facilitates entry of other cytotoxins such as serine proteases (granzymes) into the target cell (1). Granule- mediated target cell death is absolutely dependent on perforin. Mice deficient in perforin are unable to control viral infection (2,3) and patients with inactivating mutations in the perforin gene present with the disease familial heamophagocytic lymphohistocytosis (3). At higher concentrations perforin is cytolytic in its own right. At lower doses, perforin synergizes with granzymes to induce apoptosis, however the mechanism by which perforin facilitates granzyme entry into the target cell remains a topic of debate. Perforin-induced membrane damage is essential for granzyme delivery (4) and it seems likely that both complete perforin pores and http://www.jbc.org/cgi/doi/10.1074/jbc.M115.683078 The latest version is at JBC Papers in Press. Published on November 5, 2015 as Manuscript M115.683078 Copyright 2015 by The American Society for Biochemistry and Molecular Biology, Inc. by guest on September 26, 2020 http://www.jbc.org/ Downloaded from

Upload: others

Post on 25-Jul-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

1

Analysis of Perforin Assembly by Quartz Crystal Microbalance Reveals a Role for Cholesterol and

Calcium Independent Membrane Binding*

Sarah E. Stewart1, Catherina H. Bird1, Rico F. Tabor2, Michael E. D’Angelo1, Stefania

Piantavigna2, James C. Whisstock1, 3, Joseph A. Trapani4, Lisandra L. Martin2, Phillip I. Bird1

1 The Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria

3800, Australia

2 School of Chemistry, Monash University, Clayton, Australia

3 The ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton,

Victoria 3800, Australia

4 Cancer Cell Death Laboratory, Cancer Immunology Program, Peter MacCallum Cancer Centre, St

Andrew’s Place, East Melbourne, Victoria 3002, Australia

*Running title: Perforin Binding and Rearrangements Assessed by QCM-D

To whom correspondence should be addressed: Phillip I. Bird, Department of Biochemistry and

Molecular Biology, Monash University, Clayton, Victoria 3800, Australia, Tel: +61 3 9902 9365, fax:

+61 3 9902 9500, mail: [email protected]

Keywords: Perforin, pore, MACPF, Streptolysin O, calcium, cholesterol, Quartz crystal

microbalance, membrane bilayer, protein self-assembly, cellular immune response

ABSTRACT

Perforin is an essential component in the

cytotoxic lymphocyte mediated cell death

pathway. The traditional view holds that perforin

monomers assemble into pores in the target cell

membrane via a calcium–dependent process, and

facilitate translocation of cytotoxic proteases into

the cytoplasm to induce apoptosis. While many

studies have examined the structure and role of

perforin, the mechanics of pore assembly and

granzyme delivery remain unclear. Here we have

employed quartz crystal microbalance with

dissipation monitoring (QCM-D) to investigate

binding and assembly of perforin on lipid

membranes, and show that perforin monomers

bind to the membrane in a cooperative manner.

We also found that cholesterol influences perforin

binding and activity on intact cells and model

membranes. Finally, contrary to current thinking,

perforin efficiently binds membranes in the

absence of calcium. When calcium is added to

perforin already on the membrane, the QCM-D

response changes significantly, indicating that

perforin becomes membranolytic only after

calcium binding.

Cytotoxic lymphocytes have the capacity

to eliminate virally infected or otherwise

compromised cells by releasing stored cytotoxins

from granules and/or by engaging death

receptors. One stored cytotoxin is the pore-

forming protein, perforin, which facilitates entry

of other cytotoxins such as serine proteases

(granzymes) into the target cell (1). Granule-

mediated target cell death is absolutely dependent

on perforin. Mice deficient in perforin are unable

to control viral infection (2,3) and patients with

inactivating mutations in the perforin gene

present with the disease familial

heamophagocytic lymphohistocytosis (3). At

higher concentrations perforin is cytolytic in its

own right. At lower doses, perforin synergizes

with granzymes to induce apoptosis, however the

mechanism by which perforin facilitates

granzyme entry into the target cell remains a topic

of debate. Perforin-induced membrane damage is

essential for granzyme delivery (4) and it seems

likely that both complete perforin pores and

http://www.jbc.org/cgi/doi/10.1074/jbc.M115.683078The latest version is at JBC Papers in Press. Published on November 5, 2015 as Manuscript M115.683078

Copyright 2015 by The American Society for Biochemistry and Molecular Biology, Inc.

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 2: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

2

incomplete arcs may contribute to the passage of

granzymes (5).

Perforin is a 60 kDa member of the

Membrane Attack Complex/Perforin (MACPF)

family of proteins. It comprises a MACPF

domain, an epidermal growth factor-like domain

and a C2 calcium-binding domain (6,7). Perforin

monomers oligomerize to form 16-20 nm

diameter pores in the plasma membrane of target

cell (8,9). Pore formation and subsequent cell

lysis is calcium-dependent, as demonstrated in

assays using red blood cells (RBCs) and

nucleated mammalian cells (10). Calcium binding

is important for the molecular stabilization of

perforin, inducing conformational changes that

expose several aromatic residues (involved in

membrane binding) in the C2 domain (11).

Perforin has structural similarities to the

bacterial cholesterol-dependent cytolysins

(CDCs). CDCs have been extensively studied and

a robust model describing their pore formation

exists (12). CDC monomers bind to the

membrane and oligomerize into a circular pre-

pore on the membrane surface. Conformational

changes then take place within each subunit. This

involves the unfurling of two key -helices to

form -strands, together with vertical collapse of

the pre-pore towards the membrane surface. In

the case of the well-studied CDC, Streptolysin O

(SLO), a large ~30 nm pore forms in the

membrane, allowing molecules of up to 150 kDa

to enter cells (13-16). One area of controversy

centers on the relevance of incomplete CDC rings

(also termed arcs), that are frequently observed in

various in vitro settings (13,14,17-20).

We recently investigated the membrane

binding and pore-forming activity of SLO using

Quartz Crystal Microbalance with Dissipation

monitoring (QCM-D) and Atomic Force

Microscopy (AFM), finding that binding is non-

cooperative and that arcs are common and stable

in the membrane (21). A recent study further

showed conclusively that these arcs constitute

functional pores at least in vitro (22).

In contrast to CDCs, perforin pores show

no indication of vertical collapse, suggesting that

there are significant differences in their pore-

forming mechanisms (9). Both perforin arcs and

full pores are commonly observed in vitro,

however, currently it is unclear whether perforin

forms a pre-pore complex prior to insertion into

the membrane, or if there is a threshold of

oligomer size before insertion occurs (4,8,23-27).

In this study we have used QCM-D to

investigate the binding and rearrangement of

perforin on membranes. We find that perforin

exhibits a complex binding mechanism, in which

cooperation between monomers occurs during the

binding process. Perforin binding and activity is

enhanced if cholesterol is present in the

membrane. Strikingly, we find that perforin binds

membranes in the absence of calcium, and that the

introduction of calcium to membrane-bound

perforin results in perforin rearrangement in the

membrane.

EXPERIMENTAL PROCEDURES

Reagents - 1,2-dimyristoyl-sn-glycero-3-

phosphocholine (DMPC) (cat. 850345P, Avanti

polar lipids), Cholesterol (Chol) (cat. C8667,

Sigma-Aldrich), 3-mercaptopropionic acid

(MPA) (cat. M5801, Sigma-Aldrich), 28%

ammonium hydroxide solution (cat. CAS 1336-

21-6, Ajax Finechem), 30% hydrogen peroxide

solution (cat. 108597, Merck Millipore), CaCl2

(cat. 10035-04-8, Ajax Finechem), HEPES (cat.

7365-45-9, Amresco),

Ethylenediaminetetraacetic acid (EDTA) (cat.

ED, Sigma-Aldrich), L-α-Phosphatidylcholine

from egg yolk (eggPC) (cat. P3556, Sigma-

Aldrich), Methyl-β-cyclodextrin (MβCD)

(Sigma-Aldrich), NaCl (cat. 7647-14-5,

Amresco), Nystatin (Sigma-Aldrich).

.

Buffers - Hepes buffered saline (HBS):

20 mM HEPES, pH 7.5, 150 mM NaCl; HEPES

buffered saline with calcium (HBS-Ca2+) as

above with 2 mM CaCl2; Low salt buffer (LS): 20

mM HEPES, pH 7.4, 30 mM NaCl; Low salt

buffer with calcium (LS-Ca2+) as per LS plus 2

mM calcium; HBS with EDTA: 20 mM HEPES,

pH 7.5, 150 mM NaCl and 2 mM EDTA. All

buffers were made up in double distilled water

(MilliQ water) and then filtered through a 0.22

µm filter and degassed. HBSS: 10 mM HEPES,

0.2% (w/v) BSA in Hank’s balanced salts

solution, pH 7.2; HE: 10 mM HEPES, pH 7.2, 0.2

% (w/v) BSA, 150 mM NaCl, 5 mM CaCl2.

Proteins - Recombinant mouse perforin

was produced as previously described (28).

Recombinant mouse perforin mutants, were

generously provided by Dr. Voskoboinik,

purified as previously described (10,29). The

SLO expression plasmid, a gift from Prof.

Bhakdi, was produced and purified as previously

described (18).

Liposome preparation - Liposomes were

prepared as described (21,30). Briefly, DMPC,

eggPC and cholesterol powder was dissolved to 5

mM stock solution in ethanol-free chloroform.

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 3: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

3

Lipids were mixed, at ratios of 40:60 (v/v)

DMPC:Chol or 50:50 (v/v) eggPC:Chol.

Chloroform was evaporated under a gentle stream

of nitrogen gas and remaining solvent removed by

vacuum desiccator for 2 hours. Lipids were then

stored at -20°C. On the day of use lipids were

rehydrated in 1 ml HBS or HBS-Ca2+ to a final

concentration of 0.5 mM at 37°C for 1 hour,

vortexed for 2-5 minutes and sonicated for 7 min

1-3 times at ~50°C. Liposome suspensions were

stored at 4°C for up to 7 days.

QCM chip cleaning and surface

modification - Gold coated sensor chips were

cleaned by incubating in a solution of ammonium

hydroxide:hydrogen peroxide:water (1:1:3 v/v) at

70°C for 15-20 min as described previously (30).

The gold surface was then washed and modified

with 1 mM MPA solution made up in propan-2-

ol for at least 1 hour at room temperature.

QCM experiments - QCM-D

measurements were performed using an E4

system with flow cells (Q-Sense, Sweden) as

described previously (21,30). Briefly, changes to

the resonance frequency (Δf) and energy

dissipation (ΔD) were measured simultaneously.

All plots presented here are of the 7th harmonic

unless stated. All experiments were conducted at

22°C. A lipid bilayer was achieved by deposition

of liposomes in HBS or HBS-Ca2+ until a change

in frequency of 25-30 Hz (31). Lipid layers were

washed with a low salt buffer (LS or LS-Ca2+) to

encourage osmotic stress to burst any intact

liposomes. After a baseline was established in the

buffer, protein was added to the system at a rate

of 50 µl/min. Once all the protein was used the

flow was stopped and an equilibration period,

with no flow, was monitored before a wash step

introduced to remove any unbound material from

the surface with HBS or HBS-Ca2+ at 300-500

µl/min.

sRBC lysis assays - Sheep erythrocytes

were washed in 0.9% saline solution to remove

any contaminating haemoglobin in the buffer.

The erythrocytes were then counted and

resuspended at 2x108 cells/ml. Perforin was

serially diluted in HBS±Ca2+ and 2x107 cells were

added to each dilution in a final volume of 200 µl.

This was then incubated at 37°C or 22°C for 20

mins and supernatant collected and its absorbance

measured at 405 nm to detect the release of

haemoglobin.

Mammalian cell lysis assay - Jurkat cells

were washed twice with HBSS then resuspended

at 2x106 cells/ml in the same buffer. Cells were

pre-treated with 5 mM MβCD or nystatin for 30

min at 37ºC then added to an equal volume of

perforin diluted in HE with 5 mM MβCD or

nystatin. Cell viability was determined by Trypan

Blue dye exclusion following a further 30 min

incubation at 37ºC.

Theoretical model - Change in mass

(Δm) on the surface of the chip can be calculated

according to the Sauerbrey equation (32):

Δm= -C(Δf/N)

Where C is the mass sensitivity constant

(in this case 17.7 ng/cm2 for a chip with a

fundamental frequency of 5 MHz) and N is the

harmonic/overtone number (the 7th was used).

The kinetic form of the Langmuir

adsorption model provides the simplest descriptor

for the dynamics of adsorption at a solid-solution

interface (33):

Γmax = kads(1–θ) [M·Cb+kdes θ / kads(1–

θ)+M] – kdes θ

where Γmax is the maximum adsorbed

material in mol/m2 (determined using the

Sauerbrey equation for a saturating

concentration), M is a mass transport factor that

represents how quickly molecules can get to the

surface in ms–1, kads is the rate of adsorption in ms–

1, kdes is the rate of desorption in mol m–2 s–1 and

Cb is the bulk concentration mol m–3. The

fractional surface coverage, θ, is calculated by

dividing the adsorbed amount at a given time and

Γ(t) by the maximum adsorbed amount (Γmax). At

low surface coverage, the adsorption process is

assumed to be broadly controlled by diffusion of

molecules to the surface, and so the initial slope

of θ vs t, i.e. dθ/dt is used to calibrate M (34).

RESULTS

Perforin binding and assembly measured

by QCM-D - To assess the binding and assembly

of perforin, gold-coated sensors were modified

with MPA and then treated with DMPC:Chol

liposomes to form a lipid bilayer on each of the

four sensors (31). A different concentration of

perforin was introduced to each chamber and

changes in dissipation and frequency were

monitored in real time. After the flow was

stopped, changes in the dissipation and frequency

were monitored for a further 20 min. A HBS-Ca2+

solution was introduced to remove non-

specifically bound material. In this system, a

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 4: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

4

decrease in frequency over time indicates that

mass is accumulating on the surface of the lipid

bilayer, and the change in frequency (Δf) is

proportional to the accrued mass on the sensor

according to the Sauerbrey equation (32).

Perforin showed a two-phase, concentration-

dependent binding to the lipid layer (Fig 1A).

Interestingly, at higher concentrations (100 and

200 nM), ∆f continued to increase after the flow

of perforin-containing solution ceased. This was

reproducible and may be due to a small amount of

perforin binding during the equilibration period

(as the chamber solution contains perforin). The

perforin-lipid interaction appears to be very stable

as there was no loss of mass evident during the

subsequent wash step (Fig 1A).

Changes in dissipation (∆D) in real time

were also measured. Dissipation is a measure of

the energy lost (dissipated) from the sensor: if the

fluidity of the surface increases it will be more

energy dissipating and thus ∆D increases. By

contrast, if the layer becomes more rigid it will

not absorb as much of the dissipating energy and

∆D will remain constant or decrease. This allows

us to assess how perforin changes the viscoelastic

properties of the lipid layer, presumably as

monomers oligomerize and insert. We observed a

small initial increase in ∆D, followed by a

decrease, then finally a large and sustained

increase, indicating that perforin binding and

activity changes the fluidity of the lipid layer in a

multi-phase manner (Fig 1A). Like Δf, ∆D also

increased during the equilibration period, which

indicates that protein already in the chamber

continues to change the lipid layer. Perforin data

plotted as ∆f versus ∆D (∆f-∆D or signature trace)

shows several transitions consistent with a multi-

step process (Fig 1B). The overall trend, is

upward and toward the right and is indicative of

protein-protein and/or protein-lipid organization

(35,36). The Δf-ΔD profile is complex and

suggest that there are several processes occurring.

Attributing specific steps to events or pore

formation is difficult. However, along with

protein-protein associations, there are regions

where the Δf-ΔD trace reflects a decrease (or no

change) in ΔD during an increase in Δf. These

features indicate that the membrane is more rigid,

possibly be due to protein insertion into the

membrane and/or conformational changes in the

protein structure.

To further analyse changes in the

frequency and dissipation data, the first derivative

was analysed for the concentrations of perforin

shown in Fig 1A. The first derivative emphasizes

changes in the gradient of the frequency and

dissipation. These changes reflect specific kinetic

events during protein binding to the membrane,

conformational changes to the protein on the

membrane, or in some cases other effects such as

densification or solvation. The first derivative of

the frequency data (Δf’) shows two minima,

indicating that two events occur as perforin

interacts with membrane. The first minimum

appears to become saturated at 100 nM perforin

as there is no further increase in the rate with 200

nM perforin. The second minimum is strongly

concentration-dependent and does not reach

saturation (Fig 1C). Similarly, the first derivative

of ΔD (ΔD’) also shows two maxima and a

minimum, which indicates the viscoelasticity of

the composite layer is changing due to perforin

(Fig 1C). A simple interpretation of the above

results is that the first event represents initial

binding of perforin monomers to the bilayer until

a critical concentration is reached, which leads to

the second, more rapid, event reflecting

reorganization / assembly of monomers into pores

on the surface. It is conceivable that the second

event accelerates as the forming oligomers offer

high(er) affinity binding sites for incoming

monomers.

Changes in frequency and dissipation are

measured using several different harmonics,

which are integer multiples of the fundamental

frequency of the quartz sensor. Harmonics enable

measurements at various depths from sensor’s

surface, at higher harmonics the magnitude of

oscillation is less and therefore these only probe

material close to the sensor surface. Analysis of

the harmonics, for a single perforin concentration

(100 nM), shows a very small but reproducible

difference in Δf over the four harmonics (3rd –

9th). This difference is a spreading effect post

binding, where the highest Δf value was in the 3rd

harmonic which decreases to through to the 9th

harmonic. This indicates that there was more

mass detected in the 3rd (further away from the

sensor surface), potentially on top of the surface

of the membrane, compared to the 9th (closer to

the sensor surface), which reflects perforin in the

membrane (Fig 1D). Comparison of the harmonic

data for ΔD shows a more defined yet even

increase over a small ΔD range (1.5x10-6),

evident after the initial binding period. This

suggests post binding protein rearrangement on

the membrane surface.

One possible explanation for the

variation in harmonics is that during perforin

oligomerization, monomers lose coupled water

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 5: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

5

molecules and become less energy dissipating on

the surface of the membrane compared to free

monomers. Upon perforin insertion, the majority

of the protein is still on the surface with a smaller

proportion in the membrane, closer to the sensor

surface. Thus the larger portion of the oligomer

on the surface appears heavier and more energy

dissipating. Hence the spread in the harmonics is

attributed to the proportions of protein on the

surface versus protein inserted in the membrane.

When analysed as Δf-ΔD, all the

harmonics follow the same trend, as expected,

and spread out after initial binding, where the 3rd

harmonic displays the largest shift to the right

(Fig 1E). An increase in mass and dissipation in

the Δf-ΔD plot is indicative of protein-protein

association (35,36). As all ∆f and ∆D harmonics

followed the same trend, only the 7th harmonic for

is displayed for comparison in subsequent

experiments.

To assess the kinetics of perforin binding

to the lipid membrane, the data were compared

with the Langmuir isotherm model of adsorption

for a similar overall rate. This comparison clearly

indicates that perforin adsorption does not obey

simple first-order kinetics, providing additional

evidence for a more complex, cooperative

process. Specifically, over the first 12 min (during

the initial binding period) perforin does not

follow the Langmuir isotherm model, and it is

evident there is more than one rate of binding or

more than one process occurring (Fig 1F). This

may indicate an element of cooperation in

perforin binding, or a surface rearrangement step.

It is possible that as monomers bind, they provide

higher affinity binding sites for incoming

monomers, thus increasing the rate of binding

once a critical surface coverage is achieved. More

complex kinetic modelling could not be

performed due to insufficient knowledge of

perforin oligomerization and surface interactions.

Perforin activity is enhanced in the

presence of cholesterol - Composition of the

membrane plays a significant role in binding

and/or activity of pore forming proteins (e.g. the

CDCs (37), pleurotolysin (38), and Vibrio

cholerae cytolysin (39)), however, the membrane

requirements for perforin pore formation remain

largely unstudied. It has been suggested that

cholesterol may play a role in dictating the type

of pore formed by perforin, where full barrel stave

pores predominate in high cholesterol membranes

and proteo-lipidic hemi-pores are more common

when the concentration of cholesterol is lower

(27,40).

To examine the requirement for

cholesterol in perforin-mediated lysis of

nucleated mammalian cells, we used methyl-β-

cyclodextrin (MβCD) to remove cholesterol from

the plasma membrane (41,42). Perforin-mediated

lysis of MβCD treated and untreated cells was

assessed by Trypan blue exclusion, and compared

to lysis by the CDC, SLO. As expected, SLO was

unable to lyse mammalian cells pre-treated with

MβCD, confirming effective extraction of

cholesterol from the plasma membrane (data not

shown). In comparison, perforin lysed cells pre-

treated with MβCD but, compared to untreated

cells, a five-fold higher concentration was

required to lyse 50% of the target cell population

(Fig 2A). This indicates that cholesterol plays an

important role in perforin pore formation on

mammalian cells. MβCD has been used with

perforin previously, but in the context of

nucleated cells where it inhibits endocytosis (4).

These results were concordant with ours in

showing that perforin activity is suppressed by

MβCD. Furthermore, this study showed that

MβCD reduces perforin binding to the plasma

membrane (4). We also pre-treated mammalian

cells with nystatin, which disrupts cholesterol-

rich plasma membrane microdomains but does

not remove cholesterol from the bilayer (42).

Interestingly, nystatin had no effect on perforin’s

lytic ability (Fig 2A); which suggests that

cholesterol must be present but not necessarily

specifically organized, in the membrane.

Next we used QCM-D to compare

perforin binding to DMPC or DMPC:Chol. This

showed perforin can bind to both model

membranes, however less perforin bound when

cholesterol was absent (Fig 2B). Significantly, the

trace for ΔD was substantially different with and

without cholesterol. There were more pronounced

fluctuations in ΔD when cholesterol was present,

indicating a larger impact on the structure and

fluidity of the membrane (Fig 2B). This was also

clearly evident in the Δf-ΔD signature trace (Fig

2C).

Analysis of first derivative of the

frequency data indicates that the two-phase

binding pattern remains evident when cholesterol

is absent, although the rates are lower (Fig 2D).

Changes in the first derivative of the dissipation

data show a similar trend, which are reduced in

amplitude (Fig 2D). This is consistent with the

observation that lysis of intact cells by perforin is

reduced but not abolished when cholesterol is

removed from the membrane. Overall, these

results suggest that although perforin does not

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 6: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

6

require cholesterol to bind membranes such as

DMPC, post-binding events such as assembly or

pore formation require lipid configurations

including free (non-sequestered) cholesterol.

Perforin binds to membranes in the

absence of calcium - Lysis by perforin is calcium-

dependent. It is currently thought that calcium is

required for initial binding of perforin to the

plasma membrane through the C2 domain. We

investigated the requirement of calcium for

binding and activity of perforin on DMPC:Chol

membranes by QCM-D. Surprisingly, we

observed that perforin efficiently binds

membranes in the absence of calcium (with 2 mM

EDTA added to chelate any trace calcium) (Fig

3A). Binding in the absence of calcium appeared

to be as stable as when calcium was present,

because little or no material was removed from

the membrane at a wash buffer flow rate of 300

µl/min (Fig 3A). By contrast, the changes in

dissipation were very different when calcium was

absent. In the presence of EDTA the dissipation

steadily decreased, indicating that the protein had

little or no effect on the viscoelastic properties of

the membrane (Fig 3A). This was also evident in

the Δf-ΔD signature traces (Fig 3B). Equivalent

results were obtained for perforin in HBS lacking

both calcium and EDTA, confirming that this

effect was due to the absence of calcium (data not

shown). This suggests that in the absence of

calcium, perforin can associate with the

membrane but cannot form pores. To ensure that

perforin used in this experiment was competent to

form pores, we tested flow-through solutions

from the QCM-D cells for activity by red blood

cell lysis assays. As expected, solutions

containing calcium lysed the red blood cells

whereas those lacking calcium (and containing

EDTA) did not (Fig 3C). When excess calcium (5

mM) was added to the EDTA-containing

solution, lytic activity was restored (Fig 3C).

To further investigate binding of perforin

to DMPC:Chol membranes in the absence of

calcium, we used an inactive perforin mutant

(D429A), which has one calcium binding residue

mutated. This mutant reportedly does not bind to

red blood cells and hence is non-lytic (10), which

was confirmed by red blood cell lysis assay (data

not shown). Using QCM-D, in the presence of

calcium, D429A bound to the membrane,

although much less efficiently than wild type

perforin (wt Pfn, Fig 3D). Strikingly, dissipation

data showed absolutely no impact on membrane

properties (Fig 3D). To further compare the

D429A binding and viscoelastic changes to the

membrane, we analysed the first derivatives of

the frequency and dissipation data (Fig 3E). The

Δf’ trace for D429A appeared somewhat similar

to wt Pfn but with slower and a less efficient

association with the membrane (Fig 3E). The ΔD’

analysis for D429A shows little change over time

(Fig 3E). This is in stark contrast to wt Pfn and

indicates that although D429A can (weakly) bind

to the membrane, post-binding events are

inhibited.

In the absence of calcium, D429A bound

DMPC:Chol similarly to wt Pfn, confirming that

perforin has calcium-independent membrane

binding capacity (Fig 3F). ΔD traces were

identical, indicating absence of post-binding

events (Fig 3F). Removal of unbound perforin,

with a wash step, and subsequent introduction of

calcium into the system resulted in a large change

in dissipation (and smaller change in frequency)

with wt Pfn but not D429A (Fig 3F). The first

derivatives for these data are very similar for wt

Pfn and D429A until calcium is introduced

producing the dramatic change in both Δf’ and

ΔD’ for wt Pfn but not D429A (Fig 3G and H).

Thus these results indicate that (i) perforin

associates with phospholipids independently of

calcium, (ii) membrane-bound perforin is able to

bind calcium, and (iii) that calcium is critical for

post-binding events in pore formation.

Decreasing the oligomerization rate

changes the QCM-D trace for perforin membrane

interactions - To identify the QCM-D signatures

of perforin assembly and pore formation we used

the point mutant R213E, which forms pores at a

slower rate than wt Pfn (29). The slower rate of

R213E lysis was confirmed using in an RBC lysis

assay at 22°C (QCM-D working temperature) for

40 min, (data not shown). Wt Pfn and R213E

were then analysed by QCM-D. The Δf data for

wt Pfn and R213E were comparable (Fig 4A),

indicating that the mode of membrane binding of

this mutant is unaffected. However there was a

small but reproducible increase in the rate of

binding of R213E (Fig 4A). The shape of the ΔD

trace for R213E was also comparable to wt Pfn,

and again the changes produced by the mutant

occurred slightly faster (Fig 4A). A small but

notable difference in both Δf and ΔD was evident

during the equilibration period (when the flow

was stopped), in that the wt Pfn traces continued

to increase over time but the R213E traces did not

(Fig 4A). These differences were further

highlighted in the Δf-ΔD signature trace where

the R213E curve followed a similar path as the wt

Pfn curve, being only slightly temporally

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 7: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

7

displaced (events occurring slightly earlier). In

addition, there was no final extension in the trace

of the R213E mutant as seen for wt Pfn (Fig 4B).

The first derivatives of the frequency and

dissipation for R213E show that the second

process for both Δf’ and ΔD’ shifted to the left

and increased in rate compared to wt Pfn (Fig

4C). This suggests that the second process (or at

least part of the second process) reports perforin

oligomerization/insertion in the membrane.

When the data was plotted against the Langmuir

isotherm model, it was evident that the R213E

mutant resembles wt Pfn until approximately 8

min when it diverges (Fig 4D). To analyse these

differences further, increasing concentrations of

R213E mutant where tested (Fig 4E). These data

show that at the highest concentration (200 nM)

the R213E mutant does not induce an increase in

ΔD once the flow has ceased. This contrasts with

wt Pfn, where changes in ΔD during this period

were quite pronounced (Fig 4E and Fig 1A). Once

again, when plotted as Δf-ΔD, clearly the Δf-ΔD

signatures are missing a final “extension” at the

end of the trace (Fig 4F), similar to 50 nM wt Pfn

(Fig 1B). Finally, the first derivatives for Δf and

ΔD data once again indicated that R213E behaves

similarly, but not identically, to wt Pfn. A notable

difference is the decreased magnitude of the ΔD’

response during the incubation period (Fig 4G)

compared with 200 and 100 nM wt Pfn (Fig 1C).

Together, these results suggest that the rate of

oligomerization is reflected by the QCM-D

signature of perforin, and that post-binding events

can be investigated using this approach.

Perforin and SLO differ in their

membrane binding and pore forming

characteristics - Previously we characterized

SLO binding and pore formation using QCM-D

(21). The data obtained for SLO can be compared

with the results for perforin as they were obtained

under similar conditions, on Au-MPA with

DMPC:Chol (50:50). However, it is important to

note the considerable size difference between

recombinant SLO and perforin. Perforin is

expressed as a hexa-histidine tagged monomer of

60 kDa, whereas SLO is a MBP fusion protein

that is 97 kDa. Therefore, it is expected that the

same concentration of SLO would produce a

higher Δf maxima than perforin due to its larger

hydrated mass per molecule. This was indeed the

case, despite obvious differences in binding rates:

at the same concentration SLO has a larger Δf

maxima than perforin (Fig 5A). Additionally, ΔD

appears to be substantially different, again SLO

has a significantly larger ΔD compared with

perforin (Fig 5A). This implies that the larger

SLO molecule is more energy dissipating than

perforin, as expected. This is also true for the

oligomeric structures formed by SLO vs perforin.

The large SLO pores would cause a higher ΔD in

comparison to the smaller oligomers that are

formed by perforin (approximately half the size).

Comparison of the signature QCM-D Δf-

ΔD plots revealed obvious differences between

SLO and perforin, not only in magnitude but also

in their overall membrane binding trends (Fig

5B). In particular, SLO binds in an energy

dissipating manner in 3 stages, whereas perforin

shows complex multi-staged processes, two of

which are consistent with a stiffer, less energy

dissipating surface. This comparison suggests

that the mechanism of pore formation differs

between SLO and perforin, and that the

membrane responds differently as a consequence.

Comparison of the first derivative of SLO

and perforin functions reveal both traces display

two processes or events (maxima) for Δf’ at

similar time points (Fig 5C). Analysis of Δf’ for

SLO showed that the second process was related

to oligomerization (21). This may also be the case

for perforin given the similarities, however, this

remains speculative. While the Δf’ traces appear

similar, there are important discrepancies such as

the difference in the magnitude of the two

processes (minima). SLO displays two similar

events of similar magnitude which both increase

with increasing concentration, whereas the first

process for perforin is concentration independent

and only the second increases with concentration

(Fig 5C). The ΔD’ traces show that initially

perforin differs from SLO, with a decrease in ΔD’

below zero that is never seen for SLO, however,

this is followed by a subsequent process

(positive), which is a common feature at

approximately the same time point (Fig 5C).

Finally, when plotted against the

Langmuir isotherm model for single order

binding, it is obvious that only SLO follows a first

order kinetic model, suggesting there is an

element of cooperation or surface rearrangement

in perforin binding that is very different to SLO

(Fig 5D). These data highlight the complexity of

pore formation and suggest that while there are

similarities between SLO and perforin, the

mechanism of pore formation is different.

Comparing individual SLO and perforin

traces shows that there are also differences in the

response of the harmonics in both Δf and ΔD (Fig

5 E and F). In the Δf trace for 100 nM SLO there

is a spread in harmonic data, with the 9th being

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 8: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

8

smallest and increasing to the 3rd (Fig 5E). In

contrast, perforin shows less spread in Δf (Fig

5F). Comparing ΔD harmonic data for SLO they

essentially overlay, whereas, perforin shows a

large spread with the 9th being the most energy

dissipating. These differences indicate that SLO

and perforin are interacting with the membrane in

subtle but distinctly different ways. The spread in

harmonics in Δf indicates that there is mass

through the lipid layer, whereas, a spread in ΔD

indicates that the layer is not equally as

viscoelastic throughout the layer. The spread in

Δf may be larger for SLO due to the increased

mass of the individual molecules and oligomers,

compared to perforin, which shows a less

prominent spreading (but it is still evident). Both

traces indicate that there is likely to be protein

insertion into the membrane, however,

differences in the behaviour of the harmonics in

ΔD indicates that the effect (or

organization/activity) on the membrane differs.

DISCUSSION

We have used QCM-D to investigate the

association and interaction of perforin with

membranes. We find that perforin binds to

membranes in a multi-step process: binding (but

not subsequent events) is possible in the absence

of calcium, and cholesterol is important for

activity. These results help to further dissect the

mechanism of pore formation by perforin.

Role of cholesterol and calcium in

perforin–membrane interactions - Our results

indicate that cholesterol affects the lytic activity

of perforin, although unlike CDCs it is not

essential for binding. There are mixed reports on

the importance of cholesterol for perforin

function. Early work suggested that the absence

of cholesterol in liposomes makes no difference

to perforin pore formation (43,44). Recently,

however, it has been suggested that cholesterol

dictates the type of pore formed by perforin. In

cholesterol rich membranes, fully formed (barrel

stave) pores are the dominant species, whereas

incomplete proteo-lipidic pores occur more often

in membranes that have less cholesterol (27,40).

We suggest that cholesterol contributes to bilayer

structure or fluidity to provide an optimal binding

and assembly stratum for perforin.

Perforin requires calcium for lytic and

granzyme delivery function. Our experiments

show perforin binding to a phospholipid bilayer

can occur in the absence of calcium, suggesting

that calcium-independent association of

individual perforin monomers with membrane

may contribute to perforin’s function, in addition

to calcium-mediated monomer-monomer binding

and/or rearrangement on the membrane. Recent

structural studies suggest perforin has three

calcium-chelating sites in its C2 domain, two high

and one low affinity site (9,11). The two high

affinity sites are likely occupied at cytoplasmic

calcium concentrations, whereas the low affinity

site is occupied only at higher calcium

concentrations (likely >0.2 mM). D429 is an

important residue in the low affinity site

responsible for a conformational change which

allows repositioning of several residues, thus

potentiating binding of aromatic side chains at the

tip of the C2 domain to the membrane (11). In our

studies, stable calcium-independent binding is

evident for both wt Pfn and D429A, however, it

is not associated with changes to the

viscoelasticity of the lipid bilayer. When calcium

is introduced, wt Pfn produces a large change in

energy dissipation that is not seen with D429A.

This increase in the dissipation is probably due to

rearrangement of perforin into oligomers and/or

insertion into the lipid layer.

Oligomerization - Perforin

oligomerization is proposed to be facilitated by

electrostatic interaction between subunits (9,29).

Several residues are known to play a role in this

interaction, including R213. The QCM response

suggests the slowly oligomerizing mutant R213E

exhibits a subtle but faster change to the second

stage of perforin binding, as highlighted by the

first derivative data (Δf’). Unlike wt Pfn, there is

no steady increase in the dissipation (or

frequency) once the mutant binds to the

membrane, which is possibly due to a delay in

oligomerization and insertion into the membrane.

If binding is cooperative, delayed insertion may

stabilize or increase binding opportunities for free

monomers, explaining the faster second stage.

Model for pore formation - Models for

perforin pore formation have been guided by

those built to explain CDC pores. The current

model for CDC pore formation invokes the

assembly of monomers into a stable circular

intermediate (the pre-pore), which then collapses

and inserts into membrane. This model dictates

that insertion into the membrane only occurs after

complete ring formation is achieved. However, to

date a perforin pre-pore has not been described,

and by cryo-EM inserted pores show no signs of

vertical collapse (9).

Our results further highlight differences

between perforin and SLO (a CDC). This is

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 9: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

9

demonstrated by the differences in both

frequency and dissipation when each toxin

interacts with a lipid layer. Furthermore, these

differences are clearest when the frequency and

dissipation are plotted with respect to each other

(Δf-ΔD), showing that they do not overlay nor do

they follow the same trend. In addition, it seems

that the order of binding differs between SLO and

perforin, where SLO displays first order kinetics

and perforin does not. Taken together this

suggests that their mechanisms of binding and

pore formation are diverse.

The mechanism of perforin pore

formation is still elusive and many questions

remain. Based on our work, any model must now

incorporate a requirement for a specific

phospholipid configuration including cholesterol,

and accommodate cooperative monomer binding

(Fig 5G). In addition, a more complex role for

calcium should be considered as it appears to

function in both binding and post-binding events.

(Fig 5G). Whether perforin forms complete pre-

pore intermediates and then inserts into the

membrane remains unclear: it is possible that

monomers are able to insert into the membrane as

they join the growing assembly, and this creates

transient proteo-lipidic pores that may become

complete pores or remain incomplete (Fig 5G). It

has recently been reported that AFM images of

perforin indeed include a mixture of arcs and fully

formed rings (45). The full rings have a mean

diameter of 20 nm which is similar to previous

reports using EM (8,9,23-27,45).

CONCLUSION

This study reveals that the kinetics of

perforin monomer binding to membranes is a

complex cooperative process that involves

calcium and cholesterol for optimal activity.

QCM-D shows that perforin does not exhibit first

order binding, suggesting cooperation between

monomers during the binding process. Perforin

binding and activity is enhanced when cholesterol

is present in the membrane. Strikingly, perforin

binds membranes in the absence of calcium, and

the introduction of calcium to membrane-bound

perforin results in perforin chelating calcium and

subsequent rearrangement of the membrane.

QCM-D offers a new approach to understanding

the dynamics of pore formation, and evaluating

therapeutic candidates that potentiate or inhibit

perforin binding and activity.

ACKNOWLEDGEMENTS: We thank Dr. Ilia

Voskoboinik and Mrs Annette Ciccone, (Peter

MacCallum Cancer Centre, East Melbourne,

Victoria 3002, Australia) for kindly providing

recombinant perforin mutants. Thanks to Prof.

Bhakdi for providing the pMAL-SLO plasmid.

This work was supported by grants to PIB and

JCW from the National Health and Medical

Research Council (Australia) and LLM, RFT and

JCW from the Australian Research Council.

CONFLICT OF INTEREST: The authors

declare that they have no conflicts of interest with

the contents of this article.

AUTHOR CONTRIBUTIONS: PIB and SES

conceived and coordinated the study with

involvement of LLM and MED. SES performed

the QCM-D experiments. The paper was written

by SES and PIB with contributions from LLM,

JCW, MED and RFT. SP and RFT provided

expertise in QCM-D and theoretical analysis,

respectively. CHB performed and analysed

experiments shown in Figure 2A. JAT provided

recombinant wt perforin and expert advice. All

authors reviewed the results and approved the

final version of the manuscript.

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 10: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

10

REFERENCES

1. Peters, P., Borst, J., Oorschot, V., Fukuda, M., Krahenbuhl, O., Tschopp, J., Slot, J., and Geuze, H. (1991) Cytotoxic T lymphocyte granules are secretory lysosomes, containing both perforin and granzymes. J. Exp. Med. 173, 1099-1109

2. Kagi, D., Ledermann, B., Burki, K., Seiler, P., Odermatt, B., Olsen, K. J., Podack, E. R., Zinkernagel, R. M., and Hengartner, H. (1994) Cytotoxicity mediated by T cells and natural killer cells is greatly impaired in perforin-deficient mice. Nature 369, 31-37

3. Stepp, S. E., Dufourcq-Lagelouse, R., Le Deist, F., Bhawan, S., Certain, S., Mathew, P. A., Henter, J. I., Bennett, M., Fischer, A., de Saint Basile, G., and Kumar, V. (1999) Perforin gene defects in familial hemophagocytic lymphohistiocytosis. Science 286, 1957-1959

4. Lopez, J. A., Susanto, O., Jenkins, M. R., Lukoyanova, N., Sutton, V. R., Law, R. H., Johnston, A., Bird, C. H., Bird, P. I., Whisstock, J. C., Trapani, J. A., Saibil, H. R., and Voskoboinik, I. (2013) Perforin forms transient pores on the target cell plasma membrane to facilitate rapid access of granzymes during killer cell attack. Blood 121, 2659-2668

5. Stewart, S. E., D'Angelo, M. E., and Bird, P. I. (2012) Intercellular communication via the endo-lysosomal system: translocation of granzymes through membrane barriers. Biochim. Biophys. Acta. 1824, 59-67

6. Lichtenheld, M. G., Olsen, K. J., Lu, P., Lowrey, D. M., Hameed, A., Hengartner, H., and Podack, E. R. (1988) Structure and function of human perforin. Nature 335, 448-451

7. Ponting, C. P., and Parker, P. J. (1996) Extending the C2 domain family: C2s in PKCs δ, ϵ, η, θ, phospholipases, GAPs, and perforin. Protein Sci. 5, 162-166

8. Dourmashkin, R. R., Deteix, P., Simone, C. B., and Henkart, P. (1980) Electron microscopic demonstration of lesions in target cell membranes associated with antibody-dependent cellular cytotoxicity. Clin. Exp. Immunol. 42, 554-560

9. Law, R. H., Lukoyanova, N., Voskoboinik, I., Caradoc-Davies, T. T., Baran, K., Dunstone, M. A., D'Angelo, M. E., Orlova, E. V., Coulibaly, F., Verschoor, S., Browne, K. A., Ciccone, A., Kuiper, M. J., Bird, P. I., Trapani, J. A., Saibil, H. R., and Whisstock, J. C. (2010) The structural basis for membrane binding and pore formation by lymphocyte perforin. Nature 468, 447-451

10. Voskoboinik, I., Thia, M. C., Fletcher, J., Ciccone, A., Browne, K., Smyth, M. J., and Trapani, J. A. (2005) Calcium-dependent plasma membrane binding and cell lysis by perforin are mediated through its C2 domain: A critical role for aspartate residues 429, 435, 483, and 485 but not 491. J. Biol. Chem. 280, 8426-8434

11. Traore, D. A., Brennan, A. J., Law, R. H., Dogovski, C., Perugini, M. A., Lukoyanova, N., Leung, E. W., Norton, R. S., Lopez, J. A., Browne, K. A., Yagita, H., Lloyd, G. J., Ciccone, A., Verschoor, S., Trapani, J. A., Whisstock, J. C., and Voskoboinik, I. (2013) Defining the interaction of perforin with calcium and the phospholipid membrane. Biochem. J. 456, 323-335

12. Dunstone, M. A., and Tweten, R. K. (2012) Packing a punch: the mechanism of pore formation by cholesterol dependent cytolysins and membrane attack complex/perforin-like proteins. Curr. Opin. Struct. Biol. 22, 342-349

13. Bhakdi, S., Tranum-Jensen, J., and Sziegoleit, A. (1985) Mechanism of membrane damage by streptolysin-O. Infect. Immun. 47, 52-60

14. Palmer, M., Harris, R., Freytag, C., Kehoe, M., Tranum-Jensen, J., and Bhakdi, S. (1998) Assembly mechanism of the oligomeric streptolysin O pore: the early membrane lesion is lined by a free edge of the lipid membrane and is extended gradually during oligomerization. EMBO J. 17, 1598-1605

15. Walev, I., Bhakdi, S. C., Hofmann, F., Djonder, N., Valeva, A., Aktories, K., and Bhakdi, S. (2001) Delivery of proteins into living cells by reversible membrane permeabilization with streptolysin-O. Proc Natl Acad Sci U S A 98, 3185-3190

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 11: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

11

16. Stewart, S. E., Kondos, S. C., Matthews, A. Y., D'Angelo, M. E., Dunstone, M. A., Whisstock, J. C., Trapani, J. A., and Bird, P. I. (2014) The Perforin Pore Facilitates the Delivery of Cationic Cargos. J. Biol. Chem. 289, 9172-9181

17. Harris, J. R., Adrian, M., Bhakdi, S., and Palmer, M. (1998) Cholesterol-Streptolysin O Interaction: An EM Study of Wild-Type and Mutant Streptolysin O. Journal of structural biology 121, 343-355

18. Weller, U., Muller, L., Messner, M., Palmer, M., Valeva, A., Tranum-Jensen, J., Agrawal, P., Biermann, C., Dobereiner, A., Kehoe, M. A., and Bhakdi, S. (1996) Expression of active streptolysin O in Escherichia coli as a maltose-binding-protein--streptolysin-O fusion protein. The N-terminal 70 amino acids are not required for hemolytic activity. Eur. J. Biochem. 236, 34-39

19. Niedermeyer, W. (1985) Interaction of streptolysin-O with biomembranes: kinetic and morphological studies on erythrocyte membranes. Toxicon 23, 425-439

20. Sekiya, K., Danbara, H., and Futaesaku, Y. (1993) [Mechanism of pore formation on erythrocyte membrane by streptolysin-O]. Kansenshogaku zasshi. The Journal of the Japanese Association for Infectious Diseases 67, 736-740

21. Stewart, S. E., D'Angelo, M. E., Piantavigna, S., Tabor, R. F., Martin, L. L., and Bird, P. I. (2014) Assembly of streptolysin O pores assessed by quartz crystal microbalance and atomic force microscopy provides evidence for the formation of anchored but incomplete oligomers. Biochim. Biophys. Acta. 1848, 115-126

22. Leung, C., Dudkina, N. V., Lukoyanova, N., Hodel, A. W., Farabella, I., Pandurangan, A. P., Jahan, N., Pires Damaso, M., Osmanovic, D., Reboul, C. F., Dunstone, M. A., Andrew, P. W., Lonnen, R., Topf, M., Saibil, H. R., and Hoogenboom, B. W. (2014) Stepwise visualization of membrane pore formation by suilysin, a bacterial cholesterol-dependent cytolysin. eLife 3, e04247

23. Young, J. D.-E., Hengartner, H., Podack, E. R., and Cohn, Z. A. (1986) Purification and characterization of a cytolytic pore-forming protein from granules of cloned lymphocytes with natural killer activity. Cell 44, 849-859

24. Podack, E. R., and Dennert, G. (1983) Assembly of two types of tubules with putative cytolytic function by cloned natural killer cells. Nature 302, 442-445

25. Masson, D., and Tschopp, J. (1985) Isolation of a lytic, pore-forming protein (perforin) from cytolytic T-lymphocytes. J. Biol. Chem. 260, 9069-9072

26. Dennert, G., and Podack, E. R. (1983) Cytolysis by H-2-specific T killer cells. Assembly of tubular complexes on target membranes. J. Exp. Med. 157, 1483-1495

27. Praper, T., Sonnen, A., Viero, G., Kladnik, A., Froelich, C. J., Anderluh, G., Dalla Serra, M., and Gilbert, R. J. (2011) Human perforin employs different avenues to damage membranes. J. Biol. Chem. 286, 2946-2955

28. Sutton, V. R., Waterhouse, N. J., Baran, K., Browne, K., Voskoboinik, I., and Trapani, J. A. (2008) Measuring cell death mediated by cytotoxic lymphocytes or their granule effector molecules. Methods (San Diego, Calif.) 44, 241-249

29. Baran, K., Dunstone, M., Chia, J., Ciccone, A., Browne, K. A., Clarke, C. J. P., Lukoyanova, N., Saibil, H., Whisstock, J. C., Voskoboinik, I., and Trapani, J. A. (2009) The Molecular Basis for Perforin Oligomerization and Transmembrane Pore Assembly. Immunity 30, 684-695

30. Piantavigna, S., McCubbin, G. A., Boehnke, S., Graham, B., Spiccia, L., and Martin, L. L. (2011) A mechanistic investigation of cell-penetrating Tat peptides with supported lipid membranes. Biochim. Biophys. Acta. 1808, 1811-1817

31. Mechler, A., Praporski, S., Piantavigna, S., Heaton, S. M., Hall, K. N., Aguilar, M. I., and Martin, L. L. (2009) Structure and homogeneity of pseudo-physiological phospholipid bilayers and their deposition characteristics on carboxylic acid terminated self-assembled monolayers. Biomaterials 30, 682-689

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 12: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

12

32. Sauerbrey, G. (1959) Verwendung von Schwingquarzen zur Wägung dünner Schichten und zur Mikrowägung. Z. Physik 155, 206-222

33. Tabor, R. F., Eastoe, J., and Dowding, P. J. (2010) A two-step model for surfactant adsorption at solid surfaces. J. Colloid Interface Sci. 346, 424-428

34. Torn, L. H., Koopal, L. K., de Keizer, A., and Lyklema, J. (2005) Adsorption of nonionic surfactants on cellulose surfaces: adsorbed amounts and kinetics. Langmuir 21, 7768-7775

35. McCubbin, G. A., Praporski, S., Piantavigna, S., Knappe, D., Hoffmann, R., Bowie, J. H., Separovic, F., and Martin, L. L. (2011) QCM-D fingerprinting of membrane-active peptides. Eur. Biophys. J. 40, 437-446

36. Praporski, S., Ng, S. M., Nguyen, A. D., Corbin, C. J., Mechler, A., Zheng, J., Conley, A. J., and Martin, L. L. (2009) Organization of cytochrome P450 enzymes involved in sex steroid synthesis: PROTEIN-PROTEIN INTERACTIONS IN LIPID MEMBRANES. J. Biol. Chem. 284, 33224-33232

37. Flanagan, J. J., Tweten, R. K., Johnson, A. E., and Heuck, A. P. (2009) Cholesterol exposure at the membrane surface is necessary and sufficient to trigger perfringolysin O binding. Biochemistry 48, 3977-3987

38. Tomita, T., Noguchi, K., Mimuro, H., Ukaji, F., Ito, K., Sugawara-Tomita, N., and Hashimoto, Y. (2004) Pleurotolysin, a novel sphingomyelin-specific two-component cytolysin from the edible mushroom Pleurotus ostreatus, assembles into a transmembrane pore complex. J. Biol. Chem. 279, 26975-26982

39. Krasilnikov, O. V., Merzlyak, P. G., Lima, V. L. M., Zitzer, A. O., Valeva, A., and Yuldasheva, L. N. (2007) Pore formation by Vibrio cholerae cytolysin requires cholesterol in both monolayers of the target membrane. Biochimie 89, 271-277

40. Metkar, S. S., Wang, B., Catalan, E., Anderluh, G., Gilbert, R. J., Pardo, J., and Froelich, C. J. (2011) Perforin rapidly induces plasma membrane phospholipid flip-flop. PloS one 6, e24286

41. Kilsdonk, E. P. C., Yancey, P. G., Stoudt, G. W., Bangerter, F. W., Johnson, W. J., Phillips, M. C., and Rothblat, G. H. (1995) Cellular Cholesterol Efflux Mediated by Cyclodextrins. J. Biol. Chem. 270, 17250-17256

42. Foster, L. J., De Hoog, C. L., and Mann, M. (2003) Unbiased quantitative proteomics of lipid rafts reveals high specificity for signaling factors. Proc Natl Acad Sci U S A 100, 5813-5818

43. Blumenthal, R., Millard, P. J., Henkart, M. P., Reynolds, C. W., and Henkart, P. A. (1984) Liposomes as targets for granule cytolysin from cytotoxic large granular lymphocyte tumors. Proc Natl Acad Sci U S A 81, 5551-5555

44. Criado, M., Lindstrom, J. M., Anderson, C. G., and Dennert, G. (1985) Cytotoxic granules from killer cells: specificity of granules and insertion of channels of defined size into target membranes. J. Immunol. 135, 4245-4251

45. Metkar, S. S., Marchioretto, M., Antonini, V., Lunelli, L., Wang, B., Gilbert, R. J., Anderluh, G., Roth, R., Pooga, M., Pardo, J., Heuser, J. E., Serra, M. D., and Froelich, C. J. (2015) Perforin oligomers form arcs in cellular membranes: a locus for intracellular delivery of granzymes. Cell Death Differ. 22, 74-85

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 13: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

13

FIGURE LEGENDS

FIGURE 1. Perforin binding and activity monitored by QCM-D. (A) QCM-D trace for 25 nM (yellow),

50 nM (green), 100 nM (blue) and 200 nM (red) wt Pfn. Δf (right axis) plotted as solid lines and ΔD

(left axis) plotted as dotted lines. Time points where the flow rate was stopped and/or the sensors were

washed are indicated. Data shown are representative of three or more experiments for each

concentration. (B) Δf-ΔD trace for 25 nM, 50 nM, 100 nM and 200 nM wt Pfn, coloured as in (A).

Changes in direction of the trace are highlighted by small arrows and inflection points are indicated by

asterisks. (C) The first derivative of the frequency and dissipation functions for data shown in panel

(A). df (Hz)/dt (min) (Δf’) is plotted on the left axis in darker colours and dD (10-6)/dt (min) (ΔD’) is

plotted on the right axis in lighter colours. The major minima for Δf’ are indicated by the black arrows

below the x-axis and maxima in ΔD’ are indicated by the grey arrows above the trace. The open grey

arrow highlights a clear minimum in the ΔD’ trace. (D) QCM-D trace for 100 nM wt Pfn showing the

3rd, 5th, 7th and 9th harmonics (lightest to darkest respectively). Δf (right axis) is plotted as solid lines

and ΔD (left axis) is plotted as dotted lines. As per (A), time points where the flow was stopped and

restarted for the wash are indicated, and the data shown are representative of three or more experiments.

(E) Δf-ΔD trace for 100 nM wt Pfn showing the spread over the 3rd, 5th, 7th and 9th harmonics (lightest

to darkest respectively). (F) The wt Pfn binding rate does not overlay the Langmuir isotherm adsorption

model for a simple first order process. 200 nM wt Pfn, data from panel (A), is plotted as theta (θ) against

the theoretical Langmuir isotherm adsorption model over time. Experimental data (wt Pfn) is plotted as

a solid line and the model data is plotted as a broken line.

FIGURE 2. Cholesterol is important for perforin activity. (A) Perforin mediated lysis of mammalian

cells is decreased in the absence of cholesterol. Cells pre-treated with MβCD (black), nystatin (green)

or untreated (red) were incubated with increasing concentrations of wt Pfn. Specific wt Pfn induced

lysis is plotted as % cell lysis. Representative curves of four or more experiments for each treatment

are shown. (B) QCM-D trace for 100 nM wt Pfn on DMPC alone (black) or with cholesterol (red). Δf

(right axis) is plotted as solid lines and ΔD (left axis) is plotted as dotted lines. The data shown are representative of three experiments. (C) Δf-ΔD trace for data shown in panel (B). (D) The first

derivative of the frequency and dissipation functions shown in panel (B) for wt Pfn in the presence (red)

or absence (black) of cholesterol in the membrane. df (Hz)/dt (min) plotted on the left, in darker colours,

and dD (10-6)/dt (min) is plotted on the right, in lighter colours.

FIGURE 3. Perforin binds membranes in the absence of calcium. (A) QCM-D trace for 100 nM wt Pfn

in the presence of HBS-Ca2+ (black) or HBS containing 2 mM EDTA (grey). Δf (right axis) is plotted

as solid lines and ΔD (left axis) is plotted as dotted lines and are presentative of three separate

experiments. (B) Δf-ΔD trace for wt Pfn data shown in (A). (C) Lysis assay using the flow through from

the flow cells in (A): sRBC were treated with 100 µl of each flow through with or without the addition

of excess calcium (5 mM). Graph displayed as % specific lysis ((Abs595 – Background/ Max. Abs595

– Background)x100). (D) QCM-D trace of 100 nM wt Pfn (black) and D429A (green). Δf (right axis)

plotted as solid lines and ΔD (left axis) plotted as dotted lines. Data are representative of three or more

experiments. (E) The first derivative of the frequency and dissipation functions shown in panel (A) for

wt Pfn (black) and D429A (green). df (Hz)/dt (min) (Δf’) is plotted on the left axis in darker colours,

and dD (10-6)/dt (min) (ΔD’) is plotted on the right axis in lighter colours. (F) QCM-D trace of 100 nM

wt Pfn (black) and D429A (green) in HBS containing 2 mM EDTA. At the time indicated, buffer was

exchanged for HBS-Ca2+ post binding. Δf (right axis) plotted as solid lines and ΔD (left axis) plotted as

dotted lines. (C) The first derivative of the frequency function shown in panel (F) for wt Pfn (black)

and D429A (green). The time points where the flow rate was stopped and 2 mM calcium was introduced

into the system are indicated. (H) The first derivative of the dissipation function shown in panel (F) for

wt Pfn (black) and D429A (green). Time points indicated as in (F) and (G).

FIGURE 4. Rate of oligomerization impacts the QCM-D profile of perforin. (A) QCM-D trace for 100

nM wt Pfn (black) and R213E (pink). Δf (right axis) plotted as solid lines and ΔD (left axis) plotted as

dotted lines. Data are representative of three experiments. (B) Δf-ΔD trace for wt Pfn (black) and R213E

(pink), data from panel (A). (C) The first derivative of the frequency and dissipation functions shown

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 14: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

14

in panel (A) for wt Pfn (black) and R213E (pink). df (Hz)/dt (min) plotted on the left axis in darker

colours and dD (10-6)/dt (min) is plotted on the right axis in lighter colours. (D) Neither wt Pfn (black)

or R213E (pink) fit the Langmuir Isotherm model, instead they largely overlay each other. (E) QCM-D

trace for 25 nM (yellow), 50 nM (green), 100 nM (blue) and 200 nM (red) R213E perforin. Δf (right

axis) plotted as solid lines and ΔD (left axis) plotted as dotted lines. Data are representative of three or

more experiments. (F) Δf-ΔD trace of data from panel (E). (G) The first derivative of the dissipation

function shown in panel (E) for 25 nM (yellow), 50 nM (green), 100 nM (blue) and 200 nM (red) R213E

perforin. df (Hz)/dt (min) plotted on the left axis in darker colours and dD (10-6)/dt (min) is plotted on

the right axis in lighter colours.

FIGURE 5. QCM-D comparison of SLO and Perforin. (A) Representative QCM-D traces of 100 nM

SLO (black) and Pfn (red) on Au-MPA containing DMPC:Chol bilayers. Only the binding (0- ~20 min)

and rest period are shown, no wash. Δf (right axis) plotted as solid lines and ΔD (left axis) plotted as

dotted lines. (B) Δf-ΔD trace for 100 nM SLO (black) and Pfn (red) data from (A). (C) The first

derivative of the frequency and dissipation functions shown in panel (A) for 100 nM SLO (black) and

Pfn (red), data from (A). df (Hz)/dt (min) plotted on the left axis (darker solid lines) and dD (10-6)/dt

(min) is plotted on the right axis (lighter thin line). (D) Langmuir isotherm adsorption model (grey

broken line) plotted against experimental data for SLO (black) and Pfn (red). (E) QCM-D traces of 100

nM SLO on Au-MPA containing DMPC:Chol bilayers. Only the binding (0- ~20 min) and rest period

shown, no wash. Δf (right axis) plotted as solid lines and ΔD (left axis) plotted as dotted lines, 3rd to

9th harmonics are coloured from lightest to darkest as indicated. (F) QCM-D traces of 100 nM Pfn on

Au-MPA containing DMPC:Chol bilayers. Graph plotted as in (E). (G) Perforin (made up of a C2

domain (green), EGF-like domain (black line), MACPF domain (yellow) containing two bundles of

helices (red)) is able to bind to the membrane in a calcium (small orange circles) independent

conformation, that is unknown, and through the calcium dependent C2 domain (green). Once bound in

the correct orientation perforin then provides a higher affinity binding site for other monomers to bind,

thus cooperation is seen when binding to the membrane. Oligomerization occurs and either perforin is

capable of insertion whilst the ring of monomers grows, or a rate limiting number of monomers must

be bound (similar to a pre-pore) before insertion. Finally, either a complete or incomplete pore is formed

to perforate the membrane.

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 15: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

15

FIGURES

FIGURE 1

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 16: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

16

FIGURE 2

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 17: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

17

FIGURE 3

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 18: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

18

FIGURE 4

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 19: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

Perforin Binding and Rearrangements Assessed by QCM-D

19

FIGURE 5

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 20: 1, Rico F. Tabor2 Michael E. D’Angelo1, Stefania 2 1, 3 ... · Perforin Binding and Rearrangements Assessed by QCM-D 1 Analysis of Perforin Assembly by Quartz Crystal Microbalance

BirdPiantavigna, James C. Whisstock, Joseph A. Trapani, Lisandra L. Martin and Phillip I.

Sarah E. Stewart, Catherina H. Bird, Rico F. Tabor, Michael E. D'Angelo, StefaniaCholesterol and Calcium Independent Membrane Binding

Analysis of Perforin Assembly by Quartz Crystal Microbalance Reveals a Role for

published online November 5, 2015J. Biol. Chem. 

  10.1074/jbc.M115.683078Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

by guest on September 26, 2020

http://ww

w.jbc.org/

Dow

nloaded from