Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

RESEARCH ARTICLE

MECHANOSENSITIVE CHANNELS

Activation of TRPV1 channels inhibits


mechanosensitive Piezo channel activity by
depleting membrane phosphoinositides
Istvan Borbiro, Doreen Badheka, Tibor Rohacs*

Capsaicin is an activator of the heat-sensitive TRPV1 (transient receptor potential vanilloid 1) ion channels
and has been used as a local analgesic. We found that activation of TRPV1 channels with capsaicin either
in dorsal root ganglion neurons or in a heterologous expression system inhibited the mechanosensitive
Piezo1 and Piezo2 channels by depleting phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] and its precur-
sor phosphatidylinositol 4-phosphate [PI(4)P] from the plasma membrane through Ca2+-induced phos-
pholipase Cd (PLCd) activation. Experiments with chemically inducible phosphoinositide phosphatases
and receptor-induced activation of PLCb indicated that inhibition of Piezo channels required depletion of
both PI(4)P and PI(4,5)P2. The mechanically activated current amplitudes decreased substantially in the
excised inside-out configuration, where the membrane patch containing Piezo1 channels is removed from
the cell. PI(4,5)P2 and PI(4)P applied to these excised patches inhibited this decrease. Thus, we concluded
that Piezo channel activity requires the presence of phosphoinositides, and the combined depletion of
PI(4,5)P2 and PI(4)P reduces channel activity. In addition to revealing a role for distinct membrane lipids
in mechanosensitive ion channel regulation, these data suggest that inhibition of Piezo2 channels may
contribute to the analgesic effect of capsaicin.

INTRODUCTION zymatically more active on PI(4,5)P2 than on PI(4)P (21). PI(4,5)P2 has a
Mechanosensation is an essential process involved in a variety of biolog- general role in regulating ion channels (22) and the integrity of the cyto-
ical phenomena. Mechanically activated (MA) ion channels that are ac- skeleton (23). PI(4,5)P2 may also be involved in mechanotransduction
tivated by membrane stretch are generally thought to mediate many mediated by cytoskeletal elements in hair cells (24), but its role in the regu-
aspects of mechanotransduction (1). Piezo1 and Piezo2 are nonselective lation of the putative mechanosensitive channel has not been examined.
Ca2+-permeable cation channels that are emerging as important mediators Transient receptor potential vanilloid 1 (TRPV1) channels are present
of various aspects of mechanotransduction (2–4). Unlike previous candi- in nociceptive DRG neurons and are activated by noxious heat and by cap-
dates, they consistently produce large, rapidly inactivating MA currents in saicin, a pungent compound in chili peppers. Capsaicin has long been used
recombinant systems (4–10). Both channels are abundant in organs with topically as a local analgesic, but how it inhibits mechanical pain is not
mechanical functions, such as the lungs, bladder, skin, and somatosensory known (25). Deletion of the single Piezo gene in Drosophila melanogaster
dorsal root ganglion (DRG) neurons (4). Gain-of-function mutations in reduced the sensitivity of the flies to noxious mechanical stimuli (26), and
Piezo1 (11, 12) cause hereditary xerocytosis (13), a disease associated with knockdown of the mouse Piezo2 gene inhibited mechanical allodynia
dehydrated red blood cells and mild to moderate hemolytic anemia. Gain- evoked by nerve ligation (27), suggesting the involvement of Piezo chan-
of-function mutations in Piezo2 cause distal arthrogryposis (14), a disease nels in mechanical pain. Piezo2 is present in some of TRPV1-positive
characterized by multiple distal contractures, ophthalmoplegia, and re- mouse DRG neurons (4), and capsaicin-sensitive DRG neurons in rat main-
strictive lung disease. Global genetic ablation of either Piezo1 (15) or Piezo2 ly display rapidly adapting MA currents (28), which are likely mediated by
(8) is embryonic lethal in mice. Knockdown of Piezo2 with short interfering Piezo2. Here, we performed electrophysiological experiments to test the
RNA eliminates rapidly adapting MA currents in DRG neurons (4), and effects of capsaicin-induced activation of TRPV1 channels on the activity
selective genetic knockout of Piezo2 in the skin produces sensory deficits of MA currents in DRG neurons and Piezo channels expressed heterolo-
of the mechanosensitive Merkel cells (16, 17). Combined deletion of Piezo2 gously in human embryonic kidney (HEK) 293 cells.
in adult DRG neurons and Merkel cells leads to profound loss of touch We found that activation of TRPV1 by capsaicin inhibited rapidly
sensation (18). adapting MA currents in DRG neurons. Heterologously expressed Piezo2
Phosphoinositides are important regulators of many different biolog- or Piezo1 channels were also inhibited upon activation of TRPV1 by cap-
ical processes (19). Phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] saicin, and this effect required extracellular Ca2+. Inclusion of PI(4,5)P2 or
and its precursor phosphatidylinositol 4-phosphate [PI(4)P] are the most PI(4)P in the whole-cell patch pipette alleviated the inhibitory effect of
abundant phosphoinositides in the plasma membrane, each constituting capsaicin, suggesting the involvement of the Ca2+-stimulated PLCd iso-
up to 1% of the lipids therein (20). Both of these lipids are substrates for forms. In contrast, we found that Gaq-dependent activation of PLCb by the
phospholipase C (PLC) enzymes, although most PLC isoforms are en- human muscarinic receptor 1 (hM1) only marginally inhibited Piezo1 cur-
rents. Fluorescence-based monitoring of PI(4,5)P2 and PI(4)P revealed
substantial differences in phosphoinositide changes induced by the two
Department of Pharmacology and Physiology, Rutgers New Jersey Medical
PLC-activating pathways, which may explain the differential inhibition
School, Newark, NJ 07103, USA. of Piezo channels. We found that depletion of PI(4,5)P2 and its precursor
*Corresponding author. E-mail: tibor.rohacs@rutgers.edu PI(4)P using a chemically inducible phosphatase inhibited Piezo1 currents.

www.SCIENCESIGNALING.org 10 February 2015 Vol 8 Issue 363 ra15 1


RESEARCH ARTICLE

In excised inside-out patch experiments, Piezo1 currents persisted in a so- A Small DRG neurons
B Medium and large DRG neurons
lution containing PI(4,5)P2 and PI(4)P, but decreased substantially without (under 25 pF) (above 25 pF)
these lipids. In conclusion, our data show that PI(4,5)P2 and PI(4)P are im- SA (3) RA Caps + (4) SA (1) RA (6)
portant cofactors for maintaining Piezo channel activity and indicate that
Ca2+ influx through TRPV1 inhibits Piezo channels by depleting these
phosphoinositides.
IA (5) IA (10)
RA Caps - (7)

RESULTS

A subset of DRG neurons exhibits MA currents that are


inhibited by capsaicin
A subset of TRPV1-positive DRG neurons (24%) also expresses Piezo2
(4). To study the effects of the activation of TRPV1 channels by capsaicin
on MA currents in DRG neurons, we used TRPV1 reporter mice obtained
by crossing the TRPV1-Cre and ROSA-STOP-EYFP mouse lines. In this
reporter line, neurons that had expressed Trpv1 during development or are 20 µm 20 µm
actively expressing Trpv1 are labeled with yellow fluorescent protein
(YFP) (29), enabling easy visual identification of TRPV1-positive neu- C RA Caps + DRG neuron RA Caps - DRG neuron
rons in DRG preparations. To enrich for cells displaying rapidly adapting 1 µM capsaicin 1 µM capsaicin

100 pA
MA currents consistent with the presence of Piezo channels, we visually

20 pA
selected neurons on the basis of morphological criteria described earlier
(8). We analyzed small YFP-positive neurons (membrane capacitance 1 min
1 min 5 µm 5 µm
under 25 pF) and found that 70.4% of them displayed MA currents with

10 pA

20 pA
different inactivation kinetics ranging from rapid through intermediate to
slow adaptation rates (Fig. 1A). Medium and large YFP-positive neurons 50 ms 50 ms
(membrane capacitance >25 pF) displayed mainly intermediate adapting
MA currents and did not respond to capsaicin (Fig. 1B). D Before
After 1 µM capsaicin

Normalized current (before = 1)


Among small YFP-positive DRG neurons displaying rapidly adapting n=3
1.2 n=3
MA currents, 36% also responded to 1 mM capsaicin; in these neurons, n = 7n = 5
capsaicin robustly inhibited MA currents (Fig. 1, C and D). In cells that –150 1.0
Current (pA)

did not display capsaicin-induced inward currents, MA currents were not n=5 0.8
affected by capsaicin, indicating that capsaicin did not directly inhibit –100
0.6
n=7 n=4
Piezo2 channels (Fig. 1, C and D). Neither the medium and large cells that n=4 0.4
–50 *

Before
responded to mechanical stimuli nor the small neurons with intermediate or *
0.2
slowly adapting MA currents displayed capsaicin-induced currents, despite
0 0.0
their clear YFP positivity (Fig. 1, A and B). These data indicated that many Caps + Caps – IA SA Caps Caps IA SA
+ –
of the small and medium mechanosensitive DRG neurons lost Trpv1 ex- RA
RA
pression during development.
Fig. 1. Effects of capsaicin on whole-cell MA currents in DRG neurons isolated
Capsaicin inhibits MA currents when Piezo and TRPV1 from TRPV1 reporter mice. (A) Pie chart showing the distribution of the rapidly
channels are heterologously coexpressed adapting (RA), intermediate adapting (IA), and slowly adapting (SA) MA cur-
Rapidly adapting MA currents in DRG neurons are mediated by Piezo2 rents in small YFP-positive neurons. The RA group is subdivided into cells that
channels (4); thus, we hypothesized that capsaicin-induced TRPV1 activa- respond to 1 mM capsaicin with large inward currents (Caps +) and cells that
tion inhibited Piezo2 channels. To support this hypothesis, we performed do not (Caps −). The numbers in parentheses are the number of neurons
experiments in cells from a human embryonic kidney cell line (HEK293) tested in each group. None of the IA and SA cells responded to capsaicin.
transiently cotransfected with TRPV1 and Piezo2. Application of 1 mM cap- (B) Pie chart showing the distribution of the different types of MA currents in
saicin inhibited MA currents in these cells in a manner dependent on extra- medium and large YFP-positive neurons. None of these neurons responded
cellular Ca2+ (Fig. 2, A and B). to capsaicin. Bottom panels in (A) and (B) show overlay of fluorescent and
Capsaicin also inhibited MA currents in HEK293 cells cotransfected phase-contrast images of a small and a large neuron, respectively. (C) Rep-
with TRPV1 and Piezo1, a homolog of Piezo2 (Fig. 3A). MA current ac- resentative traces recorded at −60 mV with mechanical stimulation every 30 s
tivity did not recover after washout of capsaicin even after several minutes, in a capsaicin-responsive neuron (left) and a nonresponsive neuron (right);
when mechanical stimuli of the same intensity were applied (Fig. 3A), and the application of 1 mM capsaicin is indicated by the horizontal bars. The
larger mechanical stimuli could only elicit very small MA currents after downward spikes correspond to the individual MA current evoked by the re-
capsaicin treatment (Fig. 3B). Capsaicin did not inhibit Piezo1 currents in petitive mechanical stimuli. Insets show individual MA currents on enlarged
cells not transfected with TRPV1 (Fig. 3C). Similar to the results with time scales before and after the application of capsaicin; the mechanical step
Piezo2-transfected cells (Fig. 2), the rapid inhibitory effect of capsaicin protocol, indicating the displacement of the mechanical probe, is also shown.
was essentially eliminated when assayed in the absence of extracellular (D) Statistical analysis for small neurons showing the raw data (left) and nor-
Ca2+ (Fig. 3D). These data are similar to those obtained with DRG neurons malized data (right) of peak MA currents measured before and after cap-
and indicated a crucial role for Ca2+ influx through TRPV1 in mediating saicin. *P < 0.05, analysis of variance (ANOVA).

www.SCIENCESIGNALING.org 10 February 2015 Vol 8 Issue 363 ra15 2


RESEARCH ARTICLE

A inhibition was less complete and had the tendency to partially recover (fig.
2 mM Ca 2+ in EC Ca 2+-free EC
S2A). Activation of the Ca2+-permeable noxious chemical sensor TRPA1
1 µM capsaicin 1 µM capsaicin
channels with mustard oil [allyl isothiocyanate (AITC)] also induced a
200 pA

small, but statistically significant, inhibition of Piezo1 currents (fig.


S2B). The inward currents induced by mustard oil were substantially
1 min smaller than those induced by low pH or capsaicin (fig. S2C), and gener-
ally, the extent of Piezo1 inhibition correlated with the current amplitudes
induced by these agents (fig. S2C). Neither pH 5.0 nor AITC inhibited
MA currents in cells transfected with Piezo1 but not with TRPV1 or TRPA1
(fig. S2D).
Although TRPV1 activation stimulates Ca2+-regulated PLCd enzymes
(30), we investigated whether stimuli that activate other PLC isoforms
inhibited Piezo currents. We activated PLCb using the hM1 agonist car-
B Before bachol in HEK293 cells cotransfected with Piezo1 and hM1, which is a
After 1 µM capsaicin ** Gaq-coupled GPCR [G protein (heterotrimeric guanine nucleotide–binding
Normalized current (before = 1)

–160 * 1.2 n=3 protein)–coupled receptor] (32). The cotransfected HEK293 cells exhibited
** decreased Piezo1 currents after a 5-min application of carbachol in exper-
–140 1.0
iments where the pipette solution (representing the intracellular solution)
–120
Current (pA)

0.8 contained 2 mM adenosine triphosphate (ATP) (Fig. 4A). However, MA


–100 current amplitudes also moderately decreased in the absence of carbachol
–80 0.6 (control) over time (Fig. 4A), and the difference between carbachol-treated
–60 and control cells was not statistically significant when normalized to the
0.4
current amplitude at the start of the experiment (Fig. 4B). Comparison of
Before
n=4

–40
0.2 n=4 the responses to increasing mechanical stimuli at the beginning and the end
n=3

–20
of the experiments in control and carbachol-treated cells showed that the
0 0.0 stimulus-response curves were identical (Fig. 4C). We interpreted these data
2 mM Ca 2+ Ca 2+-free 2 mM Ca 2+-
Ca 2+ free to indicate that activation of PLCb by hM1 marginally inhibited Piezo1 cur-
rents when the intracellular solution contained ATP, which is required for the
Fig. 2. Effects of TRPV1 activation on Piezo2-mediated MA currents. resynthesis of phosphoinositides (33).
HEK293 cells were transiently cotransfected with Piezo2 and TRPV1, and To inhibit the resynthesis of phosphoinositides, we also tested the ef-
MA currents were measured in whole-cell patch clamp experiments at fect of carbachol in the absence of intracellular ATP. Under these con-
−60 mV. (A) Cellular responses to repeated mechanical stimuli applied ditions, carbachol slowly but robustly inhibited Piezo1 activity (Fig. 4D),
every 30 s, recorded with application of 1 mM capsaicin in 2 mM Ca2+– producing a significant decrease in MA currents after 5 min when com-
containing (left) and Ca2+-free (right) extracellular (EC) solution. (B) Sta- pared with the MA currents in control cells after 5 min (Fig. 4E). When ATP
tistical analysis shows raw data (left) and normalized data (right) before was excluded from the intracellular solution, carbachol induced a robust
and after the application of capsaicin. *P < 0.05, **P < 0.01 (ANOVA). shift to the right (reduction in sensitivity) of the MA current–mechanical
stimulus relationship and also reduced the maximal response (Fig. 4F). In
inhibition of MA currents. For the remainder of the heterologous expres- contrast to carbachol, the inhibitory effect of capsaicin was essentially the
sion studies, we used Piezo1 because these channels are much easier to same in the presence or absence of intracellular ATP (Fig. 3F).
express than Piezo2, likely due to the cellular toxicity of heterologously Protein kinase C (PKC) is activated downstream of PLC isoforms;
expressed Piezo2. therefore, we tested the effect of activating PKC with the phorbol ester
phorbol 12-myristate 13-acetate (PMA). In HEK293 cells transfected with
Stimuli that deplete membrane phosphoinositides inhibit Piezo1, PMA induced a slow but significant reduction in Piezo1 currents,
Piezo1 currents which was attenuated by the PKC inhibitor bisindolylmaleimide (fig. S3,
Ca2+ influx in response to TRPV1 activation stimulates PLCd isoforms, A to C).
leading to the depletion of PI(4,5)P2 and PI(4)P both in DRG neurons To provide additional evidence that the inhibition of MA currents was
(30) and in heterologous expression systems (31). To test whether phos- mediated by depletion of phosphoinositides rather than by activation of
phoinositide depletion plays a role in the inhibitory effect of capsaicin, we PKC resulting from activation of PLC, we also tested the effect of depleting
supplemented the whole-cell patch pipette solution with PI(4,5)P2 or PI(4)P phosphoinositides without activating PLC. We cotransfected HEK293
and tested the effect of capsaicin on Piezo1 currents. This intracellular ap- cells with Piezo1 and the chemically inducible 4′,5′-phosphatase system
plication of PI(4,5)P2 or PI(4)P attenuated the inhibition of MA currents by called pseudojanin (34). Activation of pseudojanin by rapamycin depletes
capsaicin (Fig. 3, E and F). Capsaicin-induced inward TRPV1 current PI(4,5)P2 and PI(4)P without releasing second messengers, such as diacyl-
amplitudes in cells dialyzed with PI(4,5)P2 or PI(4)P were not significantly glycerol, that could activate PKC; see Materials and Methods for further
different from those in control cells (fig. S1); thus, smaller Ca2+ influx in the details. Rapamycin inhibited MA currents in HEK293 cells coexpres-
presence of excess lipids cannot account for the differences in Piezo channel sing Piezo1 and pseudojanin in whole-cell patch clamp experiments
inhibition. Capsaicin-induced currents in the absence of Ca2+ were substan- (Fig. 5A, left). The rapamycin-induced inhibition developed over several
tially larger (fig. S1). minutes. As a control, we confirmed that MA currents were unaffected
Activation of TRPV1 with low pH (pH 5.0) also inhibited Piezo1 cur- by rapamycin-induced translocation of an inactive phosphatase (Fig. 5A,
rents in HEK293 cells cotransfected with TRPV1 and Piezo1 (fig. S2A). right). Furthermore, MA current amplitudes were significantly different
Compared to the inhibition induced by capsaicin, this low pH–mediated in rapamycin-treated cells expressing the inactive or active pseudojanin

www.SCIENCESIGNALING.org 10 February 2015 Vol 8 Issue 363 ra15 3


RESEARCH ARTICLE

Fig. 3. Effects of TRPV1 activation on Piezo1-mediated MA currents. HEK293 A B


2 mM Ca2+ in EC Before capsaicin (n = 6)
cells were transiently cotransfected with Piezo1 and TRPV1, and MA currents
1 µM capsaicin After capsaicin (n = 6)
were measured in whole-cell patch clamp experiments at −60 mV. (A) Re- –800

200 pA
sponses to repeated mechanical stimuli applied every 30 s, recorded with –700
the application of 1 mM capsaicin as indicated. Insets throughout the figure show –600

Current (pA)
individual MA currents in response to a mechanical step of the same experiment –500
before and after the application of capsaicin. Scale indicator lines identical to –400
those in this panel were used throughout the figure. (B) Stimulus-response 5 µm –300

200 pA
curves before and after the application of capsaicin. (C) Representative trace 1 min –200
in a cell not transfected with TRPV1. (D) Representative trace of a Piezo1 and –100
TRPV1–cotransfected cell assayed in the absence of extracellular Ca2+. (E) 50 ms 0
Representative trace of a Piezo1 and TRPV1–cotransfected cell assayed in 2 3 4 5 6
normal extracellular (2 mM) Ca2+ with 40 mM PI(4,5)P2 (left) or 40 mM PI(4)P Stimulus length (µm)
C D
(right) in the intracellular (IC) solution. (F) Left: Statistical summary of current HEK without TRPV1 Ca2+-free EC
amplitudes before and after the application of capsaicin including those mea- 1 µM capsaicin 1 µM capsaicin
sured in the presence of the indicated concentrations of MgATP in the patch
pipette. Right: Time course of the effects of PI(4,5)P2 (PIP2), PI(4)P (PIP), and
Ca2+-free EC solution. Data in all measurements were normalized to the cur-
rent amplitudes before the application of capsaicin; individual MA current am-
plitudes every 30 s are shown (mean ± SEM). Capsaicin (1 mM) was applied for
120 s in most measurements (range, 60 to 150 s). To obtain comparable time
courses, the time scale of each measurement was synchronized again to the
first point after the washout of capsaicin. *P < 0.05, ***P < 0.001 (ANOVA).

E
(Fig. 5B). In contrast, MA currents in cells transfected with a PI(4,5)P2 5- PI(4,5)P2 in IC PI(4)P in IC
phosphatase that converts PI(4,5)P2 into PI(4)P were reduced in response 1 µM capsaicin 1 µM capsaicin
to rapamycin, but the inhibition was not significant when compared with
the MA currents in the cells before rapamycin induction (Fig. 5B).

Stimuli that activate PLCb or PLCd produce different


changes in phosphoinositides
We hypothesized that the differences in Piezo1 current inhibition were due
to distinct PI(4,5)P2 and PI(4)P changes induced by the activation of differ-
ent PLC isoforms upon stimulation of hM1 or TRPV1. To measure dynamic
changes in phosphoinositides in the plasma membrane, we cotransfected
HEK293 cells with hM1 or TRPV1 and with different fluorescence reso- F 2 mM Ca2+ in EC
nance energy transfer (FRET)–based sensors of PI(4,5)P2 or PI(4)P and Before 1 µM capsaicin Ca2+-free EC
–1200 After PIP2 in IC
measured their emission ratios after hM1 or TRPV1 activation. In all cases, * PIP in IC
we used pairs of cyan fluorescent protein (CFP)– and YFP-tagged phos- –1000 capsaicin
Current (pA)

phoinositide binding domains. A decrease in FRET ratio, due to trans- –800


(Before capsaicin = 1)

*** *** 1.6


Normalized current

location of the sensors from the plasma membrane to the cytoplasm, –600 *** *
represents a decrease in the concentration of their respective phosphoinosi- 1.2
–400
tide target(s) in the plasma membrane (35).
The PLCd1 pleckstrin homology (PH) domain sensor monitors PI(4,5)P2 –200 0.8
(35), but the PLCd1 PH domain also binds inositol 1,4,5-trisphosphate (IP3) 0
in vitro (36). The R322H Tubby sensor monitors PI(4,5)P2 (37) and does not TRPV1 – + + + + + + 0.4 1 min
Ca2+ (mM) 2 2 2 2 0 2 2
bind IP3. We used the R322H mutant (Tubby-mut) because it is more sen- ATP (mM) 2 2 0 4 2 2 2
0.0
sitive to changes in PI(4,5)P2 than the wild-type probe (38). The sensor Lipid – – – – – PIP2 PIP MA currents MA currents
containing two copies of the PH domain of the protein OSH2 binds to PI(4)P N 7 11 9 5 6 8 6 before after
and PI(4,5)P2 and responds to combined depletion of these two lipids with
a translocation from the plasma membrane to the cytoplasm (34). to capsaicin (fig. S4B), and capsaicin had essentially no effect in the ab-
Measurements with the Tubby-mut FRET sensor indicated similar de- sence of extracellular Ca2+ (fig. S4C). PI(4,5)P2 depletion induced by car-
creases in PI(4,5)P2 in response to carbachol in hM1-expressing cells and bachol was significantly slower than that induced by capsaicin, when
to capsaicin in TRPV1-expressing cells (Fig. 6A). Experiments with CFP- detected both by the Tubby-mut sensor (Fig. 6A) or by the PLCd1 PH do-
and YFP-tagged PLCd1 PH domain sensors also resulted in a similar decrease main sensor (fig. S4A).
in FRET ratio upon stimulation of hM1-expressing cells with carbachol or With the CFP- and YFP-tagged OSH2 PH-domain sensors that re-
stimulation of TRPV1-expressing cells with capsaicin (fig. S4A). spond to dual depletion of PI(4)P and PI(4,5)P2, we assessed PI(4)P changes
In cells expressing the Tubby-mut PI(4,5)P2 sensor, the TRPV1 antag- upon TRPV1 and hM1 activation. We reasoned that because both carba-
onist capsazepine blocked the decreased FRET ratio produced in response chol and capsaicin induced robust and comparable decreases in PI(4,5)P2

www.SCIENCESIGNALING.org 10 February 2015 Vol 8 Issue 363 ra15 4


RESEARCH ARTICLE

A A
2 mM ATP in IC
100 µM CCh Flow control Active PI(4,5)P2 4,5-phosphatase Inactive PI(4,5)P2 phosphatase
200 pA

100 nM rapamycin 100 nM rapamycin

100 pA
5 µm
200 pA

1 min

5 µm

100 pA
50 ms
1 min
B C 50 ms
Before
2.5 min 100 µM CCh (n = 7)
Normalized current (before = 1)

5 min flow control (n = 5)


–1200 1.2 –1200
*
1.0
NS –1000 B Before 100 nM rapamycin
–1000

Normalezed current (before = 1)


Current (pA)

Current (pA)

–800
After 1.2 n=9
–800 0.8 ***
–600 0.6 –600 –700 1.0
–600 NS n=8
–400

Current (pA)
–400 0.4 0.8
0.2 –200 –500 ***
–200
–400 0.6 n=9
0 0.0 0
Ctrl CCh Ctrl CCh 2 3 4 5 6 –300
n=6 n=7 Stimulus (µm) 0.4
–200

Before
D

n=9

n=9

n=8
0.2
ATP free IC –100
100 µM CCh flow control 0 0.0
Inactive PJ 4,5- PJ 5- Inactive PJ 4,5- PJ 5-
ptase ptase ptase ptase ptase ptase

Fig. 5. Effects of rapamycin-induced plasma membrane translocation of


the PI(4,5)P2 4,5-phosphatase pseudojanin on Piezo1-mediated MA cur-
rents. HEK293 cells were cotransfected with Piezo1 and components of
the pseudojanin system; MA currents were measured in whole-cell patch
clamp experiments at −60 mV with mechanical stimuli applied every 30 s.
(A) Representative traces show responses to 100 nM rapamycin during re-
peated mechanical stimuli in cells expressing Piezo1 and the active PI(4,5)P2
E F 100 µM CCh (n = 7) 4,5-phosphatase pseudojanin (left) or the inactive PI(4,5)P2 phosphatase
Normalized current (before = 1)

flow control (n = 6) (right). Insets show individual MA current traces before and after the appli-
–1200 1.2 –1200
*** cation of rapamycin. (B) Plots of raw data (left) and normalized data (right)
–1000 *** –1000
1.0 show statistical analysis of peaks of MA currents in cells transfected with
Current (pA)

**
Current (pA)

–800 0.8 –800 inactive or active PI(4,5)P2 4,5-phosphatase (inactive ptase; PJ 4,5-ptase)
–600 0.6 –600 or active PI(4,5)P2 5-phosphatase (PJ 5-ptase) before and after 5 min of
–400 0.4 –400 rapamycin application. ***P < 0.001; NS, not significant (P = 0.128); ANOVA
and t test.
–200 0.2 –200
0 0.0 0
Ctrl CCh Ctrl CCh 2 3 4 5 6 on the basis of the Tubby-mut and PLCd1 PH domain sensors (Fig. 6A and
n=7 n=8 Stimulus (µm)
fig. S4A), differences in the signals obtained with the OSH2-PH probe like-
Fig. 4. Effects of hM1 activation on Piezo1-mediated MA currents. HEK293 ly reflected differential effects on PI(4)P in response to these two agonists.
cells were transiently cotransfected with Piezo1 and hM1, and MA currents Therefore, we refer to this sensor as a PI(4)P sensor, although it responds to
were measured in whole-cell patch clamp experiments at −60 mV with me- parallel changes in PI(4)P and PI(4,5)P2.
chanical stimuli applied every 30 s. (A) Representative traces for experi- In cells expressing the OSH2 PH domain sensor, carbachol induced a
ments with 2 mM ATP–containing IC solution in the patch pipette with a significantly smaller decrease in the FRET ratio than did capsaicin, indicating
5-min application of 100 mM carbachol (CCh) (left) and control solution (right). less PI(4)P depletion in the membrane upon hM1 activation (Fig. 6B). Con-
Insets show individual MA currents recorded before and after a 5-min car- sistent with the requirement for TRPV1 activity, the capsaicin-induced de-
bachol application or at identical times for the control cell. Scale indicator crease in the FRET ratio of the OSH2 PH domain sensor was eliminated in
lines identical to those in this panel were used throughout the figure. (B) Sta- the presence of capsazepine (fig. S4D). When we cotransfected HEK293
tistical analysis of carbachol-treated and control groups using raw data (left) cells with the OSH2 PH domain sensor and both hM1 and TRPV1, the
and normalized data (right) before and after 2.5 and 5 min of carbachol ap- sequential application of carbachol and capsaicin revealed that TRPV1
plication. (C) Stimulus-response curves recorded in carbachol-treated and flow activation with capsaicin further depleted PI(4)P (reduced the FRET ratio)
control groups with increasing steps of mechanical stimuli. (D to F) Identical after hM1 activation with carbachol (Fig. 6C). In contrast, carbachol did
experiments and data analysis with ATP-free IC solution in the patch pipette. not further reduce the FRET ratio of the OSH2 PH domain sensor when
*P < 0.05; **P < 0.01; ***P < 0.001; NS, not significant (P = 0.101); ANOVA. TRPV1 activation with capsaicin preceded hM1 activation (Fig. 6C). In

www.SCIENCESIGNALING.org 10 February 2015 Vol 8 Issue 363 ra15 5


RESEARCH ARTICLE

A Baseline B tion depleted PI(4,5)P2 to comparable ex-


Tubby-mut, PI(4,5)P2 sensor CCh OSH2-PH, PI(4)P sensor tents (Fig. 6D). A slight decrease in FRET
Caps *** ratio occurred when capsaicin was applied
CCh *** CCh
*** after carbachol, indicating that despite the
***
Normalized FRET ratio

Normalized FRET ratio


1.0 1.0 16 * 1.0 1.0 comparable FRET decrease evoked by the

n = 10
*** two agonists, capsaicin may induce a some-

n=6
0.9 0.9 0.9 0.9 what more complete PI(4,5)P2 depletion
12

T50 (s)
than carbachol. This decrease was only
0.8 0.8 8 0.8 0.8 significant statistically when the data were
Caps normalized to the point before the appli-

n = 13
0.7 0.7 0.7 0.7 cation of capsaicin (Fig. 6D, right panel).

n=6
n=9
4

n=9
Caps These data indicated that the hM1 receptor,
0.6 0.6 0 0.6 0.6 which signals through G proteins to PLCb,
0 30 60 90 120 Tubby-mut Tubby-mut 0 30 60 90 120 OSH2-PH
Time (s) Time (s) produced a moderate and slow decrease
C of PI(4)P and a larger, but still slow, deple-
tion of PI(4,5)P2, whereas TRPV1, which
OSH2-PH, PI(4)P sensor signals through Ca2+ to PLCd, resulted in
CCh ** * rapid and maximal depletion of both PI
Caps ** *
Normalized FRET ratio

1.0 1.0 (4,5)P2 and PI(4)P.


***

end of 1st agonist


***
Normalized to the
end of baseline

Normalized to the
1.0
0.9 0.9 Phosphoinositides inhibit Piezo1
0.9
current rundown in cell-free
0.8 0.8 excised inside-out patches
To test the direct effects of phosphoinositides
0.7 Caps on MA currents, we performed patch clamp
0.7 0.8
CCh
n=6 n=4 recordings on excised inside-out membrane
0.6 0.6 patches from Piezo1-transfected HEK293
0 30 60 90 120 150 180 CCh 1st Caps 1st CCh 1st Caps 1st
Time (s) cells. We evoked MA currents by applying
negative pressure steps through the patch
D pipette using a high-speed pressure clamp
Tubby-mut, PI(4,5)P2 sensor
*** *** device (39). In the cell-attached configuration,
CCh
Caps *** *** the Piezo1-mediated MA currents showed
Normalized FRET ratio

1.0 1.0
* fast inactivation at negative voltages and no
end of 1st agonist
Normalized to the

Normalized to the
end of baseline

1.0 inactivation at positive voltages (Fig. 7A).


0.9 0.9
Upon excision, MA current activity decreased
0.9 (rundown) (Fig. 7B, left graph). When patches
0.8 0.8 were excised into a bath solution containing
0.7 Caps PI(4,5)P2 and PI(4)P, rundown was inhibited
CCh 0.7 0.8
(Fig. 7B, right graph). However, the inhibition
0.6 n = 9 n = 6 of rundown by these two phosphoinositides
0.6
0 30 60 90 120 150 180 CCh 1st Caps 1st CCh 1st Caps 1st was only statistically significant when the re-
Time (s) sponses at positive voltages were compared
Fig. 6. Fluorescence-based PI(4,5)P2 and PI(4)P measurements in HEK cells cotransfected with hM1 or between the patches excised into control
TRPV1 and phosphoinositide sensors. (A) FRET measurements in cells expressing the Tubby-mut PI(4,5)P2 bath solution and those excised into phos-
sensor. Left panel shows individual traces in response to 100 mM carbachol (CCh) in an hM1-transfected phoinositide-containing solution (Fig. 7C).
cell and in response to 1 mM capsaicin in a TRPV1-transfected cell. Middle panel shows statistical analysis The addition of poly-lysine, which chelates
of FRET ratios; right panel shows FRET decay half-times for cells exposed to carbachol or capsaicin. (B) negatively charged lipids, inhibited the MA
Individual FRET measurements in cells expressing the OSH2 PH domain PI(4)P sensor and either hM1 or currents (Fig. 7, B and C). Once the currents
TPRV1 (left) and statistical analysis of FRET ratios (right). (C) FRET measurements in cells cotransfected had run down, we could not reactivate the
with the OSH2 PH domain sensor and both TRPV1 and hM1. The sequential applications of 1 mM capsaicin MA current by adding phosphoinositides to
and 100 mM carbachol are indicated by the horizontal lines in the representative traces (left). Statistical the bath solution in most patches (fig. S5A),
analyses of OSH2 PH domain FRET ratios normalized to the baseline (middle) or to the end of first agonist even though a slow partial reactivation oc-
application (right) are shown. (D) FRET measurements in cells cotransfected with the Tubby-mut PI(4,5)P2 curred in some cases (fig. S5B).
sensor and both TRPV1 and hM1. Data are presented as in (C). *P < 0.05, **P < 0.01, ***P < 0.001 (ANOVA We also measured MA currents in Piezo1-
and t test). expressing Xenopus oocytes in excised inside-
out patches.The MA currents inPiezo1-injected
oocytes were more than 10 times larger than
cells cotransfected with the Tubby-mut sensor, hM1, and TRPV1, the de- the endogenous MA currents (fig. S6A). The endogenous MA currents did
crease in FRET ratio was independent of the order of addition of carbachol not show inactivation, whereas Piezo1-injected oocytes showed substantial
and capsaicin (Fig. 6D), indicating that TRPV1 activation and hM1 activa- inactivation of MA currents at negative voltages (fig. S6A). This inactivation

www.SCIENCESIGNALING.org 10 February 2015 Vol 8 Issue 363 ra15 6


RESEARCH ARTICLE

A B C *
Current Pressure Voltage *
+60
(mV) PolyK 150 PolyK 1.0
200 i/o i/o in PIPs
0

Normalized current
100
–60 n=6 n=6
0.5

Current (pA)
100
(mm Hg)

0 50
0
–20 0 0.0
200 ms 200 ms
–100 –50 ca
150 pA

–0.5 After i/o


–200 –100 End
PolyK
0 2 4 6 8 10 0 2 4 6 8 10
Time (min) Time (min) Control i/o in PIPs

Fig. 7. Effects of PI(4,5)P2 and PI(4)P on Piezo1-mediated MA currents in rents (blue triangle) at +60 mV, which were plotted in (B). (B) Representative
excised inside-out patches in HEK293 cells transfected with Piezo1. (A) Volt- MA peaks and end currents [as marked in the lower part of (A)] measured
age (upper) and pressure (middle) protocols and representative MA cur- when patches were excised (i/o) into control bath solution (left) or a bath so-
rents (lower). The holding potential was 0 mV, and a 3-s voltage step to lution containing 10 mM PI(4,5)P2 and 10 mM PI(4)P (PIPs) (right). At the periods
−60 mV was applied, during which a 200-ms negative pressure pulse was indicated by the horizontal lines, the patch was perfused with poly-lysine
used to elicit MA currents; 15 s later, a second 3-s voltage step to +60 mV (30 mg/ml) (PolyK). (C) Statistical summary of peak MA currents recorded at
was applied, during which the MA currents were induced with a −20 mmHg −60 mV (downward bars) and +60 mV (upward bars) in the cell-attached
pressure step. The protocol was repeated every 30 s. In the representative configuration (ca), after excision (i/o), 5 min after excision (end), and after
traces (A, lower), marks indicate the peak currents (black square) and the application of poly-lysine (30 mg/ml). All data were normalized to the cell-
currents at the end of the pressure step (red circle) at −60 mV and peak cur- attached current values at +60 mV. *P < 0.05, ANOVA.

was slower than that observed in mammalian cells (compare fig. S6A to the pipette solution resulted in a much larger inhibition of Piezo1 by car-
Fig. 7A). Upon excision, currents showed a variable extent of rundown in bachol. ATP serves as a substrate for lipid kinases; its absence interferes
experiments in which repetitive mechanical stimuli were applied (fig. S6B). with the resynthesis of phosphoinositides. Thus, without ATP, the decrease
In patches where rundown was not observed or was slow, channel activ- in PI(4,5)P2 and PI(4)P is expected to become more pronounced and ir-
ity was generally inhibited by poly-lysine (fig. S6C). In a few oocytes, we reversible. The PKC activator PMA also inhibited Piezo1, but the minimal
succeeded in reactivating Piezo1 currents by adding PI(4,5)P2 after either effect of carbachol in the presence of intracellular ATP suggested that a sub-
poly-lysine application (fig. S6C) or spontaneous rundown (fig. S6, D stantial contribution by PKC to Piezo1 inhibition was unlikely. Further-
and E), but in many other patches, these efforts were unsuccessful (fig. S6F). more, the absence of ATP potentiated the effect of carbachol, which is
Similar manipulations in noninjected oocytes induced negligible MA cur- the opposite of what is expected if PKC was a major contributor, because
rents (fig. S6G). protein kinases also require ATP.
To test whether the different kinetics and extent of inhibition by cap-
saicin and carbachol were due to different effects on phosphoinositides,
DISCUSSION
we monitored PI(4,5)P2 and PI(4)P using various FRET-based phospho-
We found that capsaicin quickly and almost completely inhibited rapidly inositide sensors. We concluded that TRPV1 activation causes a fast and
adapting Piezo2-mediated MA currents in TRPV1-positive DRG neurons. robust depletion of both PI(4,5)P2 and PI(4)P. Muscarinic receptor acti-
Heterologously expressed Piezo2 channels were also robustly inhibited upon vation primarily induced a comparable yet somewhat smaller and slower
TRPV1 activation, showing that components required for inhibition were decrease of PI(4,5)P2 and a substantially smaller decrease in PI(4)P. These
not specific to DRG neurons. Stimulating TRPV1 with capsaicin or low findings are consistent with the slow and incomplete inhibition of Piezo1
pH also inhibited heterologously expressed Piezo1 channels. Capsaicin- currents by carbachol and the fast and complete inhibition by capsaicin.
induced inhibition of both channels required the presence of extracellular Overall, our experiments with inducible phosphatases and PLC activa-
Ca2+. This inhibitory effect persisted well after the washout of capsaicin; tion indicated that Piezo1 channel activity was only inhibited upon sub-
thus, a Ca2+-dependent signaling cascade is more likely to explain the pro- stantial reduction in both PI(4,5)P2 and PI(4)P. Piezo2 channels are likely
longed effect of capsaicin than direct inhibition by Ca2+. Because Piezo2 to behave similarly because Piezo2 is not inhibited but rather potentiated
channels appeared toxic to HEK293 cells, we performed further mecha- by bradykinin (8), which only induces a moderate decrease in PI(4,5)P2
nistic studies with Piezo1 channels, which are homologs of Piezo2 and have and does not change PI(4)P in DRG neurons (30). Piezo channels are sim-
similar channel properties (5, 9, 10, 12). ilar in this respect to TRPV1, which is also only inhibited when both of
Previously, we reported that application of capsaicin to TRPV1-positive these phosphoinositides are greatly depleted (30, 34).
DRG neurons activated Ca2+-sensitive PLCd enzymes, which lead to de- Activation of TRPV1 by low pH or activation of TRPA1, another Ca2+-
pletion of the two phosphoinositides [PI(4,5)P2 and PI(4)P] (30). Here, we permeable channel, by AITC also inhibited Piezo1 channels, suggesting
found that inclusion of PI(4,5)P2 or PI(4)P in the whole-cell patch pipette that Ca2+ influx through other pathways may also modulate Piezo channel
alleviated capsaicin-induced inhibition of Piezo currents, indicating a activity. Generally, the inhibitory effect correlated with TRPV1 or TRPA1
potential role for phosphoinositide depletion in this phenomenon. current amplitudes, but even when low pH and capsaicin produced similar
Distinct pathways can lead to activation of different PLC isoforms; for current amplitudes, capsaicin induced a more complete and less reversible
example, Gaq-coupled receptors activate PLCb enzymes, whereas Ca2+ inhibition than low pH. A likely explanation for this is that larger propor-
influx induces PLCd activation (40). In our study, application of carbachol tions of currents are carried by Ca2+ when TRPV1 is activated by capsa-
to cells expressing hM1 and Piezo1 only induced a marginal inhibition if icin, than by low pH (41). It is possible that Ca2+-dependent factors other
the intracellular pipette solution contained ATP. Exclusion of ATP from than phosphoinositide depletion also contribute to inhibition of Piezo1

www.SCIENCESIGNALING.org 10 February 2015 Vol 8 Issue 363 ra15 7


RESEARCH ARTICLE

currents upon TRPV1 activation because PI(4,5)P2 and PI(4)P only par- these channels indirectly, or their effects may depend on cellular factors
tially and transiently alleviated the capsaicin-induced inhibition. that are easily lost in excised patches.
To test whether PI(4,5)P2 and PI(4)P directly affected Piezo1 channel
activity, we performed excised inside-out patch clamp experiments, per-
fusing the internal side of the plasma membrane patch with control solu- MATERIALS AND METHODS
tions or phosphoinositide-containing solutions. Piezo1 currents measured
in HEK293 cells rapidly decreased after excision (rundown), which is a Cell culture
characteristic of phosphoinositide-dependent ion channels and reflects the HEK293 cells were obtained from the American Type Culture Collection
loss of PI(4,5)P2 and PI(4)P from the patch membrane by lipid phospha- and were cultured in minimal essential medium (MEM) (Life Technologies)
tases (22). When we excised patches into a solution containing both containing 10% (v/v) Hyclone characterized fetal bovine serum (FBS)
PI(4,5)P2 and PI(4)P, rundown of Piezo1 activity was reduced. Our efforts (Thermo Scientific), and penicillin (100 IU/ml) and streptomycin (100 mg/ml;
to reactivate Piezo1 currents after rundown were generally unsuccessful in Life Technologies). Cells were transiently transfected using the Effectene
HEK293 cells. We also measured Piezo1 currents in Xenopus oocytes in transfection reagent (Qiagen). Then, 24 hours after transfection, cells were
cell-attached and excised inside-out configurations. Although the results trypsinized and replated on poly-L-lysine–coated round coverslips; 24 to
were quite variable, we managed to reactivate Piezo1 currents in a limited 48 hours after seeding, fluorescent cells were selected for electrophysio-
number of patches. Overall, our excised patch measurements are compa- logical recordings. Cultured HEK293 cells were kept in a humidity-
tible with the phosphoinositide dependence of these channels that we ob- controlled incubator with 5% CO2 at 37°C.
served in whole-cell experiments. However, our findings also suggest that DRG neurons were isolated from 6- to 12-week-old heterozygous
some critical cellular component is rapidly lost after excision in most TRPV1-reporter mice [offspring of TRPV1-Cre+/+ (29) and R26-STOP-
experiments. We did not address here what this lost factor could be, but EYFP+/+ (54)] as described previously (55) with some modifications. All
it is well known that an excised patch, even though it is removed from the animal procedures were approved by the Institutional Animal Care and Use
cellular environment, is still a relatively complex system containing various Committee. DRG neurons were isolated from mice of either sex; ganglia
cellular components, including large portions of the cortical cytoskeleton were harvested from all spinal segments after laminectomy and removal of
(42). Our data are somewhat similar to those observed for TRPA1 chan- the spinal column and maintained in ice-cold Hanks’ balanced salt solu-
nels, where activation by PI(4,5)P2 was only observed if the lipid was ap- tion (HBSS) for the duration of the isolation. Isolated ganglia were cleaned
plied within a short period after excision (43). This contrasts with most from excess nerve tissue and incubated in type I collagenase (2 mg/ml;
PI(4,5)P2-sensitive ion channels, which are reproducibly reactivated in ex- Worthington) and dispase (5 mg/ml; Sigma) in HBSS at 37°C for 27 min,
cised patches even several minutes after complete rundown (31, 44–46), followed by mechanical trituration. Digestive enzymes were then removed
reflecting activation by direct interactions between the lipid and the channel after centrifugation of the cells at 100g for 5 min. Cells were then resuspended
(47–49). and seeded onto glass coverslips coated with a mixture of poly-D-lysine (Life
Capsaicin has been used as a topical analgesic for a long time. Its Technologies) and laminin (Sigma). Neurons were maintained in culture
effects are complex. Initially, capsaicin evokes painful burning sensation in Dulbecco’s MEM/F12 (1:1) supplemented with 10% FBS (Thermo
due to activation of TRPV1, followed by hypersensitivity mainly caused Scientific) NT-4 (50 ng/ml; Sigma), nerve growth factor (100 ng/ml; Sigma),
by neurogenic inflammation, after which it renders neurons refractory not and penicillin (100 IU/ml) and streptomycin (100 mg/ml; Life Technologies)
only to heat and capsaicin but also to other stimuli (25). Capsaicin inhibits for 16 to 48 hours before measurements.
mechanical hyperalgesia after plantar incision (50) and after spinal nerve Oocytes were extracted from mature female Xenopus laevis frogs
transection in rats (51). Our finding that capsaicin robustly and almost (Xenopus Express) and digested with collagenase (0.2 mg/ml; Sigma) in
irreversibly inhibits Piezo2 channels may underlie or contribute to this phe- OR2 solution (82.5 mM NaCl, 2 mM KCl, 1 mM MgCl2, and 5 mM Hepes,
nomenon. We note, however, that the analgesic effects of capsaicin are long- pH 7.4) overnight at 16° to 18°C. Defolliculated oocytes were selected
lasting and may also involve neuronal degeneration (25). and maintained in OR2 solution plus 1.8 mM CaCl2 and 1% penicillin/
Earlier work showed that capsaicin-responsive DRG neurons mainly streptomycin at 16° to 18°C. Complementary RNA (cRNA; 20 ng) was
display rapidly adapting MA currents (28), which were later shown to be microinjected into each oocyte using a nanoliter injector system (World
mediated by Piezo2 channels (4). We also found that capsaicin-sensitive Precision Instruments). Experiments were performed 72 hours after
DRG neurons that responded to mechanical stimuli exclusively displayed injection.
rapidly adapting MA currents. We had smaller proportion of capsaicin-
responsive cells displaying MA currents, however, than those reported in Fluorescence measurements
rat DRG neurons (28). Genetic deletion of TRPV1-positive neurons in mice For monitoring phosphoinositides, we cotransfected HEK293 cells with
had no effect on mechanosensitivity (52), whereas acute axonal silencing phosphoinositide-binding domains of different proteins fused with CFP
of TRPV1-positive fibers in rats inhibited both acute and inflammatory and YFP. Under basal conditions, there are sufficient amounts of phos-
mechanical pain (53). This discrepancy may reflect species differences but phoinositides in the plasma membrane such that these two fluorophores
could also be due to the difference between acute versus chronic intervention. are localized at the plasma membrane and undergo FRET because of their
In conclusion, our data show that Piezo channels require phospho- proximity due to the two-dimensional distribution in the plasma mem-
inositides for activity. Activation of TRPV1 by capsaicin inhibits rapidly brane. Following a decrease in the concentration of their respective lipid
adapting MA currents in DRG neurons, as well as heterologously ex- targets, the sensors translocate to the cytosol, and FRET decreases (35).
pressed Piezo1 and Piezo2 channels through robust Ca2+-induced deple- We measured FRET ratios between CFP and YFP upon hM1 or TRPV1
tion of both of these lipids, an effect that may contribute to the local activation. The following phosphoinositide sensors were used: the CFP-
analgesic effect of capsaicin. The channels likely have very high apparent and YFP-tagged PLCd1 PH domain, which is sensitive to PI(4,5)P2 (35);
affinity for phosphoinositides because the more moderate decreases in the sensor containing two copies of the OSH2 PH domain each pair tagged
PI(4,5)P2 and PI(4)P during PLCb activation only marginally and very with either CFP or YFP was used to monitor changes in PI(4)P (56) and
slowly inhibited Piezo1 channel activity. Phosphoinositides may activate the PI(4,5)P2-sensitive Tubby-R322H mutant tagged with YFP (38) together

www.SCIENCESIGNALING.org 10 February 2015 Vol 8 Issue 363 ra15 8


RESEARCH ARTICLE

with the CFP-tagged Tubby-R322H, which we created by subcloning it into We classified the mechanosensitive DRG neurons into three groups
the pECFP-N1 vector. on the basis of the inactivation properties of their MA currents. Cells dis-
FRET measurements were performed as described earlier (31). Briefly, playing currents with rapidly and completely inactivating currents (t =
we used a photomultiplier-based dual-emission system mounted on an 22.9 ± 3.1 ms) during the 200-ms stimulus were referred to as rapidly
IX-71 inverted microscope (Olympus), equipped with a DeltaRAM ex- adapting mechanosensors; cells with slow inactivation and more than
citation light source from Photon Technology International (PTI). Exci- half of the peak remaining activity at the end of stimulus were called
tation wavelength was 430 nm; emission was detected parallel at 480 and slowly adapting. Cells showing slower inactivation than rapidly adapting
535 nm using two interference filters and a dichroic mirror to separate cells (t = 61.2 ± 6.6 ms) and with significant remaining current at the end
the two emission wavelengths. Data were collected and analyzed with the of stimulus were called intermediately adapting (58).
Felix3.2 program (PTI). Phase-contrast and fluorescent images of DRG
neurons in Fig. 1 were taken with an Orca Flash 4.0 camera, mounted on Excised inside-out patch clamp
an Olympus IX-81 inverted microscope. Fluorescence excitation was pro- Excised inside-out patch clamp experiments on HEK293 cells were per-
vided by a Lambda LS light source (Sutter Instruments). formed with borosilicate glass pipettes (World Precision Instruments) of
1.1- to 1.5-megohm resistance. The electrode pipette contained normal
Whole-cell patch clamp extracellular solution. After gigohm-resistance seals were established on
For whole-cell patch clamp, HEK293 cells were transfected with the HEK293 cells, the currents were measured using an Axopatch 200B am-
following complementary DNA (cDNA) constructs: mouse Piezo1 cloned plifier (Molecular Devices). The holding potential was 0 mV, and me-
into a pcDNA3.1 IRES GFP vector (4), hM1 cloned into a pCMV vector, chanical stimulations were applied during steps to –60 mV and +60 mV.
and rat TRPV1 cloned into a pCDNA3 vector. The rapamycin-inducible The perfusion solution contained 140 mM K+ gluconate, 10 mM Hepes,
PI(4,5)P2 and PI(4)P phosphatase system pseudojanin (34) contains a 5 mM EGTA, and 1 MgCl2 (pH adjusted to 7.4).
plasma membrane–anchored FRB fragment of the protein mTOR and an- Excised inside-out macropatch experiments on X. laevis oocytes were
other cytoplasmic fusion protein with both a PI(4,5)P2 5-phosphatase and performed as described (59). Oocytes were injected with the cRNA of
a PI(4)P 4-phosphatase fused to FKBP12. Rapamycin induces a heterodi- Piezo1 transcribed from Piezo1 cloned in the pGEMSH oocyte vector
merization of FKBP12 and FRB and thus the translocation of the phos- (59). We used borosilicate glass pipettes (World Precision Instruments)
phatases from the cytosol to the plasma membrane, which results in the of 0.8- to 1.7-megohm resistance. The electrode pipette contained 96 mM
depletion of PI(4,5)P2 converting it to PI(4)P and then to PI (34). We also NaCl, 2 mM KCl, 1 mM MgCl2, and 5 mM Hepes (pH 7.4). After estab-
used the inactive PI(4,5)P2 phosphatase as a control and the construct that lishing gigohm-resistance seals on devitellinized Xenopus oocytes, we mea-
has only PI(4,5)P2 5-phosphatase activity. sured the currents using an Axopatch 200B amplifier (Molecular Devices).
Whole-cell patch clamp recordings were performed at room tempera- In some experiments, we used a step protocol in which the holding potential
ture (22° to 24°C) as described previously (55). Patch pipettes were pulled was 0 mVand mechanical stimulations were applied during steps to −80 mV
from thick-walled borosilicate glass capillaries (Sutter Instruments) on a and +80 mV. The perfusion solution contained 96 mM KCl, 5 mM EGTA,
P-97 pipette puller (Sutter Instruments) to a resistance of 4 to 6 megohms. and 10 mM Hepes (pH adjusted to 7.4).
After formation of gigohm-resistance seals, the whole-cell configuration Mechanical stimulation of excised inside-out patches was performed
was established and MA currents were measured at a holding potential of using a high-speed pressure clamp (39) (HSPC-1, ALA Scientific) controlled
−60 mV using an Axopatch 200B amplifier (Molecular Devices). Currents by pClamp 9.0 software (Molecular Devices).
were filtered at 2 kHz using the low-pass Bessel filter of the amplifier and
digitized using a Digidata 1440 unit (Molecular Devices). Measurements Materials
were conducted in normal extracellular solution containing 137 mM NaCl, Natural phosphoinositides (purified from porcine brain) mainly consisting
5 mM KCl, 1 mM MgCl2, 2 mM CaCl2, 10 mM Hepes, and 10 mM glucose of arachidonyl and stearyl side chains were purchased from Avanti Polar
(pH adjusted to 7.4 with NaOH). For experiments in Ca2+-free solutions, Lipids. Carbamyl-choline (carbachol), capsaicin, and rapamycin were
CaCl2 was omitted from the extracellular solution. The normal intracellular purchased from Sigma. Stock solutions (1000× concentration) for rapa-
solution consisted of 140 mM K+ gluconate, 1 mM MgCl2, 2 mM Na2ATP, mycin were prepared in dimethyl sulfoxide. Capsaicin stock was prepared
5 mM EGTA, 10 mM Hepes (pH adjusted to 7.2 with KOH). Intracellular in ethanol (1000× concentration or higher). Carbachol stocks were pre-
solution containing 4 mM MgATP (Fig. 3F) was also supplemented with pared in deionized water.
4 mM MgCl2. For experiments with hM1, the normal intracellular solution
was complemented with 0.2 mM Na2–guanosine triphosphate. Series re- Data analysis
sistance was not compensated for whole-cell experiments; measurements Data are presented as means ± SEM. Homogeneity of variances was
with access resistances of 5 to 15 megohms were used for analysis. determined on all data sets using Levene’s test. Groups showed equal var-
Mechanical stimulation of isolated DRG neuron somata and HEK293 iances, and thus, they were analyzed for statistically significant differences
cells in whole-cell experiments was performed using a heat-polished glass using ANOVA or one-sample Student’s t test, where applicable. Bonferroni
pipette (tip diameter, about 3 mm), controlled by a piezo-electric crystal test was applied in multiple comparisons.
drive (Physik Instrumente) positioned at 60° to the surface of the cover
glass. The probe was positioned so that a 10-mm movement did not visibly SUPPLEMENTARY MATERIALS
contact the cell but that an 11.5-mm stimulus produced an observable www.sciencesignaling.org/cgi/content/full/8/363/ra15/DC1
membrane deflection. Steadily increasing series of mechanical steps from Fig. S1. Capsaicin-induced inward (TRPV1) current amplitudes in HEK293 cells transfected
12 mm in 0.4-mm increments were applied at 15-s intervals at the beginning with Piezo1 and TRPV1.
of most experiments. Submaximal stimuli (3.6 to 5.6 mm), based on these Fig. S2. Effects of TRPV1 activation by low pH and TRPA1 activation by AITC on Piezo1
activity.
step protocols for each cell, were applied at 30-s intervals in time-course pro- Fig. S3. Effects of PKC activation on Piezo1-mediated MA currents.
tocols. When cells showed significant swelling upon repetitive mechanical Fig. S4. Fluorescence-based PI(4,5)P 2 and PI(4)P measurements in HEK293 cells
stimulation, measurements were discarded (57). cotransfected with hM1 or TRPV1 and various phosphoinositide sensors.

www.SCIENCESIGNALING.org 10 February 2015 Vol 8 Issue 363 ra15 9


RESEARCH ARTICLE

Fig. S5. Attempts to reactivate Piezo1 currents in excised inside-out patches in HEK293 25. A. Szallasi, P. M. Blumberg, Vanilloid (capsaicin) receptors and mechanisms. Pharmacol.
cells. Rev. 51, 159–212 (1999).
Fig. S6. Cell-attached and excised inside-out patch clamp measurements of MA currents 26. S. E. Kim, B. Coste, A. Chadha, B. Cook, A. Patapoutian, The role of Drosophila
in Xenopus oocytes. Piezo in mechanical nociception. Nature 483, 209–212 (2012).
27. N. Eijkelkamp, J. E. Linley, J. M. Torres, L. Bee, A. H. Dickenson, M. Gringhuis,
M. S. Minett, G. S. Hong, E. Lee, U. Oh, Y. Ishikawa, F. J. Zwartkuis, J. J. Cox, J. N. Wood,
REFERENCES AND NOTES A role for Piezo2 in EPAC1-dependent mechanical allodynia. Nat. Commun. 4, 1682
1. F. Sachs, Stretch-activated ion channels: What are they? Physiology 25, 50–56 (2013).
(2010). 28. L. J. Drew, J. N. Wood, P. Cesare, Distinct mechanosensitive properties of capsaicin-
2. P. Delmas, B. Coste, Mechano-gated ion channels in sensory systems. Cell 155, sensitive and -insensitive sensory neurons. J. Neurosci. 22, RC228 (2002).
278–284 (2013). 29. D. J. Cavanaugh, A. T. Chesler, A. C. Jackson, Y. M. Sigal, H. Yamanaka, R. Grant,
3. B. Nilius, Pressing and squeezing with Piezos. EMBO Rep. 11, 902–903 (2010). D. O’Donnell, R. A. Nicoll, N. M. Shah, D. Julius, A. I. Basbaum, Trpv1 reporter mice
4. B. Coste, J. Mathur, M. Schmidt, T. J. Earley, S. Ranade, M. J. Petrus, A. E. Dubin, reveal highly restricted brain distribution and functional expression in arteriolar
A. Patapoutian, Piezo1 and Piezo2 are essential components of distinct mechan- smooth muscle cells. J. Neurosci. 31, 5067–5077 (2011).
ically activated cation channels. Science 330, 55–60 (2010). 30. V. Lukacs, Y. Yudin, G. R. Hammond, E. Sharma, K. Fukami, T. Rohacs, Distinctive
5. C. Bae, P. A. Gottlieb, F. Sachs, Human PIEZO1: Removing inactivation. Biophys. J. changes in plasma membrane phosphoinositides underlie differential regulation of
105, 880–886 (2013). TRPV1 in nociceptive neurons. J. Neurosci. 33, 11451–11463 (2013).
6. C. Bae, F. Sachs, P. A. Gottlieb, The mechanosensitive ion channel Piezo1 is 31. V. Lukacs, B. Thyagarajan, P. Varnai, A. Balla, T. Balla, T. Rohacs, Dual regulation of
inhibited by the peptide GsMTx4. Biochemistry 50, 6295–6300 (2011). TRPV1 by phosphoinositides. J. Neurosci. 27, 7070–7080 (2007).
7. B. Coste, B. Xiao, J. S. Santos, R. Syeda, J. Grandl, K. S. Spencer, S. E. Kim, M. Schmidt, 32. A. C. Kruse, B. K. Kobilka, D. Gautam, P. M. Sexton, A. Christopoulos, J. Wess,
J. Mathur, A. E. Dubin, M. Montal, A. Patapoutian, Piezo proteins are pore-forming sub- Muscarinic acetylcholine receptors: Novel opportunities for drug development.
units of mechanically activated channels. Nature 483, 176–181 (2012). Nat. Rev. Drug Discov. 13, 549–560 (2014).
8. A. E. Dubin, M. Schmidt, J. Mathur, M. J. Petrus, B. Xiao, B. Coste, A. Patapoutian, 33. B. C. Suh, B. Hille, Recovery from muscarinic modulation of M current channels
Inflammatory signals enhance piezo2-mediated mechanosensitive currents. Cell Rep. requires phosphatidylinositol 4,5-bisphosphate synthesis. Neuron 35, 507–520
2, 511–517 (2012). (2002).
9. P. A. Gottlieb, C. Bae, F. Sachs, Gating the mechanical channel Piezo1: A comparison 34. G. R. Hammond, M. J. Fischer, K. E. Anderson, J. Holdich, A. Koteci, T. Balla, R. F. Irvine,
between whole-cell and patch recording. Channels 6, 282–289 (2012). PI4P and PI(4,5)P2 are essential but independent lipid determinants of membrane identity.
10. P. A. Gottlieb, F. Sachs, Piezo1: Properties of a cation selective mechanical channel. Science 337, 727–730 (2012).
Channels 6, 214–219 (2012). 35. J. van der Wal, R. Habets, P. Varnai, T. Balla, K. Jalink, Monitoring agonist-induced
11. J. Albuisson, S. E. Murthy, M. Bandell, B. Coste, H. Louis-Dit-Picard, J. Mathur, phospholipase C activation in live cells by fluorescence resonance energy transfer.
M. Feneant-Thibault, G. Tertian, J. P. de Jaureguiberry, P. Y. Syfuss, S. Cahalan, J. Biol. Chem. 276, 15337–15344 (2001).
L. Garcon, F. Toutain, P. Simon Rohrlich, J. Delaunay, V. Picard, X. Jeunemaitre, 36. C. Xu, J. Watras, L. M. Loew, Kinetic analysis of receptor-activated phosphoinositide
A. Patapoutian, Dehydrated hereditary stomatocytosis linked to gain-of-function turnover. J. Cell Biol. 161, 779–791 (2003).
mutations in mechanically activated PIEZO1 ion channels. Nat. Commun. 4, 37. S. Santagata, T. J. Boggon, C. L. Baird, C. A. Gomez, J. Zhao, W. S. Shan, D. G. Myszka,
1884 (2013). L. Shapiro, G-protein signaling through tubby proteins. Science 292, 2041–2050
12. C. Bae, R. Gnanasambandam, C. Nicolai, F. Sachs, P. A. Gottlieb, Xerocytosis is (2001).
caused by mutations that alter the kinetics of the mechanosensitive channel PIEZO1. 38. K. V. Quinn, P. Behe, A. Tinker, Monitoring changes in membrane phosphatidylinositol
Proc. Natl. Acad. Sci. U.S.A. 110, E1162–E1168 (2013). 4,5-bisphosphate in living cells using a domain from the transcription factor tubby.
13. R. Zarychanski, V. P. Schulz, B. L. Houston, Y. Maksimova, D. S. Houston, B. Smith, J. Physiol. 586, 2855–2871 (2008).
J. Rinehart, P. G. Gallagher, Mutations in the mechanotransduction protein PIEZO1 39. S. R. Besch, T. Suchyna, F. Sachs, High-speed pressure clamp. Pflugers Arch. 445,
are associated with hereditary xerocytosis. Blood 120, 1908–1915 (2012). 161–166 (2002).
14. B. Coste, G. Houge, M. F. Murray, N. Stitziel, M. Bandell, M. A. Giovanni, A. Philippakis, 40. T. Rohacs, Regulation of transient receptor potential channels by the phospholipase
A. Hoischen, G. Riemer, U. Steen, V. M. Steen, J. Mathur, J. Cox, M. Lebo, H. Rehm, C pathway. Adv. Biol. Regul. 53, 341–355 (2013).
S. T. Weiss, J. N. Wood, R. L. Maas, S. R. Sunyaev, A. Patapoutian, Gain-of-function 41. D. S. Samways, B. S. Khakh, T. M. Egan, Tunable calcium current through TRPV1
mutations in the mechanically activated ion channel PIEZO2 cause a subtype of Distal receptor channels. J. Biol. Chem. 283, 31274–31278 (2008).
Arthrogryposis. Proc. Natl. Acad. Sci. U.S.A. 110, 4667–4672 (2013). 42. T. M. Suchyna, V. S. Markin, F. Sachs, Biophysics and structure of the patch and the
15. S. S. Ranade, Z. Qiu, S. H. Woo, S. S. Hur, S. E. Murthy, S. M. Cahalan, J. Xu, J. Mathur, gigaseal. Biophys. J. 97, 738–747 (2009).
M. Bandell, B. Coste, Y. S. Li, S. Chien, A. Patapoutian, Piezo1, a mechanically activated 43. Y. Karashima, J. Prenen, V. Meseguer, G. Owsianik, T. Voets, B. Nilius, Modulation of
ion channel, is required for vascular development in mice. Proc. Natl. Acad. Sci. U.S.A. the transient receptor potential channel TRPA1 by phosphatidylinositol 4,5-biphosphate
111, 10347–10352 (2014). manipulators. Pflugers Arch. 457, 77–89 (2008).
16. S. Maksimovic, M. Nakatani, Y. Baba, A. M. Nelson, K. L. Marshall, S. A. Wellnitz, 44. D. W. Hilgemann, R. Ball, Regulation of cardiac Na+,Ca2+ exchange and KATP potassium
P. Firozi, S. H. Woo, S. Ranade, A. Patapoutian, E. A. Lumpkin, Epidermal Merkel channels by PIP2. Science 273, 956–959 (1996).
cells are mechanosensory cells that tune mammalian touch receptors. Nature 509, 45. T. Rohacs, C. M. Lopes, I. Michailidis, D. E. Logothetis, PI(4,5)P2 regulates the activation
617–621 (2014). and desensitization of TRPM8 channels through the TRP domain. Nat. Neurosci. 8,
17. S. H. Woo, S. Ranade, A. D. Weyer, A. E. Dubin, Y. Baba, Z. Qiu, M. Petrus, T. Miyamoto, 626–634 (2005).
K. Reddy, E. A. Lumpkin, C. L. Stucky, A. Patapoutian, Piezo2 is required for Merkel-cell 46. J. L. Sui, J. Petit-Jacques, D. E. Logothetis, Activation of the atrial KACh channel by
mechanotransduction. Nature 509, 622–626 (2014). the bg subunits of G proteins or intracellular Na+ ions depends on the presence of
18. S. S. Ranade, S. H. Woo, A. E. Dubin, R. A. Moshourab, C. Wetzel, M. Petrus, J. Mathur, phosphatidylinositol phosphates. Proc. Natl. Acad. Sci. U.S.A. 95, 1307–1312 (1998).
V. Begay, B. Coste, J. Mainquist, A. J. Wilson, A. G. Francisco, K. Reddy, Z. Qiu, 47. V. Lukacs, J. M. Rives, X. Sun, E. Zakharian, T. Rohacs, Promiscuous activation of
J. N. Wood, G. R. Lewin, A. Patapoutian, Piezo2 is the major transducer of mechanical transient receptor potential vanilloid 1 channels by negatively charged intracellular
forces for touch sensation in mice. Nature 516, 121–125 (2014). lipids: The key role of endogenous phosphoinositides in maintaining channel activity.
19. T. Balla, Phosphoinositides: Tiny lipids with giant impact on cell regulation. Physiol. J. Biol. Chem. 288, 35003–35013 (2013).
Rev. 93, 1019–1137 (2013). 48. E. Zakharian, C. Cao, T. Rohacs, Gating of transient receptor potential melastatin 8
20. D. A. Fruman, R. E. Meyers, L. C. Cantley, Phosphoinositide kinases. Annu. Rev. (TRPM8) channels activated by cold and chemical agonists in planar lipid bilayers.
Biochem. 67, 481–507 (1998). J. Neurosci. 30, 12526–12534 (2010).
21. M. J. Rebecchi, S. N. Pentyala, Structure, function, and control of phosphoinositide- 49. E. Zakharian, C. Cao, T. Rohacs, Intracellular ATP supports TRPV6 activity via lipid
specific phospholipase C. Physiol. Rev. 80, 1291–1335 (2000). kinases and the generation of PtdIns(4,5)P2. FASEB J. 25, 3915–3928 (2011).
22. B. C. Suh, B. Hille, PIP2 is a necessary cofactor for ion channel function: How and 50. E. Pospisilova, J. Palecek, Post-operative pain behavior in rats is reduced after single
why? Annu. Rev. Biophys. 37, 175–195 (2008). high-concentration capsaicin application. Pain 125, 233–243 (2006).
23. J. Saarikangas, H. Zhao, P. Lappalainen, Regulation of the actin cytoskeleton-plasma 51. S. M. Kim, J. Kim, E. Kim, S. J. Hwang, H. K. Shin, S. E. Lee, Local application of
membrane interplay by phosphoinositides. Physiol. Rev. 90, 259–289 (2010). capsaicin alleviates mechanical hyperalgesia after spinal nerve transection. Neurosci.
24. M. Hirono, C. S. Denis, G. P. Richardson, P. G. Gillespie, Hair cells require phospha- Lett. 433, 199–204 (2008).
tidylinositol 4,5-bisphosphate for mechanical transduction and adaptation. Neuron 44, 52. S. K. Mishra, S. M. Tisel, P. Orestes, S. K. Bhangoo, M. A. Hoon, TRPV1-lineage
309–320 (2004). neurons are required for thermal sensation. EMBO J. 30, 582–593 (2011).

www.SCIENCESIGNALING.org 10 February 2015 Vol 8 Issue 363 ra15 10


RESEARCH ARTICLE

53. C. Brenneis, K. Kistner, M. Puopolo, D. Segal, D. Roberson, M. Sisignano, S. Labocha, was a gift from D. E. Logothetis (Virginia Commonwealth University), and the rat TRPV1 cloned
N. Ferreiros, A. Strominger, E. J. Cobos, N. Ghasemlou, G. Geisslinger, P. W. Reeh, into a pCDNA3 vector was a gift from D. Julius [University of California, San Francisco (UCSF)].
B. P. Bean, C. J. Woolf, Phenotyping the function of TRPV1-expressing sensory neu- The plasmid for the rapamycin-inducible phosphatase system pseudojanin was a gift from
rons by targeted axonal silencing. J. Neurosci. 33, 315–326 (2013). G. R. Hammond (NIH). The CFP- and YFP-tagged PLCd1 PH domain and OSH2 PH do-
54. S. Srinivas, T. Watanabe, C. S. Lin, C. M. William, Y. Tanabe, T. M. Jessell, F. Costantini, mains were provided by T. Balla (NIH), and the Tubby-R322H mutant tagged with YFP was
Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 a gift from A. Tinker (University College London). The insightful comments of the members
locus. BMC Dev. Biol. 1, 4 (2001). of the Rohacs laboratory were highly appreciated. Funding: This work was supported by
55. Y. Yudin, V. Lukacs, C. Cao, T. Rohacs, Decrease in phosphatidylinositol 4,5-bisphosphate NIH grants NS055159 and GM093290 and a grant from the New Jersey Health Foundation
levels mediates desensitization of the cold sensor TRPM8 channels. J. Physiol. 589, to T.R. Author contributions: I.B. designed the experiments, performed most of the
6007–6027 (2011). experiments, analyzed the data, and wrote the paper. T.R. designed the experiments,
56. A. Balla, Y. J. Kim, P. Varnai, Z. Szentpetery, Z. Knight, K. M. Shokat, T. Balla, Maintenance directed the research, and wrote the paper. D.B. designed and performed some of the
of hormone-sensitive phosphoinositide pools in the plasma membrane requires phospha- experiments. Competing interests: The authors declare that they have no competing in-
tidylinositol 4-kinase IIIa. Mol. Biol. Cell 19, 711–721 (2008). terests. Data and materials availability: Plasmids are available from T.R. upon request,
57. O. P. Hamill, D. W. McBride Jr., Induced membrane hypo/hyper-mechanosensitivity: or requests are referred to the scientist supplying them.
A limitation of patch-clamp recording. Annu. Rev. Physiol. 59, 621–631 (1997).
58. F. Rugiero, L. J. Drew, J. N. Wood, Kinetic properties of mechanically activated currents
Submitted 7 July 2014
in spinal sensory neurons. J. Physiol. 588, 301–314 (2010).
Accepted 9 January 2015
59. T. Rohacs, Recording macroscopic currents in large patches from Xenopus oocytes.
Final Publication 10 February 2015
Methods Mol. Biol. 998, 119–131 (2013).
10.1126/scisignal.2005667
Citation: I. Borbiro, D. Badheka, T. Rohacs, Activation of TRPV1 channels inhibits
Acknowledgments: The mouse Piezo1 cloned into a pcDNA3.1 IRES GFP vector was a mechanosensitive Piezo channel activity by depleting membrane phosphoinositides.
gift from A. Patapoutian (Scripps Research Institute), the hM1 cloned into a pCMV vector Sci. Signal. 8, ra15 (2015).

www.SCIENCESIGNALING.org 10 February 2015 Vol 8 Issue 363 ra15 11


Activation of TRPV1 channels inhibits mechanosensitive Piezo
channel activity by depleting membrane phosphoinositides
Istvan Borbiro, Doreen Badheka and Tibor Rohacs (February 10,
2015)
Science Signaling 8 (363), ra15. [doi: 10.1126/scisignal.2005667]

The following resources related to this article are available online at http://stke.sciencemag.org.
This information is current as of February 15, 2015.

Article Tools Visit the online version of this article to access the personalization and
article tools:
http://stke.sciencemag.org/content/8/363/ra15

Supplemental "Supplementary Materials"


Materials http://stke.sciencemag.org/content/suppl/2015/02/06/8.363.ra15.DC1.html

Related Content The editors suggest related resources on Science's sites:


http://stke.sciencemag.org/content/sigtrans/7/331/ra61.full.html
http://stke.sciencemag.org/content/sigtrans/2001/111/re19.full.html
http://stke.sciencemag.org/content/sigtrans/2005/272/re3.full.html
http://stke.sciencemag.org/content/sigtrans/8/363/fs3.full.html
http://www.sciencemag.org/content/sci/330/6000/55.full.html
http://www.sciencemag.org/content/sci/347/6223/732.9.full.html

References This article cites 59 articles, 31 of which you can access for free at:
http://stke.sciencemag.org/content/8/363/ra15#BIBL

Glossary Look up definitions for abbreviations and terms found in this article:
http://stke.sciencemag.org/cgi/glossarylookup

Permissions Obtain information about reproducing this article:


http://www.sciencemag.org/about/permissions.dtl

Science Signaling (ISSN 1937-9145) is published weekly, except the last December, by the
American Association for the Advancement of Science, 1200 New York Avenue, NW, Washington,
DC 20005. Copyright 2015 by the American Association for the Advancement of Science; all rights
reserved.

You might also like