sirna12 sirna attenuates m3r-mediated activation of the pi3k/rock/mlc axis in hasmcs figure 2 –...

1
Gα 12 Facilitates Methacholine-Induced Shortening in Human Airway Smooth Muscle By Modulating Phosphoinositide 3-Kinase-Mediated Activation In A RhoA-Dependent Manner Edwin J. Yoo 1 , Gaoyuan Cao 1 , Cynthia J. Koziol-White 1 , Christie A. Ojiacku 1 , Krishna Sunder 1 , Joseph A. Jude 1 , James V. Michael 2 , Hong Lam 3 , Steven S. An 3 , Raymond B. Penn 2 , and Reynold A. Panettieri, Jr. 1 1 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, NJ; 2 Department of Medicine, Division of Pulmonary and Critical Care Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA; 3 Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; Abstract RATIONALE: Phosphoinositide 3-kinase (PI3K)-dependent activation of rho kinase (ROCK) is necessary for agonist-induced human airway smooth muscle cell (HASMC) contraction, and inhibition of PI3K promotes bronchodilation of human small airways 1 . The upstream mechanisms driving agonist-mediated PI3K/ROCK axis activation, however, remain unclear. Given the capacity of G12 family proteins to activate ROCK pathways in other cell types 2 , the role of G12 proteins in modulating M3R-stimulated PI3K/ROCK activation and HASMC contraction was examined. METHODS: siRNA and pharmacological inhibitors, as well as overexpression of a regulator of G-protein signaling (RGS) protein that limits Gα 12 activation, were used in HASMCs. Phosphorylation of AKT, myosin phosphatase targeting subunit-1 (MYPT1), and myosin light chain-20 (MLC) was measured. Gα 12 coupling was evaluated using co-immunoprecipitation and serum response element (SRE)-luciferase reporter assay. HASMC contraction was evaluated using magnetic twisting cytometry (MTC). Human precision-cut lung slices (hPCLS) were utilized to evaluate bronchoconstriction. RESULTS: Knockdown of Gα 12 attenuated carbachol-induced activation of AKT, MYPT1, and MLC phosphorylation. Gα 12 coimmunoprecipitated with the M3R, and p115RhoGEF-RGS overexpression inhibited carbachol-mediated induction of SRE-luciferase reporter. p115RhoGEF-RGS overexpression inhibited carbachol-induced activation of AKT and HASMC contraction. Moreover, pharmacological inhibition of RhoA blunted carbachol-mediated activation of PI3K. Lastly, RhoA inhibitors induced dilation of hPCLS. CONCLUSIONS: Gα 12 plays a crucial role in HASMC contraction via RhoA-dependent activation of the PI3K/ROCK axis. Inhibition of RhoA activation induces bronchodilation in hPCLS, and targeting Gα 12 signaling may elucidate novel therapeutic targets in asthma. Taken together, these findings provide alternative approaches to the clinical management of airway obstruction in asthma. Gα 12 siRNA attenuates M3R-mediated activation of the PI3K/ROCK/MLC axis in HASMCs Figure 2 – (A) Effect of Gα 12 or scrambled siRNA on protein expression. Data normalized to tubulin expression in the same samples. (B) Effect of carbachol on AKT, MYPT1, and MLC phosphorylation at S473 (pAKT), T696 (pMYPT1), and S19 (pMLC) after transfection with Gα 12 or scrambled siRNA. pAKT, pMYPT1, and pMLC data were normalized to total AKT (AKT), total MYPT1 (MYPT1), and total MLC (MLC). p115RhoGEF-RGS expression attenuates M3R- mediated SRE-luciferase reporter induction, PI3K activation, and HASMC contraction RhoA Inhibitors Induce Bronchodilation of Human Small Airways References Since G12 family proteins regulate smooth muscle tone by activating Rho Kinase, we hypothesize that Gα 12 may regulate HASMC contraction by activation of the PI3K/ ROCK axis. Figure 5 – Airways were preconstricted to carbachol (10 8 –10 4 M) prior to dilation to rhosin or formoterol (10 10 –10 4 M). Data were normalized to maximum forskolin response (10 μM) that was given after max dose of formoterol or rhosin. Each data point is expressed as mean ± SEM. Each group contains 2 airways from each of the three donors (6 total airways). 1. Koziol-White, C. J., Yoo, E. J., Cao, G., Zhang, J., Papanikolaou, E., Pushkarsky, I., Andrews, A., Himes, B. E., Damoiseaux, R. D., Liggett, S. B., Di Carlo, D., Kurten, R. C., and Panettieri, R. A. Jr. (2016) Inhibition of PI3K promotes dilation of human small airways in a rho kinase-dependent manner. British Journal of Pharmacology, 173: 2726–2738. doi: 10.1111/bph.13542. 2. Riobo, N.A., and Manning, D.R. (2005). Receptors coupled to heterotrimeric G proteins of the G12 family. Trends Pharmacol. Sci. 26: 146–154. Human Lungs Inflate & Section Slices (350 µm) Core Gα 12 plays a crucial role in HASMC contraction via RhoA-dependent activation of the PI3K/ ROCK axis. Inhibition of RhoA activation induces bronchodilation in hPCLS, and targeting Gα 12 signaling may elucidate novel therapeutic targets in asthma. Significance Hypothesis Figure 3 – (A) hTERT-immortalized HASMCs expressing p115RhoGEF-RGS and control hTERT-immortalized HASMCs were infected with SRE-luciferase reporter. After carbachol stimulation (10 μM, 6 h), cells were lysed and SRE-luciferase reporter activity was measured. (B-C) HASMCs expressing p115RhoGEF-RGS were stimulated with carbachol and assayed for AKT phosphorylation using immunoblot, and contraction using magnetic twisting cytometry. Data are representative of five independent experiments (n = 6, mean ± SD); statistical comparisons analyzed by unpaired t-test are denoted by lines between tested conditions *P < 0.05. Roles Of PI3K and ROCK in HASMCs M3 muscarinic acetylcholine receptor couples to Gα 12 in HASMCs IP: M3R Ab Control Carbachol HASMC Lysate Control Carbachol IP: Rabbit IgG IP: Gα 12 Ab IB M3R Gα 12 Figure 1 –HASMCs were stimulated with carbachol (10 μM, 1 min) and lysates were immunoprecipitated with anti-M3R or anti-Gα 12 antibody and then probed as indicated. Immunoblot is representative of five independent experiments. Gα 12 Scrambled Gα 12 Tubulin 50 nM siRNA A Scrambled siRNA Gα12 siRNA 0.0 0.5 1.0 1.5 Gα 12 /Tubulin Density (Fold/Basal) * pMYPT1 pAKT pMLC Scrambled Gα 12 50 nM siRNA 10μM Carbachol Scrambled Gα 12 AKT MYPT1 MLC B No Agonist Carbachol 0 1 2 3 4 pAKT S473 /AKT (Fold/Basal) * Scrambled Gα12 siRNA * No Agonist Carbachol 0 1 2 3 4 pMYPT1 S696 /MYPT1 (Fold/Basal) * Scrambled Gα12 siRNA * * No Agonist Carbachol 0.0 0.5 1.0 1.5 2.0 pMLC T18S19 /MLC (Fold/Basal) * Scrambled Gα12 siRNA * * p115RhoGEF-RGS-GFP Expressing hTERT -Immortalized HASMCs hTERT -Immortalized HASMCs No Agonist Carbachol 0.0 0.5 1.0 1.5 2.0 2.5 Relative Luciferase Units (Fold/Basal) * Control p115RhoGEF-RGS * A C pAKT Basal AKT Carbachol p115RhoGEF-RGS Basal Carbachol Control No Agonist Carbachol 0 2 4 6 8 pAKT S473 /AKT (Fold/Basal) * Control p115RhoGEF-RGS * B Control p115RhoGEF-RGS 0 20 40 60 80 100 120 Normalized Contraction % Over Basal * +Carbachol RhoA inhibition blunts M3R-mediated PI3K activation pAKT DMSO 10 μM Rhosin AKT Carbachol DMSO 10 μM Rhosin No Agonist Carbachol 0 2 4 6 pAKT S473 /AKT (Fold/Basal) * Control 10 μM Rhosin * Figure 4 – Effect of rhosin (10 μM, 30 min) on carbachol-induced (10 μM, 10 min) AKT phosphorylation at S473 (pAKT). -10 -8 -6 -4 -2 -25 0 25 50 75 100 125 log[Drug], M Bronchodilation (%Forskolin Response) Formoterol Rhosin Conclusions R Ca ++ Ca ++ Ca ++ Rho A/ Rho Kinase MLCP Gα q PLCβ IP 3 , RyR Ca ++ MLCK CONTRACTION RELAXATION MLC-P i MLC ? PI3K ? Ins$tute for Transla$onal Medicine and Science

Upload: others

Post on 22-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: siRNA12 siRNA attenuates M3R-mediated activation of the PI3K/ROCK/MLC axis in HASMCs Figure 2 – (A) Effect of Gα 12 or scrambled siRNA on protein expression. Data normalized to

Gα12 Facilitates Methacholine-Induced Shortening in Human Airway Smooth Muscle By Modulating Phosphoinositide 3-Kinase-Mediated Activation In A RhoA-Dependent Manner

Edwin J. Yoo1, Gaoyuan Cao 1, Cynthia J. Koziol-White1, Christie A. Ojiacku1, Krishna Sunder1, Joseph A. Jude1, James V. Michael2, Hong Lam3, Steven S. An3, Raymond B. Penn2, and Reynold A. Panettieri, Jr.1

1Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, NJ; 2Department of Medicine, Division of Pulmonary and Critical Care Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA; 3Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland;

Abstract RATIONALE: Phosphoinositide 3-kinase (PI3K)-dependent activation of rho kinase (ROCK) is necessary for agonist-induced human airway smooth muscle cell (HASMC) contraction, and inhibition of PI3K promotes bronchodilation of human small airways1. The upstream mechanisms driving agonist-mediated PI3K/ROCK axis activation, however, remain unclear. Given the capacity of G12 family proteins to activate ROCK pathways in other cell types2, the role of G12 proteins in modulating M3R-stimulated PI3K/ROCK activation and HASMC contraction was examined. METHODS: siRNA and pharmacological inhibitors, as well as overexpression of a regulator of G-protein signaling (RGS) protein that limits Gα12 activation, were used in HASMCs. Phosphorylation of AKT, myosin phosphatase targeting subunit-1 (MYPT1), and myosin light chain-20 (MLC) was measured. Gα12 coupling was evaluated using co-immunoprecipitation and serum response element (SRE)-luciferase reporter assay. HASMC contraction was evaluated using magnetic twisting cytometry (MTC). Human precision-cut lung slices (hPCLS) were utilized to evaluate bronchoconstriction. RESULTS: Knockdown of Gα12 attenuated carbachol-induced activation of AKT, MYPT1, and MLC phosphorylation. Gα12 coimmunoprecipitated with the M3R, and p115RhoGEF-RGS overexpression inhibited carbachol-mediated induction of SRE-luciferase reporter. p115RhoGEF-RGS overexpression inhibited carbachol-induced activation of AKT and HASMC contraction. Moreover, pharmacological inhibition of RhoA blunted carbachol-mediated activation of PI3K. Lastly, RhoA inhibitors induced dilation of hPCLS. CONCLUSIONS: Gα12 plays a crucial role in HASMC contraction via RhoA-dependent activation of the PI3K/ROCK axis. Inhibition of RhoA activation induces bronchodilation in hPCLS, and targeting Gα12 signaling may elucidate novel therapeutic targets in asthma. Taken together, these findings provide alternative approaches to the clinical management of airway obstruction in asthma.

Gα12 siRNA attenuates M3R-mediated activation of the PI3K/ROCK/MLC axis in HASMCs

Figure 2 – (A) Effect of Gα12 or scrambled siRNA on protein expression. Data normalized to tubulin expression in the same samples. (B) Effect of carbachol on AKT, MYPT1, and MLC phosphorylation at S473 (pAKT), T696 (pMYPT1), and S19 (pMLC) after transfection with Gα12 or scrambled siRNA. pAKT, pMYPT1, and pMLC data were normalized to total AKT (AKT), total MYPT1 (MYPT1), and total MLC (MLC).

p115RhoGEF-RGS expression attenuates M3R-mediated SRE-luciferase reporter induction, PI3K

activation, and HASMC contraction

RhoA Inhibitors Induce Bronchodilation of Human Small Airways

References

Since G12 family proteins regulate smooth muscle tone by

activating Rho Kinase, we hypothesize that Gα12 may regulate HASMC contraction by activation of the PI3K/

ROCK axis.

Figure 5 – Airways were preconstricted to carbachol (10−8–10−4 M) prior to dilation to rhosin or formoterol (10−10–10−4 M). Data were normalized to maximum forskolin response (10  µM) that was given after max dose of formoterol or rhosin. Each data point is expressed as mean ± SEM. Each group contains 2 airways from each of the three donors (6 total airways).

1.  Koziol-White, C. J., Yoo, E. J., Cao, G., Zhang, J., Papanikolaou, E., Pushkarsky, I., Andrews, A., Himes, B. E., Damoiseaux, R. D., Liggett, S. B., Di Carlo, D., Kurten, R. C., and Panettieri, R. A. Jr. (2016) Inhibition of PI3K promotes dilation of human small airways in a rho kinase-dependent manner. British Journal of Pharmacology, 173: 2726–2738. doi: 10.1111/bph.13542.

2.  Riobo, N.A., and Manning, D.R. (2005). Receptors coupled to heterotrimeric G proteins of the G12 family. Trends Pharmacol. Sci. 26: 146–154.

http://www.med.upenn.edu/airways/

Elevated Rho Kinase Activity Mediates Intrinsic Hypercontractility in Airway Smooth Muscle From Fatal Asthmatic Subjects

Edwin J. Yoo1, Joseph A. Jude1, Cynthia J. Koziol-White1, Reynold A. Panettieri Jr.1 1University of Pennsylvania, Philadelphia, PA

Hypothesis Formaldehyde induces airway

hyperresponsiveness through enhancing contractile response & mediator release in

airway smooth muscle.

Abstract RATIONALE: Asthma manifests as airway hyperresponsiveness and inflammation. Importantly, human airway smooth muscle (HASM) serves as the pivotal cell regulating bronchomotor tone. Whether an intrinsic abnormality in HASM in asthma exists is controversial. Rho-associated kinase (ROCK) mediates calcium sensitization in HASM. Since ROCK modulates agonist-mediated HASM shortening, we hypothesize that ROCK is differentially activated in HASM cells derived from fatal asthma subjects compared to non-asthma HASM cells. METHODS: HASM cells obtained from fatal asthma and non-asthma subjects were grown to confluence. Cells were treated with bradykinin (1uM) for 10 minutes and then lysed in the presence and absence of Y-27632 (10 uM). ROCK activity and mRNA expression were determined using ELISA and qPCR, respectively. RhoA expression levels were determined by immunoblot analysis. In parallel, PCLS from normal healthy human donors, each containing a small airway, were bronchoconstricted to a dose response of carbachol (10^-8 – 10^-6 M) and bronchodilated to Y27632, or formoterol (10^-9 – 10^-4 M). Maximal bronchodilation as well as area under the curve were compared between formoterol and Y-27632. RESULTS: Baseline ROCK activity was elevated in asthma HASM compared to non-asthma HASM (44%, p=.001, n=5). Peak ROCK acitvity in response to bradykinin was also amplified in asthma HASM (51%, n=2). ROCK activity was blunted in the presence of Y-27632 (60%, p<.0001, n=5). Interestingly, ROCK 1 and 2 mRNA were equally expressed and levels did not differ between asthma and non-asthma HASM. Total RhoA expression also did not differ between asthma and non-asthma HASM. Maximal bronchodilation to Y27632 was comparable to that induced by formoterol (Emax: Formoterol=81.24% +/- 6.152; Y27632=70.03% +/- 6.159). CONCLUSIONS: Rho kinase activity is elevated in asthma HASM without significant changes in ROCK and total RhoA mRNA expression when compared to non-asthma HASM. These findings suggest that Rho Kinase is an important contributor to hyperresponsiveness in asthma HASM and may serve as a new therapeutic target in treatment of allergic airway disease.

Formaldehyde exposure has little effect on agonist-induced [Ca2+]i in

HASM cells

Figure 3 – Human precision cut lung slices (PCLS) were exposed to saline, 0.2, 0.8, or 2.0 ppm formaldehyde for 1 hr, then incubated overnight (18 hr) in fresh medium. Culture supernatants were collected and assayed for A) IL-6 and B) IL-8. Data are representative of mean±SEM for n=3.

Figure 1 – Human precision cut lung slices (PCLS) were exposed to saline, 0.2, 0.8, or 2.0 ppm formaldehyde for 1 hr, then incubated overnight (18 hr). Airways were bronchoconstricted to A) incremental doses of carbachol and both B) maximal bronchocconstriction (Emax) and C) the log of the effective concentration to elicit 50% bronchoconstriction (log EC50) were calculated. Data a representative of mean ±SEM for 4 separate lung donors (*p<0.05, compared to saline).

Formaldehyde exposure promotes airway hyper-reactivity in PCLS

A

B

Formaldehyde exposure does not elicit mediator release in PCLS

A

B

Human precision-cut lung slices (PCLS)8

C

Formaldehyde exposure does not alter IL-6 secretion from HASM cells

Figure 4. Human airway smooth muscle cells were exposed to saline or 0.2-2 ppm formaldehyde for 1 hr and incubated 24 h. In Ca2+-detector dye-loaded cells, Bradykinin (BK, 1uM) or Thrombin (1 U)-induced Ca2+ response was determined. Peak Ca2+ response was determined in 5-9 cells from each treatment/per donor. (n=3, data presented as mean±SEM).

Figure 5 – Human airway smooth muscle cells were exposed to saline or 0.2-2 ppm formaldehyde for 1 h and incubated for 24 h in fresh medium. IL-6 levels in the culture supernatants were determined. TNF-α (10 ng/ml, 24 h) was used as positive control. (n=2, data shown as mean±SEM).

Figure 6– Human airway smooth muscle cells were exposed to saline or 0.2-2 ppm formaldehyde for 1 h and incubated for 24 h in fresh medium. Rho-associated kinase activity was determined in the cell lysates. IL13 (100 ng/ml, for 24 h) was used as the positive control for Rho-kinase induction. Y27632-selective inhibitor of Rho kinase. Data shown as percentage change of enzyme activity from saline; as mean±SEM (n=3).

Formaldehyde exposure decreases ciliary beat response in PCLS

Figure 2 – Human precision cut lung slices (PCLS) were exposed to saline, 0.2, or 0.8 ppm formaldehyde for 1 hr. The ciliary beat frequency (CBF, beats/min) was determined under a video microscope. Data is expressed as % increase in CBF from baseline. Representative data from 6 lung slices obtained from a single donor. Each bar represents mean±SEM in 6 slices.

Formaldehyde exposure alters Rho-Kinase activity in HASM cells

Formaldehyde exposure does not alter Rho kinase mRNA expression in HASM cells

Figure 7. Human airway smooth muscle cells were exposed to saline or 0.2-2 ppm formaldehyde for 1 h and incubated for 24 h in fresh medium. Total RNA was collected and expression of Rho-kinase 1 (ROCK1) and Rho-Kinase 2 (ROCK2) mRNA were determined by qRT-PCR. Expression was normalized to cyclophilin and saline treatment. Data represent 3 experiments as mean±SEM.

Conclusions 1.  Formaldehyde exposure enhances agonist-induced airway

contractility in PCLS, without eliciting inflammatory mediator release.

2.  In HASM cells, formaldehyde exposure has little effect on agonist-induced [Ca2+]i transients.

3.  In HASM cells, formaldehyde exposure elevates Rho-associated kinase activity, suggesting involvement of Ca2+ sensitization mechanism in formaldehyde-induced AHR.

Saline

0.2 ppm

0.8 ppm

2.0 ppm

0

50

100

150

200

250

IL-6

(pg/

ml/u

g pr

otei

n)

Saline

0.2 ppm

0.8 ppm

2.0 ppm

0

100

200

300

400

500

IL-8

(pg/

ml/u

g pr

otei

n)

Acknowledgments This work is funded by the NIH Training Grant T32-ES019851 & CEET center grant P30-ES013508

Human Lungs Inflate & Section Slices (350 µm) Core

PBS or Formalin Solution

5 L/min

Vacuum

1 hr

Fresh medium Contractility

24 hr

Mediators Ciliary Beat

Exposure of PCLS to formaldehyde

Significance Formaldehyde is an indoor air pollutant associated with asthma exacerbation. Understanding the mechanisms of formaldehyde effect on airway smooth muscle may help identify novel therapeutic targets to treat formaldehyde-induced asthma exacerbations.

ROCK1 ROCK2

References 1. Casset A, Marchand C, Purohit A, le Calve S, Uring-Lambert B, Donnay C, Meyer P, de Blay F. Inhaled formaldehyde exposure: effect on bronchial response to mite allergen in sensitized asthma patients. Allergy. 2006 Nov;61(11):1344-50. 2. Gerald McGwin, Jr., Jeffrey Lienert, John I. Kennedy, Jr. Formaldehyde Exposure and Asthma in Children: A Systematic Review. Environ Health Perspect. 2010 March; 118(3): 313–317. 3. Krzyzanowski M, Quackenboss JJ, Lebowitz MD. Chronic respiratory effects of indoor formaldehyde exposure. Environ Res. 1990 Aug;52(2):117-25. 4. Zhao Z, Zhang Z, Wang Z, Ferm M, Liang Y, Norbäck D. Asthmatic symptoms among pupils in relation to winter indoor and outdoor air pollution in schools in Taiyuan, China. Environ Health Perspect. 2008;116:90–97 5. Rumchev KB, Spickett JT, Bulsara MK, Phillips MR, Stick SM. Domestic exposure to formaldehyde significantly increases the risk of asthma in young children. Eur Respir J. 2002;20:403–408. 6. Garrett MH, Hooper MA, Hooper BM, Rayment PR, Abramson MJ. Increased risk of allergy in children due to formaldehyde exposure in homes. Allergy. 1999;54:330–337. 7. Persoz C, Achard S, Momas I, Seta N. Inflammatory response modulation of airway epithelial cells exposed to formaldehyde. Toxicol Lett. 2012 Jun 1;211(2):159-63. 8. Cooper PR, Mesaros AC, Zhang J, Christmas P, Stark CM, Douaidy K, Mittelman MA, Soberman RJ, Blair IA, Panettieri RA. 20-HETE mediates ozone-induced, neutrophil- independent airway hyper-responsiveness in mice. PLoS One. 2010; 5(4):e10235

IL-6

IL-8

0

20

40

60

80

100

10-610-710-8 10-5

Saline0.2 ppm FA0.8 ppm FA2.0 ppm FA

[CCh] M

Bron

choc

onst

rictio

n (%

)

CCh Emax values

Saline

0.2 ppm FA

0.8 ppm FA

2.0 ppm FA

0

20

40

60

80

100Saline0.2 ppm FA0.8 ppm FA2.0 ppm FA

Bron

choc

onst

rictio

n(%

)

log EC50 values

Saline

0.2 ppm FA

0.8 ppm FA

2.0 ppm FA

-0.6

-0.4

-0.2

0.0Saline0.2 ppm FA0.8 ppm FA2.0 ppm FA

Log

EC50

*

Airway smooth muscle & AHR

Hyper-contractility

Airway Remodeling Airway inflammation

Bronchospasm

Cytoplasm

MLC Kinase p-MLC

Ca2+

Rho A

MLC Phosphatase Rho-associated Kinase (ROCK)

MLC

Contractility

Agonist

Ca2+ mobilization

Ca2+ sensitization

Formaldehyde

?

?

Gα12 plays a crucial role in HASMC contraction via RhoA-dependent activation of the PI3K/ROCK axis. Inhibition of RhoA activation induces bronchodilation in hPCLS, and targeting

Gα12 signaling may elucidate novel therapeutic targets in asthma.

Significance

Hypothesis

Figure 3 – (A) hTERT-immortalized HASMCs expressing p115RhoGEF-RGS and control hTERT-immortalized HASMCs were infected with SRE-luciferase reporter. After carbachol stimulation (10  µM, 6 h), cells were lysed and SRE-luciferase reporter activity was measured. (B-C) HASMCs expressing p115RhoGEF-RGS were stimulated with carbachol and assayed for AKT phosphorylation using immunoblot, and contraction using magnetic twisting cytometry. Data are representative of five independent experiments (n = 6, mean ± SD); statistical comparisons analyzed by unpaired t-test are denoted by lines between tested conditions *P < 0.05.

Roles Of PI3K and ROCK in HASMCs

M3 muscarinic acetylcholine receptor couples to Gα12 in HASMCs

IP: M3R Ab

Con

trol

Car

bach

ol

HA

SMC

Lys

ate

Con

trol

Car

bach

ol

IP: R

abbi

t IgG

IP: Gα12 Ab

IB

M3R

Gα12

Figure 1 –HASMCs were stimulated with carbachol (10   µM, 1 min) and lysates were immunoprecipitated with anti-M3R or anti-Gα12 antibody and then probed as indicated. Immunoblot is representative of five independent experiments.

Gα12

Scra

mbl

ed

Gα 1

2

Tubulin

50 nM siRNA

GNA13&

GNA12&

Non&Targe/ng&

GNA12&

GNA13&

siRNA____________&

Tubulin&

GNA13&

GNA12&

Non&Targe/ng&

GNA12&

GNA13&

siRNA____________&

Tubulin&

A

Scrambled siRNA Gα12 siRNA0.0

0.5

1.0

1.5

12/T

ubul

in D

ensi

ty

(Fol

d/B

asal

)

*

pMYPT1

pAKT

pMLC

Scra

mbl

ed

Gα 1

2

50 nM siRNA 10µM Carbachol

Scra

mbl

ed

Gα 1

2

AKT

MYPT1

MLC

pAKT%

pMLC%

10nM

%GNA1

2%

10nM

%GNA1

3%%%

siRNA__________________________________________________________%

pERK%

pMYPT1%

1%um%Carbachol%10min%

Tubulin%

50nM

%GNA1

2%

50nM

%GNA1

3%%% 10nM

%GNA1

2%

10nM

%GNA1

3%%% 50nM

%GNA1

2%

50nM

%GNA1

3%%% 10nM

%GNA1

2%

10nM

%GNA1

3%%% 50nM

%GNA1

2%

50nM

%GNA1

3%%%

10%um%Carbachol%10min%

pAKT%

pMLC%

10nM

%GNA1

2%

10nM

%GNA1

3%%%

siRNA__________________________________________________________%

pERK%

pMYPT1%

1%um%Carbachol%10min%

Tubulin%

50nM

%GNA1

2%

50nM

%GNA1

3%%% 10nM

%GNA1

2%

10nM

%GNA1

3%%% 50nM

%GNA1

2%

50nM

%GNA1

3%%% 10nM

%GNA1

2%

10nM

%GNA1

3%%% 50nM

%GNA1

2%

50nM

%GNA1

3%%%

10%um%Carbachol%10min%

pAKT%

pMLC%

10nM%GNA12%

10nM%GNA13%

%%

siRNA__________________________________________________________%

pERK%

pMYPT1%

1%um%Carbachol%10min%

Tubulin%

50nM%GNA12%

50nM%GNA13%

%%10nM%GNA12%

10nM%GNA13%

%%50nM%GNA12%

50nM%GNA13%

%%10nM%GNA12%

10nM%GNA13%

%%50nM%GNA12%

50nM%GNA13%

%%

10%um%Carbachol%10min%

pAKT%

pMLC%

10nM%GNA12%

10nM%GNA13%

%%

siRNA__________________________________________________________%

pERK%

pMYPT1%

1%um%Carbachol%10min%

Tubulin%

50nM%GNA12%

50nM%GNA13%

%%10nM%GNA12%

10nM%GNA13%

%%50nM%GNA12%

50nM%GNA13%

%%10nM%GNA12%

10nM%GNA13%

%%50nM%GNA12%

50nM%GNA13%

%%

10%um%Carbachol%10min%

pAKT%

pMLC%

10nM

%GNA1

2%

10nM

%GNA1

3%%%

siRNA__________________________________________________________%

pERK%

pMYPT1%

1%um%Carbachol%10min%

Tubulin%

50nM

%GNA1

2%

50nM

%GNA1

3%%% 10nM

%GNA1

2%

10nM

%GNA1

3%%% 50nM

%GNA1

2%

50nM

%GNA1

3%%% 10nM

%GNA1

2%

10nM

%GNA1

3%%% 50nM

%GNA1

2%

50nM

%GNA1

3%%%

10%um%Carbachol%10min%

pAKT%

pMLC%

10nM%GNA12%

10nM%GNA13%

%%

siRNA__________________________________________________________%

pERK%

pMYPT1%

1%um%Carbachol%10min%

Tubulin%

50nM%GNA12%

50nM%GNA13%

%%10nM%GNA12%

10nM%GNA13%

%%50nM%GNA12%

50nM%GNA13%

%%10nM%GNA12%

10nM%GNA13%

%%50nM%GNA12%

50nM%GNA13%

%%

10%um%Carbachol%10min%B

No Agonist Carbachol0

1

2

3

4

pA

KTS4

73/A

KT

(Fol

d/B

asal

)

*ScrambledGα12 siRNA

*

No Agonist Carbachol0

1

2

3

4

pM

YPT1

S696

/MYP

T1

(Fol

d/B

asal

)

*

ScrambledGα12 siRNA

*

*

No Agonist Carbachol0.0

0.5

1.0

1.5

2.0

pM

LCT1

8S19

/MLC

(F

old/

Bas

al)

*

ScrambledGα12 siRNA

**

p115RhoGEF-RGS-GFP Expressing hTERT-Immortalized HASMCs

hTERT-Immortalized HASMCs

No Agonist Carbachol0.0

0.5

1.0

1.5

2.0

2.5

Rel

ativ

e Lu

cife

rase

Uni

ts(F

old/

Bas

al)

*Controlp115RhoGEF-RGS

*

A

C

pAKT

Bas

al

AKT

Car

bach

ol

No

Ago

nist

30 µ

M C

arba

chol

pAKT

pMLC

10 µ

M H

ista

min

e

1U/m

l Thr

ombi

n

50 m

M K

Cl

No

Ago

nist

30 µ

M C

arba

chol

10 µ

M H

ista

min

e

1U/m

l Thr

ombi

n

1ng/

ml P

DG

F

GAPDH

p115RhoGEF-RGS Expressing hTERT Immortalized HASM Cell Line

hTERT D12 hTERT D12 P115RhoGEF-RGS

AKT

No

Ago

nist

30 µ

M C

arba

chol

pAKT

pMLC

10 µ

M H

ista

min

e

1U/m

l Thr

ombi

n

50 m

M K

Cl

No

Ago

nist

30 µ

M C

arba

chol

10 µ

M H

ista

min

e

1U/m

l Thr

ombi

n

1ng/

ml P

DG

F

GAPDH

p115RhoGEF-RGS Expressing hTERT Immortalized HASM Cell Line

hTERT D12 hTERT D12 P115RhoGEF-RGS

AKT

No

Ago

nist

30 µ

M C

arba

chol

pAKT

pMLC 10

µM

His

tam

ine

1U/m

l Thr

ombi

n

50 m

M K

Cl

No

Ago

nist

30 µ

M C

arba

chol

10 µ

M H

ista

min

e

1U/m

l Thr

ombi

n

1ng/

ml P

DG

F

GAPDH

p115RhoGEF-RGS Expressing hTERT Immortalized HASM Cell Line

hTERT D12 hTERT D12 P115RhoGEF-RGS

AKT

No

Ago

nist

30 µ

M C

arba

chol

pAKT

pMLC 10

µM

His

tam

ine

1U/m

l Thr

ombi

n

50 m

M K

Cl

No

Ago

nist

30 µ

M C

arba

chol

10 µ

M H

ista

min

e

1U/m

l Thr

ombi

n

1ng/

ml P

DG

F

GAPDH

p115RhoGEF-RGS Expressing hTERT Immortalized HASM Cell Line

hTERT D12 hTERT D12 P115RhoGEF-RGS

AKT

p115RhoGEF-RGS

Bas

al

Car

bach

ol Control

No Agonist Carbachol0

2

4

6

8

pA

KTS4

73/A

KT

(Fol

d/B

asal

)

*

Controlp115RhoGEF-RGS

*

B

Control p115RhoGEF-RGS0

20

40

60

80

100

120

Nor

mal

ized

Con

trac

tion

% O

ver B

asal

*

+Carbachol

RhoA inhibition blunts M3R-mediated PI3K activation

pAKT

DM

SO

10 µ

M R

hosi

n

AKT

Carbachol

DM

SO

10 µ

M R

hosi

n

No Agonist Carbachol0

2

4

6

pA

KTS4

73/A

KT

(Fol

d/B

asal

)

*Control10 µM Rhosin

*

Figure 4 – Effect of rhosin (10  µM, 30 min) on carbachol-induced (10  µM, 10 min) AKT phosphorylation at S473 (pAKT).

-10 -8 -6 -4 -2-25

0

25

50

75

100

125

log[Drug], M

Bro

ncho

dila

tion

(%Fo

rsko

lin R

espo

nse) Formoterol

Rhosin

Conclusions

R

Ca++ Ca++ Ca++

Rho A/ Rho Kinase

MLCP

Gαq

PLCβ

IP3 , RyR

Ca++

MLCK

CONTRACTION

RELAXATION

MLC-Pi

MLC

?

PI3K

?

                               Ins$tute  for  Transla$onal                            Medicine  and  Science