Articles in PresS. Am J Physiol Cell Physiol (October 15, 2014). doi:10.1152/ajpcell.00037.2014

1

Caveolin-1 Regulates P2X7Receptor Signaling in Osteoblasts

2

Vimal Gangadharan1, Anja Nohe1, Jeffrey Caplan1,2,

3

Kirk Czymmek1,2, Randall L. Duncan1, 2

4 5

Author Affiliation: 1Department of Biological Sciences, University of Delaware, Newark, DE 19716

6

2

Bioimaging Center, Delaware Biotechnology Institute, Newark, DE 19711

7 8 9 10 11 12 13 14 15 16 17 18 19 20 21

Corresponding Author: Randall Duncan, Ph.D. Department of Biological Sciences 118 Wolf Hall University of Delaware Newark, DE 19716, USA Tel: 302.831.4296 Fax: 302.831.2281 Email: [email protected]

Running Head: CAV1 regulates P2X7R signaling in Osteoblasts

22 23 24 25 26 27

Copyright © 2014 by the American Physiological Society.

28

Abstract

29

The synthesis of new bone in response to a novel applied mechanical load requires a

30

complex series of cellular signaling events in osteoblasts and osteocytes. The activation

31

of the purinergic receptor, P2X7R, is central to this mechanotransduction signaling

32

cascade. Recently, the P2X7R have been found to be associated with caveolae, a

33

subset of lipid microdomains found in several cell types. Deletion of caveolin-1 (CAV1),

34

the primary protein constituent of caveolae in osteoblasts, results in increased bone

35

mass, leading us to hypothesize that the P2X7R is scaffolded to caveolae in

36

osteoblasts. Thus, upon activation of the P2X7R, we postulate that caveolae are

37

endocytosed, thereby modulating the downstream signal. Sucrose gradient fractionation

38

of MC3T3-E1 pre-osteoblasts showed that CAV1 was translocated to the denser

39

cytosolic fractions upon stimulation with ATP. Both ATP and the more specific P2X7R

40

agonist, BzATP, induced endocytosis of CAV1, which was inhibited when MC3T3-E1

41

cells were pretreated with the specific P2X7R antagonist, A-839977. The P2X7R co-

42

fractionated with CAV1 but, using Superresolution Structured Illumination Microscopy

43

(SR-SIM), we found that only a sub-population of the P2X7R existed in these lipid

44

microdomains on the membrane of MC3T3-E1 cells. Suppression of CAV1 enhanced

45

the intracellular Ca2+ response to BzATP, suggesting that caveolae regulate P2X7R

46

signaling. This proposed mechanism is supported by increased mineralization observed

47

in CAV1 knockdown MC3T3-E1 cells treated with BzATP. These data suggest that

48

caveolae regulate P2X7R signaling upon activation by undergoing endocytosis and

49

potentially carrying with it other signaling proteins, hence controlling the spatio-temporal

50

signaling of P2X7R in osteoblasts.

2

51

Keywords: Caveolin-1, Purinergic signaling, P2X7R, Endocytosis, Osteoblasts

52

3

53

Introduction

54

Purinergic signaling plays a critical role in regulating skeletal mechanotransduction, an

55

intricate process through which physical stimulus is converted into a biochemical

56

response (7, 23). We, and others, have shown that ATP, and other nucleotides, are

57

released in response to fluid shear stress in vitro and activate purinergic (P2) receptors

58

on the membrane of the osteoblasts (13, 20, 29, 38). Although there are many subtypes

59

of P2 receptors present in osteoblasts (4), knockout of the P2X7R, a ligand gated ion

60

channel, resulted in an osteopenic phenotype (22), suggesting that this receptor is

61

important in skeletal maintenance. Further in vivo loading experiments show that load-

62

induced bone formation is significantly abrogated when the P2X7R is genetically

63

deleted, demonstrating the importance of this receptor in mechanotransduction (23).

64

Several characteristics distinguish the P2X7R from the other P2X receptors. This

65

receptor is 10-30 times more specific for 2’,3’-(benzoyl-4-benzoyl)-ATP (BzATP) than

66

ATP (28) and activation of the receptor by BzATP induces dynamic membrane blebbing

67

(39). P2X7R activation has been associated with the opening of a large pore on the

68

plasma membrane that allows the entry of molecules up to 900 Da in size (28, 46),

69

although recent reports suggests that this pore is actually a pannexin (19). Though the

70

prolonged activation of this receptor has been known to trigger apoptosis in certain cell

71

types, we have shown there is no caspase-3 activation observed in osteoblasts when

72

stimulated with BzATP (23). Stimulation of the P2X7R with BzATP in cells of the

73

osteoblastic lineage results in the expression of osteogenic markers such as osterix and

74

osteocalcin (31) and the release of PGE2 (23), all of which can eventually lead to

75

osteogenesis. Signaling via the P2X7R is also known to activate PLD and PLA2, leading 4

76

to the synthesis of LPA that, in turn, has been attributed to membrane blebbing (31).

77

P2X7R activation also induces ERK phosphorylation in a PKC-dependent manner in

78

osteoblasts (26). The receptor has a significant role in inflammatory responses, wherein

79

it was shown the receptor activation induces the release of inflammatory cytokines from

80

macrophages and osteoclasts (42). Overall this ligand gated ion channel has a major

81

role in several intracellular processes in a cell type specific manner.

82

Recent reports suggest that a sub-population of the P2X7R exists in caveolae, 60-100

83

nm invaginations of the plasma membrane (30, 49), in cells of the submandibular

84

glands, lung alveolae and macrophages (3, 12, 16). These plasma membrane

85

structures are widely expressed in adipocytes, fibroblasts, endothelial cells and

86

osteoblasts (34, 43, 44) and are formed from a subset of components of lipid rafts, such

87

as sphingolipids and cholesterol, and are stabilized by 22 kD integral proteins called

88

caveolins (34).There are three isoforms of caveolin, CAV1, CAV2 and CAV3. CAV1 and

89

CAV2 are found in non-muscle cells, whereas CAV3 is the primarily found in skeletal

90

and some smooth muscle cells (47). The removal of CAV1 and CAV3 prevents the

91

formation of caveolae in these cells, but there was no apparent changes when CAV2

92

was suppressed (5, 11). Numerous studies have demonstrated an important role for

93

caveolae in cell signaling and scaffolding of signaling molecules in several cell types

94

(24, 27, 35). Besides anchoring second messengers, caveolae can also attenuate

95

signaling by endocytosis therefore regulating the presence of the receptors along with

96

other signaling proteins on the plasma membrane (17, 36).

97

The importance of CAV1 and caveolae in regulation of the skeleton is emphasized by a

98

study that demonstrate the knockout of CAV1 in mice increases trabecular and cortical 5

99

bone which appears to result from increased osteoblast function (40). Because both

100

P2X7R and CAV1 are important in skeletal maintenance and the response of bone to

101

mechanical loading, we hypothesized that the P2X7R is present in caveolae of

102

osteoblasts and further postulated that disruption of caveolae will alter P2X7R mediated

103

signaling.

104

Materials and Methods

105

Cell culture: MC3T3-E1 cells, a murine preosteoblast-like cell line (ATCC), were grown

106

in α-Modified Eagles Medium (α-MEM) that was supplemented with 10% FBS (Gemini),

107

100 U/ml penicillin and 100 μg/ml streptomycin. Cells were maintained in a humidified

108

incubator in a 5% CO2/95% air mixture and subcultured every 72 hrs. MC3T3-E1 cells

109

were serum starved for 12-16 hrs prior to all experiments. All cell culture media and

110

antibiotics were purchased from Sigma-Aldrich, St Louis, MO.

111

Reagents and antibodies: Adenosine 5’ Triphosphate disodium salt (ATP); 2’, (3’)-O-

112

(4-Benzoylbenzoyl) adenosine 5’ triphosphate triethylammonium salt (BzATP) and

113

puromycin dihydrochloride (Puromycin) were purchased from Sigma-Aldrich. ATP (250

114

µM; from a stock of 5 mM in PBS) and BzATP (150 µM; from a stock of 6 mM in dH2O)

115

were used for activation of P2X7R. A-839977 (500 nM; from stock of 500 µM in DMSO),

116

a potent P2X7R antagonist was purchased from Tocris Bioscience. Rabbit polyclonal

117

anti-CAV1 and mouse monoclonal anti-CAV1 were obtained from BD Biosciences (San

118

Jose, CA). Rabbit polyclonal anti-P2X7R and FITC conjugated rabbit anti-P2X7R were

119

purchased from Alomone Labs (Jerusalem, Israel). Anti-GAPDH antibody obtained from

120

Abcam (Cambridge, MA) was used as loading control in Western blots. The caveolar

6

121

marker, Alexa Fluor®-555 Cholera Toxin B (CTX-B) and AF-488 donkey anti-rabbit IgG

122

were purchased from Life Technologies (Carlsbad, CA). siRNA against CAV1 was

123

obtained from Santa Cruz Biotechnology (Santa Cruz, CA) and the Sure Silencing

124

plasmid vectors coding shRNA against CAV1 with Puromycin resistance (KM03639P)

125

were purchased from SABiosciences (Frederick, MD).

126

Detergent free isolation of caveolin enriched membrane fractions: Sucrose

127

gradient fractionation was used to isolate caveolin enriched membrane fractions as

128

described by Song, et al. with some modifications (45). Briefly, MC3T3-E1 cells were

129

grown to 80-90% confluence in 150 mm Petri dishes and washed with cold PBS. In the

130

experiments with agonist treatment, the cells were grown in reduced serum (0.2%) for

131

14-16 hrs prior to treatment. The cells were scraped into 2 ml of ice cold 500 mM

132

sodium carbonate containing protease inhibitors (Calbiochem, San Deigo, CA). The cell

133

lysate was then homogenized by sonication, centrifuged at 9000xg for 10 min, and the

134

supernatant was collected. Protein concentration was measured using BCA assay and

135

equal concentrations were subjected to discontinuous sucrose gradient fractionation.

136

A 45% sucrose-lysate solution was made by adding 90% sucrose in MBS (25 mM MES,

137

150 mM NaCl, pH 6.5) to the lysate. This mixture was then placed at the bottom of an

138

ultracentrifuge tube and a discontinuous gradient was prepared on top of this mixture by

139

layering with 4 ml of 35% and 5% sucrose in MBS containing 250 mM sodium

140

carbonate. The gradient was subjected to ultracentrifugation for 18 hrs at 39,000 rpm at

141

4°C with a SW41-TI rotor (Beckman Coulter). A light scattering band was typically

142

observed at the 5%-35% interface where most of the caveolae was enriched. Twelve 1

143

ml fractions were collected from the top of the gradient and the proteins present in each 7

144

fraction were precipitated using TCA-Acetone. The precipitates were then resuspended

145

in sample buffer and equal volumes of the fractions were subjected to Western blotting

146

for the respective proteins.

147

Immunocytochemistry: Cells were seeded at a density of 2.5x103/cm2 onto glass

148

cover slips coated with rat tail type I collagen (100 µg/ml in 0.02 N acetic acid, BD,

149

Franklin Lakes, NJ) and grown to 80-90% confluency. Experimentally, cells were treated

150

with 250 µM ATP or 150 µM BzATP. In one group, cells were pretreated with 500 nM of

151

A-839977 for 30 min prior to the addition of 150 µM BzATP. After 10 min of stimulation,

152

cells were fixed in 4% paraformaldehyde (Electron Microscopy Sciences, Hatfield, PA)

153

containing 0.1% Triton X-100. Fixed cells were washed with PBS and blocked with

154

blocking buffer containing 3% BSA and 10% donkey serum for 2 hrs. Rabbit anti-

155

caveolin-1 antibody (0.5 µg) was then added and the samples were incubated overnight

156

at 4°C. Following incubation, the samples were washed with blocking buffer, and then

157

the secondary antibody, AF-488 donkey anti-rabbit IgG, was added. The cells were then

158

mounted on microscopy slides with Prolong Gold (Life Technologies) and visualized

159

using the Zeiss LSM 780 confocal microscope with a EC Plan Neofluar 40x/1.3 Oil

160

objective lens. Quantification was carried out by manually counting the number of cells

161

with strong membrane staining pattern for CAV1 within the field of view. A minimum of

162

30 cells were counted for each treatment and the percentage of cells with plasma

163

membrane CAV1 was calculated. Each experiment was repeated three times with

164

different cell passages. The line profile of CAV1 staining illustrating the intensity of

165

labeling across the cell was created using the ImageJ software.

8

166

Triton X-100 extraction: MC3T3-E1 cells grown on 100mm Petri dishes were washed

167

three times with ice cold PBS and lysed in 500 µl of cold Triton X-100 lysis buffer (1%

168

Triton X-100, 50 mM Tris, 150 mM NaCl, pH 6.5) containing protease inhibitors. The

169

cells were scraped off the plate and proteins were extracted for 30 min at 4°C. The

170

lysate was then centrifuged at 9000xg for 30 min and supernatant was collected as the

171

soluble fraction (S). The pellet containing the detergent-resistant caveolae was

172

resuspended in 500 µl RIPA buffer (50 mM Tris, 150 mM NaCl, 0.1% SDS, 0.5%

173

sodium deoxycholate, 1% Triton X-100 and protease inhibitors) and sonicated to obtain

174

the insoluble fraction (IS). Equal volumes of both fractions were subjected to Western

175

blotting and probed for CAV1 and the P2X7 receptor.

176

siRNA strategy for transient knockdown of CAV1: Small interfering RNA was used

177

to transiently knockdown CAV1 in MC3T3-E1 cells as per manufacturer’s instructions. In

178

brief, MC3T3-E1 cells were seeded at a cell density of 2.5x103/cm2 onto rat tail type-1

179

collagen-coated (100 µg/ml; BD) glass bottom dishes (MatTek Corporation, Ashland,

180

MA) and grown to 50-60% prior to transfection. For transfection, the culture medium

181

was removed and the cells washed with PBS, followed by addition of 800 µl Opti MEM

182

(Life Technologies). siRNA against CAV1 (0.5 µM; sc-29942) and 6 µl of Oligofectamine

183

(Life Technologies) were diluted in 100 µl of OptiMEM, respectively. The reagents were

184

then gently mixed and incubated for 20 min at room temperature before being added to

185

each dish. After 5-7 hrs of incubation with the siRNA, fresh culture media containing

186

10% FBS was added to the cells. Experiments were conducted after 72 hrs post

187

transfection since Western blotting demonstrated maximum suppression was achieved

188

after 72 hrs. Scrambled siRNA (sc-37007) and PBS were used as controls. 9

189

Generation of stable CAV1 knockdown (CAV1 KD) cells: To generate a clonal line of

190

MC3T3-E1 cells with stable knockdown of CAV1, SureSilencing shRNA plasmid with

191

Puromycin resistance was used. Five sets of plasmids were provided; of which four

192

were specifically targeted against CAV1 mRNA and the fifth plasmid was a non-specific

193

scrambled plasmid that served as a control. Following the manufacturers protocol, each

194

of the plasmids was amplified after transformation into the competent DH5-alpha cells.

195

The plasmids were isolated using QIAGEN Mini Prep and concentration was measured.

196

Prior to transfection with the shRNA encoding plasmids, MC3T3-E1 cells (2.5x103/cm2)

197

were grown on a 12 well plate for 24 hrs. 6 µl of the transfection reagent, Lipofectin (Life

198

Technologies) was diluted in 100 µl of OptiMEM and mixed with 100 µl of OptiMEM

199

containing 1.2 µg of the plasmid and incubated for 20 min at room temperature.

200

Meanwhile the growth medium on the cells was replaced with 800 µl of fresh OptiMEM.

201

After incubation, the transfection mixture was added to the MC3T3-E1 cells and

202

incubated for 5-7 hrs at 37°C. The transfection reagents were then removed and fresh

203

culture media containing 10% FBS was added. After 72 hrs post transfection, the

204

culture media was supplemented with 5 µg/ml of Puromycin, the minimum inhibitory

205

concentration based on the dose response for MC3T3-E1 cells. Cells that did not take

206

up the plasmid failed to develop a resistance to the antibiotic and died. Within a week,

207

colonies of MC3T3-E1 cells with Puromycin resistance were beginning to develop.

208

These colonies were isolated for approximately 8 weeks and were continuously

209

screened for antibiotic resistance. A control group of untransfected MC3T3-E1 cells

210

were also screened to ensure the antibiotic concentration remained effective to

211

completely kill the non-transfected cells. Four different clones were isolated of which 10

212

one was the scrambled control and the extent of CAV1 knockdown was measured using

213

immunoblotting. GAPDH was used as the loading control.

214

Western Blotting: To determine the protein levels of CAV1 following knockdown the

215

siRNA or shRNA treatment, MC3T3-E1 cells were lysed using RIPA buffer and equal

216

amounts of protein were subjected to SDS-PAGE. To detect CAV1, P2X7R and GAPDH

217

in the sucrose fractionation studies and in the Triton-X100 extraction experiments, equal

218

volumes of the samples were loaded onto 4-12% gradient NuPAGE gels (Life

219

Technologies). After electrophoresis, the proteins were transferred to nitrocellulose

220

membrane and blocked ON with 5% milk in TBS/T (TBS containing 0.1% Tween-20).

221

The blots were probed with antibodies against CAV1, P2X7R and GAPDH using the

222

respective antibodies.

223

Calcium imaging: Intracellular Ca2+ levels [Ca2+]i were measured in MC3T3-E1 cells

224

using a fluorescent Ca2+ chelator, Fluo-4 and imaged using Zeiss 510 confocal

225

microscope with a Zeiss 10X C-Apochromat water immersion lens (0.45 numerical

226

aperture). As previously described, MC3T3-E1 cells were grown on rat tail type I

227

collagen-coated glass bottom dishes until cells reached 70% - 80% confluency. Cells

228

were then loaded with the calcium indicator, 10 µM Fluo-4 (Life Technologies) for 20

229

min at 37ºC. The cells were then washed with HBSS to remove excess dye and allowed

230

to recover for 15 min before observation. A 488nm excitation laser was used to visualize

231

[Ca2+]i and the changes in intensity were measured as a time lapse. Images were

232

collected over a period of 1 min prior to the addition of BzATP to establish the baseline

233

and continued for 4 min after the addition of BzATP. The data was analyzed using the

234

Zeiss ZEN Physiology software module (version 2011) and the change in intensity over 11

235

baseline was calculated to determine the increase in intracellular calcium over baseline.

236

The calcium response to BzATP was measured in control and CAV1 suppressed

237

MC3T3-E1 cells. The experiment was repeated 3 times from different cell passages,

238

with a minimum of 15 cells under each condition.

239

Co-localization of P2X7R and cholera toxin B subunit: AF 555-Cholera toxin B (CTX-

240

B; 5 μg/ml) subunit was added to MC3T3-E1 cells grown on coverslips and incubated

241

on ice for 20 min. Subsequently, the cells were washed with PBS and fixed in 4%

242

paraformaldehyde for 20 min at 4ºC. FITC conjugated rabbit anti-P2X7R antibody was

243

used to target the extracellular domain of the receptor after blocking the fixed cells with

244

3% BSA. Prolong Gold was used to mount the immunostained cells on a slide and

245

imaged using the Zeiss ELYRA PS.1 superresolution microscope with the

246

PlanApochromat 63X oil immersion objective (1.4 numerical aperture). Structural

247

illumination data was acquired with 5 rotations and 3 phases to obtain improved

248

resolution and quantification. The excitation lasers used were 488 nm and 561 nm. To

249

minimize artifacts, channels were aligned with an affine channel alignment from a multi-

250

color bead calibration, auto noise filtered, and displayed using a baseline cut for

251

accurate representation for colocalization analysis. Mander’s correlation coefficient was

252

measured by setting an appropriate threshold for each channel using the Zen 2011

253

software. The values describe the total amount of P2X7R colocalized with the CTX-B

254

over the entire CTX-B bound region (8). Mander’s correlation coefficient were measured

255

for 12 randomly selected individual cells over two different experiments and averaged.

256

Mineralization of CAV1 KD cells with BzATP treatment: CAV1 KD cells were seeded

257

in a 12 well plate at 2.5x103/cm2 cell density and grown to confluency after which, the 12

258

cell culture media supplemented with 50 μg/ml of ascorbic acid and 10 mM of β-

259

glycerophosphate (BGP). The cells were treated with the P2X7R agonist, BzATP (150

260

μM) and after 24 hrs fresh media was added. The same process was carried out for 21

261

days with BzATP addition on alternative days. The control cells with the scrambled

262

shRNA were also subjected to the same treatment. The extent of mineralization was

263

then measured either by using von Kossa staining or Alizarin red staining.

264

von Kossa Staining: After 21 days in culture, the cells were washed with cold PBS and

265

fixed in 4% paraformaldehyde for 20 min at 4ºC. 5% aqueous silver nitrate was added to

266

the cells after rinsing with distilled water. The samples were then exposed to UV light for

267

30 min; then the excess silver nitrate was washed off with H2O. The regions of mineral

268

deposits were seen as dark nodules on the plate. Digital images of the wells were taken

269

and the extent of mineralization was quantified using ImageJ software. The intensity of

270

the control untreated cells was set as the threshold and comparisons were made with

271

the suppression of CAV1 and with BzATP treatment in CAV1 KD cells. The data was

272

normalized to the extent of mineralization in the control untreated cells and expressed

273

as fold change.

274

Alizarin Red Staining: After 21 days in culture the cells were washed gently with PBS

275

and fixed in 100% ethanol for 20 min, then air dried. Alizarin red stain (Lifeline Cell

276

Technology, Fredrick MD) was added to the samples and washed away with running

277

water after 15 min. Images were taken randomly encompassing different regions of the

278

well using the Nikon SMZ 1500 light microscope and quantified using ImageJ

13

279

Statistical Analysis: Densitometric analysis was carried out to quantify the extent of

280

CAV1 translocation from the buoyant fractions to the denser fractions using ImageJ

281

software. The samples were all normalized to the protein concentration of the lysate.

282

Mean peak [Ca2+]i response was expressed as a percentage increase in intensity over

283

baseline intensity levels. Experiments were repeated at least three times with cells from

284

3 different cell passages and the data are presented as mean ± SE. Significance of all

285

experiments were considered by One way analysis of variance (ANOVA) and the

286

significance of multiple comparisons was made using the Tukey-Kramer post hoc test.

287

Results:

288

P2X7R activation induces CAV1 endocytosis: To determine if purinergic signaling

289

can stimulate caveolar endocytosis, sucrose gradient fractionation was performed on

290

MC3T3-E1 cells treated with 250 µM ATP. Due to the high concentration of cholesterol

291

and sphingolipids in the caveolae, CAV1 is found in the less dense fractions of the

292

gradient when subjected to ultracentrifugation. However when CAV1 translocates from

293

the membrane microdomains, it loses the buoyant characteristic associated with the

294

lipid microdomains and is found in the denser fractions with other cytosolic components

295

(21). Fig 1A shows that in control conditions, CAV1 was mostly found in the 3-6

296

fractions of the gradient, equivalent to the 5%-35% gradient interface. However, when

297

the cells were stimulated with 250 µM of ATP for 10 min at 37ºC, most of the CAV1 is

298

seen in the denser fractions (9-12 fractions). Normalized densitometric analysis of 3

299

separate experiments shows a significant reduction in the CAV1 present in lipid

300

microdomain fractions in Fig 1B.

14

301

To test our hypothesis that CAV1 is endocytosed due to the activation of P2X7R,

302

MC3T3-E1 cells were treated with ATP, BzATP or BzATP after pretreatment with the

303

specific P2X7R inhibitor, A-839977, for 10 min. Changes in CAV1 localization was

304

determined using immunocytochemistry. Under control conditions, CAV1 was localized

305

to the plasma membrane as indicated by the arrows (Fig. 1C, i), however, when the

306

cells were treated with 250 µM ATP (Fig 1C, ii) or 150 µM BzATP (Fig 1C, iii), the

307

characteristic membrane CAV1 staining was significantly reduced. Treatment with

308

P2X7R agonists resulted in a diffuse and cytosolic staining. Finally, this change in CAV1

309

localization to purinergic stimulation was significantly attenuated when the cells were

310

pretreated with 500 nM A-839977 (Fig 1C, iv), which blocks BzATP induced [Ca2+]i

311

influx (18). Quantification the images shows, under control conditions, approximately

312

60% of cells showed the characteristic CAV1 staining on the plasma membrane and it

313

was significantly reduced with either ATP or BzATP treatment (p

Caveolin-1 regulates P2X7 receptor signaling in osteoblasts.

The synthesis of new bone in response to a novel applied mechanical load requires a complex series of cellular signaling events in osteoblasts and ost...
3MB Sizes 0 Downloads 7 Views