Articles in PresS. Am J Physiol Cell Physiol (December 3, 2014). doi:10.1152/ajpcell.00275.2013

1

TRPM7 regulates vascular endothelial cell adhesion and tube formation

2 3

Zhao Zeng1,2, Koichi Inoue2, Huawei Sun2, Tiandong Leng2, Xuechao Feng2, Li Zhu1*,

4

Zhi-Gang Xiong2*

5 6

1

7

Key Lab of Thrombosis and Hemostasis, Jiangsu Key Lab of Preventive and translational

8

Medicine for Geriatric Diseases, Soochow University, Suzhou, China

9 10 11 12 13 14

Cyrus Tang Hematology center, Collaborative Innovation Center of Hematology, MOH

2

Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA

*Co-corresponding authors. Li Zhu. Tel.: +86 (512) 65880899 Ext 509; Fax: +86 (512) 65880929; Email: [email protected]. Zhi-Gang Xiong. Tel.: +1 (404) 752 -8683; Fax: +1 (404) 752 -1041; Email: [email protected]; Abstract:

15 16

Transient receptor potential melastatin 7 (TRPM7) is a non-selective cation channel

17

with an α-kinase domain in its C-terminal, known to play a role in diverse physiological

18

and pathological processes such as Mg2+ homeostasis, cell proliferation, and hypoxic

19

neuronal injury. Increasing evidence suggests that TRPM7 contributes to the

20

physiology/pathology of vascular systems. For example, we recently demonstrated that

21

silencing TRPM7 promotes growth and proliferation and protects against hyperglycemia-

22

induced injury in human umbilical vein endothelial cells (HUVECs). Here we

23

investigated the potential effects of TRPM7 on morphology, adhesion, migration, and

24

tube formation of vascular endothelial cells and the potential underlying mechanism. We

25

showed that inhibition of TRPM7 function in HUVECs by silencing TRPM7 decreases

1

Copyright © 2014 by the American Physiological Society.

26

the density of TRPM7-like current and cell surface area, and inhibits cell adhesion to

27

Matrigel. Silencing TRPM7 also promotes cell migration, wound healing, and tube

28

formation. Further studies showed that extracellular signal regulated kinase (ERK)

29

pathway is involved in the change of cell morphology and the increase in HUVEC

30

migration induced by TRPM7 silencing. We also demonstrated that silencing TRPM7

31

enhances the phosphorylation of myosin light chain (MLC) in HUVECs, which might be

32

involved in the enhancement of cell contractility and motility. Collectively, our data

33

suggest that TRPM7 channel negatively regulate the function of vascular endothelial

34

cells. Further studies on the underlying mechanism may facilitate the development of

35

TRPM7 channel as a target for the therapeutic intervention of vascular diseases.

36 37

Key words: TRPM7; HUVECs; adhesion; migration; angiogenesis

38 39

2

40

Introduction

41 42

Vascular endothelial cells play an important role in the regulation of vascular tone,

43

vascular permeability, angiogenesis, and vascular inflammatory response (12, 53). Those

44

cells express a variety of ion channels, including TRP channels that constitute a large

45

family including canonical (TRPC), vanilloid (TRPV), and melastatin (TRPM) sub

46

families (73). At least 19 members among the 28 mammalian TRP channel isoforms have

47

been identified in vascular endothelial cells (38, 73). These channels appear to be

48

involved in diverse physiological/pathological functions.

49 50

Transient receptor potential melastatin 7 (TRPM7), a member of TRPM subfamily, is a

51

Ca2+ and Mg2+-permeant channel protein in possession of its own kinase domain (49, 57-

52

59). It has been reported to be expressed in many types of cells including tumor cells,

53

neurons, hematopoietic cells, granulocytes, leukocytes, and microglia (22, 27, 33, 45, 57,

54

63). As one of two unique channel-kinases along with TRPM6, TRPM7 not only

55

possesses the ion permeability but also phosphorylates bio-molecules such as annexin I,

56

myosin IIA heavy chain, and m-calpain (14, 16, 66). Recent studies have demonstrated

57

that TRPM7 plays an important role in a large number of physiological and

58

pathophysiological processes including cell growth/proliferation, adhesion and migration,

59

motility and movements, oxidative stress and anoxic neuronal death (1, 13, 15, 30, 40,

60

56, 63, 66).

61

3

62

TRPM7 has been shown to be expressed in human vascular system and to play a role in

63

vascular functions (3, 4, 6, 25, 31, 52, 68, 69, 74, 76). For example, in vascular smooth

64

muscle cells, TRPM7 modulates Mg2+ homeostasis (6, 25). In vascular endothelial cells,

65

TRPM7 modulates cell growth and proliferation (4, 31). It was also reported that the level

66

of TRPM7 expression in endothelial cells can be up-regulated by high glucose, oxidative

67

stress and laminar shear stress (3, 68, 69). All of these studies suggest that TRPM7 might

68

be involved in cardiovascular diseases involving endothelial dysfunction. However, the

69

role of TRPM7 on adhesion and angiogenic activity of vascular endothelial cells remains

70

to be elucidated. Here we investigated the effect of TRPM7 on these processes in

71

HUVECs and explored the potential underlying mechanisms.

72 73

4

74

Materials and Methods

75 76 77 78

Regents and antibodies

79

STIM1 and anti-TRPC3 antibodies were from Alamone Labs (Jerusalem, Israel). Anti-

80

phospho-MLC, anti-total MLC, anti-phospho-MEK1/2, anti-total MEK1/2 antibodies and

81

U0126 were purchased from Cell Signaling (Beverly, MA). Growth factor-reduced

82

Matrigel was purchased from BD Biosciences (Bedford, MA). Protease inhibitors

83

cocktail were purchased from Sigma (St. Louis, MO). Phosphatase inhibitors cocktail

84

was from Roche (Indianapolis, IN).

Anti-β-actin antibody was purchased from Abcam (Cambridge, MA). Anti-Orai1, anti-

85 86

Cell culture

87

HUVECs were purchased from ATCC and grown in EGM-2 medium containing 2% fetal

88

bovine serum (FBS) and trophic factors (Lonza, Walkersville, MD), as described in our

89

previous studies (31, 32). In some experiments, HUVECs were cultured in endothelial

90

basal medium (EBM) containing 10% or 2% FBS without trophic factors.

91

Human embryonic kidney (HEK-293) cells, with inducible expression of human TRPM7

92

channels (HEK:TRPM7 cells), were cultured in DMEM supplemented with 10% FBS

93

(30). For the induction of TRPM7, the cells were treated with 1 g/ml of tetracycline.

94 95

RNA interference

96

Knockdown of TRPM7 experiments were performed as described (31, 68). Briefly,

97

dsRNAs 406-426 (TRPM7 siRNA1) and 450-470 (TRPM7 siRNA2) of TRPM7

5

98

(NM_017672.4) were synthesized from Invitrogen. Cells were transfected with 50 nM

99

siRNA using transfection reagent lipofectamineTM RNAiMAX (Invitrogen) according to

100

the manufacturer’s instructions. Non-targeting siRNA (Invitrogen) was used as a negative

101

control.

102 103

Real time PCR

104

Total RNAs were extracted with RNA purification kit (Qiagen, Valencia, CA) and

105

transcribed to cDNA using superscript® First-strand synthesis system (Invitrogen). Real

106

time PCR was performed using SYBR® Green supermix (Bio-Rad, Richmond, CA) in

107

C1000TM Thermal cycler (Bio-Rad) as described (72). 1) 95 °C for 10 min, 2) 95 °C for

108

10 s, 3) 61 °C for 10 s, 4) 72 °C for 15 s, plate read, 5) go to 2, 40 more times, 6) 95 °C

109

for 10 s, 7) Melt curve 65 °C to 95 °C: increment 0.5 °C for 5 s, plate read. Primer sets

110

were described in the “Table 1”.

111 112

Electrophysiology

113

Whole-cell voltage-clamp recordings were performed as described (31). Three to four

114

days after transfection, cells were set on the stage of an inverted microscope (Ti-S;

115

Nikon) and superfused at room temperature with an extracellular solution containing (in

116

mM) 140 NaCl, 5.4 KCl, 1 CaCl2, 10 glucose, and 20 HEPES (pH 7.4 with NaOH, 320–

117

335 mOsm). Patch electrodes were fabricated from borosilicate capillary tubing of 1.5

118

mm diameter (WPI) using a vertical puller (PP-83, Narishige). The electrode resistance

119

ranged from 4 to 5 MΩ when filled with the intracellular solution (see below).

6

120

For current–voltage (I–V) relationship, voltage steps ranged between -100 and +100 mV

121

from a holding potential of -60 mV were applied. All recordings were performed at least

122

5 min after establishing the whole-cell configuration. Membrane currents were recorded

123

using an Axopatch 200B amplifier. Data were filtered at 2 kHz and digitized at 5 kHz by

124

using a Digidata 1440A data-acquisition system and pClamp 10 software (Axon

125

Instruments). Pipette solution contained (in mM) 140 Cs-methanesulfonate, 8 NaCl, 4.1

126

CaCl2, 10 EGTA, 5 tetraethyl-ammonium chloride, 2 Na2-ATP and 10 HEPES (pH 7.3,

127

with CsOH).

128 129

Mg2+ imaging

130

The intracellular Mg2+ levels were examined using Mag-Fura-2 (Invitrogen). Cells were

131

incubated with 5 μM Mag-Fura-2-AM for 30 min at 37 °C in Ca2+-free extracellular

132

solution, which contains (in mM) 140 NaCl, 5.4 KCl, 1 MgCl2, 10 glucose, and 20

133

HEPES (pH 7.4 with NaOH, 320–335 mOsm), followed by de-esterification of the dye

134

for another 30 min at room temperature. The coverslips containing dye-loaded cells were

135

held in a recording chamber placed on the stage of an inverted microscope (Eclipse

136

TE2000-U; Nikon). Mag-Fura-2 was excited at wavelengths of 340 nm and 380 nm.

137

Fluorescence was detected with a 40× objective lens (Super Fluor ×40, numerical

138

aperture = 0.90; Nikon) and a CCD camera (Cool- SNAP ES2; Photometrics). Digitized

139

images were acquired, stored, and analyzed in a personal computer controlled by Axon

140

Imaging Workbench software (INDEC Biosystems). The shutter and filter wheel

141

(Lambda 10-3, Sutter Instrument) were also controlled by the software to allow timed

142

illumination of cells at either 340 or 380 nm excitation wavelengths. Fluorescence was

7

143

detected at an emission wavelength of 510 nm. Ratio images of 340/380 were analyzed

144

by averaging pixel ratio values in circumscribed regions of randomly selected 7-10 cells

145

within the field of view.

146 147

Adhesion assay

148

Ninety-six-well plates were coated with Matrigel (BD Biosciences, 1 mg/ml) overnight at

149

4 °C and washed with sterilized H2O. HUVECs were harvested after transfection with

150

siRNA for 48 h and seeded into 96-well plates (2-4 × 104 /well). After incubation for 1 h

151

at 37 °C, the non-adherent cells were washed away with PBS. Adhered cells were fixed

152

with 4% paraformaldehyde and stained with 0.5% crystal violet for 10 min. Three to five

153

random fields were captured as representative images using light microscope (4×)

154

connected with Nikon camera (D90). Crystal violet was extracted with 10% acetic acid,

155

and optical density was measured at 595 nm.

156 157

Transwell assay

158

Transwell assays were performed using a modified Boyden chamber (8-μm pores,

159

Corning 3422). After transfected with siRNA for 48 h and starved for 24 h, HUVECs (3-

160

4 × 105 cells/ml) in 100 μl of serum-free medium were plated in the upper chamber,

161

whereas 670 μl of medium containing 10% FBS were added to the lower chamber. After

162

incubation for ~18 h in an incubator, cells that did not migrate through the pores were

163

scraped off thoroughly with Q-Tips. Attached cells were fixed with 4%

164

paraformaldehyde for 10 min and stained with 0.5% crystal violet/methanol for 10 min.

165

Pictures of six to nine randomly picked fields for each group were taken by Nikon invert

8

166

light microscope (10×) and the migrated cells were counted. Cells under different

167

treatments were normalized to the control value and expressed as fold change in

168

migration.

169 170

Wound healing assay

171

HUVECs transfected with siRNA for 48 h were seeded to 24-well plates at a density of 1

172

× 105 cells/well. After adhesion to the plate, cells were starved with serum-free medium

173

for 24 h and the cell monolayer was scratched with 1000-μl tip. After washing three times

174

with serum-free medium, cells were incubated with EBM-2 medium containing 2% FBS

175

for 24 h at 37 °C. Pictures were taken from the field of wound area with Nikon camera

176

under light microscope (4×) and analyzed using NIH Image J software. Wound healing

177

rate was calculated as: % wound closure = [(Area of original wound – Area of wound

178

after healing)/Area of original wound] × 100%.

179 180

Tube formation assay

181

Tube formation on Matrigel was evaluated as a model of in vitro angiogenesis. Briefly,

182

60 μl of EBM-2 containing 5 mg/ml Matrigel was added into 96-well plates and

183

incubated at 37 °C for 30 min. HUVECs were harvested after tranfection with siRNA for

184

48-72 h, and then seeded into the 96-well plates pre-coated with Matrigel at a density of 3

185

× 104/well. Capillary tube structures were observed and the representative images were

186

captured with an inverted microscope (4×) equipped with Nikon camera. Tube length was

187

quantified using NIH Image J software.

188

9

189 190

Immunoblotting

191

Immunoblotting was performed as described (30, 31). Briefly, cells were lysed in RIPA

192

buffer (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 1% Triton X-100, 0.5% Sodium

193

deoxycholate, 0.1% SDS, protease inhibitor and phosphatase inhibitor cocktail). After

194

centrifugation at 13,000g at 4 °C for 10 min, the lysates were collected. The aliquots were

195

mixed with laemmli sample buffer and boiled at 95 °C for 10 min. Proteins were

196

separated by 10% SDS-PAGE, transfered to PVDF membranes, and probed with

197

antibodies against phospho-MLC (1:500), total-MLC (1:500), β-actin (1:2000), TRPC3

198

(1:200), Orai1 (1:200), STIM1 (1:500), total MEK1/2 (1:250) and phospho-MEK1/2 (1:

199

1000) followed by HRP-conjugated secondary antibodies (1:2000). The signals were

200

visualized by chemiluminescence using an ECL kit (Millipore).

201 202

Statistical analysis

203

All data were expressed as means ± SEM. Statistical analyses were done by Student t test

204

or one-way analysis of variance on data collected from at least 3 independent

205

experiments. Dunnett’s test was used to compensate for multiple experimental

206

procedures. The differences were considered statistically significant when P < 0.05.

207

10

208

Results:

209 210

In order to investigate the effect of TRPM7 on HUVEC cytoskeleton reorganization,

211

adhesion, migration, and tube formation, specific siRNAs targeting nucleotides 406-426

212

and 450-470 of human TRPM7 (NM_017672.4) were synthesized according to our

213

previous study (31). Data of quantitative real time PCR (Fig. 1A) showed that the level of

214

TRPM7 mRNA in HUVECs transfected with TRPM7 siRNAs for 72 h was decreased as

215

compared to cells transfected with non-targeting control siRNA (TRPM7 siRNA1; 39.67

216

± 12.44 % and TRPM7 siRNA2; 29.49 ± 2.74 % of control, n=3-6, P

TRPM7 regulates vascular endothelial cell adhesion and tube formation.

Transient receptor potential melastatin 7 (TRPM7) is a nonselective cation channel with an α-kinase domain in its COOH terminal, known to play a role ...
2MB Sizes 0 Downloads 6 Views