Articles in PresS. Am J Physiol Lung Cell Mol Physiol (October 17, 2014). doi:10.1152/ajplung.00180.2014

1

Cigarette smoke extract affects mitochondrial function in alveolar epithelial cells

2 3

Korbinian Ballweg1, Kathrin Mutze1, Melanie Königshoff1, Oliver Eickelberg1, Silke

4

Meiners1

5 6

1

7

University, Helmholtz Zentrum München, Munich, Member of the German Center for

8

Lung Research (DZL), Germany.

Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians

9 10

Running Head: Cigarette smoke extract affects mitochondria in alveolar epithelial cells

11 12

Correspondence to:

13

Silke Meiners, PhD

14

Comprehensive

15

Germany

16

Phone:

0049 89 3187 4673

17

Fax:

0049 89 3187 194673

18

Email:

[email protected]

Pneumology

Center,

Max-Lebsche-Platz

31,

81377

München,

19 20 1 Copyright © 2014 by the American Physiological Society.

21

Authors contributions:

22

KB and SM designed the experiments, analyzed the data and wrote the manuscript. KB

23

performed the experiments. KM and MK isolated pmATII cells. KB, KM, MK, OE and SM

24

edited and revised the manuscript.

2

25

Abstract

26

Cigarette smoke is the main risk factor for chronic obstructive pulmonary disease

27

(COPD). Exposure of cells to cigarette smoke induces an initial adaptive cellular stress

28

response involving increased oxidative stress and induction of inflammatory signaling

29

pathways. Exposure of mitochondria to cellular stress alters their fusion/fission

30

dynamics. While mild stress induces a pro-survival response termed stress induced

31

mitochondrial hyperfusion, severe stress results in mitochondrial fragmentation and

32

mitophagy.

33

In the present study, we analyzed the mitochondrial response to mild and non-toxic

34

doses of cigarette smoke extract (CSE) in alveolar epithelial cells. We characterized

35

mitochondrial morphology, expression of mitochondrial fusion and fission genes,

36

markers of mitochondrial proteostasis as well as mitochondrial functions such as

37

membrane potential and oxygen consumption.

38

Murine lung epithelial (MLE)12, as well as primary mouse alveolar epithelial cells

39

revealed pronounced mitochondrial hyperfusion upon treatment with CSE, accompanied

40

by increased expression of the mitochondrial fusion protein mitofusin (MFN) 2 and

41

increased metabolic activity. We did not observe any alterations in mitochondrial

42

proteostasis, i.e. induction of the mitochondrial unfolded protein response or mitophagy.

43

Therefore, our data indicate an adaptive pro-survival response of mitochondria of

44

alveolar epithelial cells to non-toxic concentrations of CSE. A hyperfused mitochondrial

45

network, however, renders the cell more vulnerable to additional stress such as

46

sustained cigarette smoke exposure. As such cigarette smoke induced mitochondrial

3

47

hyperfusion - although being part of a beneficial adaptive stress response in the first

48

place - may contribute to the pathogenesis of COPD.

49

Key words

50

COPD, emphysema, proteostasis, stress-induced-mitochondrial-hyperfusion

51 52

4

53

Introduction

54

Chronic obstructive pulmonary disease (COPD) is one of the leading causes of death

55

worldwide. It is characterized by progressive and irreversible airflow limitation.(4, 44) In

56

the western world the main risk factor for development of COPD is cigarette smoke (CS)

57

(4, 44), which is a complex mixture of thousands of injurious agents and reactive

58

oxidants.(7, 33, 44) Exposure of lung epithelial cells to CS initiates an adaptive cell

59

response such as induction of autophagy, impaired proteasome function, and

60

proinflammatory and oxidative responses.(33, 7, 35)

61

Mitochondria are crucial cellular organelles for cellular energetics, signaling and

62

apoptosis. Differences in cellular energy demands or cellular stress rapidly alter

63

mitochondrial

64

dynamics.(5, 9, 26) Mitochondrial hyperfusion provides a stress resolving mechanism

65

for the cell as elongated mitochondria are protected from degradation via autophagy

66

and show increased efficiency in ATP synthesis(15, 40), thereby contributing to the

67

cells’ ability to repair cellular damage. Furthermore mitochondrial fusion is part of the

68

mitochondrial quality control as increased fusion allows enhanced mixing of

69

mitochondrial components and thus complementation and dilution of mitochondrial

70

damage.(49) In contrast, mitochondrial fragmentation is induced upon severe stress and

71

is an early event upon induction of apoptosis. (13, 25) Damaged and fragmented

72

mitochondria are then removed by mitochondrial autophagy to prevent spreading of

73

damage in the mitochondrial network.(39, 45, 49) Hence, mitochondrial morphology and

74

dynamics represent an important adaptive mechanism in response to cellular stress.

behavior

mainly

by

changing

mitochondrial

fusion

and

fission

5

75

Recently, cigarette smoke-induced alterations in mitochondrial morphology were

76

described in bronchial epithelial cells and asthmatic smooth muscle cells. Hara et al.

77

reported increased mitochondrial fragmentation.(18) In contrast, Hoffmann et al.,

78

observed elongated mitochondria with increased branching in bronchial epithelial cells

79

from COPD patients.(19) In airway smooth muscle cells, CSE induces mitochondrial

80

fragmentation and upregulation of the mitochondrial fission protein DRP1 and

81

downregulation of the fusion protein MFN2.(3) Taken together, these potentially

82

conflicting data suggest cell-type specific differences of mitochondrial morphology in

83

response to cigarette smoke, and emphasize the need for further studies.

84

In this report, we show, for the first time, that cigarette smoke affects mitochondrial

85

morphology of alveolar epithelial cells. Treatment with non-toxic doses of cigarette

86

smoke extract results in hyperfusion of mitochondria and increased expression levels of

87

the mitochondrial fusion protein MFN2. This effect is accompanied by an increase in

88

metabolic activity and mitochondrial membrane potential.

6

89

Materials and Methods

90

Reagents:

91

Carbonyl cyanide 3-chlorophenylhydrazone (CCCP) was dissolved in DMSO to obtain a

92

25 mM stock solution. Antimycin A and rotenone were dissolved in DMSO to a 50 mM

93

stock solution, respectively. Oligomycin A was dissolved to a 5 mM stock solution in

94

DMSO. Tetramethylrhodamin-methylester perchlorate (TMRM) was diluted in DMSO to

95

a 1 mM stock solution. Stock solutions were stored at -20°C and diluted to the

96

appropriate final concentration directly before use. 2,5-diphenyltetrazolium bromide

97

(MTT) was stored at 4°C and dissolved in PBS to a working solution of 5 mg/ml directly

98

before use. All reagents except TMRM were obtained from Sigma-Aldrich (St. Louis,

99

MO). TMRM was purchased from Life Technologies (Carlsbad, CA).

100

Antibodies used were: anti-MFN2 (Abcam, Cambridge, UK), anti-OPA1 (GeneTex,

101

Irvine, CA), anti-DRP1 (Cell Signaling, Cambridge, UK), anti-Calreticulin (Abcam), anti-

102

ATP5A (Abcam), anti-α-Tubulin (GeneTex), anti-PINK1 (Novus Biologicals, Littleton,

103

CO), anti-MFN1 (Novus Biologicals), anti-β-Actin (Sigma-Aldrich), anti-HSP60 (Cell

104

Signaling) and anti-cytochrome C (BD Bioscience, San Jose, CA). Secondary

105

antibodies used were HRP conjugated goat anti-mouse IgG, HRP conjugated goat anti-

106

rabbit IgG (GE Healthcare, Chalfont St Giles UK), and AlexaFluor488 conjugated goat

107

anti-rabbit IgG (Life Technologies).

108

Cell culture

109

The mouse lung epithelial cell line (MLE12) was cultured in RPMI Medium (Life

110

Technologies) containing 10% FCS (PAN Biotech) and 1% Penicillin/Streptomycin (Life

7

111

Technologies). Cells were grown at 37°C in a humidified atmosphere containing 5%

112

CO2. Treatment medium for MLE12 cells consisted of FCS-free RPMI Medium

113

supplemented with 1% Penicillin/Streptomycin.

114

Primary murine alveolar epithelial type II (pmATII) cell isolation and culture

115

pmATII cells were isolated from C57BL/6N mice (Charles River GmbH, Sulzfeld,

116

Germany) at 8-14 weeks of age as describes before. (10, 23) Briefly, mouse lungs were

117

lavaged with 500 µl of sterile PBS twice and flushed through the right heart using 0.9%

118

NaCl solution (B. Braun Melsungen AG, Melsungen, Germany). Lungs were

119

subsequently inflated with 1.5 ml dispase (BD Bioscience, San Jose, CA) and 300 µl of

120

1% low melting point agarose (Sigma-Aldrich) and incubated for 45 min at room

121

temperature (RT). Lungs were minced and consecutively filtered through 100 µm,

122

20 µm, and 10 μm nylon meshes (Sefar, Heiden, Switzerland). Single cell suspension

123

was centrifuged at 200 g for 10 min and the cell pellet was resuspended in DMEM cell

124

culture medium (Sigma Aldrich). Incubation of the single cell suspension on petri dishes

125

coated with antibodies against CD45 and CD16/32 (both BD Bioscience) for 30 min at

126

37°C was performed for depletion of macrophages and lymphocytes. Non-adherent

127

cells were collected and negative selection for fibroblasts was performed by adherence

128

for 25 minutes on cell culture dishes. Again, non-adherent cells were collected and cell

129

viability was determined by trypan blue exclusion. Cell purity was assessed by

130

immunofluorescence staining of cytospin preparations using antibodies for proSFTPC

131

(Merck Millipore, Darmstadt, Germany), panCK (Dako, Hamburg, Germany), CD45 (BD

132

Bioscience), and αSMA (Sigma Aldrich). pmATII cells were resuspended in DMEM

133

supplemented with 10% FCS (PAA Laboratories, Pasching, Austria), 2 mM l-glutamine, 8

134

1% penicillin/streptomycin (both Life Technologies), 3.6 mg/ml glucose (Applichem

135

GmbH, Darmstadt, Germany), and 10 mM HEPES (PAA Laboratories) and cells were

136

cultured for 24 h to allow attachment. Medium was changed and cells were cultured up

137

to 5 days in a humidified atmosphere of 5% CO2 at 37°C. Treatment medium for pmATII

138

cells consisted of FCS free DMEM supplemented with 1% penicillin/streptomycin.

139

Cigarette smoke extract preparation

140

Cigarette smoke extract (CSE) was prepared by bubbling smoke from 6 cigarettes

141

(Research-grade cigarettes (3R4F); Kentucky Tobacco Research and Development

142

Center at the University of Kentucky) through 100 ml of FCS-free cell culture medium at

143

a constant airflow. Smoked medium was then sterile filtered through a 0.20 µm filter

144

(Minisart; Satorius Stedim Biotech, Göttingen, Germany), split into aliquots, stored at

145

-20°C and served as the 100% CSE stock solution. For treatment, CSE was

146

supplemented with 1% penicillin/streptomycin and diluted with treatment medium to the

147

indicated concentrations directly before use. To assure comparable potency of CSE,

148

each stock solution was tested by MTT assays.

149

Cytotoxicity testing

150

Cytotoxicity was assessed using the cytotoxicity detection kit (Roche, Basel,

151

Switzerland) according to the manufacturer’s instructions. For MLE12 cells, 450,000

152

cells/well were seeded into 6-well plates and grown to approx. 80% confluency and

153

treated for the indicated times. 15 minutes before the end of the treatment, one well was

154

treated with 2% Triton X-100 to serve as the control for maximal cellular release of LDH.

155

Supernatants were collected, cleared by centrifugation, and LDH content was measured

156

in technical triplicates according to the manufacturer’s instructions using a Tristar LB 9

157

941 plate reader (Berthold Technologies, Bad Wildbad, Germany). Data were

158

normalized as follows: Untreated cells were set to be 0% cytotoxicity and 2% Triton X-

159

100 treated cells were set to be 100% cytotoxicity.

160

Metabolic activity assay

161

Metabolic activity was measured using the 2,5-diphenyltetrazolium bromide (MTT)

162

assay. Briefly, 100,000 cells/well were seeded into 24-well plates, grown to approx. 80%

163

confluency, and treated for the indicated times. Each concentration was treated in at

164

least six technical replicates. After treatment, 100 ml of freshly prepared thiazolyl blue

165

tetrazolium bromide (Sigma) solution (5 mg/ml in PBS) was added to each well and

166

incubated for 30 min at 37°C. The supernatant was aspirated, and blue crystals were

167

dissolved in 500 ml isopropanol + 0.1% Triton X-100. Absorbance was measured at

168

570 nm using a Tristar LB 941 plate reader (Berthold Technologies).

169

Mitochondrial membrane potential assessment

170

Mitochondrial membrane potential was measured by TMRM fluorescence. 450,000

171

cells/well were seeded in 6-well plates and grown to approx. 80% confluency before

172

treatment for the indicated times. 10 µM CCCP treatment was used as a positive

173

control. After the treatment, cells were stained for 30 minutes in medium containing

174

5 nM TMRM (Life Technologies). Cells were washed with PBS, trypsinized, and

175

resuspended in 500 µl FACS Buffer (2% FCS + 20 µM EDTA in PBS). Samples were

176

then analyzed by FACS analysis and mitochondrial membrane potential was measured

177

as mean TMRM fluorescence intensity using the Becton-Dickinson-LSRII. To ensure

178

total depolarization, CCCP was added to a final concentration of 100 µM to the samples

179

and measurements were repeated after 5, 30, and 60 minutes. 10

180

Analysis of mitochondrial superoxide production

181

Mitochondrial superoxide generation was analyzed using MitoSOX Red. 450,000

182

cells/well were seeded in 6-well plates, grown to approx. 80% confluency, and treated

183

for the indicated times. Antimycin A was used as a positive control to induce

184

mitochondrial superoxide generation. After the treatment, cells were stained for

185

30 minutes in medium containing 5 µM MitoSOX Red (Life Technologies). Cells were

186

washed with PBS, trypsinized, and resuspended in 500 µl FACS Buffer. Samples were

187

then analyzed by FACS analysis and superoxide production was measured as mean

188

MitoSOX fluorescence intensity (BD LSRII).

189

Measurement of cellular ATP levels

190

Cellular ATP levels were measured using CellTiter-Glo assay kit (Promega, Madiso,

191

WI). Briefly, 450,000 cells were seeded in 6 well plates and grown to approx. 80%

192

confluency. Cells were treated with control or CSE containing medium for 24h. After the

193

treatment, cells were trypsinized and 40,000 cells/well were transferred to a 96 well

194

plate. CellTiter-Glo reagent was added and the luminescent signal was measured after

195

shaking the plate thoroughly in a Tristar LB 941 plate reader (Berthold Technologies)

196

Analysis of mitochondrial morphology

197

Analysis of mitochondrial morphology was performed as described elsewhere.(27)

198

Briefly, cells were grown on 15 mm glass coverslips to approx. 50% confluency and

199

treated for the indicated times with CSE or with CCCP as a positive control. After the

200

treatment, cells were fixed with 4% PFA for 10 minutes, permeabilized with 0.1% Triton

201

X-100 in PBS and unspecific binding sites were blocked with Roti-Immunoblock (Roth,

202

Karlsruhe, Germany) for 1 hour at room temperature. After blocking, cells were 11

203

incubated with anti-cytochrome C antibody for 2 hours at room temperature, washed,

204

and incubated with an Alexa Fluor488 coupled secondary antibody for 1 hour at room

205

temperature. All subsequent steps were performed with minimal light exposure. Cells

206

were washed twice for 10 minutes with PBS and nuclei were stained with DAPI (300 nM

207

in PBS for 5 minutes). Finally, cells were mounted on microscopic slides using

208

fluorescent mounting medium (Dako, Hamburg, Germany) and imaged using confocal

209

laser-scanning microscopy (Zeiss LSM710, Oberkochen, Germany).

210

Cells were categorized into three classes according to their mitochondrial morphology:

211

Cells displaying an intact network of tubular mitochondria were classified as normal.

212

When this network was disrupted and mitochondria appeared predominantly spherical

213

or rod-like, they were classified as fragmented. Cells with considerably elongated

214

mitochondria, which were more interconnected, were classified as hyperfused. The

215

mitochondrial morphology of on average 100 cells was determined by a blinded

216

observer.

217

Preparation of cell lysates and mitochondrial fractions

218

For protein lysate preparation, 450,000 cells/well were seeded in 6-well plates and

219

grown to approx. 80% confluency. Cells were treated for the indicated times with CSE

220

or control medium. After treatment, cells were trypsinized, washed with PBS, and lysed

221

in RIPA buffer (50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1% Nonidet P-40, 0.5% sodium

222

deoxycholate, and 0.1% SDS) supplemented with protease inhibitor cocktail (Complete,

223

Roche).

224

Mitochondria were isolated by ultracentrifugation after cellular lysis using a cell

225

disruption bomb (Parr Instruments, Moline, IL). In short, cells were plated in 15 cm 12

226

dishes, grown to 80% confluency, and treated for 24h with 25% CSE. After the

227

treatment, cells were scraped, pelleted and resuspended in mitochondrial isolation

228

buffer (220 mM mannitol, 70 mM sucrose, 5 mM HEPES-KOH, 1 mM EGTA-KOH)

229

supplemented with protease inhibitor cocktail (Complete; Roche). Cells were lysed by

230

stirring 15 min in a cell disruption bomb at 800 psi nitrogen pressure and subsequent

231

rapid depressurizing. Cell lysates were then centrifuged at 600 g to remove nuclei. The

232

supernatant was centrifuged at 8000 g to obtain a crude mitochondrial pellet. The

233

mitochondrial pellet was resuspended in mitochondrial isolation buffer to serve as the

234

mitochondrial fraction, while the supernatant contained the cytosolic fraction. Protein

235

content was determined using the Pierce BCA protein assay kit (Thermo Fisher

236

Scientific, Waltham, MA).

237

Western blot analysis

238

For Western blot analysis, 10-20 µg of protein were subjected to electrophoresis on

239

7.5% SDS-PAGE gels and blotted onto polyvinylidenedifluoride (PVDF) membranes.

240

Membranes were treated with antibodies using standard Western blot techniques. The

241

ECL Plus Detection Reagent (GE Healthcare) and Super Signal West Femto (Thermo

242

Fisher Scientific) were used for chemiluminescent detection, and membranes were

243

analyzed using X-Omat LS films (Carestream, Rochester, NY) in a Curix 60 developer

244

(Agfa, Mortsel, Belgium). Densitometry analysis was performed using the ImageLab

245

Software (Biorad, Hercules, CA).

246

Quantitative real-time RT-PCR

247

Total RNA from cells was isolated using Roti-Quick-Kit (Carl Roth, Karlsruhe,

248

Germany). 100-1000 ng per sample of total RNA were reverse-transcribed using 13

249

random hexamers (Life Technologies) and M-MLV reverse transcriptase (Sigma-

250

Aldrich). Quantitative PCR was performed using the SYBR Green LC480 System

251

(Roche Diagnostics, Mannheim, Germany). The following gene-specific primer were

252

used

253

Fw-MFN1: 5’-TGTGTTCGGATTTTCAAGAGGACA-3’

254

Rv-MFN1: 5’-CTCCTGGGCTGCATTATCCG-3’

255

Fw-MFN2: 5’-CATGTCCACGATGCCCAAC-3’

256

Rv-MFN2: 5’-GACAAAGTGCTTGAGAGGGG-3’

257

Fw-DRP1: 5’-AGGAGATGCAGAGGATCATTCAG-3’

258

Rv-DRP1: 5’-ATCAGCAAAGTCGGGGTGTT-3’

259

Fw-OPA1: 5’-ATGATTGGGCCAGACTGGAA-3’

260

Rv-OPA1: 5’-AGGTAAGCTGGGTGCTCATC-3’

261

Measurement of oxygen consumption

262

Cells were seeded in 10 cm dishes, grown to approx. 80% confluency and treated with

263

control medium or 25% CSE containing medium. After 24h of treatment, cells were

264

washed with PBS, trypsinized, and 5x106 cells were transferred in an Oxygraph

265

chamber (Oroboros Instruments, Innsbruck, Austria) in respiration buffer (0.5 mM

266

EGTA, 3 mM MgCl2, 60 mM K-lactobionate, 20 mM Taurine, 10 mM KH2PO4, 20 mM

267

HEPES-KOH, 110 mM Mannitol and 1 g/l BSA). Oxygen consumption was measured at

268

basal conditions (basal respiration), after addition of 2.5 µM Oligomycin (blocking proton

269

backflow through complex V to measure residual proton leak), after addition of 1 µM

14

270

CCCP (uncoupling the respiratory chain to enable unlimited proton flow through the

271

mitochondrial membrane to induce maximum respiration) and after addition of 2.5 µM

272

Antimycin A/Rotenone (blocking Complex I and III to inhibit mitochondrial respiration).

273

These concentrations were also previously shown to be suitable for measuring oxygen

274

consumption rates in MLE12 cells.(11)

275

Statistical analysis

276

One-way ANOVA with Dunnett’s multiple comparison test was used for statistical

277

analysis of mitochondrial membrane potential and mitochondrial superoxide production.

278

Differences in protein expression data were evaluated using t-test or one-way ANOVA

279

with Dunnett’s multiple comparison test. Alterations in mitochondrial morphology were

280

analyzed using the chi-square test. Statistical analysis was performed using GraphPad

281

Prism software (version 5.00).

282

15

283

Results

284

Cigarette smoke induces mitochondrial elongation in MLE12 mouse alveolar epithelial

285

cells

286

In order to analyze the effect of cigarette smoke on mitochondrial function of alveolar

287

epithelial cells, we treated the mouse alveolar epithelial cell line MLE12 with non-toxic

288

doses of cigarette smoke extract (CSE) and stained mitochondria with cytochrome-C

289

antibody. CSE doses up to 25% CSE were non-toxic as assessed by LDH release

290

assays and absence of caspase 3 cleavage (Figures 1A&B) and shown before (35). Of

291

note, non-toxic doses of CSE induced a striking elongation of mitochondria (Figure 2A,

292

Panel B). In order to quantify CSE-induced changes in mitochondrial morphology, we

293

classified mitochondrial morphology into three types: “fragmented”, “normal” or

294

“hyperfused” morphology (for details see materials & methods part). CSE significantly

295

increased the fraction of cells with hyperfused mitochondria compared to untreated

296

control cells after 6, 24, or 48h of treatment (Figure 2B). As a positive control for altered

297

mitochondrial morphology, we treated cells with CCCP, a mitochondrial uncoupling

298

reagent that is well-known to induce fragmentation of mitochondria.(12, 39) Indeed,

299

CCCP treatment led to pronounced and time-dependent mitochondrial fragmentation,

300

confirming that our classification strategy is a valid tool for semiquantitative analysis of

301

mitochondrial morphology (Figures 2A&B).

302

The observed changes in mitochondrial morphology were further analyzed by

303

expression analysis for the mitochondrial fusion and fission proteins MFN1 and MFN2

304

(involved in fusion of outer mitochondrial membranes), OPA1 (inner mitochondrial

305

membrane fusion), and DRP1 (involved in mitochondrial fission). MFN2 levels 16

306

significantly increased within 24h of treatment with 10% or 25% CSE (Figures 3A&B)

307

and returned to baseline after 48h. In contrast, no significant changes in response to

308

CSE were observed for MFN1, OPA1, and DRP1 expression (Figures 3A&B). Again,

309

CCCP served as a positive control to detect changes in mitochondrial morphology on

310

the level of fusion and fission proteins. We observed 5 bands for OPA1 and 2 bands for

311

DRP1 according to different isoforms of the proteins. In accordance with published data

312

(12, 39), we observed reduction in MFN2 levels and proteolytic cleavage of long OPA1

313

isoforms (a,b) at early time points of CCCP treatment (Figures 3A&B). Protein levels of

314

the fission protein DRP1, which translocates from the cytosol to mitochondria for

315

triggering mitochondrial fission (16), were analyzed in isolated mitochondrial fractions of

316

CSE treated cells. We found no consistent change in DRP1 levels in our enriched

317

mitochondrial fractions (Figures 3C&D). Similar results were obtained by analysis of

318

mRNA levels of mitochondrial fusion and fission proteins. Only MFN2 mRNA showed a

319

transient increase after 24h of 25% CSE treatment (Figure 3E). Taken together, our

320

data reveal increased mitochondrial fusion as an adaptive response to treatment with

321

low and non-toxic doses of cigarette smoke extract.

322

CSE-mediated mitochondrial elongation is associated with increased mitochondrial

323

function

324

As mitochondrial hyperfusion has previously been described as a mechanism to

325

increase mitochondrial ATP production (15, 40), we comprehensively investigated

326

mitochondrial function in CSE treated MLE12 cells by analyzing several hallmarks of

327

mitochondrial function. Metabolic activity was significantly increased at non-toxic doses

328

of CSE, as analyzed by the MTT assay while increasing doses of CSE up to 75% dose17

329

dependently decreased metabolic activity and survival of alveolar epithelial cells

330

(Figure 4A). Concordantly, ATP levels were increased in MLE12 cells after 24h of CSE

331

exposure (Figure 4B). Furthermore, CSE treatment resulted in a significant increase in

332

mitochondrial membrane potential after 6h, 24h, and 48h of treatment while the

333

uncoupling agent CCCP efficiently decreased the mitochondrial membrane potential as

334

assessed by measuring TMRM fluorescence (Figure 5A). Specificity of TMRM

335

dependent assessment of mitochondrial membrane potential was confirmed by

336

uncoupling of the respiratory chain and concomitant loss of TMRM fluorescence with

337

high concentrations of CCCP (data not shown).

338

Elevation of the mitochondrial membrane potential is often associated with increased

339

formation of mitochondrial ROS.(37) Hence, we measured mitochondria-derived

340

superoxides using the mitochondria specific probe MitoSOX Red. Remarkably,

341

mitochondrial superoxide production was not altered after CSE treatment (Figure 5B)

342

indicating that the increase in mitochondrial membrane potential does not induce

343

enhanced mitochondrial superoxide formation. The function of our probe was confirmed

344

by adding Antimycin A, a commonly used inducer of mitochondrial superoxide (31),

345

which markedly increased MitoSOX-specific fluorescence (data not shown).

346

To assess the underlying mechanism for the increase in mitochondrial membrane

347

potential, we further analyzed mitochondrial oxygen consumption in CSE-treated cells.

348

An increase in mitochondrial membrane potential can be caused by increased flow of

349

protons into the intermembrane space and enhanced rate of oxygen consumption

350

through increased activity of the respiratory chain. Alternatively, reducing proton

351

backflow into the mitochondrial matrix due to inhibition of complex V of the respiratory 18

352

chain can also increase mitochondrial membrane potential. The rate of basal oxygen

353

consumption and respiration was slightly increased in CSE treated alveolar epithelial

354

cells. Proton leakage through the mitochondrial inner membrane was not affected by

355

CSE treatment as revealed by inhibition of proton backflow by Oligomycin-induced

356

inhibition of complex V. Maximal mitochondrial oxygen consumption can be assessed

357

by triggering unlimited proton flow through the membrane by CCCP. Maximum

358

respiration was also slightly elevated in CSE treated cells. Finally, blocking complex I

359

and III by Rotenone and Antimycin A demonstrated that oxygen consumption was

360

indeed due to respiratory chain activity (Figures 5C&E). Importantly, mitochondrial ATP

361

production measured as Oligomycin sensitive respiration was also increased in CSE

362

treated cells (Figure 5D).

363

Altogether, these data indicate an increase in mitochondrial activity in alveolar epithelial

364

cells in response to CSE in the absence of elevated levels of mitochondrial superoxides.

365

CSE treatment of MLE12 cells does not alter mitochondrial proteostasis

366

Damage of mitochondria was reported to affect mitochondrial proteostasis as indicated

367

by upregulation of the mitochondrial chaperone HSP60 as part of the mitochondrial

368

unfolded protein response (mitoUPR) (52) and to trigger disposal of defective

369

mitochondria

370

macroautophagy.(50) To analyze whether treatment of alveolar epithelial cells with low

371

doses of CSE affects mitochondrial proteostasis, we analyzed expression levels of the

372

mitochondrial chaperone HSP60 and of ATP5A as a surrogate marker for mitochondrial

373

mass in cell lysates. We furthermore analyzed PINK1 expression in isolated

374

mitochondrial fractions. CSE treatment did not affect expression of HSP60 or ATP5A in

by

PINK1/Parkin-mediated

mitophagy,

a

specialized

form

of

19

375

MLE12 cells after 6, 24, or 48h of treatment (Figures 6A&B). Accumulation of full length

376

PINK1 on the mitochondrial outer membrane leads to the initiation of mitophagy. (20) In

377

MLE12 cells, we detected a double band for full length PINK1 with a molecular weight of

378

67 kDA and one band for cleaved PINK1 (55 kDA) which was not altered in

379

mitochondrial fractions of CSE or control treated cells (Figures 6C&D). These results

380

suggest that CSE treatment with low doses induces an adaptive mitochondrial response

381

in alveolar epithelial cells that does not involve pronounced alterations in mitochondrial

382

proteostasis.

383

Cigarette smoke induces mitochondrial elongation in primary mouse alveolar epithelial

384

type II cells

385

To shed light on the effects of CSE on mitochondrial morphology in primary alveolar

386

epithelial cells, we treated primary mouse alveolar epithelial type II (pmATII) cells with

387

CSE for 24h. The pmATII cells showed increased resistance to CSE compared to

388

MLE12 cells as analyzed by LDH release assays (data not shown). Cells treated with

389

50% CSE exhibited pronounced mitochondrial hyperfusion, similar to the hyperfused

390

mitochondria observed for MLE12 cells (Figures 7A&B). Additionally, as observed in

391

MLE12 cells, expression of MFN2 was slightly elevated in CSE treated pmATII. No

392

change in different OPA1 isoforms or in MFN1 and DRP1 levels was detected (Figure

393

8A&B). These data confirm that mitochondrial hyperfusion is a conserved feature of

394

alveolar epithelial cells in the adaptive response to low and non-toxic doses of cigarette

395

smoke.

396

20

397

Discussion

398

In the present study, we demonstrate that MLE12 alveolar epithelial cells respond to

399

non-toxic doses of CSE with mitochondrial hyperfusion involving increased levels of the

400

mitochondrial fusion protein MFN2. Elongation is accompanied by functional

401

mitochondrial changes such as increased metabolic activity and membrane potential

402

upon CSE treatment. Mitochondrial elongation and upregulation of MFN2 were

403

confirmed in primary mouse ATII cells.

404

Non-toxic doses of cigarette smoke extract do not impair mitochondrial proteostasis of

405

alveolar epithelial cells

406

Cigarette smoke exposes lung cells to a plethora of harmful and highly reactive

407

chemicals and is a potent inducer of cellular damage.(7, 33, 44) We exposed cells to

408

cigarette smoke extract which is a commonly used model to assess the effect of

409

cigarette smoke in a cell culture system. In this model system, we found the applied

410

cigarette smoke doses to be non-toxic as assessed by the absence of apoptotic and

411

necrotic cell death markers. Non-toxic doses of cigarette smoke have been shown to

412

affect function and activity of numerous cellular proteins and impair overall protein

413

homeostasis in lung alveolar and bronchial cells (6, 29, 35). Furthermore, cigarette

414

smoke affects organelle-specific proteostasis as shown for activation of the unfolded

415

protein response in the ER in lung tissue homogenates of chronic smokers and in CSE

416

treated cells.(22, 47) Damaged and misfolded proteins in the mitochondria activate the

417

mitochondrial unfolded protein response (mitoUPR). Upregulation of the mitochondrial

418

matrix chaperone HSP60 is a hallmark for mitoUPR induction.(21, 34, 52) Severely

419

damaged mitochondria depolarize and are degraded via mitophagy involving 21

420

PINK1/Parkin activation.(20, 39, 45) We therefore assessed mitochondrial quality

421

control in response to cigarette smoke extract. However, we found no signs for impaired

422

mitochondrial function or induction of mitochondrial damage in alveolar epithelial cells

423

upon non-toxic CSE treatment. HSP60 levels did not increase in CSE treated lung

424

epithelial cells, suggesting the absence of mitoUPR activation. Furthermore, we did not

425

observe any signs of mitochondrial autophagy as levels of the mitophagy marker PINK1

426

and the mitochondrial mass (measured by ATP5A expression) remained unaltered by

427

non-toxic CSE treatment. We thus conclude that our applied non-toxic doses of

428

cigarette smoke extract do not harm mitochondria of alveolar epithelial cells to such an

429

extent that seriously challenges mitochondrial proteostasis. Indeed, our CSE treatment

430

regimen induced only mild oxidative stress to mitochondria as indicated by our finding

431

that mitochondrial superoxide production was not increased by CSE treatment. These

432

mild and probably physiologically more relevant treatment conditions are clearly

433

different from those used in another study that observed induction of necrotic cell death

434

by cigarette smoke extract in primary bronchial epithelial cells.(41) During the

435

preparation of this manuscript another study was published showing increased PINK1

436

expression and mitophagy in bronchial epithelial cells and CS exposed mice. (30) In this

437

study, mitophagy was an upstream event of cigarette smoke induced necroptosis. (30)

438

This seems plausible given the fact that mitochondria are an integral component of

439

apoptosis and necroptosis pathways. (25, 46) It also indicates that the effect of cigarette

440

smoke extract on pulmonary epithelial cells is strongly dependent on the dose and the

441

effects on cell survival. Additionally, there might be not only a dose- but also cell type

442

dependent responsiveness between alveolar and bronchial epithelial cells to cigarette

22

443

smoke extract. Furthermore, a time dependency was observed in wood smoke treated

444

guinea pigs showing early decrease of mitochondrial function, but full recovery and

445

overcompensation later on. (17) In our study, we cannot rule out that mitochondrial

446

function is transiently decreased at earlier time points than 6 hours and improves later.

447

Non-toxic doses of cigarette smoke extract induce adaptive mitochondrial hyperfusion

448

CSE treatment of alveolar epithelial cells induced an adaptive mitochondrial response

449

involving mitochondrial elongation and fusion. Mitochondrial hyperfusion was associated

450

with increased levels of the mitochondrial fusion protein MFN2. Expression of the

451

mitochondrial mass marker ATP5A, the fusion protein MFN1, the inner membrane

452

fusion protein OPA1, and the mitochondrial fission protein DRP1 were not altered in the

453

respiratory epithelial cells. Mitochondrial hyperfusion was initially described to be

454

dependent on OPA1 and MFN1, but not MFN2, however, without expressional changes

455

in any of these proteins. (40) In contrast, starvation induced hyperfusion was dependent

456

on DRP1 phosphorylation and its decreased translocation to mitochondria but

457

expression levels of fusion and fission protein remained unchanged. (15) Of note, we

458

did not observe any mitochondrial translocation of DRP1 in our study indicating that the

459

mechanism of CSE induced hyperfusion is different to starvation induced elongation

460

and probably not mediated by decreased fission activity. Lastly, increased

461

oligomerization of fusion proteins was observed in oxidative stress mediated

462

hyperfusion of mitochondria without changes of absolute fusion and fission protein

463

levels. (38) This clearly shows that expressional changes are not necessary to trigger

464

stress induced hyperfusion but rather altered activity of the fusion and fission machinery

465

contributes to mitochondrial hyperfusion. Concordantly, we find hyperfusion as early as 23

466

6h of treatment where no changes in Mitofusin levels are present and hyperfusion lasts

467

up to 48h were MFN2 levels are already declining to baseline arguing that changes in

468

activity are the critical factor for the observed hyperfusion. As all mitochondrial fusion

469

and fission proteins are present in the cells we cannot determine which proteins actually

470

are responsible for the observed morphology. However, overexpression of MFN2 in

471

neurons or in pulmonary artery smooth muscle cells induced mitochondrial elongation

472

(8, 36) hence, increased fusion and especially increased levels of MFN2 may

473

functionally influence mitochondrial hyperfusion. Thus, upregulation of MFN2 levels

474

might be an adaptive response which is additional to the described stress induced

475

hyperfusion pathways. However, as levels and activity of mitochondrial fusion and

476

fission proteins need to be tightly balanced elevated MFN2 levels are probably not

477

maintained over longer periods.

478

Our results differ from data obtained for CSE exposure of bronchial epithelial cells: One

479

study observed mitochondrial fragmentation in primary human bronchial epithelial cells

480

treated with CSE for 48h. Mitochondrial fragmentation was associated with increased

481

mitochondrial ROS production and localization of the fission protein DRP1 to

482

mitochondria. This study did not observe any changes for MFN2 or other mitochondrial

483

fusion proteins.(18) Another study, however described elongated and branched

484

mitochondria in long term (6 months) cigarette smoke exposure of bronchial epithelial

485

cells and in primary bronchial epithelial cells isolated from COPD patients.(19) We

486

analyzed mitochondrial morphology in 16HBE cells, a human bronchial epithelial cell

487

line, and did not detect alterations in mitochondrial fusion and fission dynamics in

488

control vs. CSE treated cells (data not shown). Thus, we conclude that there is a strong

24

489

dependency on cell type for triggering an adaptive mitochondrial hyperfusion response

490

of lung epithelial cells to cigarette smoke extract.

491

Functional consequences of mitochondrial elongation

492

Mitochondrial hyperfusion was reported to be connected with an increased efficiency of

493

ATP production.(15, 40) Accordingly, we observed that hyperfused mitochondria in

494

MLE12 cells were associated with enhanced metabolic activity, increased cellular ATP

495

levels,

496

consumption rates. These observations are well in agreement with the concept of stress

497

induced mitochondrial hyperfusion as a protective response to increase the cell’s ability

498

to repair damage. This was previously shown for different stressors such as starvation,

499

UV, actinomycin, or cycloheximide treatment. (15, 40) As mitochondrial hyperfusion can

500

also be induced by oxidative stress (38, 48), it seems plausible that this survival

501

response is also part of the oxidative stress response to cigarette smoke extract.

502

We observed mitochondrial hyperfusion in the ATI-like MLE12 alveolar epithelial cells

503

and in primary ATII alveolar epithelial cells. Although MLE12 and pmATII cells probably

504

differ in their metabolic phenotype we speculate that the mitochondrial hyperfusion

505

observed in pmATII cells also affects mitochondrial metabolism in these cells. Increased

506

respiration rates have previously been described for primary ATII cells of smoke-

507

exposed mice but depended on the available substrates: Agarwal et al. observed

508

significantly increased mitochondrial activity and oxygen consumption when applying

509

pyruvate or palmitate as a substrate, while respiration decreased when using

510

glucose.(1, 2) Thus, although we didn’t measure mitochondrial activity in pmATII cells

511

ourselves it is well feasible that mitochondrial hyperfusion results in increased oxygen

augmented

mitochondrial

membrane

potential,

and

elevated

oxygen

25

512

consumption in CS exposed pmATII when respiring on pyruvate. Similar to our results,

513

Hoffmann et al. showed increased ATP levels in CSE treated bronchial epithelial cells

514

with elongated mitochondria (19), further supporting the notion of increased

515

mitochondrial respiratory chain activity in response to cigarette smoke. In summary,

516

cigarette-smoke induced mitochondrial hyperfusion might serve as an adaptive

517

response to increase mitochondrial ATP production in order to cope with CSE induced

518

cell damage. A hyperfused mitochondrial network, however, may render the cell more

519

vulnerable to additional stress as it attenuates mitochondrial quality control.(14)

520

Moreover, because elongated mitochondria are spared from removal by autophagy

521

(16), sustained mitochondrial hyperfusion i.e. due to continuous and chronic exposure

522

to stress might further increase susceptibility upon additional stress of the mitochondrial

523

network. This mechanism was suggested to account for the occurrence of hyperfused

524

dysfunctional mitochondria in aging muscles.(28) Of note, sustained mitochondrial

525

elongation has also been associated with increased cellular senescence.(24) Vice

526

versa, induction of senescence is accompanied by mitochondrial elongation in

527

senescent cells.(48) Alveolar epithelial cell senescence is known to be induced by

528

cigarette smoke treatment (42) and senescence of pulmonary cells has been proposed

529

as a pathogenetic mechanisms for progression of COPD.(32, 43) Furthermore,

530

mitochondrial hyperfusion was recently shown to be accompanied by NF-κb

531

activation.(51) It is thus tempting to speculate that cigarette smoke extract induced

532

mitochondrial hyperfusion - although being part of a beneficial adaptive stress response

533

in the first place - may contribute to age-related COPD pathogenesis via promoting

26

534

diminished mitochondrial quality control, impaired cellular stress resistance, and cellular

535

senescence.

536

Acknowledgement

537

We are very grateful to Otmar Schmid, Fabiana Perocchi, Jennifer Wettmarshausen,

538

Konstanze Winklhofer, and Anne Kathrin Müller-Rischart for their support and helpful

539

advice during the preparation of the manuscript. The authors would also like to thank

540

Christina Lukas, Julia Kipp, and Anastasia van den Berg for excellent technical

541

assistance.

542

Disclosure

543

The authors declare that they have no conflicts of interests.

27

544

References

545 546 547

1.

Agarwal AR, Yin F, Cadenas E. METABOLIC SHIFT IN LUNG ALVEOLAR CELL MITOCHONDRIA FOLLOWING ACROLEIN EXPOSURE. AJP Lung Cell. Mol. Physiol. (September 20, 2013). doi: 10.1152/ajplung.00165.2013.

548 549 550

2.

Agarwal AR, Yin F, Cadenas E. Short-term cigarette smoke exposure leads to metabolic alterations in lung alveolar cells. Am. J. Respir. Cell Mol. Biol. (March 13, 2014). doi: 10.1165/rcmb.2013-0523OC.

551 552 553 554

3.

Aravamudan B, Kiel A, Freeman M, Delmotte PF, Thompson MA, Vassallo R, Sieck GC, Pabelick CM, Prakash YS. Cigarette Smoke-induced Mitochondrial Fragmentation and Dysfunction in Human Airway Smooth Muscle. AJP Lung Cell. Mol. Physiol. (March 7, 2014). doi: 10.1152/ajplung.00155.2013.

555 556

4.

Barnes PJ. Chronic Obstructive Pulmonary Disease. N Engl J Med 343: 269–280, 2000.

557 558

5.

Blackstone C, Chang C-R. Mitochondria unite to survive. Nat Cell Biol 13: 521– 522, 2011.

559 560

6.

Bouchecareilh M, Balch WE. Proteostasis: A New Therapeutic Paradigm for Pulmonary Disease. Proc Am Thorac Soc 8: 189–195, 2011.

561 562 563

7.

Bowler RP, Barnes PJ, Crapo JD. The Role of Oxidative Stress in Chronic Obstructive Pulmonary Disease. COPD J Chronic Obstr Pulm Dis 1: 255–277, 2004.

564 565 566

8.

Cagalinec M, Safiulina D, Liiv M, Liiv J, Choubey V, Wareski P, Veksler V, Kaasik A. Principles of the mitochondrial fusion and fission cycle in neurons. J. Cell Sci. (March 22, 2013). doi: 10.1242/jcs.118844.

567 568 569

9.

Chauhan A, Vera J, Wolkenhauer O. The systems biology of mitochondrial fission and fusion and implications for disease and aging. Biogerontology (October 12, 2013). doi: 10.1007/s10522-013-9474-z.

570 571

10. Corti M, Brody AR, Harrison JH. Isolation and primary culture of murine alveolar type II cells. Am J Respir Cell Mol Biol 14: 309–315, 1996.

572 573 574 575

11. Das KC. Hyperoxia Decreases Glycolytic Capacity, Glycolytic Reserve and Oxidative Phosphorylation in MLE-12 Cells and Inhibits Complex I and II Function, but Not Complex IV in Isolated Mouse Lung Mitochondria. PLoS ONE 8: e73358, 2013.

576 577

12. Duvezin-Caubet S, Jagasia R, Wagener J, Hofmann S, Trifunovic A, Hansson A, Chomyn A, Bauer MF, Attardi G, Larsson N-G, Neupert W, Reichert AS.

28

578 579

Proteolytic Processing of OPA1 Links Mitochondrial Dysfunction to Alterations in Mitochondrial Morphology. J Biol Chem 281: 37972–37979, 2006.

580 581 582

13. Elgass K, Pakay J, Ryan MT, Palmer CS. Recent advances into the understanding of mitochondrial fission. Biochim. Biophys. Acta BBA - Mol. Cell Res. (May 2012). doi: 10.1016/j.bbamcr.2012.05.002.

583 584 585

14. Figge MT, Osiewacz HD, Reichert AS. Quality control of mitochondria during aging: Is there a good and a bad side of mitochondrial dynamics? BioEssays (January 29, 2013). doi: 10.1002/bies.201200125.

586 587 588

15. Gomes LC, Benedetto GD, Scorrano L. During autophagy mitochondria elongate, are spared from degradation and sustain cell viability. Nat Cell Biol 13: 589–598, 2011.

589 590

16. Gomes LC, Scorrano L. Mitochondrial morphology in mitophagy and macroautophagy. Biochim Biophys Acta BBA - Mol Cell Res 1833: 205–212, 2013.

591 592 593 594 595

17. Granados-Castro LF, Rodríguez-Rangel DS, Montaño M, Ramos C, PedrazaChaverri J. Wood smoke exposure induces a decrease in respiration parameters and in the activity of respiratory complexes I and IV in lung mitochondria from guinea pigs: Wood Smoke Exposure Induces a Decrease in Respiration Parameters. Environ. Toxicol. (November 2013). doi: 10.1002/tox.21922.

596 597 598 599 600

18. Hara H, Araya J, Ito S, Kobayashi K, Takasaka N, Yoshii Y, Wakui H, Kojima J, Shimizu K, Numata T, Kawaishi M, Kamiya N, Odaka M, Morikawa T, Kaneko Y, Nakayama K, Kuwano K. Mitochondrial fragmentation in cigarette smoke induced-bronchial epithelial cell senescence. AJP Lung Cell. Mol. Physiol. (September 20, 2013). doi: 10.1152/ajplung.00146.2013.

601 602 603 604 605

19. Hoffmann RF, Zarrintan S, Brandenburg SM, Kol A, de Bruin HG, Jafari S, Dijk F, Kalicharan D, Kelders M, Gosker HR, ten Hacken NH, van der Want JJ, van Oosterhout AJ, Heijink IH. Prolonged cigarette smoke exposure alters mitochondrial structure and function in airway epithelial cells. Respir Res 14: 97, 2013.

606 607

20. Jin SM, Youle RJ. PINK1- and Parkin-mediated mitophagy at a glance. J Cell Sci 125: 795–799, 2012.

608 609 610

21. Jovaisaite V, Mouchiroud L, Auwerx J. The mitochondrial unfolded protein response, a conserved stress response pathway with implications in health and disease. J Exp Biol 217: 137–143, 2013.

611 612 613

22. Kelsen SG, Duan X, Ji R, Perez O, Liu C, Merali S. Cigarette Smoke Induces an Unfolded Protein Response in the Human Lung: A Proteomic Approach. Am J Respir Cell Mol Biol 38: 541–550, 2008.

29

614 615 616 617 618

23. Königshoff M, Kramer M, Balsara N, Wilhelm J, Amarie OV, Jahn A, Rose F, Fink L, Seeger W, Schaefer L, Günther A, Eickelberg O. WNT1-inducible signaling protein–1 mediates pulmonary fibrosis in mice and is upregulated in humans with idiopathic pulmonary fibrosis. J. Clin. Invest. (March 16, 2009). doi: 10.1172/JCI33950.

619 620 621

24. Lee S, Jeong S-Y, Lim W-C, Kim S, Park Y-Y, Sun X, Youle RJ, Cho H. Mitochondrial Fission and Fusion Mediators, hFis1 and OPA1, Modulate Cellular Senescence. J Biol Chem 282: 22977–22983, 2007.

622 623

25. Liesa M, Palacin M, Zorzano A. Mitochondrial Dynamics in Mammalian Health and Disease. Physiol Rev 89: 799–845, 2009.

624 625

26. Liesa M, Shirihai OS. Mitochondrial Dynamics in the Regulation of Nutrient Utilization and Energy Expenditure. Cell Metab 17: 491–506, 2013.

626 627 628 629 630

27. Lutz AK, Exner N, Fett ME, Schlehe JS, Kloos K, Lammermann K, Brunner B, Kurz-Drexler A, Vogel F, Reichert AS, Bouman L, Vogt-Weisenhorn D, Wurst W, Tatzelt J, Haass C, Winklhofer KF. Loss of Parkin or PINK1 Function Increases Drp1-dependent Mitochondrial Fragmentation. J Biol Chem 284: 22938– 22951, 2009.

631 632 633

28. Marzetti E, Calvani R, Cesari M, Buford TW, Lorenzi M, Behnke BJ, Leeuwenburgh C. Mitochondrial dysfunction and sarcopenia of aging: From signaling pathways to clinical trials. Int J Biochem Cell Biol 45: 2288–2301, 2013.

634 635

29. Meiners S, Eickelberg O. What shall we do with the damaged proteins in lung disease? Ask the proteasome! Eur Respir J 40: 1260–1268, 2012.

636 637 638 639

30. Mizumura K, Cloonan SM, Nakahira K, Bhashyam AR, Cervo M, Kitada T, Glass K, Owen CA, Mahmood A, Washko GR, Hashimoto S, Ryter SW, Choi AMK. Mitophagy-dependent necroptosis contributes to the pathogenesis of COPD. J. Clin. Invest. (August 1, 2014). doi: 10.1172/JCI74985.

640 641 642 643

31. Mukhopadhyay P, Rajesh M, Haskó G, Hawkins BJ, Madesh M, Pacher P. Simultaneous detection of apoptosis and mitochondrial superoxide production in live cells by flow cytometry and confocal microscopy. Nat Protoc 2: 2295–2301, 2007.

644 645 646 647

32. Muller K-C, Welker L, Paasch K, Feindt B, Erpenbeck VJ, Hohlfeld JM, Krug N, Nakashima M, Branscheid D, Magnussen H, Jorres RA, Holz O. Lung fibroblasts from patients with emphysema show markers of senescence in vitro. Respir Res 7: 32, 2006.

648 649 650

33. Nyunoya T, Mebratu Y, Contreras A, Delgado M, Chand HS, Tesfaigzi Y. Molecular Processes that Drive Cigarette Smoke–Induced Epithelial Cell Fate of the Lung. Am J Respir Cell Mol Biol 50: 471–482, 2014. 30

651 652

34. Pellegrino MW, Nargund AM, Haynes CM. Signaling the mitochondrial unfolded protein response. Biochim Biophys Acta BBA - Mol Cell Res , [date unknown].

653 654 655

35. Van Rijt SH, Keller IE, John G, Kohse K, Yildirim AO, Eickelberg O, Meiners S. Acute cigarette smoke exposure impairs proteasome function in the lung. Am. J. Physiol. - Lung Cell. Mol. Physiol. .

656 657 658 659 660

36. Ryan JJ, Marsboom G, Fang Y-H, Toth PT, Morrow E, Luo N, Piao L, Hong Z, Ericson K, Zhang HJ, Han M, Haney CR, Chen C-T, Sharp WW, Archer SL. PGC1 -Mediated Mitofusin-2 Deficiency in Female Rats and Humans With Pulmonary Arterial Hypertension. Am. J. Respir. Crit. Care Med. (February 28, 2013). doi: 10.1164/rccm.201209-1687OC.

661 662

37. Sena LA, Chandel NS. Physiological Roles of Mitochondrial Reactive Oxygen Species. Mol Cell 48: 158–167, 2012.

663 664

38. Shutt T, Geoffrion M, Milne R, McBride HM. The intracellular redox state is a core determinant of mitochondrial fusion. EMBO Rep 13: 909–915, 2012.

665 666 667

39. Tanaka A, Cleland MM, Xu S, Narendra DP, Suen D-F, Karbowski M, Youle RJ. Proteasome and p97 mediate mitophagy and degradation of mitofusins induced by Parkin. J Cell Biol 191: 1367–1380, 2010.

668 669 670 671 672

40. Tondera D, Grandemange S, Jourdain A, Karbowski M, Mattenberger Y, Herzig S, Da Cruz S, Clerc P, Raschke I, Merkwirth C, Ehses S, Krause F, Chan DC, Alexander C, Bauer C, Youle R, Langer T, Martinou J-C. SLP-2 is required for stress-induced mitochondrial hyperfusion. EMBO J 28: 1589–1600, 2009.

673 674 675 676

41. Van der Toorn M, Slebos D-J, de Bruin HG, Leuvenink HG, Bakker SJL, Gans ROB, Koeter GH, van Oosterhout AJM, Kauffman HF. Cigarette smoke-induced blockade of the mitochondrial respiratory chain switches lung epithelial cell apoptosis into necrosis. AJP Lung Cell Mol Physiol 292: L1211–L1218, 2007.

677 678

42. Tsuji T, Aoshiba K, Nagai A. Cigarette Smoke Induces Senescence in Alveolar Epithelial Cells. Am J Respir Cell Mol Biol 31: 643–649, 2004.

679 680

43. Tsuji T, Aoshiba K, Nagai A. Alveolar Cell Senescence in Patients with Pulmonary Emphysema. Am J Respir Crit Care Med 174: 886–893, 2006.

681 682

44. Tuder RM, Petrache I. Pathogenesis of chronic obstructive pulmonary disease. J Clin Invest 122: 2749–2755, 2012.

683 684 685 686

45. Twig G, Elorza A, Molina AJA, Mohamed H, Wikstrom JD, Walzer G, Stiles L, Haigh SE, Katz S, Las G, Alroy J, Wu M, Py BF, Yuan J, Deeney JT, Corkey BE, Shirihai OS. Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. EMBO J 27: 433–446, 2008. 31

687 688 689

46. Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G. Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol 11: 700–714, 2010.

690 691 692

47. Wei J, Rahman S, Ayaub EA, Dickhout JG, Ask K. Protein Misfolding and Endoplasmic Reticulum Stress in Chronic Lung Disease. CHEST J 143: 1098, 2013.

693 694 695 696

48. Yoon Y-S, Yoon D-S, Lim IK, Yoon S-H, Chung H-Y, Rojo M, Malka F, Jou M-J, Martinou J-C, Yoon G. Formation of elongated giant mitochondria in DFO-induced cellular senescence: Involvement of enhanced fusion process through modulation of Fis1. J Cell Physiol 209: 468–480, 2006.

697 698

49. Youle RJ, van der Bliek AM. Mitochondrial Fission, Fusion, and Stress. Science 337: 1062–1065, 2012.

699 700

50. Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol 12: 9– 14, 2011.

701 702 703

51. Zemirli N, Pourcelot M, Ambroise G, Hatchi E, Vazquez A, Arnoult D. Mitochondrial hyperfusion promotes nf-κb activation via the mitochondrial e3 ligase mulan. FEBS J. (May 2014). doi: 10.1111/febs.12846.

704 705

52. Zhao Q. A mitochondrial specific stress response in mammalian cells. EMBO J 21: 4411–4419, 2002.

706 707

32

708

Figure Legends

709

Figure 1: Low-doses of CSE are nontoxic in MLE12 cells.

710

(A)

711

dose CSE for the indicated times. Each band corresponds to an independent

712

experiment. Positive control: 24h 75% CSE and 6h 75% CSE treated MLE12 cells.

713

(B)

Western blot analysis of Caspase 3 cleavage in MLE12 cells treated with low

LDH assay of CSE treated MLE12 cells after 6, 24, and 48h. n=3 + SEM

714 715

Figure 2: Mitochondria of MLE12 alveolar epithelial cells hyperfuse in response to CSE

716

treatment.

717

(A)

718

medium, 25% CSE, or 10 µM CCCP for 48h. Scale bars: 10 µm

719

(B)

720

of treatment. n=3 + SEM; Differences between the groups were analyzed using the

721

chi2-test: ***:p

Cigarette smoke extract affects mitochondrial function in alveolar epithelial cells.

Cigarette smoke is the main risk factor for chronic obstructive pulmonary disease (COPD). Exposure of cells to cigarette smoke induces an initial adap...
2MB Sizes 2 Downloads 9 Views