Articles in PresS. Am J Physiol Lung Cell Mol Physiol (January 30, 2015). doi:10.1152/ajplung.00309.2014

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45

Pulmonary epithelial barrier function- some new players and mechanisms Kieran Brune1, James Frank2, Andreas Schwingshackl3, James Finigan4, Venkataramana K. Sidhaye1 1

Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore MD, USA, 2The

Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco VA Medical Center, and NCIRE/Veterans Health Research Institute, San Francisco, California USA, 3 Department of Pediatrics, University of Tennessee Health Science Center, 4 Division of Oncology, Cancer Center, National Jewish Health

Correspondence: Venkataramana K. Sidhaye, MD Department of Medicine, Division of Pulmonary and Critical Care Medicine Johns Hopkins Asthma and Allergy Center 4B.64 5501 Hopkins Bayview Circle Baltimore, MD 21224 Tel.: 410-550-4893 Fax: 410-550-2612 E-mail: [email protected]

Running Head: New players modulating the epithelial barrier Abstract: The pulmonary epithelium serves as a barrier to prevent access of the inspired luminal contents to the sub epithelium. In addition, the epithelium dictates the lung’s initial responses to both infectious and non-infectious stimuli. One mechanism that the epithelium does this is by coordinating transport of diffusible molecules across the epithelial barrier, both through the cell and between cells. In this review we will discuss a few emerging paradigms of permeability changes through altered ion transport and paracellular regulation by which the epithelium gates its response to potentially detrimental luminal stimuli. This review is a summary of talks presented during a symposium in Experimental Biology geared towards novel and less recognized methods of epithelial barrier regulation. First, we will discuss mechanisms of dynamic regulation of cell-cell contacts in the context of repetitive exposure to inhaled infectious and non-infectious insults. In the second section, we will briefly discuss mechanisms of transcellular ion homeostasis specifically focused on the role of claudins and paracellular ion channel regulation in chronic barrier dysfunction. In the next section, we will address transcellular ion transport and highlight the role of Trek-1 in epithelial responses to lung injury. In the final section, we will outline the role of epithelial growth receptor (EGFR) in barrier regulation in baseline, acute lung injury and airway disease. We will then end with a summary of mechanisms of epithelial control as well as discuss emerging paradigms of the epithelium role in shifting between a structural element that maintains tight cell-cell adhesion to a cell that initiates and participates in immune responses.

Copyright © 2015 by the American Physiological Society.

46 47

Keywords: Lung, epithelial barrier, permeability, transport, ions Introduction

48

The lung epithelium is a mucosal surface composed of ciliated cells, mucous producing

49

cells, and undifferentiated basal cells that forms the interface between the lumen and

50

the parenchyma from the nasal passage to the alveoli. It is the initial site of contact for

51

all inspired substances and therefore acts as both a physical and an immunological

52

barrier that protects the sub-epithelial tissue.

53

The physical barrier, comprised of the mucocilary apparatus, secreted antimicrobial

54

substances, and the intercellular junctions, prevents inhaled toxins and pathogens from

55

contacting the sub-epithelial tissue. The mucociliary apparatus traps foreign particles

56

and removes them from the respiratory tract. Defects in this apparatus can lead to

57

recurrent infections as seen in patients with cystic fibrosis(104), COPD(14, 79), and

58

ciliary dyskinesis(18, 19). Secreted antimicrobial substances including enzymes,

59

protease inhibitors, and antimicrobial peptides further contribute to the epithelial cell’s

60

immune response(134). Epithelial secretions contain enzymes such as lysozyme that

61

have antimicrobial effects against both gram positive and gram negative bacteria(40,

62

76) as well as protease inhibitors including serine protease inhibitor, secretory leukocyte

63

protease inhibitor, and elafin which reduce the effects of proteases that are produced by

64

pathogens. In addition, surface antimicrobial peptides like human beta defensins

65

further protect against numerous pathogens including bacteria and viruses(116, 176).

66

The intercellular junctions form adhesive forces that connect neighboring cells and

67

separate the external environment from the sub-epithelial tissue. There are three main

68

types of intercellular junctions: the tight junctions, the adherens junctions, and the

69

desmosomes(137). The tight junctions are located more apically when compared to the

70

other junctions and are multi-protein complexes that consist of transmembrane proteins

71

including claudins, occludins, and JAMs as well as scaffolding proteins like the zona

72

occludens. These junctions form a continuous circumferential ring around the epithelial

73

cells which regulates the paracellular transport of ions and certain molecules(63, 157).

74

The adherens junctions, composed of E-cadherin and several proteins from the catenin

75

family including alpha catenin, beta catenin, and p120 anchor the actin cytoskeleton and

76

mediate cell to cell adhesion(41, 63, 78). Together, the tight and adherens junctions

77

form the apicoadherens junctions which separate the apical and basolateral membranes

78

and block the movement of integral membrane proteins between surfaces to maintain

79

epithelial cell polarity(169). The desmosomes, adhesive junctions located on the lateral

80

cell membrane, link to the intermediate filaments and help protect against mechanical

81

stress. Inhaled toxins such as cigarette smoke damage these intercellular junctions

82

resulting in a leaky, hyper-proliferative, and abnormally differentiated epithelium(22, 68,

83

73, 130, 150, 191).

84

It is increasingly recognized that the epithelium helps to maintain homeostasis in the

85

lung and disruption of the epithelial barrier can lead the development of inflammation

86

and diseases such as COPD and acute lung injury (ALI). Moreover, novel studies

87

demonstrating that restoration of the epithelial barrier through transfer of mitochondria

88

from bone marrow derived stromal cells to epithelial cells can protect against ALI further

89

establishes the importance of the epithelial barrier (77). In this review we will further

90

discuss how the airway epithelium dynamically regulates its response to luminal stimuli

91

by altering ion transport and paracellular permeability

92

Section I: The lung epithelium: insights into the role of regulation of paracellular

93

permeability in epithelial signaling

94

The role of cell-cell adhesion in modulating epithelial signaling pathways

95

The integrity of the physical barrier and epithelial cell polarity affect the interaction of

96

cytokines and growth factors with their receptors thereby modulating the epithelium’s

97

immunological response to environmental stimuli. In this respect, the epithelium also

98

acts as an immunological barrier. Subtle changes in paracellular permeability can

99

impact ligand interaction with its receptor. Moreover, certain cytokines are prevented

100

from interacting with their cytokine receptors due to segregation to different sides of the

101

apicoadherens junctional complex. Damage to the epithelium can affect the polarized

102

distribution of cell surface receptors allowing for increased receptor-ligand interaction

103

and the activation of signaling transduction cascades and secretory activity(73, 171,

104

200). One specific example of such regulation is the epidermal growth factor (EGF),

105

which regulates migration, proliferation, and differentiation following injury and

106

enhances epithelial repair(186). In normal human airways, epidermal growth factor

107

receptor (EGFR) expression is limited and isolated to the basolateral membrane (187)

108

however, expression is increased in response to cigarette smoke(165, 188), mechanical

109

stress(139), and inhaled noxious substances like bleomycin(110). In a homeostatic

110

state, the EGF ligand is located on the apical surface and therefore separated from its

111

receptor on the basolateral surface. Moreover, the apically located growth factor

112

heregulin is separated from its receptor erbB2/3 (also known as HER2/3) which is

113

located on the basolateral membrane. These growth factors therefore only have access

114

to their receptors when the epithelium is damaged allowing for rapid targeted repair of

115

the epithelial injury(200). The binding of heregulin to the erbB2/3 receptor leads to cell

116

proliferation, differentiation, and mucous secretion(193, 201, 209) which allows the

117

damaged epithelium to protect and repair itself. Therefore although the epithelium

118

expresses both the growth factors and their receptors, it can regulate the interaction

119

such that the effects occur only at times of injury, and one mechanism by which the

120

epithelium does this is by regulating paracellular permeability.

121

Dynamic regulation of paracellular permeability

122

The intercellular junctions do not statically resist entry to all substances but act as

123

dynamic structures that can open or close in response to both physiological and

124

pathological stimuli(157, 166). By tightening cell-cell adhesion in response to

125

mechanical stimuli such as shear stress or chemical environmental exposures such as

126

inhaled particulate matter, the epithelia dynamically regulates receptor-ligand

127

interactions (Fig. 1)(171). For example, airflow over the epithelium during breathing

128

creates shear stress which can lead to changes in barrier properties. Low,

129

physiological levels of shear stress are generated during normal tidal volume breathing

130

but can increase by several fold during exercise, coughing, and bronchospasm.

131

Exposure to shear stress leads to actin rearrangement and barrier enhancement(173).

132

In addition, in response to both physiological levels of shear stress as well as pathologic

133

stimuli like particulate matter- pollution formed from the mixture of solid particles and

134

liquid droplets found in air- there is increased membrane localization of septin 2 that

135

decreases paracellular permeability by altering cortical actin rearrangement(171).

136

Septin 2 is a member of the family of GTP binding proteins that function in cytoskeletal

137

arrangement and cell polarization. Membrane localization of septin 2 allows for

138

increased interaction with actin which regulates the rearrangement of cortical actin and

139

enhances barrier function. Physiological levels of shear stress also lead to a selective

140

decrease in the abundance of the apical water channel aquaporin 5(172, 173), which

141

helps to modulate the paracellular permeability in response to the stress. Aquaporin 5

142

stabilizes the microtubules(172) and antagonizes cell to cell adhesion(173) thereby

143

affecting paracellular permeability. Thus, the epithelium is able to dynamically regulate

144

its response to experienced luminal stimuli.

145

In summary, the airway epithelium is the first line of defense and prevents inspired

146

substances from contacting the sub-epithelial tissue. The epithelium acts as both a

147

physical and immunological barrier that continuously responds to both physiologic and

148

pathologic stimuli in the environment and dynamically regulates its barrier in response

149

to these stimuli. The apicoadherens junctions separate the apical and basolateral

150

membranes and therefore membrane integrity can affect cell signaling and the defenses

151

mounted in response to environmental exposures. By tightly coordinating epithelial

152

responses to environmental exposures and altering epithelial signaling, secreted

153

products, and ion and water flux in response to stimuli, the epithelium is able to provide

154

a dynamic barrier at the interface between the lung parenchyma and the luminal

155

environment. Noxious stimuli such as chronic tobacco exposure, infections, or

156

particulate matter and air pollution, can lead to a breakdown in this regulatory response

157

leading to chronic epithelial activation which can further propagate the damage resulting

158

in a leaky barrier. In the following sections, we will discuss in more detail how the tight

159

junction proteins, ion transport, and EGFR contribute to the dynamic epithelial barrier.

160

161

Section II: The lung epithelium: insights in ion transport across the barrier

162

Interactions between tight junctions and ion transporters to maintain lung fluid

163

balance

164

Control of airspace fluid volume and composition in the alveolus is primarily determined

165

by transcellular ion transport, which requires a polarized epithelium with intact tight

166

junctions. However, paracellular ion, macromolecule, and water transport through intact

167

tight junctions also makes a significant contribution to lung fluid balance (115, 199).

168

Furthermore, the effects of both bacterial and viral infections are quite complex, with

169

data suggesting that a multitude of ion channels are altered in response to infection,

170

and there is potential creation of new channels by a host of viruses, termed viroporins.

171

The detailed discussions of these effects are beyond the scope of this review. During

172

the resolution of pulmonary edema, clearance of fluid from the airspaces is largely

173

dependent on a transepithelial sodium concentration gradient driven by Na+/K+-ATPase.

174

Apical sodium conductance through ENaC is central to fluid clearance, but chloride and

175

potassium channels also make important contributions (13, 37, 98). Apical-to-

176

basolateral chloride transport may be particularly important because the maximal rate of

177

sodium and water transport from the airspaces appears to be limited by the concomitant

178

transport of chloride in many studies (9, 43, 122). Because experimental data suggest

179

that a sizable fraction of transepithelial chloride transport occurs through the

180

paracellular route in the alveolar epithelium (87), regulated paracellular ion transport

181

could influence alveolar fluid clearance rates by this mechanism. In other words,

182

changes in the selectivity and magnitude of paracellular ion conductance may influence

183

rates of alveolar fluid clearance, either by limiting sodium leak or increasing chloride

184

currents. The mechanisms for the regulation of paracellular transport in the lung and the

185

potential interrelationship with transcellular transport is not fully understood, but recent

186

work has begun to describe some of the major determinants of paracellular

187

permselectivity in the alveolar epithelium. These studies have also provided some

188

insight into deeper associations between the paracellular and transcellular routes of

189

transport that is the regulation of transcellular transporters by cell junction proteins. One

190

area of interest is the potential association between Na+/K+-ATPase and tight junction

191

claudins.

192

Sodium/Potassium ATPase as a regulator of cell junctions

193

Sodium/potassium ATPase is made up of catalytic (α) and regulatory (β) subunits.

194

Additional regulatory subunits of the FXYD family, such as the γ subunit of Na+/K+-

195

ATPase, may also associate with α/β heterodimers and affect function. Pump activity in

196

polarized epithelial cells is essential for lung fluid balance and increased pump activity is

197

associated with increased rates of vectorial fluid clearance across the alveolar

198

epithelium (4, 107). Recent work has uncovered an additional role for Na+/K+-ATPase in

199

tight junction formation and maintenance in epithelia (194). For example, the β subunit

200

of Na+/K+-ATPase associates with PDZ domain-containing proteins and localizes to tight

201

junctions (109, 141). Although the β1 subunit has been reported to function as a cell

202

adhesion molecule, enzymatic pump activity of Na+/K+-ATPase is required for tight

203

junction formation in MDCK cells (144). The mechanism for this function appears to be

204

through Na+/K+-ATPase-mediated activation of RhoA signaling and cytoskeletal

205

remodeling at the periphery of epithelial cells. Na+/K+-ATPase may also be required for

206

the maintenance of tight junctions. In monolayers of retinal pigment epithelial cells and

207

in pancreatic ductal cell (HPAF-II) monolayers with established tight junctions, inhibition

208

of Na+/K+-ATPase with ouabain increased paracellular permeability to ions and

209

macromolecules (142, 143). However, the junction promoting effects of Na+/K+-ATPase

210

may be context or cell-type specific because ouabain has also been reported to

211

increase expression of claudins-4 and -1 in MDCK cells in association with decreased

212

paracellular macromolecule permeability (97). Although the mechanisms for the

213

regulation of tight junction formation and function by Na+/K+-ATPase are not fully

214

understood, it appears that Na+/K+-ATPase can modulate tight junction assembly and

215

function.

216

Claudins as regulators of paracellular permeability

217

Differential expression of the claudin family of tight junction proteins is a fundamental

218

regulatory mechanism of paracellular permselectivity. Claudins are transmembrane

219

proteins that are necessary and sufficient for tight junction strand formation (51). The

220

spatially restricted expression of specific claudins along the course of the nephron

221

exemplifies their permselectivity function (7). Moreover, point mutations in certain

222

claudins result in clinically important changes in ion transport, and substantial

223

experimental evidence supports a direct role for claudins in charge-selective

224

paracellular ion conductance (6, 102, 195). Claudins on adjacent cells form paracellular

225

pores with an approximately 3 Angstrom diameter that function as relatively high

226

conductance, un-gated ion channels (101, 145, 197). Claudins also influence

227

paracellular macromolecule transport and individual claudins appear to convey different

228

properties to this charge-independent, lower conductance pathway (167, 168). A

229

leading hypothesis is that individual claudin strands within a tight junction establish the

230

ion pore pathway while the persistence or continuity of claudin strands within the

231

membrane influences the macromolecule leak pathway (167). Because tight junctions

232

consist of multiple layers of claudin-based strands that undergo continuous turnover,

233

both conductance pathways can exist simultaneously. Three claudins, -3, -4, and -18.1,

234

are abundantly expressed in the alveolar epithelium, although other claudins are

235

expressed at low levels (95).

236

Lung-specific claudin-18.1 is a requirement for the alveolar epithelial permeability

237

barrier

238

Claudin-18.1 is of particular interest because it is the only known lung-specific tight

239

junction protein and it is the most abundantly expressed claudin in type 1 alveolar

240

epithelial cells (95). The role of claudin-18 in alveolar epithelial barrier function has

241

recently been examined in knockout mice. Claudin-18 does not appear to be required

242

for prenatal lung development; however, upon conversion to air breathing,

243

alveolarization is abnormal in claudin-18 knockout mice(96). Claudin-18 null mice have

244

a 3-fold increase in alveolar epithelial permeability to macromolecules. There is a similar

245

increase in macromolecule permeability in cultured primary alveolar epithelial cells from

246

knockout mice and with siRNA knock down of claudin-18 in wild type cells(96). These

247

data indicate that claudin-18 provides a macromolecule permeability barrier in the

248

alveolar epithelium and suggest that the loss of this function results in abnormal alveolar

249

homeostasis and impaired alveolarization. Interestingly, although macromolecule

250

permeability is increased, claudin-18 null mice do not develop pulmonary edema(100).

251

The reason for this may be that alveolar epithelial sodium and chloride transport is

252

increased. The basal alveolar fluid clearance rate in claudin-18 null mice is

253

approximately twice that of wild type littermates. Examination of the expression levels of

254

ion transporters in whole lung revealed a 2-fold increase in the β1 subunit of Na+/K+-

255

ATPase and increased pump activity, but no change or a slight decrease in the α, β

256

and γ subunits of ENaC at the mRNA level. Because over-expression of Na+/K+-ATPase

257

results in higher rates of alveolar fluid transport in animal models, the increase in β1

258

subunit expression may account for most of the increase in alveolar fluid clearance in

259

claudin-18 null mice.

260

Chloride transport and alveolar fluid clearance in claudin-18 null mice

261

As mentioned above, sodium transport in the lung can be limited by the co-transport of

262

chloride, so increases in the levels of chloride transporters might be expected to

263

facilitate higher rates of sodium and fluid transport. It is notable that CFTR mRNA

264

expression is increased by approximately 4-fold in claudin-18 knockout mice, but

265

whether CFTR inhibition decreases alveolar fluid clearance in this context has not been

266

reported. In addition to CFTR, several other regulators of chloride transport show

267

increased expression in the lungs of claudin-18 null mice, including NKCC1, CLC2, and

268

SLC26A9. It is possible that these data point to a larger role for chloride transport in

269

alveolar fluid clearance than has been previously appreciated. For example, chloride-

270

dependent sodium transport into the airspaces appears to be an important mechanism

271

for edema formation during hydrostatic stress (183). In the nephron, chloride-dependent

272

sodium absorption is a mechanism for increased fluid retention and hypertension. This

273

mechanism depends on the coupling of sodium transport to chloride transport via

274

NKCC1. Although NKCC1 transport in a basolateral-to-apical direction is well

275

established, transport in an apical-to-basolateral direction (or bi-directional transport) is

276

possible (183). Therefore, the observed increase in chloride transporters in claudin-18

277

null mice is consistent with compensation of fluid clearance facilitated by maximizing

278

chloride-limited sodium transport. Further investigation is required, but increased

279

NKCC1 expression in claudin-18 null mice might also support the hypothesis that

280

apical-to-basolateral chloride-dependent sodium transport is a mechanism for alveolar

281

fluid clearance. In summary, the loss of claudin-18 results in increased paracellular

282

permeability in the absence of increased lung water. Higher rates of fluid transport in

283

these mice may be due to increased expression of the beta subunit of Na+/K+-ATPase

284

along with chloride transporters. The mechanisms for the up-regulation of ion

285

transporters in the context of claudin-18 deficiency are not known, but one possibility is

286

co-regulation of cell junction constituents and ion transporters.

287

Functional implications of increased claudin-4 expression in claudin-18 null mice

288

Claudin-4 mRNA and protein levels are increased by 4-fold in claudin-18 knockout mice.

289

Claudin-4 plays a distinct role in trans-epithelial ion transport and junction formation and

290

may partly compensate for the loss of claudin-18. Claudin-4 acts as a relative sodium

291

barrier and is also specifically up-regulated during epithelial repair (72, 196, 207). In

292

human and murine lungs, higher claudin-4 expression is associated with higher rates of

293

alveolar fluid clearance(82, 152, 207), which is suggestive of a protective or reparative

294

role for this claudin. Claudin-4 may also play a unique role in trans-epithelial ion

295

transport. For example, in zebra fish, the claudin-4 ortholog, claudin-b, associates with

296

Na+/K+-ATPase to effect decreased paracellular sodium transport in low ion

297

concentration conditions (92, 93). In mouse studies, the loss of claudin-4 results in

298

decreased alveolar fluid clearance and more severe pulmonary edema with injury(82,

299

207). These data are consistent with the hypothesis that lung epithelial tight junctions

300

containing high levels of claudin-4 support higher rates of alveolar fluid clearance. This

301

may be because claudin-4 forms high resistance junctions that limit sodium

302

conductance resulting in a larger trans-epithelial sodium gradient. Alternatively, claduin-

303

4 may play a role in the regulation of Na+/K+-ATPase, but further studies are needed. In

304

claudin-18 null mice, higher levels of claudin-4 may partly explain the observed increase

305

in alveolar fluid clearance.

306

Functional association between claudins and transcellular ion transport

307

Taken together, available data suggest a functional association between tight junction

308

claudins and transcellular ion transport. The hypothesis that claudin-4 and Na+/K+-

309

ATPase have a cooperative role in trans-epithelial sodium transport in the lung requires

310

further study, but it is clear that claudin-4 is necessary for normal barrier function and

311

maximal rates of fluid clearance. It is also clear that claudin-18 forms a unique

312

permeability barrier in the alveolar epithelium and is a requirement for alveolar

313

homeostasis. The loss of claudin-18 results in a compensatory increase in claudin-4

314

along with numerous changes in transcellular ion transporters in association with

315

increased rates of alveolar fluid clearance. The adaptation to chronic barrier leak in

316

claudin-18 null mice may provide novel insights into the mechanisms of alveolar ion and

317

water transport regulation in diseases characterized by epithelial barrier dysfunction.

318

319

Section III: The lung epithelium: The role of 2-pore domain potassium channels in

320

lung epithelium

321

Transcellular ion homeostasis in the lung epithelium

322

Maintenance of the integrity and the homeostatic properties of the alveolar capillary

323

barrier plays a vital role in pulmonary health and disease(111). By separating the air-

324

and blood-filled spaces in the lung, the alveolar epithelium is strategically positioned to

325

interact closely with both environments and to regulate the electrolyte and water content

326

of the alveolar spaces. In this section we will briefly review (1) the major ion transport

327

mechanisms regulating the composition of the alveolar lining fluid (ALF) under

328

physiologic conditions, and (2) the role of stretch-activated ion channels in the

329

development of hyperoxia (HO) - and mechanical stretch-induced Acute Lung Injury

330

(ALI) and Acute Respiratory Distress Syndrome (ARDS), with particular emphasis on

331

epithelial barrier function and inflammatory cytokine secretion.

332

Maintaining tight control over the intra-alveolar environment requires complex

333

synchronization of multiple epithelial ion transport processes, and a dynamic balance

334

between continuous secretion and re-absorption of Na+ and Cl- determines the

335

composition of the ALF(131). When an increase in alveolar fluid absorption is required,

336

as in the immediate postnatal period or in conditions associated with pulmonary edema

337

like pneumonia and congestive heart failure(136, 205), Na+ is internalized via apical

338

amiloride-sensitive ENaC channels and cyclic nucleotide-gated cation channels (CNG)

339

in both alveolar type I (AT I) and type II (AT II) cells(33, 36, 37, 45, 74). Internalized Na+

340

is then secreted into the interstitial space via the basolateral Na/K ATPase(4, 31). To

341

maintain intracellular electroneutrality, apical Cl- absorption occurs mainly via CFTR

342

channels or paracellular pathways(36). In combination, these electrolyte shifts create an

343

osmotic gradient for water molecules to move from the alveolar into the interstitial space

344

either via aquaporin channels or by diffusion (1, 88, 177, 198).

345

In contrast, when increased production of ALF is required, as part of physiologic ALF

346

turnover and aging, to maintain surfactant homeostasis, or in response to infectious or

347

environmental pollution stimuli(67, 81, 106, 120), Cl- anions are actively secreted into

348

the alveolar space mainly via apical CFTR channels, ClC channels, and Ca-dependent

349

Cl- channels (35, 50, 69, 108). An active driving force for Cl- secretion is maintained by

350

basolateral Na/K/2Cl co-transporters, K-Cl co-transporters, HCO3/Cl exchangers(5, 91,

351

151, 175, 190), as well as basolateral inwardly and outwardly rectifying Cl- channels(10,

352

69). Na+ ions are thought to follow Cl- ions from the interstitial into the alveolar space

353

but the molecular mechanisms underlying Na+ secretion are still not fully understood.

354

Similarly, basolateral K+ channels play an important role in maintaining ALF by creating

355

a favorable electrochemical gradient for Na+ absorption and Cl- secretion, by restoring

356

intracellular electroneutrality, and by stabilizing the resting membrane potential(112). A

357

variety of K+ channels have been described in both AT I and AT II cells, including

358

voltage-gated (Kv), ATP-dependent (KATP), inwardly rectifying (Kir), Ca2+-activated (KCa)

359

K+ channels(8, 57), and small (SK) and large conductance (BK) K+ channels(11, 113).

360

In addition to homeostatic processes, K+ channels also appear to be involved in

361

epithelial wound healing, cell migration, cell proliferation and cytokine secretion in in

362

vitro and in vivo models of ALI/ARDS(8, 149, 161, 162, 192) and large conductance

363

BKCa channels may act as oxygen sensors in the lung epithelium(80).

364

Further complexity is added to this system when accounting for the intricate interactions

365

between these ion channels and transporter proteins. For example, CFTR can

366

downregulate ENaC expression(158); inhibition of KATP channels can inhibit ENaC

367

currents(90); activation of KATP channels can increase ENaC and CFTR currents(98);

368

and inhibition of KCa and Kv channels can inhibit ENaC and CFTR currents(17, 62).

369

There are numerous limitations in the study of epithelial ion transport processes

370

however, more recent approaches employing siRNA and shRNA knockdown of specific

371

ion channels(163), and the broader availability of knockout (KO) mice deficient in

372

specific ion channels (32, 124) have significantly facilitated research in this field.

373

Nevertheless, conditional KO models for specific epithelial ion channels, where a

374

particular ion channel is absent from AT I or AT II cells only, are not always readily

375

available.

376

The role of ENaC channels in lung fluid balance

377

The ENaC channels are composed of 4 subunits (α,β, γ, and δ) coded for by the

378

SCNN1A, SCNN1B, SCNN1G, and theSCNN1D genes respectively. The α subunit is

379

required to form a functional ENaC channel and the β and γ subunits amplify the

380

channel’s activity. In mice, homozygous knockout of the α-ENaC subunit leads to

381

retention of fetal lung liquid, respiratory distress, and ultimately death (74). In humans,

382

mutations in the ENaC subunit genes SCNN1A, SCNN1B, and SCNN1G can lead to

383

autosomal recessive pseudohypoaldosteronism (PHA) characterized by life-long salt

384

wasting, hyperkalemia, and dehydration(25). Children who have PHA type 1 with

385

mutations in the genes encoding the α and β subunit have an increased volume of fluid

386

in the lungs due to problems with fluid absorption (84) leading to recurrent respiratory

387

problems. There are, however, case reports of preterm infants with a single

388

homozygous mutation in the α-ENaC subunit who did not have immediate post natal

389

difficulty with lung fluid clearance indicating that the α-ENaC may not be pivotal in the

390

clearance of lung fluid at early stages of lung maturation(75).

391

TGF-β has been shown to be important in the regulation of ENaC activity which has

392

important implications for lung fluid balance. Peters et al. demonstrated that TGF-β can

393

drive the abnormal internalization of the ENaC complex leading to a reduction in this

394

complex at the lung epithelial cell surface resulting in reduced sodium and fluid

395

absorption (135). This data suggests a unique TGF-β dependent mechanism to

396

regulate ion and fluid transport in the lung and potentially a new pathological

397

mechanism of ALI and ARDS.

398

The role of 2-pore domain potassium channels in ALI and ARDS

399

The role of stretch-activated ion channels (SACs) in the development of hyperoxia (HO)

400

and mechanical stretch-induced ALI/ARDS and the consequences of SAC dysregulation

401

on epithelial barrier function and inflammatory cytokine secretion is of particular

402

interest(181, 182). Both in vitro and in vivo studies have demonstrated that mechanical

403

forces can alter trans-epithelial ion transport, which results in a net decrease in alveolar

404

fluid clearance(50, 146, 204) and is directly associated with an increased length of

405

mechanical ventilation and hospital mortality in patients with ALI/ARDS(70, 181).

406

The term SACs refers to a variety of ion channels, including Na+ channels such as

407

ENaC, Cl- channels such as the ClC family, Ca2+ channels such as the TRPV (transient

408

receptor potential vanilloid) family, and numerous K+ channels including 2-pore domain

409

potassium (K2P) channels, KATP and Kir channels(23, 126). SAC channels are

410

ubiquitously expressed in body tissues including the heart, kidneys, genitourinary tract,

411

gastrointestinal tract, leukocytes and great blood vessels(179, 202, 203) and regulate a

412

variety of cellular functions including the resting membrane potential, cell volume, gene

413

expression, cell differentiation and cell contraction processes(16, 39, 154).

414

The K2P channel family consists of 15 members and is divided into 6 subfamilies:

415

TWIK, THIK, TREK, TASK, TALK, TRESK(156). A unique property of K2P channels is

416

that they can be constitutively open creating so-called “K+ leak currents” that maintain

417

the negative resting membrane potential of a cell (12, 127) and in tissues other than the

418

lung, K2P channels have been shown to act as mechano-sensors and mechano-

419

transducers(212). For this review we will focus specifically the role of TREK-1 in

420

ALI/ARDS.

421

Regulation and function of TREK-1 channels in models of ALI/ARDS

422

TREK-1 expression was first detected in mouse, rat and human AECs(163) but also

423

appears to be expressed in airway epithelial cells(213). Preliminary data shows TREK-

424

1 deficient cultured mouse AECs and human bronchial epithelial cells appear protected

425

from a HO- and TNF-α-induced loss of trans-epithelial electrical resistance suggesting a

426

role for TREK-1 in epithelial barrier (unpublished data). In addition, stimulation of

427

TREK-1 deficient mouse and human epithelial cells with TNF-α decreased IL-6 and

428

RANTES secretion and increased in MCP-1 secretion (Fig. 2). In contrast, KC/IL-8

429

secretion remained unchanged between TREK-1 deficient and control cells. While

430

several TNF-α-activated signaling pathways, including p38 kinase, PKC, and c-Jun N-

431

terminal kinases (JNK1/2/3) were altered in TREK-1 deficient AECs, decreased IL-6

432

secretion from TREK-1 deficient AECs was related to impaired phosphorylation of the

433

PKC isoform θ after TNF-α stimulation, whereas increased MCP-1 secretion appeared

434

unrelated to the increased phosphorylation of JNK1/2/3 in TREK-1 deficient AECs. The

435

specific molecular signaling mechanisms underlying these alterations in cytokine

436

secretion is currently being investigated.

437

Based on in vitro findings, it was anticipated that TREK-1 KO mice would be protected

438

from HO- and mechanical stretch-induced lung injury. However, exposure of TREK-1

439

KO mice to 24 hours HO resulted in increased lung injury as evidenced by gross lung

440

anatomy, H&E staining of lung sections (increased inflammatory cell infiltration and

441

hemorrhage), increased lung injury scoring, decreased lung compliance, and increased

442

macrophage and neutrophil accumulation in the BAL fluid of TREK-1 KO mice(164).

443

Interestingly, exposure of TREK-1 KO mice to HO plus mechanical ventilation resulted

444

in no further injury when compared to HO exposure alone(164).

445

Similar to in vitro studies(163), in vivo models show HO exposure is a weak stimulus for

446

cytokine secretion and only HO plus mechanical ventilation increased IL-6, MCP-1, and

447

TNF-α levels in the BAL fluid of both WT and TREK-1 KO mice. In accordance with in

448

vitro findings, there were decreased amounts of IL-6 in HO plus mechanically ventilated

449

TREK-1 KO mice(161). However, in contrast to in vitro findings(162), MCP-1 levels

450

were decreased in HO plus mechanically ventilated TREK-1 KO mice, whereas TNF-α

451

levels were unchanged between control and TREK-1 KO mice(164). Therefore, in this

452

model alterations in cytokine secretion between TREK-1 KO and WT mice were unlikely

453

the cause for the observed increase in lung injury in the TREK-1 KO mice.

454

Nevertheless, the contributions of cytokines such as IL-6, IL-1β and TNF-α to the

455

development of ALI/ARDS in other animal models and in human ARDS patients are well

456

documented(118, 119). The pro-inflammatory environment associated with the

457

secretion of these mediators promotes neutrophil-, macrophage-, T cell- and epithelium-

458

mediated alveolar injury, causes loss of alveolar barrier function, and decreases

459

alveolar surfactant levels(61, 94, 119, 185, 206). Similar to in vitro studies, the specific

460

molecular mechanisms underlying these alterations in BAL cytokine levels in vivo

461

models remain to be determined. In addition, it will be important to investigate the

462

cellular sources for these inflammatory mediators and quantify their relative

463

contributions. Furthermore, it is of major importance to determine the time course of

464

cytokine secretion in these ARDS models. It is quite possible that although after 24

465

hours HO exposure and 4 hours mechanical ventilation BAL IL-6 and MCP-1 levels

466

were decreased in TREK-1 deficient mice, these cytokines were increased at earlier

467

time points and contributed to the observed lung injury. Of note, while TNF-α is a known

468

pro-inflammatory cytokine in the early stages of ARDS, in chronic pulmonary

469

inflammation it may actually exert anti-inflammatory, beneficial effects and promote

470

physiologic wound healing(117).

471

Interestingly, alveolar barrier function appeared uncompromised in the mouse ARDS

472

model since BAL protein levels were unchanged between control and TREK-1 deficient

473

mice(164). HO alone caused only a minor disruption in alveolar barrier function whereas

474

the combination of HO and mechanical ventilation resulted in significant BAL protein

475

accumulation in both mouse types. The protective effect of TREK-1 deficiency observed

476

in vitro on TNF-α- and HO-induced loss of barrier function was not seen in the whole

477

animal model. Whether a close interaction between TREK-1 and epithelial tight junction

478

proteins, as seen in cultured alveolar epithelial cells (unpublished data), also exists in

479

vivo remains to be confirmed.

480

One explanation for the increased lung damage observed in HO exposed TREK-1 KO

481

mice could be an increase in AT I and AT II cell apoptosis in these animals as

482

evidenced by TUNEL staining and corroborated by increased PARP-1 cleavage, one of

483

the distal signaling molecules of the caspase cascade(164). Another possible

484

explanation could be a decrease in surfactant protein-C detected in TREK-1 deficient

485

mice after prolonged HO exposure, potentially making lungs more vulnerable to HO-

486

and mechanical stretch-induced injury(164). The molecular mechanisms linking TREK-1

487

deficiency to epithelial apoptosis and decreased surfactant production are currently

488

unknown. As described above, we have observed substantial alterations in p38, PKC

489

and JNK phosphorylation in TREK-1 deficient epithelial cells after TNF-α

490

stimulation(161, 162), but whether these kinases are involved in apoptosis or surfactant

491

production needs to be determined. Another question that needs to be addressed in

492

future studies is whether K+ flux through TREK-1 channels is required for the observed

493

in vitro and in vivo effects, or whether TREK-1 could exert K+-independent functions and

494

act as a regulatory molecule rather than an ion channel, as reported for example for

495

CFTR(159).

496

In conclusion, while in vitro epithelial TREK-1 deficiency appeared protective against the

497

inflammatory stimuli responsible for the development of ALI/ARDS, in an in vivo model

498

of ALI/ARDS TREK-1 deficiency resulted in increased lung damage. The mechanisms

499

accounting for these differences are currently unknown. Conditional TREK-1 deficient

500

mice should give us insights into the molecular mechanisms responsible for the

501

observed lung damage and help us resolve the discrepancies between in vitro and in

502

vivo models. Once TREK-1 signaling pathways are delineated, the development of

503

TREK-1 activating or inhibiting agents may steer us towards new therapeutic

504

approaches in ALI/ARDS. As with all known ion channel modulators, drug specificity

505

issues will likely constitute future challenges, especially since K2P channels are quite

506

ubiquitously expressed in the body tissues.

507

Section IV: The lung epithelium: NRG-1-HER2/3 Signaling in Epithelial Barrier

508

regulation

509

HER Family Signaling: Background

510

Receptor tyrosine kinases, such as members of the Human Epidermal Growth Factor

511

Receptor (HER) family, and their ligands serve as important signaling molecules that

512

regulate the epithelial response to extracellular signals and environmental stimuli. HER

513

family signaling has been shown to regulate epithelial migration, growth and

514

differentiation as well as epithelial barrier function in numerous tissue beds including

515

skin (184), intestine (38), cornea (123, 211, 215), and the lung (30). Identification of

516

mutations of HER family genes as pathogenic in certain epithelial cancers (85, 180) has

517

led to the successful deployment of targeted tyrosine kinase inhibitors (TKIs) aimed at

518

inhibiting these receptors, particularly in breast and lung cancer (24, 105). However,

519

recent data demonstrate that these receptors regulate pulmonary epithelial barrier

520

function in non-transformed pulmonary epithelia, in particularly in the context of

521

inflammatory lung injury (46, 47).

522

The HER family is a complex signaling network consisting of 4 transmembrane

523

receptors, HER1 or the epidermal growth factor receptor (EGFR), HER2, HER3 and

524

HER4 and 10 known ligands. The HER family is also referred to as the ErbB family after

525

it was discovered that the avian erytheroblastosis virus gene encoded a mutant form of

526

EGFR/HER1 (64, 65, 138). Members of the HER family are expressed on the

527

basolateral surface of airway and alveolar type I and II epithelial cells. The 4 HER

528

receptors all share a similar structure including an extracellular ligand binding domain, a

529

single transmembrane domain, and a cytoplasmic tyrosine kinase domain (209). HER

530

receptors exist in quiescent cells as single “closed” monomers. Ligand binding induces

531

a conformational change to an “open” configuration allowing for homo or hetero-

532

dimerization leading to phosphorylation of intracellular tyrosine residues and activation

533

of the kinase domain. This allows for docking of cytoplasmic proteins that facilitate

534

initiation of downstream signaling. Certain structural distinctions influence how specific

535

monomers interact to form signaling competent dimer pairs. HER2 has no ligand yet,

536

under basal conditions, exists in a receptive “open” confirmation making it the preferred

537

binding partner of other HER receptors. In contrast, HER3 has at least 4 different

538

ligands yet does not have a functioning kinase and partners with other receptors to

539

influence signaling. Though it does not directly phosphorylate other proteins, HER3

540

interacts with other intra-cellular proteins and signaling pathways such as the PI3 kinase

541

pathway (103). Additionally, in cancer cells, HER3-depedent signaling is a resistance

542

mechanism against HER2-targeted TKI therapy (52). Broadly, ligands are organized

543

depending on which receptors they bind and activate. Epidermal growth factor (EGF),

544

transforming growth factor alpha (TGF-α) and amphiregulin bind EGFR/HER1

545

exclusively while betacellulin, heparin binding EGF (HB-EGF) and epiregulin can bind

546

EGFR/HER1 or HER4. The final group of ligands includes the neuregulins (NRG-1-4).

547

The 4 NRG proteins all share a similar EGF like domain which is sufficient for receptor

548

activation yet differ in tissue distribution structurally which can influence signaling, NRG-

549

1 being the isoform most commonly studied.

550

HER family signaling is a complex, multilayered network influenced by factors such as

551

dimerization partner, cell type (e.g. cancer vs. untransformed) and ligand. (148). For

552

example, activated EGFR phosphorylates αCbl and Grb2 but only when activated in

553

partnership with HER2 as opposed to other HER family members (58, 129).

554

Additionally, while several ligands can bind and activate different HER receptors, each

555

ligand elicits specific intra-cellular responses depending cell type (e.g. transformed vs.

556

non-transformed) and the availability of other HER receptors. In HER2 deficient cells,

557

NRG-1 can induce EGFR/HER3 dimerization and activation but when HER2 is

558

available, NRG-1 preferentially induces HER2/3 heterodimers eliciting specific

559

intracellular signaling patterns (46, 58).

560

Accumulating data suggests that the HER family is important in regulating epithelial

561

barrier function in the lung. Activation of EGFR with TGF-α leads to epithelial

562

proliferation, spreading and motility (99, 153) (86) and enhances wound closure in

563

cultured alveolar epithelial cells (86). Additionally, epithelial injury leads to shedding of

564

HER family ligands and wound closure is delayed after exposure to an EGFR inhibitor.

565

Similarly, NRG-1 shedding and HER2 activation is also observed in a scratch model of

566

wound closure and blockade of HER2 impedes healing following wounding (200).

567

Notably, exogenous NRG-1 increases epithelial permeability similar in a monolayer of

568

epithelial cells (46). These data suggest that HER activation can enhance, improve, or

569

worsen epithelial barrier function, possibly depending on the continuity of the barrier at

570

the time of receptor activation.

571

NRG-1-HER2/3: Regulation of the Pulmonary Epithelial Barrier

572

Recent evidence has identified the NRG-1-HER2/3 system as activated in and a critical

573

modulator of pulmonary epithelial barrier function in acute lung injury (ALI) and the

574

Acute Respiratory Distress Syndrome (ARDS). ALI and ARDS are syndromes marked

575

by a hyper-inflammatory state with an increase in cytokines such as IL-6 and IL-8 that

576

are felt to participate in disease pathogenesis. In particular, the cytokine IL-1β has been

577

implicated as a central mediator of ALI, both in animal models and humans(53, 71, 114,

578

132, 133, 140, 174). IL-1β is increased in the lavage of ARDS patients and exogenous

579

IL-1β leads to increased epithelial permeability in an epithelial monolayer as well as in

580

animal models (89). Therapeutically, strategies to block IL-1β signaling is protective in

581

bleomycin and ventilator-induced models of lung injury (53, 132) (49). Epithelial NRG-1-

582

HER2 signaling has been recently identified as activated by IL-1β and central in the

583

pathogenesis of epithelial injury and permeability induced by IL-1β. In cultured airway

584

and alveolar cells, IL-1β-mediated increases in epithelial permeability are HER2

585

dependent (46). IL-1β induces shedding of NRG-1 and HER2 activation without

586

evidence of EGFR activation. The protease ADAM17 has been identified as the

587

sheddase responsible for NRG-1 shedding. Pharmacologic (inhibitors) or genetic

588

(siRNA) strategies to inhibit ADAM17, NRG-1 or HER2 significantly attenuate IL-1β-

589

mediated changes in epithelial barrier function. Hence, an overall pathway of IL-1β-

590

ADAM17-NRG-1-HER2/3 leading to increased epithelial permeability has emerged as a

591

new important regulator of epithelial barrier function in ALI.

592

NRG-1-HER2/3 signaling also participates in animal models of lung injury. Bleomycin

593

induced ALI is associated with both increased broncho-alveolar lavage NRG-1

594

indicative of surface shedding as well as pulmonary HER2 activation. Pharmacologic

595

ADAM17 inhibition attenuates NRG-1 cleavage, pulmonary HER2 phosphorylation and

596

indices of injury in mice exposed to bleomycin. Bleomycin serves as a model of both

597

acute and chronic, fibrotic lung injury and targeting HER2 is protective in the fibrotic

598

phase as well as the acute phase of lung injury following bleomycin. Use of an inhibitor

599

that prevents HER2 dimerization with HER3 prevented both HER2/3 activation as well

600

as fibrosis at 21 days after bleomycin exposure (44). Similarly, transgenic mice lacking

601

pulmonary epithelial HER3 are protected from bleomycin-induced fibrosis (125). Though

602

in these studies indices of acute injury were not specifically studied, there was

603

suggestion of protection in alveolar leak early following bleomycin. Mechanistically,

604

alterations in alveolar epithelial permeability have been linked to a pro-fibrotic response

605

in the lung (2, 3, 15, 178) possibly providing a link between early acute injury marked by

606

compromised alveolar epithelial barrier function and delayed fibrotic injury.

607

Proteases are critical participants in the HER family signaling as they translate external

608

signals into HER receptor activation through shedding of surface ligands. Numerous

609

agonists which induce epithelia barrier dysfunction do so via protease-mediated HER

610

ligand shedding leading to receptor activation. Examples of this include IL-1β,

611

mechanical stretch, and hypertonic stress. In particular, ADAM17 has been identified as

612

the sheddase responsible for cleaving numerous HER family ligands and has been

613

linked to disease marked by inflammation and increased permeability (21, 27, 29, 55,

614

211). In vitro, blocking ADAM17 reduces IL-1β-induced epithelial permeability through

615

blocking NRG-1 shedding and subsequent HER2 activation. In vivo, intra-tracheal

616

instillation of the ADAM17 inhibitor attenuated bleomycin induced ALI including protein

617

leak. This was associated with decreased NRG-1 lavage levels and HER2 activation.

618

While ADAM17 KO mice display embryonic lethality, mice deficient in the endogenous

619

inhibitor of ADAM17- Tissue Inhibitor of Metalloprotease (TIMP)-3- have delayed

620

resolution of lung injury following bleomycin administration supporting the importance of

621

this sheddase in ALI, though the role of HER ligands and receptors was not specifically

622

investigated.

623

Perhaps the most compelling data indicating that the HER family participates in

624

epithelial barrier disruption in the lung is that patients with ARDS have evidence of

625

active ligand shedding in the lung. TGF-α is elevated in lavage from patients suggesting

626

pathway activation. Similarly, increased NRG-1 lavage levels have been detected from

627

patients with ARDS. As a biomarker, NRG-1 levels were significantly correlated with

628

inflammation as measured by inflammatory cytokines. Notably, NRG-1 elevations were

629

associated with worse outcome as measured by ventilator free days. Plasma NRG-1 is

630

also increased in ARDS patients suggesting that the source of NRG-1 leading to HER2

631

activation could be extra-pulmonary.

632 633

NRG-1-HER2/3 Signaling- Mechanisms Regulating Epithelial Cell-Cell adhesion

634

and Permeability.

635

Knowledge regarding the mechanisms by which HER2 and the other HER receptors

636

influence epithelial permeability is incomplete but several pathways have been outlined.

637

HER proteins modulate cell-cell adhesion proteins of the tight junction and the adherens

638

junction. EGFR alters claudin expression and spatial re-arrangement of claudins and

639

ZO-1 (28, 189). HER2 has also been linked to the tight junction proteins ZO-1 and

640

occludin (54, 56) as well as integrin β4 in regulating cell-cell adhesion (66). Recent data

641

suggest that the adherens junction protein β-catenin is a required intermediary

642

influencing HER2 mediated barrier function in the lung. Phosphorylation at tyrosine Y-

643

654 of β-catenin has been linked to altered barrier function. NRG-1-mediated HER2

644

activation leads to β-catenin Y-654 phosphorylation as well as disruption of β-catenin–

645

E-cadherin interaction at the cell junction in pulmonary epithelial cells (48). This is

646

associated with impaired E-cadherin-mediated cell-cell adhesion. Finally loss of β-

647

catenin via shRNA knock down results in a significant attenuation in NRG-1- HER2/3

648

induced barrier dysfunction. Together, this data suggest that NRG-1-HER3-HER2

649

activation leads to β-catenin tyrosine phosphorylation, disruption of adherens junction,

650

and alteration of the pulmonary epithelial barrier (Fig. 3).

651

Additionally, processes inherent in barrier function such as proliferation and cell

652

migration suggest other pathways by which HER proteins might influence the epithelial

653

barrier. The GTPases Rho and Rac have both been linked to HER family signaling. In a

654

breast cancer epithelial cell line, HER2-dependent Rac activation was required for

655

NRG-1 mediated cell migration (208). Rac activation following NRG-1 is likely via

656

HER2-dependent P-Rex1 activation as down-regulation of P-Rex1 inhibited Rac

657

activation and cell migration following NRG-1 (121). Similarly, EGFR activation leads to

658

Rac activation and Rac-dependent cell migration (34). During epithelial wound closure,

659

Rho activation follows EGFR activation and, in a corneal epithelial wound model, Rho

660

inhibition decreased both cell migration and proliferation resulting in delayed wound

661

closure (210).

662

Proteins involved in formation of tight junctions and adherens junctions are also

663

mobilized after HER family activation. EGFR activation alters expression levels of

664

claudins and the cellular localization of claudins and ZO-1 (28, 189). NRG-1-HER2

665

mediated changes in epithelial permeability are also associated with alterations in

666

cellular localization of the tight junction proteins ZO-1 and occludin (54, 56). Integrin β4,

667

an adhesion molecule which regulates cell-cell junctions (66) and epithelial proliferation

668

(26), amplifies HER2 activation in epithelial cells to activate STAT and disrupt cell-cell-

669

adhesion (60). Previous reports have suggested a structural and functional link between

670

the adherens junction protein β-catenin and HER2. Structurally, HER2 co-localizes to

671

the basolateral plasma membrane via binding to the PDZ domain of erbin (20). Erbin

672

has been identified as a binding partner for the p120 catenin protein p0071 as well as

673

direct binding partner for β-catenin (59, 147). In gastric and breast cancer epithelial

674

cells, TGF-α induces EGFR dependent HER2 activation, β-catenin phosphorylation and

675

β-catenin-HER2 association, though the role of HER2 in β-catenin phosphorylation was

676

not specifically studied (83, 128, 160, 170). Functionally, HER2 and β-catenin have both

677

been linked to breast cancer pathogenesis. In murine breast cancer models utilizing a

678

constitutively active HER2, β-catenin signaling was activated and required for tumor

679

initiation and metastasis (155). Blockade of HER2 using trastuzumab blocked both

680

HER2 activation and β-catenin phosphorylation as well as β-catenin dependent

681

transcription of surviving breast cancer cells (214).

682 683

Conclusion

684

The airway epithelium is a physical and immunological barrier that sits at the interface

685

between the lumen and the lung parenchyma and is the initial site of contact for all

686

inspired substances. It is constantly exposed to both physiological and pathological

687

stimuli and dynamically regulates its barrier. This dynamic regulation occurs via

688

changes in cell-cell adhesion due to altered cytoskeletal and junctional protein

689

arrangement and expression, as well as changes in transcellular and paracellular ion

690

permeability in response to these stimuli to help maintain homeostasis. The ion

691

channels on the apical and basolateral cell surfaces and the junctional complexes,

692

which directly connect adjacent epithelial cells, play a key role in controlling the

693

movement of substances from the lumen into the sub-epithelial space both via

694

paracellular and transcellular transport. The junctions also separate the apical and

695

basolateral cell surface which maintains the specialized function of each cell surface,

696

controls the interaction between various cytokines, growth factors, and their receptors,

697

and modulates the extent and location of inflammatory response to stimuli. Exposure to

698

noxious stimuli such as mechanical stretch/trauma, hyperoxia, infection, cigarette

699

smoke, or particulate matter, damages the epithelial barrier resulting in a loss of

700

compartmentalization and a hyperproliferative, inflammatory state that is seen in many

701

pulmonary diseases.

702

The junction proteins and ion channels interact to maintain lung fluid balance. Initial

703

studies indicate that changes in the levels of the tight junction claudins can alter fluid

704

balance. However, further work needs to be done to characterize the compensatory

705

shifts in junction proteins that occur in response to physiological and pathological stimuli

706

in order to maintain tissue homeostasis.

707

As outlined earlier, stretch activated ion channels play a role in epithelial barrier

708

function. TREK-1 has been linked to the pathogenesis of ALI/ARDS however there are

709

conflicting results in in vitro and in vivo models of ALI/ARDS. Future studies delineating

710

the role of TREK-1 in ALI/ARDS could potentially lead to the development of new

711

targeted therapies for this disease. The ENaC complex is another ion channel shown to

712

be important in lung fluid balance but better defining the role of the individual subunits in

713

fluid balance as well as identifying other pathways that contribute to the trafficking of

714

this complex will be important in further understanding the pathogenesis of diseases

715

such as ARDS.

716

Although it is hypothesized that changes in the physical barrier in response to chronic

717

stimulation by both physiological and pathological stimuli precipitate the immunological

718

changes that lead to diseases such as COPD, ALI, and ARDs, the sequence and time

719

course of these changes is yet to be determined. As discussed in this review, the

720

epithelium dynamically regulates its response to a variety of stimuli to promote repair

721

and regeneration however; we do not understand mechanisms of dysregulation of this

722

dynamic repair process. Future studies should further explore the link between altered

723

transcellular and paracellular transport and the activation of the inflammatory cascades

724

that result in chronic epithelial barrier dysfunction and injury. In addition, much attention

725

has been devoted to pathways that mediate lung inflammation and injury however,

726

pathways that protect against lung injury are not well studied and therefore few have

727

been detailed in the literature. It is critical to note that Islam et al. demonstrated that

728

both epithelial injury and the downstream consequences of disease can be reversed by

729

targeting the epithelium, which was done using mitochondrial transfer (77). However,

730

this raises a future directive to not only identify pathways involved in damage, but to

731

also investigate protective mechanisms. Recently, the apelin-APJ pathway has been

732

shown to be protective against ARDS/ALI but more work needs to be done to identify

733

other pathways that protect against lung inflammation and injury(42). Our future

734

directive is not to simply define and dissect disrupted molecular pathways in the

735

epithelium, but target and manipulate protective mechanisms to alter the course of

736

disease. In conditions such as COPD, where there is a long lead time prior to the onset

737

of disease, there is ample opportunity to prevent the development by manipulating

738

epithelial targets. In conditions such as ARDS, we need to fine tune mechanisms to

739

target the epithelium to ameliorate the severity of disease and amplify repair and

740

resolution.

741

As discussed earlier in this review, the maintenance of ion and fluid homeostasis in the

742

lung is complex and requires the dynamic interaction between numerous ion channels

743

and transporter proteins. Studies have started to explore the connections between

744

paracellular and transcellular transport in maintaining fluid balance and ion

745

homeostasis. However, our knowledge regarding this subject is still in its infancy. We

746

have already identified certain transporters, such as Aquaporin 5, which have roles in

747

dictating both transcellular water transport and paracellular permeability as outlined in

748

Section I. Further defining the contribution of these transporters to the pathogenesis of

749

different pulmonary diseases could potentially lead to the development of novel

750

therapeutics. Indeed, Ivacaftor, a drug approved to treat patients with cystic fibrosis

751

who have specific mutations in the CFTR protein, is one such example, which could

752

have reaches into non-cystic fibrosis lung diseases as well.

753

In summary, the airway epithelium is a dynamic barrier that constantly responds to

754

luminal stimuli and coordinates its response to maintain homeostasis in the lung.

755

Breakdown in this coordinated response can lead to a myriad of lung diseases.

756

References

757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781

1. Ablimit A, Hasan B, Lu W, Qin W, Wushouer Q, Zhong N, and Upur H. Changes in water channel aquaporin 1 and aquaporin 5 in the small airways and the alveoli in a rat asthma model. Micron (Oxford, England : 1993) 45: 68-73, 2013. 2. Adamson IY, Hedgecock C, and Bowden DH. Epithelial cell-fibroblast interactions in lung injury and repair. The American journal of pathology 137: 385-392, 1990. 3. Adamson IY, Young L, and Bowden DH. Relationship of alveolar epithelial injury and repair to the induction of pulmonary fibrosis. The American journal of pathology 130: 377-383, 1988. 4. Adir Y, Welch LC, Dumasius V, Factor P, Sznajder JI, and Ridge KM. Overexpression of the Na-KATPase alpha2-subunit improves lung liquid clearance during ventilation-induced lung injury. Am J Physiol Lung Cell Mol Physiol 294: L1233-1237, 2008. 5. Al-Bazzaz FJ, Hafez N, Tyagi S, Gailey CA, Toofanfard M, Alrefai WA, Nazir TM, Ramaswamy K, and Dudeja PK. Detection of Cl--HCO3- and Na+-H+ exchangers in human airways epithelium. JOP : Journal of the pancreas 2: 285-290, 2001. 6. Al-Haggar M, Bakr A, Tajima T, Fujieda K, Hammad A, Soliman O, Darwish A, Al-Said A, Yahia S, and Abdel-Hady D. Familial hypomagnesemia with hypercalciuria and nephrocalcinosis: unusual clinical associations and novel claudin16 mutation in an Egyptian family. Clin Exp Nephrol 13: 288-294, 2009. 7. Angelow S, Ahlstrom R, and Yu AS. Biology of claudins. Am J Physiol Renal Physiol 295: F867876, 2008. 8. Bardou O, Trinh NT, and Brochiero E. Molecular diversity and function of K+ channels in airway and alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 296: L145-155, 2009. 9. Basset G, Crone C, and Saumon G. Fluid absorption by rat lung in situ: pathways for sodium entry in the luminal membrane of alveolar epithelium. J Physiol 384: 325-345, 1987. 10. Berger J, Richter K, Clauss WG, and Fronius M. Evidence for basolateral Cl- channels as modulators of apical Cl- secretion in pulmonary epithelia of Xenopus laevis. Am J Physiol Regul Integr Comp Physiol 300: R616-623, 2011.

782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828

11. Bernard K, Bogliolo S, Soriani O, and Ehrenfeld J. Modulation of calcium-dependent chloride secretion by basolateral SK4-like channels in a human bronchial cell line. The Journal of membrane biology 196: 15-31, 2003. 12. Berrier C, Pozza A, de Lacroix de Lavalette A, Chardonnet S, Mesneau A, Jaxel C, le Maire M, and Ghazi A. The purified mechanosensitive channel TREK-1 is directly sensitive to membrane tension. J Biol Chem 288: 27307-27314, 2013. 13. Berthiaume Y and Matthay MA. Alveolar edema fluid clearance and acute lung injury. Respir Physiol Neurobiol 159: 350-359, 2007. 14. Bhowmik A, Chahal K, Austin G, and Chakravorty I. Improving mucociliary clearance in chronic obstructive pulmonary disease. Respir Med 103: 496-502, 2009. 15. Bitterman PB. Pathogenesis of fibrosis in acute lung injury. Am J Med 92: 39S-43S, 1992. 16. Bittner S, Ruck T, Schuhmann MK, Herrmann AM, Moha ou Maati H, Bobak N, Gobel K, Langhauser F, Stegner D, Ehling P, Borsotto M, Pape HC, Nieswandt B, Kleinschnitz C, Heurteaux C, Galla HJ, Budde T, Wiendl H, and Meuth SG. Endothelial TWIK-related potassium channel-1 (TREK1) regulates immune-cell trafficking into the CNS. Nature medicine 19: 1161-1165, 2013. 17. Boiko N, Kucher V, Eaton BA, and Stockand JD. Inhibition of neuronal degenerin/epithelial Na+ channels by the multiple sclerosis drug 4-aminopyridine. J Biol Chem 288: 9418-9427, 2013. 18. Boon M, De Boeck K, Jorissen M, and Meyts I. Primary ciliary dyskinesia and humoral immunodeficiency--is there a missing link? Respir Med 108: 931-934. 19. Boon M, Jorissen M, Proesmans M, and De Boeck K. Primary ciliary dyskinesia, an orphan disease. Eur J Pediatr 172: 151-162. 20. Borg J-P, Marchetto S, Le Bivic A, Ollendorff V, Jaulin-Bastard F, Saito H, Fournier E, Adélaïde J, Margolis B, and Birnbaum D. ERBIN: a basolateral PDZ protein that interacts with the mammalian ERBB2/HER2 receptor. Nature Cell Biology 2: 407, 2000. 21. Borrell-Pages M, Rojo F, Albanell J, Baselga J, and Arribas J. TACE is required for the activation of the EGFR by TGF-alpha in tumors. EMBO J 22: 1114-1124, 2003. 22. Burke WM, Roberts CM, Bryant DH, Cairns D, Yeates M, Morgan GW, Martin BJ, Blake H, Penny R, Zaunders JJ, and et al. Smoking-induced changes in epithelial lining fluid volume, cell density and protein. Eur Respir J 5: 780-784, 1992. 23. Buxton IL, Heyman N, Wu YY, Barnett S, and Ulrich C. A role of stretch-activated potassium currents in the regulation of uterine smooth muscle contraction. Acta Pharmacol Sin 32: 758-764, 2011. 24. Carr LL, Finigan JH, and Kern JA. Evaluation and treatment of patients with non-small cell lung cancer. Med Clin North Am 95: 1041-1054, 2011. 25. Chang SS, Grunder S, Hanukoglu A, Rosler A, Mathew PM, Hanukoglu I, Schild L, Lu Y, Shimkets RA, Nelson-Williams C, Rossier BC, and Lifton RP. Mutations in subunits of the epithelial sodium channel cause salt wasting with hyperkalaemic acidosis, pseudohypoaldosteronism type 1. Nat Genet 12: 248-253, 1996. 26. Chapman HA, Li X, Alexander JP, Brumwell A, Lorizio W, Tan K, Sonnenberg A, Wei Y, and Vu TH. Integrin alpha6beta4 identifies an adult distal lung epithelial population with regenerative potential in mice. J Clin Invest 121: 2855-2862, 2011. 27. Charbonneau M, Harper K, Grondin F, Pelmus M, McDonald PP, and Dubois CM. Hypoxiainducible factor mediates hypoxic and tumor necrosis factor alpha-induced increases in tumor necrosis factor-alpha converting enzyme/ADAM17 expression by synovial cells. J Biol Chem 282: 33714-33724, 2007. 28. Chen SP, Zhou B, Willis BC, Sandoval AJ, Liebler JM, Kim KJ, Ann DK, Crandall ED, and Borok Z. Effects of transdifferentiation and EGF on claudin isoform expression in alveolar epithelial cells. J Appl Physiol 98: 322-328, 2005.

829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875

29. Colon AL, Menchen LA, Hurtado O, De Cristobal J, Lizasoain I, Leza JC, Lorenzo P, and Moro MA. Implication of TNF-alpha convertase (TACE/ADAM17) in inducible nitric oxide synthase expression and inflammation in an experimental model of colitis. Cytokine 16: 220-226, 2001. 30. Crosby LM and Waters CM. Epithelial repair mechanisms in the lung. Am J Physiol Lung Cell Mol Physiol 298: L715-731, 2010. 31. Crump RG, Askew GR, Wert SE, Lingrel JB, and Joiner CH. In situ localization of sodiumpotassium ATPase mRNA in developing mouse lung epithelium. Am J Physiol 269: L299-308, 1995. 32. De Lisle RC. Disrupted tight junctions in the small intestine of cystic fibrosis mice. Cell and tissue research 355: 131-142, 2014. 33. Demaio L, Tseng W, Balverde Z, Alvarez JR, Kim KJ, Kelley DG, Senior RM, Crandall ED, and Borok Z. Characterization of mouse alveolar epithelial cell monolayers. Am J Physiol Lung Cell Mol Physiol 296: L1051-1058, 2009. 34. Dise RS, Frey MR, Whitehead RH, and Polk DB. Epidermal growth factor stimulates Rac activation through Src and phosphatidylinositol 3-kinase to promote colonic epithelial cell migration. Am J Physiol Gastrointest Liver Physiol 294: G276-285, 2008. 35. Dobbs LG and Johnson MD. Alveolar epithelial transport in the adult lung. Respiratory physiology & neurobiology 159: 283-300, 2007. 36. Eaton DC, Chen J, Ramosevac S, Matalon S, and Jain L. Regulation of Na+ channels in lung alveolar type II epithelial cells. Proceedings of the American Thoracic Society 1: 10-16, 2004. 37. Eaton DC, Helms MN, Koval M, Bao HF, and Jain L. The contribution of epithelial sodium channels to alveolar function in health and disease. Annu Rev Physiol 71: 403-423, 2009. 38. Egan LJ, de Lecea A, Lehrman ED, Myhre GM, Eckmann L, and Kagnoff MF. Nuclear factorkappa B activation promotes restitution of wounded intestinal epithelial monolayers. Am J Physiol Cell Physiol 285: C1028-1035, 2003. 39. Eisenhut M and Wallace H. Ion channels in inflammation. Pflugers Arch 461: 401-421, 2011. 40. Ellison RT, 3rd and Giehl TJ. Killing of gram-negative bacteria by lactoferrin and lysozyme. J Clin Invest 88: 1080-1091, 1991. 41. Falk MM. Adherens junctions remain dynamic. BMC Biol 8: 34. 42. Fan XF, Xue F, Zhang YQ, Xing XP, Liu H, Mao SZ, Kong XX, Gao YQ, Liu SF, and Gong YS. The Apelin-APJ Axis Is An Endogenous Counter-injury Mechanism In Experimental Acute Lung Injury. Chest. 43. Fang X, Song Y, Hirsch J, Galietta LJ, Pedemonte N, Zemans RL, Dolganov G, Verkman AS, and Matthay MA. Contribution of CFTR to apical-basolateral fluid transport in cultured human alveolar epithelial type II cells. Am J Physiol Lung Cell Mol Physiol 290: L242-249, 2006. 44. Faress JA, Nethery DE, Kern EF, Eisenberg R, Jacono FJ, Allen CL, and Kern JA. Bleomycininduced pulmonary fibrosis is attenuated by a monoclonal antibody targeting HER2. J Appl Physiol 103: 2077-2083, 2007. 45. Fesenko EE, Kolesnikov SS, and Lyubarsky AL. Induction by cyclic GMP of cationic conductance in plasma membrane of retinal rod outer segment. Nature 313: 310-313, 1985. 46. Finigan JH, Faress JA, Wilkinson E, Mishra RS, Nethery DE, Wyler D, Shatat M, Ware LB, Matthay MA, Mason R, Silver RF, and Kern JA. Neuregulin-1-human epidermal receptor-2 signaling is a central regulator of pulmonary epithelial permeability and acute lung injury. J Biol Chem 286: 1066010670, 2011. 47. Finigan JH, Mishra R, Vasu VT, Silveira LJ, Nethery DE, Standiford TJ, Burnham EL, Moss M, and Kern JA. Bal neuregulin-1 is elevated in acute lung injury and correlates with inflammation. Eur Respir J, 2012. 48. Finigan M and Warm J. Ventilatory considerations in cardiac patients. Nurs Clin North Am 7: 541-548, 1972.

876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921

49. Frank JA, Pittet JF, Wray C, and Matthay MA. Protection from experimental ventilator-induced acute lung injury by IL-1 receptor blockade. Thorax 63: 147-153, 2008. 50. Fronius M, Clauss WG, and Althaus M. Why Do We have to Move Fluid to be Able to Breathe? Frontiers in physiology 3: 146, 2012. 51. Furuse M, Sasaki H, Fujimoto K, and Tsukita S. A single gene product, claudin-1 or -2, reconstitutes tight junction strands and recruits occludin in fibroblasts. J Cell Biol 143: 391-401, 1998. 52. Garrett JT, Olivares MG, Rinehart C, Granja-Ingram ND, Sanchez V, Chakrabarty A, Dave B, Cook RS, Pao W, McKinely E, Manning HC, Chang J, and Arteaga CL. Transcriptional and posttranslational up-regulation of HER3 (ErbB3) compensates for inhibition of the HER2 tyrosine kinase. Proc Natl Acad Sci U S A 108: 5021-5026, 2011. 53. Gasse P, Mary C, Guenon I, Noulin N, Charron S, Schnyder-Candrian S, Schnyder B, Akira S, Quesniaux VF, Lagente V, Ryffel B, and Couillin I. IL-1R1/MyD88 signaling and the inflammasome are essential in pulmonary inflammation and fibrosis in mice. J Clin Invest 117: 3786-3799, 2007. 54. Giri S, Poindexter KM, Sundar SN, and Firestone GL. Arecoline induced disruption of expression and localization of the tight junctional protein ZO-1 is dependent on the HER 2 expression in human endometrial Ishikawa cells. BMC Cell Biol 11: 53, 2010. 55. Gloushankova NA. Changes in regulation of cell-cell adhesion during tumor transformation. Biochemistry (Mosc) 73: 742-750, 2008. 56. Gon Y, Matsumoto K, Terakado M, Sekiyama A, Maruoka S, Takeshita I, Kozu Y, Okayama Y, Ra C, and Hashimoto S. Heregulin activation of ErbB2/ErbB3 signaling potentiates the integrity of airway epithelial barrier. Exp Cell Res 317: 1947-1953, 2011. 57. Gonzalez C, Baez-Nieto D, Valencia I, Oyarzun I, Rojas P, Naranjo D, and Latorre R. K(+) channels: function-structural overview. Comprehensive Physiology 2: 2087-2149, 2012. 58. Graus-Porta D, Beerli RR, Daly JM, and Hynes NE. ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J 16: 1647-1655, 1997. 59. Gujral Taranjit S, Karp Ethan S, Chan M, Chang Bryan H, and MacBeath G. Family-wide Investigation of PDZ Domain-Mediated Protein-Protein Interactions Implicates β-Catenin in Maintaining the Integrity of Tight Junctions. Chemistry & Biology 20: 816-827, 2013. 60. Guo W, Pylayeva Y, Pepe A, Yoshioka T, Muller WJ, Inghirami G, and Giancotti FG. Beta 4 integrin amplifies ErbB2 signaling to promote mammary tumorigenesis. Cell 126: 489-502, 2006. 61. Halbertsma FJ, Vaneker M, Scheffer GJ, and van der Hoeven JG. Cytokines and biotrauma in ventilator-induced lung injury: a critical review of the literature. The Netherlands journal of medicine 63: 382-392, 2005. 62. Han DY, Nie HG, Gu X, Nayak RC, Su XF, Fu J, Chang Y, Rao V, and Ji HL. K+ channel openers restore verapamil-inhibited lung fluid resolution and transepithelial ion transport. Respir Res 11: 65, 2010. 63. Hartsock A and Nelson WJ. Adherens and tight junctions: structure, function and connections to the actin cytoskeleton. Biochim Biophys Acta 1778: 660-669, 2008. 64. Hayman MJ and Beug H. Identification of a form of the avian erythroblastosis virus erb-B gene product at the cell surface. Nature 309: 460-462, 1984. 65. Hayman MJ, Ramsay GM, Savin K, Kitchener G, Graf T, and Beug H. Identification and characterization of the avian erythroblastosis virus erbB gene product as a membrane glycoprotein. Cell 32: 579-588, 1983. 66. Hintermann E, Yang N, O'Sullivan D, Higgins JM, and Quaranta V. Integrin alpha6beta4-erbB2 complex inhibits haptotaxis by up-regulating E-cadherin cell-cell junctions in keratinocytes. J Biol Chem 280: 8004-8015, 2005.

922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968

67. Hobi N, Siber G, Bouzas V, Ravasio A, Perez-Gil J, and Haller T. Physiological variables affecting surface film formation by native lamellar body-like pulmonary surfactant particles. Biochim Biophys Acta 1838: 1842-1850, 2014. 68. Hogg JC and Timens W. The pathology of chronic obstructive pulmonary disease. Annu Rev Pathol 4: 435-459, 2009. 69. Hollenhorst MI, Richter K, and Fronius M. Ion transport by pulmonary epithelia. Journal of biomedicine & biotechnology 2011: 174306, 2011. 70. Holter JF, Weiland JE, Pacht ER, Gadek JE, and Davis WB. Protein permeability in the adult respiratory distress syndrome. Loss of size selectivity of the alveolar epithelium. J Clin Invest 78: 15131522, 1986. 71. Hoshino T, Okamoto M, Sakazaki Y, Kato S, Young HA, and Aizawa H. Role of proinflammatory cytokines IL-18 and IL-1beta in bleomycin-induced lung injury in humans and mice. Am J Respir Cell Mol Biol 41: 661-670, 2009. 72. Hou J, Gomes AS, Paul DL, and Goodenough DA. Study of claudin function by RNA interference. J Biol Chem 281: 36117-36123, 2006. 73. Humlicek AL, Manzel LJ, Chin CL, Shi L, Excoffon KJ, Winter MC, Shasby DM, and Look DC. Paracellular permeability restricts airway epithelial responses to selectively allow activation by mediators at the basolateral surface. J Immunol 178: 6395-6403, 2007. 74. Hummler E, Barker P, Gatzy J, Beermann F, Verdumo C, Schmidt A, Boucher R, and Rossier BC. Early death due to defective neonatal lung liquid clearance in alpha-ENaC-deficient mice. Nature genetics 12: 325-328, 1996. 75. Huppmann S, Lankes E, Schnabel D, and Buhrer C. Unimpaired postnatal respiratory adaptation in a preterm human infant with a homozygous ENaC-alpha unit loss-of-function mutation. J Perinatol 31: 802-803. 76. Ibrahim HR, Aoki T, and Pellegrini A. Strategies for new antimicrobial proteins and peptides: lysozyme and aprotinin as model molecules. Curr Pharm Des 8: 671-693, 2002. 77. Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K, Rowlands DJ, Quadri SK, Bhattacharya S, and Bhattacharya J. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med 18: 759-765. 78. Jamora C and Fuchs E. Intercellular adhesion, signalling and the cytoskeleton. Nat Cell Biol 4: E101-108, 2002. 79. Jansen HM, Sachs AP, and van Alphen L. Predisposing conditions to bacterial infections in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 151: 2073-2080, 1995. 80. Jovanovic S, Crawford RM, Ranki HJ, and Jovanovic A. Large conductance Ca2+-activated K+ channels sense acute changes in oxygen tension in alveolar epithelial cells. Am J Respir Cell Mol Biol 28: 363-372, 2003. 81. Joyce-Brady M and Hiratake J. Inhibiting Glutathione Metabolism in Lung Lining Fluid as a Strategy to Augment Antioxidant Defense. Current enzyme inhibition 7: 71-78, 2011. 82. Kage H, Flodby P, Gao D, Kim YH, Marconett CN, DeMaio L, Kim KJ, Crandall ED, and Borok Z. Claudin 4 knockout mice: normal physiological phenotype with increased susceptibility to lung injury. Am J Physiol Lung Cell Mol Physiol 307: L524-536. 83. Kanai Y, Ochiai A, Shibata T, Oyama T, Ushijima S, Akimoto S, and Hirohashi S. c-erbB-2 gene product directly associates with beta-catenin and plakoglobin. Biochemical and biophysical research communications 208: 1067-1072, 1995. 84. Kerem E, Bistritzer T, Hanukoglu A, Hofmann T, Zhou Z, Bennett W, MacLaughlin E, Barker P, Nash M, Quittell L, Boucher R, and Knowles MR. Pulmonary epithelial sodium-channel dysfunction and excess airway liquid in pseudohypoaldosteronism. N Engl J Med 341: 156-162, 1999.

969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016

85. Kern JA, Robinson RA, Gazdar A, Torney L, and Weiner DB. Mechanisms of p185HER2 expression in human non-small cell lung cancer cell lines. Am J Respir Cell Mol Biol 6: 359-363, 1992. 86. Kheradmand F, Folkesson HG, Shum L, Derynk R, Pytela R, and Matthay MA. Transforming growth factor-alpha enhances alveolar epithelial cell repair in a new in vitro model. The American journal of physiology 267: L728-738, 1994. 87. Kim KJ, Cheek JM, and Crandall ED. Contribution of active Na+ and Cl- fluxes to net ion transport by alveolar epithelium. Respir Physiol 85: 245-256, 1991. 88. King LS and Yasui M. Aquaporins and disease: lessons from mice to humans. Trends in endocrinology and metabolism: TEM 13: 355-360, 2002. 89. Kolb M, Margetts PJ, Anthony DC, Pitossi F, and Gauldie J. Transient expression of IL-1beta induces acute lung injury and chronic repair leading to pulmonary fibrosis. J Clin Invest 107: 1529-1536, 2001. 90. Konstas AA, Bielfeld-Ackermann A, and Korbmacher C. Sulfonylurea receptors inhibit the epithelial sodium channel (ENaC) by reducing surface expression. Pflugers Arch 442: 752-761, 2001. 91. Kunzelmann K, Schreiber R, Kmit A, Jantarajit W, Martins JR, Faria D, Kongsuphol P, Ousingsawat J, and Tian Y. Expression and function of epithelial anoctamins. Experimental physiology 97: 184-192, 2012. 92. Kwong RW, Kumai Y, and Perry SF. Evidence for a role of tight junctions in regulating sodium permeability in zebrafish (Danio rerio) acclimated to ion-poor water. J Comp Physiol B 183: 203-213, 2013. 93. Kwong RW and Perry SF. The tight junction protein claudin-b regulates epithelial permeability and sodium handling in larval zebrafish, Danio rerio. Am J Physiol Regul Integr Comp Physiol 304: R504513, 2013. 94. Lacherade JC, Van De Louw A, Planus E, Escudier E, D'Ortho MP, Lafuma C, Harf A, and Delclaux C. Evaluation of basement membrane degradation during TNF-alpha-induced increase in epithelial permeability. Am J Physiol Lung Cell Mol Physiol 281: L134-143, 2001. 95. LaFemina MJ, Rokkam D, Chandrasena A, Pan J, Bajaj A, Johnson M, and Frank JA. Keratinocyte growth factor enhances barrier function without altering claudin expression in primary alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 299: L724-734, 2010. 96. LaFemina MJ, Sutherland KM, Bentley T, Gonzales LW, Allen L, Chapin CJ, Rokkam D, Sweerus KA, Dobbs LG, Ballard PL, and Frank JA. Claudin-18 deficiency results in alveolar barrier dysfunction and impaired alveologenesis in mice. Am J Respir Cell Mol Biol 51: 550-558. 97. Larre I, Lazaro A, Contreras RG, Balda MS, Matter K, Flores-Maldonado C, Ponce A, FloresBenitez D, Rincon-Heredia R, Padilla-Benavides T, Castillo A, Shoshani L, and Cereijido M. Ouabain modulates epithelial cell tight junction. Proc Natl Acad Sci U S A 107: 11387-11392, 2010. 98. Leroy C, Prive A, Bourret JC, Berthiaume Y, Ferraro P, and Brochiero E. Regulation of ENaC and CFTR expression with K+ channel modulators and effect on fluid absorption across alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 291: L1207-1219, 2006. 99. Leslie CC, McCormick-Shannon K, Shannon JM, Garrick B, Damm D, Abraham JA, and Mason RJ. Heparin-binding EGF-like growth factor is a mitogen for rat alveolar type II cells. Am J Respir Cell Mol Biol 16: 379-387, 1997. 100. Li G, Flodby P, Luo J, Kage H, Sipos A, Gao D, Ji Y, Beard LL, Marconett CN, DeMaio L, Kim YH, Kim KJ, Laird-Offringa IA, Minoo P, Liebler JM, Zhou B, Crandall ED, and Borok Z. Knockout mice reveal key roles for claudin 18 in alveolar barrier properties and fluid homeostasis. Am J Respir Cell Mol Biol 51: 210-222. 101. Li J, Zhuo M, Pei L, Rajagopal M, and Yu AS. Comprehensive cysteine-scanning mutagenesis reveals Claudin-2 pore-lining residues with different intrapore locations. J Biol Chem 289: 6475-6484, 2014.

1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064

102. Liang GH and Weber CR. Molecular aspects of tight junction barrier function. Curr Opin Pharmacol 19C: 84-89, 2014. 103. Liu J, Nethery D, and Kern JA. Neuregulin-1 induces branching morphogenesis in the developing lung through a P13K signal pathway. Exp Lung Res 30: 465-478, 2004. 104. Livraghi A and Randell SH. Cystic fibrosis and other respiratory diseases of impaired mucus clearance. Toxicol Pathol 35: 116-129, 2007. 105. Loi S, de Azambuja E, Pugliano L, Sotiriou C, and Piccart MJ. HER2-overexpressing breast cancer: time for the cure with less chemotherapy? Curr Opin Oncol 23: 547-558, 2011. 106. Londino JD, Lazrak A, Jurkuvenaite A, Collawn JF, Noah JW, and Matalon S. Influenza matrix protein 2 alters CFTR expression and function through its ion channel activity. Am J Physiol Lung Cell Mol Physiol 304: L582-592, 2013. 107. Looney MR, Sartori C, Chakraborty S, James PF, Lingrel JB, and Matthay MA. Decreased expression of both the alpha1- and alpha2-subunits of the Na-K-ATPase reduces maximal alveolar epithelial fluid clearance. Am J Physiol Lung Cell Mol Physiol 289: L104-110, 2005. 108. MacVinish LJ, Cope G, Ropenga A, and Cuthbert AW. Chloride transporting capability of Calu-3 epithelia following persistent knockdown of the cystic fibrosis transmembrane conductance regulator, CFTR. Br J Pharmacol 150: 1055-1065, 2007. 109. Madan P, Rose K, and Watson AJ. Na/K-ATPase beta1 subunit expression is required for blastocyst formation and normal assembly of trophectoderm tight junction-associated proteins. J Biol Chem 282: 12127-12134, 2007. 110. Madtes DK, Busby HK, Strandjord TP, and Clark JG. Expression of transforming growth factoralpha and epidermal growth factor receptor is increased following bleomycin-induced lung injury in rats. Am J Respir Cell Mol Biol 11: 540-551, 1994. 111. Maina JN and West JB. Thin and strong! The bioengineering dilemma in the structural and functional design of the blood-gas barrier. Physiological reviews 85: 811-844, 2005. 112. Mall M, Wissner A, Schreiber R, Kuehr J, Seydewitz HH, Brandis M, Greger R, and Kunzelmann K. Role of K(V)LQT1 in cyclic adenosine monophosphate-mediated Cl(-) secretion in human airway epithelia. Am J Respir Cell Mol Biol 23: 283-289, 2000. 113. Manzanares D, Gonzalez C, Ivonnet P, Chen RS, Valencia-Gattas M, Conner GE, Larsson HP, and Salathe M. Functional apical large conductance, Ca2+-activated, and voltage-dependent K+ channels are required for maintenance of airway surface liquid volume. J Biol Chem 286: 19830-19839, 2011. 114. Mato N, Fujii M, Hakamata Y, Kobayashi E, Sato A, Hayakawa M, Ohto-Ozaki H, Bando M, Ohno S, Tominaga S, and Sugiyama Y. Interleukin-1 receptor-related protein ST2 suppresses the initial stage of bleomycin-induced lung injury. Eur Respir J 33: 1415-1428, 2009. 115. Matthay MA. Resolution of pulmonary edema. Thirty years of progress. Am J Respir Crit Care Med 189: 1301-1308, 2014. 116. McCray PB, Jr. and Bentley L. Human airway epithelia express a beta-defensin. Am J Respir Cell Mol Biol 16: 343-349, 1997. 117. McGrath EE, Lawrie A, Marriott HM, Mercer P, Cross SS, Arnold N, Singleton V, Thompson AA, Walmsley SR, Renshaw SA, Sabroe I, Chambers RC, Dockrell DH, and Whyte MK. Deficiency of tumour necrosis factor-related apoptosis-inducing ligand exacerbates lung injury and fibrosis. Thorax 67: 796803, 2012. 118. Meduri GU, Annane D, Chrousos GP, Marik PE, and Sinclair SE. Activation and regulation of systemic inflammation in ARDS: rationale for prolonged glucocorticoid therapy. Chest 136: 1631-1643, 2009. 119. Meduri GU, Headley S, Kohler G, Stentz F, Tolley E, Umberger R, and Leeper K. Persistent elevation of inflammatory cytokines predicts a poor outcome in ARDS. Plasma IL-1 beta and IL-6 levels are consistent and efficient predictors of outcome over time. Chest 107: 1062-1073, 1995.

1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110

120. Moliva JI, Rajaram MV, Sidiki S, Sasindran SJ, Guirado E, Pan XJ, Wang SH, Ross P, Jr., Lafuse WP, Schlesinger LS, Turner J, and Torrelles JB. Molecular composition of the alveolar lining fluid in the aging lung. Age (Dordrecht, Netherlands) 36: 9633, 2014. 121. Montero JC, Seoane S, Ocana A, and Pandiella A. P-Rex1 participates in Neuregulin-ErbB signal transduction and its expression correlates with patient outcome in breast cancer. Oncogene 30: 10591071, 2011. 122. Mutlu GM, Adir Y, Jameel M, Akhmedov AT, Welch L, Dumasius V, Meng FJ, Zabner J, Koenig C, Lewis ER, Balagani R, Traver G, Sznajder JI, and Factor P. Interdependency of beta-adrenergic receptors and CFTR in regulation of alveolar active Na+ transport. Circ Res 96: 999-1005, 2005. 123. Nakamura Y, Sotozono C, and Kinoshita S. The epidermal growth factor receptor (EGFR): role in corneal wound healing and homeostasis. Exp Eye Res 72: 511-517, 2001. 124. Namiranian K, Brink CD, Goodman JC, Robertson CS, and Bryan RM, Jr. Traumatic brain injury in mice lacking the K channel, TREK-1. J Cereb Blood Flow Metab 31: e1-6, 2011. 125. Nethery DE, Moore BB, Minowada G, Carroll J, Faress JA, and Kern JA. Expression of mutant human epidermal receptor 3 attenuates lung fibrosis and improves survival in mice. J Appl Physiol 99: 298-307, 2005. 126. Nilius B and Honore E. Sensing pressure with ion channels. Trends in neurosciences 35: 477-486, 2012. 127. Noel J, Sandoz G, and Lesage F. Molecular regulations governing TREK and TRAAK channel functions. Channels (Austin, Tex) 5: 402-409, 2011. 128. Ochiai A, Akimoto S, Kanai Y, Shibata T, Oyama T, and Hirohashi S. c-erbB-2 gene product associates with catenins in human cancer cells. Biochemical and biophysical research communications 205: 73-78, 1994. 129. Olayioye MA, Graus-Porta D, Beerli RR, Rohrer J, Gay B, and Hynes NE. ErbB-1 and ErbB-2 acquire distinct signaling properties dependent upon their dimerization partner. Mol Cell Biol 18: 50425051, 1998. 130. Olivera DS, Boggs SE, Beenhouwer C, Aden J, and Knall C. Cellular mechanisms of mainstream cigarette smoke-induced lung epithelial tight junction permeability changes in vitro. Inhal Toxicol 19: 1322, 2007. 131. Olver RE and Strang LB. Ion fluxes across the pulmonary epithelium and the secretion of lung liquid in the foetal lamb. J Physiol 241: 327-357, 1974. 132. Ortiz LA, Dutreil M, Fattman C, Pandey AC, Torres G, Go K, and Phinney DG. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc Natl Acad Sci U S A 104: 11002-11007, 2007. 133. Park WY, Goodman RB, Steinberg KP, Ruzinski JT, Radella F, 2nd, Park DR, Pugin J, Skerrett SJ, Hudson LD, and Martin TR. Cytokine balance in the lungs of patients with acute respiratory distress syndrome. Am J Respir Crit Care Med 164: 1896-1903, 2001. 134. Parker D and Prince A. Innate immunity in the respiratory epithelium. Am J Respir Cell Mol Biol 45: 189-201. 135. Peters DM, Vadasz I, Wujak L, Wygrecka M, Olschewski A, Becker C, Herold S, Papp R, Mayer K, Rummel S, Brandes RP, Gunther A, Waldegger S, Eickelberg O, Seeger W, and Morty RE. TGF-beta directs trafficking of the epithelial sodium channel ENaC which has implications for ion and fluid transport in acute lung injury. Proc Natl Acad Sci U S A 111: E374-383. 136. Planes C, Randrianarison NH, Charles RP, Frateschi S, Cluzeaud F, Vuagniaux G, Soler P, Clerici C, Rossier BC, and Hummler E. ENaC-mediated alveolar fluid clearance and lung fluid balance depend on the channel-activating protease 1. EMBO molecular medicine 2: 26-37, 2010.

1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158

137. Pohl C, Hermanns MI, Uboldi C, Bock M, Fuchs S, Dei-Anang J, Mayer E, Kehe K, Kummer W, and Kirkpatrick CJ. Barrier functions and paracellular integrity in human cell culture models of the proximal respiratory unit. Eur J Pharm Biopharm 72: 339-349, 2009. 138. Privalsky ML, Sealy L, Bishop JM, McGrath JP, and Levinson AD. The product of the avian erythroblastosis virus erbB locus is a glycoprotein. Cell 32: 1257-1267, 1983. 139. Puddicombe SM, Polosa R, Richter A, Krishna MT, Howarth PH, Holgate ST, and Davies DE. Involvement of the epidermal growth factor receptor in epithelial repair in asthma. Faseb J 14: 13621374, 2000. 140. Pugin J, Ricou B, Steinberg KP, Suter PM, and Martin TR. Proinflammatory activity in bronchoalveolar lavage fluids from patients with ARDS, a prominent role for interleukin-1. Am J Respir Crit Care Med 153: 1850-1856, 1996. 141. Rajasekaran AK and Rajasekaran SA. Role of Na-K-ATPase in the assembly of tight junctions. Am J Physiol Renal Physiol 285: F388-396, 2003. 142. Rajasekaran SA, Barwe SP, Gopal J, Ryazantsev S, Schneeberger EE, and Rajasekaran AK. Na-KATPase regulates tight junction permeability through occludin phosphorylation in pancreatic epithelial cells. Am J Physiol Gastrointest Liver Physiol 292: G124-133, 2007. 143. Rajasekaran SA, Hu J, Gopal J, Gallemore R, Ryazantsev S, Bok D, and Rajasekaran AK. Na,KATPase inhibition alters tight junction structure and permeability in human retinal pigment epithelial cells. Am J Physiol Cell Physiol 284: C1497-1507, 2003. 144. Rajasekaran SA, Palmer LG, Moon SY, Peralta Soler A, Apodaca GL, Harper JF, Zheng Y, and Rajasekaran AK. Na,K-ATPase activity is required for formation of tight junctions, desmosomes, and induction of polarity in epithelial cells. Mol Biol Cell 12: 3717-3732, 2001. 145. Raleigh DR, Boe DM, Yu D, Weber CR, Marchiando AM, Bradford EM, Wang Y, Wu L, Schneeberger EE, Shen L, and Turner JR. Occludin S408 phosphorylation regulates tight junction protein interactions and barrier function. J Cell Biol 193: 565-582, 2011. 146. Ramsingh R, Grygorczyk A, Solecki A, Cherkaoui LS, Berthiaume Y, and Grygorczyk R. Cell deformation at the air-liquid interface induces Ca2+-dependent ATP release from lung epithelial cells. Am J Physiol Lung Cell Mol Physiol 300: L587-595, 2011. 147. Ress A and Moelling K. The PDZ protein erbin modulates beta-catenin-dependent transcription. Eur Surg Res 41: 284-289, 2008. 148. Riese DJ, 2nd, van Raaij TM, Plowman GD, Andrews GC, and Stern DF. The cellular response to neuregulins is governed by complex interactions of the erbB receptor family. Mol Cell Biol 15: 57705776, 1995. 149. Roan E, Waters CM, Teng B, Ghosh M, and Schwingshackl A. The 2-pore domain potassium channel TREK-1 regulates stretch-induced detachment of alveolar epithelial cells. PLoS One 9: e89429, 2014. 150. Roberts CM, Cairns D, Bryant DH, Burke WM, Yeates M, Blake H, Penny R, Shelley L, Zaunders JJ, and Breit SN. Changes in epithelial lining fluid albumin associated with smoking and interstitial lung disease. Eur Respir J 6: 110-115, 1993. 151. Rock JR, O'Neal WK, Gabriel SE, Randell SH, Harfe BD, Boucher RC, and Grubb BR. Transmembrane protein 16A (TMEM16A) is a Ca2+-regulated Cl- secretory channel in mouse airways. J Biol Chem 284: 14875-14880, 2009. 152. Rokkam D, Lafemina MJ, Lee JW, Matthay MA, and Frank JA. Claudin-4 levels are associated with intact alveolar fluid clearance in human lungs. Am J Pathol 179: 1081-1087. 153. Ryan RM, Mineo-Kuhn MM, Kramer CM, and Finkelstein JN. Growth factors alter neonatal type II alveolar epithelial cell proliferation. The American journal of physiology 266: L17-22, 1994. 154. Sasaki S, Yui N, and Noda Y. Actin directly interacts with different membrane channel proteins and influences channel activities: AQP2 as a model. Biochim Biophys Acta 1838: 514-520, 2014.

1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206

155. Schade B, Lesurf R, Sanguin-Gendreau V, Bui T, Deblois G, O'Toole SA, Millar EKA, Zardawi SJ, Lopez-Knowles E, Sutherland RL, Giguère V, Kahn M, Hallett M, and Muller WJ. β-Catenin Signaling Is a Critical Event in ErbB2-Mediated Mammary Tumor Progression. Cancer Research 73: 4474-4487, 2013. 156. Schmidt C, Wiedmann F, Schweizer PA, Katus HA, and Thomas D. [Cardiac two-pore-domain potassium channels (K2P): Physiology, pharmacology, and therapeutic potential]. Deutsche medizinische Wochenschrift (1946) 137: 1654-1658, 2012. 157. Schneeberger EE and Lynch RD. The tight junction: a multifunctional complex. Am J Physiol Cell Physiol 286: C1213-1228, 2004. 158. Schreiber R, Hopf A, Mall M, Greger R, and Kunzelmann K. The first-nucleotide binding domain of the cystic-fibrosis transmembrane conductance regulator is important for inhibition of the epithelial Na+ channel. Proc Natl Acad Sci U S A 96: 5310-5315, 1999. 159. Schreiber R, Nitschke R, Greger R, and Kunzelmann K. The cystic fibrosis transmembrane conductance regulator activates aquaporin 3 in airway epithelial cells. J Biol Chem 274: 11811-11816, 1999. 160. Schroeder JA, Adriance MC, McConnell EJ, Thompson MC, Pockaj B, and Gendler SJ. ErbB-betacatenin complexes are associated with human infiltrating ductal breast and murine mammary tumor virus (MMTV)-Wnt-1 and MMTV-c-Neu transgenic carcinomas. J Biol Chem 277: 22692-22698, 2002. 161. Schwingshackl A, Teng B, Ghosh M, Lim KG, Tigyi G, Narayanan D, Jaggar JH, and Waters CM. Regulation of interleukin-6 secretion by the two-pore-domain potassium channel Trek-1 in alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 304: L276-286. 162. Schwingshackl A, Teng B, Ghosh M, and Waters CM. Regulation of Monocyte Chemotactic Protein-1 secretion by the Two-Pore-Domain Potassium (K2P) channel TREK-1 in human alveolar epithelial cells. Am J Transl Res 5: 530-542, 2013. 163. Schwingshackl A, Teng B, Ghosh M, West AN, Makena P, Gorantla V, Sinclair SE, and Waters CM. Regulation and function of the two-pore-domain (K2P) potassium channel Trek-1 in alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 302: L93-L102, 2012. 164. Schwingshackl A, Teng B, Makena P, Ghosh M, Sinclair SE, Luellen C, Balasz L, Rovnaghi C, Bryan RM, Lloyd EE, Fitzpatrick E, Saravia JS, Cormier SA, and Waters CM. Deficiency of the two-poredomain potassium channel TREK-1 promotes hyperoxia-induced lung injury. Crit Care Med 42: e692-701. 165. Shaykhiev R, Zuo WL, Chao I, Fukui T, Witover B, Brekman A, and Crystal RG. EGF shifts human airway basal cell fate toward a smoking-associated airway epithelial phenotype. Proc Natl Acad Sci U S A 110: 12102-12107. 166. Shen L, Weber CR, Raleigh DR, Yu D, and Turner JR. Tight junction pore and leak pathways: a dynamic duo. Annu Rev Physiol 73: 283-309. 167. Shen L, Weber CR, Raleigh DR, Yu D, and Turner JR. Tight junction pore and leak pathways: a dynamic duo. Annu Rev Physiol 73: 283-309, 2011. 168. Shen L, Weber CR, and Turner JR. The tight junction protein complex undergoes rapid and continuous molecular remodeling at steady state. J Cell Biol 181: 683-695, 2008. 169. Shin K, Fogg VC, and Margolis B. Tight junctions and cell polarity. Annu Rev Cell Dev Biol 22: 207-235, 2006. 170. Shomori K, Ochiai A, Akimoto S, Ino Y, Shudo K, Ito H, and Hirohashi S. Tyrosinephosphorylation of the 12th armadillo-repeat of beta-catenin is associated with cadherin dysfunction in human cancer. International journal of oncology 35: 517-524, 2009. 171. Sidhaye VK, Chau E, Breysse PN, and King LS. Septin-2 mediates airway epithelial barrier function in physiologic and pathologic conditions. Am J Respir Cell Mol Biol 45: 120-126. 172. Sidhaye VK, Chau E, Srivastava V, Sirimalle S, Balabhadrapatruni C, Aggarwal NR, D'Alessio FR, Robinson DN, and King LS. A novel role for aquaporin-5 in enhancing microtubule organization and stability. PLoS One 7: e38717.

1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254

173. Sidhaye VK, Schweitzer KS, Caterina MJ, Shimoda L, and King LS. Shear stress regulates aquaporin-5 and airway epithelial barrier function. Proc Natl Acad Sci U S A 105: 3345-3350, 2008. 174. Siler TM, Swierkosz JE, Hyers TM, Fowler AA, and Webster RO. Immunoreactive interleukin-1 in bronchoalveolar lavage fluid of high-risk patients and patients with the adult respiratory distress syndrome. Exp Lung Res 15: 881-894, 1989. 175. Simard CF, Bergeron MJ, Frenette-Cotton R, Carpentier GA, Pelchat ME, Caron L, and Isenring P. Homooligomeric and heterooligomeric associations between K+-Cl- cotransporter isoforms and between K+-Cl- and Na+-K+-Cl- cotransporters. J Biol Chem 282: 18083-18093, 2007. 176. Singh PK, Jia HP, Wiles K, Hesselberth J, Liu L, Conway BA, Greenberg EP, Valore EV, Welsh MJ, Ganz T, Tack BF, and McCray PB, Jr. Production of beta-defensins by human airway epithelia. Proc Natl Acad Sci U S A 95: 14961-14966, 1998. 177. Singha O, Kengkoom K, Chaimongkolnukul K, Cherdyu S, Pongponratn E, Ketjareon T, Panavechkijkul Y, and Ampawong S. Pulmonary edema due to oral gavage in a toxicological study related to aquaporin-1, -4 and -5 expression. Journal of toxicologic pathology 26: 283-291, 2013. 178. Sisson TH, Mendez M, Choi K, Subbotina N, Courey A, Cunningham A, Dave A, Engelhardt JF, Liu X, White ES, Thannickal VJ, Moore BB, Christensen PJ, and Simon RH. Targeted injury of type II alveolar epithelial cells induces pulmonary fibrosis. Am J Respir Crit Care Med 181: 254-263, 2010. 179. Skryma R, Prevarskaya N, Gkika D, and Shuba Y. From urgency to frequency: facts and controversies of TRPs in the lower urinary tract. Nature reviews Urology 8: 617-630, 2011. 180. Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE, Levin WJ, Stuart SG, Udove J, Ullrich A, and et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 244: 707-712, 1989. 181. Slutsky AS and Ranieri VM. Ventilator-induced lung injury. The New England journal of medicine 370: 980, 2014. 182. Smith LS, Zimmerman JJ, and Martin TR. Mechanisms of acute respiratory distress syndrome in children and adults: a review and suggestions for future research. Pediatric critical care medicine : a journal of the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies 14: 631-643, 2013. 183. Solymosi EA, Kaestle-Gembardt SM, Vadasz I, Wang L, Neye N, Chupin CJ, Rozowsky S, Ruehl R, Tabuchi A, Schulz H, Kapus A, Morty RE, and Kuebler WM. Chloride transport-driven alveolar fluid secretion is a major contributor to cardiogenic lung edema. Proc Natl Acad Sci U S A 110: E2308-2316, 2013. 184. Stoll S, Garner W, and Elder J. Heparin-binding ligands mediate autocrine epidermal growth factor receptor activation In skin organ culture. J Clin Invest 100: 1271-1281, 1997. 185. Strieter RM and Kunkel SL. Acute lung injury: the role of cytokines in the elicitation of neutrophils. Journal of investigative medicine : the official publication of the American Federation for Clinical Research 42: 640-651, 1994. 186. Takeyama K, Dabbagh K, Lee HM, Agusti C, Lausier JA, Ueki IF, Grattan KM, and Nadel JA. Epidermal growth factor system regulates mucin production in airways. Proc Natl Acad Sci U S A 96: 3081-3086, 1999. 187. Takeyama K, Fahy JV, and Nadel JA. Relationship of epidermal growth factor receptors to goblet cell production in human bronchi. Am J Respir Crit Care Med 163: 511-516, 2001. 188. Takeyama K, Jung B, Shim JJ, Burgel PR, Dao-Pick T, Ueki IF, Protin U, Kroschel P, and Nadel JA. Activation of epidermal growth factor receptors is responsible for mucin synthesis induced by cigarette smoke. Am J Physiol Lung Cell Mol Physiol 280: L165-172, 2001. 189. Terakado M, Gon Y, Sekiyama A, Takeshita I, Kozu Y, Matsumoto K, Takahashi N, and Hashimoto S. The Rac1/JNK pathway is critical for EGFR-dependent barrier formation in human airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 300: L56-63, 2011.

1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302

190. Tessier GJ, Traynor TR, Kannan MS, and O'Grady SM. Mechanisms of sodium and chloride transport across equine tracheal epithelium. Am J Physiol 259: L459-467, 1990. 191. Thorley AJ and Tetley TD. Pulmonary epithelium, cigarette smoke, and chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2: 409-428, 2007. 192. Trinh NT, Prive A, Kheir L, Bourret JC, Hijazi T, Amraei MG, Noel J, and Brochiero E. Involvement of KATP and KvLQT1 K+ channels in EGF-stimulated alveolar epithelial cell repair processes. Am J Physiol Lung Cell Mol Physiol 293: L870-882, 2007. 193. van der Geer P, Hunter T, and Lindberg RA. Receptor protein-tyrosine kinases and their signal transduction pathways. Annu Rev Cell Biol 10: 251-337, 1994. 194. Van Driessche W, Kreindler JL, Malik AB, Margulies S, Lewis SA, and Kim KJ. Interrelations/cross talk between transcellular transport function and paracellular tight junctional properties in lung epithelial and endothelial barriers. Am J Physiol Lung Cell Mol Physiol 293: L520-524, 2007. 195. Van Itallie CM and Anderson JM. The molecular physiology of tight junction pores. Physiology (Bethesda) 19: 331-338, 2004. 196. Van Itallie CM, Fanning AS, and Anderson JM. Reversal of charge selectivity in cation or anionselective epithelial lines by expression of different claudins. Am J Physiol Renal Physiol 285: F1078-1084, 2003. 197. Van Itallie CM, Holmes J, Bridges A, Gookin JL, Coccaro MR, Proctor W, Colegio OR, and Anderson JM. The density of small tight junction pores varies among cell types and is increased by expression of claudin-2. J Cell Sci 121: 298-305, 2008. 198. Verkman AS. Role of aquaporins in lung liquid physiology. Respiratory physiology & neurobiology 159: 324-330, 2007. 199. Verkman AS, Matthay MA, and Song Y. Aquaporin water channels and lung physiology. Am J Physiol Lung Cell Mol Physiol 278: L867-879, 2000. 200. Vermeer PD, Einwalter LA, Moninger TO, Rokhlina T, Kern JA, Zabner J, and Welsh MJ. Segregation of receptor and ligand regulates activation of epithelial growth factor receptor. Nature 422: 322-326, 2003. 201. Walker RA. The erbB/HER type 1 tyrosine kinase receptor family. J Pathol 185: 234-235, 1998. 202. Wang JH and Thampatty BP. An introductory review of cell mechanobiology. Biomechanics and modeling in mechanobiology 5: 1-16, 2006. 203. Watanabe H, Murakami M, Ohba T, Takahashi Y, and Ito H. TRP channel and cardiovascular disease. Pharmacology & therapeutics 118: 337-351, 2008. 204. Waters CM, Ridge KM, Sunio G, Venetsanou K, and Sznajder JI. Mechanical stretching of alveolar epithelial cells increases Na(+)-K(+)-ATPase activity. J Appl Physiol 87: 715-721, 1999. 205. Wilson SM, Olver RE, and Walters DV. Developmental regulation of lumenal lung fluid and electrolyte transport. Respiratory physiology & neurobiology 159: 247-255, 2007. 206. Wispe JR, Clark JC, Warner BB, Fajardo D, Hull WE, Holtzman RB, and Whitsett JA. Tumor necrosis factor-alpha inhibits expression of pulmonary surfactant protein. J Clin Invest 86: 1954-1960, 1990. 207. Wray C, Mao Y, Pan J, Chandrasena A, Piasta F, and Frank JA. Claudin-4 augments alveolar epithelial barrier function and is induced in acute lung injury. Am J Physiol Lung Cell Mol Physiol 297: L219-227, 2009. 208. Yang C, Liu Y, Lemmon MA, and Kazanietz MG. Essential role for Rac in heregulin beta1 mitogenic signaling: a mechanism that involves epidermal growth factor receptor and is independent of ErbB4. Mol Cell Biol 26: 831-842, 2006. 209. Yarden Y and Sliwkowski MX. Untangling the ErbB signalling network. Nature reviews Molecular cell biology 2: 127-137, 2001.

1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343

210. Yin J, Lu J, and Yu FS. Role of small GTPase Rho in regulating corneal epithelial wound healing. Invest Ophthalmol Vis Sci 49: 900-909, 2008. 211. Yin J and Yu FS. ERK1/2 mediate wounding- and G-protein-coupled receptor ligands-induced EGFR activation via regulating ADAM17 and HB-EGF shedding. Invest Ophthalmol Vis Sci 50: 132-139, 2009. 212. Zhao F, Dong L, Cheng L, Zeng Q, and Su F. Effects of acute mechanical stretch on the expression of mechanosensitive potassium channel TREK-1 in rat left ventricle. J Huazhong Univ Sci Technolog Med Sci 27: 385-387, 2007. 213. Zhao KQ, Xiong G, Wilber M, Cohen NA, and Kreindler JL. A role for two-pore K(+) channels in modulating Na(+) absorption and Cl(-) secretion in normal human bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol 302: L4-L12, 2012. 214. Zhu H, Zhang G, Wang Y, Xu N, He S, Zhang W, Chen M, Liu M, Quan L, Bai J, and Xu N. Inhibition of ErbB2 by Herceptin reduces survivin expression via the ErbB2–β-catenin/TCF4-survivin pathway in ErbB2-overexpressed breast cancer cells. Cancer Science 101: 1156-1162, 2010. 215. Zieske JD, Takahashi H, Hutcheon AE, and Dalbone AC. Activation of epidermal growth factor receptor during corneal epithelial migration. Invest Ophthalmol Vis Sci 41: 1346-1355, 2000.

Fig.1. The EGFR is restricted to the basal surface of the airway epithelial cells. This figure serves to highlight how the integrity of the apicoadherens junction can regulate epithelial cell signaling. Under conditions of low paracellular permeability created by the cytoskeleton and cell-cell contacts, there is segregation of EGF ligand and receptor, thereby inhibiting constant cellular signaling. In conditions of increased paracellular permeability, EGF can bind its receptor and activate its pathway where phosphorylated ERK serves as readout of EGFR activation. Thus, the epithelial barrier can modulate epithelial signaling pathways. Fig. 2: We propose that hyperoxia, mechanical stretch and inflammatory cytokines alter the regulation of K2P channel function. This can then impact on cytokine secretion and epithelial barrier function and this dysregulation of these proteins can contribute to the pathogenesis of ALI through downstream. Fig. 3. Cytokine activation can lead to increased free NRG-1. Initial disruption of the epithelial barrier allows for NRG-1-HER3-HER2 activation. The activation of this complex can leads to β-catenin tyrosine phosphorylation, which further propogates disruption of adherens junction, and alteration of the pulmonary epithelial barrier.

Fig. 1

Fig. 2

IL-1β NRG-1 Free NRG-1

Pulmonary Epithelial Cell IL-1R

Occludin ADAM17

ZO-1

Claudin

JAM/CAR HER3

HER2 Epithelial Barrier Disruption

P P

PP P Actin β-catenin

E-cadherin

EGFR

Fig. 3

Pulmonary epithelial barrier function: some new players and mechanisms.

The pulmonary epithelium serves as a barrier to prevent access of the inspired luminal contents to the subepithelium. In addition, the epithelium dict...
771KB Sizes 0 Downloads 9 Views