Review Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Respirol. Nov 28, 2013; 3(3): 48-56
Published online Nov 28, 2013. doi: 10.5320/wjr.v3.i3.48
Von Hippel-Lindau protein and respiratory diseases
Tianji Chen, Miranda Sun, Guofei Zhou, Department of Pediatrics, University of Illinois at Chicago, University of Illinois Hospital and Health Sciences System, Chicago, IL 60612, United States
Author contributions: Zhou G conceived the study; Chen T, Sun M, and Zhou G wrote the manuscript.
Supported by Grants from Pulmonary Hypertension Association and American Lung Association to Dr. Guofei Zhou
Correspondence to: Guofei Zhou, PhD, Department of Pediatrics, University of Illinois at Chicago, University of Illinois Hospital and Health Sciences System, 840 S. Wood Street, M/C 856, Ste 1206, Chicago, IL 60612, United States. guofei@uic.edu
Telephone: +1-312-3550073 Fax: +1-312-9968204
Received: June 27, 2013
Revised: July 9, 2013
Accepted: July 17, 2013
Published online: November 28, 2013

Abstract

Von Hippel-Lindau protein (pVHL) was first identified as a tumor suppressor gene as mutations in the VHL gene predispose individuals to systemic benign or malignant tumors and cysts in many organs, including renal cell carcinoma of the clear-cell type and hemangioblastoma. Although pVHL is best known to act as a component of ubiquitin protein ligase for the proteasomal degradation of hypoxia inducible factor (HIF)-α, pVHL also interacts with extracellular matrix proteins and cytoskeleton, regulating extracellular matrix assembly, cell signaling, and many other cellular functions. Recent studies suggest that pVHL contributes to many lung diseases, including pulmonary arterial hypertension, lung cancer, pulmonary fibrosis, and acute respiratory distress syndrome. Mutation or loss of function of pVHL activates HIF and induced expression of vascular endothelial growth factor, endothelin-1, and FoxM1, leading to pulmonary arterial hypertension. Loss of pVHL in lung cancer cells promotes epithelial-mesenchymal transition and cancer migration and invasion while decreasing lung cancer cell proliferation and colonization. In patients of idiopathic pulmonary fibrosis, elevated expression of pVHL induces expression of fibronectin/integrin α5β1/focal adhesion kinase signaling, resulting in fibroproliferation and fibrosis. In alveolar epithelial cells, pVHL mediates Na-K-ATPase degradation in an HIF independent pathway, causing decreased edema clearance during hypoxia. These studies suggest that pVHL plays key roles in the pathogenesis of many lung diseases, and further investigations are warranted to elucidate the underlying molecular mechanisms.

Key Words: Von Hippel-Lindau protein, Lung cancer, Pulmonary fibrosis, Pulmonary hypertension

Core tip: Although von Hippel-Lindau protein (pVHL) was first identified as a tumor suppressor and is best known as a component of ubiquitin protein ligase for the proteasomal degradation of hypoxia inducible factor (HIF)-α, recent studies suggest that pVHL contributes to many lung diseases in both HIF-dependent and HIF-independent pathways. Loss of pVHL promotes pulmonary arterial hypertension via activation of HIF. In lung cancer, loss of pVHL promotes epithelial-mesenchymal transition and cancer migration and invasion while decreasing cell proliferation and colonization. pVHL also induces fibronectin/integrin α5β1/focal adhesion kinase signaling to facilitate fibrogenesis. pVHL mediates Na-K-ATPase degradation to cause decreased edema clearance during hypoxia.



VON HIPPEL-LINDAU PROTEIN

The von Hippel-Lindau (VHL) gene was first identified in patients with von Hippel-Lindau syndrome, an autosomal dominant disease with an incidence of one in 36000 births[1]. Germline mutation of the VHL gene, which is located in human chromosome 3p25[2], predisposes individuals to benign or malignant tumors and cysts in many organ systems and causes clear-cell renal cell carcinoma, hemangioblastoma, pheochromocytoma in adrenal glands, pancreas tumors, endolymphatic sac tumors of middle ear, and epididymal cystadenomas in testes. There are also findings that VHL contributes to liver and pulmonary hemangiomas[3]. Individuals with VHL disease usually have an inherited mutant copy of the VHL gene and an inactivated wild-type allele through somatic mutation or hypermethylation.

pVHL regulates HIF activity

The VHL gene, cloned in 1993[4], consists of three exons and produces two alternatively spliced mRNAs, translating into two proteins pVHL30 and pVHL19[5,6]. The larger protein contains 213 amino acids and the shorter is internally translated and is missing the first 53 amino acids. VHL30 is found in the nuclear, cytosolic and membrane fractions while VHL19 localizes mostly in the nucleus. The VHL protein contains two domains, α and β domain. The α domain serves as an elongin C binding site while the β domain acts as a substrate-recognition/docking site. VHL is found in a multiple-protein complex with Elongin B, Elongin C, Cullin (Cul) 2, and Rbx 1 (also known as ROC1/Hrt1), forming an ubiquitin protein ligase (E3) complex named VEC[7,8]. Elongin C brings VHL and Cul2 together. Cul2 is associated with Rbx1, which recognizes a cognate ubiquitin conjugating enzyme (E2).

pVHL is best known as a component of E3 for the proteasomal degradation of hypoxia inducible factor (HIF)-1/2α[9,10]. In normoxic conditions, HIF-α is hydroxylated at the conserved proline residues within the oxygen-dependent degradation domain by the prolyl-hydroxylase domain proteins[9-11]. The prolyl-hydroxylated HIF-α is recognized by pVHL, leading to poly-ubiquitination and degradation[9,10]. During hypoxia, the prolyl -hydroxylases are inhibited, HIF-α is not hydroxylated and is unable to bind pVHL, leading to its stabilization. Stabilized HIF-α forms a heterodimer with a HIF-β family member and acts as a transcriptional factor to activate expression of downstream genes that contain the HIF-responsive elements in their promoters[12]. Many of these genes are involved in promoting adaption to hypoxic conditions, including the angiogenic genes vascular endothelial growth factor (VEGF) and erythropoietin (EPO), glycolysis-involving gene phosphoglycerate kinase, and glucose transporter Glut-1, etc.[12,13].

pVHL was identified as a tumor suppressor because the VHL gene mutation was shown to be associated with tumors in the kidney[3,14]. Iliopoulos et al[14] and Kondo et al[15] demonstrated that reintroduction of the wild-type VHL into the VHL-mutated renal carcinoma cell line 768-O inhibited the tumor formation in nude mice after injection of the renal carcinoma cells. HIF activation is a crucial for carcinogenesis in absence of pVHL since HIF induces expression of angiogenic factors VEGF, EPO and platelet-derived growth factor[3,12,16]. pVHL also regulates a proliferative response to hypoxia since the loss of pVHL leads to constitutively elevated Cyclin D1 and abnormal proliferation rate of the renal carcinoma cells[17,18]. Additionally, the pVHL mutant is known to increase Akt-mTOR signaling[19], induce fibroblast growth factor receptor signaling[20,21], disrupt cilia formation[22], and downregulate p53[23] to promote cancer initiation and progression.

pVHL regulates extracellular matrix assembly

In the other characterized pVHL pathway, fibronectin interacts with pVHL in cells and co-localizes with a fraction of pVHL on the endoplasmic reticulum (ER)[24]. Moreover, renal carcinoma cells with loss of pVHL have a defective assembly of extracellular fibronectin matrix[24]. These data support a direct role of pVHL in fibronectin matrix assembly[24]. Although pVHL null cells have intact extracellular matrix (ECM) expression and secretion levels[25,26], pVHL deficient 786-O renal cancer cells are unable to organize an adequate matrix even in the presence of an excess of exogenous fibronectin, suggesting that proper assembly of fibronectin matrix requires adequate interaction between fibronectin and its receptor[27]. Further studies demonstrated that association of fibronectin with αvβ1 integrin is crucial for the fibronectin matrix assembly. In cells with pVHL expression, αv integrin forms “patch-like” focal contacts and relocates to the intercellular junctions where αv and β1 integrin form large fibrillar adhesions and anchor firmly to the fibronectin substrate. In the absence of pVHL, αv integrin focal contacts remain unchanged; however, they are unable to assemble β1 fibrillar adhesions[27]. Activation of β1 integrin by exogenous divalent cations or activating antibodies partially restores the capability of VHL null cells to assemble β1 fibrillar adhesions and fibronectin fibers[27]. These studies suggest that pVHL is an important regulator of αvβ1 integrins and is essential for the formation of β1 fibrillar adhesions and the organization of extracellular fibronectin.

The interaction with pVHL is also necessary for collagen to be assembled into the ECM. Failure of pVHL to interact with collagen correlates with the loss of collagen network and collagen remodeling in vitro and in vivo[24,28,29]. Although pVHL can interact with type I, II, IV, and V collagen in denatured conditions, the most specific binding occurs with type IV collagen, specifically the collagen IV α2 (COL4A2)[28,29]. The pVHL-COL4A2 interaction occurs on the ER where the N-terminal tail of COL4A2 protrudes from the ER lumen into the cytosol to associate with pVHL. This association requires a collagenous domain of Gly-X-Y triplets in the full-length α-chains of collagen (X and Y can be any amino acid residue but are commonly proline or hydroxyproline). However, hydroxylation of the N-terminal domain leads to disassociation of collagen from pVHL, and collagen folds into the matured triple helical conformation[29]. Taken together, these findings suggest that pVHL can bind directly to a variety of collagen chains and that this association is critical for collagen matrix assembly.

pVHL and cytoskeleton dynamics and epithelial cilia maintenance

Accumulating evidence suggests a novel function of pVHL in cytoskeleton dynamics and epithelial cilia maintenance[30,31]. Hergovich et al[32,33] reported that pVHL associates with microtubules and protects them from depolymerization. Amino acids 95-123 of pVHL, particularly point mutations such as pVHL (Y98H) and pVHL (Y112H), are critical for the pVHL-microtubule interaction[32]. The pVHL-microtubule interaction appears to be indirect and is mediated by microtubule motor Kinesin-2, which is responsible for the transport of pVHL to the cell periphery along microtubules, which also stabilizes them[34,35]. Accordingly, pVHL affects the normal function of primary cilium, a microtubule-based cellular sensory organelle in the kidney. pVHL functions to stabilize microtubules in the axoneme and localizes to primary cilia, but loss of pVHL alone does not affect primary cilia structure in primary cells[36]. However, inactivation of both the pVHL and GSK3β leads to loss of cilia, suggesting that mutation of pVHL may sensitize cells to lose their primary cilia and promote the formation of cysts in the kidney[36]. In absence of functional pVHL, loss of GSK-3β activates Akt and extracellular signal-regulated kinase, inducing epithelial cell proliferation and kidney cyst formation[37]. Conversely, mice with the double deletion of VHL and phosphatase and tensin homolog, which mimics Akt activity, display cilia loss and cyst formation in the kidney[37]. These findings indicate that, although loss of pVHL function alone is insufficient to cause uncontrolled cellular proliferation and cyst formation, additional signaling such as activation of Akt and inactivation GSK-3β may allow cyst formation.

pVHL also regulates actin cytoskeletal organization and cell motility. Overexpression of pVHL translocates vinculin from the cytoplasm to the cell membrane and induces focal adhesion formation and adhesion[38]. In contrast, overexpression of pVHL also increases stress fibers which exhibit a spreading morphology, leading to reduced cell motility[38]. These results suggest that the loss of pVHL may promote cancer progression via destabilized actin organization and increased motility in cancer cells. Additional studies suggest that loss of pVHL results in the loss of Brick1 (Brk1), a component of the Wave/Scar pathway that regulates branched nucleation of actin fibers. Consistently, suppression of Brk1 causes abnormal vinculin distribution, loss of Arp2/3 and Wave proteins at the cellular protrusions, and abnormal actin stress fiber formation. Furthermore, suppression of Brk1 decreases proliferation, migration, and invasion in renal carcinoma cells[39]. A recent genetic study shows that germline and somatic mutations in VHL is associated with loss of HSPC300 gene, a regulator of actin dynamics and cytoskeleton organization. Depletion of HSPC300 causes cytoskeleton abnormalities and cytokinesis arrest in tumor cells[40]. The underlying mechanism of how pVHL regulates Brk1 and HSPC300, however, remains unclear.

VHL AND PULMONARY ARTERIAL HYPERTENSION

Pulmonary artery hypertension (PAH) is a devastating disease that results in a progressive increase in pulmonary vascular resistance, right ventricular failure, and ultimately death[41,42]. Despite recent advances in management of PAH, there is currently no cure for PAH. PAH is characterized by pulmonary arterial remodeling that includes vascular cell proliferation[41,42]. Hypoxia is a well established stimulus for the induction of pulmonary hypertension (PH) in several animal models that exhibit pulmonary artery smooth muscle cell proliferation and de-differentiation after exposure to hypoxia[43]. HIF is a master transcription factor that regulates cellular adaptation in hypoxia[44,45] and has been implicated in PH[46,47]. Furthermore, inhibition of HIF by Digoxin prevents and reverses hypoxia-induced PH[48].

Distinct from the classic VHL-associated inherited cancer syndrome, Chuvash polycythemia is defined as a new form of VHL-associated disease which is caused by the homozygosity for the C598T mutation of the VHL gene[49]. The C598T VHL mutation increases the HIF-α level and the expression of several HIF target genes including EPO and VEGF, leading to the development of polycythemia[49]. Smith et al[50] reported that a small group of patients with Chuvash polycythemia showed striking abnormalities in the respiratory and pulmonary vascular systems. Compared to control individuals, Chuvash polycythemia patients displayed elevated basal ventilation and pulmonary vascular tone as well as increased ventilatory, pulmonary vasoconstrictive, and heart rate responses to acute hypoxia, suggesting a role of pVHL/HIF signaling in calibration and homeostasis of the respiratory and cardiovascular system[50]. Other studies have also shown that patients with Chuvash polycythemia had a higher systolic pulmonary artery pressure and plasma concentration of endothelin-1 and VEGF than control individuals[51,52], suggesting that, in Chuvash polycythemia patients, the VHL mutation may activate HIF and upregulate endothelin-1 and VEGF, leading to pulmonary hypertension.

In a strain of mice harboring the C598T VHL mutation (VhlR/R mice) as a model for Chuvash disease, Hickey et al[53] found that lungs from VhlR/R mice showed pulmonary vascular remodeling, hemorrhage, edema, and macrophage infiltration resembling pulmonary hypertension. Interestingly, they found that the C598T VHL mutation in mouse lungs did not change the expression of HIF-1α and its targets whereas the expression of HIF-2α protein and HIF-2α-regulated genes such as Serpine1 were induced[53]. Moreover, heterozygosity of HIF-2α (HIF-2α+/-), but not HIF-1α (HIF-1α+/-), suppressed both polycythemia and pulmonary hypertension in the VhlR/R mice and attenuated vascular remodeling in VhlR/R lungs, suggesting a selective and critical role for HIF-2α in the pulmonary pathology in C598T VHL mutants[53]. This concept is strengthened by the study of Formenti et al[54] in which they showed that patients with HIF-2α gain-of-function mutations developed pulmonary hypertension with increased cardiac output, heart rate, and pulmonary ventilation. Recently, we found that HIF-2α, but not HIF-1α induced expression of FoxM1, leading to the induction of Aurora A kinase and Cyclin D1 and cell cycle progression which resulted in the proliferation of pulmonary artery smooth muscle cells[55]. A recent study reported novel VHL mutations in exon2 (G376A) and exon3 (C548T) of the lungs in a 2-month-old boy with severe polycythemia and pulmonary arterial hypertension[56]. Together, these studies suggest that, in Chuvash polycythemia, VHL mutations activate HIF, particularly HIF-2α, and contribute to the development of pulmonary arterial hypertension.

VHL AND LUNG CANCER

Lung cancer is the most common cause of cancer-related death in the United States[57]. There are two main types of lung cancer, small cell lung cancer (SCLC), and non-small cell lung cancer (NSCLC)[57]. SCLC is more aggressive and is strongly associated with cigarette smoking. SCLC arises in the larger airways: the primary and secondary bronchi. NSCLC accounts for 80% of lung cancer cases and is divided into three subcategories: squamous cell carcinoma, large cell carcinoma, and adenocarcinomas. Adenocarcinomas account for 30% of NSCLC cases and usually arise from epithelial cells in peripheral lung tissue. Recently, the population of adenocarcinomas cases in nonsmokers has been rising[58,59]. As in other solid tumors, lung cancer cells proliferate at a rate that exceeds the oxygen supply, resulting in regions of low oxygen tension (hypoxia)[60,61]. Tumor cells adapt to hypoxia by inducing genes involved in angiogenesis or glucose metabolism via HIF[61-63]. Hypoxia induces epithelial-mesenchymal transition (EMT) in tumor cells. EMT is a molecular and cellular process during which epithelial cells lose cell-cell interactions and apico-basal polarity and acquire mesenchymal and migratory properties[64-67]. EMT is featured with changes on cell morphology and genetic markers including the disappearance of an epithelial marker such as E-cadherin and acquisition of mesenchymal markers such as α-smooth muscle actin (α-SMA) and vimentin[68]. The significance of EMT in tumor metastasis is recently evidenced in a few tumor models[69]. Many researchers observed the loss of epithelial characteristics paired with the gain of mesenchymal markers in the invasive front of various cancers, pointing to a possible contribution of EMT in tumor metastasis[70]. EMT leads to increased tumor cell invasiveness and metastatic potential and resistance to radiation-induced cell death, which are the main causes of cancer death[61-63,66,71].

It is well established that in renal carcinoma, mutation of pVHL leads to stabilization of HIF, which drives angiogenesis and cancer development. However, the role of pVHL in lung cancer is less known. Lungs express levels of pVHL comparable to kidneys[72,73], and loss of VHL allele frequently occurs in patients with NSCLC[74-76], suggesting that pVHL likely contributes to lung cancer development. Indeed, we showed that suppression of pVHL in lung cancer cells induced EMT and increased migration and invasion[77], which is consistent with the observation that loss of pVHL promotes EMT and invasion in renal carcinoma cells[78-80]. We have shown that hypoxia induces lung cancer cell EMT and invasion and migration via a HIF-dependent pathway[81,82]. Therefore, loss of pVHL may mediate EMT through HIF signaling. However, we have found that HIF activation alone is not sufficient to induce EMT in lung cancer cells[81], indicating that a HIF independent pathway is also contributing to EMT when pVHL is absent. Surprisingly, our study suggests that loss of pVHL repressed lung cancer cell proliferation in vitro and decreased colonization in vivo[77], suggesting that loss of pVHL may limit lung cancer development. Consistently, loss of pVHL also reduces cell proliferation in fibroblasts, mammary epithelial cells, and chondrocytes and inhibits fibrosarcoma[83-85]. Since loss of pVHL induces HIF, we investigated the role of HIF in lung cancer cell proliferation and colonization. We found that constitutively active HIF increased lung cancer cell colonization whereas dominant negative HIF inhibited lung cancer colonization[77]. Thus, loss of pVHL causes reduced lung cancer cell proliferation and colonization in a HIF-independent pathway. We showed that suppression of pVHL decreased integrin and phosphorylated focal adhesion kinase (FAK) levels, suggesting that knockdown of pVHL represses lung cancer cell proliferation and colonization via decreased integrin/FAK signaling. Taking these together, we conclude that the role of pVHL/HIF in lung cancer development and progression may be determined by the status of HIF activity and pVHL status and stage of the lung cancer. Further studies with spatially and temporally controlled pVHL expression are warranted.

VHL AND PULMONARY FIBROSIS

Idiopathic pulmonary fibrosis (IPF) is another devastating lung disease with poor patient survival. It is characterized by progressive scarring of the lungs and elevated respiratory failure[86-89]. The etiology of IPF is unknown, and currently there is no effective treatment[90,91]. Pulmonary fibrosis is characterized by proliferation of lung fibroblasts and exaggerated deposition of ECM proteins, especially collagen and fibronectin[86,89]. ECM proteins interact with integrins, which act as their cell membrane receptors and thus initiate FAK signaling cascades involved in cell proliferation[92,93]. The proper assembly of fibronectin and collagen matrix requires the presence of pVHL, and loss of pVHL prevents fibroblast proliferation[24,28,29,83], suggesting that pVHL may play a role in the pathogenesis of pulmonary fibrosis.

A microarray study showed that lungs of IPF patients expressed higher levels of pVHL mRNA than lungs of control individuals[94]. We have reported that lungs of fibrotic patients expressed elevated levels of pVHL in fibroblastic foci. In an experimental lung fibrosis model induced by Bleomycin, pVHL expression is also elevated in mouse lung fibroblasts but not in alveolar type II cells. Ectopic overexpression of pVHL in lung fibroblasts increased the expression of fibronectin, collagen, and the α5 integrin subunit as well as lung fibroblast proliferation[95]. In addition, overexpression of pVHL induced FAK phosphorylation and activation[95]. Consistently, inhibition of the fibronectin/integrin α5β1/FAK signaling pathway diminished pVHL mediated cell proliferation while activation of α5 and β1 integrin fibronectin/integrin α5β1/FAK signaling pathway increased proliferation of fibroblasts. Moreover, pVHL is necessary for fibroblast proliferation after treatment of TGF-β1, a potent pro-fibrotic cytokine. These results suggest that elevated expression of pVHL results in the aberrant fibronectin expression and activation of integrin/FAK signaling, leading to fibroblast proliferation and fibrosis[95]. Although fibroblast activation (increased expression of collagen) and differentiation to myofibroblast (elevated levels of α-smooth muscle actin) are critical steps in fibrogenesis, we found that pVHL increased collagen expression but not the expression of α-smooth muscle actin, indicating pVHL may participate in earlier events in the formation of the fibroblastic foci, such as fibroblast proliferation[95]. Interestingly, this gain of function of pVHL did not alter HIF activity, suggesting that pVHL plays a HIF-independent role in the pathogenesis of pulmonary fibrosis. However, Hickey and colleagues have recently reported that the mutation of pVHL at codon 200 (R200W) causes Chuvash disease with pulmonary vascular remodeling and hypertension[53]. Older mice with this mutation contain elevated ECM deposition and develop fibrosis. In terms of the mechanism underlying the fibrosis in R200W mutation mice, the authors speculated that fibrosis is partially secondary to hemorrhage, edema, and impaired endothelial integrity and partially due to HIF2 activity[53]. Thus, additional studies are warranted to elucidate the HIF-dependent and HIF-independent roles of pVHL in pulmonary fibrosis.

VHL AND ACUTE RESPIRATORY DISTRESS SYNDROME

The acute respiratory distress syndrome (ARDS) is a key cause of acute respiratory failure. ARDS can be caused by pneumonia, sepsis, aspiration of gastric contents, and major trauma, and it is characterized by pulmonary edema and severe hypoxemia[96]. Most patients with ARDS who cannot clear alveolar edema efficiently have worse outcomes[97-99]. Alveolar fluid clearance is effected by active Na+ transport across the alveolar epithelium through apical Na+ channels and basolateral Na, K-ATPase[100]. Na+ enters alveolar epithelial cells via the apical Na+ channels and is extruded by the basolateral Na, K-ATPase, with water following the osmotic gradient into the interstitium and pulmonary circulation, leading to the clearance of edema[100-103]. Over-expression of Na, K-ATPase has been shown to increase alveolar fluid clearance[104-107]. In contrast, in several models of lung injury, Na, K-ATPase activity in alveolar epithelial cells is decreased, resulting in decreased lung fluid clearance[97,102,108-111].

Hypoxia is common in the lungs of patients with ARDS and inhibits Na, K-ATPase activity, thereby decreasing the rate of alveolar fluid reabsorption and worsening the outcomes of ARDS patients[110-112]. Dada et al[113] reported that hypoxia increases mitochondrial reactive oxygen species (ROS) production, which activates protein kinase C (PKC). PKC phosphorylates Ser18 of Na-K-ATPase α1 subunit, triggering the endocytosis of Na-K-ATPase[113]. Although short-term effects of hypoxia appear to be reversible upon reoxygenation with no significant degradation of total Na-K-ATPase in the whole cell lysate, prolonged hypoxia results in degradation of alveolar epithelial Na-K-ATPase via the ubiquitination/proteosome pathway[113,114].

We found that alveolar epithelial Na-K-ATPase is downregulated in a pVHL-dependent manner[115]. In the presence of pVHL, hypoxia promoted the degradation of plasma membrane Na-K-ATPase; in the absence of pVHL, hypoxia is unable to degrade plasma membrane Na-K-ATPase. pVHL mutants and dominant-negative Ubc5 (an ubiquitin conjugating enzyme, E2) prevented Na-K-ATPase from degradation, suggesting a functional pVHL E3 ligase is essential for Na-K-ATPase degradation during hypoxia[115]. Interestingly, HIF overexpression is not sufficient to induce Na-K-ATPase degradation, and loss of HIF does not prevent hypoxia-mediated degradation of Na-K-ATPase. Therefore, pVHL-mediated Na-K-ATPase degradation is likely HIF independent[115]. Moreover, generation of reactive oxygen species was necessary for pVHL-mediated Na-K-ATPase degradation during hypoxia[115].

CONCLUSION

Although pVHL was first identified to be a tumor suppressor in renal carcinoma, accumulating evidence suggests that pVHL has a much broader spectrum of functions and is key to many organ dysfunctions. In this review, we summarized recent evidence that pVHL plays an essential role in a few respiratory diseases, including pulmonary arterial hypertension, lung cancer, pulmonary fibrosis, and ARDS. As in other tissues, pVHL functions in two distinct pathway in the lungs: HIF independent (fibrosis, lung cancer, and ARDS) and HIF dependent (lung cancer and pulmonary arterial hypertension). Certainly, this is only the beginning of uncovering the complex role of pVHL in lung diseases, as evidenced by a recent report which suggests pVHL expression is elevated in skeletal muscles of patients with chronic obstructive pulmonary disease (COPD)[116]. However, the challenge to dissect the function of VHL disease is to investigate the role of pVHL in a cell-type-specific, context-specific, and temporal fashion. A more challenging task is to elucidate in detail the molecular mechanisms by which pVHL regulate these functions in the lungs as an E3 ligase and adaptor protein.

Footnotes

P- Reviewer: Wu M S- Editor: Song XX L- Editor: A E- Editor: Wang CH

References
1.  Lonser RR, Glenn GM, Walther M, Chew EY, Libutti SK, Linehan WM, Oldfield EH. von Hippel-Lindau disease. Lancet. 2003;361:2059-2067.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1083]  [Cited by in F6Publishing: 948]  [Article Influence: 45.1]  [Reference Citation Analysis (0)]
2.  Seizinger BR, Rouleau GA, Ozelius LJ, Lane AH, Farmer GE, Lamiell JM, Haines J, Yuen JW, Collins D, Majoor-Krakauer D. Von Hippel-Lindau disease maps to the region of chromosome 3 associated with renal cell carcinoma. Nature. 1988;332:268-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 459]  [Cited by in F6Publishing: 408]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
3.  Kaelin WG. The von Hippel-Lindau tumour suppressor protein: O2 sensing and cancer. Nat Rev Cancer. 2008;8:865-873.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 519]  [Cited by in F6Publishing: 517]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
4.  Latif F, Tory K, Gnarra J, Yao M, Duh FM, Orcutt ML, Stackhouse T, Kuzmin I, Modi W, Geil L. Identification of the von Hippel-Lindau disease tumor suppressor gene. Science. 1993;260:1317-1320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 11]  [Reference Citation Analysis (0)]
5.  Kaelin WG. The von Hippel-Lindau tumor suppressor gene and kidney cancer. Clin Cancer Res. 2004;10:6290S-6295S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 223]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
6.  Maher ER. Von Hippel-Lindau disease. Curr Mol Med. 2004;4:833-842.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Iwai K, Yamanaka K, Kamura T, Minato N, Conaway RC, Conaway JW, Klausner RD, Pause A. Identification of the von Hippel-lindau tumor-suppressor protein as part of an active E3 ubiquitin ligase complex. Proc Natl Acad Sci USA. 1999;96:12436-12441.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Yan Q, Kamura T, Cai Y, Jin J, Ivan M, Mushegian A, Conaway RC, Conaway JW. Identification of Elongin C and Skp1 sequences that determine Cullin selection. J Biol Chem. 2004;279:43019-43026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
9.  Ivan M, Kondo K, Yang H, Kim W, Valiando J, Ohh M, Salic A, Asara JM, Lane WS, Kaelin WG. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science. 2001;292:464-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3582]  [Cited by in F6Publishing: 3553]  [Article Influence: 154.5]  [Reference Citation Analysis (0)]
10.  Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, von Kriegsheim A, Hebestreit HF, Mukherji M, Schofield CJ. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science. 2001;292:468-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4121]  [Cited by in F6Publishing: 4097]  [Article Influence: 178.1]  [Reference Citation Analysis (0)]
11.  Masson N, Willam C, Maxwell PH, Pugh CW, Ratcliffe PJ. Independent function of two destruction domains in hypoxia-inducible factor-alpha chains activated by prolyl hydroxylation. EMBO J. 2001;20:5197-5206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 776]  [Cited by in F6Publishing: 775]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
12.  Semenza GL. O2-regulated gene expression: transcriptional control of cardiorespiratory physiology by HIF-1. J Appl Physiol. 2004;96:1173-117; discussion 1173-1177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 190]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
13.  Kim W, Kaelin WG. The von Hippel-Lindau tumor suppressor protein: new insights into oxygen sensing and cancer. Curr Opin Genet Dev. 2003;13:55-60.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Iliopoulos O, Kibel A, Gray S, Kaelin WG. Tumour suppression by the human von Hippel-Lindau gene product. Nat Med. 1995;1:822-826.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Kondo K, Klco J, Nakamura E, Lechpammer M, Kaelin WG. Inhibition of HIF is necessary for tumor suppression by the von Hippel-Lindau protein. Cancer Cell. 2002;1:237-246.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Semenza GL. VHL and p53: tumor suppressors team up to prevent cancer. Mol Cell. 2006;22:437-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 19]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
17.  Bindra RS, Vasselli JR, Stearman R, Linehan WM, Klausner RD. VHL-mediated hypoxia regulation of cyclin D1 in renal carcinoma cells. Cancer Res. 2002;62:3014-3019.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Zatyka M, da Silva NF, Clifford SC, Morris MR, Wiesener MS, Eckardt KU, Houlston RS, Richards FM, Latif F, Maher ER. Identification of cyclin D1 and other novel targets for the von Hippel-Lindau tumor suppressor gene by expression array analysis and investigation of cyclin D1 genotype as a modifier in von Hippel-Lindau disease. Cancer Res. 2002;62:3803-3811.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Clark PE. The role of VHL in clear-cell renal cell carcinoma and its relation to targeted therapy. Kidney Int. 2009;76:939-945.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 90]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
20.  Dammai V, Adryan B, Lavenburg KR, Hsu T. Drosophila awd, the homolog of human nm23, regulates FGF receptor levels and functions synergistically with shi/dynamin during tracheal development. Genes Dev. 2003;17:2812-2824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 82]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
21.  Champion KJ, Guinea M, Dammai V, Hsu T. Endothelial function of von Hippel-Lindau tumor suppressor gene: control of fibroblast growth factor receptor signaling. Cancer Res. 2008;68:4649-4657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 26]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
22.  Lutz MS, Burk RD. Primary cilium formation requires von hippel-lindau gene function in renal-derived cells. Cancer Res. 2006;66:6903-6907.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 112]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
23.  Roe JS, Kim H, Lee SM, Kim ST, Cho EJ, Youn HD. p53 stabilization and transactivation by a von Hippel-Lindau protein. Mol Cell. 2006;22:395-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 192]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
24.  Ohh M, Yauch RL, Lonergan KM, Whaley JM, Stemmer-Rachamimov AO, Louis DN, Gavin BJ, Kley N, Kaelin WG, Iliopoulos O. The von Hippel-Lindau tumor suppressor protein is required for proper assembly of an extracellular fibronectin matrix. Mol Cell. 1998;1:959-968.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Kurban G, Hudon V, Duplan E, Ohh M, Pause A. Characterization of a von Hippel Lindau pathway involved in extracellular matrix remodeling, cell invasion, and angiogenesis. Cancer Res. 2006;66:1313-1319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 103]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
26.  Tang N, Mack F, Haase VH, Simon MC, Johnson RS. pVHL function is essential for endothelial extracellular matrix deposition. Mol Cell Biol. 2006;26:2519-2530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 75]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
27.  Esteban-Barragán MA, Avila P, Alvarez-Tejado M, Gutiérrez MD, García-Pardo A, Sánchez-Madrid F, Landázuri MO. Role of the von Hippel-Lindau tumor suppressor gene in the formation of beta1-integrin fibrillar adhesions. Cancer Res. 2002;62:2929-2936.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Kurban G, Duplan E, Ramlal N, Hudon V, Sado Y, Ninomiya Y, Pause A. Collagen matrix assembly is driven by the interaction of von Hippel-Lindau tumor suppressor protein with hydroxylated collagen IV alpha 2. Oncogene. 2008;27:1004-1012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 72]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
29.  Grosfeld A, Stolze IP, Cockman ME, Pugh CW, Edelmann M, Kessler B, Bullock AN, Ratcliffe PJ, Masson N. Interaction of hydroxylated collagen IV with the von hippel-lindau tumor suppressor. J Biol Chem. 2007;282:13264-13269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
30.  Frew IJ, Krek W. pVHL: a multipurpose adaptor protein. Sci Signal. 2008;1:pe30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 75]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
31.  Frew IJ, Krek W. Multitasking by pVHL in tumour suppression. Curr Opin Cell Biol. 2007;19:685-690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
32.  Hergovich A, Lisztwan J, Barry R, Ballschmieter P, Krek W. Regulation of microtubule stability by the von Hippel-Lindau tumour suppressor protein pVHL. Nat Cell Biol. 2003;5:64-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 267]  [Cited by in F6Publishing: 274]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
33.  Lolkema MP, Mehra N, Jorna AS, van Beest M, Giles RH, Voest EE. The von Hippel-Lindau tumor suppressor protein influences microtubule dynamics at the cell periphery. Exp Cell Res. 2004;301:139-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 40]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
34.  Mans DA, Lolkema MP, van Beest M, Daenen LG, Voest EE, Giles RH. Mobility of the von Hippel-Lindau tumour suppressor protein is regulated by kinesin-2. Exp Cell Res. 2008;314:1229-1236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
35.  Lolkema MP, Mans DA, Snijckers CM, van Noort M, van Beest M, Voest EE, Giles RH. The von Hippel-Lindau tumour suppressor interacts with microtubules through kinesin-2. FEBS Lett. 2007;581:4571-4576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 37]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
36.  Thoma CR, Frew IJ, Hoerner CR, Montani M, Moch H, Krek W. pVHL and GSK3beta are components of a primary cilium-maintenance signalling network. Nat Cell Biol. 2007;9:588-595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 193]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
37.  Frew IJ, Thoma CR, Georgiev S, Minola A, Hitz M, Montani M, Moch H, Krek W. pVHL and PTEN tumour suppressor proteins cooperatively suppress kidney cyst formation. EMBO J. 2008;27:1747-1757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 126]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
38.  Kamada M, Suzuki K, Kato Y, Okuda H, Shuin T. von Hippel-Lindau protein promotes the assembly of actin and vinculin and inhibits cell motility. Cancer Res. 2001;61:4184-4189.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Escobar B, de Cárcer G, Fernández-Miranda G, Cascón A, Bravo-Cordero JJ, Montoya MC, Robledo M, Cañamero M, Malumbres M. Brick1 is an essential regulator of actin cytoskeleton required for embryonic development and cell transformation. Cancer Res. 2010;70:9349-9359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
40.  Cascón A, Escobar B, Montero-Conde C, Rodríguez-Antona C, Ruiz-Llorente S, Osorio A, Mercadillo F, Letón R, Campos JM, García-Sagredo JM. Loss of the actin regulator HSPC300 results in clear cell renal cell carcinoma protection in Von Hippel-Lindau patients. Hum Mutat. 2007;28:613-621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
41.  Humbert M, Sitbon O, Simonneau G. Treatment of pulmonary arterial hypertension. N Engl J Med. 2004;351:1425-1436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1235]  [Cited by in F6Publishing: 1128]  [Article Influence: 56.4]  [Reference Citation Analysis (0)]
42.  McLaughlin VV, Archer SL, Badesch DB, Barst RJ, Farber HW, Lindner JR, Mathier MA, McGoon MD, Park MH, Rosenson RS. ACCF/AHA 2009 expert consensus document on pulmonary hypertension: a report of the American College of Cardiology Foundation Task Force on Expert Consensus Documents and the American Heart Association: developed in collaboration with the American College of Chest Physicians, American Thoracic Society, Inc., and the Pulmonary Hypertension Association. Circulation. 2009;119:2250-2294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 768]  [Cited by in F6Publishing: 745]  [Article Influence: 49.7]  [Reference Citation Analysis (0)]
43.  Stenmark KR, Fagan KA, Frid MG. Hypoxia-induced pulmonary vascular remodeling: cellular and molecular mechanisms. Circ Res. 2006;99:675-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 739]  [Cited by in F6Publishing: 741]  [Article Influence: 41.2]  [Reference Citation Analysis (0)]
44.  Semenza GL. Hypoxia-inducible factor 1 (HIF-1) pathway. Sci STKE. 2007;2007:cm8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 557]  [Cited by in F6Publishing: 614]  [Article Influence: 36.1]  [Reference Citation Analysis (0)]
45.  Simon MC, Keith B. The role of oxygen availability in embryonic development and stem cell function. Nat Rev Mol Cell Biol. 2008;9:285-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 730]  [Cited by in F6Publishing: 683]  [Article Influence: 42.7]  [Reference Citation Analysis (0)]
46.  Brusselmans K, Compernolle V, Tjwa M, Wiesener MS, Maxwell PH, Collen D, Carmeliet P. Heterozygous deficiency of hypoxia-inducible factor-2alpha protects mice against pulmonary hypertension and right ventricular dysfunction during prolonged hypoxia. J Clin Invest. 2003;111:1519-1527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 236]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
47.  Yu AY, Shimoda LA, Iyer NV, Huso DL, Sun X, McWilliams R, Beaty T, Sham JS, Wiener CM, Sylvester JT. Impaired physiological responses to chronic hypoxia in mice partially deficient for hypoxia-inducible factor 1alpha. J Clin Invest. 1999;103:691-696.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 533]  [Cited by in F6Publishing: 503]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
48.  Abud EM, Maylor J, Undem C, Punjabi A, Zaiman AL, Myers AC, Sylvester JT, Semenza GL, Shimoda LA. Digoxin inhibits development of hypoxic pulmonary hypertension in mice. Proc Natl Acad Sci USA. 2012;109:1239-1244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 79]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
49.  Pastore YD, Jelinek J, Ang S, Guan Y, Liu E, Jedlickova K, Krishnamurti L, Prchal JT. Mutations in the VHL gene in sporadic apparently congenital polycythemia. Blood. 2003;101:1591-1595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 116]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
50.  Smith TG, Brooks JT, Balanos GM, Lappin TR, Layton DM, Leedham DL, Liu C, Maxwell PH, McMullin MF, McNamara CJ. Mutation of von Hippel-Lindau tumour suppressor and human cardiopulmonary physiology. PLoS Med. 2006;3:e290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 153]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
51.  Bushuev VI, Miasnikova GY, Sergueeva AI, Polyakova LA, Okhotin D, Gaskin PR, Debebe Z, Nekhai S, Castro OL, Prchal JT. Endothelin-1, vascular endothelial growth factor and systolic pulmonary artery pressure in patients with Chuvash polycythemia. Haematologica. 2006;91:744-749.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Sable CA, Aliyu ZY, Dham N, Nouraie M, Sachdev V, Sidenko S, Miasnikova GY, Polyakova LA, Sergueeva AI, Okhotin DJ. Pulmonary artery pressure and iron deficiency in patients with upregulation of hypoxia sensing due to homozygous VHL(R200W) mutation (Chuvash polycythemia). Haematologica. 2012;97:193-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 23]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
53.  Hickey MM, Richardson T, Wang T, Mosqueira M, Arguiri E, Yu H, Yu QC, Solomides CC, Morrisey EE, Khurana TS. The von Hippel-Lindau Chuvash mutation promotes pulmonary hypertension and fibrosis in mice. J Clin Invest. 2010;120:827-839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 115]  [Article Influence: 8.2]  [Reference Citation Analysis (1)]
54.  Formenti F, Beer PA, Croft QP, Dorrington KL, Gale DP, Lappin TR, Lucas GS, Maher ER, Maxwell PH, McMullin MF. Cardiopulmonary function in two human disorders of the hypoxia-inducible factor (HIF) pathway: von Hippel-Lindau disease and HIF-2alpha gain-of-function mutation. FASEB J. 2011;25:2001-2011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 69]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
55.  Raghavan A, Zhou G, Zhou Q, Ibe JC, Ramchandran R, Yang Q, Racherla H, Raychaudhuri P, Raj JU. Hypoxia-induced pulmonary arterial smooth muscle cell proliferation is controlled by forkhead box M1. Am J Respir Cell Mol Biol. 2012;46:431-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 36]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
56.  Bond J, Gale DP, Connor T, Adams S, de Boer J, Gascoyne DM, Williams O, Maxwell PH, Ancliff PJ. Dysregulation of the HIF pathway due to VHL mutation causing severe erythrocytosis and pulmonary arterial hypertension. Blood. 2011;117:3699-3701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 36]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
57.  Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ. Cancer statistics, 2008. CA Cancer J Clin. 2008;58:71-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8283]  [Cited by in F6Publishing: 8159]  [Article Influence: 509.9]  [Reference Citation Analysis (0)]
58.  Sun S, Schiller JH, Gazdar AF. Lung cancer in never smokers--a different disease. Nat Rev Cancer. 2007;7:778-790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1019]  [Cited by in F6Publishing: 1053]  [Article Influence: 61.9]  [Reference Citation Analysis (0)]
59.  Herbst RS, Heymach JV, Lippman SM. Lung cancer. N Engl J Med. 2008;359:1367-1380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1876]  [Cited by in F6Publishing: 1905]  [Article Influence: 119.1]  [Reference Citation Analysis (0)]
60.  Erler JT, Bennewith KL, Nicolau M, Dornhöfer N, Kong C, Le QT, Chi JT, Jeffrey SS, Giaccia AJ. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature. 2006;440:1222-1226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1058]  [Cited by in F6Publishing: 1031]  [Article Influence: 57.3]  [Reference Citation Analysis (0)]
61.  Harris AL. Hypoxia--a key regulatory factor in tumour growth. Nat Rev Cancer. 2002;2:38-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3808]  [Cited by in F6Publishing: 3776]  [Article Influence: 171.6]  [Reference Citation Analysis (0)]
62.  Erler JT, Giaccia AJ. Lysyl oxidase mediates hypoxic control of metastasis. Cancer Res. 2006;66:10238-10241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 155]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
63.  Peinado H, Cano A. A hypoxic twist in metastasis. Nat Cell Biol. 2008;10:253-254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
64.  Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7:131-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2893]  [Cited by in F6Publishing: 3008]  [Article Influence: 167.1]  [Reference Citation Analysis (0)]
65.  Peinado H, Olmeda D, Cano A. Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer. 2007;7:415-428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2365]  [Cited by in F6Publishing: 2426]  [Article Influence: 142.7]  [Reference Citation Analysis (0)]
66.  Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2:563-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2801]  [Cited by in F6Publishing: 2694]  [Article Influence: 122.5]  [Reference Citation Analysis (0)]
67.  MacDonald IC, Groom AC, Chambers AF. Cancer spread and micrometastasis development: quantitative approaches for in vivo models. Bioessays. 2002;24:885-893.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 131]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
68.  Zavadil J, Böttinger EP. TGF-beta and epithelial-to-mesenchymal transitions. Oncogene. 2005;24:5764-5774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1207]  [Cited by in F6Publishing: 1271]  [Article Influence: 66.9]  [Reference Citation Analysis (0)]
69.  Hugo H, Ackland ML, Blick T, Lawrence MG, Clements JA, Williams ED, Thompson EW. Epithelial--mesenchymal and mesenchymal--epithelial transitions in carcinoma progression. J Cell Physiol. 2007;213:374-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 783]  [Cited by in F6Publishing: 802]  [Article Influence: 50.1]  [Reference Citation Analysis (0)]
70.  Tse JC, Kalluri R. Mechanisms of metastasis: epithelial-to-mesenchymal transition and contribution of tumor microenvironment. J Cell Biochem. 2007;101:816-829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 252]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
71.  Pantel K, Brakenhoff RH. Dissecting the metastatic cascade. Nat Rev Cancer. 2004;4:448-456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 954]  [Cited by in F6Publishing: 931]  [Article Influence: 46.6]  [Reference Citation Analysis (0)]
72.  Corless CL, Kibel AS, Iliopoulos O, Kaelin WG. Immunostaining of the von Hippel-Lindau gene product in normal and neoplastic human tissues. Hum Pathol. 1997;28:459-464.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Los M, Jansen GH, Kaelin WG, Lips CJ, Blijham GH, Voest EE. Expression pattern of the von Hippel-Lindau protein in human tissues. Lab Invest. 1996;75:231-238.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Ho WL, Chang JW, Tseng RC, Chen JT, Chen CY, Jou YS, Wang YC. Loss of heterozygosity at loci of candidate tumor suppressor genes in microdissected primary non-small cell lung cancer. Cancer Detect Prev. 2002;26:343-349.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  An Q, Liu Y, Gao Y, Huang J, Fong X, Liu L, Zhang D, Zhang J, Cheng S. Deletion of tumor suppressor genes in Chinese non-small cell lung cancer. Cancer Lett. 2002;184:189-195.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Pitterle DM, Jolicoeur EM, Bepler G. Hot spots for molecular genetic alterations in lung cancer. In Vivo. 1998;12:643-658.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Zhou Q, Chen T, Ibe JC, Raj JU, Zhou G. Knockdown of von Hippel-Lindau protein decreases lung cancer cell proliferation and colonization. FEBS Lett. 2012;586:1510-1515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
78.  Esteban MA, Tran MG, Harten SK, Hill P, Castellanos MC, Chandra A, Raval R, O’brien TS, Maxwell PH. Regulation of E-cadherin expression by VHL and hypoxia-inducible factor. Cancer Res. 2006;66:3567-3575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 209]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
79.  Evans AJ, Russell RC, Roche O, Burry TN, Fish JE, Chow VW, Kim WY, Saravanan A, Maynard MA, Gervais ML. VHL promotes E2 box-dependent E-cadherin transcription by HIF-mediated regulation of SIP1 and snail. Mol Cell Biol. 2007;27:157-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 210]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
80.  Krishnamachary B, Zagzag D, Nagasawa H, Rainey K, Okuyama H, Baek JH, Semenza GL. Hypoxia-inducible factor-1-dependent repression of E-cadherin in von Hippel-Lindau tumor suppressor-null renal cell carcinoma mediated by TCF3, ZFHX1A, and ZFHX1B. Cancer Res. 2006;66:2725-2731.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 318]  [Cited by in F6Publishing: 323]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
81.  Zhou G, Dada LA, Wu M, Kelly A, Trejo H, Zhou Q, Varga J, Sznajder JI. Hypoxia-induced alveolar epithelial-mesenchymal transition requires mitochondrial ROS and hypoxia-inducible factor 1. Am J Physiol Lung Cell Mol Physiol. 2009;297:L1120-L1130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 162]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
82.  Shaikh D, Zhou Q, Chen T, Ibe JC, Raj JU, Zhou G. cAMP-dependent protein kinase is essential for hypoxia-mediated epithelial-mesenchymal transition, migration, and invasion in lung cancer cells. Cell Signal. 2012;24:2396-2406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 68]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
83.  Mack FA, Patel JH, Biju MP, Haase VH, Simon MC. Decreased growth of Vhl-/- fibrosarcomas is associated with elevated levels of cyclin kinase inhibitors p21 and p27. Mol Cell Biol. 2005;25:4565-4578.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 65]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
84.  Pfander D, Kobayashi T, Knight MC, Zelzer E, Chan DA, Olsen BR, Giaccia AJ, Johnson RS, Haase VH, Schipani E. Deletion of Vhlh in chondrocytes reduces cell proliferation and increases matrix deposition during growth plate development. Development. 2004;131:2497-2508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 108]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
85.  Seagroves TN, Peacock DL, Liao D, Schwab LP, Krueger R, Handorf CR, Haase VH, Johnson RS. VHL deletion impairs mammary alveologenesis but is not sufficient for mammary tumorigenesis. Am J Pathol. 2010;176:2269-2282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
86.  Coultas DB, Zumwalt RE, Black WC, Sobonya RE. The epidemiology of interstitial lung diseases. Am J Respir Crit Care Med. 1994;150:967-972.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 616]  [Cited by in F6Publishing: 653]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
87.  American Thoracic Society. Idiopathic pulmonary fibrosis: diagnosis and treatment. International consensus statement. American Thoracic Society (ATS), and the European Respiratory Society (ERS). Am J Respir Crit Care Med. 2000;161:646-664.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Schwartz DA, Helmers RA, Galvin JR, Van Fossen DS, Frees KL, Dayton CS, Burmeister LF, Hunninghake GW. Determinants of survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 1994;149:450-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 228]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
89.  Selman M, Pardo A. The epithelial/fibroblastic pathway in the pathogenesis of idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol. 2003;29:S93-S97.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Nathan SD, Noble PW, Tuder RM. Idiopathic pulmonary fibrosis and pulmonary hypertension: connecting the dots. Am J Respir Crit Care Med. 2007;175:875-880.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 169]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
91.  Strieter RM, Gomperts BN, Keane MP. The role of CXC chemokines in pulmonary fibrosis. J Clin Invest. 2007;117:549-556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 197]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
92.  Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell. 2002;110:673-687.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Janes SM, Watt FM. New roles for integrins in squamous-cell carcinoma. Nat Rev Cancer. 2006;6:175-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 145]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
94.  Pardo A, Gibson K, Cisneros J, Richards TJ, Yang Y, Becerril C, Yousem S, Herrera I, Ruiz V, Selman M. Up-regulation and profibrotic role of osteopontin in human idiopathic pulmonary fibrosis. PLoS Med. 2005;2:e251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in F6Publishing: 358]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
95.  Zhou Q, Pardo A, Königshoff M, Eickelberg O, Budinger GR, Thavarajah K, Gottardi CJ, Jones J, Varga J, Selman M. Role of von Hippel-Lindau protein in fibroblast proliferation and fibrosis. FASEB J. 2011;25:3032-3044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
96.  Matthay MA, Ware LB, Zimmerman GA. The acute respiratory distress syndrome. J Clin Invest. 2012;122:2731-2740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1156]  [Cited by in F6Publishing: 1276]  [Article Influence: 106.3]  [Reference Citation Analysis (0)]
97.  Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med. 2000;342:1334-1349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3976]  [Cited by in F6Publishing: 3767]  [Article Influence: 157.0]  [Reference Citation Analysis (0)]
98.  Sznajder JI. Alveolar edema must be cleared for the acute respiratory distress syndrome patient to survive. Am J Respir Crit Care Med. 2001;163:1293-1294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 85]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
99.  Ware LB, Matthay MA. Alveolar fluid clearance is impaired in the majority of patients with acute lung injury and the acute respiratory distress syndrome. Am J Respir Crit Care Med. 2001;163:1376-1383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 669]  [Cited by in F6Publishing: 658]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
100.  Matalon S, Lazrak A, Jain L, Eaton DC. Invited review: biophysical properties of sodium channels in lung alveolar epithelial cells. J Appl Physiol. 2002;93:1852-1859.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Sznajder JI. Strategies to increase alveolar epithelial fluid removal in the injured lung. Am J Respir Crit Care Med. 1999;160:1441-1442.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Sznajder JI, Factor P, Ingbar DH. Invited review: lung edema clearance: role of Na(+)-K(+)-ATPase. J Appl Physiol. 2002;93:1860-1866.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Therien AG, Blostein R. Mechanisms of sodium pump regulation. Am J Physiol Cell Physiol. 2000;279:C541-C566.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Azzam ZS, Dumasius V, Saldias FJ, Adir Y, Sznajder JI, Factor P. Na,K-ATPase overexpression improves alveolar fluid clearance in a rat model of elevated left atrial pressure. Circulation. 2002;105:497-501.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Machado-Aranda D, Adir Y, Young JL, Briva A, Budinger GR, Yeldandi AV, Sznajder JI, Dean DA. Gene transfer of the Na+,K+-ATPase beta1 subunit using electroporation increases lung liquid clearance. Am J Respir Crit Care Med. 2005;171:204-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 59]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
106.  Thome U, Chen L, Factor P, Dumasius V, Freeman B, Sznajder JI, Matalon S. Na,K-ATPase gene transfer mitigates an oxidant-induced decrease of active sodium transport in rat fetal ATII cells. Am J Respir Cell Mol Biol. 2001;24:245-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 36]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
107.  Factor P, Dumasius V, Saldias F, Brown LA, Sznajder JI. Adenovirus-mediated transfer of an Na+/K+-ATPase beta1 subunit gene improves alveolar fluid clearance and survival in hyperoxic rats. Hum Gene Ther. 2000;11:2231-2242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 86]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
108.  Zhou G, Dada LA, Sznajder JI. Regulation of alveolar epithelial function by hypoxia. Eur Respir J. 2008;31:1107-1113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 35]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
109.  Dada LA, Sznajder JI. Mechanisms of pulmonary edema clearance during acute hypoxemic respiratory failure: role of the Na,K-ATPase. Crit Care Med. 2003;31:S248-S252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
110.  Clerici C, Matthay MA. Hypoxia regulates gene expression of alveolar epithelial transport proteins. J Appl Physiol. 2000;88:1890-1896.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Wodopia R, Ko HS, Billian J, Wiesner R, Bärtsch P, Mairbäurl H. Hypoxia decreases proteins involved in epithelial electrolyte transport in A549 cells and rat lung. Am J Physiol Lung Cell Mol Physiol. 2000;279:L1110-L1119.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Vivona ML, Matthay M, Chabaud MB, Friedlander G, Clerici C. Hypoxia reduces alveolar epithelial sodium and fluid transport in rats: reversal by beta-adrenergic agonist treatment. Am J Respir Cell Mol Biol. 2001;25:554-561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 144]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
113.  Dada LA, Chandel NS, Ridge KM, Pedemonte C, Bertorello AM, Sznajder JI. Hypoxia-induced endocytosis of Na,K-ATPase in alveolar epithelial cells is mediated by mitochondrial reactive oxygen species and PKC-zeta. J Clin Invest. 2003;111:1057-1064.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 206]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
114.  Comellas AP, Dada LA, Lecuona E, Pesce LM, Chandel NS, Quesada N, Budinger GR, Strous GJ, Ciechanover A, Sznajder JI. Hypoxia-mediated degradation of Na,K-ATPase via mitochondrial reactive oxygen species and the ubiquitin-conjugating system. Circ Res. 2006;98:1314-1322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 83]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
115.  Zhou G, Dada LA, Chandel NS, Iwai K, Lecuona E, Ciechanover A, Sznajder JI. Hypoxia-mediated Na-K-ATPase degradation requires von Hippel Lindau protein. FASEB J. 2008;22:1335-1342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 34]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
116.  Jatta K, Eliason G, Portela-Gomes GM, Grimelius L, Caro O, Nilholm L, Sirjsö A, Piehl-Aulin K, Abdel-Halim SM. Overexpression of von Hippel-Lindau protein in skeletal muscles of patients with chronic obstructive pulmonary disease. J Clin Pathol. 2009;62:70-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]