REVIEW URRENT C OPINION

Role of epidermal growth factor receptor in vascular structure and function Barbara Schreier, Michael Gekle, and Claudia Grossmann

Purpose of the review The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase with a wide implication in tumor biology, wound healing and development. Besides acting as a growth factor receptor activated by ligands such as EGF, the EGFR can also be transactivated and thereby mediate cross-talk with different signaling pathways. The aim of this review is to illustrate the Janus-faced function of the EGFR in the vasculature with its relevance for vascular biology and disease. Recent findings Over recent years, the number of identified signaling partners of the EGFR has steadily increased, as have the biological processes in which the EGFR is thought to be involved. Recently, new models have allowed investigation of EGFR effects in vivo, shedding some light on the overall function of the EGFR in the vasculature. At the same time, EGFR inhibitors and antibodies have become increasingly established in cancer therapy, providing potential therapeutic tools for decreasing EGFR signaling. Summary The EGFR is a versatile signaling pathway integrator associated with vascular homeostasis and disease. In addition to modulating basal vascular tone and tissue homeostasis, the EGFR also seems to be involved in proinflammatory, proliferative, migratory and remodeling processes, with enhanced deposition of extracellular matrix components, thereby promoting vascular diseases such as hypertension or atherosclerosis. Keywords atherosclerosis, diabetes, endothelial cells, hypertension, vascular smooth muscle cells

INTRODUCTION In humans, 58 receptor tyrosine kinases have so far been identified. Of those, the epidermal growth factor receptor (EGFR), also known as ErbB1, is a prominent member [1]. It belongs to the ErbB family and is composed of an N-terminal extracellular ligand-binding region, a single alpha helical transmembrane region, a juxtamembrane domain that is conserved between the family members and a c-terminal cytoplasmic region with tyrosine kinase activity and phosphorylation sites. Activation of EGFR occurs either by binding of ligands such as EGF (epidermal growth factor) and heparin boundEGF, or by transactivation. Upon binding of ligand to a single receptor, conformational changes occur that allow dimerization and allosteric activation of the tyrosine kinase domain in the cytoplasm [2], resulting in phosphorylation of tyrosine residues, enabling docking of different signaling components [3,4]. Transactivation may be mediated by activation of matrix metalloproteases (MMPs)/a

disintegrin and metalloproteases (ADAMS) and cleavage of membrane-bound ligand precursor molecules of the EGFR or by activation of the EGFR through intracellular protein kinases. This flexibility enables the EGFR to mediate the cross-talk between different signaling pathways and to act as an important signal integrator (Fig. 1). The EGFR is widely acknowledged for its influence in tumor biology and wound healing but an additional role in maintaining organ and cellular homeostasis in general is becoming more and more evident especially in the cardiovascular

Julius Bernstein Institute of Physiology, Martin Luther University HalleWittenberg, Halle, Germany Correspondence to Claudia Grossmann, Julius-Bernstein-Institut fu¨r Physiologie, Universita¨t Halle-Wittenberg, Magdeburger Strasse 6, 06097 Halle/Saale, Germany. Tel: 49 345 557 1886; fax: +49 345 557 4019; e-mail: [email protected] Curr Opin Nephrol Hypertens 2014, 23:113–121 DOI:10.1097/01.mnh.0000441152.62943.29

1062-4821 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

www.co-nephrolhypertens.com

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Pathophysiology of hypertension

VASCULAR SMOOTH MUSCLE CELLS

KEY POINTS  The EGFR is an important signaling pathway integrator of G-protein-coupled receptors, steroid receptors and (receptor) tyrosine kinases.  It seems to be a Janus-faced receptor for vascular biology because it is involved in homeostatic and also pathological actions.  Regarding vascular homeostasis, the EGFR supports basal vascular tone and enhances ROS defense capacity.  During the pathogenesis of vascular disease, the EGFR enhances inflammation, proliferation, migration and extracellular matrix deposition, potentially promoting atherosclerotic processes, diabetic vascular complications and hypertension.

&&

system [1,5 ]. The EGFR is expressed in vascular smooth muscle cells (VSMC), endothelial cells, macrophages and regulatory T lymphocytes, and all of these cells also secrete EGFR ligands.

Dopamine

Aldosterone

Angiotensin II ATP

Thrombin

Opioids

EGFR ligands

15-HETE

Estrogen

Research so far has mainly focused on describing the effects of EGFR in VSMC, and evidence of an involvement in pathophysiology is beginning to emerge. The EGFR seems to mediate the generation of a proinflammatory phenotype, for example by promoting formation of inflammatory mediators, such as prostaglandins and sphingosin-1-phosphate via the protein kinase C (PKC) delta/c-Src/EGFR/ PI3K/Akt/Elk-1 pathway [6]. Vasoactive sphingosin-1-phosphate in turn can transactivate the EGFR to stimulate inflammatory signaling, a response enhanced in spontaneously hypertensive strokeprone rats, providing a novel link between hypertension and vascular inflammation [7]. Transactivation of the EGFR by renin-angiotensin-aldosterone system (RAAS) components leads to oxidative stress and also a proinflammatory phenotype [8]. Interestingly, attenuation of angiotensin II-induced inflammation and growth by the hepatocyte growth factor/c-Met system has been shown to rely on EGFR degradation, thereby preventing atherosclerotic changes [9]. During aging a shift to a

(EGF, TGF-α, amphiregulin, epigen, HB-EGF, betacellulin, epiregulin)

oxLDL

MMPs

Endothelin-1

ADAMs 9, 10, 12, 17

Norepinephrine Urokinase Thrombospondin

β

NOX γ

P P



P

Src

– Ca2+ – PI3K – Pyk2 – PKC ?

P P P P P P P P P P P EGFR

– Ras – ERK 1/2 – Src – PI3K

– mTOR – Akt – FAK – STAT ?

FIGURE 1. Model of the epidermal growth factor receptor (EGFR) transactivation signaling cascade. Transactivation of the EGFR occurs by binding of transactivating substances to their specific receptors. They can then induce phosphorylation of the EGFR either by intracellular signaling cascades or a triple-membrane-spanning mechanism involving shedding of EGFR-ligands by a disintegrin and metalloproteases/matrix metalloproteases (ADAMS/MMPs). Examples of the resulting different downstream signaling events are depicted in the figure. FAK, focal adhesion kinase; HB-EGF, heparin-binding epidermal growth factor-like growth factor; mTOR, mammalian target of rapamycin; oxLDL, oxidized low-density lipoproteins; PKC, protein kinase C; TGF, transforming growth factor. 114

www.co-nephrolhypertens.com

Volume 23  Number 2  March 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Epidermal growth factor receptor in vasculature Schreier et al.

proinflammatory phenotype within the vessel wall occurs that resembles the beginning of vessel disease and leads to endothelial dysfunction, vascular stiffening and thickening of intima and media [10]. In parallel, aging increases EGFR expression and enhances the expression of proinflammatory markers such as transforming growth factor-beta (TGF-b) or intercellular adhesion molecule (ICAM) through an EGFR-dependent mechanism [11]. Conversely, reactive oxygen species (ROS)-defense capacity, measured as glucose-6-phosphate dehydrogenase activity, was reduced in VSMC from EGFR knockout mice [5 ]. EGFR-dependent VSMC proliferation and migration with enhanced deposition of extracellular matrix components may contribute to vascular remodeling, as has been suggested for vascular injury or disease [12]. Ligands such as EGF, heparin-binding epidermal growth factor-like growth factor (HB-EGF) [13,14 ,15,16] or other stimuli, such as angiotensin II [17–20], urokinase [16,21], thrombin [22], 15(S)hydroxyeicosatetraenic acid [23], adenosine triphophate [6], ROS [18,19,21,23,24] and oxidized lowdensity lipoproteins (oxLDL) [25,26], induce migration and proliferation of VSMC mediated by EGFR. Thrombin, for example, induces the activation of Gab1 via EGFR, with subsequent RhoA/cdc42/ Rac-1/PAK-1 activation, which causes stress fiber formation [22]. In the case of migration, the turnover of focal adhesions, mediated by focal adhesion kinase (FAK), plays a central role [14 ] and is supported by EGFR. Despite these examples, further investigations are required to elucidate the molecular mechanisms underlying EGFR-induced VSMC migration. There are several indications that EGFR mediates enhanced expression of matrix components when transactivated by members of the RAAS system such as angiotensin II and the mineralocorticoid receptor [27–30]. More recently, the importance of the ErbB-family members for the profibrotic TGF-b response in the vasculature has been recognized, for which a cross-talk with platelet-derived growth factor receptor (PDGFR) is required, and connective tissue growth factor (CTGF) has been suggested as a new EGFR ligand [31,32 ]. Consequently, combined inhibition of EGFR and PDGFR seems a promising approach to target organ fibrosis. Activated EGFR may contribute to fibrotic responses by supporting the induction of profibrotic mediators such as plasminogen activator inhibitor-1 (PAI-1) that also influences VSMC migration, proliferation and apoptosis [33]. In addition to inducing angiogenic effects in tumors and wounds [34–37], the EGFR contributes to the angiogenic effect of vasoactive petides such as angiotensin II, chemokines, and blood coagulation factor XII [38–41]. &&

&&

&&

&&

ENDOTHELIAL CELLS In endothelial cells, enhanced EGFR activation has been associated with nitric oxide (NO) homeostasis, which is important as an attenuator of endothelial activation and influences inflammation, migration, proliferation and angiogenesis [42]. On the one hand, EGFR has been shown to mediate vasoprotective antiatherosclerotic effects. In bovine aortic, human umbilicalvein,and humanmicrovascularendothelial cells, estradiol nongenomically can induce endothelial nitric oxide synthase (eNOS) via activation of estrogen receptor (ER)-a and b followed by rapid activation of c-Src, MMP and EGFR transactivation [43]. On the other hand, under conditions favoring eNOS uncoupling, activation of eNOS can stimulate O2 – generation that furthers endothelial dysfunction and atherosclerosis. Factors favoring uncoupling include angiotensin II, aldosterone and aging, and may involve EGFR as has been shown for angiotensin II [44–46]. Additionally, stimuli such as 5-HETE and aldosterone can enhance ACE expression via EGFR, which leads to enhanced angiotensin II generation and endothelial dysfunction, thereby imbedding a potential positive feedback loop [47 ,48]. &

IN-VIVO DATA First indications of an in-vivo relevance of the EGFR in vascular homeostasis and pathophysiology came from studies in spontaneously hypertensive rats (SHR) that displayed polygenetic hypertension with vascular remodeling. Their VSMCs showed enhanced EGFR phosphorylation and proliferated more rapidly [49]. This effect was attributed to elevated angiotensin II and endothelin I concentrations acting on angiotensin II-type 1, endothelin-A and endothelin-B receptors to induce ROS production, c-Src phosphorylation, EGFR transactivation and MAPK activation, potentially leading to Gi alpha overexpression [50,51]. The next step in deciphering the relevance of EGFR for vascular biology and disease were experiments in waved-2 mice, a spontaneous mutant with 90% reduced EGFR tyrosine kinase activity in all tissues. No difference in mean basal blood pressure was observed but the vasoconstrictor response to endothelin-1 was weaker in EGFR-deficient mice or after EGFR inhibition, suggesting that EGFR mediates endothelin-induced vasoconstriction. Additionally, endothelin-1 was able to activate the procollagen-2(I)-gene in freshly isolated aortas in an EGFR- and ERK1/2-dependent manner. EGFR therefore seems to support fibrogenesis and contraction of the vascular wall [52]. Recently, more detailed investigations regarding the vascular phenotype of the EGFR were performed in waved-2 mice [53]. No difference in arterial wall

1062-4821 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

www.co-nephrolhypertens.com

115

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Pathophysiology of hypertension

structure and morphological parameters of the carotid artery was detected compared with controls and the maximal vascular contraction in response to KCl and phenylephrine was also comparable. However, eNOS expression was reduced in the aorta of waved-2 mice and was associated with an increase in oxidative stress. Moreover, aorta relaxation in response to acetylcholine was weaker in waved-2 mice, whereas the response to NO donors was similar, suggesting that EGFR activation may be beneficial under basal conditions. As expected, control animals responded to nephrectomy-aldosterone-salt treatment (NAS) with reduced eNOS expression and increased oxidative stress markers in the aorta, indicating endothelial dysfunction. Additionally, functional vasomotoric effects occurred, such as an impaired vasorelaxation in response to acetylcholine and a potentiation of the vasoconstrictive response to phenylephrine and angiotensin II. In waved-2 mice, no further changes in oxidative stress markers or eNOS expression were detected and the functional changes induced by NAS were blunted or unchanged compared to wildtype mice. These data suggest that EGFR is involved in NAS-induced functional changes in the vasculature without affecting remodeling. Nevertheless, EGFR deficiency resulted in increased collagen 1 and 3 mRNA in NAS-treated mice. As a more straightforward approach of testing the overall significance of the EGFR signaling pathway for vascular biology and disease, tissuespecific knockout mice were generated [5 ,54] as global EGFR knockout mice die during in-utero development or shortly after birth [55]. In an EGFR-SM22 VSMC knockout model, which allows differentiation between effects mediated by VSMC and endothelial cell EGFR, systolic blood pressure was not different between genotypes; however, total peripheral vascular resistance, diastolic blood pressure and mean blood pressure were reduced. Loss of VSMC-EGFR resulted in a dilated phenotype of small vessels with minor signs of fibrosis and inflammation, suggesting that in the vasculature EGFR contributes to the appropriate vascular wall architecture and vessel reactivity, thereby supporting a role in physiological vascular tone. No differences in NOX2 or NOX4 expression were detected, suggesting that changes in oxidative stress measured by Griol-Charhbili originate from the endothelium and not the VSMCs [5 ]. To summarize, the importance of EGFR for vascular tone and vascular remodeling can be deduced from the above studies. &&

inflammatory response and there are indications that EGFR transactivation contributes to this pathophysiology. The presence of HB-EGF and EGFR in atherosclerotic plaques from different species has been confirmed [15,22,56 ,57 ,58] and seem to be involved on multiple levels in plaque formation. In macrophages, the proinflammatory metalloprotease meprin-a induces the activation of EGFR [56 ]. In Ox-PAPC-stimulated human aortic endothelial cells, the expression of IL-8, inducing migration of macrophages into the vessel wall, is reduced if these cells are treated with siRNA against HB-EGF, EGFR or with AG1478, an EGFR inhibitor. It is reported that ox-PAPC can interact with ADAM 10, 19 and a disintegrin and metalloprotease with thrombospondin motifs (ADAMTS) 4 by covalent binding, which would be an additional mechanism for EGFR receptor transactivation [59 ]. Furthermore, oxLDL-induced and angiotensin II-type 1 receptor-mediated transformation of macrophages into foam cells can be inhibited by AG1478 [60]. IL-8 in turn can activate CXCR2 (interleukin-8-receptor b) and thereby induces HB-EGF shedding and EGFR transactivation, resulting in migration of microvascular endothelial cells [61]. In contrast to the brief EGFR transactivation achieved by ROS, EGFR transactivation by oxLDL lasts several hours, suggesting a different mechanism and damaging potential [62,63]. Nevertheless, H2O2-induced Src, EGFR and JNK activation was also shown to potentially mediate oxidative stress response [64]. An increase in vasoactive peptides such as endothelin is typical for patients with cardiovascular disease. Hsieh et al. report that this increase leads to enhanced transactivation of the EGFR followed by PI3K, Akt and MAPK activation, resulting in AP1-induced enhancement of COX2 (cyclooxygenase-2) activity and prostaglandin-E2 biosynthesis. Thereby, a proinflammatory milieu enhancing remodeling processes is promoted [65]. Stimuli enhancing neointima formation by EGFR transactivation include thrombin [22,66], 15-HETE [23], angiotensin II [67], uremic toxins and oxLDL[25,68]. Recently, Wong et al. [69 ] demonstrated that inhibition of the mammalian target of rapamycin (mTOR) induces migration and differentiation of vascular progenitor cells (Sca-1þ) through a CXCR4/EGFR/b-catenin pathway, suggesting an involvement in restenosis and possibly atherosclerosis. &

&&

&

&

&&

&&

ATHEROSCLEROSIS During the pathogenesis of atherosclerosis, oxLDL and fatty acids play a major role in initiating an 116

www.co-nephrolhypertens.com

DIABETES There is accumulating evidence that EGFR signaling participates in the pathogenesis of diabetic vascular complications. Case reports of patients receiving receptor tyrosine kinase inhibitors for the treatment of malignancies demonstrate an improvement of Volume 23  Number 2  March 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Epidermal growth factor receptor in vasculature Schreier et al.

preexisting type 2 diabetes [70–72]. In mesenteric vessels of diabetic rats, the EGFR was significantly overexpressed and EGFR inhibition attenuated the majority of gene expression changes detected in these rats compared with control animals [73]. Incubation of endothelial cells or VSMC with high glucose levels can lead to increased EGFR phosphorylation [74,75] via ROS-dependent c-Src phosphorylation, resulting in enhanced MAPK and PI3K activity as well as enhanced Gq/11 alpha expression [74,76,77]. In addition to glucose, fatty acids, advanced glycation end products (AGEs) and ROS are also elevated during diabetes and seem to participate in the pathogenesis of vascular complications in an EGFR-dependent fashion. Notably, glucose tolerance, insulin resistance and signaling were improved after long-term EGFR inhibition and subclinical inflammation was attenuated [78]. Additionally, AGEs stimulated NADPH oxidase with subsequent EGFR-dependent PKC-delta and ERK1/2 activation and enhanced proinflammatory NF-KB signaling in endothelial cells [79,80]. Similarly, leptin, known to be elevated in patients with metabolic disorders, can enhance EGFR signaling via ROS and c-Src as well as ErbB2 activity, providing another link between metabolic and cardiovascular disease [81]. In coronary and mesenteric arteries of diabetic mice, EGFR phosphorylation and pressure-induced myogenic tone were increased and endothelial function measured as eNOS expression and phosphorylation in response to shear stress and acetylcholine was impaired [75]. Perfused mesenteric beds and isolated renal artery ring segments of diabetic rats showed an EGFR-dependent increased response to vasoconstrictors and a decreased response to vasodilators [73,82]. In addition, mesenteric resistance arteries of diabetic mice showed increased NADPH oxidase activity and ER stress marker expression leading to impaired endothelium-dependent and independent relaxation [83 ]. In a fructose-induced insulin resistance rat model, the MMP-EGFR-ERK1/2 signaling pathway was activated and phosphorylation of the contractile protein myosin light chain II (MLCII) and the corresponding regulatory proteins P90RSK and SRF was enhanced, coinciding with hypertension that could be prevented by EGFR inhibition [74]. Vasoactive substances further enhanced expression of MLCII and myosin light chain kinase in insulin-resistant VSMC and may thereby aggravate the pathological effects induced by diabetes [74]. EGFR has also been suggested as mediating vascular dysfunction in diabetes as a heterodimer with ErbB2, as hyperreactivity to vasoconstrictors and reduced responsiveness to vasodilators in diabetic rats could be attenuated by EGFR or ErbB2 inhibitors [82,84,85]. &

Another consequence of elevated EGFR signaling in diabetes seems to be vascular remodeling, leading to smaller passive diameters of resistance arteries, higher wall thickness to lumen diameter ratios and an increased stiffness with elevated collagen type 1 content. Treatment with AG1478 attenuated the morphological alterations [86]. Impaired blood flow recovery, vascular and capillary density and endothelial nitric oxide synthase activity after unilateral femoral artery ligation in diabetic db-db- mice could be significantly normalized by inhibition of EGFR and ERK1/2 activity, suggesting that EGFR und ERK1/2 inhibitors may be a therapeutic option in ischemia-induced vascular pathology in diabetes type 2 [87 ]. &&

SYSTEMIC HYPERTENSION The effect of EGF and HB-EGF on blood pressure seems to be primarily vasoconstrictive but vasodilator effects have been reported as well [88–91]. Under physiological conditions, EGFR shows little influence on systolic blood pressure [53,74,92,93]. However, in VSMC-specific knockout mice, a reduction in diastolic blood pressure without alteration in systolic blood pressure was demonstrated [5 ]. EGFR-induced vasoconstriction was observed in diseased vessels and different settings of experimental hypertension such as an infusion of angiotensin II, endothelin-1, aldosterone and phenylephrine [53,73,94–96]. Under these pathophysiological conditions, EGFR receptor tyrosine kinase blockade attenuated elevated blood pressure. The EGFR may also affect blood pressure through its actions on vessel remodeling as indicated above. &&

PULMONARY HYPERTENSION Pulmonary arterial hypertension (PAH) is elicited by a disequilibrium between vasoconstrictors and vasodilators and a chronic-proliferative component leading to vascular remodeling of small lung vessels. Different receptor tyrosine kinases have been suggested as important signal mediators and promising therapeutic targets, among them the EGFR [97,98 ,99]. In rats with monocrotaline-induced PAH, some EGFR/HER2 inhibitors led to an increased survival with a decrease in muscularization of small arteries and a reduction in pulmonary pressure and right ventricular hypertrophy, whereas others showed no beneficial effect [100]. EGFR inhibitors such as erlotinib and gefitinib reduced PASMC proliferation but showed no, or only slight, reduction in vascular remodeling measured as medial wall thickness and muscularization of pulmonary arteries [101]. Hypoxia, another inductor of PAH, has been identified as a trigger for EGFR activation and

1062-4821 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

&

www.co-nephrolhypertens.com

117

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Pathophysiology of hypertension

Vascular homeostasis • Vascular tone • ROS defense • NO balance

Endothelial cells • NO homeostasis • Endothelial dysfunction • Inflammation • Angiogenesis? VSMC • Vascular tone • ROS homeostasis • Inflammation • Migration • Proliferation • Fibrosis

Impact of EGFR

Atherosclerosis • Transactivation: oxLDL/fatty acids • IL-8 expression h • Migration: macrophages, VSMC, vascular progenitor cells • Macrophages g foam cells • Neointima formation h Diabetes (vessels) • Transactivation: glucose, fatty acids, AGEs, ROS • Proinflammatory • Endothelial dysfunction • Myogenic tone h • Vascular remodeling h

• Vascular tone • Oxidative stress • Vascular remodeling • Endothelial dysfunction • Fibrosis • Systemic hypertension

Pulmonary hypertension • Transactivation: hypoxia • Vascular remodeling h • Vasoconstrictor response h • NO availability i • Proinflammatory

FIGURE 2. Summary of the impact of the epidermal growth factor receptor on the vasculature. AGEs, advanced glycation end products; EGFR, epidermal growth factor receptor; NO, nitric oxide; oxLDL, oxidized low-density lipoproteins; ROS, reactive oxygen species; VSMC, vascular smooth muscle cells.

expression, leading to an increased vasoconstrictor response in pulmonary arteries by EGFR-dependent activation of Rac1 and NOX2 and subsequent calcium sensitization of myofilaments [98 ]. Hypoxia also induced EGFR-dependent proliferation of pulmonary endothelial cells and VSMCs, leading to wall thickening and distal muscularization in fetal pulmonary arteries, possibly by enhancing the proliferative response of SMC to such mitogens as PDGF, FGF-2 and EGF [102,103]. EGFR also mediated inflammation-dependent arginase activation, thereby reducing arginin and therefore NO availability [103,104]. In addition, EGFR has also been shown to interact with cytokine secretion, chemokine receptor activation, NFKB-/STAT3, PAF and PDGFR signaling during inflammation [105–107]. Furthermore, thrombospondin 1 can activate EGFR/HER2 and thereby cause phosphorylation of zonula adherens proteins and opening of the paracellular pathway in pulmonary microvascular endothelial cells [108]. Overall, EGFR is involved in the signaling events leading to pulmonary hypertension and therefore multireceptor tyrosine kinase inhibitors seem to be a promising therapeutic target for pulmonary hypertension. &

CONCLUSION The EGFR participates in a cross-talk with the signaling of G-protein-coupled receptors, steroid receptors 118

www.co-nephrolhypertens.com

and other (receptor) tyrosine kinases. In the vasculature, the EGFR is essential for adequate basal vascular tone and tissue homeostasis (Fig. 2). In contrast, the EGFR may also be involved in induction of proinflammatory mediators, altered secretion of matrix components, enhanced proliferation, and migration of cells and vessel remodeling, thereby promoting vascular diseases such as atherosclerosis and hypertension. Overall, the EGFR is a versatile signaling pathway integrator that is required for basal vascular homeostasis and function but that may also be involved in pathophysiological actions. Acknowledgements None. Conflicts of interest This work was funded by the DFG grant GE 905/19–1. There are no conflicts of interest.

REFERENCES AND RECOMMENDED READING Papers of particular interest, published within the annual period of review, have been highlighted as: & of special interest && of outstanding interest 1. Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell 2010; 141:1117–1134. 2. Burgess AW, Cho HS, Eigenbrot C, et al. An open-and-shut case? Recent insights into the activation of EGF/ErbB receptors. Mol Cell 2003; 12:541– 552.

Volume 23  Number 2  March 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Epidermal growth factor receptor in vasculature Schreier et al. 3. Dreux AC, Lamb DJ, Modjtahedi H, Ferns GAA. The epidermal growth factor receptors and their family of ligands: their putative role in atherogenesis. Atherosclerosis 2006; 186:38–53. 4. Batchu SN, Korshunov VA. Novel tyrosine kinase signaling pathways: implications in vascular remodeling. Curr Opin Nephrol Hypertens 2012; 21:122–127. 5. Schreier B, Rabe S, Schneider B, et al. Loss of epidermal growth factor && receptor in vascular smooth muscle cells and cardiomyocytes causes arterial hypotension and cardiac hypertrophy. Hypertension 2013; 61:333–340. This is the first study using a VSMC-specific EGFR knockout mouse model to characterize the in-vivo impact of the EGFR for vascular structure and function. 6. Lin CC, Lin WN, Cheng SE, et al. Transactivation of EGFR/PI3K/Akt involved in ATP-induced inflammatory protein expression and cell motility. J Cell Physiol 2012; 227:1628–1638. 7. Yogi A, Callera GE, Aranha AB, et al. Sphingosine-1-phosphate-induced inflammation involves receptor tyrosine kinase transactivation in vascular cells: upregulation in hypertension. Hypertension 2011; 57:809–818. 8. Fiebeler A, Luft FC. The mineralocorticoid receptor oxidative stress. Heart Failure Rev 2005; 10:47–52. 9. Sanada F, Taniyama Y, Iekushi K, et al. Negative action of hepatocyte growth factor/c-Met system on angiotensin II signaling via ligand-dependent epithelial growth factor receptor degradation mechanism in vascular smooth muscle cells. Circ Res 2009; 105:667–675. 10. Lakatta EG. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: part III: cellular and molecular clues to heart and arterial aging. Circulation 2003; 107:490–497. 11. Krug AW, Allenhofer L, Monticone R, et al. Elevated mineralocorticoid receptor activity in aged rat vascular smooth muscle cells promotes a proinflammatory phenotype via extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase and epidermal growth factor receptordependent pathways. Hypertension 2010; 55:1476–1483. 12. Gerthoffer WT. Mechanisms of vascular smooth muscle cell migration. Circ Res 2007; 100:607–621. 13. Tu L, De Man FS, Girerd B, et al. A critical role for p130Cas in the progression of pulmonary hypertension in humans and rodents. Am J Respir Crit Care Med 2012; 186:666–676. 14. Song GJ, Leslie KL, Barrick S, et al. EBP50 promotes focal adhesion turnover && and vascular smooth muscle cells migration. J Mol Cell Cardiol 2012; 53:809–819. The study provides new insights into the mechanism of EGFR-dependent migration by showing that EGFR is involved in focal adhesion turnover. 15. Shafi S, Lamb D, Modjtahedi H, Ferns G. Periadventitial delivery of anti-EGF receptor antibody inhibits neointimal macrophage accumulation after angioplasty in a hypercholesterolaemic rabbit. Int J Exp Pathol 2010; 91:224– 234. 16. Bakken AM, Protack CD, Roztocil E, et al. Cell migration in response to the amino-terminal fragment of urokinase requires epidermal growth factor receptor activation through an ADAM-mediated mechanism. J Vasc Surg 2009; 49:1296–1303. 17. Takaguri A, Shirai H, Kimura K, et al. Caveolin-1 negatively regulates a metalloprotease-dependent epidermal growth factor receptor transactivation by angiotensin II. J Mol Cell Cardiol 2011; 50:545–551. 18. Shimizu H, Hirose Y, Goto S, et al. Indoxyl sulfate enhances angiotensin II signaling through upregulation of epidermal growth factor receptor expression in vascular smooth muscle cells. Life Sci 2012; 91 (5–6):172–177. 19. Sakurada T, Ishizawa K, Imanishi M, et al. Nitrosonifedipine ameliorates angiotensin II-induced vascular remodeling via antioxidative effects. Naunyn Schmiedebergs Arch Pharmacol 2013; 386:29–39. 20. Mugabe BE, Yaghini FA, Song CY, et al. Angiotensin II-induced migration of vascular smooth muscle cells is mediated by p38 mitogen-activated protein kinase-activated c-Src through spleen tyrosine kinase and epidermal growth factor receptor transactivation. J Pharmacol Exp Ther 2010; 332:116–124. 21. Duru EA, Fu Y, Davies MG. Urokinase requires NAD(P)H oxidase to transactivate the epidermal growth factor receptor. Surgery 2012; 152:879–885. 22. Wang D, Paria BC, Zhang Q, et al. A role for Gab1/SHP2 in thrombin activation of PAK1: gene transfer of kinase-dead PAK1 inhibits injury-induced restenosis. Circ Res 2009; 104:1066–1075. 23. Singh NK, Wang D, Kundumani-Sridharan V, et al. 15-Lipoxygenase-1enhanced Src-Janus kinase 2-signal transducer and activator of transcription 3 stimulation and monocyte chemoattractant protein-1 expression require redox-sensitive activation of epidermal growth factor receptor in vascular wall remodeling. J Biol Chem 2011; 286:22478–22488. 24. Jagadeesha DK, Takapoo M, Banfi B, et al. Nox1 transactivation of epidermal growth factor receptor promotes N-cadherin shedding and smooth muscle cell migration. Cardiovasc Res 2012; 93:406–413. 25. Suc I, Meilhac O, Lajoie-mazenc I, et al. Activation of EGF receptor by oxidized LDL. FASEB J 1998; 12:665–671. 26. Liao PL, Cheng YW, Li CH, et al. 7-Ketocholesterol and cholesterol-5alpha,6alpha-epoxide induce smooth muscle cell migration and proliferation through the epidermal growth factor receptor/phosphoinositide 3-kinase/Akt signaling pathways. Toxicol Lett 2010; 197:88–96. 27. Gekle M, Mildenberger S, Freudinger R, Grossmann C. Altered collagen homeostasis in human aortic smooth muscle cells (HAoSMCs) induced by aldosterone. Pfluegers Archiv Eur J Physiol 2007; 454:403–413.

28. Grossmann C, Krug AW, Freudinger R, et al. Aldosterone-induced EGFR expression: interaction between the human mineralocorticoid receptor and the human EGFR promoter. Am J Physiol Endocrinol Metab 2007; 292:E1790–E1800. 29. Letavernier E, Perez J, Bellocq A, et al. Targeting the calpain/calpastatin system as a new strategy to prevent cardiovascular remodeling in angiotensin II-induced hypertension. Circ Res 2008; 102:720–728. 30. Schreier B, Rabe S, Schneider B, et al. Aldosterone/NaCl-induced renal and cardiac fibrosis is modulated by TGF-[beta] responsiveness of T cells. Hypertens Res 2011; 34:623–629. 31. Rayego-Mateos S, Rodrigues-Diez R, Morgado-Pascual JL, et al. Connective tissue growth factor is a new ligand of epidermal growth factor receptor. J Mol Cell Biol 2013; 5:323–335. 32. Andrianifahanana M, Wilkes MC, Gupta SK, et al. Profibrotic TGF beta && responses require the cooperative action of PDGF and ErbB receptor tyrosine kinases. FASEB J 2013; 27:4444–4454. The authors show that for a profibrotic TGF-b response, a complex interaction between PDGF and ErbB receptors is necessary, suggesting that targeting multiple pathways may be a promising option to treat fibrosis. 33. Samarakoon R, Dobberfuhl AD, Cooley C, et al. Induction of renal fibrotic genes by TGF-beta1 requires EGFR activation, p53 and reactive oxygen species. Cell Signall 2013; 25:2198–2209. 34. Karashima T, Sweeney P, Slaton JW, et al. Inhibition of angiogenesis by the antiepidermal growth factor receptor antibody ImClone C225 in androgenindependent prostate cancer growing orthotopically in nude mice. Clin Cancer Res 2002; 8:1253–1264. 35. Huang SM, Harari PM. Modulation of radiation response after epidermal growth factor receptor blockade in squamous cell carcinomas: inhibition of damage repair, cell cycle kinetics, and tumor angiogenesis. Clin Cancer Res 2000; 6:2166–2174. 36. Repertinger SK, Campagnaro E, Fuhrman J, et al. EGFR enhances early healing after cutaneous incisional wounding. J Investig Dermatol 2004; 123:982–989. 37. Casanova ML, Larcher F, Casanova B, et al. A critical role for ras-mediated, epidermal growth factor receptor-dependent angiogenesis in mouse skin carcinogenesis. Cancer Res 2002; 62:3402–3407. 38. Michaelis UR, Fisslthaler B, Medhora M, et al. Cytochrome P450 2C9derived epoxyeicosatrienoic acids induce angiogenesis via cross-talk with the epidermal growth factor receptor. FASEB J 2003; 17:770–772. 39. LaRusch GA, Mahdi F, Shariat-Madar Z, et al. Factor XII stimulates ERK1/2 and Akt through uPAR, integrins, and the EGFR to initiate angiogenesis. Blood 2010; 115:5111–5120. 40. McMellen ME, Wakeman D, Erwin CR, et al. Epidermal growth factor receptor signaling modulates chemokine (CXC) ligand 5 expression and is associated with villus angiogenesis after small bowel resection. Surgery 2010; 148:364–370. 41. Buharalioglu C, Song C, Yaghini F, et al. Angiotensin II-induced process of angiogenesis is mediated by spleen tyrosine kinase via VEGF receptor-1 phosphorylation. Am J Physiol Heart Circ Physiol 2011; 301:H1043–H1055. 42. Tai SC, Robb GB, Marsden PA. Endothelial nitric oxide synthase: a new paradigm for gene regulation in the injured blood vessel. Arterioscler Thromb Vasc Biol 2004; 24:405–412. 43. Klinge CM, Blankenship KA, Risinger KE, et al. Resveratrol and estradiol rapidly activate MAPK signaling through estrogen receptors alpha and beta in endothelial cells. J Biol Chem 2005; 280:7460–7468. 44. Yang YM, Huang A, Kaley G, Sun D. eNOS uncoupling and endothelial dysfunction in aged vessels. Am J Physiol Heart Circ Physiol 2009; 297:H1829– H1836. 45. Nagata D, Takahashi M, Sawai K, et al. Molecular mechanism of the inhibitory effect of aldosterone on endothelial NO synthase activity. Hypertension 2006; 48:165–171. 46. Oak JH, Cai H. Attenuation of angiotensin II signaling recouples eNOS and inhibits nonendothelial NOX activity in diabetic mice. Diabetes 2007; 56:118–126. 47. Cheng J, Garcia V, Ding Y, et al. Induction of angiotensin-converting enzyme & and activation of the renin angiotensin system contribute to 20-hydroxyeicosatetraenoic acidmediated endothelial dysfunction. Arterioscler Thromb Vasc Biol 2012; 32:1917–1924. The contribution of (EGFR-dependent) activation of the renin-angiotensin-aldosterone system for 20 HETE-induced endothelial dysfunction is explored, providing a new mechanism with therapeutical potential. 48. Sugiyama T, Yoshimoto T, Tsuchiya K, et al. Aldosterone induces angiotensin converting enzyme (ACE) gene expression via JAK2-dependent pathway in rat endothelial cells. Endocrinology 2005; 146:3900–3906. 49. Limas C, Westrum B, Limas CJ. The evolution of vascular changes in the spontaneously hypertensive rat. Am J Pathol 1980; 98:357–384. 50. Li Y, Levesque LO, Anand-Srivastava MB. Epidermal growth factor receptor transactivation by endogenous vasoactive peptides contributes to hyperproliferation of vascular smooth muscle cells of SHR. Am J Physiol Heart Circ Physiol 2010; 299:H1959–H1967. 51. Sandoval YHG, Li Y, Anand-Srivastava MB. Transactivation of epidermal growth factor receptor by enhanced levels of endogenous angiotensin II contributes to the overexpression of Gialpha proteins in vascular smooth muscle cells from SHR. Cell Signall 2011; 23:1716–1726.

1062-4821 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

www.co-nephrolhypertens.com

119

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Pathophysiology of hypertension 52. Francois H, Placier S, Flamant M, et al. Prevention of renal vascular and glomerular fibrosis by epidermal growth factor receptor inhibition. FASEB J 2004; 18:926–928. 53. Griol-Charhbili V, Fassot C, Messaoudi S, et al. Epidermal growth factor receptor mediates the vascular dysfunction but not the remodeling induced by aldosterone/salt. Hypertension 2011; 57:238–244. 54. Schreier B, Doehler M, Rabe S, et al. Consequences of epidermal growth factor receptor (ErbB1) loss for vascular smooth muscle cells from mice with targeted deletion of ErbB1. Arterioscler Thromb Vasc Biol 2011; 31:1643– 1652. 55. Threadgill DW, Dlugosz AA, Hansen LA, et al. Targeted disruption of mouse EGF receptor: effect of genetic background on mutant phenotype. Science 1995; 269:230–234. 56. Gao P, Wang XM, Qian DH, et al. Induction of oxidative stress by oxidized & LDL via meprin alpha-activated epidermal growth factor receptor in macrophages. Cardiovasc Res 2013; 97:533–543. oxLDL via meprin alpha induces oxidative stress in macrophages through HB-EGF release and EGFR transactivation and thereby promotes atherosclerotic plaque formation. 57. Stanic B, Pandey D, Fulton DJ, Miller FJ. Increased epidermal growth && factorlike ligands are associated with elevated vascular nicotinamide adenine dinucleotide phosphate oxidase in a primate model of atherosclerosis. Arterioscler Thromb Vasc Biol 2012; 32:2452–2460. In primates, a high-fat diet increases EGF-like ligand shedding, EGFR activation and NADPH-oxidase activity thereby modulating atherogenesis. This could be reversed by switching the animals back to a normal fat diet. 58. Miyagawa J, Higashiyama S, Kawata S, et al. Localization of heparin-binding EGF-like growth factor in the smooth muscle cells and macrophages of human atherosclerotic plaques. J Clin Invest 1995; 95:404–411. 59. Lee S, Springstead JR, Parks BW, et al. Metalloproteinase processing of & HBEGF is a proximal event in the response of human aortic endothelial cells to oxidized phospholipids. Arterioscler Thromb Vasc Biol 2012; 32:1246– 1254. This is the first study to demonstrate that Ox-PAPC activates ADAM/HB-EGF/ EGFR, which then leads to induction of interleukin-8 in endothelium. 60. Osada-Oka M, Kita H, Yagi S, et al. Angiotensin AT1 receptor blockers suppress oxidized low-density lipoprotein-derived formation of foam cells. Eur J Pharmacol 2012; 679 (13):9–15. 61. Schraufstatter IU, Zhao M, Khaldoyanidi SK, Discipio RG. The chemokine CCL18 causes maturation of cultured monocytes to macrophages in the M2 spectrum. Immunology 2012; 135:287–298. 62. Gamou S, Shimizu N. Hydrogen peroxide preferentially enhances the tyrosine phosphorylation of epidermal growth factor receptor. FEBS Lett 1995; 357:161–164. 63. Sachsenmaier C, Radler-Pohl A, Zinck R, et al. Involvement of growth factor receptors in the mammalian UVC response. Cell 1994; 78:963–972. 64. Chen K, Vita JA, Berk BC, Keaney JF. c-Jun N-terminal kinase activation by hydrogen peroxide in endothelial cells involves Src-dependent epidermal growth factor receptor transactivation. J Biol Chem 2001; 276:16045– 16050. 65. Hsieh H, Lin C, Chan H, Yang C. c-Src-dependent EGF receptor transactivation contributes to ET-1-induced COX-2 expression in brain microvascular endothelial cells. J Neurinflammation 2012; 9:152. 66. Hsieh HL, Tung WH, Wu CY, et al. Thrombin induces EGF receptor expression and cell proliferation via a PKC(delta)/c-Src-dependent pathway in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2009; 29:1594–1601. 67. Fukuda D, Enomoto S, Hirata Y, et al. The angiotensin receptor blocker, telmisartan, reduces and stabilizes atherosclerosis in ApoE and AT1aR double deficient mice. Biomed Pharmacother 2010; 64:712–717. 68. Fries DM, Penha RG, Damico EA, et al. Oxidized low-density lipoprotein stimulates nitric oxide release by rabbit aortic endothelial cells. Biochem Biophys Res Comm 1995; 207:231–237. 69. Wong MM, Winkler B, Karamariti E, et al. Sirolimus stimulates vascular stem/ && progenitor cell migration and differentiation into smooth muscle cells via epidermal growth factor receptor/extracellular signal-regulated kinase/betacatenin signaling pathway. Arterioscler Thromb Vasc Biol 2013; 33:2397– 2406. The study demonstrates that rapamycin EGFR dependently increases differentiation of vascular progenitor cells and their migration into the intima of blood vessels, thereby providing a new mechanism for restenosis. 70. Templeton A, Braendle M, Cerny T, Gillessen S. Remission of diabetes while on sunitinib treatment for renal cell carcinoma. Ann Oncol 2008; 19:824– 825. 71. Costa DB, Huberman MS. Improvement of type 2 diabetes in a lung cancer patient treated with erlotinib. Diabetes Care 2006; 29:1711. 72. Veneri D, Franchini M, Bonora E. Imatinib and regression of type 2 diabetes. N Engl J Med 2005; 352:1049–1050. 73. Benter IF, Benboubetra M, Hollins A J, et al. Early inhibition of EGFR signaling prevents diabetes-induced up-regulation of multiple gene pathways in the mesenteric vasculature. Vasc Pharmacol 2009; 51:236–245. 74. Nagareddy PR, MacLeod KM, McNeill JH. GPCR agonist-induced transactivation of the EGFR upregulates MLC II expression and promotes hypertension in insulin-resistant rats. Cardiovasc Res 2010; 87:177–186.

120

www.co-nephrolhypertens.com

75. Belmadani S, Palen DI, Gonzalez-Villalobos RA, et al. Elevated epidermal growth factor receptor phosphorylation induces resistance artery dysfunction in diabetic db/db mice. Diabetes 2008; 57:1629–1637. 76. Konishi A, Berk BC. Epidermal growth factor receptor transactivation is regulated by glucose in vascular smooth muscle cells. J Biol Chem 2003; 278:35049–35056. 77. Descorbeth M, Anand-Srivastava MB. Role of growth factor receptor transactivation in high glucose-induced increased levels of Gq/11alpha and signaling in vascular smooth muscle cells. J Mol Cell Cardiol 2010; 49:221–233. 78. Prada PO, Ropelle ER, Mourao RH, et al. EGFR tyrosine kinase inhibitor (PD153035) improves glucose tolerance and insulin action in high-fat dietfed mice. Diabetes 2009; 58:2910–2919. 79. Cai W, Torreggiani M, Zhu L, et al. AGER1 regulates endothelial cell NADPH oxidase-dependent oxidant stress via PKC-delta: implications for vascular disease. Am J Physiol Cell Physiol 2010; 298:C624–C634. 80. Cai W, He JC, Zhu L, et al. Advanced glycation end product (AGE) receptor 1 suppresses cell oxidant stress and activation signaling via EGF receptor. Proc Natl Acad Sci 2006; 103:13801–13806. 81. Jamroz-Wisniewska A, Wojcicka G, Lowicka E, et al. Transactivation of epidermal growth factor receptor in vascular and renal systems in rats with experimental hyperleptinemia: role in leptin-induced hypertension. Biochem Pharmacol 2008; 75:1623–1638. 82. Benter IF, Yousif MHM, Hollins AJ, et al. Diabetes-induced renal vascular dysfunction is normalized by inhibition of epidermal growth factor receptor tyrosine kinase. J Vasc Res 2005; 42:284–291. 83. Galan RG, Kassan M, Choi SK, et al. A novel role for epidermal growth factor & receptor tyrosine kinase and its downstream endoplasmic reticulum stress in cardiac damage and microvascular dysfunction in type 1 diabetes mellitus. Hypertension 2012; 60:71–80. The work provides new insights into the relationship between EGFR activation, ER stress and microvascular endothelial dysfunction and cardiac fibrosis in type 1 diabetes mellitus. 84. Yousif MHM, Benter IF, Dunn KMJ, et al. Role of 20-hydroxyeicosatetraenoic acid in altering vascular reactivity in diabetes. Auton Autacoid Pharmacol 2009; 29 (1–2):1–12. 85. Akhtar S, Yousif MHM, Dhaunsi GS, et al. Activation of ErbB2 and downstream signalling via Rho kinases and ERK1/2 contributes to diabetesinduced vascular dysfunction. PLoS One 2013; 8:e67813. 86. Palen DI, Matrougui K. Role of elevated EGFR phosphorylation in the induction of structural remodelling and altered mechanical properties of resistance artery from type 2 diabetic mice. Diabetes Metab Res Rev 2008; 24:651–656. 87. Choi SK, Galan M, Partyka M, et al. Chronic inhibition of epidermal growth && factor receptor tyrosine kinase and extracellular signal-regulated kinases 1 and 2 (ERK1/2) augments vascular response to limb ischemia in type 2 diabetic mice. Am J Pathol 2012; 180:410–418. Data suggest that inhibition of EGFR and ERK1/2 may be a therapeutic option to protect from ischemia-induced vascular pathology in type 2 diabetes. 88. Berk BC, Brock TA, Webb RC, et al. Epidermal growth factor, a vascular smooth muscle mitogen, induces rat aortic contraction. J Clin Invest 1985; 75:1083–1086. 89. Florian JA, Watts SW. Epidermal growth factor: a potent vasoconstrictor in experimental hypertension. Am J Physiol 1999; 276:H976–H983. 90. Keiser JA, Ryan MJ. Hemodynamic effects of epidermal growth factor in conscious rats and monkeys. Proc Natl Acad Sci 1996; 93:4957– 4961. 91. Zhou Y, Brigstock D, Besner GE. Heparin-binding EGF-like growth factor is a potent dilator of terminal mesenteric arterioles. Microvasc Res 2009; 78:78– 85. 92. McEwen ST, Balus SF, Durand MJ, Lombard JH. Angiotensin II maintains cerebral vascular relaxation via EGF receptor transactivation and ERK1/2. Am J Physiol Heart Circ Physiol 2009; 297:H1296–H1303. 93. Yousif MHM, Benter IF, Akhtar S. The role of tyrosine kinase-mediated pathways in diabetes-induced alterations in responsiveness of rat carotid artery. Auton Autacoid Pharmacol 2005; 25:69–78. 94. Flamant M, Tharaux PL, Placier S, et al. Epidermal growth factor receptor transactivation mediates the tonic and fibrogenic effects of endothelin in the aortic wall of transgenic mice. FASEB J 2003; 17:327–329. 95. Kagiyama S, Eguchi S, Frank GD, et al. Angiotensin II-induced cardiac hypertrophy and hypertension are attenuated by epidermal growth factor receptor antisense. Circulation 2002; 106:909–912. 96. Ulu N, Gurdal H, Landheer SW, et al. Alpha1-Adrenoceptor-mediated contraction of rat aorta is partly mediated via transactivation of the epidermal growth factor receptor. Brit J Pharmacol 2010; 161:1301–1310. 97. Montani D, Chaumais MC, Guignabert C, et al. Targeted therapies in pulmonary arterial hypertension. Pharmacol Ther 2013. [Epub ahead of print] 98. Norton C, Broughton B, Jernigan N, et al. Enhanced depolarization-induced & pulmonary vasoconstriction following chronic hypoxia requires EGFR-dependent activation of NAD(P)H oxidase 2. Antioxid Redox Signal 2013; 18:1777–1788. This article describes a novel signaling cascade in VSMC by which chronic hypoxia enhances membrane depolarization-induced pulmonary Caþþ sensitization and vasoconstriction through EGFR-dependent stimulation of Rac1 and NOX2.

Volume 23  Number 2  March 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Epidermal growth factor receptor in vasculature Schreier et al. 99. Izikki M, Mercier O, Lecerf F, et al. The beneficial effect of suramin on monocrotaline-induced pulmonary hypertension in rats. PLoS One 2013; 8:e77073. 100. Merklinger SL, Jones PL, Martinez EC, Rabinovitch M. Epidermal growth factor receptor blockade mediates smooth muscle cell apoptosis and improves survival in rats with pulmonary hypertension. Circulation 2005; 112:423–431. 101. Dahal BK, Cornitescu T, Tretyn A, et al. Role of epidermal growth factor inhibition in experimental pulmonary hypertension. Am J Respir Crit Care Med 2010; 181:158–167. 102. Schultz K, Fanburg B, Beasley D. Hypoxia and hypoxia-inducible factor1alpha promote growth factor-induced proliferation of human vascular smooth muscle cells. Am J Physiol Heart Circ Physiol 2006; 290: H2528–H2534. 103. Nelin LD, Chicoine LG, Reber KM, et al. Cytokine-induced endothelial arginase expression is dependent on epidermal growth factor receptor. Am J Respir Cell Mol Biol 2005; 33:394–401.

104. Toby IT, Chicoine LG, Cui H, et al. Hypoxia-induced proliferation of human pulmonary microvascular endothelial cells depends on epidermal growth factor receptor tyrosine kinase activation. Am J Physiol Lung Cell Mol Physiol 2010; 298:L600–L606. 105. Janakidevi K, Fisher M, Del Vecchio P, et al. Endothelin-1 stimulates DNA synthesis and proliferation of pulmonary artery smooth muscle cells. Am J Physiol 1992; 263 (6 Pt 1):C1295–C1301. 106. Lee KS, Park JH, Lee S, et al. HB-EGF induces delayed STAT3 activation via NF-KB mediated IL-6 secretion in vascular smooth muscle cell. Biochim Biophys Acta 2007; 1773:1637–1644. 107. Robitaille G, Christin MS, Clement I, et al. Nuclear autoantigen CENP-B transactivation of the epidermal growth factor receptor via chemokine receptor 3 in vascular smooth muscle cells. Arthritis Rheum 2009; 60:2805–2816. 108. Garg P, Yang S, Liu A, et al. Thrombospondin-1 opens the paracellular pathway in pulmonary microvascular endothelia through EGFR/ErbB2 activation. Am J Physiol Lung Cell Mol Physiol 2011; 301:L79–L90.

1062-4821 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

www.co-nephrolhypertens.com

121

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Role of epidermal growth factor receptor in vascular structure and function.

The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase with a wide implication in tumor biology, wound healing and development. Bes...
1MB Sizes 0 Downloads 0 Views