An emerging role for Hippo-YAP signaling in cardiovascular development

2014-02-11 06:40JiliangZhou
THE JOURNAL OF BIOMEDICAL RESEARCH 2014年4期

Jiliang Zhou

Department of Pharmacology&Toxicology,Medical College of Georgia,Georgia Regents University,Augusta,GA 30912,USA.

An emerging role for Hippo-YAP signaling in cardiovascular development

Jiliang Zhou✉

Department of Pharmacology&Toxicology,Medical College of Georgia,Georgia Regents University,Augusta,GA 30912,USA.

The Hippo signaling pathway was originally discovered in Drosophila and shown to be critical for organ size con–trol and tumorigenesis.This pathway consists of a cascade of kinases and several adaptors that lead to the phosphor–ylation and inhibition,through nuclear exclusion,of the transcriptional cofactor Yorkie in Drosophila or YAP(yes associated protein)in mammals.Recent studies demonstrate that cardiac–specific deletion of the Hippo pathway kinase Mst(STE20–like protein kinases)co–activator WW45(WW domain–containing adaptor 45),Mst1,Mst2,or Lats2(large tumor suppressor homologue 2)in mice result in over–grown hearts with elevated cardiomyocyte pro–liferation.Consistent with these observations,over–expression of YAP in the mouse embryonic heart increases heart size and promotes cardiac regeneration and contractility after myocardial infarction by inducing cardiomyocyte pro–liferation,whereas deletion of YAP in the mouse heart impedes cardiomyocyte proliferation,causing myocardial hypoplasia and embryonic or premature lethality.YAP has also been shown to play an important role in the vascular system.Specific–deletion of YAP from vascular smooth muscle cells in mice results in aberrant development of large arteries with a hypoplastic arterial wall phenotype.Hippo–YAP signaling cross–talks with other signaling pathways such as IGF(insulin–like growth factor)and Wnt signaling to promote heart growth by increasing expression of cell cycle genes.The purpose of this review is to summarize these recent findings and discuss potential diagnostic or ther–apeutic strategies in cardiovascular system based on manipulating the Hippo–YAP signaling.

Hippo,YAP,cardiac development,vascular smooth muscle,proliferation

INTRODUCTION

The heart is the first organ to form during mamma–lian embryogenesis.Together with blood vessels,these circulatory organs provide nutrient perfusion necessary for further organogenesis.In the mouse,heart forma–tion starts from cardiogenic precursor cells located in the cardiac crescent,to form the linear heart tube at about embryonic day(E)8.0.The heart tube then initi–ates rightward looping and chamber specification,fol–lowed by septation,and valvulogenesis[1].The mouse embryo starts to form blood vessels through vasculo–genesis at E7.5.A coordination of multiple cell types including endothelial and smooth muscle cells(SMCs) is required for a proper pattering of the vasculature. Soon after establishment of a capillary network by endothelial cells,vascular SMC progenitors begin to invest the vessel wall through migration and prolifera–tion,meanwhile undergoing differentiation to acquire a unique array of smooth muscle–contractile proteins[2]. Understanding the transcriptional networks that control cardiovascular development and their underlying controlmechanisms is not only critical for identifying genes whose defects cause congenital cardiovascular disorders in humans but also provides a molecular basis for diag–nostic and therapeutic strategies.

The Hippo signaling pathway was originally dis–covered in Drosophila and subsequent genetic and biochemical studies revealed that the pathway compo–nents and functions are evolutionarily conserved through humans[3].This pathway includes a cascade of kinases and several adaptors that lead to the phosphor–ylation and inhibition of the transcriptional cofactor Yorkie by preventing its nuclear translocation.The core components of the pathway consist of the Ste20–like kinases Hippo and its regulatory protein Salvador,the downstream kinase Warts and its regulatory protein Mats.The Hippo–Salvador complex phosphorylates the Warts–Mats complex,which,in turn,phosphorylates the oncoprotein Yorkie,which inactivates Yorkie by retaining it in the cytoplasm.In the absence of Hippo signaling,unphosphorylated,active Yorkie translocates into the nucleus and functions as a co–activator with the TEAD(TEA domain)family transcription factor Scalloped,to induce genes that promote cell growth and oncogenic transformation[4,5].Due to the high evo–lutionary conversation,Hippo,Salvador,Warts,Mats, Yorkie and Scalloped in Drosophila are structurally and functionally homologous to mammalian MST1/2, WW45,LATS1/2,Mob1(Mps one binder),YAP and TEAD,respectively.YAP has been shown to be a cri–tical determinant of organ size[6].YAP is also considered as a potent oncoprotein by promoting cell growth and inhibiting cell apoptosis[7].

Recently,several studies showed that the Hippo–YAP pathway plays a critical role in mouse cardiovas–cular development.The focus of this review is to sum–marize recent progress in understanding the molecular basis underlying Hippo–YAP signaling in cardiovas–cular development.Other functions of Hippo–YAP signaling have been discussed in several recent excel–lent review articles[8–10].

ROLE OF HIPPO PATHWAY COMPONENTS IN MOUSE CARDIAC DEVELOPMENT

Extensive genetic studies have shown Hippo pathway components are critical for mouse embryogenesis and tumorigenesis in mice.Salvador is a key component of the Hippo pathway that restricts cell proliferation and promotes apoptosis[11,12].Conventional ablation of the Salvador homologue WW45 in mice leads to embryonic lethality with hyper–proliferative epithelial cells of the skin and intestine accompanied by immature differentiation[13].Many organs in the WW45 mutant embryos display hyperplasia.However,this study did not examine the possibility that embryonic lethality is attributable to a cardiac phenotype as it would be a little surprising if the skin or intestinal phenotypes resulted in embryonic mortality.Although Mst1/2 are tumor sup–pressors due to their ability to restrict cell prolifera–tion[14],unexpectedly,mice lacking both Mst1 and 2 do not exhibit a hyper–proliferative phenotype.Rather, the Mst1/2 double knockout mice exhibit severe growth retardation,failed placental development,impaired yolk sac/embryo vascular patterning and primitive hemato–poiesis and die at approximately E8.5[15].The majority of mice with global deletion of the Mst1/2 downstream kinase Lats1 die by postnatal day 1 and surviving mice develop large soft tissue sarcomas,ovarian stromal cell tumors andare susceptible to carcinogenic treatments[16]. Lats2 global knockout mice are embryonic lethal between E10.5–12.5 and display pleiotrophic develop–mental defects[17],suggesting unique functions of Lats2 in development.The severe phonotypes observed in the mice harboring genetic deletion of Hippo pathway components described above suggest Hippo signaling is critical for embryogenesis and tumorigenesis in mice.

To begin to examine the effects of the Hippo pathway more specifically during cardiac development a number of groups have generated cardiac–specific knockouts of the pathway components.The cardiac specific knock–out of the Mst kinase co–activator WW45(Salvador homologue in mammals)in mice resulted in over–grown hearts with elevated cardiomyocyte proliferation[18]. WW45 cardiac–specific KO hearts display expansion of trabecular and subcompact ventricular myocardial layers and have thickened ventricular walls without a change in cardiomyocyte cell size.Lats2 and Mst1/2 cardiac–conditional KO hearts have similar myocardial expansion phenotypes[18].Mechanistically,the study reported that the Hippo signaling interacts with the Wnt signaling effector β–catenin to regulate several cell cycle genes thereby promoting cardiomyocytes cell prolifera–tion.However,how the Hippo signaling cross–talks to Wnt signaling in the heart is still elusive.Furthermore, the function of upstream components above Mst/ WW45 in Hippo signaling in the cardiac and vascular development remains to be determined.

ROLE OF YAP IN MOUSE CARDIOVASCULAR DEVELOPMENT

YAP null mice die at E8.5 with defects in yolk sac vasculogenesis,chorioallantoic attachment,and embryonic axis elongation[19],precluding an analysis of heart development.Cardiac–specific inactivation ofYAP,early during development mediated by either Nkx2.5CreorTnnt2Cre,impedescardiomyocyteprolif–eration,causing myocardial hypoplasia and embryonic lethality in mice[20,21].By using α–MHC cre,which deletes YAP at later stages of heart development,YAP knockout mice complete embryonic development but mutant mice died prematurely starting around 11 weeks of age due to dilated cardiomyopathy resulted from thinning ventricu–lar walls[22,23].These YAP knockout mice also exhibit impaired neonatal heart regeneration,leading to a default fibrotic response and exacerbated myocardial infarction induced cardiac impairment[22,23].Similar to the mice with loss–of–function of Hippo pathway kinases,over–expres–sion of YAP in mouse embryonic heart increases heart size by promoting cardiomyocyte proliferation without significantly changing cardiomyocyte size[20,22].The gain–of–function YAP phenotype in the mouse heart lar–gely phenocopies the cardiac–specific knockout of Salvador,Mst1/2 and Lats2 in the mouse heart. However,the cardiac–specific WW45knockout mice dis–play a ventricular septal defect[18],which is not observed in mice in which YAP overexpression is driven by either the cardiac–specific α-MHC,β-MHC or Tnnt2 promo–ters.This discrepancy suggests that there must be other effectors of down–stream of Hippo signaling in the heart in addition to YAP,although YAP is the major effector that regulates cardiomyocyte proliferation.Consistent with the impaired regeneration seen in YAP knockout mice,over–expression of YAP in the mouse heart pro–motes cardiac regeneration and improves contractility after myocardial infarction[22].These exciting findings suggest that therapeutic strategies to increase YAP expression or activate the existing YAP would be bene–ficial for treatment of myocardial infarctions.Together, these gain–of–function and loss–of–function assays unequivocally demonstrate that YAP is necessary for embryonic and neonatal heart growth.Mechanistically, all of reports described above unambiguously demon–strate that YAP functions in cardiomyocytes to promote proliferative gene programs.Although manipulation of YAP expression levels in the heart provides a potential strategy to repair heart injury by inducing cardiomyocyte proliferation,the role of upstream Hippo kinases in the cardiac regeneration warrants further investigation.

In addition to being important for cardiac develop–ment,we have recently shown that YAP also plays an important role in vascular development[24].Previously we found that YAP plays a novel integrative role in smooth muscle phenotypic modulation by inhibiting smooth muscle–specific gene expression while pro–moting smooth muscle proliferation and migration in vitro and in vivo[25].Similar to the‘‘synthetic’’pheno–type that SMCs exhibit in response to arterial injury, SMCs are highly proliferative and migratory during vasculogenesis.Given the important role of YAP in regulating cell proliferation,we tested the importance of YAP in regulating SMC proliferation during vascular development.We found that cardiac and smooth muscle cell–specific deletion of YAP directed by SM22α–Cre resulted in perinatal lethality in mice due to profound cardiac defects.The cardiac/smooth muscle–specific YAP knockout mice also displayed severe vascular defects[24].Unexpectedly,deletion of YAP in mouse vas–cular SMCs did not alter the expression of smooth mus–cle–contractile proteins or the proliferative genes that are involved in cardiomyocyte proliferation[18,20,21].Rather, YAP knockout induced expression of a subset of cell cycle arrest genes including Gpr132[24].Over–expression of Gpr132 attenuated SMC growth by arresting cell cycle in G0/G1 phase,suggesting deletion of YAP induced impairment of SMC proliferation is mediated, at least in part,by induction of Gpr132 expression. Our study not only suggests a crucial role of YAP in arterial development in mice,but also identifies a novel mechanism through which YAP promotes vascular SMC proliferation by repressing cell cycle arrest gene expres–sion.Due to the embryonic and perinatal lethality of the YAP mutant mice directed by the SM22α promoter dri–ven cre transgene,a smooth muscle–specific inducible KO model is required to further study role of YAP in adult smooth muscle cells.Whether deletion of YAP in adult SMCs can attenuate the vascular remodeling induced by vascular injury is currently under investiga–tion in our group by using an inducible SM MHC pro–moter–driven cre mouse line[26].

FUTURE PERSPECTIVES

In a relative short period of time,several studies have highlighted the importance of the Hippo–YAP pathway in heart development and regeneration.It is quite likely that mutation of genes or disregulation of the Hippo–YAP pathway may be involved in patho–genesis of congenital heart diseases associated with abnormalities of myocardial growth in humans. These studies suggest that analysis of Hippo–YAP components may be a useful diagnostic strategy for identification of congenital cardiovascular disorders in humans.Moreover,the strong effects of deletion of Hippo kinases or overexpression of YAP on cardio–myoctye proliferation offers great promise of targeting this pathway as a therapeutic strategy in cardiac regen–erative medicine.Simply,by activation of YAP or/and inactivation Hippo kinases in the heart,it may be possi–ble to promote cardiomyocyte proliferation to repair the heart after injury and thereby limit the deleterious effectsof myocardial infarctions.However,YAP′s ability to promote cell proliferation may be a‘‘double–edged sword’’for vascular SMCs because the increased prolif–eration of vascular SMCs is a major contributing event leading to the development of occlusive vascular dis–eases after arterial injury.YAP is significantly induced in rodent carotid artery injury models and deletion of YAP attenuates injury–induced neointima formation[25]. Therefore blocking the induction of YAP would be a potential therapeutic approach for ameliorating vascular occlusive diseases.These data suggest that any therapies aimed at targeting the Hippo–YAP pathway in the cardi–ovascular system will need to be spatially and temporally targeted carefully.

Acknowledgements

I′d like to thank my lifelong mentor Dr.Paul Herring for critically reading the manuscript.

[1] Xin M,Olson EN,Bassel–Duby R.Mending broken hearts:Cardiac development as a basis for adult heart regeneration and repair.Nat Rev Mol Cell Biol 2013;14: 529–41.

[2] Hungerford JE,Little CD.Developmental biology of the vascular smooth muscle cell:Building a multilayered vessel wall.Journal of vascular research 1999;36:2–27.

[3] Edgar BA.From cell structure to transcription:Hippo forges a new path.Cell 2006;124:267–73.

[4] Zhao B,Lei QY,Guan KL.The hippo–yap pathway:New connections between regulation of organ size and cancer. Curr Opin Cell Biol 2008;20:638–46.

[5] Pan D.The hippo signaling pathway in development and cancer.Developmental cell 2010;19:491–505.

[6] Dong J,Feldmann G,Huang J,Wu S,Zhang N,Comerford SA,et al.Elucidation of a universal size–control mechan–ism in drosophila and mammals.Cell 2007;130:1120–33.

[7] Zhao B,Li L,Lei Q,Guan KL.The hippo–yap pathway in organ size control and tumorigenesis:An updated version. Genes&development 2010;24:862–74.

[8] Harvey KF,Zhang X,Thomas DM.The hippo pathway and human cancer.Nature reviews.Cancer 2013;13:246–57.

[9] Yu FX,Guan KL.The hippo pathway:Regulators and regulations.Genes Dev 2013;27:355–71.

[10]Barry ER,Camargo FD.The hippo superhighway: Signaling crossroads converging on the hippo/yap path–way in stem cells and development.Curr Opin Cell Biol 2013;25:247–53.

[11]Kango–Singh M,Nolo R,Tao C,Verstreken P,Hiesinger PR,Bellen HJ,et al.Shar–pei mediates cell proliferation arrest during imaginal disc growth in drosophila. Development 2002;129:5719–30.

[12]Tapon N,Harvey KF,Bell DW,Wahrer DC,Schiripo TA,Haber D,et al.Salvador promotes both cell cycle exit and apoptosis in drosophila and is mutated in human can–cer cell lines.Cell 2002;110:467–78.

[13]Lee JH,Kim TS,Yang TH,Koo BK,Oh SP,Lee KP, et al.A crucial role of ww45 in developing epithelial tis–sues in the mouse.EMBO J 2008;27:1231–42.

[14]Wu S,Huang J,Dong J,Pan D.Hippo encodes a ste–20 family protein kinase that restricts cell proliferation and promotes apoptosis in conjunction with salvador and warts.Cell 2003;114:445–56.

[15]Oh S,Lee D,Kim T,Kim TS,Oh HJ,Hwang CY,et al. Crucial role for mst1 and mst2 kinases in early embryonic development of the mouse.Mol Cell Biol 2009;29:6309–20.

[16]St John MA,Tao W,Fei X,Fukumoto R,Carcangiu ML, Brownstein DG,et al.Mice deficient of lats1 develop soft–tissue sarcomas,ovarian tumours and pituitary dys–function.Nat Genet 1999;21:182–6.

[17]McPherson JP,Tamblyn L,Elia A,Migon E,Shehabeldin A,Matysiak–Zablocki E,et al.Lats2/kpm is required for embryonic development,proliferation control and geno–mic integrity.EMBO J 2004;23:3677–88.

[18]Heallen T,Zhang M,Wang J,Bonilla–Claudio M,Klysik E,Johnson RL,et al.Hippo pathway inhibits wnt signal–ing to restrain cardiomyocyte proliferation and heart size. Science 2011;332:458–61.

[19]Morin–Kensicki EM,Boone BN,Howell M,Stonebraker JR,Teed J,Alb JG,et al.Defects in yolk sac vasculogen–esis,chorioallantoic fusion,and embryonic axis elonga–tion in mice with targeted disruption of yap65.Mol Cell Biol 2006;26:77–87.

[20]von Gise A,Lin Z,Schlegelmilch K,Honor LB,Pan GM, Buck JN,et al.Yap1,the nuclear target of hippo signal–ing,stimulates heart growth through cardiomyocyte pro–liferation but not hypertrophy.Proc Natl Acad Sci U S A 2012;109:2394–9.

[21]Xin M,Kim Y,Sutherland LB,Qi X,McAnally J, Schwartz RJ,et al.Regulation of insulin–like growth factor signaling by yap governs cardiomyocyte prolifera–tion and embryonic heart size.Science signaling 2011; 4:ra70.

[22]Xin M,Kim Y,Sutherland LB,Murakami M,Qi X, McAnally J,et al.Hippo pathway effector yap promotes cardiac regeneration.Proc Natl Acad Sci U S A 2013;110: 13839–44.

[23]Del Re DP,Yang Y,Nakano N,Cho J,Zhai P, Yamamoto T,et al.Yes–associated protein isoform 1 (yap1)promotes cardiomyocyte survival and growth to protect against myocardial ischemic injury.J Biol Chem 2013;288:3977–88.

[24]Wang Y,Hu G,Liu F,Wang X,Wu M,Schwarz JJ,et al. Deletion of yap specifically in cardiac and vascular smooth muscle cells reveals a crucial role for yap in mouse cardi–ovascular development.Circ Res 2014;114:957–65.

[25]Wang X,Hu G,Gao X,Wang Y,Zhang W,Harmon EY, et al.The induction of yes–associated protein expression after arterial injury is crucial for smooth muscle phenoty–pic modulation and neointima formation.Arterioscler Thromb Vasc Biol 2012;32:2662–9.

[26]Wirth A,Benyo Z,Lukasova M,Leutgeb B,Wettschureck N,Gorbey S,et al.G12–g13–larg–mediated signaling in vascular smooth muscle is required for salt–induced hyper–tension.Nat Med 2008;14:64–8.

Received 04 February 2014,Accepted 24 February 2014,Epub 22 March 2014

This study was supported by a grant(No.R01HL109605)from the National Heart,Lung,and Blood Institute,NIH.

✉Corresponding author:Jiliang Zhou,MD/PhD,Department of Pharmacology&Toxicology,Medical College of Georgia,GeorgiaRegents University,CB–3628(Office);CB–3606(Lab),1459 Laney Walker Blvd,Augusta,GA 30912,USA.Tel/Fax:706–721–7582(Office)/ 706–721–7583(Lab)/706–721–2347,E–mail:JIZHOU@gru.edu.

The author reported no conflict of interests.

©2014 by the Journal of Biomedical Research.All rights reserved.

10.7555/JBR.28.20140020