<<

Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

Bone Morphogenetic in Vascular Homeostasis and Disease

Marie-Jose´Goumans,1 An Zwijsen,2,3 Peter ten Dijke,1,4 and Sabine Bailly5,6,7

1Department of Molecular Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands 2VIB Center for the Biology of Disease, 3000 Leuven, Belgium 3KU Leuven Department of Human Genetics, 3000 Leuven, Belgium 4Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands 5Institut National de la Sante´ et de la Recherche Me´cale (INSERM), U1036, 38000 Grenoble, France 6Laboratoire Biologie du Cancer et de l’Infection, Commissariat a` l’E´nergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France 7University of Grenoble Alpes, 38000 Grenoble, France Correspondence: [email protected]; [email protected]

It is well established that control of vascular morphogenesis and homeostasis is regulated by vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), Delta-like 4 (Dll4), angiopoietin, and ephrin signaling. It has become clear that signaling by bone morphogenetic proteins (BMPs), which have a long history of studies in bone and early heart development, are also essential for regulating vascular function. Indeed, mutations that cause deregulated BMP signaling are linked to two human vascular diseases, hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension. These observations are corroborated by data obtained with vascular cells in cell culture and in mouse models. BMPs are required for normal endothelial cell differentiation and for venous/arterial and lymphatic specification. In adult life, BMP signaling orchestrates neo- as well as vascular inflammation, remodeling, and calcification responses to shear and oxidative stress. This review emphasizes the pivotal role of BMPs in the vascular system, based on studies of mouse models and human vascular disorders.

one morphogenetic proteins (BMPs) were ing growth factor-b (TGF-b) family (Bragdon Boriginally discovered based on their capacity et al. 2011; Katagiri and Watabe 2016). Most to induce bone and cartilage formation at ec- BMPs have been reported to elicit some effects topic sites (Urist 1965). As their pleiotropic on vascular cells. Based on their sequence sim- functions have become apparent, it has been ilarity, receptor affinities and function, the BMP proposed to call them body morphogenetic polypeptides are classified into several proteins (Reddi 2005). BMPs comprise a dozen subgroups including the BMP-2 and -4 group; of the 33 known polypeptides of the transform- BMP-5, -6, -7, -8a, -8b group; BMP-9, -10

Editors: Rik Derynck and Kohei Miyazono Additional Perspectives on The Biology of the TGF-b Family available at www.cshperspectives.org Copyright # 2017 Cold Spring Harbor Laboratory Press; all rights reserved Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989

1 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

group; and BMP-3, -3b, -11, -12, -13, -14, -15, each perform highly specific functions (De and -16 group (Katagiri and Watabe 2016; Mo- Smet et al. 2009). ECs can transition between rikawa et al. 2016). Among the different BMPs, subtypes, which is termed EC plasticity. During BMP-2, BMP-4, BMP-6, and BMP-7 have been the activation phase of angiogenesis, increased shown to function in vascular biology. Addi- vascular permeability and basement membrane tionally, BMP-9 and -10 have emerged as crucial degradation allows EC migration and prolifera- factors in endothelial function and vascular dis- tion into the extracellular space. These new eases (David et al. 2009). sprouts are spearheaded by the tip cell and elon- Several reviews discuss the emerging role of gated by proliferation of the trailing stalk cells. In BMPs in vascular remodeling, focusing on vas- the resolution phase of angiogenesis, EC migra- cular biology (Garcia de Vinuesa et al. 2016; tion and proliferation are stalled, the basement Morrell et al. 2016) and their molecular targets membrane is reconstituted, and vessel matura- (Luo et al. 2015). Here, we will describe human tion is promoted. Lumen formation will occur vascular disorders and animal models that have via fusion of neighboring sprouts (Gebala et al. contributed to our understanding of the role of 2016). Next, mesenchymal cells are recruited BMP signaling in cardiovascular diseases and and subsequently differentiate into pericytes that may help us to find more effective treat- or VSMCs, which are attached to the newly ment modalities. A review on TGF-b signaling formed vessel (Eilken and Adams 2010; Korn in control of cardiovascular function has also and Augustin 2015). Although blood vessels been published (Goumans and ten Dijke 2017). predominantly remain quiescent throughout adult life, they retain the capacity to rapidly form new vasculature in response to injury or VASCULAR DEVELOPMENT IN A NUTSHELL pathological conditions. During this process, The vascular system circulates blood and lymph transitions between the resolution phase and throughout the body and is critical for tissue the activation phase of angiogenesis are deter- growth, homeostasis and repair. Blood vessels mined by an intricately regulated balance be- supply tissues with oxygen and nutrients, tween the inducers and the inhibitors of these whereas lymphatic vessels absorb and filter in- phases. terstitial fluids from these tissues. Blood vessels Angiogenesis requires the coordinated inte- comprise endothelial cells (ECs) and mural cells, gration of multiple signaling cascades, including such as vascular smooth muscle cells (VSMCs) the vascular endothelial growth factor (VEGF), and pericytes. Interactions and interplay be- fibroblast growth factor (FGF), Notch/Delta- tween ECs and mural cells play pivotal roles in like 4 (Dll4), angiopoietin, ephrin, vascular biology (Risau 1997; Carmeliet and TGF-b, and BMP pathways. The BMP pathway Jain 2011). plays an important role in maintaining vascular Vessel formation during embryonic devel- homeostasis and affects several processes, in- opment begins with vasculogenesis (i.e., the de cluding the endothelial responses to shear stress, novo formation of blood vessels from locally hypoxia, and inflammation. differentiating ECs). This is followed by angio- genesis, which entails remodeling of the honey- BONE MORPHOGENETIC PROTEINS comb-like pattern of the primary plexus into a stable branched network of vessels. The vascula- BMP ligands are synthesized as dimers of large ture further expands through sprouting angio- proproteins that include an amino-terminal sig- genesis (i.e., formation of new blood vessels nal peptide, a long propeptide, and a carboxy- from pre-existing ones) and vascular pruning terminal mature peptide that contains a cystine (Eilken and Adams 2010; Korn and Augustin knot (Mueller and Nickel 2012). These BMP 2015). Capillary sprouting requires interactions proproteins are proteolytically cleaved by pro- among three different EC subtypes—tip cells, convertases to generate the active dimeric form stalk cells, and quiescent phalanx cells—that (Miyazono et al. 2010). Although BMP process-

2 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

ing is predominantly intracellular, it might also Awell-established mechanism for regulating occur at the plasma membrane or extracellularly the extracellular availability of BMPs involves a (Yadinet al. 2016). Dimerization to generate the gradient of BMP antagonists that prevents BMP active ligand requires a covalent disulphide binding to receptors and BMP action (Chang bridge at the seventh conserved cysteine within 2016; Hinck et al. 2016). Multiple secreted pro- each monomer outside of the cystine knot struc- teins (e.g., , , , matrix ture formed by the six other cysteines. Active Gla protein [MGP], and ) can seques- BMPs are homodimeric but some heterodimers ter BMP ligands and impair their interactions (i.e., BMP-2/5, BMP-2/6, and BMP-2/7) are with receptor complexes. Other interacting pro- more potent than the corresponding homo- teins, such as cross-veinless 2, also known as dimers in certain experimental settings (Sieber BMP-binding EC precursor-derived regulator et al. 2009). Mature BMP dimers remain non- or BMPER, may exert more complex effects by covalently associated with their cleaved prodo- either decreasing or increasing BMP signaling mains. The fact that prodomains confer latency (Yao et al. 2012). BMPER was originally identi- to TGF-b (and some other family members) but fied in a screen for proteins that are differentially not to the BMPs remains unresolved and is cur- expressed in embryonic endothelial precursor rently being investigated for certain BMPs, in- cells (Moser et al. 2003), suggesting its potential cluding BMP-10 (Sengle et al. 2011; Jiang et al. role in vascular remodeling (see below). These 2016b; Yadin et al. 2016). The prodomain of different antagonists and agonists bind to BMPs BMP-9 does not inhibit its biological activity with different affinities, and can preferentially and is rapidly released from the mature protein interfere with one family member and not oth- on binding to the receptors (Kienast et al. 2016). ers. For example, noggin binds with strong af- Posttranslational modifications, including N- finity to BMP-2, BMP-4, and BMP-7, with lower and O-glycosylation, strongly influence BMP affinity to BMP-6, and not to BMP-9 and BMP- activity, because glycosylation is important for 10 (David et al. 2008; Song et al. 2010). Also, BMP secretion, prolongs a BMP’s half-life, and MGP can either inhibit or enhance the BMP-2 modulates BMP receptor binding and function and BMP-4 activity, depending on its concen- (Saremba et al. 2008). tration (Zebboudj et al. 2002; Yao et al. 2006). Regulation of cell differentiation, prolifera- tion, and apoptosis require appropriate ligand concentration and activity, as well as tight reg- BMP SIGNALING ulation of the local availability of BMPs (Umulis et al. 2009). Upon secretion, ligand binding to BMPs bind as dimers at the cell surface to het- proteins may induce a con- erotetrameric receptor complexes that include formational change that prevents ligand access two types of dual specificity kinase receptors to the BMP receptors (Wohl et al. 2016). Some (Table 1, Fig. 1): the BMP type I receptors prodomains (e.g., of BMP-2, -4, -7, and -10, and (i.e., -like kinase [ALK]-1, growth and differentiation factor [GDF-]5) ALK-2, ALK-3, and ALK-6) and the BMP form stable complexes with extracellular pro- type II receptors (i.e., BMPRII, ActRIIA, and teins (e.g., fibrillin-1 or collagen IV), thereby ActRIIB) (Yadin et al. 2016). These type I and restricting their diffusion in the extracellular type II receptors bind a variety of BMP ligands, space (Sengle et al. 2008; Wang et al. 2008; Ra- whereas the activin type II receptors also bind mirez and Rifkin 2009). BMP-4, BMP-6, BMP-9, activins (Upton and Morrell 2009). Regardless and BMP-10 are detected in the circulation, and of the receptor combination, the type II recep- can therefore affect tissues and organs at a dis- tor contains a constitutively active kinase do- tance, as well as the endothelial lining of the main, even in the absence of ligand. Binding vessels, through luminal activation of BMP re- of intracellular FK506-binding protein (FKBP) ceptors (David et al. 2008; Souza et al. 2008; 12 acts as a gatekeeper mechanism, setting a Herrera and Inman 2009; Bidart et al. 2012). threshold for type I receptor activation by the

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 3 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

Table 1. Bone morphogenetic protein (BMP) ligands and their receptors BMP Type II Type I R-Smad Coreceptor BMP-2 BMPRII ALK-3 Smadl, 5, 8 RGM BMP-4 ALK-6 ACTRIIA/B ALK-3 Smadl, 5, 8 ALK-6 BMP-6 BMPRII ALK-3 Smadl, 5, 8 RGM BMP-7 ALK-6 ACTRIIA/B ALK-2 Smadl, 5, 8 ALK-3 ALK-6 BMP-9 BMPRII ALK-1 Smadl, 5, 8 BMP-10 ALK-2 ACTRIIA/B ALK-1 Smadl, 5, 8 Smad2 BMPs bind to a combination of type I and type II receptors resulting in the carboxy-terminal phosphorylation and activation of Smad proteins and Smad-mediated modulation of transcription. Although there is redundancy, BMPs have a preference related to which receptor complex they bind and which coreceptor may modulate the signal. BMP,bone morphogenetic protein; ALK, activin receptor-like kinase, BMPRII, BMP type II receptor; ActRIIA/B, activin type II receptor A or B; RGM, repulsive guidance molecules.

type II receptor in the absence of ligand (Huse binding affinities of the other BMPs for their et al. 1999). type I receptors (Mahlawat et al. 2012). On the other hand, BMP-9 shows substantial dif- ferences with BMP-10 in type II receptor bind- BMP Receptors ing. BMP-9 binds most strongly to ActRIIB, and BMPs are classified into different subgroups shows weaker binding to BMPRII and weakest based on their sequence similarity and affinities binding to ActRIIA, although these data remain for specific receptors (Table 1) (Bragdon et al. to be confirmed in ECs (Townson et al. 2012; 2011; Katagiri and Watabe 2016). BMP-2 and Kienast et al. 2016). In contrast, BMP-10 does BMP-4 preferentially bind to ALK-3 and ALK-6 not show such a clear preference for the differ- (also known as BMPRIA and BMPRIB, respec- ent type II receptors (Townson et al. 2012). tively), and BMPRII or ActRIIA/B. BMP-6 and BMP-7 bind to ActRIIA and form a complex BMP Coreceptors with ALK-2, also known as ActRI. BMP-9 and BMP-10 bind with high affinity to ALK-1, also The binding of a BMP to its receptor can be known as TSR-I or SKR3, in combination with modulated by coreceptors, which lack an intrin- BMPRII, ActRIIA, or ActRIIB. Moreover, BMP- sic signaling motif (Table 1, Fig. 1). Two trans- 9 binds ALK-2 only in the presence of a type II membrane receptors, betaglycan, also called the receptor, whereas BMP-10 cannot bind to ALK- TGF-b type III receptor or TbRIII, and endo- 2 (Scharpfenecker et al. 2007; Olsen et al. 2014). glin play roles in the vasculature. Betaglycan Interestingly, ALK-2 can form a heterodimer increases the binding of BMP-2, -4, -7, and with other type I receptors, thereby increasing -14 to ALK-3 and ALK-6 (Kirkbride et al. the signaling complexity (Yadin et al. 2016). 2008), whereas endoglin has been shown to pro- Various BMPs show different affinities for their mote or repress signaling via distinct mecha- receptors. The binding affinities of BMP-9 and nisms that depend on the levels of endoglin BMP-10 for ALK-1 are in the picomolar range, and the concentrations of receptors and ligands which is exceptionally high (Townson et al. (ten Dijke et al. 2008). Endoglin interaction 2012; Kienast et al. 2016) compared with the with ALK-1 increases the responses to BMP-9

4 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

Extracellular regulators Noggin, MGP, BMPER

Type I Co-receptors Type II ALK-1 Endoglin ActRIIA ALK-2 Betaglycan ActRIIB BMP ALK-3 + LRP1 BAMBI BMPRII ALK-6 RGM

Smad1,5,8 No signal Smad6/7 Smurf1/2 PP1Aa MAPK Smad4 ErK p38 JNK

Co- factor

Figure 1. Bone morphogenetic protein (BMP) signaling pathway. Schematic representation of the BMP signaling pathway. BMPs interact with specific type I and type II receptors to form a heterotetrameric complex. Complex formation and ligand binding can be potentiated by a coreceptor—that is, endoglin, betaglycan, lipoprotein receptor-related protein 1 (LRP-1), or repulsive guidance molecule (RGM). After complex formation, the type II receptor phosphorylates the type I receptor, which then carboxy-terminally phosphorylates Smad1, Smad5, and Smad8 (canonical BMP signaling). Phosphorylated Smads propagate the signal via complex formation with Smad4, translocation into the nucleus, and regulation of the expression of target . Besides Smad-depen- dent signaling, BMPs can also transduce signals by mitogen-activated proteins kinases (MAPKs) (noncanonical BMP signaling). Canonical BMP signaling is intracellularly inhibited by inhibitory Smads—that is, Smad6 and/ or Smad7, E3 ubiquitin ligases, such as Smurf1 or Smurf2, or phosphatases (PPA1a). BMPs are extracellularly inhibited from binding to the receptor complex by secreted inhibitors, like noggin, matrix Gla protein (MGP), and the decoy receptor BAMBI. BMP signaling can be extracellularly stimulated or inhibited by BMPER.

and BMP-10 (Blanco et al. 2005; David et al. ALK-1 (Lebrin et al. 2004; Pece-Barbara et al. 2007; Scharpfenecker et al. 2007; Castonguay 2005). The interaction of endoglin with ALK-1 et al. 2011; Alt et al. 2012). The present model is also enhanced by fibronectin and integrin a5b1 is that endoglin and ALK-1 at the cell surface act leading to increases of BMP-9- and TGF-b-in- together to bind and capture BMP-9, and that duced phosphorylation of the BMP signaling ef- subsequent BMP-9 binding to the type II recep- fectors Smad1, Smad5, and Smad8 (Tian et al. tor displaces the bound endoglin to form a li- 2012). Endoglin can be shed from the cell surface gand-bound type I and type II receptor signal- through the actions of matrix metalloprotei- ing complex. The affinity of BMP-9 for endoglin nases, and the soluble endoglin ectodomain is in the nanomolar range (Castonguay et al. may thereby act as a released receptor trap for 2011; Kienast et al. 2016). Endoglin can also BMPs that can affect vascular remodeling (Ven- form a complex with the TGF-b type II recep- katesha et al. 2006; Hawinkels et al. 2010; Ra- tor, and either promote or inhibit TGF-b-in- thouska et al. 2015; Gallardo-Vara et al. 2016). duced Smad1 and Smad5 phosphorylation by Soluble endoglin was also shown to scavenge

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 5 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

BMP-9 and prevent BMP-9-induced Smad1, sponses to BMP-2 and BMP-4 in pulmonary Smad5 and Smad8 phosphorylation, whereas it arterial (PA) smooth muscle cells (SMCs). cannot bind TGF-b (Gregory et al. 2014). ALK-1, ALK-2, BMPRII, ActRIIA, and ActRIIB BMP signaling is potentiated by the gly- are also expressed in cultured murine VSMCs, cosylphosphatidylinositol-anchored repulsive and treatment with BMP-9 induces calcification guidance molecule (RGM) proteins (Corradini (Zhu et al. 2015). et al. 2009) and low-density lipoprotein recep- tor-related protein 1 (LRP-1) (Pi et al. 2012), BMP whereas the decoy receptor BMP and activin membrane-bound inhibitor (BAMBI) seques- On formation of a ligand–receptor complex, ters ligands from type I receptors and inhibits type II receptors activate type I receptors BMP signaling in ECs (Onichtchouk et al. 1999; through phosphorylation of serine and threo- Guillot et al. 2012). nine residues in their glycine-serine-rich (GS) domain. Activated type I receptors phosphory- late and thus activate the receptor-regulated Heterotetrameric BMP Receptor Complexes Smads (R-Smads), which then form hetero- Although BMP receptors are widely expressed, meric complexes with the common mediator not all cell types express all BMP receptors, and Smad, Smad4 (Fig. 1) (Hill 2016; Xu et al. the type I–type II receptor combinations for 2016). These complexestranslocate to the nucle- each BMP may therefore be cell-type specific. us where they regulate target gene transcription The available type I, type II, and type III recep- by binding to Smad-binding elements (SBEs) tors will determine which BMP will preferen- and interacting with other coactivators or core- tially signal and what the cellular response will pressors (Ross and Hill 2008). The specific type I be. ALK-2 and ALK-3 are widely expressed, receptor in the ligand–receptor complex deter- whereas ALK-1 is more selectively expressed on mines which R-Smads are activated. Most BMPs ECs, and ALK-6 is expressed by only a few cell induce phosphorylation of Smad1, Smad5, and types (Garcia de Vinuesa et al. 2016). BMPRII Smad8 (also named Smad9), although, in pul- and ActRIIA are ubiquitously expressed, where- monary ECs, BMP-9 can also activate Smad2, as ActRIIB expression is restricted to certain cell which is normallyassociated with TGF-b signal- types, including ECs. Among these type II re- ing (Upton et al. 2009). The underlying mecha- ceptors, only BMPRII is highlyexpressed in ECs. nism for this Smad2 response remains to be de- The high binding affinityof BMP-9 and BMP-10 termined. The Smad-dependent pathway is for ALK-1 and BMPRII, together with the high directly activated by the type I receptor, al- ALK-1 and BMPRII expression levels in ECs, though Smads may also be activated by alterna- may explain the major role of these two BMPs tive mechanisms that must be further explored. in angiogenesis. ALK-1 mayalso form acomplex Indeed, TGF-b-activated kinase 1 (TAK1), with ActRIIA or ActRIIB enabling BMP-9- or which is generally thought to be involved in BMP-10-induced activation (David et al. 2007; Smad-independent pathways, has been reported Scharpfenecker et al. 2007). BMP-2 and BMP-4 to phosphorylate Smad1 at the carboxyl termi- bind to ALK-3 or ALK-6; however, because nus (Shim et al. 2009). ALK-3 and ALK-6 are not highly expressed in On ligand binding, BMP receptors can also ECs, BMP-2 and BMP-4 are unlikely to induce activate non-Smad pathways that are important strong responses in these cells. BMP-6 and BMP- for correct vascular system establishment 7 bind to ActRIIA and ALK-2, thus inducing (Zhang 2017). Such non-Smad pathways in- activation of Smad1 and Smad5. VSMCs express clude signaling by mitogen-activated proteins BMPRII and ALK-3 at high levels, whereas ALK- kinases (MAPKs), such as p38, Erk, and JNK 1 and endoglin are not highly expressed in (Gallea et al. 2001; Guicheux et al. 2003), the VSMCs under physiological conditions (Upton phosphatidylinositol 3-kinase (PI3K)—Akt et al. 2008). BMPRII and ALK-3 mediate re- pathway and small Rho-like GTPases (Gamell

6 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

et al. 2008). Cross-talk between VEGF and degradation (Kavsak et al. 2000; Ebisawa et al. BMP-6 can enhance p38 MAPK activation and 2001; Murakami et al. 2003). Smad6 and Smad7 inhibit Akt activation, but does not impact the can also recruit protein phosphatases, such as carboxy-terminal phosphorylation of Smad1, protein phosphatase 1 a (PPA1a), to dephos- Smad5 and Smad8 or Erk MAPK activation phorylate ALK-1 (Valdimarsdottir et al. 2006). (Li et al. 2015a). BMP-9 inhibits JNK activation Interestingly, Smad6 and Smad7 were originally in ECs (Long et al. 2015). Endoglin and Ga- identified as vascular Smads because their ex- interacting protein carboxyl terminus interact- pression is induced in ECs by laminar shear ing protein (GIPC)-mediated trafficking of stress (Topper et al. 1997). The expression of PI3K regulates endothelial signaling and func- both inhibitory Smads is induced by BMPs, tion (Lee et al. 2008, 2012). The BMPRII car- thus creating a negative feedback loop (Ishisaki boxy-terminal extension also interacts with et al. 1999). Smad7 inhibits both BMPand TGF- effectors that can be involved in non-Smad sig- b signaling by competing with recruitment of naling (Hassel et al. 2004; Chan et al. 2007). phosphorylated R-Smads and by promoting BMP signaling pathways interact with other the degradation of the type I receptors, whereas growth factor pathways that are essential for vas- Smad6 more selectively inhibits BMP signaling cular development and homeostasis, including by recruiting Smurf1 to BMP type I receptors or the FGF, Notch, and WNT pathways, and possi- by competing with R-Smad for binding to bly also the Hippo pathway (Guo and Wang Smad4 (Hata et al. 1998; Murakami et al. 2003). 2009; Garcia de Vinuesa et al. 2016). BMPs, to- gether with other vascular signaling pathways, BMP SIGNALING MUTATIONS IN VASCULAR also interact with vascular endothelial (VE) cad- DISEASES herin, an essential component of endothelial ad- herent junctions, which plays a crucial role in Two human genetic vascular disorders have vascular permeability (Weis and Cheresh 2005; been linked to mutations in BMP signaling Lagendijk and Hogan 2015). It has been shown components showing that BMPs are key players that VE-cadherin can associate with ALK-2 and in vascular biology. Hereditary hemorraghic tel- BMPRII in a BMP-6-dependent manner (Benn angiectasia (HHT) or Osler–Weber–Rendu et al. 2016). BMP-6 induces c-Src phosphoryla- syndrome has been linked mostly to mutations tion leading to VE-cadherin internalization and in ENG, the gene encoding endoglin, and in an increase in vessel permeabilization. ACVRL1, which encodes the ALK-1 receptor. The second vascular disease caused by muta- tions in a BMP signaling component is heredi- BMP Signal Termination tary pulmonary arterial hypertension (hPAH). As the duration of BMP signaling will determine Mutations in BMPR2, and occasionally in its effects invascular development and disease, it ACVRL1 or ENG, have been linked to hPAH. has to be tightly controlled (Fig. 1). BMP signal- Fibrodysplasia ossificans progressiva (FOP) is ing can be terminated in many ways. Inhibitory a rare autosomal-dominant disorder that is Smads (I-Smads, Smad6, and Smad7) inhibit mainly characterized by episodic heterotopic BMP signaling through various mechanisms, ossification (HO) of muscle, fascia, ligaments, including interfering with the interactions and tendons. FOP is driven by activating muta- between R-Smads and type I receptors, down- tions in ACVR1 in the coding region of the ALK- regulation of cell surface type I receptors, pre- 2 intracellular domain. It is under debate wheth- vention of complex formation by R-Smads and er this disease categorizes as a vascular disease. co-Smads, and transcriptional regulation in the nucleus (Miyazawa and Miyazono 2016). For Hereditary Hemorrhagic Telangiectasia example, inhibitory Smads can recruit the Smad E3 ubiquitin ligases Smurf1 or Smurf2 The first report describing that disrupting BMP to the type I receptors, thus promoting receptor signaling can cause a vascular disorder was the

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 7 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

identification of mutations in ENG that led to tance (Simonneau et al. 2013). The increased the haploinsufficient vascular disease HHT1 pulmonary vascular resistance in PAH is attrib- (Fig. 2) (McAllister et al. 1994). Soon after, uted to small pulmonary artery constrictions mutations in ACVRL1, which encodes ALK-1, caused by profound vascular remodeling were identified in HHT2 (Johnson et al. 1996). (Guignabert et al. 2015). ECs, VSMCs, and fi- Several years later, SMAD4 mutations in com- broblasts show aberrant proliferation and resis- bined juvenile polyposis-HHT syndrome (JP- tance to apoptosis, resulting in reduced luminal HHT) were reported (Gallione et al. 2004). diameter, increased blood pressure and, ulti- Mutations in GDF2, which encodes BMP-9, mately, lethality from right ventricular failure have now also been described in a related form (Fig. 3). of HHT, HHT5 (Wooderchak-Donahue et al. Three forms of hypertension have been de- 2013; Hernandez et al. 2015). HHT is a rare fined: idiopathic PAH (iPAH), familial or heri- autosomal-dominant vascular disorder (inci- table PAH (hPAH), and environmental PAH. dence of 1/8000) (McDonald et al. 2015), Approximately 70% of hPAHpatients show mu- which is characterized by frequent epistaxis, tations in the BMPR2 gene, and 25% of spora- skin and mucosa telangiectasia, arteriovenous dic iPAHcases show de novo BMPR2 mutations. malformations (AVMs) in the lungs, liver or Mutations in BMPR2 have been identified in brain, and hemorrhages associated with these nearly all exons of the coding sequence—in- vascular lesions (Dupuis-Girod et al. 2010; cluding exons encoding parts of the ligand bind- Shovlin 2010). HHT1 patients more commonly ing domain, kinase domain, and cytoplasmic show pulmonary and cerebral AVMs, whereas tail. The vast majority of BMPR2 mutations HHT2 patients show higher incidences of he- are frame-shift mutations leading to a prema- patic AVMs and gastrointestinal telangiectasia ture stop codon and resulting in haploinsuffi- (Letteboer et al. 2006; Lesca et al. 2007). These ciency owing to nonsense-mediated decay of differences may be partly explained by the dif- mutant BMPRII mRNA transcripts (Fessel ferential distribution and expression levels of et al. 2011). The remaining mutations lead to BMP signaling components in these tissues, loss of function via a variety of mechanisms, suggesting overlapping but nonidentical func- including loss of kinase activity and impair- tions for endoglin and ALK-1. AVMs are ments of receptor folding and trafficking (Ru- thought to arise from enlargement of capillary darakanchana et al. 2002). A meta-analysis of vessels, creating a shunt between an artery and a data of 1550 patients with PAH revealed that vein, although the molecular mechanism is still PAH patients harboring a BMPR2 mutation not completely understood (Fig. 2) (Park et al. present at a younger age a more severe disease 2009). It has been proposed that idiopathic and carry an increased risk of death (Evans et al. AVM and Notch-induced AVMs are distinct 2016). There is no full penetrance of the disease from those of HHT (Peacock et al. 2016). onset, and women carrying a BMPR2 mutation show a greater incidence of PAH (Shapiro et al. 2012), likely caused by estrogen-induced inhibi- Pulmonary Arterial Hypertension tion of BMPRII signaling in VSMCs (Mair et al. The second vascular disease reported with mu- 2015). tations in BMP signaling is pulmonary arterial Although mutations in ACVRL1 usually hypertension (PAH), which is mainly due to cause HHT2, cases of severe PAH that are in- mutation in BMPR2, which encodes the distinguishable from iPAH have been docu- BMPRII receptor (Deng et al. 2000; Lane et al. mented in families segregating with HHT (Har- 2000). PAH is a rare vascular disorder with an rison et al. 2003; Girerd et al. 2010). Of note, incidence of 1/25,000, and is characterized by mutations in the genes encoding endoglin an elevated mean resting pulmonary arterial (ENG), Smad8 (SMAD9), and BMP-9 (GDF2) pressure of .25 mmHg, with normal left atrial have also been described in PAH (Machado pressure and high pulmonary vascular resis- et al. 2015; Wang et al. 2016a). As in HHT,

8 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from dacdOln ril.Ct hsatceas article this Cite Article. Online Advanced http://cshperspectives.cshlp.org/

A HHT Gene mutationB Artery Vein C Capillary bed Endoglin ENG (HHT1) BMP BMP9 GDF2 (HHT5) 9 ALK-1 ACVRL1 (HHT2) Healthy

P onOctober8,2021-PublishedbyColdSpringHarborLaboratoryPress odSrn abPrpc Biol Perspect Harb Spring Cold Type II Smad1,5,8 HHT receptor P Ateriovenous shunt

Smad4 SMAD4 (JP-HHT) P D E P

P o:10.1101 doi: P Id1/3 Disease and Homeostatis Vascular in BMPs Co- factor / cshperspect.a031989

Figure 2. Mutations in bone morphogenetic protein (BMP) signaling components result in hereditary hemorrhagic telangiectasia (HHT). (A) The genetic vascular disorder HHT, also known as the Osler–Weber–Rendu syndrome, is linked to mutations (shown in red) in the ENG gene, which encodes the coreceptor endoglin (HHT1), ACVRL1, which encodes the activin receptor-like kinase (ALK)-1 type I receptor (HHT2), GDF2, which encodes BMP-9 (HHT5), and SMAD4 ( juvenile polyposis [JP]-HHT). (B) Mutations in these genes result in disturbed endothelial cell (EC) growth and specification, the development of tortuous fragile vessels, and loss of arterial venous specification that lead to arteriovenous shunts (AVMs).(C,D) Typical features of the disease are pulmonary AVMs(C) and telangiectasia visible as red dots on lips and tongue (D). 9 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

ABPAH Gene mutation Smooth muscle cells Endothelial cells BMP BMP 9 9 ALK-1 ACVRL1 Genetic TGF-β factors hypoxia

P PAH Healthy Smad1,5,8 BMPRII BMPR2 P C

Endothelial proliferation/ plexiform core Smad4 P P Former Fibroblastic/ native artery myofibroblastic proliferation

P P Co- Id1/3 factor 200 μm Dilation lesion

Figure 3. Mutations in bone morphogenetic protein (BMP) signaling cascade results in pulmonary arterial hypertension (PAH). (A) The genetic vascular disorder PAH has been linked to mutations mainly in the BMPR2 gene for the BMPRII receptor, as well as the ACVRL1 gene, encoding the activin receptor-like kinase (ALK)-1 receptor (mutations indicated in red). Recently, a mutation in GDF2, encoding BMP-9, was described in a PAH patient. Rare cases of mutation in ENG or SMAD8 have also been described but are not shown here. (B) Mutations in these genes increase endothelial cell (EC) and vascular smooth muscle cell (VSMC) proliferation, resulting in a multilayered vessel that will lead to vessel obstruction. (C) Plexiform lesion distal to a pulmonary artery in the lung of a patient with idiopathic PAH, which is characteristic for plexogenic pulmonary arterio- pathy. The section is stained with hematoxylin and eosin.

BMP-9 and BMP-10 signaling via BMPRII and these issues and guide our design of improved ALK-1 likely plays a central role in PAH patho- targeted therapies for patients. Indeed, for most biology, implicating ECs as an important cell of the genes encoding BMP signaling mediators type in the initiation of both diseases (Morrell that are found to be mutated in vascular dis- et al. 2016). eases, genetic deletion results in strong and Despite advances in our understanding of often lethal vascular phenotypes in mice, fur- the pathogenesis of these genetic diseases in hu- ther supporting the view that this pathway mans, many important questions remain unan- plays a role in vascular development. Further swered. For example, it is unknown how loss-of- insights into the disease mechanisms have function mutations in BMPR2, ACVRL1,or been obtained using heterozygote animal mod- ENG can cause a disease that seems confined els of tissue-specific deletions, which mimic to pulmonary, liver, or brain vascular beds. We a human patient’s phenotype. These analyses also lack an explanation for the variable pene- have characterized different (non-)cell-autono- trance of these diseases—with 100% pene- mous players involved in disease development. trance for carriers of HHTmutations compared In the following section, we will discuss the with only 20% for carriers of PAH mutations. mouse models that have helped elucidate the Genetic studies in mice have clarified some of role of BMPs in the human pathologies.

10 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

LOSS-OF-FUNCTION ANIMAL MODELS IN vessels, a stalled vascular development at the BMP SIGNALING LINKED TO VASCULAR primary capillary plexus stage, and presence DISEASES of AVMs(Chen et al. 2013), which are reminis- cent of the vascular phenotype observed in Defects in Blood Vascular Remodeling Acvrl12/2 embryos (Oh et al. 2000; Urness In mice, genetic deletion of Eng, Acvrl1,or et al. 2000). The same research group generated Smad5 causes embryonic lethality because of a Bmp9 knockin mouse line, inwhich the Bmp10 cardiovascular system defects (Goumans and coding region was replaced with that of Bmp9. Mummery 2000), suggesting that these genes These mice showed normal vascular develop- function in a common pathway. Embryos defi- ment up to E16.5, supporting the view that cient for Eng (Li et al. 1999; Arthur et al. 2000; BMP-9 and BMP-10 are functionally inter- Bourdeau et al. 2000), Acvrl1 (Oh et al. 2000; changeable during vascular development. How- Urness et al. 2000), or Smad5 (Chang et al. ever, at E17.5, the heart was enlarged with pro- 1999; Yang et al. 1999) each show impaired nounced ventricular septal defects consistent yolk sac and embryonic vascular development, with previous reports showing that BMP-10 is resulting in embryonic lethality around embry- critical for heart development (Chen et al. onic day (E) 11.5. The phenotypes look remark- 2004). Interestingly, ALK-1 and BMP-10 are first ably similar, but there are subtle differences. En- expressed at E8.5 (Seki et al. 2003; Chen et al. doglin-deficient embryos do not have a layer of 2013), whereas BMP-9 expression begins 2 days VSMCs covering their major vessels at E9.5 (Li later, at E10.5 (Chen et al. 2013). Collectively, et al. 1999), and this VSMC developmental these data show that BMP-10 plays a crucial deformity precedes the vascular remodeling de- role in early vascular development. Moreover, fects. Eng2/2 ECs produce less TGF-b1, ham- at the onset of BMP-9 synthesis, both BMPs pering the recruitment of mesenchymal cells are interchangeably involved in blood vascular and their differentiation into pericytes and development, and in ensuring vascular quies- VSMCs, and thus preventing formation of the cence. In contrast, BMP-10 plays a specific role VSMC layer (Carvalho et al. 2004). In addition in heart development, likely because of the dif- to this vascular phenotype, Eng2/2 embryos ferent affinities of BMP-9 and BMP-10 for their also show an enlarged ventricle and outflow receptors (Townson et al. 2012). tract, abnormal cardiac looping, and truncal The BMP pathway is controlled by extracel- cushions with a disorganized endothelial lular BMP modulators, such as BMPER and layer—which are all signs of abnormal cardiac twisted gastrulation (TSG), which finely tune development (Arthur et al. 2000). Acvrl12/2 the BMP-induced proangiogenic events and embryos also show large vessel dilation and de- maintain vascular homeostasis (Heinke et al. layed VSMC differentiation and migration. Un- 2013). In the embryonic heart, BMPER expres- like Eng2/2 embryos, Acvrl12/2 embryos show sion is restricted to the ECs and the endotheli- excessive fusion of capillary networks in the em- um-derived cushions. Bmper2/2 mice die at bryo proper. Furthermore, they form AVMsbe- birth because of skeletal and kidney defects cause of fusion of major arteries and veins (Oh (Ikeya et al. 2006), and they have coronary heart et al. 2000; Urness et al. 2000; Park et al. 2008). anomalies because of impaired migration of Both BMP-9 and BMP-10 are physiological ventricular ECs (Dyer et al. 2015). Furthermore, ligands of ALK-1. This finding, coupled to the these hearts develop mitral valve prolapse (Wil- fact that Acvrl12/2 mice show a strong vascular lis et al. 2013) because of dysregulation of en- phenotype and Bmp92/2 mice show normal dothelial-to-mesenchymal transition (EndMT) blood vascular development, has led to a re- in the cardiac cushions (Dyer et al. 2015). Final- evaluation of Bmp102/2 mice that were de- ly, BMPER was shown to coregulate the BMP- scribed to have a cardiac phenotype (Chen mediated inflammation associated with lami- et al. 2004). Bmp10-deficiency leads to defects nar and oscillatory shear stress (Pi et al. 2012) in the embryo proper, including absent vitelline and may regulate the osteoblast-like differenti-

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 11 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

ation of human coronary artery VSMCs (Sa- The essential role of BMP signaling in the tomi-Kobayashi et al. 2012). Interestingly, lymphatic development of mice was first shown Mgp2/2 mice develop AVMs of the lung and by the disrupted lymphatic development ob- kidney, which are characteristic of vascular dys- served in the intestine and tail of newborn plasia HHT (Yao et al. 2013). mice following injection of the extracellular do- Another modulator of BMP signaling is main of ALK-1 fused to the Fc fragment of hu- TAK1, which is activated in response to both man IgG1 (ALK1-FC) or of a blocking anti- BMP (Shibuya et al. 1998) and TGF-b (Yama- ALK-1 (Niessen et al. 2010). Mice guchi et al. 1995) stimulation, as well as inter- with an induced genetic deletion of Acvrl1 leukin-1 (IL-1) and several other unrelated have enlarged lymphatic vessels within the intes- cytokines. Among other defects, Tak12/2 em- tine, cornea, and diaphragm, consistent with the bryos show dilated blood vessels associated with antiproliferative role of ALK-1 in postnatal lym- the loss of smooth muscle differentiation at phatic development (Yoshimatsu et al. 2013). mid-gestation (Jadrich et al. 2006). Endotheli- Bmp92/2 mice also show enlarged lymphatic al-specific Tak1 deletion reveals the importance capillaries and collecting lymphatic vessels in of TAK1 for embryonic EC survival and migra- mesentery, ear skin, and back skin, suggesting tion (Morioka et al. 2012). The phenotype of that BMP-9 could be a major ALK-1 ligand re- Tak1 deletion in mice is strikingly similar to that quired for lymphatic vascular development of Acvrl1 and Eng deletion, which is in line with (Levet et al. 2013; Yoshimatsu et al. 2013). Fur- the notion that TAK1 is an effector of this path- thermore, Bmp92/2 mice show reduced num- way. Consistently, TAK1 overexpression can res- bers of lymphatic valves and decreased drainage cue the vascular defect elicited by silencing efficiency (Levet et al. 2013), whereas adenovi- Acvrl1 expression using morpholino oligonu- rus-mediated BMP-9 overexpression diminish- cleotides in zebrafish (Jadrich et al. 2006). es inflammatory lymphangiogenesis in mice (Yoshimatsu et al. 2013). No lymphatic defects have been described so far in Bmp102/2 mice, Defects in Lymphatic Vascular Remodeling suggesting a specific role for BMP-9 signaling Embryonic lymphatic ECs (LECs) transdiffer- through ALK-1 in lymphaticdevelopment. Oth- entiate from embryonic venous ECs through the er studies report that BMP-2 signaling through cooperation of the SOX18 and COUP-TFII Alk-3, Alk-3b, BMPRIIa, BMPRIIb, and Smad5 transcription factors to promote PROX1 ex- can decrease the number of LECs in zebrafish pression (Francois et al. 2008; Yang et al. 2012; (Dunworth et al. 2014; Kim and Kim 2014). Aranguren et al. 2013; Srinivasan et al. 2014). However, zebrafish and mice may show different Migration of PROX1-expressing cells from em- context-dependent molecular and cellular roles bryonic veins and the subsequent podoplanin- for BMP signaling in vascular remodeling. mediated lymphaticovenous separation require VEGF-C (Zheng et al. 2014). Following estab- lishment of the primary lymphatic vasculature, GENETICALLY MODIFIED MOUSE MODELS FOR HUMAN VASCULAR DISEASES AND lymphatic vessels undergo further remodeling TREATMENTS to form a functional and hierarchical lymphatic vessel network comprising lymphatic capillar- Mice homozygous for null mutations in Eng or ies, precollectors, and collecting vessels. Fluid is Acvrl1 are embryonic lethal at mid-gestation taken up by lymphatic capillaries via discontin- because of cardiovascular defects. Although uous VE-cadherin-positive button junctions. these models are very helpful for determining The fluid passes into precollectors that contain the function of endoglin or ALK-1, they do not intraluminal valves to prevent retrograde flow, phenocopy the human disease. Therefore, het- ensuring unidirectional flow of lymph to the erozygous or inducible homozygous null mice, blood circulation (Coso et al. 2014; Vittet which have grown normally to adulthood before 2014; Yang and Oliver 2014). deletion, may be very useful models of BMP-

12 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

related genetic vascular diseases. These models istration of ALK1-FC as a ligand trap in wild- will also aid the design of new treatments. type mice (Larrivee et al. 2012), or anti-BMP-10 antibody in Bmp92/2 mice, or neutralizing anti-BMP9 and anti-BMP10 in wild-type mice Hereditary Hemorrhagic Telangiectasia further revealed that both BMP-9 and BMP-10 Mice that are heterozygous for mutations in are involved in retinal angiogenesis and can Acvrl1 or Eng develop vascular abnormalities modulate the Notch pathway (Ricard et al. that resemble the clinical pathology of HHT 2012; Chen et al. 2013; Baeyens et al. 2016; Ola (Bourdeau et al. 1999; Srinivasan et al. 2003; et al. 2016; Ruiz et al. 2016). It has been shown Torsney et al. 2003). These phenotypes are, like that Smad1 and Smad5 are required for modu- in patients, quite variable and mild, occur with a lation of the Notch pathway during angiogene- late onset, and depend on the genetic back- sis (Moya et al. 2012). ground (observed in 129/Ola background Together, these mouse models led to the only), suggesting that additional factors are re- identification of several key events in HHT de- quired for HHT development, such as genetic velopment: (1) ECs are the critical cell type in- modifiers or environmental triggers (Bourdeau volved in HHT; (2) loss of Acvrl1 or Eng is not et al. 1999; Torsneyet al. 2003). Angiogenesis has required in every EC of an AVM,because mosa- been proposed to act as an additional trigger or icism can account for the phenotype (Tual- secondary hit, because Engþ/2 and Acvrl1þ/2 Chalot et al. 2014); and (3) genetic modifiers mice injected with VEGF-expressing adenoviral may influence the susceptibility to HHT disease particles show increased vascular dysplasia (Xu (Bourdeau et al. 2001). Studies in these mouse et al. 2004; Hao et al. 2010). Homozygous dele- models also led to the proposal that AVM for- tion of Eng or Acvrl1 using cell type-specific or mation requires three events: (1) heterozygosity time-dependent Cre drivers results in consistent of Eng or Acvrl1 mutations; (2) loss of hetero- and robust AVMs in a pro-angiogenic and in- zygosity by somatic mutation, which has not yet flammatory environment (e.g., VEGF adminis- been described in patients, or alternatively by tration, normal angiogenesis in early postnatal protein shedding of endoglin, or potentially life, inflammation, wounding) (Park et al. 2009; also by ALK-1; and (3) an additional proangio- Walker et al. 2011; Choi et al. 2012, 2014). The genic or inflammatory trigger. use of -specific expression of Cre Heterozygous and homozygous Eng and recombinase clearly shows that ECs are the Acvrl1 mice are also used in experiments to prime cell type affected by the mutation, be- test potential new HHT treatments. Thalido- cause no AVMs are detected when the loss of mide treatment can restore the reduced VSMC Eng or Acvrl1 occurs in VSMCs, pericytes or coverage of vessels in Engþ/2 mice (Lebrin et al. (Choi et al. 2014; Garrido-Martin 2010). Anti-VEGF treatment of inducible et al. 2014). Acvrl12/2 mice has led to attenuation of brain The neonatal retina is initially avascular, and AVMs(Walker et al. 2012), and topical applica- the vascular plexus develops during the first tion of a VEGF-neutralizing antibody shortly week of life in a stereotypical fashion (Fruttiger after wounding can prevent AVM formation, 2007). The neonatal mouse retina is a widely block the progression of established AVMs, used model for studying angiogenesis because and even induce regression of early AVMs in it allows a detailed studyof the different stages of mice with induced endothelial Acvrl1 inactiva- vascular development. This retinal angiogenesis tion (Han et al. 2014). For an extensive dis- model was used to show the roles of ALK-1 and cussion on this topic we refer to the review by endoglin in retinal vascular differentiation Tual-Chalot et al. (2015). In agreement with the (Mahmoud et al. 2010; Tual-Chalot et al. results obtained in animal models, therapeutic 2014). Induced endothelium-specific Eng or approaches to block angiogenesis, such as anti- Acvrl1 inactivation result in delayed vascular re- VEGF , thalidomide, or PI3-kinase modeling of the retina capillary plexus. Admin- inhibition, have been developed and show ben-

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 13 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

eficial effects in HHT patients (Lebrin et al. combined with VEGF receptor inhibition using 2010; Dupuis-Girod et al. 2012; Ola et al. the SU-5416 kinase inhibitor). These models 2016). Taken together, these results support show pronounced pulmonary hypertension the hypothesis that HHT pathogenesis is caused with plexiform lesions and right ventricular hy- by an enhanced response to angiogenic cues pertrophy (Gomez-Arroyo et al. 2012; Nicolls when ALK-1 or endoglin expression are dimin- et al. 2012; Dickinson et al. 2013). Strain-spe- ished (Choi et al. 2013). cific differences in the SU-5416 rat model of PAH have been reported, supporting the notion that genetic modifiers play an important role in Pulmonary Arterial Hypertension the PAH phenotype (Jiang et al. 2016a). A Mice heterozygous for Bmpr2 do not develop Bmpr2 mutant rat model has been generated or show only minimal signs of PAH without that carries a heterozygous 140-bp deletion in additional stimuli (Beppu et al. 2004; Song the first Bmpr2 exon (Bmpr2D140Ex1/þ), as iden- et al. 2005; Long et al. 2006). Expression of a tified in a PAH patient (Ranchoux et al. 2015). dominant-negative form of BMPRII represses Although this rat does not spontaneously de- BMPRII signaling in smooth muscle cells and velop the hemodynamic features of pulmonary causes elevated right ventricle systolic pressure hypertension, by three months of age, it displays (RVSP) (West et al. 2004). Interestingly, intense pulmonary vascular remodeling and Bmpr22/2 deficiency in pulmonary ECs neomuscularization of intraparenchymal distal (pECs) elicits PAH in some mice (Hong et al. arterioles (Ranchoux et al. 2015). Further anal- 2008), providing in vivo evidence that absence yses will reveal whether this genetic rat model of BMPRII signaling in ECs is sufficient to cause will be a useful model for PAH. No existing a predisposition to PAH. Interference with BMP single model can recapitulate the many diverse signaling at the level of the Smads has shown forms of PAH; however, they do show endothe- that EC- or VSMC-specific Smad1 deletion lial dysfunction, an imbalance of proliferation yields mutant mice with elevated pulmonary and apoptosis, and a glycolytic metabolic pro- pressure, right ventricular hypertrophy, and file (Maarman et al. 2013; Tuder et al. 2013; thickened pulmonary arterioles (Han et al. Lawrie 2014; Rabinovitch et al. 2014). 2013). Smad82/2 mice show a defective pul- Experimental PAH animal models have monary vasculature (Huang et al. 2009). Addi- already been successfully used to test new thera- tionally, VSMC-specific homozygous replace- peutic approaches. To date, two approaches to ment of the wild-type Bmpr2 sequence with enhance BMP signaling in animal models are Bmpr2R899X, corresponding to the BMPR2R899X showing great promise; however, several ques- mutation in a PAH patient that results in a trun- tions remain unanswered and further work is cated BMPRII, induces an increase of RVSP in needed (Guignabert et al. 2016). In animals approximately one-third of the transgenic mice with experimentally induced pulmonary hyper- (West et al. 2008). Additionally, heterozygous tension, treatment with either FK506/tacrolimus Bmpr2þ/R899X mice developed RVSP by six (Spiekerkoetter et al. 2013) or BMP-9 (Long et al. months of age (Long et al. 2015). Signs of pul- 2015) reverse the established PAH. Tacrolimus monary hypertension have also been described treatment has also shown some benefits in end- in heterozygote Acvrl1þ/2 or Engþ/2 mice stage PAH patients (Spiekerkoetter et al. 2015). (Toporsian et al. 2010; Jerkic et al. 2011). Because mice rarely develop pronounced Fibrodysplasia Ossificans Progressiva pulmonary hypertension and their vessels are difficult to image and catheterize, rat models FOP is a rare autosomal-dominant disorder are often preferred for studies of PAH. Several that is mainly characterized by episodic hetero- rat models of PAH have been developed using topic ossification (HO) of muscle, fascia, liga- environmental induction (e.g., chronic hypox- ments, and tendons. FOP is driven by ACVR1 ia, monocrotaline injection, or chronic hypoxia mutations in the coding region of the ALK-2

14 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

intracellular domain (Fig. 4). The most com- chondrocyte- and osteoblast-like cells. When mon mutation, accounting for 97% of cases, this happens, the surrounding extracellular is a gain-of-function Arg206His (R206H) ami- matrix can become mineralized. When subject- no acid change in the ALK-2 intracellular juxta- ed to high shear stress, ECs disassemble their membrane GS subdomain (Shore et al. 2006). A primary cilium, which renders them more mouse model carrying the human R206H mu- sensitive to shear-induced EndMT (Egorova tation in the Acvr1 gene confirmed that this et al. 2011; Tkachenko et al. 2013) and BMP- mutation can lead to hyperactivation of Smad induced, Slug-dependent osteogenic differenti- signaling, thus identifying an underlying mo- ation (Sanchez-Duffhues et al. 2015). Increased lecular mechanism for the development of this BMP-2 and BMP-4 expression at atherosclerotic disease. Normally, ALK-2, together with the sites potentiates such calcification (Bostro¨m type II receptors, bind activin but the resulting et al. 1993; Dhore et al. 2001), whereas pharma- complex does not stimulate Smad1/5/8 phos- cological inhibitors of BMP signaling or the phorylation, thus activin acts as an inhibitor of BMP antagonist MGP can reduce vascular in- canonical BMP-mediated signaling through flammation and calcification (Cai et al. 2012). ALK-2. However, the R206H variant of ALK-2 Indeed, MGP overexpression attenuates vascu- responds to activin, inducing signaling via lar calcification in ApoE2/2 mice (Bostro¨m Smad1 and Smad5 (Hatsell et al. 2015). This et al. 2001; Yao et al. 2010), and treatment with has led to a new therapeutic approach using a the extracellular domain of ALK-3 (ALK3ECD) neutralizing activin A antibody that prevents or LDN-193189, a small BMPRI kinase inhibi- heterotopic ossification in Acvr1R206H mice tor, reduces vascular inflammation and calcifi- (Hatsell et al. 2015). Interestingly, ECs are in- cation in Ldlr2/2 mice (Derwall et al. 2012). corporated into the heterotopic cartilage and Furthermore, oxidized-LDL (oxLDL) and bone of FOP patients, which was not observed BMP-6 synergistically promote osteogenic dif- in the bone of healthy controls (Medici et al. ferentiation in ECs, providing a potential mech- 2010). Furthermore, the ACVR1 mutation and anism for the interactions between BMP signal- resulting activation of ALK-2 signaling in ECs ing, oxidative stress, and inflammation in the leads to EndMT, and the newly generated mes- induction of atherosclerosis-associated vascular enchymal-like cells can further differentiate into calcification (Yung et al. 2015). osteoblast-like cells (Fig. 4). This effect is medi- The vascular calcification process also re- ated by TGF-b2 and BMP-4, but not BMP-7, in quires an inflammatory response and a pheno- a manner dependent on ALK-2 and ALK-5. typic transformation of VSMCs into osteogenic These data support the idea that BMP-depen- cells under chronic inflammatory conditions dent vascular dysfunction could be important (New and Aikawa 2011). Activation of the in the development of FOP. BMP-2 pathway increases both valvular and vas- cular calcification (Nakagawa et al. 2010; Song et al. 2015). Tumor necrosis factor (TNF)-a can BMP-RELATED VASCULAR DISEASES induce vascular calcification both indirectly WITHOUT MUTATIONS IN THE BMP (Buendia et al. 2015) and directly. In a paracrine SIGNALING CASCADE manner, TNF-a induces BMP-2 expression in ECs and stimulates the release of BMP-2-con- Vascular Calcification taining endothelial microparticles (EMPs) (Li Vascular calcification is a tightly regulated pro- et al. 2010). Vascular injury may facilitate fusion cessthat results from the phenotypic plasticityof of the EMPs with VSMCs and favor osteogenic vascular cells. It has been observed in several transformation of VSMCs, thus promoting vas- pathologies, including atherosclerosis, chronic cular calcification (Buendia et al. 2015). Fur- kidney disease, diabetes, and hypertension. thermore, Smad62/2 mice develop vascular ECs can undergo EndMT to form myofibro- and valvular calcification, suggesting that blast-like cells or they can differentiate into Smad6 plays a protective role in this process

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 15 Downloaded from .J omn tal. et Goumans M.-J. 16 http://cshperspectives.cshlp.org/

ABFOP Gene mutation

Activin dacdOln ril.Ct hsatceas article this Cite Article. Online Advanced A ALK-2 ACVR1

Bone onOctober8,2021-PublishedbyColdSpringHarborLaboratoryPress

Smad1,5,8 BMPRII Healthy

Mesenchymal Smad4 progenitors odSrn abPrpc Biol Perspect Harb Spring Cold

End MT FOP

Co- Id1/3 factor Cartilage o:10.1101 doi:

Figure 4. Mutations in the bone morphogenetic protein (BMP) type I receptor activin receptor-like kinase (ALK)-2 results in fibrodysplasia ossificans progressiva (FOP). (A) The genetic vascular disorder FOP is linked to activating mutations in / cshperspect.a031989 ACVR1 encoding the receptor ALK-2 (indicated in red). (B) These mutations enhance activin and BMP signaling and heterotopic endochondral ossification. Cells of endothelial origin have been found in bone, which could result from endothelial-to-mesenchymal transition (EndMT). These mesenchymal precursors will then differentiate into chondro- cytes and osteoblasts. Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

(Galvin et al. 2000). TNF-a also reduces Smad6 (Kim et al. 2013). ApoE2/2;Ldlr2/2 mice expression, highlighting a third potential mech- that were fed a cholesterol diet showed signifi- anism for enhancing BMP-2 signaling levels in cantly higher levels of soluble endoglin com- vascular cells (Li et al. 2015b). pared with ApoE2/2 C57BL/6J mice and ApoE2/2;Ldlr2/2 mice that were fed a chow diet (Strasky et al. 2011). Furthermore, soluble Atherosclerosis endoglin interferes with rolling and adhesion of Atherosclerosis is a chronic inflammatory re- leukocytes to ECs in culture (Walsheet al. 2009). sponse in the arterial wall, which is character- Finally, BMP-7 combined with a partial left ca- ized by vascular plaque formation (Libby et al. rotid artery ligation inhibits plaque formation 2011). Stable plaques contain more collagen in vivo in ApoE2/2 mice (Singla et al. 2016). and VSMCs, whereas unstable plaques possess a large lipid core covered with a thin fibrous cap Cerebral Cavernous Malformation containing macrophages. An unstable plaque is more likely to rupture, which can cause myo- Cerebral cavernous malformation (CCM) is a cardial infarction or cerebrovascular accidents. vascular dysplasia that is characterized by en- Within atherosclerotic lesions, TGF-b signaling larged and irregular capillaries, mainly localized components are detectable in ECs, VSMCs, my- within the brain (Fischer et al. 2013). The af- ofibroblasts, dendritic cells, T cells, monocytes, fected capillaries have abnormally thin and and macrophages, and their expression is rap- fragile walls that make them prone to leakage, idly enhanced during vascular injury (Bobik resulting in cerebral hemorrhages. CCM occurs et al. 1999; Frostegard et al. 1999; Kalinina et in both sporadic and familial forms. Loss-of- al. 2004; Bot et al. 2009). Atherosclerosis, plaque function mutations in KRIT1 (also known as instability, and vascular calcification are also CCM1), which encodes the cerebral cavernous linked to altered BMP signaling, although no malformations 1 protein CCM1, CCM2,or mutations in BMP signaling components have CCM3 account for 90% of all cases of familial as yet been identified (Yao et al. 2010; Simoes CCM, resulting in weakened cell-to-cell junc- Sato et al. 2014; Grgurevic et al. 2016). tions and increased leakage from vessels (Stahl BMPs are expressed in atherosclerotic et al. 2008). EndMToccurs in ECs of the lesions plaques (Dhore et al. 2001) and colocalize in both the familial (Akers et al. 2009) and with the valvular fibrosa (i.e., the calcified sur- sporadic forms (Bravi et al. 2016) of CCM, face of the valve leaflets) (Mohler et al. 2001). and EndMT in Ccm12/2 ECs is mediated by Among patients with type 2 diabetes mellitus, KLF-4-dependent induction of BMP-6 expres- plasma BMP-2 levels correlate positively with sion (Maddaluno et al. 2013; Cuttano et al. plaque burden and calcification (Zhang et al. 2016). Inhibitors of the TGF-b or BMP path- 2015). Furthermore, increased BMP-2 levels ex- ways can partially reduce both the number and ert pro-inflammatory and pro-atherogenic ef- the size of vascular lesions in Ccm12/2 mice fects by stimulating oxidative stress and endo- (Maddaluno et al. 2013; Cuttano et al. 2016). thelial dysfunction as well as promoting plaque calcification by inducing an osteogenic pheno- Preeclampsia type in VSMCs (Li et al. 2008). Unexpectedly, BMPRII expression was found to be reduced in Preeclampsia (PE) is characterized by hyperten- advanced human coronary atherosclerotic le- sion and proteinuria and is associated with ma- sions, and Bmpr2 heterozygosity in ApoE32/2 ternal endothelial dysfunction. Soluble endo- mice leads to robust inflammation and athero- glin is increased in PE, and the level of soluble sclerosis (Kim et al. 2013). Bmpr2 deficiency endoglin has been proposed as a biomarker for increases monocyte adhesion owing to elevated PE that may reflect the extent of endothelial ICAM-1 and VCAM-1 levels, whereas silencing dysfunction (Levine et al. 2006; Venkatesha of Acvrl1 decreases endothelial inflammation et al. 2006; Rathouska et al. 2015). Injection of

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 17 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

soluble endoglin acts in concert with the soluble the disease. Therefore, a second hit is required Flt1 ectodomain to induce severe PE (Venkate- for disease onset and progression. Analyses of sha et al. 2006). Furthermore, a transgenic different animal models suggest that the second mouse model overexpressing human soluble hit that triggers disease onset and progression endoglin developed a PE-like phenotype, in- may include genetic modifiers, angiogenesis cluding increased arterial pressure (Valbuena- (discussed above), inflammation, metabolic Diez et al. 2012). When the levels of membra- stress, and/or hypoxia. Here, we discuss these nous endoglin are suppressed and replaced by putative additional triggers in the context of soluble endoglin, adhesion between vascular BMP signaling. ECs and mural cells is inhibited (Rossi et al. 2016), suggesting that endoglin plays a critical Genetic Modifiers role in mural cell adhesion and differentiation. As discussed above, the genetic background of an animal model determines the disease onset Arteriogenesis and Ischemic and severity of both HHT and PAH, highlight- Neovascularization ing the importance of (epi)genetic modifiers. Vasculogenesis and angiogenesis also play piv- Accordingly, the PTPN14 gene—encoding the otal roles invarious ischemic disorders in adults. protein tyrosine phosphatase, nonreceptor type Researchers are exploring several therapeutic 14 (PTPN14)—is identified as a modifier of approaches that use endothelial progenitor pulmonary AVMincidence in HHT (Benzinou cells (EPCs) to promote reendothelialization et al. 2012). Other weaker genetic associations of damaged vessels, and enhance neovasculari- have also been identified between ENG and zation following ischemic diseases, such as heart EMILIN2, which encodes the extracellular ma- and limb ischemia. BMP-9 administration en- trix protein elastin microfibril interfacer 2 hances blood flow recovery in vivo in a mouse (Pawlikowska et al. 2005; Letteboer et al. 2015). model of hindlimb ischemia (Kim et al. 2015). This is corroborated by laser Doppler perfusion BMPs and Inflammation in Vascular Disease imaging results showing a significant decrease in blood flow recovery in Engþ/2 and Acvrl1þ/2 Inflammation plays a fundamental role in the mice with hindlimb ischemia. Interestingly, initiation and progression of many vascular compared with controls, collateral artery size is disorders, including HHT and PAH. During significantly reduced in Engþ/2 mice, but not in inflammation, monocytes migrate to and infil- Acvrl1þ/2 mice (Seghers et al. 2012). This sug- trate the injured tissue, where they differentiate gests that endoglin contributes to both shear- into macrophages. Macrophages can be divided induced collateral artery growth and ischemia- in two main types, pro-inflammatory M1 induced angiogenesis, whereas ALK-1 may only macrophages that enhance the inflammatory be involved in ischemia-induced angiogenesis. response by secreting factors that stimulate dis- ease progression, such as TNF-a and monocyte chemoattractant protein (MCP)-1, and anti-in- SECOND HIT IN HHT AND PAH DISEASE flammatory M2 macrophages that promote PROGRESSION wound healing and tissue repair (Dutta and Accumulating evidence suggests that haploin- Nahrendorf 2014; Dingenouts et al. 2015). sufficiency of BMPRII, ENG,orACVRL1 are BMPs play roles in differentia- insufficient for HHTor PAH development. Fur- tion and modulate the inflammatory response. thermore, although all cells carry the same mu- BMP-2 and BMP-4, which can be produced by tations, the pathology is only seen at specific atherosclerotic VSMCs, promote BMPRII-de- locations and in specific vascular beds. Finally, pendent monocyte attraction leading to inflam- different patients with the same mutations, mation of the atherosclerotic lesion (Simoes show different timing of onset and severity of Sato et al. 2014). BMP-6 can activate M1 mac-

18 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

rophages, and stimulates IL-6 expression de- ment with elevated IL-6 levels in the lungs and pendent on ALK-2, Smad1, and p38 MAPK circulation (Soon et al. 2015). Inflammation has (Lee et al. 2010). On the other hand, BMP-7 also been proposed to be a trigger for HHT, be- induces macrophage differentiation toward an cause Engþ/2 mice with blepharitis have more M2 phenotype and stimulates the production of pronounced bleeding lesions (Torsney et al. anti-inflammatory cytokines, such as IL-10, 2003). Lipopolysaccharide was also shown to while reducing IL-6 expression in a Smad- and trigger the formation of skin AVMs(Han et al. PI3K-dependent manner (Rocher and Singla 2014). 2013; Rocher et al. 2012). BMP-7 treatment of ApoE2/2 mice inhibits monocyte infiltration BMPs and Metabolism and reduces the production of pro-inflamma- tory cytokines (Singla et al. 2016). Even though ECs are in close proximity to ox- Monocytes express low endoglin levels, ygenated blood, they primarily rely on glycolysis which are up-regulated during differentiation rather than oxidative metabolism for the pro- into macrophages (Lastres et al. 1992; O’Con- duction of ATP (Goveia et al. 2014). Under nell et al. 1992). Endoglin is a key determinant physiological conditions, ECs produce .80% for both monocyte migration and differentia- of their ATP by converting glucose into lactate, tion (van Laake et al. 2006). Indeed, Eng hetero- and ,1% of glucose-derived pyruvate enters zygosity hampers the ability of monocytes both the mitochondria for oxidative metabolism to respond to stromal cell-derived factor-1 through the tricarboxylic acid (TCA) cycle. (SDF1) and to migrate toward and invade dam- The switch from a quiescent to an angiogenic aged tissue (Post et al. 2010). Moreover, mono- phenotype is mediated by alterations of EC en- cytes with perturbed endoglin levels show im- ergy metabolism. Although such adaptations paired macrophage differentiation (van Laake were first described in tumor angiogenesis, et al. 2006; Aristorena et al. 2014; Dingenouts emerging evidence suggest that similar meta- et al. 2015; Ojeda-Fernandez et al. 2016). It is bolic abnormalities characterize PAH. Metabol- unknown which ligand causes this effect and ic features of ECs from PAH patients include how it influences the development of HHT. high aerobic glycolysis, low oxidative metabo- Ubiquitous expression of a cytoplasmically lism, upregulation of the pentose phosphate truncated BMPRII receptor (BMPRIIdelx4/þ), pathway, and increases in the nucleotide salvage which acts as dominant-negative inhibitor of and polyamine biosynthesis pathways (Fessel endogenous BMPRII, results in mild hyperten- et al. 2012). ECs isolated from mice with EC- sion but a substantial increase in pulmonary in- specific Bmpr2 inactivation show similarly in- flammation, in miceat 8 weeks of age, suggesting creased expression of phosphoglycerate kinase that BMPRII has anti-inflammatory effects (Ta- (PGK) 1 (Majka et al. 2011). The similarities lati et al. 2014). This effect appears to be unique between the metabolic adaptations in PAH to BMPRII, as silencing the expression of and the metabolic profile of angiogenic ECs ActRIIA and ActRIIB does not induce an in- could represent an opportunity for therapeuti- flammatory phenotype (Kim et al. 2013). Fur- cally targeting of EC metabolism in PAH by thermore, BMPRIIdelx4-expressing macrophag- pharmacological inhibition of the glycolytic es show an M1 phenotype, with increased enzyme 6-phosphofructo-2-kinase/fructose-2,6- transcription factor NF-kB activation and en- biphosphatase isoform 3 (PFKFB3), for exam- hanced IL-6 secretion (Talati et al. 2014). Con- ple, by using 3-(3-pyridinyl)-1-(4-pyridinyl)- ditioned medium of the activated macrophages 2-propen-1-one (3PO) (Clem et al. 2008). stimulates SMC migration, which is dependent on the BMP type I receptor. Finally, chronic li- BMP and Hypoxia popolysaccharide administration to Bmpr2þ/2 mice induces an exacerbated inflammatory Hypoxia causes oxidative stress by increasing response and pulmonary hypertension develop- mitochondrial superoxide production. Both

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 19 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

BMPRII and endoglin levels are affected by hy- crucial role in vascular remodeling. BMPs exert poxia indicating that oxidative stress and meta- potent effects on all blood and lymphatic cell bolic dysfunction play direct pathogenic roles in lineages, affecting the capacity of ECs to mi- HHTand PAH. Hypoxia increases endoglin ex- grate, proliferate, and form basic tubular struc- pression in ECs in cell culture and infarcted tures. BMPs also help determine the fates of the murine hearts in vivo (van Laake et al. 2006). surrounding pericytes, VSMCs, and adventitial In hypoxic ECs, increased endoglin expression cells that contribute to vessel structural integrity promotes expression of ALK-1 but not ALK-5 and function. BMP inhibition may be useful (Tian et al. 2010). Pulmonary endothelial ex- in the treatment of vascular disorders, such as pression of BMP-2 or BMP-4, but not BMP-5, CCM, vascular calcification and atherosclerosis, BMP-6 or BMP-7, is increased following expo- whereas BMP stimulation could be an effective sure to hypoxia (Frank et al. 2005). In mice treatment for HHT, PE, and PAH. Current heterozygous for Bmp4 (Bmp4LacZ/þ), loss of knowledge suggests that BMP-9 and BMP-10, hypoxia-induced BMP-4 expression is associat- which share 65% amino acid sequence identity, ed with decreased pulmonary vascular remod- are the most important BMP ligands in vascular eling and VSMC proliferation, and reduced development. This is supported by their bind- PH (Anderson et al. 2010). BMP-9 signaling ing with high affinity to BMPRII, ALK-1, and through BMPRII, ALK-1, and the transcription endoglin, which are expressed at relatively high factor Id1 protects ECs from hypoxia-induced levels by ECs and VSMCs, and their presence in apoptosis partly by inducing expression of the blood (David et al. 2008). BMP-4 and BMP-6 anti-apoptotic protein crystallin-aB (CRYAB) also exist in serum, supporting their potential (Ciumas et al. 2013). Hypoxia induces BMPRII role in vascular development (Herrera and In- down-regulation, and concomitantly reduces man 2009; Vukicevic and Grgurevic 2009), Smad1 and Smad5 phosphorylation and Id1 which is also supported by their involvement expression in pulmonary artery SMCs (Takaha- in vascular dysfunction like shear-mediated vas- shi et al. 2006; Maruyama et al. 2016). Hypoxia- cular remodeling and CCM. Furthermore, induced HIF-1a activates the expression of BMP-2, in particular in zebrafish (Wakayama miR-322, which in turn inhibits expression of et al. 2015), is gaining importance in vascular ALK3 and SMAD5 in pulmonary artery smooth biology, including also in flow-mediated vascu- muscle cells (PASMCs,) promoting hypoxia- lar calcification. induced PASMC proliferation and migration BMP-9 is produced by the liver (Miller et al. (Zeng et al. 2015). Moreover, hypoxia represses 2000; Bidart et al. 2012) and BMP-10 is pro- the expression of BMPER, resulting in en- duced in the heart, chiefly by the right atrium hanced BMP signaling and revascularization in adults (Neuhaus et al. 1999). The physiolog- of the hypoxic retina (Moreno-Miralles et al. ical levels of BMP-9 and BMP-10 are similar 2011). Together, these findings suggest that hy- (0.5–15 ng/ml) in both human and mouse se- poxia modulates BMP signaling and may, there- rum (David et al. 2007, 2008; Souza et al. 2008; fore, be an interesting target for HHTand PAH Chen et al. 2013; Kienast et al. 2016). BMP-9 is treatment. considered as the main bioactive form in adult circulation, and indeed most BMP-Smad re- porter activity is inhibited by a neutralizing BMP Signaling Goes Vascular anti-BMP-9 antibody (David et al. 2007; Ricard Unbalanced BMP signaling is involved in several et al. 2012; Chen et al. 2013), and plasma from human vascular diseases, caused directly by Bmp92/2 mice cannot activate this reporter mutations in BMP signaling components or in- (Ricard et al. 2012). directly by alteration of signaling levels. Togeth- In vivo data clearly indicate that BMP-9 and er with evidence from numerous genetic animal BMP-10 signaling play major roles in vascular models, these genetic correlations provide clear remodeling through BMPRII and ALK-1 (Ri- support for the view that BMP signaling plays a card et al. 2012; Chen et al. 2013). However,

20 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

the precise cellular mechanisms and molecular influencing the ratio between Smad-dependent responses induced by these ligands in ECs and and Smad-independent cascades (Hassel et al. VSMCs remain incompletely understood. One 2003). In addition, some mutant receptors possible explanation for contradictory findings may become activated by uncommon ligands, is that context-specific mechanisms may be at as shown for activin binding to the mutant play (David et al. 2009; Suzuki et al. 2010; Tillet ALK-2 in FOP (Hatsell et al. 2015). Improving and Bailly 2014). our understanding of how receptor complex lo- One major difference between HHT and calization and composition are disturbed in dis- PAH is that they involve mutations in different eased ECs and VSMCs in different vascular beds types of receptors. Although HHT is linked to may help explain why some tissues are more mutations in genes encoding the type I receptor, prone to develop vascular lesions than others. ACVRL1, or the coreceptor, ENG, PAH corre- The specific endothelial expression of ALK- lates with mutations of the type II receptor 1 and endoglin and their roles in vessel network BMPR2, although in few cases mutations in formation has led to the hypothesis that target- ACVRL1 have also been found. This raises the ing ALK-1 or endoglin could be a new thera- question as to whether the etiologies of these peutic approach to regulate tumor angiogenesis diseases can be explained by differences in li- and lymphangiogenesis. Thus, recent strategies gand binding or downstream Smad-dependent for cancer treatment have focused on simulta- or -independent signaling pathways. Another neous inhibition of VEGF and ALK-1 or endo- interesting point is that the type II receptor glin (Bhatt and Atkins 2014; Gupta et al. 2015). ActRIIA or ActRIIB cannot rescue BMPRII A neutralizing ALK-1 antibody (Necchi et al. deficiency. Also, unlike ALK-1 and endoglin, 2014), or the ALK1-Fc (Wang et al. 2016b) or which are primarily expressed in ECs, BMPRII endoglin-Fc ligand traps (Gordon et al. 2014), is highly expressed in both ECs and VSMCs and, combined with a VEGF inhibitor or chemother- thus, is likely to directly influence EC and apy have been reported to inhibit tumor VSMC function. growth. Although these trials are encouraging, It remains unclear as to how and why a dis- the highly context-dependent effects of ALK-1 ease that is caused by a mutation in a gene ex- and endoglin in controlling vascular function pressed widely throughout different vascular warrant further research to prevent unwanted beds manifests primarily in only a few organs side-effects of these approaches. (e.g., the lung, brain, gastrointestinal tract, and liver). Vascular bed-specific endothelial effects CONCLUSION of BMP-4 have been observed. BMP-4 exerts pro-oxidant, prohypertensive, and pro-inflam- Genetic vascular diseases have revealed major matory effects only in the systemic circulation, roles of BMPs in vascular development and whereas pulmonary arteries are protected from dysfunction. Unraveling their functions has these adverse effects of BMP-4 (Csiszar et al. enabled the development of new potential 2008). Moreover, differences in shear stress and therapeutics for several vascular-related pathol- responses in different vascular beds likely con- ogies such as PAH and FOP. In the future, the tribute in the selective development of defects. development of selective small molecules mod- The expression levels of various BMP receptors ifiers of BMP signaling will be an important and attenuators at the cell surface, as well as the challenge and should lead to new therapeutic ligand availability invarious vascular beds, likely opportunities. contribute to the greater sensitivity of some vas- cular beds to mutations and phenotypic out- ACKNOWLEDGMENTS comes. A change in receptor availability may alsoalter the compositionofpreformed receptor Weapologize to those whose work was not cited complexes, such that BMPs might bind to owing to lack of space. We thank David Baker different type I and type II receptors, thereby for valuable comments on our manuscript and

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 21 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

Drs. J.J. Mager, A. Hosman, and E. Nossent for eloid lineage and their role during aging and macrophage the images of HHTand PAH pathologies. Work polarization. J Cell Sci 127: 2723–2735. Arthur HM, Ure J, Smith AJ, Renforth G, Wilson DI, in the laboratory of S.B. is funded by the IN- Torsney E, Charlton R, Parums DV, Jowett T, Marchuk SERM, CEA, University of Grenoble Alpes DA, et al. 2000. Endoglin, an ancillary TGF-b receptor, is (UGA), Association pour la Recherche sur le required for extraembryonic angiogenesis and plays a key Cancer (ARC), the Ligue Contre le Cancer de role in heart development. Dev Biol 217: 42–53. Baeyens N, Larrivee B, Ola R, Hayward-Piatkowskyi B, Du- la Loire et de la Savoie, and the Association brac A, Huang B, Ross TD, Coon BG, Min E, Tsarfati M, Maladie de Rendu-Osler (AMRO). A.Z.’s work et al. 2016. Defective fluid shear stress mechanotransduc- is supported by FWO G0542N13 and C12/16/ tion mediates hereditary hemorrhagic telangiectasia. J Cell Biol 214: 807–816. 023. Work in the M.-J.G. laboratory is support- Benn A, Bredow C, Casanova I, Vukicevic S, Knaus P.2016. ed by the Netherlands CardioVascular Research VE-cadherin facilitates BMP-induced endothelial cell Initiative: an initiative with support of the permeability and signaling. J Cell Sci 129: 206–218. Dutch Heart Foundation, Dutch Federation of Benzinou M, Clermont FF,Letteboer TG, Kim JH, Espejel S, Harradine KA, Arbelaez J, Luu MT, Roy R, Quigley D, et University Medical Centers, the Netherlands al. 2012. Mouse and human strategies identify PTPN14 as Organisation for Health Research and Develop- a modifier of angiogenesis and hereditary haemorrhagic ment, and the Royal Netherlands Academy of telangiectasia. Nat Commun 3: 616. Sciences (CVON-PHAEDRA CVON2012-08 Beppu H, Ichinose F,Kawai N, Jones RC, Yu PB, Zapol WM, Miyazono K, Li E, Bloch KD. 2004. BMPR-II heterozy- and CVON-RECONNECT CVON 2014-11), gous mice have mild pulmonary hypertension and an and NHS-BAV.The P.t.D. laboratory is support- impaired pulmonary vascular remodeling response to ed by Cancer Genomics Centre Netherlands and prolonged hypoxia. Am J Physiol Lung Cell Mol Physiol the Netherlands CardioVascular Research Ini- 287: L1241–L1247. Bhatt RS, Atkins MB. 2014. Molecular pathways: Can acti- tiative: the Dutch Heart Foundation, Dutch vin-like kinase pathway inhibition enhance the limited Federation of University Medical Centers, the efficacy of VEGF inhibitors? Clin Cancer Res 20: 2838– Netherlands Organisation for Health Research 2845. and Development, and the Royal Netherlands Bidart M, Ricard N, Levet S, Samson M, Mallet C, David L, Subileau M, Tillet E, Feige JJ, Bailly S. 2012. BMP9 is Academy of Sciences CVON-PHAEDRA produced by hepatocytes and circulates mainly in an ac- CVON2012-08. tive mature form complexed to its prodomain. Cell Mol Life Sci 69: 313–324. Blanco FJ, Santibanez JF, Guerrero-Esteo M, Langa C, Vary CP, Bernabe´u C. 2005. Interaction and functional inter- REFERENCES play between endoglin and ALK-1, two components of Reference is also in this subject collection. the endothelial transforming growth factor-b receptor complex. J Cell Physiol 204: 574–584. Akers AL, Johnson E, Steinberg GK, Zabramski JM, Bobik A, Agrotis A, Kanellakis P, Dilley R, Krushinsky A, Marchuk DA. 2009. Biallelic somatic and germline mu- Smirnov V, Tararak E, Condron M, Kostolias G. 1999. tations in cerebral cavernous malformations (CCMs): Distinct patterns of transforming growth factor-b iso- Evidence for a two-hit mechanism of CCM pathogenesis. form and receptor expression in human atherosclerotic Hum Mol Genet 18: 919–930. lesions. Colocalization implicates TGF-b in fibrofatty le- Alt A, Miguel-Romero L, Donderis J, Aristorena M, Blanco sion development. Circulation 99: 2883–2891. FJ, Round A, Rubio V, Bernabe´u C, Marina A. 2012. Bostro¨m K, Watson KE, Horn S, Wortham C, Herman IM, Structural and functional insights into endoglin ligand Demer LL. 1993. Bone morphogenetic protein expres- recognition and binding. PLoS ONE 7: e29948. sion in human atherosclerotic lesions. J Clin Invest 91: Anderson L, Lowery JW, Frank DB, Novitskaya T, Jones M, 1800–1809. Mortlock DP, Chandler RL, de Caestecker MP. 2010. Bostro¨m K, Tsao D, Shen S, WangY,Demer LL. 2001. Matrix Bmp2 and Bmp4 exert opposing effects in hypoxic pul- GLA protein modulates differentiation induced by bone monary hypertension. Am J Physiol Regul Integr Comp morphogenetic protein-2 in C3H10T1/2 cells. J Biol Physiol 298: R833–842. Chem 276: 14044–14052. Aranguren XL, Beerens M, Coppiello G, Wiese C, Vanders- Bot PT, Hoefer IE, Sluijter JP,van Vliet P,Smits AM, Lebrin missen I, Lo Nigro A, VerfaillieCM, Gessler M, Luttun A. F, Moll F, de Vries JP, Doevendans P, Piek JJ, et al. 2009. 2013. COUP-TFII orchestrates venous and lymphatic en- Increased expression of the transforming growth factor-b dothelial identity by homo- or hetero-dimerisation with signaling pathway, endoglin, and early growth response-1 PROX1. J Cell Sci 126: 1164–1175. in stable plaques. Stroke 40: 439–447. Aristorena M, Blanco FJ, de Las Casas-Engel M, Ojeda-Fer- Bourdeau A, Dumont DJ, Letarte M. 1999. A murine model nandez L, Gallardo-Vara E, Corbı´ A, Botella LM, Berna- of hereditary hemorrhagic telangiectasia. J Clin Invest be´u C. 2014. Expression of endoglin isoforms in the my- 104: 1343–1351.

22 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

Bourdeau A, Faughnan ME, Letarte M. 2000. Endoglin-de- of Eng with VEGF stimulation is sufficient to cause cere- ficient mice, a unique model to study hereditary hemor- brovascular dysplasia in the adult mouse. Cerebrovasc Dis rhagic telangiectasia. Trends Cardiovasc Med 10: 279– 33: 540–547. 285. Choi EJ, Kim YH, Choe SW, Tak YG, Garrido-Martin EM, Bourdeau A, Faughnan ME, McDonald ML, Paterson AD, Chang M, Lee YJ, Oh SP. 2013. Enhanced responses to Wanless IR, Letarte M. 2001. Potential role of modifier angiogenic cues underlie the pathogenesis of hereditary genes influencing transforming growth factor-b1 levels hemorrhagic telangiectasia 2. PLoS ONE 8: e63138. in the development of vascular defects in endoglin het- Choi EJ, Chen W,Jun K, Arthur HM, YoungWL, Su H. 2014. erozygous mice with hereditary hemorrhagic telangiec- Novel brain arteriovenous malformation mouse models tasia. Am J Pathol 158: 2011–2020. for type 1 hereditary hemorrhagic telangiectasia. PLoS Bragdon B, Moseychuk O, Saldanha S, King D, Julian J, ONE 9: e88511. Nohe A. 2011. Bone morphogenetic proteins: A critical Ciumas M, Eyries M, Poirier O, Maugenre S, Dierick F, review. Cell Signal 23: 609–620. Gambaryan N, Montagne K, Nadaud S, Soubrier F. Bravi L, Malinverno M, Pisati F,Rudini N, Cuttano R, Pallini 2013. Bone morphogenetic proteins protect pulmonary R, Martini M, Larocca LM, Locatelli M, Levi V,et al. 2016. microvascular endothelial cells from apoptosis by up- Endothelial cells lining sporadic cerebral cavernous mal- regulating a-B-crystallin. Arterioscler Thromb Vasc Biol formation cavernomas undergo endothelial-to-mesen- 33: 2577–2584. chymal transition. Stroke 47: 886–890. Clem B, Telang S, Clem A, Yalcin A, Meier J, Simmons A, Buendia P,Montes de Oca A, Madueno JA, Merino A, Mar- Rasku MA, Arumugam S, Dean WL, Eaton J, et al. 2008. tin-Malo A, Aljama P, Ramirez R, Rodriguez M, Carra- Small-molecule inhibition of 6-phosphofructo-2-kinase cedo J. 2015. Endothelial microparticles mediate inflam- activity suppresses glycolytic flux and tumor growth. Mol mation-induced vascular calcification. FASEB J 29: 173– Cancer Ther 7: 110–120. 181. Corradini E, Babitt JL, Lin HY. 2009. The RGM/DRAGON Cai J, Pardali E, Sanchez-Duffhues G, ten Dijke P.2012. BMP family of BMP co-receptors. Cytokine Growth Factor Rev signaling in vascular diseases. FEBS Lett 586: 1993–2002. 20: 389–398. Carmeliet P, Jain RK. 2011. Molecular mechanisms and Coso S, Bovay E, Petrova TV. 2014. Pressing the right but- clinical applications of angiogenesis. Nature 473: 298– tons: Signaling in lymphangiogenesis. Blood 123: 2614– 307. 2624. Carvalho RL, Jonker L, Goumans MJ, Larsson J, Bouwman Csiszar A, Labinskyy N, Jo H, Ballabh P, Ungvari Z. 2008. P, Karlsson S, ten Dijke P, Arthur HM, Mummery CL. Differential proinflammatory and prooxidant effects of 2004. Defective paracrine signalling by TGFb in yolk sac bone morphogenetic protein-4 in coronary and pulmo- vasculature of endoglin mutant mice: A paradigm for nary arterial endothelial cells. Am J Physiol Heart Circ hereditary haemorrhagic telangiectasia. Development Physiol 295: H569–H577. 131: 6237–6247. Cuttano R, Rudini N, Bravi L, Corada M, Giampietro C, Castonguay R, Werner ED, Matthews RG, Presman E, Mu- Papa E, Morini MF, Maddaluno L, Baeyens N, Adams livor AW,Solban N, Sako D, Pearsall RS, Underwood KW, RH, et al. 2016. KLF4 is a key determinant in the devel- Seehra J, et al. 2011. Soluble endoglin specifically binds opment and progression of cerebral cavernous malfor- bone morphogenetic proteins 9 and 10 via its orphan mations. EMBO Mol Med 8: 6–24. domain, inhibits blood vessel formation, and suppresses David L, Mallet C, Mazerbourg S, Feige JJ, Bailly S. 2007. tumor growth. J Biol Chem 286: 30034–30046. Identification of BMP9 and BMP10 as functional activa- Chan MC, Nguyen PH, Davis BN, Ohoka N, Hayashi H, Du tors of the orphan activin receptor-like kinase 1 (ALK1) K, Lagna G, Hata A. 2007. A novel regulatory mechanism in endothelial cells. Blood 109: 1953–1961. of the bone morphogenetic protein (BMP) signaling David L, Mallet C, Keramidas M, Lamande N, Gasc JM, pathway involving the carboxyl-terminal tail domain of Dupuis-Girod S, Plauchu H, Feige JJ, Bailly S. 2008. BMP type II receptor. Mol Cell Biol 27: 5776–5789. Bone morphogenetic protein-9 is a circulating vascular Chang C. 2016. Agonists and antagonists of TGF-b family quiescence factor. Circ Res 102: 914–922. ligands. Cold Spring Harb Perspect Biol 8: a021923. David L, Feige JJ, Bailly S. 2009. Emerging role of bone Chang H, Huylebroeck D, Verschueren K, Guo Q, Matzuk morphogenetic proteins in angiogenesis. Cytokine MM, Zwijsen A. 1999. Smad5 knockout mice die at mid- Growth Factor Rev 20: 203–212. gestation due to multiple embryonic and extraembryonic Deng Z, Morse JH, Slager SL, Cuervo N, Moore KJ, Venetos defects. Development 126: 1631–1642. G, Kalachikov S, Cayanis E, Fischer SG, Barst RJ, et al. Chen H, Shi S, Acosta L, Li W, Lu J, Bao S, Chen Z, Yang Z, 2000. Familial primary pulmonary hypertension (gene Schneider MD, Chien KR, et al. 2004. BMP10 is essential PPH1) is caused by mutations in the bone morphogenet- for maintaining cardiac growth during murine cardio- ic protein receptor-II gene. Am J Hum Genet 67: 737– genesis. Development 131: 2219–2231. 744. Chen H, Brady Ridgway J, Sai T, Lai J, Warming S, Chen H, Derwall M, Malhotra R, Lai CS, Beppu Y, Aikawa E, Seehra Roose-Girma M, Zhang G, Shou W, Yan M. 2013. Con- JS, Zapol WM, Bloch KD, Yu PB. 2012. Inhibition of text-dependent signaling defines roles of BMP9 and bone morphogenetic protein signaling reduces vascular BMP10 in embryonic and postnatal development. Proc calcification and atherosclerosis. Arterioscler Thromb Vasc Natl Acad Sci 110: 11887–11892. Biol 32: 613–622. Choi EJ, Walker EJ, Shen F, Oh SP, Arthur HM, Young WL, De Smet F, Segura I, De Bock K, Hohensinner PJ, Carmeliet Su H. 2012. Minimal homozygous endothelial deletion P. 2009. Mechanisms of vessel branching: Filopodia on

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 23 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

endothelial tip cells lead the way. Arterioscler Thromb Vasc Fischer A, Zalvide J, Faurobert E, Albiges-Rizo C, Tournier- Biol 29: 639–649. Lasserve E. 2013. Cerebral cavernous malformations: Dhore CR, Cleutjens JP, Lutgens E, Cleutjens KB, Geusens From CCM genes to endothelial cell homeostasis. Trends PP,Kitslaar PJ, Tordoir JH, Spronk HM, VermeerC, Dae- Mol Med 19: 302–308. men MJ. 2001. Differential expression of bone matrix Francois M, Caprini A, Hosking B, Orsenigo F, Wilhelm D, regulatory proteins in human atherosclerotic plaques. Browne C, Paavonen K, Karnezis T,Shayan R, Downes M, Arterioscler Thromb Vasc Biol 21: 1998–2003. et al. 2008. Sox18 induces development of the lymphatic Dickinson MG, Bartelds B, Borgdorff MA, Berger RM. 2013. vasculature in mice. Nature 456: 643–647. The role of disturbed blood flow in the development of Frank DB, Abtahi A, Yamaguchi DJ, Manning S, Shyr Y, pulmonary arterial hypertension: Lessons from preclini- Pozzi A, Baldwin HS, Johnson JE, de Caestecker MP. cal animal models. Am J Physiol Lung Cell Mol Physiol 2005. Bone morphogenetic protein 4 promotes pulmo- 305: –L14. nary vascular remodeling in hypoxic pulmonary hyper- tension. Circ Res 97: 496–504. Dingenouts CK, Goumans MJ, Bakker W. 2015. Mononu- clear cells and vascular repair in HHT. Front Genet 6: 114. Frostegard J, Ulfgren AK, Nyberg P,Hedin U, Swedenborg J, Andersson U, Hansson GK. 1999. Cytokine expression in Dunworth WP, Cardona-Costa J, Bozkulak EC, Kim JD, advanced human atherosclerotic plaques: of Meadows S, Fischer JC, Wang Y, Cleaver O, Qyang Y, pro-inflammatory (Th1) and macrophage-stimulating Ober EA, et al. 2014. Bone morphogenetic protein 2 sig- cytokines. Atherosclerosis 145: 33–43. naling negatively modulates lymphatic development in Fruttiger M. 2007. Development of the retinal vasculature. vertebrate embryos. Circ Res 114: 56–66. Angiogenesis 10: 77–88. Dupuis-Girod S, Bailly S, Plauchu H. 2010. Hereditary hem- Gallardo-Vara E, Blanco FJ, Roque M, Friedman SL, Suzuki orrhagic telangiectasia (HHT): From molecular biology T, Botella LM, Bernabe´u C. 2016. Transcription factor to patient care. J Thromb Haemost 8: 1447–1456. KLF6 upregulates expression of metalloprotease Dupuis-Girod S, Ginon I, Saurin JC, Marion D, Guillot E, MMP14 and subsequent release of soluble endoglin dur- Decullier E, Roux A, Carette MF, Gilbert-Dussardier B, ing vascular injury. Angiogenesis 19: 155–171. Hatron PY, et al. 2012. Bevacizumab in patients with Gallea S, Lallemand F, Atfi A, Rawadi G, Ramez V,Spinella- hereditary hemorrhagic telangiectasia and severe hepatic Jaegle S, Kawai S, Faucheu C, Huet L, Baron R, et al. 2001. vascular malformations and high cardiac output. JAMA Activation of mitogen-activated protein kinase cascades 307: 948–955. is involved in regulation of bone morphogenetic protein- Dutta P,Nahrendorf M. 2014. Regulation and consequences 2-induced osteoblast differentiation in pluripotent of monocytosis. Immunol Rev 262: 167–178. C2C12 cells. Bone 28: 491–498. Dyer L, Lockyer P, Wu Y, Saha A, Cyr C, Moser M, Pi X, Gallione CJ, Repetto GM, Legius E, Rustgi AK, Schelley SL, Patterson C. 2015. BMPER promotes epithelial-mesen- Tejpar S, Mitchell G, Drouin E, Westermann CJ, Marchuk chymal transition in the developing cardiac cushions. DA. 2004. A combined syndrome of juvenile polyposis PLoS ONE 10: e0139209. and hereditary haemorrhagic telangiectasia associated with mutations in MADH4 (SMAD4). Lancet 363: Ebisawa T, Fukuchi M, Murakami G, Chiba T, Tanaka K, 852–859. Imamura T, Miyazono K. 2001. Smurf1 interacts with transforming growth factor-b type I receptor through Galvin KM, Donovan MJ, Lynch CA, Meyer RI, Paul RJ, Smad7 and induces receptor degradation. J Biol Chem Lorenz JN, Fairchild-Huntress V, Dixon KL, Dunmore 276: 12477–12480. JH, Gimbrone MAJr, et al. 2000. A role for Smad6 in development and homeostasis of the cardiovascular sys- Egorova AD, Khedoe PP,Goumans MJ, YoderBK, Nauli SM, tem. Nat Genet 24: 171–174. ten Dijke P, Poelmann RE, Hierck BP. 2011. Lack of pri- Gamell C, Osses N, Bartrons R, Ruckle T,Camps M, Rosa JL, mary cilia primes shear-induced endothelial-to-mesen- Ventura F. 2008. BMP2 induction of actin cytoskeleton chymal transition. Circ Res 108: 1093–1101. reorganization and cell migration requires PI3-kinase Eilken HM, Adams RH. 2010. Dynamics of endothelial cell and Cdc42 activity. J Cell Sci 121: 3960–3970. behavior in sprouting angiogenesis. Curr Opin Cell Biol Garcia de Vinuesa A, Abdelilah-Seyfried S, Knaus P,Zwijsen 22: 617–625. A, Bailly S. 2016. BMP signaling in vascular biology and Evans JD, Girerd B, Montani D, Wang XJ, Galie N, Austin dysfunction. Cytokine Growth Factor Rev 27: 65–79. ED, Elliott G, Asano K, Grunig E, Yan Y, et al. 2016. Garrido-Martin EM, Nguyen HL, Cunningham TA, Choe BMPR2 mutations and survival in pulmonary arterial SW, Jiang Z, Arthur HM, Lee YJ, Oh SP. 2014. Common hypertension: An individual participant data meta-anal- and distinctive pathogenetic features of arteriovenous ysis. Lancet Respir Med 4: 129–137. malformations in hereditary hemorrhagic telangiectasia Fessel JP,Loyd JE, Austin ED. 2011. The genetics of pulmo- 1 and hereditary hemorrhagic telangiectasia 2 animal nary arterial hypertension in the post-BMPR2 era. Pulm models—Brief report. Arterioscler Thromb Vasc Biol 34: Circ 1: 305–319. 2232–2236. Fessel JP, Hamid R, Wittmann BM, Robinson LJ, Blackwell Gebala V,Collins R, Geudens I, Phng LK, Gerhardt H. 2016. T, Tada Y, Tanabe N, Tatsumi K, Hemnes AR, West JD. Blood flow drives lumen formation by inverse membrane 2012. Metabolomic analysis of bone morphogenetic pro- blebbing during angiogenesis in vivo. Nat Cell Biol 18: tein receptor type 2 mutations in human pulmonary 443–450. endothelium reveals widespread metabolic reprogram- Girerd B, Montani D, Coulet F, Sztrymf B, Yaici A, Jais X, ming. Pulm Circ 2: 201–213. Tregouet D, Reis A, Drouin-Garraud V, Fraisse A, et al.

24 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

2010. Clinical outcomes of pulmonary arterial hyperten- an animal model for hereditary hemorrhagic telangiec- sion in patients carrying an ACVRL1 (ALK1) mutation. tasia 2. Angiogenesis 17: 823–830. Am J Respir Crit Care Med 181: 851–861. Hao Q, Zhu Y, Su H, Shen F, Yang GY, Kim H, Young WL. Gomez-Arroyo JG, Farkas L, Alhussaini AA, Farkas D, Kras- 2010. VEGF induces more severe cerebrovascular dyspla- kauskas D, Voelkel NF, Bogaard HJ. 2012. The monocro- sia in Engþ/2 than in Alk1þ/2 mice. Transl Stroke Res 1: taline model of pulmonary hypertension in perspective. 197–201. Am J Physiol Lung Cell Mol Physiol 302: L363–L369. Harrison RE, Flanagan JA, Sankelo M, Abdalla SA, Rowell J, Gordon MS, Robert F, Matei D, Mendelson DS, Goldman Machado RD, Elliott CG, Robbins IM, Olschewski H, JW, Chiorean EG, Strother RM, Seon BK, Figg WD, Peer McLaughlin V,et al. 2003. Molecular and functional anal- CJ, et al. 2014. An open-label phase Ib dose-escalation ysis identifies ALK-1 as the predominant cause of pul- study of TRC105 (anti-endoglin antibody) with bevaci- monary hypertension related to hereditary haemorrhagic zumab in patients with advanced cancer. Clin Cancer Res telangiectasia. J Med Genet 40: 865–871. 20: 5918–5926. Hassel S, Schmitt S, Hartung A, Roth M, Nohe A, Petersen Goumans MJ, Mummery C. 2000. Functional analysis of the N, Ehrlich M, Henis YI, Sebald W, Knaus P. 2003. Initi- TGF-b receptor/Smad pathway through gene ablation in ation of Smad-dependent and Smad-independent signal- mice. Int J Dev Biol 44: 253–265. ing via distinct BMP-receptor complexes. J Bone Joint Goumans MJ, ten Dijke P.2017. TGF-b signaling in control Surg Am 85-A: 44–51. of cardiovascular function. Cold Spring Harb Perspect Biol Hassel S, Eichner A, Yakymovych M, Hellman U, Knaus P, doi: 10.1101/cshperspect.a022210. Souchelnytskyi S. 2004. Proteins associated with type II Goveia J, Stapor P,Carmeliet P.2014. Principles of targeting bone morphogenetic protein receptor (BMPR-II) and endothelial cell metabolism to treat angiogenesis and en- identified by two-dimensional gel electrophoresis and dothelial cell dysfunction in disease. EMBO Mol Med 6: mass spectrometry. Proteomics 4: 1346–1358. 1105–1120. Hata A, Lagna G, Massague´ J, Hemmati-Brivanlou A. 1998. Gregory AL, Xu G, Sotov V, Letarte M. 2014. Review: the Smad6 inhibits BMP/Smad1 signaling by specifically enigmatic role of endoglin in the placenta. Placenta 35: competing with the Smad4 tumor suppressor. Genes S93–S99. Dev 12: 186–197. Grgurevic L, Christensen GL, Schulz TJ, Vukicevic S. 2016. Hatsell SJ, Idone V,Wolken DM, Huang L, Kim HJ, Wang L, Bone morphogenetic proteins in inflammation, glucose Wen X, Nannuru KC, Jimenez J, Xie L, et al. 2015. homeostasis and adipose tissue energy metabolism. Cy- ACVR1R206H receptor mutation causes fibrodysplasia os- tokine Growth Factor Rev 27: 105–118. sificans progressiva by imparting responsiveness to acti- Guicheux J, Lemonnier J, Ghayor C, Suzuki A, Palmer G, vin A. Sci Transl Med 7: 303ra137. Caverzasio J. 2003. Activation of p38 mitogen-activated Hawinkels LJ, Kuiper P,Wiercinska E, Verspaget HW,Liu Z, protein kinase and c-Jun-NH2-terminal kinase by BMP- Pardali E, Sier CF, ten Dijke P. 2010. Matrix metallopro- 2 and their implication in the stimulation of osteoblastic cell differentiation. J Bone Miner Res 18: 2060–2068. teinase-14 (MT1-MMP)-mediated endoglin shedding inhibits tumor angiogenesis. Cancer Res 70: 4141–4150. Guignabert C, TuL, Girerd B, Ricard N, Huertas A, Montani D, Humbert M. 2015. New molecular targets of pulmo- Heinke J, Juschkat M, Charlet A, Mnich L, Helbing T, Bode nary vascular remodeling in pulmonary arterial hyper- C, Patterson C, Moser M. 2013. Antagonism and synergy tension: Importance of endothelial communication. between extracellular BMP modulators Tsg and BMPER Chest 147: 529–537. balance blood vessel formation. J Cell Sci 126: 3082– 3094. Guignabert C, Bailly S, Humbert M. 2016. Restoring BMPRII functions in pulmonary arterial hypertension: Hernandez F,Huether R, Carter L, Johnston T,Thompson J, Opportunities, challenges and limitations. Expert Opin Gossage JR, Chao E, Elliott AM. 2015. Mutations in Ther Targets 21: 181–190. RASA1 and GDF2 identified in patients with clinical fea- Guillot N, Kollins D, Gilbert V, Xavier S, Chen J, Gentle M, tures of hereditary hemorrhagic telangiectasia. Hum Ge- Reddy A, Bo¨ttinger E, Jiang R, Rastaldi MP, et al. 2012. nome Var 2: 15040. BAMBI regulates angiogenesis and endothelial homeo- Herrera B, Inman GJ. 2009. A rapid and sensitive bioassay stasis through modulation of alternative TGF-b signal- for the simultaneous measurement of multiple bone ing. PLoS ONE 7: e39406. morphogenetic proteins. Identification and quantifica- Guo X, Wang XF. 2009. Signaling cross-talk between TGF- tion of BMP4, BMP6 and BMP9 in bovine and human b/BMP and other pathways. Cell Res 19: 71–88. serum. BMC Cell Biol 10: 20. Gupta S, Gill D, Pal SK, Agarwal N. 2015. Activin receptor Hill CS. 2016. Transcriptional control by the SMADs. Cold inhibitors—Dalantercept. Curr Oncol Rep 17: 14. Spring Harb Perspect Biol 8: a022079. Han C, Hong KH, Kim YH, Kim MJ, Song C, Kim MJ, Kim Hinck AP, Mueller TD, Springer TA. 2016. Structural biol- SJ, Raizada MK, Oh SP.2013. SMAD1 deficiency in either ogy and evolution of the TGF-b family. Cold Spring Harb endothelial or smooth muscle cells can predispose mice Perspect Biol 8: a022103. to pulmonary hypertension. Hypertension 61: 1044– Hong KH, Lee YJ, Lee E, Park SO, Han C, Beppu H, Li E, 1052. Raizada MK, Bloch KD, Oh SP.2008. Genetic ablation of Han C, Choe SW, Kim YH, Acharya AP, Keselowsky BG, the Bmpr2 gene in pulmonary endothelium is sufficient Sorg BS, Lee YJ, Oh SP. 2014. VEGF neutralization can to predispose to pulmonary arterial hypertension. Circu- prevent and normalize arteriovenous malformations in lation 118: 722–730.

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 25 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

Huang Z, Wang D, Ihida-Stansbury K, Jones PL, Martin JF. Kim J, Kim M, Jeong Y,Lee WB, Park H, Kwon JY,Kim YM, 2009. Defective pulmonary vascular remodeling in Hwang D, Kwon YG. 2015. BMP9 induces cord blood- Smad8 mutant mice. Hum Mol Genet 18: 2791–2801. derived endothelial progenitor cell differentiation and Huse M, Chen YG, Massague´ J, Kuriyan J. 1999. Crystal ischemic neovascularization via ALK1. Arterioscler structure of the cytoplasmic domain of the type I TGF- Thromb Vasc Biol 35: 2020–2031. b receptor in complex with FKBP12. Cell 96: 425–436. Kirkbride KC, Townsend TA, Bruinsma MW, Barnett JV, Ikeya M, Kawada M, Kiyonari H, Sasai N, Nakao K, Furuta Y, Blobe GC. 2008. Bone morphogenetic proteins signal Sasai Y.2006. Essential pro-Bmp roles of crossveinless 2 in through the transforming growth factor-b type III recep- mouse organogenesis. Development 133: 4463–4473. tor. J Biol Chem 283: 7628–7637. Ishisaki A, Yamato K, Hashimoto S, Nakao A, Tamaki K, Korn C, Augustin HG. 2015. Mechanisms of vessel pruning Nonaka K, ten Dijke P, Sugino H, Nishihara T. 1999. and regression. Dev Cell 34: 5–17. Differential inhibition of Smad6 and Smad7 on bone Lagendijk AK, Hogan BM. 2015. VE-cadherin in vascular morphogenetic protein- and activin-mediated growth development: A coordinator of and tissue arrest and apoptosis in B cells. J Biol Chem 274: morphogenesis. Curr Top Dev Biol 112: 325–352. 13637–13642. Lane KB, Machado RD, Pauciulo MW,Thomson JR, Phillips Jadrich JL, O’Connor MB, Coucouvanis E. 2006. The TGF-b JAIII, Loyd JE, Nichols WC, Trembath RC. 2000. Hetero- activated kinase TAK1 regulates vascular development in zygous germline mutations in BMPR2, encoding a TGF- vivo. Development 133: 1529–1541. b receptor, cause familial primary pulmonary hyperten- sion. The International PPH Consortium. Nat Genet 26: Jerkic M, Kabir MG, Davies A, Yu LX, McIntyre BA, Husain 81–84. NW, Enomoto M, Sotov V, Husain M, Henkelman M, et al. 2011. Pulmonary hypertension in adult Alk1 hetero- Larrivee B, Prahst C, Gordon E, Del Toro R, Mathivet T, zygous mice due to oxidative stress. Cardiovasc Res 92: Duarte A, Simons M, Eichmann A. 2012. ALK1 signaling 375–384. inhibits angiogenesis by cooperating with the Notch pathway. Dev Cell 22: 489–500. Jiang B, Deng Y,Suen C, TahaM, Chaudhary KR, Courtman ´ DW, Stewart DJ. 2016a. Marked strain-specific differenc- Lastres P, Bellon T, Cabanas C, Sanchez-Madrid F, Acevedo ´ es in the SU5416 rat model of severe pulmonary arterial A, Gougos A, Letarte M, Bernabeu C. 1992. Regulated hypertension. Am J Respir Cell Mol Biol 54: 461–468. expression on human macrophages of endoglin, an Arg- Gly-Asp-containing surface antigen. Eur J Immunol 22: Jiang H, Salmon RM, Upton PD, Wei Z, Lawera A, Daven- 393–397. port AP, Morrell NW, Li W. 2016b. The prodomain- Lawrie A. 2014. A report on the use of animal models and bound form of bone morphogenetic protein 10 is bio- phenotyping methods in pulmonary hypertension re- logically active on endothelial cells. J Biol Chem 291: search. Pulm Circ 4: 2–9. 2954–2966. Lebrin F, Goumans MJ, Jonker L, Carvalho RL, Valdimars- Johnson DW, Berg JN, Baldwin MA, Gallione CJ, Marondel dottir G, Thorikay M, Mummery C, Arthur HM, ten I, Yoon SJ, Stenzel TT, Speer M, Pericak-Vance MA, Di- Dijke P.2004. Endoglin promotes endothelial cell prolif- amond A, et al. 1996. Mutations in the activin receptor- eration and TGF-b/ALK1 signal transduction. EMBO J like kinase 1 gene in hereditary haemorrhagic telangiec- 23: 4018–4028. tasia type 2. Nat Genet 13: 189–195. Lebrin F, Srun S, Raymond K, Martin S, van den Brink S, Kalinina N, Agrotis A, Antropova Y, Ilyinskaya O, Smirnov Freitas C, Breant C, Mathivet T,Larrivee B, Thomas JL, et V, Tararak E, Bobik A. 2004. Smad expression in human al. 2010. Thalidomide stimulates vessel maturation and atherosclerotic lesions: Evidence for impaired TGF-b/ reduces epistaxis in individuals with hereditary hemor- Smad signaling in smooth muscle cells of fibrofatty le- rhagic telangiectasia. Nat Med 16: 420–428. sions. Arterioscler Thromb Vasc Biol 24: 1391–1396. Lee NY, Ray B, How T, Blobe GC. 2008. Endoglin promotes Katagiri T, Watabe T. 2016. Bone morphogenetic proteins. transforming growth factor-b-mediated Smad 1/5/8 sig- Cold Spring Harb Perspect Biol 8: a021899. naling and inhibits endothelial cell migration through its Kavsak P, Rasmussen RK, Causing CG, Bonni S, Zhu H, association with GIPC. J Biol Chem 283: 32527–32533. Thomsen GH, Wrana JL. 2000. Smad7 binds to Smurf2 Lee GT, Kwon SJ, Lee JH, Jeon SS, Jang KT, Choi HY, Lee to form an E3 ubiquitin ligase that targets the TGF-b HM, Kim WJ, Kim SJ, Kim IY. 2010. Induction of inter- receptor for degradation. Mol Cell 6: 1365–1375. leukin-6 expression by bone morphogenetic protein-6 in Kienast Y,Jucknischke U, Scheiblich S, Thier M, de Wouters macrophages requires both SMAD and p38 signaling M, Haas A, Lehmann C, Brand V,Bernicke D, Honold K, pathways. J Biol Chem 285: 39401–39408. et al. 2016. Rapid activation of bone morphogenic pro- Lee NY, Golzio C, Gatza CE, Sharma A, Katsanis N, Blobe tein 9 by receptor-mediated displacement of pro-do- GC. 2012. Endoglin regulates PI3-kinase/Akt trafficking mains. J Biol Chem 291: 3395–3410. and signaling to alter endothelial capillary stability dur- Kim JD, Kim J. 2014. Alk3/Alk3b and Smad5 mediate BMP ing angiogenesis. Mol Biol Cell 23: 2412–2423. signaling during lymphatic development in zebrafish. Lesca G, Olivieri C, Burnichon N, Pagella F, Carette MF, Mol Cells 37: 270–274. Gilbert-Dussardier B, Goizet C, Roume J, Rabilloud M, Kim CW, Song H, Kumar S, Nam D, Kwon HS, Chang KH, Saurin JC, et al. 2007. Genotype-phenotype correlations Son DJ, Kang DW, Brodie SA, Weiss D, et al. 2013. Anti- in hereditary hemorrhagic telangiectasia: Data from the inflammatory and antiatherogenic role of BMP receptor French-Italian HHT network. Genet Med 9: 14–22. II in endothelial cells. Arterioscler Thromb Vasc Biol 33: Letteboer TG, Mager JJ, Snijder RJ, Koeleman BP,Lindhout 1350–1359. D, Ploos van Amstel JK, Westermann CJ. 2006. Geno-

26 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

type-phenotype relationship in hereditary haemorrhagic current perspective on established and emerging molec- telangiectasia. J Med Genet 43: 371–377. ular genetic defects. Hum Mutat 36: 1113–1127. Letteboer TG, Benzinou M, Merrick CB, Quigley DA, Zhau Maddaluno L, Rudini N, Cuttano R, Bravi L, Giampietro C, K, Kim IJ, To MD, Jablons DM, van Amstel JK, West- Corada M, Ferrarini L, Orsenigo F, Papa E, Boulday G, et ermann CJ, et al. 2015. Genetic variation in the functional al. 2013. EndMT contributes to the onset and progression ENG allele inherited from the non-affected parent asso- of cerebral cavernous malformations. Nature 498: 492– ciates with presence of pulmonary arteriovenous mal- 496. formation in hereditary hemorrhagic telangiectasia 1 Mahlawat P,Ilangovan U, Biswas T,Sun LZ, Hinck AP.2012. (HHT1) and may influence expression of PTPN14. Front Structure of the Alk1 extracellular domain and charac- Genet 6: 67. terization of its bone morphogenetic protein (BMP) Levet S, Ciais D, Merdzhanova G, Mallet C, Zimmers TA, binding properties. Biochemistry 51: 6328–6341. Lee SJ, Navarro FP, Texier I, Feige JJ, Bailly S, et al. 2013. Bone morphogenetic protein 9 (BMP9) controls lym- Mahmoud M, Allinson KR, Zhai Z, Oakenfull R, Ghandi P, phatic vessel maturation and valve formation. Blood Adams RH, Fruttiger M, Arthur HM. 2010. Pathogenesis 122: 598–607. of arteriovenous malformations in the absence of endo- glin. Circ Res 106: 1425–1433. Levine RJ, Lam C, Qian C, Yu KF, Maynard SE, Sachs BP, Sibai BM, Epstein FH, Romero R, Thadhani R, et al. 2006. Mair KM, Yang XD, Long L, White K, Wallace E, Ewart MA, Soluble endoglin and other circulating antiangiogenic Docherty CK, Morrell NW, MacLean MR. 2015. Sex af- factors in preeclampsia. N Engl J Med 355: 992–1005. fects bone morphogenetic protein type II receptor signal- Li DY,Sorensen LK, Brooke BS, Urness LD, Davis EC, Taylor ing in pulmonary artery smooth muscle cells. Am J Respir DG, Boak BB, WendelDP.1999. Defective angiogenesis in Crit Care Med 191: 693–703. mice lacking endoglin. Science 284: 1534–1537. Majka S, Hagen M, Blackwell T, Harral J, Johnson JA, Gen- Li X, Yang HY, Giachelli CM. 2008. BMP-2 promotes phos- dron R, Paradis H, Crona D, Loyd JE, Nozik-Grayck E, et phate uptake, phenotypic modulation, and calcification al. 2011. Physiologic and molecular consequences of en- of human vascular smooth muscle cells. Atherosclerosis dothelial Bmpr2 mutation. Respir Res 12: 84. 199: 271–277. Maruyama H, Dewachter C, Sakai S, Belhaj A, Rondelet B, Li Y, Messina C, Bendaoud M, Fine DH, Schreiner H, Remmelink M, Vachiery JL, Naeije R, Dewachter L. 2016. Tsiagbe VK. 2010. Adaptive immune response in osteo- Bosentan reverses the hypoxia-induced downregulation clastic bone resorption induced by orally administered of the bone morphogenetic protein signaling in pulmo- Aggregatibacter actinomycetemcomitans in a rat model nary artery smooth muscle cells. Life Sci 159: 111–115. of periodontal disease. Mol Oral Microbiol 25: 275–292. McAllister KA, Grogg KM, Johnson DW, Gallione CJ, Bald- Li CJ, Madhu V, Balian G, Dighe AS, Cui Q. 2015a. Cross- win MA, Jackson CE, Helmbold EA, Markel DS, McKin- talk between VEGF and BMP-6 pathways accelerates os- non WC, Murrell J, et al. 1994. Endoglin, a TGF-b bind- teogenic differentiation of human adipose-derived stem ing protein of endothelial cells, is the gene for hereditary cells. J Cell Physiol 230: 2671–2682. haemorrhagic telangiectasia type 1. Nat Genet 8: 345– Li X, Lim J, Lu J, Pedego TM, Demer L, Tintut Y. 2015b. 351. Protective role of Smad6 in inflammation-induced val- McDonald J, Wooderchak-Donahue W, VanSant Webb C, vular cell calcification. J Cell Biochem 116: 2354–2364. Whitehead K, Stevenson DA, Bayrak-Toydemir P. 2015. Libby P, Ridker PM, Hansson GK. 2011. Progress and chal- Hereditary hemorrhagic telangiectasia: Genetics and lenges in translating the biology of atherosclerosis. Nature molecular diagnostics in a new era. Front Genet 6: 1. 473: 317–325. Medici D, Shore EM, Lounev VY, Kaplan FS, Kalluri R, Long L, MacLean MR, Jeffery TK, Morecroft I, Yang X, Olsen BR. 2010. Conversion of vascular endothelial cells Rudarakanchana N, Southwood M, James V, Trembath into multipotent stem-like cells. Nat Med 16: 1400–1406. RC, Morrell NW.2006. Serotonin increases susceptibility Miller AF, Harvey SA, Thies RS, Olson MS. 2000. Bone to pulmonary hypertension in BMPR2-deficient mice. morphogenetic protein-9. An autocrine/paracrine cyto- Circ Res 98: 818–827. kine in the liver. J Biol Chem 275: 17937–17945. Long L, Ormiston ML, Yang X, Southwood M, Graf S, Ma- Miyazawa K, Miyazono K. 2016. Regulation of TGF-b fam- chado RD, Mueller M, Kinzel B, Yung LM, Wilkinson JM, et al. 2015. Selective enhancement of endothelial BMPR- ily signaling by inhibitory Smads. Cold Spring Harb Per- II with BMP9 reverses pulmonary arterial hypertension. spect Biol 9: a022095. Nat Med 21: 777–785. Miyazono K, Kamiya Y, Morikawa M. 2010. Bone morpho- Luo W,Zhao X, Jin H, TaoL, Zhu J, WangH, Hemmings BA, genetic protein receptors and signal transduction. J Bio- Yang Z. 2015. Akt1 signaling coordinates BMP signaling chem 147: 35–51. and b-catenin activity to regulate second heart field pro- Mohler ERIII, Gannon F,Reynolds C, Zimmerman R, Keane genitor development. Development 142: 732–742. MG, Kaplan FS. 2001. Bone formation and inflammation Maarman G, Lecour S, Butrous G, Thienemann F, Sliwa K. in cardiac valves. Circulation 103: 1522–1528. 2013. A comprehensive review: The evolution of animal Moreno-Miralles I, Ren R, Moser M, Hartnett ME, Patter- models in pulmonary hypertension research; are we there son C. 2011. Bone morphogenetic protein endothelial yet? Pulm Circ 3: 739–756. cell precursor-derived regulator regulates retinal angio- Machado RD, Southgate L, Eichstaedt CA, Aldred MA, Aus- genesis in vivo in a mouse model of oxygen-induced tin ED, Best DH, Chung WK, Benjamin N, Elliott CG, retinopathy. Arterioscler Thromb Vasc Biol 31: 2216– Eyries M, et al. 2015. Pulmonary arterial hypertension: A 2222.

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 27 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

Morikawa M, Derynck R, Miyazono K. 2016. TGF-b and the factor-b1 signaling in the regulation of angiogenesis. TGF-b family: Context-dependent roles in cell and tissue Proc Natl Acad Sci 97: 2626–2631. physiology. Cold Spring Harb Perspect Biol 8: a021873. Ojeda-Fernandez L, Recio-Poveda L, Aristorena M, Lastres Morioka S, Inagaki M, Komatsu Y,Mishina Y,Matsumoto K, P, Blanco FJ, Sanz-Rodriguez F, Gallardo-Vara E, de Las Ninomiya-Tsuji J. 2012. TAK1 kinase signaling regulates Casas-Engel M, Corbı´ A, Arthur HM, et al. 2016. Mice embryonic angiogenesis by modulating endothelial cell lacking endoglin in macrophages show an impaired im- survival and migration. Blood 120: 3846–3857. mune response. PLoS Genet 12: e1005935. Morrell NW, Bloch DB, ten Dijke P, Goumans MJ, Hata A, Ola R, Dubrac A, Han J, Zhang F,Fang JS, Larrivee B, Lee M, Smith J, Yu PB, Bloch KD. 2016. Targeting BMP signal- Urarte AA, Kraehling JR, Genet G, et al. 2016. PI3 kinase ling in cardiovascular disease and anaemia. Nat Rev Car- inhibition improves vascular malformations in mouse diol 13: 106–120. models of hereditary haemorrhagic telangiectasia. Nat Moser M, Binder O, Wu Y, Aitsebaomo J, Ren R, Bode C, Commun 7: 13650. Bautch VL, Conlon FL, Patterson C. 2003. BMPER, a Olsen OE, Wader KF,Misund K, Vatsveen TK, Ro TB, Mylin novel endothelial cell precursor-derived protein, antago- AK, Turesson I, Stordal BF, Moen SH, Standal T, et al. nizes bone morphogenetic protein signaling and endo- 2014. Bone morphogenetic protein-9 suppresses growth thelial cell differentiation. Mol Cell Biol 23: 5664–5679. of myeloma cells by signaling through ALK2 but is inhib- Moya IM, Umans L, Maas E, Pereira PN, Beets K, Francis A, ited by endoglin. Blood Cancer J 4: e196. Sents W,Robertson EJ, Mummery CL, Huylebroeck D, et Onichtchouk D, Chen YG, Dosch R, Gawantka V,Delius H, al. 2012. Stalk cell phenotype depends on integration of Massague´ J, Niehrs C. 1999. Silencing of TGF-b signal- Notch and Smad1/5 signaling cascades. Dev Cell 22: ling by the pseudoreceptor BAMBI. Nature 401: 480– 501–514. 485. Mueller TD, Nickel J. 2012. Promiscuity and specificity in Park SO, Lee YJ, Seki T, Hong KH, Fliess N, Jiang Z, Park A, BMP receptor activation. FEBS Lett 586: 1846–1859. Wu X, Kaartinen V, Roman BL, et al. 2008. ALK5- and Murakami G, Watabe T, Takaoka K, Miyazono K, Imamura TGFBR2-independent role of ALK1 in the pathogenesis T. 2003. Cooperative inhibition of bone morphogenetic of hereditary hemorrhagic telangiectasia type 2. Blood protein signaling by Smurf1 and inhibitory Smads. Mol 111: 633–642. Biol Cell 14: 2809–2817. Park SO, Wankhede M, Lee YJ, Choi EJ, Fliess N, Choe SW, Nakagawa Y, Ikeda K, Akakabe Y, Koide M, Uraoka M, Yu- Oh SH, Walter G, Raizada MK, Sorg BS, et al. 2009. Real- taka KT, Kurimoto-Nakano R, Takahashi T, Matoba S, time imaging of de novo arteriovenous malformation in a Yamada H, et al. 2010. Paracrine osteogenic signals via mouse model of hereditary hemorrhagic telangiectasia. J bone morphogenetic protein-2 accelerate the atheroscle- Clin Invest 119: 3487–3496. rotic intimal calcification in vivo. Arterioscler Thromb Pawlikowska L, Tran MN, Achrol AS, Ha C, Burchard E, Vasc Biol 30: 1908–1915. Choudhry S, Zaroff J, Lawton MT, Castro R, McCulloch Necchi A, Giannatempo P,Mariani L, Fare E, Raggi D, Pen- CE, et al. 2005. Polymorphisms in transforming growth nati M, Zaffaroni N, Crippa F,Marchiano A, Nicolai N, et factor-b-related genes ALK1 and ENG are associated al. 2014. PF-03446962, a fully-human monoclonal anti- with sporadic brain arteriovenous malformations. Stroke body against transforming growth-factor-b (TGF-b)re- 36: 2278–2280. ceptor ALK1, in pre-treated patients with urothelial can- Peacock HM, Caolo V, Jones EA. 2016. Arteriovenous mal- cer: An open label, single-group, phase 2 trial. Invest New formations in hereditary haemorrhagic telangiectasia: Drugs 32: 555–560. Looking beyond ALK1-NOTCH interactions. Cardiovasc Neuhaus H, Rosen V, Thies RS. 1999. Heart specific expres- Res 109: 196–203. sion of mouse BMP-10 a novel member of the TGF-b Pece-Barbara N, Vera S, Kathirkamathamby K, Liebner S, Di superfamily. Mech Dev 80: 181–184. Guglielmo GM, Dejana E, Wrana JL, Letarte M. 2005. New SE, Aikawa E. 2011. Cardiovascular calcification: An Endoglin null endothelial cells proliferate faster and are inflammatory disease. Circ J 75: 1305–1313. more responsive to transforming growth factor-b1 with Nicolls MR, Mizuno S, Taraseviciene-Stewart L, Farkas L, higher affinity receptors and an activated Alk1 pathway. J Drake JI, Al Husseini A, Gomez-Arroyo JG, Voelkel NF, Biol Chem 280: 27800–27808. Bogaard HJ. 2012. New models of pulmonary hyperten- Pi X, Schmitt CE, Xie L, Portbury AL, Wu Y,Lockyer P,Dyer sion based on VEGF receptor blockade-induced endo- LA, Moser M, Bu G, Flynn EJIII, et al. 2012. LRP1-de- thelial cell apoptosis. Pulm Circ 2: 434–442. pendent endocytic mechanism governs the signaling out- Niessen K, Zhang G, Ridgway JB, Chen H, Yan M. 2010. put of the bmp system in endothelial cells and in angio- ALK1 signaling regulates early postnatal lymphatic vessel genesis. Circ Res 111: 564–574. development. Blood 115: 1654–1661. Post S, Smits AM, van den Broek AJ, Sluijter JP, Hoefer IE, O’Connell PJ, McKenzie A, Fisicaro N, Rockman SP,Pearse Janssen BJ, Snijder RJ, Mager JJ, Pasterkamp G, Mummery MJ, d’Apice AJ. 1992. Endoglin: A 180-kD endothelial CL, et al. 2010. Impaired recruitment of HHT-1 mono- cell and macrophage restricted differentiation molecule. nuclear cells to the ischaemic heart is due to an altered Clin Exp Immunol 90: 154–159. CXCR4/CD26 balance. Cardiovasc Res 85: 494–502. Oh SP,Seki T, Goss KA, Imamura T, Yi Y,Donahoe PK, Li L, Rabinovitch M, Guignabert C, Humbert M, Nicolls MR. 2014. Miyazono K, ten Dijke P, Kim S, et al. 2000. Activin Inflammation and immunity in the pathogenesis of pul- receptor-like kinase 1 modulates transforming growth monary arterial hypertension. Circ Res 115: 165–175.

28 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

Ramirez F, Rifkin DB. 2009. Extracellular microfibrils: Con- Scharpfenecker M, van Dinther M, Liu Z, van Bezooijen RL, textual platforms for TGF-b and BMP signaling. Curr Zhao Q, Pukac L, Lowik CW, ten Dijke P. 2007. BMP-9 Opin Cell Biol 21: 616–622. signals via ALK1 and inhibits bFGF-induced endothelial Ranchoux B, Antigny F, Rucker-Martin C, Hautefort A, Pe- cell proliferation and VEGF-stimulated angiogenesis. J choux C, Bogaard HJ, Dorfmuller P, Remy S, Lecerf F, Cell Sci 120: 964–972. Plante S, et al. 2015. Endothelial-to-mesenchymal tran- Seghers L, de Vries MR, Pardali E, Hoefer IE, Hierck BP,ten sition in pulmonary hypertension. Circulation 131: Dijke P, Goumans MJ, Quax PH. 2012. Shear induced 1006–1018. collateral artery growth modulated by endoglin but not Rathouska J, Jezkova K, Nemeckova I, Nachtigal P. 2015. by ALK1. J Cell Mol Med 16: 2440–2450. Soluble endoglin, hypercholesterolemia and endothelial Seki T, Yun J, Oh SP. 2003. Arterial endothelium-specific dysfunction. Atherosclerosis 243: 383–388. activin receptor-like kinase 1 expression suggests its role Reddi AH. 2005. BMPs: From bone morphogenetic proteins in arterialization and vascular remodeling. Circ Res 93: to body morphogenetic proteins. Cytokine Growth Factor 682–689. Rev 16: 249–250. Sengle G, Ono RN, Lyons KM, Bachinger HP,Sakai LY.2008. A new model for growth factor activation: Type II recep- Ricard N, Ciais D, Levet S, Subileau M, Mallet C, Zimmers tors compete with the prodomain for BMP-7. J Mol Biol TA, Lee SJ, Bidart M, Feige JJ, Bailly S. 2012. BMP9 and 381: 1025–1039. BMP10 are critical for postnatal retinal vascular remod- eling. Blood 119: 6162–6171. Sengle G, Ono RN, Sasaki T, Sakai LY. 2011. Prodomains of transforming growth factor-b (TGF-b) superfamily Risau W. 1997. Mechanisms of angiogenesis. Nature 386: members specify different functions: Extracellular matrix 671–674. interactions and growth factor bioavailability. J Biol Rocher C, Singla DK. 2013. SMAD-PI3K-Akt-mTOR path- Chem 286: 5087–5099. way mediates BMP-7 polarization of monocytes into M2 Shapiro S, Traiger GL, Turner M, McGoon MD, Wason P, macrophages. PLoS ONE 8: e84009. Barst RJ. 2012. Sex differences in the diagnosis, treatment, Rocher C, Singla R, Singal PK, Parthasarathy S, Singla DK. and outcome of patients with pulmonary arterial hyper- 2012. Bone morphogenetic protein 7 polarizes THP-1 tension enrolled in the registry to evaluate early and long- cells into M2 macrophages. Can J Physiol Pharmacol 90: term pulmonary arterial hypertension disease manage- 947–951. ment. Chest 141: 363–373. Ross S, Hill CS. 2008. How the Smads regulate transcription. Shibuya H, Iwata H, Masuyama N, Gotoh Y, Yamaguchi K, Int J Biochem Cell Biol 40: 383–408. Irie K, Matsumoto K, Nishida E, Ueno N. 1998. Role of Rossi E, Smadja DM, Boscolo E, Langa C, Arevalo MA, TAK1 and TAB1 in BMP signaling in early Xenopus de- Pericacho M, Gamella-Pozuelo L, Kauskot A, Botella velopment. EMBO J 17: 1019–1028. LM, Gaussem P, et al. 2016. Endoglin regulates mural Shim JH, Greenblatt MB, Xie M, Schneider MD, Zou W, cell adhesion in the circulatory system. Cell Mol Life Sci Zhai B, Gygi S, Glimcher LH. 2009. TAK1 is an essential 73: 1715–1739. regulator of BMP signalling in cartilage. EMBO J 28: Rudarakanchana N, Flanagan JA, Chen H, Upton PD, Ma- 2028–2041. chado R, Patel D, Trembath RC, Morrell NW.2002. Func- Shore EM, Xu M, Feldman GJ, Fenstermacher DA, Cho TJ, tional analysis of bone morphogenetic protein type II Choi IH, Connor JM, Delai P,Glaser DL, LeMerrer M, et receptor mutations underlying primary pulmonary hy- al. 2006. A recurrent mutation in the BMP type I receptor pertension. Hum Mol Genet 11: 1517–1525. ACVR1 causes inherited and sporadic fibrodysplasia os- Ruiz S, Zhao H, Chandakkar P, Chatterjee PK, Papoin J, sificans progressiva. Nat Genet 38: 525–527. Blanc L, Metz CN, Campagne F, Marambaud P. 2016. A Shovlin CL. 2010. Hereditary haemorrhagic telangiectasia: mouse model of hereditary hemorrhagic telangiectasia Pathophysiology, diagnosis and treatment. Blood Rev 24: generated by transmammary-delivered immunoblocking 203–219. of BMP9 and BMP10. Sci Rep 5: 37366. Sieber C, Kopf J, Hiepen C, Knaus P.2009. Recent advances Sanchez-Duffhues G, de Vinuesa AG, Lindeman JH, in BMP receptor signaling. Cytokine Growth Factor Rev Mulder-Stapel A, DeRuiter MC, Van Munsteren C, Gou- 20: 343–355. mans MJ, Hierck BP,ten Dijke P.2015. SLUG is expressed Simoes Sato AY, Bub GL, Campos AH. 2014. BMP-2 and -4 in endothelial cells lacking primary cilia to promote cel- produced by vascular smooth muscle cells from athero- lular calcification. Arterioscler Thromb Vasc Biol 35: 616– sclerotic lesions induce monocyte chemotaxis through 627. direct BMPRII activation. Atherosclerosis 235: 45–55. Saremba S, Nickel J, Seher A, Kotzsch A, Sebald W, Mueller Simonneau G, Gatzoulis MA, Adatia I, Celermajer D, Den- TD. 2008. Type I receptor binding of bone morphogenet- ton C, Ghofrani A, Gomez Sanchez MA, Krishna Kumar ic protein 6 is dependent on N-glycosylation of the li- R, Landzberg M, Machado RF, et al. 2013. Updated clin- gand. FEBS J 275: 172–183. ical classification of pulmonary hypertension. J Am Coll Satomi-Kobayashi S, Kinugasa M, Kobayashi R, Hata- Cardiol 62: D34–D41. keyama K, Kurogane Y,Ishida T,Emoto N, Asada Y,Takai Singla DK, Singla R, Wang J. 2016. BMP-7 treatment in- Y, Hirata K, et al. 2012. Osteoblast-like differentiation of creases M2 macrophage differentiation and reduces in- – / – cultured human coronary artery smooth muscle cells by flammation and plaque formation in Apo E mice. bone morphogenetic protein endothelial cell precursor- PLoS ONE 11: e0147897. derived regulator (BMPER). J Biol Chem 287: 30336– Song Y, Jones JE, Beppu H, Keaney JFJr, Loscalzo J, Zhang 30345. YY. 2005. Increased susceptibility to pulmonary hyper-

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 29 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

tension in heterozygous BMPR2-mutant mice. Circula- Talati M, West J, Zaynagetdinov R, Hong CC, Han W,Black- tion 112: 553–562. well T, Robinson L, Blackwell TS, Lane K. 2014. BMP Song K, Krause C, Shi S, Patterson M, Suto R, Grgurevic L, pathway regulation of and by macrophages. PLoS ONE Vukicevic S, van Dinther M, Falb D, ten Dijke P, et al. 9: e94119. 2010. Identification of a key residue mediating bone mor- ten Dijke P, Goumans MJ, Pardali E. 2008. Endoglin in an- phogenetic protein (BMP)-6 resistance to noggin inhibi- giogenesis and vascular diseases. Angiogenesis 11: 79–89. tion allows for engineered BMPs with superior agonist Tian F, Zhou AX, Smits AM, Larsson E, Goumans MJ, Hel- activity. J Biol Chem 285: 12169–12180. din CH, Boren J, Akyurek LM. 2010. Endothelial cells are Song R, Fullerton DA, Ao L, Zheng D, Zhao KS, Meng X. activated during hypoxia via endoglin/ALK-1/SMAD1/ 2015. BMP-2 and TGF-b1 mediate biglycan-induced 5 signaling in vivo and in vitro. Biochem Biophys Res pro-osteogenic reprogramming in aortic valve interstitial Commun 392: 283–288. cells. J Mol Med (Berl) 93: 403–412. Tian H, Mythreye K, Golzio C, Katsanis N, Blobe GC. 2012. Soon E, Crosby A, Southwood M, Yang P, Tajsic T, Toshner Endoglin mediates fibronectin/a5b1 integrin and TGF- M, Appleby S, Shanahan CM, Bloch KD, Pepke-Zaba J, et b pathway crosstalk in endothelial cells. EMBO J 31: al. 2015. Bone morphogenetic protein receptor type II 3885–3900. deficiency and increased inflammatory cytokine produc- Tillet E, Bailly S. 2014. Emerging roles of BMP9 and BMP10 tion. A gateway to pulmonary arterial hypertension. Am J in hereditary hemorrhagic telangiectasia. Front Genet 5: Respir Crit Care Med 192: 859–872. 456. Souza TA, Chen X, Guo Y, Sava P,Zhang J, Hill JJ, Yaworsky Tkachenko E, Gutierrez E, Saikin SK, Fogelstrand P,Kim C, PJ, Qiu Y. 2008. Proteomic identification and functional Groisman A, Ginsberg MH. 2013. The nucleus of endo- validation of activins and bone morphogenetic protein thelial cell as a sensor of blood flow direction. Biology 11 as candidate novel muscle mass regulators. Mol Endo- Open 2: 1007–1012. crinol 22: 2689–2702. Toporsian M, Jerkic M, Zhou YQ, Kabir MG, Yu LX, McIn- Spiekerkoetter E, Tian X, Cai J, Hopper RK, Sudheendra D, tyre BA, Davis A, WangYJ, Stewart DJ, Belik J, et al. 2010. Li CG, El-Bizri N, Sawada H, Haghighat R, Chan R, et al. Spontaneous adult-onset pulmonary arterial hyperten- 2013. FK506 activates BMPR2, rescues endothelial dys- sion attributable to increased endothelial oxidative stress function, and reverses pulmonary hypertension. J Clin in a murine model of hereditary hemorrhagic telangiec- Invest 123: 3600–3613. tasia. Arterioscler Thromb Vasc Biol 30: 509–517. Spiekerkoetter E, Sung YK, Sudheendra D, Bill M, Aldred Topper JN, Cai J, Qiu Y, Anderson KR, Xu YY, Deeds JD, MA, van de Veerdonk MC, Vonk Noordegraaf A, Long- Feeley R, Gimeno CJ, Woolf EA, Tayber O, et al. 1997. Boyle J, Dash R, Yang PC, et al. 2015. Low-dose FK506 Vascular MADs: Two novel MAD-related genes selective- (tacrolimus) in end-stage pulmonary arterial hyperten- ly inducible by flow in human vascular endothelium. Proc sion. Am J Respir Crit Care Med 192: 254–257. Natl Acad Sci 94: 9314–9319. Srinivasan S, Hanes MA, Dickens T, Porteous ME, Oh SP, Torsney E, Charlton R, Diamond AG, Burn J, Soames JV, Hale LP, Marchuk DA. 2003. A mouse model for hered- Arthur HM. 2003. Mouse model for hereditary hemor- itary hemorrhagic telangiectasia (HHT) type 2. Hum Mol rhagic telangiectasia has a generalized vascular abnor- Genet 12: 473–482. mality. Circulation 107: 1653–1657. Srinivasan RS, Escobedo N, YangY,Interiano A, Dillard ME, Townson SA, Martinez-Hackert E, Greppi C, Lowden P, Finkelstein D, Mukatira S, Gil HJ, Nurmi H, Alitalo K, et Sako D, Liu J, Ucran JA, Liharska K, Underwood KW, al. 2014. The Prox1–Vegfr3 feedback loop maintains the Seehra J, et al. 2012. Specificity and structure of a high identity and the number of lymphatic endothelial cell affinity activin receptor-like kinase 1 (ALK1) signaling progenitors. Genes Dev 28: 2175–2187. complex. J Biol Chem 287: 27313–27325. Stahl S, Gaetzner S, Voss K, Brackertz B, Schleider E, Surucu Tual-Chalot S, Mahmoud M, Allinson KR, Redgrave RE, O, Kunze E, Netzer C, Korenke C, Finckh U, et al. 2008. Zhai Z, Oh SP,Fruttiger M, Arthur HM. 2014. Endothe- Novel CCM1, CCM2, and CCM3 mutations in patients lial depletion of Acvrl1 in mice leads to arteriovenous with cerebral cavernous malformations: In-frame dele- malformations associated with reduced endoglin expres- tion in CCM2 prevents formation of a CCM1/CCM2/ sion. PLoS ONE 9: e98646. CCM3 protein complex. Hum Mutat 29: 709–717. Tual-Chalot S, Oh SP, Arthur HM. 2015. Mouse models of Strasky Z, Vecerova L, Rathouska J, Slanarova M, Brcakova hereditary hemorrhagic telangiectasia: Recent advances E, Kudlackova Z, Andrys C, Micuda S, Nachtigal P.2011. and future challenges. Front Genet 6: 25. Cholesterol effects on endoglin and its downstream path- ways in ApoE/LDLR double knockout mice. Circ J 75: Tuder RM, Stacher E, Robinson J, Kumar R, Graham BB. 1747–1755. 2013. Pathology of pulmonary hypertension. Clin Chest Med 34: 639–650. Suzuki Y,Ohga N, Morishita Y,Hida K, Miyazono K, Watabe T.2010. BMP-9 induces proliferation of multiple types of Umulis D, O’Connor MB, Blair SS. 2009. The extracellular endothelial cells in vitro and in vivo. J Cell Sci 123: 1684– regulation of bone morphogenetic protein signaling. De- 1692. velopment 136: 3715–3728. Takahashi H, Goto N, Kojima Y, Tsuda Y, Morio Y, Mura- Upton PD, Morrell NW. 2009. TGF-b and BMPR-II phar- matsu M, Fukuchi Y. 2006. Downregulation of type II macology—Implications for pulmonary vascular diseas- bone morphogenetic protein receptor in hypoxic pulmo- es. Curr Opin Pharmacol 9: 274–280. nary hypertension. Am J Physiol Lung Cell Mol Physiol Upton PD, Long L, Trembath RC, Morrell NW. 2008. Func- 290: L450–L458. tional characterization of bone morphogenetic protein

30 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

BMPs in Vascular Homeostatis and Disease

binding sites and Smad1/5 activation in human vascular combined with VEGFR TKI leads to tumor stasis in renal cells. Mol Pharmacol 73: 539–552. cell carcinoma. Oncotarget 7: 41857–41869. Upton PD, Davies RJ, Trembath RC, Morrell NW. 2009. Weis SM, Cheresh DA. 2005. Pathophysiological conse- Bone morphogenetic protein (BMP) and activin type II quences of VEGF-induced vascular permeability. Nature receptors balance BMP9 signals mediated by activin re- 437: 497–504. ceptor-like kinase-1 in human pulmonary artery endo- West J, Fagan K, Steudel W, Fouty B, Lane K, Harral J, thelial cells. J Biol Chem 284: 15794–15804. Hoedt-Miller M, Tada Y, Ozimek J, Tuder R, et al. 2004. Urist MR. 1965. Bone: Formation by autoinduction. Science Pulmonary hypertension in transgenic mice expressing a 150: 893–899. dominant-negative BMPRII gene in smooth muscle. Circ Urness LD, Sorensen LK, Li DY. 2000. Arteriovenous mal- Res 94: 1109–1114. formations in mice lacking activin receptor-like kinase-1. West J, Harral J, Lane K, Deng Y, Ickes B, Crona D, Albu S, Nat Genet 26: 328–331. Stewart D, Fagan K. 2008. Mice expressing BMPR2R899X Valbuena-Diez AC, Blanco FJ, Oujo B, Langa C, Gonzalez- transgene in smooth muscle develop pulmonary vascular Nunez M, Llano E, Pendas AM, Diaz M, Castrillo A, lesions. Am J Physiol Lung Cell Mol Physiol 295: L744– Lopez-Novoa JM, et al. 2012. Oxysterol-induced soluble L755. endoglin release and its involvement in hypertension. Willis MS, Dyer LA, Ren R, Lockyer P, Moreno-Miralles I, Circulation 126: 2612–2624. Schisler JC, Patterson C. 2013. BMPER regulates cardio- Valdimarsdottir G, Goumans MJ, Itoh F,Itoh S, Heldin CH, myocyte size and vessel density in vivo. Cardiovasc Pathol ten Dijke P.2006. Smad7 and protein phosphatase 1a are 22: 228–240. critical determinants in the duration of TGF-b/ALK1 Wohl AP, Troilo H, Collins RF, Baldock C, Sengle G. 2016. signaling in endothelial cells. BMC Cell Biol 7: 16. Extracellular regulation of bone morphogenetic protein van Laake LW, van den Driesche S, Post S, Feijen A, Jansen activity by the microfibril component fibrillin-1. J Biol MA, Driessens MH, Mager JJ, Snijder RJ, Westermann CJ, Chem 291: 12732–12746. Doevendans PA, et al. 2006. Endoglin has a crucial role in Wooderchak-Donahue WL, McDonald J, O’Fallon B, Upton blood cell-mediated vascular repair. Circulation 114: PD, Li W, Roman BL, Young S, Plant P, Fulop GT, Langa 2288–2297. C, et al. 2013. BMP9 mutations cause a vascular-anomaly Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, syndrome with phenotypic overlap with hereditary hem- Kim YM, Bdolah Y,Lim KH, Yuan HT, Libermann TA, et orrhagic telangiectasia. Am J Hum Genet 93: 530–537. al. 2006. Soluble endoglin contributes to the pathogene- sis of preeclampsia. Nat Med 12: 642–649. Xu B, Wu YQ, Huey M, Arthur HM, Marchuk DA, Hashi- moto T, Young WL, Yang GY. 2004. Vascular endothelial Vittet D. 2014. Lymphatic collecting vessel maturation and growth factor induces abnormal microvasculature in the valve morphogenesis. Microvasc Res 96: 31–37. endoglin heterozygous mouse brain. J Cereb Blood Flow Vukicevic S, Grgurevic L. 2009. BMP-6 and mesenchymal Metab 24: 237–244. differentiation. Cytokine Growth Factor Rev 20: Xu P,Lin X, Feng X-H. 2016. Posttranslational regulation of 441–448. Smads. Cold Spring Harb Perspect Biol 8: a022087. Wakayama Y, Fukuhara S, Ando K, Matsuda M, Mochizuki YadinD, Knaus P,Mueller TD. 2016. Structural insights into N. 2015. Cdc42 mediates Bmp-induced sprouting angio- BMP receptors: Specificity, activation and inhibition. Cy- genesis through Fmnl3-driven assembly of endothelial tokine Growth Factor Rev 27: 13–34. filopodia in zebrafish. Dev Cell 32: 109–122. Walker EJ, Su H, Shen F, Choi EJ, Oh SP, Chen G, Lawton Yamaguchi K, Shirakabe K, Shibuya H, Irie K, Oishi I, Ueno MT, Kim H, Chen Y, Chen W, et al. 2011. Arteriovenous N, Taniguchi T, Nishida E, Matsumoto K. 1995. Identi- malformation in the adult mouse brain resembling the fication of a member of the MAPKKK family as a poten- human disease. Ann Neurol 69: 954–962. tial mediator of TGF-b signal transduction. Science 270: 2008–2011. Walker EJ, Su H, Shen F, Degos V, Amend G, Jun K, Young WL. 2012. Bevacizumab attenuates VEGF-induced an- Yang Y, Oliver G. 2014. Development of the mammalian giogenesis and vascular malformations in the adult lymphatic vasculature. J Clin Invest 124: 888–897. mouse brain. Stroke 43: 1925–1930. Yang X, Castilla LH, Xu X, Li C, Gotay J, Weinstein M, Liu Walshe TE, Dole VS, Maharaj AS, Patten IS, Wagner DD, PP,Deng CX. 1999. Angiogenesis defects and mesenchy- D’Amore PA. 2009. Inhibition of VEGF or TGF-b signal- mal apoptosis in mice lacking SMAD5. Development 126: ing activates endothelium and increases leukocyte roll- 1571–1580. ing. Arterioscler Thromb Vasc Biol 29: 1185–1192. YangY,Garcia-Verdugo JM, Soriano-Navarro M, Srinivasan Wang X, Harris RE, Bayston LJ, Ashe HL. 2008. Type IV RS, Scallan JP, Singh MK, Epstein JA, Oliver G. 2012. collagens regulate BMP signalling in Drosophila. Nature Lymphatic endothelial progenitors bud from the cardinal 455: 72–77. vein and intersomitic vessels in mammalian embryos. Wang G, Fan R, Ji R, Zou W, Penny DJ, Varghese NP,Fan Y. Blood 120: 2340–2348. 2016a. Novel homozygous BMP9 nonsense mutation Yao Y, Zebboudj AF, Shao E, Perez M, Bostro¨m K. 2006. causes pulmonary arterial hypertension: A case report. Regulation of bone morphogenetic protein-4 by matrix BMC Pulm Med 16: 17. GLA protein in vascular endothelial cells involves activin- Wang X, Solban N, Khanna P, Callea M, Song J, Alsop DC, like kinase receptor 1. J Biol Chem 281: 33921–33930. Pearsall RS, Atkins MB, Mier JW, Signoretti S, et al. Yao Y,Bennett BJ, Wang X, Rosenfeld ME, Giachelli C, Lusis 2016b. Inhibition of ALK1 signaling with dalantercept AJ, Bostro¨m KI. 2010. Inhibition of bone morphogenetic

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 31 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

M.-J. Goumans et al.

proteins protects against atherosclerosis and vascular cal- Zebboudj AF, Imura M, Bostrom K. 2002. Matrix GLA pro- cification. Circ Res 107: 485–494. tein, a regulatory protein for bone morphogenetic pro- YaoY,Jumabay M, Ly A, Radparvar M, Wang AH, Abdmau- tein-2. J Biol Chem 277: 4388–4394. len R, Bostro¨m KI. 2012. Crossveinless 2 regulates bone Zeng Y, Liu H, Kang K, Wang Z, Hui G, Zhang X, Zhong J, morphogenetic protein 9 in human and mouse vascular Peng W, Ramchandran R, Raj JU, et al. 2015. Hypoxia endothelium. Blood 119: 5037–5047. inducible factor-1 mediates expression of miR-322: Po- tential role in proliferation and migration of pulmonary Yao Y, Yao J, Radparvar M, Blazquez-Medela AM, Guihard arterial smooth muscle cells. Sci Rep 5: 12098. PJ, Jumabay M, Bostro¨m KI. 2013. Reducing Jagged 1 and 2 levels prevents cerebral arteriovenous malformations in Zhang YE. 2017. Non-Smad signaling pathways of the TGF- b family. Cold Spring Harb Perspect Biol 9: a022129. matrix Gla protein deficiency. Proc Natl Acad Sci 110: 19071–19076. Zhang M, Sara JD, WangFL, Liu LP,Su LX, Zhe J, Wu X, Liu JH. 2015. Increased plasma BMP-2 levels are associated Yoshimatsu Y, Lee YG, Akatsu Y, Taguchi L, Suzuki HI, with atherosclerosis burden and coronary calcification in Cunha SI, Maruyama K, Suzuki Y, Yamazaki T, Katsura type 2 diabetic patients. Cardiovasc Diabetol 14: 64. A, et al. 2013. Bone morphogenetic protein-9 inhibits Zheng W, Aspelund A, Alitalo K. 2014. Lymphangiogenic lymphatic vessel formation via activin receptor-like ki- factors, mechanisms, and applications. J Clin Invest 124: nase 1 during development and cancer progression. Proc 878–887. Natl Acad Sci 110: 18940–18945. Zhu D, Mackenzie NC, Shanahan CM, Shroff RC, Farqu- Yung LM, Sanchez-Duffhues G, ten Dijke P, Yu PB. 2015. harson C, MacRae VE. 2015. BMP-9 regulates the osteo- Bone morphogenetic protein 6 and oxidized low-density blastic differentiation and calcification of vascular lipoprotein synergistically recruit osteogenic differentia- smooth muscle cells through an ALK1 mediated pathway. tion in endothelial cells. Cardiovasc Res 108: 278–287. J Cell Mol Med 19: 165–174.

32 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a031989 Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

Bone Morphogenetic Proteins in Vascular Homeostasis and Disease

Marie-José Goumans, An Zwijsen, Peter ten Dijke and Sabine Bailly

Cold Spring Harb Perspect Biol published online March 27, 2017

Subject Collection The Biology of the TGF-β Family

TGF-β Family Signaling in Early Vertebrate TGF-β Family Signaling in Mesenchymal Development Differentiation Joseph Zinski, Benjamin Tajer and Mary C. Mullins Ingo Grafe, Stefanie Alexander, Jonathan R. Peterson, et al. Bone Morphogenetic Protein−Based Therapeutic TGF-β1 Signaling and Tissue Fibrosis Approaches Kevin K. Kim, Dean Sheppard and Harold A. Jonathan W. Lowery and Vicki Rosen Chapman TGF-β Family Signaling in Ductal Differentiation Bone Morphogenetic Proteins in Vascular and Branching Morphogenesis Homeostasis and Disease Kaoru Kahata, Varun Maturi and Aristidis Marie-José Goumans, An Zwijsen, Peter ten Dijke, Moustakas et al. TGF-β Signaling in Control of Cardiovascular TGF-β Family Signaling in Epithelial Function Differentiation and Epithelial−Mesenchymal Marie-José Goumans and Peter ten Dijke Transition Kaoru Kahata, Mahsa Shahidi Dadras and Aristidis Moustakas TGF-β Family Signaling in Tumor Suppression TGF-β Family Signaling in Connective Tissue and and Cancer Progression Skeletal Diseases Joan Seoane and Roger R. Gomis Elena Gallo MacFarlane, Julia Haupt, Harry C. Dietz, et al. Targeting TGF-β Signaling for Therapeutic Gain The TGF-β Family in the Reproductive Tract Rosemary J. Akhurst Diana Monsivais, Martin M. Matzuk and Stephanie A. Pangas Regulation of Hematopoiesis and Hematological TGF-β Family Signaling in Drosophila Disease by TGF- β Family Signaling Molecules Ambuj Upadhyay, Lindsay Moss-Taylor, Myung-Jun Kazuhito Naka and Atsushi Hirao Kim, et al. TGF-β Family Signaling in Neural and Neuronal Signaling Cross Talk between TGF-β/Smad and Differentiation, Development, and Function Other Signaling Pathways Emily A. Meyers and John A. Kessler Kunxin Luo For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2017 Cold Spring Harbor Laboratory Press; all rights reserved Downloaded from http://cshperspectives.cshlp.org/ on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2017 Cold Spring Harbor Laboratory Press; all rights reserved