+ All Categories
Home > Documents > Cerebral cavernous malformation protein CCM1 inhibits sprouting ... · men formation, and sprouting...

Cerebral cavernous malformation protein CCM1 inhibits sprouting ... · men formation, and sprouting...

Date post: 18-Feb-2019
Category:
Upload: trandan
View: 219 times
Download: 0 times
Share this document with a friend
6
Cerebral cavernous malformation protein CCM1 inhibits sprouting angiogenesis by activating DELTA-NOTCH signaling Joycelyn Wüstehube a,b,1 , Arne Bartol a,b,1 , Sven S. Liebler a,b , René Brütsch a , Yuan Zhu c , Ute Felbor d , Ulrich Sure c , Hellmut G. Augustin a,b , and Andreas Fischer a,b,2 a Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim, Heidelberg University, D-68167 Mannheim, Germany; b Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), D-69120 Heidelberg, Germany; c Department of Neurosurgery, University Hospital Essen, D-45122 Essen, Germany; and d Institute of Human Genetics, University Hospital Greifswald, D-17475 Greifswald, Germany Edited by Napoleone Ferrara, Genentech, Inc., South San Francisco, CA, and approved June 7, 2010 (received for review January 7, 2010) Cerebral cavernous malformations (CCM) are frequent vascular abnormalities caused by mutations in one of the CCM genes. CCM1 (also known as KRIT1) stabilizes endothelial junctions and is essential for vascular morphogenesis in mouse embryos. However, cellular functions of CCM1 during the early steps of the CCM path- ogenesis remain unknown. We show here that CCM1 represents an antiangiogenic protein to keep the human endothelium quiescent. CCM1 inhibits endothelial proliferation, apoptosis, migration, lu- men formation, and sprouting angiogenesis in primary human en- dothelial cells. CCM1 strongly induces DLL4-NOTCH signaling, which promotes AKT phosphorylation but reduces phosphorylation of the mitogen-activated protein kinase ERK. Consistently, blocking of NOTCH activity alleviates CCM1 effects. ERK phosphorylation is increased in human CCM lesions. Transplantation of CCM1-silenced human endothelial cells into SCID mice recapitulates hallmarks of the CCM pathology and serves as a unique CCM model system. In this setting, the multikinase inhibitor Sorafenib can ameliorate loss of CCM1-induced excessive microvascular growth, reducing the microvessel density to levels of normal wild-type endothelial cells. Collectively, our data suggest that the origin of CCM lesions is caused by perturbed Notch signaling-induced excessive capillary sprouting, which can be therapeutically targeted. endothelial cells | vascular malformations | CCM | KRIT1 C erebral cavernous malformations (CCM) [OMIM: 116860] are frequent vascular abnormalities, predominately localized in the brain, affecting up to 0.5% of the human population (1). CCM lesions are characterized by grossly enlarged vascular chan- nels, often lacking support of mural cells. The carriers can develop a symptomatic disease with headaches, seizures, focal neurological decits, or hemorrhages. The disease is caused by mutations in one of the three known CCM genes, namely KRIT1 (CCM1), OSM (CCM2), or PDCD10 (CCM3), and can occur sporadically or fa- milial with high penetrance (2). Recent data suggest a two-hit mechanismfor the cause of localized lesions, where a germ-line loss of one allele is followed by a loss of the second allele of in- dividual endothelial cells (3, 4). CCM1 is expressed in astrocytes and endothelial cells, and can be associated to microtubules, membranes, and adherens junctions, but also the nucleus (58). The CCM1 protein is part of a large protein complex together with CCM2 and CCM3, components of the cytoskeleton and cell junctions, as well as components of signal transduction pathways and lipids (9). The interaction with RAP1 is essential for stabilizing endothelial cell-cell contacts (5). Addi- tionally, the strong binding to the cytoplasmic β1-integrin binding protein-1 ICAP1 (ITGB1BP1) could affect β1-integrin conforma- tion and endothelial-extracellular matrix interactions (10); how- ever, detailed molecular and cellular functions remain elusive. A Ccm1 null mutation in mouse causes embryonic lethality. Many major blood vessels of mutant embryos are dilated, and others are narrowed and the expression of arterial marker genes is strongly reduced, suggesting a defect in arterial/venous differenti- ation (11). In contrast, loss of ccm1 in zebrash does not alter ar- terial marker gene expression, but leads to dilation of major vessels because of excessive spreading of endothelial cells (12). The clinical observation that CCM lesions grow in response to VEGF (13) suggests that their formation could be driven by angiogenesis: the growth of new blood vessels from preexisting ones. We conse- quently hypothesized that CCM lesions may reect a pathological angiogenesis. The experiments revealed a role of CCM1 as an ac- tivator of vascular DELTA-NOTCH signaling. Correspondingly, perturbed DELTA-NOTCH signaling was identied as a pathoge- netic cause of CCM1 mutation-mediated vascular malformations. Results CCM1 Is a Negative Regulator of Sprouting Angiogenesis. Human umbilical vein endothelial cells (HUVEC) express CCM1 mRNA as determined by quantitative real-time RT-PCR (qPCR). When cultured on Matrigel, HUVEC spontaneously organized into cords and formed a honeycomb-like network. Forced expression of CCM1 inhibited such network formation (Fig. 1 A and B). HUVEC remained isolated or in clumps of cells and formed fewer tubes and branch points compared with GFP overexpression as control. CCM1 expression caused a drastic reduction of VEGF- or FGF2-induced sprout formation in a 3D spheroidal system of endothelial differentiation and capillary formation (14) (Fig. 1C). The short sprouts failed to form a lumen. Because CCM lesions are typically caused by a loss of CCM1 function, we investigated whether down-regulation of this gene would lead to enhanced angiogenesis. Two independent CCM1 siRNAs, as well as a len- tiviral shRNA vector, led to substantial silencing of CCM1 mRNA expression (Fig. 1D). HUVEC with silenced CCM1 expression formed signicantly more long capillary sprouts compared with nonsilencing siRNA-transfected control cells (Fig. 1E). When CCM1 shRNA transduced HUVEC (green uorescent) were mixed with control cells (red uorescent), the CCM1-depleted HUVEC formed more and larger sprouts (Fig. 1F and Movie S1). Cells overexpressing CCM1 formed only very few and short sprouts when mixed with control HUVEC (Fig. 1G and Movie S2). Author contributions: J.W., A.B., S.S.L., R.B., Y.Z., H.G.A., and A.F. designed research; J.W., A.B., S.S.L., R.B., Y.Z., and A.F. performed research; U.F., U.S., and H.G.A. contributed new reagents/analytic tools; J.W., A.B., S.S.L., R.B., Y.Z., and A.F. analyzed data; and J.W., H.G.A., and A.F. wrote the paper. The authors declare no conict of interest. This article is a PNAS Direct Submission. Data deposition: The data reported in this paper have been deposited in the Gene Ex- pression Omnibus (GEO) database, www.ncbi.nlm.nih.gov/geo (accession no. GSE18014). 1 J.W. and A.B. contributed equally to this work. 2 To whom correspondence should be addressed. E-mail: andreas.[email protected] heidelberg.de. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1000132107/-/DCSupplemental. 1264012645 | PNAS | July 13, 2010 | vol. 107 | no. 28 www.pnas.org/cgi/doi/10.1073/pnas.1000132107
Transcript
Page 1: Cerebral cavernous malformation protein CCM1 inhibits sprouting ... · men formation, and sprouting angiogenesis in primary human en- dothelialcells.CCM1stronglyinducesDLL4-NOTCHsignaling,which

Cerebral cavernous malformation protein CCM1inhibits sprouting angiogenesis by activatingDELTA-NOTCH signalingJoycelyn Wüstehubea,b,1, Arne Bartola,b,1, Sven S. Lieblera,b, René Brütscha, Yuan Zhuc, Ute Felbord, Ulrich Surec,Hellmut G. Augustina,b, and Andreas Fischera,b,2

aVascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim, Heidelberg University, D-68167 Mannheim, Germany; bVascular Oncology andMetastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), D-69120 Heidelberg, Germany; cDepartment of Neurosurgery, University Hospital Essen,D-45122 Essen, Germany; and dInstitute of Human Genetics, University Hospital Greifswald, D-17475 Greifswald, Germany

Edited by Napoleone Ferrara, Genentech, Inc., South San Francisco, CA, and approved June 7, 2010 (received for review January 7, 2010)

Cerebral cavernous malformations (CCM) are frequent vascularabnormalities caused by mutations in one of the CCM genes.CCM1 (also known as KRIT1) stabilizes endothelial junctions and isessential for vascular morphogenesis in mouse embryos. However,cellular functions of CCM1 during the early steps of the CCM path-ogenesis remain unknown. We show here that CCM1 represents anantiangiogenic protein to keep the human endothelium quiescent.CCM1 inhibits endothelial proliferation, apoptosis, migration, lu-men formation, and sprouting angiogenesis in primary human en-dothelial cells. CCM1 strongly induces DLL4-NOTCH signaling,whichpromotes AKT phosphorylation but reduces phosphorylation ofthe mitogen-activated protein kinase ERK. Consistently, blockingof NOTCH activity alleviates CCM1 effects. ERK phosphorylation isincreased in human CCM lesions. Transplantation of CCM1-silencedhuman endothelial cells into SCID mice recapitulates hallmarks ofthe CCM pathology and serves as a unique CCM model system. Inthis setting, the multikinase inhibitor Sorafenib can ameliorate lossof CCM1-induced excessive microvascular growth, reducing themicrovessel density to levels of normal wild-type endothelial cells.Collectively, our data suggest that the origin of CCM lesions iscaused by perturbed Notch signaling-induced excessive capillarysprouting, which can be therapeutically targeted.

endothelial cells | vascular malformations | CCM | KRIT1

Cerebral cavernous malformations (CCM) [OMIM: 116860]are frequent vascular abnormalities, predominately localized

in the brain, affecting up to 0.5% of the human population (1).CCM lesions are characterized by grossly enlarged vascular chan-nels, often lacking support of mural cells. The carriers can developa symptomatic disease with headaches, seizures, focal neurologicaldeficits, or hemorrhages. The disease is caused by mutations inone of the three known CCM genes, namely KRIT1 (CCM1),OSM(CCM2), or PDCD10 (CCM3), and can occur sporadically or fa-milial with high penetrance (2). Recent data suggest a “two-hitmechanism” for the cause of localized lesions, where a germ-lineloss of one allele is followed by a loss of the second allele of in-dividual endothelial cells (3, 4).CCM1 is expressed in astrocytes and endothelial cells, and canbe

associated to microtubules, membranes, and adherens junctions,but also the nucleus (5–8). The CCM1 protein is part of a largeprotein complex together with CCM2 and CCM3, components ofthe cytoskeleton and cell junctions, as well as components of signaltransduction pathways and lipids (9). The interaction with RAP1 isessential for stabilizing endothelial cell-cell contacts (5). Addi-tionally, the strong binding to the cytoplasmic β1-integrin bindingprotein-1 ICAP1 (ITGB1BP1) could affect β1-integrin conforma-tion and endothelial-extracellular matrix interactions (10); how-ever, detailed molecular and cellular functions remain elusive.A Ccm1 null mutation in mouse causes embryonic lethality.

Many major blood vessels of mutant embryos are dilated, andothers are narrowed and the expression of arterial marker genes is

strongly reduced, suggesting a defect in arterial/venous differenti-ation (11). In contrast, loss of ccm1 in zebrafish does not alter ar-terial marker gene expression, but leads to dilation of major vesselsbecauseof excessive spreading of endothelial cells (12). The clinicalobservation that CCM lesions grow in response to VEGF (13)suggests that their formation could be driven by angiogenesis: thegrowth of new blood vessels from preexisting ones. We conse-quently hypothesized that CCM lesions may reflect a pathologicalangiogenesis. The experiments revealed a role of CCM1 as an ac-tivator of vascular DELTA-NOTCH signaling. Correspondingly,perturbed DELTA-NOTCH signaling was identified as a pathoge-netic cause of CCM1mutation-mediated vascular malformations.

ResultsCCM1 Is a Negative Regulator of Sprouting Angiogenesis. Humanumbilical vein endothelial cells (HUVEC) express CCM1 mRNAas determined by quantitative real-time RT-PCR (qPCR). Whencultured onMatrigel,HUVECspontaneously organized into cordsand formed a honeycomb-like network. Forced expression ofCCM1 inhibited suchnetwork formation (Fig. 1A andB).HUVECremained isolated or in clumps of cells and formed fewer tubes andbranch points compared with GFP overexpression as control.CCM1 expression caused a drastic reduction of VEGF- or

FGF2-induced sprout formation in a 3D spheroidal system ofendothelial differentiation and capillary formation (14) (Fig. 1C).The short sprouts failed to form a lumen. Because CCM lesionsare typically caused by a loss of CCM1 function, we investigatedwhether down-regulation of this gene would lead to enhancedangiogenesis. Two independent CCM1 siRNAs, as well as a len-tiviral shRNA vector, led to substantial silencing ofCCM1mRNAexpression (Fig. 1D). HUVEC with silenced CCM1 expressionformed significantly more long capillary sprouts compared withnonsilencing siRNA-transfected control cells (Fig. 1E). WhenCCM1 shRNA transduced HUVEC (green fluorescent) weremixed with control cells (red fluorescent), the CCM1-depletedHUVEC formed more and larger sprouts (Fig. 1F andMovie S1).Cells overexpressing CCM1 formed only very few and short sproutswhen mixed with control HUVEC (Fig. 1G and Movie S2).

Author contributions: J.W., A.B., S.S.L., R.B., Y.Z., H.G.A., and A.F. designed research; J.W.,A.B., S.S.L., R.B., Y.Z., and A.F. performed research; U.F., U.S., and H.G.A. contributed newreagents/analytic tools; J.W., A.B., S.S.L., R.B., Y.Z., and A.F. analyzed data; and J.W., H.G.A., andA.F. wrote the paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

Data deposition: The data reported in this paper have been deposited in the Gene Ex-pression Omnibus (GEO) database, www.ncbi.nlm.nih.gov/geo (accession no. GSE18014).1J.W. and A.B. contributed equally to this work.2To whom correspondence should be addressed. E-mail: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1000132107/-/DCSupplemental.

12640–12645 | PNAS | July 13, 2010 | vol. 107 | no. 28 www.pnas.org/cgi/doi/10.1073/pnas.1000132107

Page 2: Cerebral cavernous malformation protein CCM1 inhibits sprouting ... · men formation, and sprouting angiogenesis in primary human en- dothelialcells.CCM1stronglyinducesDLL4-NOTCHsignaling,which

Control cells exhibited normal sprouting behavior, suggestinga cell-autonomous function of CCM1. These experiments estab-lished CCM1 as a negative regulator of angiogenesis.

CCM1 Impairs Endothelial Migration, Proliferation, and Apoptosis.Angiogenesis involves multiple cellular processes, includingendothelial cell migration and proliferation. Migration of CCM1-expressing HUVEC was significantly delayed compared with con-trol. This delay was detected in a scratch wound assay (Fig. 2A) andin the Boyden chamber using VEGF or FGF2 as chemoattractants(Fig. 2B). Conversely, CCM1 silencing enhanced endothelial mi-gration (Fig. 2 A and B), which was associated with an increasednumber of polarized focal adhesions at cellular protrusions (Fig. 2C).Silencing of CCM1 caused a moderate, nonsignificant en-

hancement of HUVEC proliferation as determined by BrdU in-corporation and cell-number counting. The apoptosis rate underbasal conditions and in response to the apoptosis-inducing agentstaurosporin was also not significantly altered (Fig. S1). Con-versely, forced CCM1 expression led to reduced cell proliferation(Fig. 2D). This finding was associated with a reduced fraction of S-phase cells, an increased G1-phase population (Fig. 2E), and el-evatedmRNA levels of the cell cycle inhibitors p21 (Waf1 orCIP1)and p27 (KIP1) (Fig. 2F). CCM1 expression significantly reducedERK phosphorylation, whereas higher phospho-ERK levels could

be detected after CCM1 silencing (Fig. 2G). These findings werecorroborated by analysis of protein lysates derived from surgicalmaterial of human CCM lesions. Compared with control braintissue, lesions from six CCM patients (15) had significantly higherlevels of phosphorylated ERK protein (Fig. 2H).CCM1 expression did not induce apoptosis under normal culture

conditions and even protected HUVEC from staurosporine-induced apoptosis (Fig. 2I). In line with the potent antiapoptoticand prosurvival functions of activeAKT (16, 17), CCM1 expressionsignificantly elevated the amount of active AKT protein phosphor-ylated at serine 473 (Fig. 2J), whereas knockdown of CCM1decreased AKT phosphorylation (Fig. 2K). Collectively, the dataestablish a role of CCM1 in shifting the balance from ERK-mediated proliferation and migration to AKT-mediated cellsurvival and endothelial quiescence.

CCM1 Regulates Sprouting Angiogenesis in Vivo.Human endothelialcells during angiogenesis can functionally be studied in vivo bygrafting them in immunocompromized mice, where they will forma network of perfused capillaries that anastomoses with the mousevasculature (18, 19). We sought to advance this assay to mimica genetic human vascular disease in mice (Fig. 3A). LentivirallyCCM1-silenced HUVEC and control shRNA transduced cellswere comparatively studied for their ability to form vascular net-

Fig. 1. CCM1 inhibits sprouting angiogenesis. (A–C) HUVEC were adenovirally transduced with CCM1 or GFP as control leading to an about 5-fold over-expression of CCM1 mRNA as determined by qPCR (A). (B) CCM1 inhibited tube formation and branching on a Matrigel matrix. (Scale bars, 2,000 μm.)(C) CCM1 inhibited VEGF (25 ng/mL) or FGF2 (25 ng/mL) stimulated sprouting angiogenesis in a collagen matrix. (Scale bar, 200 μm.) (D) Down-regulation ofCCM1 mRNA expression by two siRNAs (Ambion) led to about 80% reduction of mRNA levels. (E) Small interfering RNA-mediated CCM1 silencing stronglyenhanced endothelial sprouting in collagen gels under basal conditions and after VEGF or FGF2 stimulation. (Scale bar, 200 μm.) (F and G) Control HUVECwere labeled with the red fluorescent membrane dye PKH26 and mixed with CCM1 shRNA (F) or cDNA (G) expressing HUVEC stained with the greenmembrane dye PKH67. After sprouting in collagen beds (25 ng/mL VEGF), spheroids were imaged with confocal laser microscopy and 3D images were cal-culated. (F) CCM1 depletion caused more and larger sprouts. (Scale bar, 100 μm.) (G) CCM1-expressing HUVEC formed few regular sprouts compared withcontrol. *P < 0.05. (Scale bar, 100 μm.) Error bars are means ± SD of n = 10 in C and E.

Wüstehube et al. PNAS | July 13, 2010 | vol. 107 | no. 28 | 12641

MED

ICALSC

IENCE

S

Page 3: Cerebral cavernous malformation protein CCM1 inhibits sprouting ... · men formation, and sprouting angiogenesis in primary human en- dothelialcells.CCM1stronglyinducesDLL4-NOTCHsignaling,which

works following xenotransplantation. Twenty-eight days aftertransplantation, CCM1-silenced HUVEC had formed a signifi-cantly denser vessel network in vivo (Fig. 3C). These vessels alsohad more protrusions and had larger diameters than controlHUVEC vasculatures (Fig. 3 B and D and Movie S3 and MovieS4), thus representing hallmarks of the CCM disease.

Global Gene Expression Analysis to Uncover Molecular CCM1 Effects.Comparative microarray analyses of CCM1 expressing and controlGFP-transduced HUVEC identified 262 transcripts with at least2-fold changed expression (Fig. S2 and Table S1). The transcrip-tional changes of 12 candidates were verified by qPCR Table S2.Themajority of the regulated transcripts were involved in cell-cycleprogression, proliferation, and cell migration (Fig. 4B) and thesegenes were also highly functional connected with each other (Fig.S3 and Fig. S4).Among the regulated genes, we identified a considerable num-

ber of transcripts associated with angiogenesis (Fig. S2C). An-giogenic factors (EGR1, ESM1, ROBO4) and genes involved incell migration (CDC42EP2, ETV5,HMMR, IQGAP3) were down-regulated. In contrast, extracellular matrix molecules (COL1A2,FIBULIN2) and BMP/TGFβ related/regulated genes (ANGPTL4,BAMBI, COL1A2, ESM1, GDF3, TGFA) were up-regulated. No-

tably, CCM1 induced HEY1, a direct target of NOTCH signaling(20), which acts as a strong inhibitor of sprouting angiogenesis (21).

CCM1 Acts Upstream of the DELTA-NOTCH Pathway. In addition tothe up-regulation of HEY1, we identified a significant inductionof the NOTCH ligand DLL4 and the NOTCH target gene HEY2(Fig. 4A), which was accompanied by higher levels of cleaved,and thus activated, NOTCH protein (Fig. 4B). Active NOTCHsignaling inhibits endothelial proliferation, migration, sprouting,and branching, whereas loss-of-function causes the oppositeeffects with excessive sprouting angiogenesis (22).Silencing of CCM1 expression significantly diminished DLL4,

HEY1, andHEY2mRNA levels (Fig. 4A). The down-regulation ofthe NOTCH ligand DLL4 was between 50 and 80%, a sufficientreduction known to cause excessive angiogenesis (23–25). NOTCHactivity is tightly linked to CCM1 because the down-regulation ofCCM1 mRNA after 8 h was followed by rapid DLL4, HEY1, andHEY2 mRNA reduction. Conversely, CCM1 mRNA expression,observable after 18 h of transduction, was accompanied by rapidDLL4, HEY1, and HEY2 induction (Fig. S5).

DLL4-NOTCH Signaling Regulates AKT and ERK Phosphorylation.CCM1 increased AKT phosphorylation (Figs. 2 and 4C). AKT is

Fig. 2. CCM1 inhibits endothelial migration and proliferation. (A) Confluent HUVEC were wounded and migration was assayed. Short hairpin RNA againstCCM1 enhanced closure of the gap, whereas lentiviral CCM1 expression inhibited cell migration (arrows). Because shRNA vectors also expressed GFP, fluo-rescence pictures are shown, whereas CCM1 over expression is shown as bright field image. (Scale bars, 200 μm.) (B) Transmigration through a collagen coatedfilter (8-μm pore size) toward VEGF (25 ng/mL) or FGF2 (25 ng/mL) was inhibited by CCM1 and enhanced after CCM1 knockdown. (C) Antibody stainingagainst focal adhesion kinase (FAK) revealed a higher number and more polarized focal adhesion contacts at the cell periphery after CCM1 knockdown. (Scalebar, 50 μm.) (D–G) CCM1 inhibited endothelial proliferation as shown by reduced BrdU incorporation (D), decreased fraction of cells in S-phase as determinedby FACS (E), and up-regulation of the cell cycle inhibitors p21 and p27 mRNA (F). (G) Western blot analysis 48 h after CCM1 adenovirus transduction showedthat CCM1 reduced phosphorylation of ERK1/2 proteins, whereas siRNA treatment elevated phospho-ERK1/2 levels. (H) Protein lysates of human CCM lesionexhibit high phospho-ERK1/2 amounts. (I) Apoptosis of HUVEC under normal culture conditions and 2 h after addition of 250 nmol/L staurosporine wasmeasured by detection of caspase-3 and -7 activities with a luminescent substrate. CCM1 expression reduced the rate of staurosporine-induced cell death.(J and K) CCM1 expression increased the amount of phosphorylated AKT protein at serine 473. *P < 0.05. Error bars are means ± SD of n = 5.

12642 | www.pnas.org/cgi/doi/10.1073/pnas.1000132107 Wüstehube et al.

Page 4: Cerebral cavernous malformation protein CCM1 inhibits sprouting ... · men formation, and sprouting angiogenesis in primary human en- dothelialcells.CCM1stronglyinducesDLL4-NOTCHsignaling,which

phosphorylated after interaction with phosphatidylinositol tris-phosphates (PIP3) at the cell membrane. PIP3 is generated fromPIP2 by phosphatidylinositol-3-kinase (PI3K), and CCM proteinscan interact with PIP2 or PIP3 molecules (9, 26). The PI3Kinhibitors Wortmannin and LY294002 almost completely abol-ished CCM1-induced AKT phosphorylation (Fig. 4), implicatingthatCCM1 functions are transmitted in aPIP3-dependentmanner.Active NOTCH1 could strongly induce phosphorylation of

AKT (Fig. 4D). However, we ruled out the possibility thatCCM1-induced AKT phosphorylation was only a secondary ef-fect because of induction of NOTCH signaling. The blockage ofNOTCH with the γ-secretase inhibitor DAPT did not preventphosphorylation of AKT1 after CCM1 expression (Fig. 4C).Furthermore, CCM1 inhibited ERK phosphorylation (Figs. 2

and 4E). However, this inhibition was abolished when HUVECwere treated with DAPT, suggesting that this CCM1 effect is fullymediated through NOTCH signaling. Our assumption was sup-ported by the finding that constitutive active NOTCH1 stronglyinhibited ERK1/2 phosphorylation (Fig. 4F). Taken together,these data imply that CCM1 promotes AKT phosphorylation ina NOTCH-dependent and independent manner, and it inhibitsEKR1/2 phosphorylation indirectly through activation of theDELTA-NOTCH cascade (Fig. 4G).

Rescue of the CCM1 Phenotype. To rescue the endothelial defectscaused by CCM1 gain-of-function, cells overexpressing CCM1

were treated with the γ-secretase inhibitor DAPT to preventNOTCH activation. This treatment was sufficient to restore theresponsiveness to VEGF and caused an almost normal sproutingbehavior of HUVEC (Fig. 5 A and B).Finally, we aimed at developing a unique therapeutic strategy

to improve the treatment of CCM. Our data showed that loss ofCCM1 leads to enhanced ERK signaling and angiogenesis. Themultiple kinase inhibitor Sorafenib (Nexavar) is a clinically ap-proved anti-angiogenic drug which inhibits the Raf/ERKpathway,as well as VEGF receptor signaling, besides some other tyrosinekinases (27). Although doses smaller than 5 μM were not effec-tive, the addition of 10 μM Sorafenib could almost completelyblockVEGF-induced sprouting after CCM1-silencing inHUVEC(Fig. 5C andD). Thus, antiangiogenic therapy might be beneficialto prevent disease progression. To test this theory, we graftedCCM1-depleted HUVEC s.c. into the flanks of SCID mice. Twogroups were formed after 28 d. One group received 40 mg/kg perday Sorafenib per oral gavage, whereas the control group wasmock-treated with the solvent. The plugs were analyzed aftertreatment for 7 d. The mock-treated mice showed again the for-mation of a hyper-dense and irregular vasculature with largevessel diameters. Treatment with Sorafenib was able to stronglyameliorate the excessive vasculature. Sorafenib decreased themicrovessel density to rates comparable with untreated endo-thelial cells (Fig. 5E–G). Taking these data together, we concludethat the elucidation of CCM1-regulated signaling pathways

Fig. 3. CCM1 inhibits angiogenesis in transplanted endothelial cells. (A) Scheme of the spheroid-based angiogenesis assay. (B) Representative sections throughplugs stained against human CD34 (green) for endothelial cells. HUVEC expressing control shRNA formed a regular capillary vascular network. CCM1 silencedHUVEC formed a denser networkwith larger vessels. (Scale bar, 50 μm.) (C) Quantification ofmicrovessel density determined by the number of vessels per squaremilimeter. n = 7 plugs of control and CCM1 shRNA. (D) Thick sections (50 μm) were stained with human specific anti-CD31 to stain the endothelial network andassessed by confocal laser microscopy (LSM710; Zeiss). shRNA against CCM1 led to formation of a denser network with larger vessels compared with control.

Fig. 4. CCM1 activates DELTA-NOTCH signaling. (A) Quantitative RT-PCR showing significantly elevated mRNA levels of the NOTCH ligand DLL4 and thetarget genes HEY1 and HEY2 48 h after adenoviral CCM1 transduction. Small interfering RNA treatment against CCM1 down-regulated expression of DLL4and NOTCH target genes. (B) CCM1 expression in HUVEC increased the amounts of cleaved NOTCH receptor proteins as detected by a cleavage-specificantibody. (C) Western blotting revealed strongly increased levels of phospho-AKT after 36 h of adenoviral CCM1 transduction. The NOTCH inhibitor DAPT(25 μM) could not prevent AKT phosphorylation. Inhibition of PI3K activity by Wortmannin (2 μM) prevented AKT phosphorylation independent of CCM1expression. (D) Adenoviral expression of constitutive active NOTCH1 (intracellular domain, ICD) increased phospho-AKT levels. (E) Adenoviral CCM1 expressiondecreased ERK1/2 phosphorylation and this could fully be prevented with the NOTCH inhibitor DAPT (25 μM). (F) Active NOTCH1 strongly inhibited ERK1/2phosphorylation. (G) Proposed scheme of CCM1-mediated regulation of endothelial functions. *P < 0.05.

Wüstehube et al. PNAS | July 13, 2010 | vol. 107 | no. 28 | 12643

MED

ICALSC

IENCE

S

Page 5: Cerebral cavernous malformation protein CCM1 inhibits sprouting ... · men formation, and sprouting angiogenesis in primary human en- dothelialcells.CCM1stronglyinducesDLL4-NOTCHsignaling,which

should open previously unexplored therapeutic avenues to pre-vent progression of cavernous malformations with antiangiogenicsubstances.

DiscussionCCMs are common vascular lesions that can dynamically change innumber and size over time.Althoughmutations inCCM1 (KRIT1),CCM2 (OSM), and CCM3 (PDCD10) have been detected in themajority of cases, little is known about their cellular functions andthus about the pathogenesis of this disease (2). Here, we show thatCCM1 is a pivotal inhibitor of angiogenesis. Thus, we suggest thatuncontrolled sprouting significantly contributes to the pathogen-esis of CCM and could be therapeutically targeted in the future.We hypothesized, based on the chaotic vascular architecture and

thedynamic progression ofCCMlesions in response to localVEGF(13), that CCM lesions are originally caused by enhanced angio-genesis. Our data strongly supported this hypothesis and showedthat CCM1 is needed to keep the endothelium quiescent. CCM1mutations are typically loss-of-function mutations and recentlya two-hitmechanismwith biallelic loss of aCCM genewas proposedto explain the local development of CCM lesions (3, 4). This studysupports such a scenario. The reduction or loss of CCM1 in anendothelial cell would make it very susceptible to growth factorsto initiate angiogenesis. The decreased formation of cell junctionsandenhancedpermeability (5)would support angiogenic sprouting.Because we found that CCM1 loss-of-function also caused a loss ofappropriateDELTA-NOTCHsignaling, one could assume that thesprouting processmust be highly disorganizedwithmany additionalcapillary branches (22). Three studies have recently reported thatCCM2 is essential for cardiovascular development andmaintainingvascular integrity (28–30). Thus, it appears that the CCM proteins,which are not related but can form a complex (9), together keepadult endothelial cells quiescent.

Beside the existing CCMmousemodelCcm1+/−;Trp53+/− (31),there is a need to develop a suitable in vivoCCMmodel that allowsrapid screening of a larger set of therapeutic substances. Wemodified a spheroid-based xenotransplantation assay (18, 19) tomimic the human CCM pathology in mice. CCM1-silencedHUVEC formed a much denser vascular network with moreprotrusions and drastically enlarged microvessels. Thus, thismodel can be used to generate a human vasculature in miceshowing hallmarks of the CCMdisease within only 4 w. Themodelis also well suited for drug testing, as demonstrated with Sorafenibtreatment. The model also has the great advantage that the newlydeveloped vessels are solely formed by human endothelial cells, sothat human-specific drugs can be screened. The use of nonbrain-derived endothelial cells transplanted s.c. did not prevent theformation of CCMhallmarks, suggesting that CCM1 seems to playa pan-endothelial role. This finding is also reflected by the wide-spread cardiovascular defects in Ccm1−/− mice, and by the mani-festation of CCM lesions outside of the central nervous system (2).We could demonstrate that CCM1 acts genetically upstream of

NOTCH signaling. CCM1 robustly induced expression of theNOTCH ligand DLL4 and target genes HEY1 and HEY2. Thephenotype of our CCM1 manipulation experiments is almostidentical to the effects of endothelial NOTCH signaling inHUVEC and in mice (22), and we could successfully rescue thedefects caused by CCM1 expression with a NOTCH cleavage in-hibitor. We showed that CCM1 reduced phospho-ERK1/2 levelsbut increasedAKT phosphorylation in endothelial cells. The lattercould not be prevented by the NOTCH inhibitor DAPT, sug-gesting that CCM1 can induce AKT phosphorylation in the ab-sence of NOTCH. Because CCM proteins interact with the lipidsPIP2 and PIP3 (9, 26), we suggest that the CCM complex supportsthe phosphorylation of AKT, which is recruited by PIP3. BlockingPI3K-AKT diminished NOTCH signaling in HUVEC and this

Fig. 5. Rescue of the CCM1 phenotype. (A and B) CCM1 expression inhibited sprouting angiogenesis of HUVEC after stimulation with VEGF (25 ng/mL). TheNOTCH cleavage inhibitor DAPT (25 μM) could almost completely counteract this defect. (Scale bar in A, 200 μm.) (C and D) Sorafenib (10 μM) could blockVEGF-induced sprouting in control and CCM1 siRNA silenced HUVEC. (Scale bar in C, 200 μm.) (E–G) The spheroid-based angiogenesis assay was employed totest Sorafenib in vivo. Twenty-eight days after implantation, mice were treated with Sorafenib or solvent for 7 d. (E) Quantification of microvascular density.(F) Representative sections show a hyperdense vascular network of CCM1-silenced endothelial cells which can be reverted by Sorafenib. (Scale bar, 50 μm.)(G) Three-dimensional reconstruction of the vascular networks by confocal microscopy (LSM710, Zeiss). Sorafenib normalizes the CCM vasculature. Error barsare means ± SD of n = 3 plugs; *P < 0.05.

12644 | www.pnas.org/cgi/doi/10.1073/pnas.1000132107 Wüstehube et al.

Page 6: Cerebral cavernous malformation protein CCM1 inhibits sprouting ... · men formation, and sprouting angiogenesis in primary human en- dothelialcells.CCM1stronglyinducesDLL4-NOTCHsignaling,which

is in line with a recent report showing that AKT1 can activateNOTCH1 in melanoma cells (32). Thus, we propose that CCM1activates NOTCH by stimulation of AKT phosphorylation andcleaved NOTCH1 further stimulates AKT phosphorylation ina positive feedback loop. Additionally, we found that activeNOTCH1 could strongly diminish ERK phoshorylation. Thus,CCM1 leads to potent reduction of ERK-mediated cell pro-liferation but to an increase of AKT-driven endothelial survivalvia NOTCH signaling.We envisage that antiangiogenic therapy could be beneficial to

prevent progression of CCM lesions. Sorafenib, an approvedantiangiogenic drug, targets VEGF receptors and the Raf/ERKpathway (27). Our data suggest that ERK signaling is enhanced inthe angiogenic endothelium of CCM lesions, making Sorafenib anideal candidate for initial drug testing. Indeed, Sorafenib effi-ciently blocked angiogenic sprouting of HUVEC silenced forCCM1 expression. In vivo, Sorafenib led to a drastic regression ofCCM1-deficient blood vessels. Thus, antiangiogenic therapymightbe beneficial for the treatment of cerebral cavernous malforma-tions in the future.

Materials and MethodsPlasmids, RNAi. CCM1 cDNA was cloned into pENTR3c-IRES2-EGFP. ControlsiRNA-1andsiRNAsagainstCCM1(15469, 214883) (5)were fromAmbion.CCM1shRNAs (RHS4430-98913140, -98820292, RMM4431-98978703) were fromOpen Biosystems. RHS4430-98820292 achieved excellent knockdown.

Endothelial Migration, Proliferation, Apoptosis, Tube Formation, and Sprouting.Endothelial behavior was analyzed as described (33). Apoptosis was de-termined using the Caspase-Glo 3/7 Assay (Promega).

Spheroid-Based Transplant Assay. HUVEC were transduced with lentivirus andselected with 0.37 μg/mL Puromycin. Spheroids were s.c. injected into 6- to 8-week-old female CB17 SCID mice (Charles River) (18, 19).

Microarray Analyses. Total RNA was harvested, transcriped to biotin-labeledcRNA, and hybridized on Illumina Human Sentrix-8 BeadChips. Data analysiswas performed with Illumina BeadStudio V3.

Western Blotting and Human Lesion Samples. Proteins were separated withPAGE and blotted to nitrocellulose. Antibodies: cleaved NOTCH (Val-1744,Cell Signaling), ERK1 (K-23, Santa Cruz), phospho-ERK (E-4, Santa Cruz),phospho-AKT (Ser473) (D9E, Cell Signaling), AKT (9272, Cell Signaling) andGAPDH (6C5, Abcam). Human CCM specimens were described (15).

Statistical Analysis. Results are expressed as mean ± SD. Comparisons be-tween groups were analyzed by t test (two-sided). P values <0.05 wereconsidered as statistically significant.

For further descriptions, please see SI Materials and Methods.

ACKNOWLEDGMENTS. We thank Anja Telzerow for excellent technical assis-tance and the Genomics and Proteomics Core Facility at the DKFZ Heidelbergfor performing the microarray studies. This work was supported by Grants DFGFI-1568/1-1 from the Deutsche Forschungsgemeinschaft (to A.F.), SFB/TR23 (toA.F. and H.G.A.), and GRK880 (to A.F. and H.G.A.), and BayGene (U.F.).

1. Moriarity JL, et al. (1999) The natural history of cavernous malformations: Aprospective study of 68 patients. Neurosurgery 44:1166–1171.

2. Labauge P, Denier C, Bergametti F, Tournier-Lasserve E (2007) Genetics of cavernousangiomas. Lancet Neurol 6:237–244.

3. Akers AL, Johnson E, Steinberg GK, Zabramski JM, Marchuk DA (2009) Biallelic somaticand germline mutations in cerebral cavernous malformations (CCMs): Evidence fora two-hit mechanism of CCM pathogenesis. HumMol Genet 18:919–930.

4. Pagenstecher A, Stahl S, Sure U, Felbor U (2009) A two-hit mechanism causes cerebralcavernous malformations: Complete inactivation of CCM1, CCM2 or CCM3 in affectedendothelial cells. Hum Mol Genet 18:911–918.

5. Glading A, Han J, Stockton RA, Ginsberg MH (2007) KRIT-1/CCM1 is a Rap1 effectorthat regulates endothelial cell cell junctions. J Cell Biol 179:247–254.

6. Gunel M, et al. (2002) KRIT1, a gene mutated in cerebral cavernous malformation,encodes a microtubule-associated protein. Proc Natl Acad Sci USA 99:10677–10682.

7. Guzeloglu-Kayisli O, et al. (2004) KRIT1/cerebral cavernous malformation 1 proteinlocalizes to vascular endothelium, astrocytes, and pyramidal cells of the adult humancerebral cortex. Neurosurgery 54:943–949.

8. Zhang J, Rigamonti D, Dietz HC, Clatterbuck RE (2007) Interaction between krit1 andmalcavernin: Implications for the pathogenesis of cerebral cavernous malformations.Neurosurgery 60:353–359.

9. Hilder TL, et al. (2007) Proteomic identification of the cerebral cavernous malformationsignaling complex. J Proteome Res 6:4343–4355.

10. Zawistowski JS, Serebriiskii IG, LeeMF, Golemis EA,MarchukDA (2002) KRIT1 associationwith the integrin-binding protein ICAP-1: A new direction in the elucidation of cerebralcavernous malformations (CCM1) pathogenesis. Hum Mol Genet 11:389–396.

11. Whitehead KJ, Plummer NW, Adams JA, Marchuk DA, Li DY (2004) Ccm1 is requiredfor arterial morphogenesis: Implications for the etiology of human cavernous malfor-mations. Development 131:1437–1448.

12. Hogan BM, Bussmann J, Wolburg H, Schulte-Merker S (2008) Ccm1 cell autonomouslyregulates endothelial cellular morphogenesis and vascular tubulogenesis in zebrafish.Hum Mol Genet 17:2424–2432.

13. Jung KH, et al. (2003) Cerebral cavernousmalformations with dynamic and progressivecourse: Correlation study with vascular endothelial growth factor. Arch Neurol 60:1613–1618.

14. Korff T, Augustin HG (1998) Integration of endothelial cells in multicellular spheroidsprevents apoptosis and induces differentiation. J Cell Biol 143:1341–1352.

15. Zhu Y, et al. (2009) Involvement of PTEN promoter methylation in cerebral cavernousmalformations. Stroke 40:820–826.

16. Dudek H, et al. (1997) Regulation of neuronal survival by the serine-threonine proteinkinase Akt. Science 275:661–665.

17. Hemmings BA (1997) Akt signaling: Linking membrane events to life and deathdecisions. Science 275:628–630.

18. Alajati A, et al. (2008) Spheroid-based engineering of a human vasculature in mice.

Nat Methods 5:439–445.19. Laib AM, et al. (2009) Spheroid-based human endothelial cell microvessel formation

in vivo. Nat Protoc 4:1202–1215.20. Fischer A, Gessler M (2007) Delta-Notch—and then? Protein interactions and

proposed modes of repression by Hes and Hey bHLH factors. Nucleic Acids Res 35:

4583–4596.21. Taylor KL, Henderson AM, Hughes CC (2002) Notch activation during endothelial cell

network formation in vitro targets the basic HLH transcription factor HESR-1 and

downregulates VEGFR-2/KDR expression. Microvasc Res 64:372–383.22. Roca C, Adams RH (2007) Regulation of vascular morphogenesis by Notch signaling.

Genes Dev 21:2511–2524.23. Duarte A, et al. (2004) Dosage-sensitive requirement for mouse Dll4 in artery deve-

lopment. Genes Dev 18:2474–2478.24. Gale NW, et al. (2004) Haploinsufficiency of delta-like 4 ligand results in embryonic

lethality due to major defects in arterial and vascular development. Proc Natl Acad Sci

USA 101:15949–15954.25. Krebs LT, et al. (2004) Haploinsufficient lethality and formation of arteriovenous

malformations in Notch pathway mutants. Genes Dev 18:2469–2473.26. Béraud-Dufour S, Gautier R, Albiges-Rizo C, Chardin P, Faurobert E (2007) Krit 1

interactions with microtubules and membranes are regulated by Rap1 and integrin

cytoplasmic domain associated protein-1. FEBS J 274:5518–5532.27. Wilhelm S, et al. (2006) Discovery and development of sorafenib: A multikinase

inhibitor for treating cancer. Nat Rev Drug Discov 5:835–844.28. Boulday G, et al. (2009) Tissue-specific conditional CCM2 knockout mice establish the

essential role of endothelial CCM2 in angiogenesis: Implications for human cerebral

cavernous malformations. Dis Model Mech 2:168–177.29. Kleaveland B, et al. (2009) Regulation of cardiovascular development and integrity by

the heart of glass-cerebral cavernous malformation protein pathway. Nat Med 15:

169–176.30. Whitehead KJ, et al. (2009) The cerebral cavernous malformation signaling pathway

promotes vascular integrity via Rho GTPases. Nat Med 15:177–184.31. Plummer NW, et al. (2004) Loss of p53 sensitizes mice with a mutation in Ccm1

(KRIT1) to development of cerebral vascular malformations. Am J Pathol 165:

1509–1518.32. Bedogni B, Warneke JA, Nickoloff BJ, Giaccia AJ, Powell MB (2008) Notch1 is

an effector of Akt and hypoxia in melanoma development. J Clin Invest 118:

3660–3670.33. Nacak TG, et al. (2007) The BTB-Kelch protein KLEIP controls endothelial migration

and sprouting angiogenesis. Circ Res 100:1155–1163.

Wüstehube et al. PNAS | July 13, 2010 | vol. 107 | no. 28 | 12645

MED

ICALSC

IENCE

S


Recommended