Translational Research - Circulation...

26
857 E ndocardial fibroelastosis (EFE) refers to a pronounced, fibroelastic thickening of the ventricular endocardium. 1 The term EFE was introduced by Weinberg and Himelfarb in 1943, when EFE presented as an unexplained heart failure and common cause of death in infants and children. 1,2 Although, for unknown reasons, the primary form, which is not associ- ated with any significant structural anomaly of the heart, has become rare, EFE is still a major feature of several congenital heart diseases, most notably lesions with left heart obstruc- tions including hypoplastic left heart syndrome (HLHS). HLHS is a term to describe a spectrum of congenital heart disease, which is lethal if it is not treated. 3 Children diag- nosed with HLHS had left ventricular outflow tract obstruc- tion with various degrees of left ventricular hypoplasia. 3 To prevent the evolution of the full syndrome, balloon aortic valvotomy was introduced to re-establish flow through Translational Research © 2015 American Heart Association, Inc. Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.116.305629 Rationale: Endocardial fibroelastosis (EFE) is a unique form of fibrosis, which forms a de novo subendocardial tissue layer encapsulating the myocardium and stunting its growth, and which is typically associated with congenital heart diseases of heterogeneous origin, such as hypoplastic left heart syndrome. Relevance of EFE was only recently highlighted through the establishment of staged biventricular repair surgery in infant patients with hypoplastic left heart syndrome, where surgical removal of EFE tissue has resulted in improvement in the restrictive physiology leading to the growth of the left ventricle in parallel with somatic growth. However, pathomechanisms underlying EFE formation are still scarce, and specific therapeutic targets are not yet known. Objective: Here, we aimed to investigate the cellular origins of EFE tissue and to gain insights into the underlying molecular mechanisms to ultimately develop novel therapeutic strategies. Methods and Results: By utilizing a novel EFE model of heterotopic transplantation of hearts from newborn reporter mice and by analyzing human EFE tissue, we demonstrate for the first time that fibrogenic cells within EFE tissue originate from endocardial endothelial cells via aberrant endothelial to mesenchymal transition. We further demonstrate that such aberrant endothelial to mesenchymal transition involving endocardial endothelial cells is caused by dysregulated transforming growth factor beta/bone morphogenetic proteins signaling and that this imbalance is at least in part caused by aberrant promoter methylation and subsequent transcriptional suppression of bone morphogenetic proteins 5 and 7. Finally, we provide evidence that supplementation of exogenous recombinant bone morphogenetic proteins 7 effectively ameliorates endothelial to mesenchymal transition and experimental EFE in rats. Conclusions: In summary, our data point to aberrant endothelial to mesenchymal transition as a common denominator of infant EFE development in heterogeneous, congenital heart diseases, and to bone morphogenetic proteins 7 as an effective treatment for EFE and its restriction of heart growth. (Circ Res. 2015;116:857-866. DOI: 10.1161/CIRCRESAHA.116.305629.) Key Words: congenital heart disease endocardial fibroelastosis endocardium endothelial cell fibrosis hypoplastic left heart syndrome Original received November 13, 2014; revision received January 9, 2015; accepted January 13, 2015. In December, 2014, the average time from submission to first decision for all original research papers submitted to Circulation Research was 14.47 days. From the Department of Cardiology and Pneumology (X.X., F.A., E.M.Z.), Department of Nephrology and Rheumatology (B.T., M.Z.), University Medical Center of Göttingen, Georg-August University, Göttingen, Germany; Department of Cardiac Surgery, Boston Children’s Hospital, Harvard Medical School, MA (I.F., I.V., P.J.d N.); Division of Matrix Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (T.Z.H., R.K., E.M.Z.); Lab Genetica Molecolare, Papa Giovanni XXIII Hospital, Bergamo, Italy (M.I.); Department of Cancer Biology and the Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston (R.K.); and DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Germany (E.M.Z.). *These authors contributed equally to this article. †These authors share last authorship. The online-only Data Supplement is available with this article at http://circres.ahajournals.org/lookup/suppl/doi:10.1161/CIRCRESAHA. 116.305629/-/DC1. Correspondence to Prof Dr Elisabeth Zeisberg, Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August- University, Robert-Koch-Str 40, 37075 Göttingen, Germany. E-mail [email protected] Endocardial Fibroelastosis Is Caused by Aberrant Endothelial to Mesenchymal Transition Xingbo Xu,* Ingeborg Friehs,* Tachi Zhong Hu, Ivan Melnychenko, Björn Tampe, Fouzi Alnour, Maria Iascone, Raghu Kalluri, Michael Zeisberg, Pedro J. del Nido,† Elisabeth M. Zeisberg† by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from by guest on July 16, 2018 http://circres.ahajournals.org/ Downloaded from

Transcript of Translational Research - Circulation...

Page 1: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

857

Endocardial fibroelastosis (EFE) refers to a pronounced, fibroelastic thickening of the ventricular endocardium.1

The term EFE was introduced by Weinberg and Himelfarb in 1943, when EFE presented as an unexplained heart failure and common cause of death in infants and children.1,2 Although, for unknown reasons, the primary form, which is not associ-ated with any significant structural anomaly of the heart, has become rare, EFE is still a major feature of several congenital

heart diseases, most notably lesions with left heart obstruc-tions including hypoplastic left heart syndrome (HLHS).

HLHS is a term to describe a spectrum of congenital heart disease, which is lethal if it is not treated.3 Children diag-nosed with HLHS had left ventricular outflow tract obstruc-tion with various degrees of left ventricular hypoplasia.3 To prevent the evolution of the full syndrome, balloon aortic valvotomy was introduced to re-establish flow through

Translational Research

© 2015 American Heart Association, Inc.

Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.116.305629

Rationale: Endocardial fibroelastosis (EFE) is a unique form of fibrosis, which forms a de novo subendocardial tissue layer encapsulating the myocardium and stunting its growth, and which is typically associated with congenital heart diseases of heterogeneous origin, such as hypoplastic left heart syndrome. Relevance of EFE was only recently highlighted through the establishment of staged biventricular repair surgery in infant patients with hypoplastic left heart syndrome, where surgical removal of EFE tissue has resulted in improvement in the restrictive physiology leading to the growth of the left ventricle in parallel with somatic growth. However, pathomechanisms underlying EFE formation are still scarce, and specific therapeutic targets are not yet known.

Objective: Here, we aimed to investigate the cellular origins of EFE tissue and to gain insights into the underlying molecular mechanisms to ultimately develop novel therapeutic strategies.

Methods and Results: By utilizing a novel EFE model of heterotopic transplantation of hearts from newborn reporter mice and by analyzing human EFE tissue, we demonstrate for the first time that fibrogenic cells within EFE tissue originate from endocardial endothelial cells via aberrant endothelial to mesenchymal transition. We further demonstrate that such aberrant endothelial to mesenchymal transition involving endocardial endothelial cells is caused by dysregulated transforming growth factor beta/bone morphogenetic proteins signaling and that this imbalance is at least in part caused by aberrant promoter methylation and subsequent transcriptional suppression of bone morphogenetic proteins 5 and 7. Finally, we provide evidence that supplementation of exogenous recombinant bone morphogenetic proteins 7 effectively ameliorates endothelial to mesenchymal transition and experimental EFE in rats.

Conclusions: In summary, our data point to aberrant endothelial to mesenchymal transition as a common denominator of infant EFE development in heterogeneous, congenital heart diseases, and to bone morphogenetic proteins 7 as an effective treatment for EFE and its restriction of heart growth. (Circ Res. 2015;116:857-866. DOI: 10.1161/CIRCRESAHA.116.305629.)

Key Words: congenital heart disease ■ endocardial fibroelastosis ■ endocardium ■ endothelial cell ■ fibrosis ■ hypoplastic left heart syndrome

Original received November 13, 2014; revision received January 9, 2015; accepted January 13, 2015. In December, 2014, the average time from submission to first decision for all original research papers submitted to Circulation Research was 14.47 days.

From the Department of Cardiology and Pneumology (X.X., F.A., E.M.Z.), Department of Nephrology and Rheumatology (B.T., M.Z.), University Medical Center of Göttingen, Georg-August University, Göttingen, Germany; Department of Cardiac Surgery, Boston Children’s Hospital, Harvard Medical School, MA (I.F., I.V., P.J.d N.); Division of Matrix Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (T.Z.H., R.K., E.M.Z.); Lab Genetica Molecolare, Papa Giovanni XXIII Hospital, Bergamo, Italy (M.I.); Department of Cancer Biology and the Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston (R.K.); and DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Germany (E.M.Z.).

*These authors contributed equally to this article.†These authors share last authorship.The online-only Data Supplement is available with this article at http://circres.ahajournals.org/lookup/suppl/doi:10.1161/CIRCRESAHA.

116.305629/-/DC1. Correspondence to Prof Dr Elisabeth Zeisberg, Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August-

University, Robert-Koch-Str 40, 37075 Göttingen, Germany. E-mail [email protected]

Endocardial Fibroelastosis Is Caused by Aberrant Endothelial to Mesenchymal Transition

Xingbo Xu,* Ingeborg Friehs,* Tachi Zhong Hu, Ivan Melnychenko, Björn Tampe, Fouzi Alnour, Maria Iascone, Raghu Kalluri, Michael Zeisberg, Pedro J. del Nido,† Elisabeth M. Zeisberg†

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 2: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

858 Circulation Research February 27, 2015

the LV outflow tract, which led to renewed LV growth in some fetuses.4–6 In untreated children, as well as in nonre-sponders, a thick endocardial layer of cellular fibroelastic tissue, termed EFE forms, which restricts growth of the LV. Clinical observation indicates a direct observation between the LV growth of the fetus and the extent of EFE present at the time of the intervention on the valve.4,7 Thus, fetal intervention was combined with postnatal surgical remov-al of EFE tissue.8 Technically successful EFE removal in both previously untreated children, as well as in the fetal nonresponders to valvotomy resulted in catch-up growth of left-sided structures of the heart, restoring biventricular physiology in a subset of cases, whereas in the children, where EFE could not be removed surgically, there was lim-ited LV growth.8,9 These clinical findings underscore the likely fundamental role of EFE in HLHS. Therefore, pre-ventive measures to inhibit formation of EFE tissue would be highly desirable. However, pathomechanisms underlying EFE formation are still scarce and specific therapeutic tar-gets are not yet known. Here, we aimed to investigate the cellular origins of EFE tissue and to gain insights into the underlying molecular mechanisms.

EFE is a special type of fibrosis, which forms a de novo subendocardial tissue layer encapsulating the myocardium. It can be surgically removed without myocardial contami-nation, suggesting that it is derived from the endocardium and not from the myocardial stroma.9 Furthermore, during embryonic heart development, the endocardial endothelium plays a significant role in the development of the valves and septa. Through a process termed endothelial to mes-enchymal transition (EndMT), the endocardial endothelial cells form the (mesenchymal) endocardial cushion, which later gives rise to the valves and septum10 Thus, we hy-pothesized that fibroblasts within the EFE layer originate from the endothelial cells of the endocardium via EndMT. Here, we demonstrate that the fibroblasts within the EFE layer indeed derive from endocardial endothelial cells via EndMT. Although several of the mutations that have been linked to HLHS directly affect EndMT, previous studies de-tected no genetic mutation in the vast majority of patients. We therefore explored whether the transcription level of af-fected genes could be regulated by the epigenetic mecha-nism of promoter hypermethylation. We demonstrate here that transcriptional suppression of bone morphogenetic

proteins (BMP)7 through promoter methylation contributes to EndMT and thus EFE formation and that supplementa-tion with exogenous recombinant BMP7 impedes EFE formation.

MethodsEthics StatementThis research was approved by and conducted in accordance with the standards of the local institutional review committees of the Boston Children’s Hospital and the Beth Israel Deaconess Medical Center. All studies with human tissue were performed with the approval of the institutional review board of Boston Children’s Hospital.

All animals received humane care from the Animal Resources of Boston Children’s Hospital or the Beth Israel Deaconess Medical Center, respectively, and all protocols were reviewed and approved by the Institutional Animal Care and Use Committee at Boston Children’s Hospital or Beth Israel Deaconess Medical Center.

SubjectsEFE tissue from patients with HLHS were obtained as discarded ma-terial during open heart surgery between May 2009 and June 2010 at the Boston Children’s Hospital and snap frozen for further analysis. Left ventricular control tissue was obtained from autopsy of infants who have passed away for reasons other than congenital heart disease.

Statistical AnalysisAll qPCR data for RNA expression analysis (two or more biologi-cal replicates) were calculated using the ΔΔCT method. Student t test (GraphPad Prism 5.1) was used to obtain calculations of statisti-cal significance. For multiple parameter comparison, 1-way Anova was used with Bonferroni adjustment for the VE-Cadherin luciferase experiment.

All further methods are described in detail in the Online Data Supplement.

Results Evidence for EndMT in EFE Tissue From HLHS PatientsIn young infants with HLHS, the endocardial fibroelastic tissue forms a distinct subendocardial tissue layer with high collagen and elastin content, encapsulating the underlying myocardium (Figure 1A and 1B). This EFE layer is removed from the underlying myocardium to allow the left ventricle to grow, as part of the operative therapy during biventricular repair,11 enabling us to directly analyze fresh EFE tissue (for RNA, DNA, and microscopic analysis) without contamination from myocardial tissue. The main cell population in patients’ EFE tissue is fibroblasts. To gain insights into the possible endocardial origin of EFE fibroblasts, we performed confocal analy-sis of the EndMT marker TWIST, as well as of double immunofluo-rescence for fibroblast markers FSP1 and αSMA, and the endothelial marker CD31, and expression analysis of the EndMT marker genes SNAIL, TWIST, and SLUG.12 In 27 out of 30 patients, we detected both FSP1+/CD31+ and αSMA+/CD31+ double-positive cells and twist-positive cells. 54.4% of FSP1-positive fibroblasts and 30.3% of αSMA-positive fibroblasts also expressed CD31, suggesting that they are transitioning from endocardial cells into fibroblasts via EndMT (Figure 1C and 1D; Online Figure I). SNAIL, TWIST, and SLUG as well as mesenchymal markers αSMA and FSP1 were all signifi-cantly upregulated in EFE tissue as compared with healthy control samples, whereas endothelial markers CD31 and VE-Cadherin were downregulated (Figure 1E), suggesting that fibroblasts within EFE tissue possibly derive from the endocardium via undergoing EndMT.

BMP7 Promoter Is Hypermethylated in EFE TissueDuring normal embryonic heart development, the endocardial en-dothelial cells, which line the atrioventricular canal undergo an endothelial to mesenchymal transition to form the endocardial mes-enchymal cushion which later gives rise to the septum and mitral and tricuspid valves,10 prompting us to hypothesize that the EndMT

Nonstandard Abbreviations and Acronyms

ALK activin receptor-like kinase

BMP bone morphogenetic proteins

EFE endocardial fibroelastosis

EndMT endothelial to mesenchymal transition

HCAEC human coronary artery endothelial cells

HLHS hypoplastic left heart syndrome

LV left ventricle

MeDIP methylated DNA immunoprecipitation

PCR polymerase chain reaction

TGFβ transforming growth factor beta

YFP yellow fluorescent protein

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 3: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

Xu et al Endocardial Fibroelastosis and HLHS 859

observed in EFE formation is a reflection of the unique susceptibil-ity of the embryonic endocardial endothelium to undergo EndMT. Because EndMT during embryonic heart development is regulated to a great extent by the TGFβ/BMP signaling pathways,13–18 we hypothesized that aberrant TGFβ/BMP signaling contributes to ab-errant EndMT associated with EFE. We performed a qPCR array, which included 84 genes in the TGFβ/BMP signaling family, and we compared EFE tissue to normal neonatal left ventricular myocar-dium without heart disease (Figure 2A). Select regulated genes were then validated by real-time PCR (Figure 2B). Among the most regu-lated genes, we found BMP7, BMP5, and their signaling molecule SMAD5 downregulated (Figure 2B). In contrast, BMP2 and Noggin as well as collagen1A1 (the main constituent of cardiac matrix) were markedly upregulated in EFE tissue (Figure 2B). TGFβ1 was simi-larly significantly upregulated albeit less pronounced (Figure 2B). In addition, we tested relevant genes in the TGFβ/signaling pathway

ID1-3 (as target genes for BMP signals) as well as TGFβ3 (which were not included in the PCR array) by real-time analysis. Both ID1 and ID2 were downregulated in EFE, whereas ID3 and TGFβ3 were not significantly regulated in EFE when compared with healthy con-trol tissue (Figure 2B).

We next aimed to gain insights into the mechanisms underlying the persistent transcriptional suppression of select members of the BMP/TGF signaling family. Because BMP7, BMP5, and SMAD5 all contain CpG-rich motifs within their promoter regions, we hy-pothesized that observed persistent transcriptional suppression could be because of promoter hypermethylation. To explore whether de-creased expression of BMP7, BMP5, and SMAD5 in EFE tissue is associated with promoter hypermethylation of these genes, a com-mon epigenetic mechanism to reduce gene expression, we performed immunoprecipitation of methylated DNA, followed by PCRs for the BMP7, BMP5, and SMAD5 promoters (MeDIP assay; Figure 2C and

Figure 1. A, Macroscopic pictures of endocardial fibroelastosis (EFE) tissue lining the left ventricle of a human heart from an hypoplastic left heart syndrome (HLHS) patient at autopsy (upper panel) and resected from an HLHS patient during surgery (lower panel). B, Representative photomicrographs of HE and Masson’s Trichome stained sections of human EFE tissue obtained during surgery. C and D, Representative confocal photomicrographs of surgically removed EFE tissue from patients with HLHS (C) double stained for endothelial (CD31) and fibroblast specific markers (αSMA) or (D) single stained with EndMT transcription factor (TWIST). White arrows denote representative double positive cells; dotted areas denote the regions were magnified in the lower panel. E, Quantitative real-time polymerase chain reaction analysis of the mRNA expression levels of EndMT transcriptional factors (SNAIL, SLUG, and TWIST), mesenchymal markers (FSP-1, a-SMA) and endothelial markers (CD31, VE-Cadherin) in healthy controls and EFE tissues. Results were normalized to reference gene GAPDH. The size of the scale bars represents as noted above. Data are shown as the mean±SD of 3 experiments. ***P<0.001.

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 4: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

860 Circulation Research February 27, 2015

2D; Online Figure II). Results revealed higher BMP7 and BMP5 pro-moter methylation in EFE tissue compared with controls, whereas SMAD5 promoter methylation levels were unchanged in EFE tissue (Figure 2C and 2D; Online Figure II). To validate MeDIP results, we also performed methylation-sensitive high-resolution melting analysis (Figure 2E; Online Figure V) as well as bisulfite genomic se-quencing (Figure 2F) for the BMP7 promoter, which both confirmed significantly increased both relative, as well as absolute BMP7 pro-moter CpG methylation in EFE when compared with healthy control tissue (Figure 2E and 2F).

To test the biological impact of BMP7 promoter methylation, we cloned the BMP7 promoter into a CpG-free backbone and performed an in vitro methylation luciferase assay, demonstrat-ing that hypermethylation of BMP7 promoter leads to reduced

BMP7 expression (Figure 2G). This result suggests that the ob-served BMP7 promoter hypermethylation in EFE tissue likely causally contributes to the similar documented decrease of BMP7 expression.

BMP7 Blocks TGFβ-Induced EndMT Through Regulation of VE-Cadherin Promoter ActivityBecause analysis of EFE tissues revealed that observed EndMT is as-sociated with persistent suppression of BMP signaling and increased TGFβ signaling, we next aimed to gain further insights into the impact of BMP/TGFβ dysregulation on EndMT. Although BMP7 antagoniz-es TGFβ1-induced EndMT (Figure 3A), the molecular level of this mechanism has not yet been addressed. BMP7 and TGFβ1 each bind to distinct type II receptors, which then form complexes with specific

Figure 2. A, TGF-β/BMP qPCR array compared endocardial fibroelastosis (EFE) with healthy control heart tissues. The black arrow highlights the candidate genes which were altered in the EFE sample. B, Quantitative real-time polymerase chain reaction (PCR) analysis to validate the candidate genes selected from the TGF-β/BMP qPCR array. C and D, Methylated DNA immunoprecipitation (MeDIP) to assess BMP7 methylation in healthy and EFE hearts. C, Real-time PCR analysis quantitatively showed the methylation levels in control and EFE samples and (D) virtual gel images of BMP7 PCR products (upper panel) of immunoprecipitated 5mC DNA and the lower panel shows PCR products of input DNA as loading controls. E, BMP7 promoter methylation was determined by methylation-sensitive high-resolution melting analysis. F, BMP7 promoter methylation status of the individual CpG sites in the EFE samples and in healthy hearts by bisulfite sequencing. Open and filled circles indicate unmethylated and methylated status, respectively. G, Luciferase assay assessed promoter activity of methylated human BMP7 promoter. Around 2.1 kb (from 2271 to 161 upstream of transcriptional starting site) human BMP7 promoter fragment was cloned into a CpG-free luciferase vector (upper panel). The luciferase activity from the methylated BMP7 promoter construct was significantly reduced as compared with unmethylated control construct. Data are shown as the mean±SD of 3 experiments. n.s., no significance; **P<0.01; ***P<0.001.

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 5: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

Xu et al Endocardial Fibroelastosis and HLHS 861

type I serine-threonine kinase receptors (activin receptor-like kinase (ALK) receptors).19–22 BMP7 binds specifically to ALK3 and ALK6 receptors, whereas TGFβ1 binds to ALK2 and ALK5 receptors.19,20,22 Complex formation between BMP7 or TGFβ1 with type II and ALK type I receptors then leads to the intracellular signaling pathway me-diated by SMAD proteins. SMAD1, SMAD5, and SMAD8 transduce BMP7 action, whereas SMAD2 and SMAD3 mediate TGFβ action in epithelial cells. SMAD4 is common to both pathways (Figure 3B).

We therefore transfected endothelial cells with a VE-cadherin luciferase construct and mimicked TGFβ1 and BMP7 signaling by additionally transfecting endothelial cells with constitutively active ALK3 (to mimic BMP7) and ALK5 (to mimic TGFβ1) receptors or with SMAD3, SMAD5, or SMAD4.

Mimicry of the BMP7 signaling pathway increased the activity of full-length VE-cadherin promoter in HCAEC. In contrast, trans-fection with CA-ALK5 and SMAD3 (TGFβ pathway) resulted in a >50% decrease in VE-cadherin promoter activity (Figure 3B and 3C). Additional transfection in the same cells with CA-ALK3 and SMAD5 (BMP7 pathway) stimulated reinduction of VE-cadherin promoter activity by ≈2-fold, restoring it back to the basal level (Figure 3C).

Because Smad signaling pathways are not entirely specific for BMP7 and TGFβ, a similar cross talk by other members of the TGFβ- superfamily is possible. Nevertheless, these results provide evidence for a direct SMAD-dependent counteraction of the TGFβ pathway by BMP7 signaling, and vice versa, in cardiac endothelial cells.

To investigate potential therapeutic efficacy of exogenous recom-binant human BMP7 (rhBMP7) in HLHS, we analyzed the presence of BMP7 type 1 (ALK3) and type 2 (BMPR2) receptors in human EFE tissue by immunofluorescence labeling. Both receptors were present abundantly in all human EFE tissues. An average of 91% of fibroblasts coexpressed ALK3 and 71% BMPRII. An average of 44% of CD31-positive endothelial cells also coexpressed ALK3 and 65% BMPRII (Online Figures III and IV).

EFE in the Tie2Cre;Rosa-Stop-YFP Reporter Mouse Is Derived From EndMTTo clarify the origin of EFE tissue in the left ventricle, a rodent model was developed in which neonatal hearts were heterotopically trans-planted in the abdomen of adult animals.23 The rationale for this het-erotopic transplantation was that human EFE develops in the fetus and in association with reduced blood flow, and this surgical tech-nique allows to mimic this situation, by using immature donor hearts where intracavitary blood flow is reduced. Implantation of the heart was performed as a heterotopic infrarenal graft unloaded with aorta to the aorta and pulmonary artery to inferior vena cava anastomoses (Figure 4A and 4B). These unloaded neonatal rodent hearts develop EFE tissue resembling human EFE tissue observed in patients with HLHS (Figure 4A). We have applied this technique, which was origi-nally developed in rats to transgenic Tie2Cre;Rosa-Stop-YFP report-er mice (Figure 4C and 4D). In these mice, cells of (Tie2-positive) endothelial origin express YFP, and they continue to express YFP, even if they change their phenotype. These neonatal donor reporter mouse hearts developed massive EFE 2-weeks after surgery, simi-lar to the previously used neonatal rat hearts. Confocal analysis of the thickened endocardium revealed presence of YFP throughout the endocardium, revealing that cells within the EFE tissue are of endo-cardial origin (Figure 4C and 4D), whereas neonatal control reporter mouse hearts display only 1 thin (YFP-expressing) layer of endo-cardium (Figure 4C). Co-labeling of these hearts with the fibroblast markers FSP1 and αSMA showed that the FSP1- and αSMA-positive cells coexpressed YFP, indicating the endothelial origin of the fibro-blasts (Figure 4D).

Because Tie2 is also expressed in hematopoietic cells, we aimed to address if immune cells infiltrate into the fibroelastic endocardium (and thereby account for the YFP-labeling not derived from Tie2-positive endocardial cells but from Tie2-positive hematopoietic pro-genitor cells). We therefore obtained ubiquitous EGFP rats, where all cells are labeled green (Figure 4E, upper panel) and performed addi-tional transplantation experiments, engrafting wild-type donor hearts into ubiquitously EGFP-positive recipient rats. No EGFP-positive cells are detected in the newly formed EFE tissue of the donor heart, indicating that immune cells from the recipient do not contribute to the EFE formation (Figure 4E, lower panel).

Exogenous BMP7 Ameliorates Experimental EFEWe next sought to test the in vivo effect of rhBMP7 on inhibition of EFE. Because of the substantially bigger size of neonatal rat hearts when compared with neonatal mice, we chose to use the rat heart transplantation model for the BMP7 treatment study to decrease im-pact because of handling artifacts. Wild-type neonatal rat hearts were unloaded by heterotopic heart transplantation in wild type adult rats as previously described. Recipient rats were administered 30µg/kg rhBMP7 or vehicle buffer intraperitoneally every other day, starting on the day of surgery, with n=6 in each group. Transplanted hearts were harvested 2 weeks after surgery; sectioned from base to apex and analyzed for fibrosis with Masson’s Trichrome stains (Figure 5A through 5D; Online Figure VII). The fibrotic area was significant-ly reduced, and cardiomyocyte area was significantly increased in BMP7 administered rats when compared with vehicle-treated ani-mals (Figure 5A through 5D).

Figure 3. A, Representative confocal photomicrographs of human coronary artery endothelial cells (HCAEC) double stained for CD31 and FSP1. In nontreated HCAEC (left), the endothelial specific marker CD31 is visible (red), while there is FSP1 expression (green) detectable. On TGFβ1-treatment (middle), CD31 expression is reduced, whereas expression of FSP1 is induced. Addition of BMP7 to TGFβ1 (right) leads to loss of FSP1 and reinduction of CD31. B, Human telomerase reverse transcriptase (hTERT) immortalized HCAEC cells were transfected with VE-Cadherin luciferase reporter construct and control Renilla expression vector. To mimic TGFβ1 and BMP7 signaling, the cells were also transfected with expression vectors encoding constitutively active type 1 receptor (ALK5 or ALK3) or SMAD3, SMAD4 or SMAD5. C, Different plasmid combinations (+) or with empty control vector (-) were used to cotransfect the cells. The bar graphs display relative VE-Cadherin promoter activities presented as a ratio to Renilla control activities. ***P<0.001. The size of the scale bars represents as noted above.

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 6: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

862 Circulation Research February 27, 2015

DiscussionFibroelastosis of the endocardium is a congenital condition of unknown cause in which there occurs a diffuse thickening of the mural endocardium associated in most cases with myocar-dial hypertrophy, and leading to early death. This definition by Gowing from 1953 is still valid today.11 Because various attempts have been made to unravel its cause, but until now a definite mechanism of EFE development could not be identi-fied. Genetic predispositions have been proposed and infec-tious causes or hypoxia during fetal cardiac development.24–30

Today, most forms of EFE are associated with congenital heart disease, most notably with hypoplastic left heart syndrome (HLHS), where clinical data suggest that EFE plays a role in the pathogenesis of the disease and is functionally most rel-evant.4,9,31 Here, we provide evidence that EFE tissue derives from the endocardial endothelium involving aberrant EndMT associated with epigenetically induced BMP/TGFβ signal-ing dysregulation. We also provide evidence that such insight can be utilized to inhibit EndMT and EFE formation through supplementation of exogenous recombinant BMP7.

Figure 4. A, Representative macroscopic picture and photomicrographs of Masson’s Trichome and HE stained sections of unloaded rodent hearts after heterotopic transplantation. B, Sketch of the rodent unloaded heterotopic heart transplant model; the bold line indicates the position of the cross-sections. C, Sketch of heart cross section, dotted line rectangle indicates the area used for confocal microscopy (left). Representative confocal photomicrographs of heart sections from neonatal unloaded Tie2Cre; Rosa-Stop-YFP mice 2-weeks after heterotopic transplantation (right panel) and Tie2Cre; Rosa-Stop-YFP control hearts (middle panel). D, Representative confocal photomicrographs of sections from neonatal unloaded heterotopic transplanted Tie2Cre; Rosa-Stop-YFP mouse heart stained for mesenchymal marker FSP1 (red, upper panel) or αSMA (red, bottom panel). Endogenous YFP expression is shown in green. Dotted areas denote the region, which is shown magnified in the middle panel; white arrows denote representative double positive cells. E, Representative confocal photomicrographs of sections from loaded control UBC-EGFP rat heart and unloaded rat heart. E indicates endocardium; LV, left ventricular lumen; and M, myocardium.

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 7: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

Xu et al Endocardial Fibroelastosis and HLHS 863

Our finding that EFE tissue derives via aberrant EndMT from the endocardial endothelium is in agreement with sev-eral independent lines of research: EFE comprises a distinct tissue layer, which encapsulates the myocardium and which can be surgically removed without affecting the myocardial tissue, suggesting that it is of endocardial and not of myocar-dial stroma origin. Furthermore, clinical observations suggest that EFE development is directly associated with reduction of blood flow through the left ventricular cavity (in case of HLHS because of obstruction of the aortic valve), which further suggests that EFE formation is sensed and initiated in the endocardium and not within the myocardial compart-ment.4,9,31 During embryonic heart development the endocar-dial endothelial cells of the atrioventricular canal undergo an endothelial-mesenchymal transition to form the mesenchymal endocardial cushion, which later develops into the septum and the valves of the heart suggesting that even in later stages the endocardial endothelial cells retain a unique propensity to undergo EndMT.10,32 In this regard, previous studies demon-strated that endocardial cells generate the endothelial cells of coronary arteries,32 which may explain, why human coronary endothelial cells retain the capacity to undergo EndMT.33 A recent study, which questioned contribution, of EndMT to cardiac fibrosis also reported the presence of fibroblasts of en-docardial ancestry within myocardial fibrosis.34 In this regard, our findings of EndMT involving endocardial endothelial cells provide additional evidence that heterogeneous embryonic an-cestry of endothelial cells impacts their susceptibility to un-dergo EndMT and that the clinical context of EFE highlights the unique propensity of endocardial endothelial cells to un-dergo EndMT. The causal contribution of EndMT involving endocardial endothelial cells in the formation of EFE is also compatible with several lines of research which went alternate routes to identify underlying mechanisms: Several genetic mutations or copy number variations found in patients with HLHS, this far, although overall rare, are in genes which regu-late EndMT via different pathways (eg,Notch1, Smad3, GJA1,

NR2F2, and GATA4).26–30,35–38 Hypoxia, another possible fac-tor in the development of EFE, is also a strong inducer of EMT and EndMT in cell culture experiments.39,40 Various vi-ruses have been suspected to cause primary EFE without other associated cardiac anomaly, and frequency of primary EFE has declined in association with eradication of the mumps vi-rus through vaccination.25 Several viruses have been shown to induce EMT, which makes viruses potential inducers of aber-rant EndMT.41–43 Of note, it has been described that in patients with primary EFE, adventitial perivascular hyperelastosis and fibrosis are often found in medium-sized and small arteries of parenchymatous organs other than the heart, suggesting a generalized activation of EndMT in these patients.2

We further provide evidence that observed EndMT in EFE tissue is at least in part caused by imbalance of TGFβ/BMP signaling (TGFβ is increased and BMP signaling is impaired) and that EndMT and ultimately forming of EFE can be ame-liorated through supplementation of exogenous recombinant BMP7. Through utilization of a ligand-free cell culture sys-tem, we demonstrate that TGF and BMP signaling directly counteract each other in the regulation of EndMT. Our studies further suggest that impaired BMP signaling in the context of HLHS-associated EFE is directly linked to transcriptional suppression of BMP5 and BMP7. During mouse develop-ment, BMP5 and BMP7 are coexpressed in the heart, and they can substitute for each other. The simultaneous lack of both BMP5 and BMP7 leads to embryonic lethality with endocar-dial cushion defects, whereas the individual BMP5 or BMP7 mutants have negligible developmental defects.44 BMP2, which is not downregulated in EFE tissue, on the contrary, is necessary and sufficient to drive EndMT of endocardial cells to form the endocardial cushion.45

In the vast majority of patients with hypoplastic left heart syndrome whole-exome sequencing to date failed to detect causal mutations. Our studies demonstrate that the observed imbalance of TGFβ /BMP in EFE tissue is at least in part because of aberrant promoter hypermethylation of BMP7.

Figure 5. A, Representative photomicrographs of Masson’s Trichrome-stained sections from rat hearts, 2-weeks after heterotopic transplantation without (left) and after rhBMP7 treatment (right). B, Quantification of fibrotic and cardiomyocyte area in control and rhBMP7 treated hearts. C and D, Measurement of the endocardial fibroelastosis (EFE) area of slices located from the base to the apex of rat hearts from the left ventricle and the valvular endocardium after heterotopic transplantation. The left picture shows the anatomic position of the slices. The graph shows the measurement of the mean EFE area in mm2. Each data point reflects at least 6 measured slices. Data series of slices from hearts without rhBMP7 treatment are highlighted in red; those from slices of hearts after rhBMP7 treatment are highlighted in green (*P<0.05).

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 8: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

864 Circulation Research February 27, 2015

Thus, our approach to link HLHS to epigenetic silencing of select genes may provide an important new aspect of the mo-lecular roots of this heterogeneous congenital heart disease. Mechanisms why specific genes are subject to aberrant hyper-methylation in the context of EFE remain unclear, but to the best of our knowledge this issue has not been solved in any disease context yet.

Finally, we provide evidence that EndMT involving endo-cardial endothelial cells and formation of experimental EFE can be ameliorated through supplementation of exogenous recombinant BMP7. This is in line with previous studies from our group, which demonstrated that aberrant EndMT of vascular endothelial cells contributes to adult cardiac fibrosis and that by counteraction of TGFβ, BMP7 inhibits EndMT of adult cardiac endothelial cells and thereby ameliorates cardiac fibrosis in several mouse models of cardiac fibrosis,33 suggest-ing that EndMT of coronary artery and of endocardial endo-thelial cells is controlled by common mechanisms. Because the 2 BMP7 receptors ALK3 and BMPRII are present in a significant portion of both endothelial cells and fibroblasts in EFE tissue, it is conceivable that rhBMP7 could be effective in ameliorating EFE in patients with HLHS. Because the dif-ferent pathways of EndMT ultimately all lead to VE-cadherin suppression, and BMP7-induced SMAD5 activity is capable of reinducing VE-Cadherin expression (even outside the TGFβ pathway), we think that the ubiquitous rescue mechanism by BMP7 is most relevant, irrespective of the underlying EndMT mechanism, which may be not be same in all children with EFE. A schematic to illustrate the effect of BMP7 in different settings of known EndMT pathways is shown in Figure 6. Of note, our data do not exclude the possibility that other BMPs are equally effective in inhibiting EndMT and EFE formation, but those to our knowledge have not yet been developed for

systemic use, (while recombinant pro-BMP7 is in clinical use for compound fractures).

We are aware of a few limitations of our studies: because human endocardial endothelial cells to our knowledge have not yet been successfully cultured, we have used primary hu-man coronary artery endothelial cells (HCAEC), which were isolated from the coronary arteries from a single donor, to in-duce EndMT in vitro. Coronary artery endothelial cells (un-like aortic) originate to a significant extent from endocardial cells,32 and, at least to some degree retain similar biological potential as original endocardial cells, such as the capability to undergo EndMT on TGFβ treatment. Thus we think that HCAEC are well suited to substitute for the lack of primary endocardial cells with respect to understanding molecular mechanisms of both vascular and endocardial EndMT.

Furthermore, we induced EFE in neonatal Tie2Cre;Rosa-Stop-YFP mouse hearts and demonstrate here that in this animal model cells of the EFE tissue are of endothelial ori-gin. Because Tie2, in addition to endothelial cells, is also ex-pressed in hematopoietic progenitor cells in the bone marrow, we also induced EFE in wildtype rats transplanted in recipi-ent rats with ubiquitous EGFP expression. No EGFP-positive cells were found in the donor heart EFE tissue, ruling out the contribution of bone marrow cells to EFE formation.

No EFE develops if 2-week-old mouse hearts are used in this model of unloading, suggesting that immaturity is a nec-essary factor during EndMT in EFE formation.23 We therefore think that most EndMT of endocardial cells happens during fetal EFE development. However, we still detect a fraction of cells undergoing active EndMT (by colabeling of CD31 and mesenchymal markers or of CD31 and EndMT transcription factors such as TWIST) in EFE tissue from children with HLHS, aged between a few days until preschool, suggesting that part of EFE tissue is maintained by active EndMT even at later developmental stages, while naturally, an absolute num-ber of EndMT-derived fibroblasts cannot be provided by this technique.

Although we provide evidence that aberrant EndMT causes EFE, additional studies are needed to address how disturbed EndMT relates to abnormal valve formation and reduced ven-tricular growth in HLHS and in other congenital heart diseases associated with EFE. Because aberrant EndMT also contrib-utes to cardiac fibrosis in adult forms of heart disease, study-ing EndMT in the context of fetal EFE may well contribute valuable knowledge to unravel fibrogenesis in chronic heart disease.

AcknowledgmentsWe are strongly indebted to Kohei Miyazono (The University of Tokyo, Japan) for providing us with the SMAD3-5 as well as the constitutively active ALK3 and ALK5 plasmids and to Philippe Huber (University Joseph Fourier, Grenoble, France) for providing the VE-cadherin_pGL3 plasmid. We also would like to especially thank Michael Rehli (University of Regensburg, Germany) for shar-ing pCpGL-basic plasmids.

Sources of FundingThis study was funded in part by funds of the University Medical Center of Göttingen, the Boston Children’s Hospital, the Beth Israel Deaconess Medical Center Boston, the Harvard Catalyst Pilot Grant

Figure 6. Schematic representation of endothelial to mesenchymal transition (EndMT) regulated by different signaling pathways. Extrinsic signals stimulate endothelial cells to undergo EndMT through ligands binding to various receptors such as receptor tyrosine kinases (RTK), transforming growth factor receptor (TGFBR), and NOTCH receptor. Once getting activated, these pathways either independently trigger unknown mechanisms or work in concert through common pathway intermediates like SMAD2/3, thereby activating EndMT transcription factor, which leads to loss of endothelial cell characteristics, ie, expression of VE-Cadherin. The presence of BMP7 can activate the BMP pathway through binding to bone morphogenetic protein receptors (BMPR), thereby activating SMAD1/5/8 which can block the EndMT-inducing signals to ultimately maintain endothelial cell characteristics.

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 9: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

Xu et al Endocardial Fibroelastosis and HLHS 865

NIH-1UL1RR025758-01 (to E.M. Zeisberg and I. Melnychenko), NHLB-KO8 HL-075430 (to I. Friehs) and NIH RO1 – HL63095 (to P.J. del Nido). E.M. Zeisberg is further supported by grants SFB1002/C01 from the DFG and F/55/13 from the German Heart Foundation. M. Zeisberg is supported by DFG grant ZE523/2-1, ZE523/3-1 and the Else-Kröner Memorial Stipend 2005/59. X. Xu and B. Tampe were supported by seed funding “Research program” of the Faculty of Medicine, Georg-August-University Göttingen. Confocal micros-copy was performed at the Molecular & Optical Live Cell Imaging (MOLCI) core facility at the University Medicine Göttingen.

DisclosuresNone.

References 1. Weinberg T, Himelfarb AJ. Endocardial fibroelastosis (so-called fetal

endocarditis). A report of two cases occurring in siblings. Bull Johns Hopkins Hosp. 1943;72:299.

2. Rosahn PD. Endocardial fibroelastosis: old and new concepts. Bull N Y Acad Med. 1955;31:453–472.

3. Hickey EJ, Caldarone CA, McCrindle BW. Left ventricular hypoplasia: a spectrum of disease involving the left ventricular outflow tract, aortic valve, and aorta. J Am Coll Cardiol. 2012;59(1 Suppl):S43–S54.

4. McElhinney DB, Vogel M, Benson CB, Marshall AC, Wilkins-Haug LE, Silva V, Tworetzky W. Assessment of left ventricular endocardial fibro-elastosis in fetuses with aortic stenosis and evolving hypoplastic left heart syndrome. Am J Cardiol. 2010;106(12):1792–1797.

5. Vogel M, Wilkins-Haug LE, McElhinney DB, Marshall AC, Benson CB, Silva V, Tworetzky W. Reversible ductus arteriosus constriction due to maternal indomethacin after fetal intervention for hypoplastic left heart syndrome with intact/restrictive atrial septum. Fetal Diagn Ther. 2010;27(1):40–45.

6. Selamet Tierney ES, Wald RM, McElhinney DB, Marshall AC, Benson CB, Colan SD, Marcus EN, Marx GR, Levine JC, Wilkins-Haug L, Lock JE, Tworetzky W. Changes in left heart hemodynamics after technically successful in-utero aortic valvuloplasty. Ultrasound Obstet Gynecol. 2007;30(5):715–720.

7. McElhinney DB, Marshall AC, Wilkins-Haug LE, Brown DW, Benson CB, Silva V, Marx GR, Mizrahi-Arnaud A, Lock JE, Tworetzky W. Predictors of technical success and postnatal biventricular outcome after in utero aortic valvuloplasty for aortic stenosis with evolving hypoplastic left heart syndrome. Circulation. 2009;120(15):1482–1490.

8. Emani SM, Bacha EA, McElhinney DB, Marx GR, Tworetzky W, Pigula FA, del Nido PJ. Primary left ventricular rehabilitation is effective in maintaining two-ventricle physiology in the borderline left heart. J Thorac Cardiovasc Surg. 2009;138(6):1276–1282.

9. Emani SM, McElhinney DB, Tworetzky W, Myers PO, Schroeder B, Zurakowski D, Pigula FA, Marx GR, Lock JE, del Nido PJ. Staged left ventricular recruitment after single-ventricle palliation in patients with bor-derline left heart hypoplasia. J Am Coll Cardiol. 2012;60(19):1966–1974.

10. Eisenberg LM, Markwald RR. Molecular regulation of atrioventricular valvuloseptal morphogenesis. Circ Res. 1995;77:1–6.

11. Gowing NFC. Congenital fibroelastosis of the endocardium. J Path Bact. 1953;65:13–28.

12. Kokudo T, Suzuki Y, Yoshimatsu Y, Yamazaki T, Watabe T, Miyazono K. Snail is required for TGFbeta-induced endothelial-mesenchymal transition of embryonic stem cell-derived endothelial cells. J Cell Sci. 2008;121(Pt 20):3317–3324.

13. Liebner S, Cattelino A, Gallini R, Rudini N, Iurlaro M, Piccolo S, Dejana E. Beta-catenin is required for endothelial-mesenchymal trans-formation during heart cushion development in the mouse. J Cell Biol. 2004;166(3):359–367.

14. Nakajima Y, Yamagishi T, Hokari S, Nakamura H. Mechanisms involved in valvuloseptal endocardial cushion formation in early cardiogenesis: roles of transforming growth factor (TGF)-beta and bone morphogenetic protein (BMP). Anat Rec. 2000;258:119–127.

15. Boyer AS, Ayerinskas, II, Vincent EB, McKinney LA, Weeks DL, Runyan RB. TGFbeta2 and TGFbeta3 have separate and sequential activities dur-ing epithelial-mesenchymal cell transformation in the embryonic heart. Dev Biol. 1999;208(2):530–545.

16. Camenisch TD, Molin DG, Person A, Runyan RB, Gittenberger-de Groot AC, McDonald JA, Klewer SE. Temporal and distinct TGFbeta ligand

requirements during mouse and avian endocardial cushion morphogen-esis. Dev Biol. 2002;248:170–181.

17. Lijnen PJ, Petrov VV, Fagard RH. Induction of cardiac fibro-sis by transforming growth factor-beta(1). Mol Genet Metab. 2000;71(1–2):418–435.

18. Ramsdell AF, Moreno-Rodriguez RA, Wienecke MM, Sugi Y, Turner DK, Mjaatvedt CH, Markwald RR. Identification of an autocrine signaling pathway that amplifies induction of endocardial cushion tissue in the avian heart. Acta Anat (Basel). 1998;162:1–15.

19. Heldin CH, Miyazono K, ten Dijke P. TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature. 1997;390(6659):465–471.

20. Wrana JL. Regulation of Smad activity. Cell. 2000;100:189–192. 21. Derynck R, Zhang Y, Feng XH. Smads: transcriptional activators of TGF-

beta responses. Cell. 1998;95:737–740. 22. Massague J. How cells read TGF-beta signals. Nat Rev Mol Cell Biol.

2000;1(3):169–178. 23. Friehs I, Illigens B, Melnychenko I, Zhong-Hu T, Zeisberg E, Del

Nido PJ. An animal model of endocardial fibroelastosis. J Surg Res. 2013;182:94100.

24. Purevjav E, Varela J, Morgado M, Kearney DL, Li H, Taylor MD, Arimura T, Moncman CL, McKenna W, Murphy RT, Labeit S, Vatta M, Bowles NE, Kimura A, Boriek AM, Towbin JA. Nebulette mutations are associ-ated with dilated cardiomyopathy and endocardial fibroelastosis. J Am Coll Cardiol. 2010;56(18):1493–1502.

25. Ni J, Bowles NE, Kim YH, Demmler G, Kearney D, Bricker JT, Towbin JA. Viral infection of the myocardium in endocardial fibroelastosis. Molecular evidence for the role of mumps virus as an etiologic agent. Circulation. 1997;95:133–139.

26. Iascone M, Ciccone R, Galletti L, Marchetti D, Seddio F, Lincesso AR, Pezzoli L, Vetro A, Barachetti D, Boni L, Federici D, Soto AM, Comas JV, Ferrazzi P, Zuffardi O. Identification of de novo mutations and rare vari-ants in hypoplastic left heart syndrome. Clin Genet. 2012;81(6):542–554.

27. Fitzgerald KK, Bhat AM, Conard K, Hyland J, Pizarro C. Novel SMAD3 mutation in a patient with hypoplastic left heart syndrome with significant aortic aneurysm. Case Rep Genet. 2014;2014:591516.

28. Dasgupta C, Martinez AM, Zuppan CW, Shah MM, Bailey LL, Fletcher WH. Identification of connexin43 (alpha1) gap junction gene mutations in patients with hypoplastic left heart syndrome by denaturing gradient gel electrophoresis (DGGE). Mutat Res. 2001;479:173–186.

29. Al Turki S, Manickaraj AK, Mercer CL, et al. Rare variants in NR2F2 cause congenital heart defects in humans. Am J Hum Genet. 2014;94(4):574–585.

30. Warburton D, Ronemus M, Kline J, Jobanputra V, Williams I, Anyane-Yeboa K, Chung W, Yu L, Wong N, Awad D, Yu CY, Leotta A, Kendall J, Yamrom B, Lee YH, Wigler M, Levy D. The contribution of de novo and rare inherited copy number changes to congenital heart disease in an unselected sample of children with conotruncal defects or hypoplastic left heart disease. Hum Genet. 2014;133(1):11–27.

31. Sharland GK, Chita SK, Fagg NL, Anderson RH, Tynan M, Cook AC, Allan LD. Left ventricular dysfunction in the fetus: relation to aortic valve anomalies and endocardial fibroelastosis. Br Heart J. 1991;66:419–424.

32. Wu B, Zhang Z, Lui W, Chen X, Wang Y, Chamberlain AA, Moreno-Rodriguez RA, Markwald RR, O’Rourke BP, Sharp DJ, Zheng D, Lenz J, Baldwin HS, Chang CP, Zhou B. Endocardial cells form the coronary arteries by angiogenesis through myocardial-endocardial VEGF signaling. Cell. 2012;151(5):1083–1096.

33. Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, Gustafsson E, Chandraker A, Yuan X, Pu WT, Roberts AB, Neilson EG, Sayegh MH, Izumo S, Kalluri R. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med. 2007;13(8):952–961.

34. Bursac N. Cardiac fibroblasts in pressure overload hypertrophy: the enemy within? J Clin Invest. 2014;124(7):2850–2853.

35. Mihira H, Suzuki HI, Akatsu Y, Yoshimatsu Y, Igarashi T, Miyazono K, Watabe T. TGF-beta-induced mesenchymal transition of MS-1 endothelial cells requires Smad-dependent cooperative activation of Rho signals and MRTF-A. J Biochem. 2012;151(2):145–156.

36. Nakano Y, Oyamada M, Dai P, Nakagami T, Kinoshita S, Takamatsu T. Connexin43 knockdown accelerates wound healing but inhibits mesenchy-mal transition after corneal endothelial injury in vivo. Invest Ophthalmol Vis Sci. 2008;49(1):93–104.

37. Rivera-Feliciano J, Lee KH, Kong SW, Rajagopal S, Ma Q, Springer Z, Izumo S, Tabin CJ, Pu WT. Development of heart valves re-quires Gata4 expression in endothelial-derived cells. Development. 2006;133(18):3607–3618.

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 10: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

866 Circulation Research February 27, 2015

38. Noseda M, McLean G, Niessen K, Chang L, Pollet I, Montpetit R, Shahidi R, Dorovini-Zis K, Li L, Beckstead B, Durand RE, Hoodless PA, Karsan A. Notch activation results in phenotypic and functional changes consistent with endothelial-to-mesenchymal transformation. Circ Res. 2004;94(7):910–917.

39. Luo D, Wang J, Li J, Post M. Mouse snail is a target gene for HIF. Mol Cancer Res. 2011;9(2):234–245.

40. Misra A, Pandey C, Sze SK, Thanabalu T. Hypoxia activated EGFR sig-naling induces epithelial to mesenchymal transition (EMT). PLoS One. 2012;7(11):e49766.

41. Horikawa T, Yoshizaki T, Kondo S, Furukawa M, Kaizaki Y, Pagano JS. Epstein-Barr Virus latent membrane protein 1 induces Snail and epitheli-al-mesenchymal transition in metastatic nasopharyngeal carcinoma. Br J Cancer. 2011;104(7):1160–1167.

42. Shimamura M, Murphy-Ullrich JE, Britt WJ. Human cytomegalovirus in-duces TGF-beta1 activation in renal tubular epithelial cells after epithelial-to-mesenchymal transition. PLoS Pathog. 2010;6(11):e1001170.

43. Sides MD, Klingsberg RC, Shan B, Gordon KA, Nguyen HT, Lin Z, Takahashi T, Flemington EK, Lasky JA. The Epstein-Barr virus latent membrane protein 1 and transforming growth factor–beta1 synergistically induce epithelial–mesenchymal transition in lung epithelial cells. Am J Respir Cell Mol Biol. 2011;44(6):852–862.

44. Solloway MJ, Robertson EJ. Early embryonic lethality in Bmp5;Bmp7 double mutant mice suggests functional redundancy within the 60A sub-group. Development. 1999;126:1753–1768.

45. Rivera-Feliciano J, Tabin CJ. Bmp2 instructs cardiac progenitors to form the heart-valve-inducing field. Dev Biol. 2006;295(2):580–588.

What Is Known?

• Endocardial fibroelastosis (EFE) is a unique form of subendocardial fibrosis that complicates congenital heart disease.

• During embryonic heart development the endocardial endothelium undergoes an endothelial to mesenchymal transition (EndMT) to form the mesenchymal endocardial cushion.

What New Information Does This Article Contribute?

• Fibroblasts contributing to EFE originate from the endocardium via an aberrant EndMT.

• Hypermethylation of CpG island promoters of Bone Morphogenic Proteins (BMP)-5 and -7 cause an imbalanced growth factor microen-vironment and subsequently aberrant EndMT.

• In rodent models of EFE, administration of recombinant BMP-7 ame-liorates EndMT and the formation of EFE tissue.

Endocardial fibroelastosis (EFE) is a unique form of subendocar-dial fibrosis that complicates congenital heart disease, including

hypoplastic left heart syndrome (HLHS). While surgical removal of EFE tissue improves outcome of HLHS, preventive measures to inhibit EFE would be highly desirable. However, the mechanisms underlying EFE formation remains unclear and specific therapeu-tic targets have not been identified. During embryonic heart de-velopment the endocardial endothelium undergoes an endothelial to mesenchymal transition (EndMT) to form the (mesenchymal) endocardial cushion (which later gives rise to the valves and sep-tum reference). Hence, we hypothesized that fibroblasts within the EFE layer originate from the endothelial cells of the endocar-dium via EndMT. Here, we demonstrate that the fibroblasts within the EFE layer derive from endocardial endothelial cells via EndMT. We also report that transcriptional suppression of BMP7 through promoter methylation contributes to EndMT and EFE formation and that supplementation with exogenous recombinant BMP7 impedes EFE formation. On the basis of these findings, it might be useful to assess the utility of the intrauterine administration of BMP7 in fetuses diagnosed with HLHS.

Novelty and Significance

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 11: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

Maria Iascone, Raghu Kalluri, Michael Zeisberg, Pedro J. del Nido and Elisabeth M. ZeisbergXingbo Xu, Ingeborg Friehs, Tachi Zhong Hu, Ivan Melnychenko, Björn Tampe, Fouzi Alnour,Endocardial Fibroelastosis Is Caused by Aberrant Endothelial to Mesenchymal Transition

Print ISSN: 0009-7330. Online ISSN: 1524-4571 Copyright © 2015 American Heart Association, Inc. All rights reserved.is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231Circulation Research

doi: 10.1161/CIRCRESAHA.116.3056292015;116:857-866; originally published online January 13, 2015;Circ Res. 

http://circres.ahajournals.org/content/116/5/857World Wide Web at:

The online version of this article, along with updated information and services, is located on the

http://circres.ahajournals.org/content/suppl/2015/01/13/CIRCRESAHA.116.305629.DC1Data Supplement (unedited) at:

  http://circres.ahajournals.org//subscriptions/

is online at: Circulation Research Information about subscribing to Subscriptions: 

http://www.lww.com/reprints Information about reprints can be found online at: Reprints:

  document. Permissions and Rights Question and Answer about this process is available in the

located, click Request Permissions in the middle column of the Web page under Services. Further informationEditorial Office. Once the online version of the published article for which permission is being requested is

can be obtained via RightsLink, a service of the Copyright Clearance Center, not theCirculation Researchin Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions:

by guest on July 16, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 12: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

1

Supplemental Material

Page 13: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

2

Page 14: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

3

Page 15: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

4

Page 16: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

5

Page 17: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

6

Page 18: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

7

Page 19: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

8

Page 20: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

9

Methods in detail

Animals Model of Unloaded Heart Transplantation

Heterotopic heart transplantation was performed in C57/BL6 mice and Lewis rats as well as

ubiquitous EGFP rats (strain F344-Tg(UBC-EGFP)F455Rrrc) from the Rat Resource &

Research Center (RRRC-P40OD011062) following a method previously described1. Young

adult Lewis rats (100-120g) or C57/BL6 mice (20-25g) served as recipients and donor

hearts were obtained from neonatal rats (2-4 days of age) or Tie2Cre;Rosa-Stop-YFP mice

(7-10 days of age) respectively. Tie2Cre and Rosa-Stop-YFP mice have been described

previously. Animals were anesthetized with ketamine (40-60mg/kg i.p.)/xylazine (10mg/kg

i.p.) and isoflurane via endotracheal tube. The donor rats and mice received 300IU and

60IU heparin respectively, and the heart was explanted through a midline thoracic incision

followed by storage in cold high potassium Krebs-Henseleit solution as previously

described1. Implantation of the heart was performed as a heterotopic infra-renal graft

unloaded with aorta to aorta and pulmonary artery (PA) to inferior vena cava (IVC)

anastomoses. Animals were survived for 2 weeks. The recipient rats received either 30

µg/kg rhBMP7 or vehicle buffer i.p. every other day, starting on the day of surgery.

DNA isolation

Tissues were first lysed in DNA lysis buffer (Qiagen, Hilden, Germany) then precipitated

and isolated using DNeasy Blood & Tissue Kit (Qiagen, Hilden, Germany) followed by

manufacturer’s protocol.

Histological Analysis of Transplanted Hearts and EFE tissue

Snap frozen human EFE tissue embedded in OCT was sectioned and stained with

Hematoxylin & Eosin and Masson Trichrome staining. The same staining was applied to the

paraffin-embedded rat and mouse hearts, which were sectioned, de-paraffinized and

rehydrated with xylene and graded alcohol series prior to staining. Mouse hearts from

Tie2Cre; Rosa-Stop-YFP mice and EGFP rats were additionally equilibrated into 30%

sucrose prior to embedding to preserve endogenous YFP signal.

Fibrosis in rat hearts was quantified using the Image Pro Plus Software.

Immunofluorescence

Page 21: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

10

Frozen human EFE tissue sections were fixed with ice cold acetone for 10min at -20°C,

after 3 times washing with PBS, and then blocked with 1%BSA in PBS for 30min at room

temperature. To examine the marker gene expression in the tissue, samples were

incubated with primary antibody overnight at 4°C, washed three times in PBS and

incubated with the secondary antibody for one hour at room temperature. The primary and

secondary antibodies used in this study with dilution factors are listed below. Microscopic

images were acquired using an Olympus Confocal Microscope FV-1000 and Fluoview

program. The acquired images were processed using Photoshop CS3 software.

Antibody Product code

Dilution Company

Rabbit Anti-Human αSMA ab32575 1:100 Abcam

Mouse Anti-Human CD31 M0823 1:100 Dako

Rabbit Anti-Human Twist1 ABD29 1:100 Millipore

Rabbit Anti-Human FSP1 A5114 1:50 Dako

Goat Anti-Human BMPR-1A sc-5676 1:50 Santa Cruz

Rabbit Anti-Human BMPR-1A sc-20736 1:50 Santa Cruz

Rabbit Anti-Human BMPR2 ab106266 1:100 Abcam

Alexa Fluor 488 donkey anti rabbit A21206 1:200 Life technologies

Alexa Fluor 568 donkey anti mouse A10037 1:200 Life technologies

Alexa Fluor 488 donkey anti goat A11055 1:200 Life technologies

Alexa Fluor 555 donkey anti rabbit A31572 1:200 Life technologies

In vitro EndMT assay

HCAEC (6 x 105 cells per plate) were plated in 10cm cell culture plates and cultured for 24

hours. Then the cells were starved with endothelial cell basal medium (Genlantis) overnight.

EndMT induction was performed by 1:10 dilution of HCAEC growth medium by endothelial

basic medium (Genlantis, San Diego, USA) with 10 ng/ml TGFβ1 (R&D Systems, USA).

Medium with TGFß1 was changed every other day. BMP7 was added together with TGFß1

using 100 ng/ml for EndMT rescue.

Methylated DNA immunoprecipitation (MeDIP)

Methylated DNA was captured using Methylamp Methylated DNA capture Kit (Epigentek,

Farmingdale, USA). 1.0µg of sonicated DNA was used in each antibody coated well and

incubated at room temperature for 2 hours on a horizontal shaker. The captured DNA was

released with proteinase K at 65 °C for 1 hour. DNA was eluted from the column and

adjusted to a final volume of 100µl with nuclease-free water. For each sample, an input vial

was performed using total sonicated DNA as loading control. To visualize the

Page 22: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

11

immunoprecipitated products, PCR products were loaded on the Bioanalyzer 2100

electrophoresis system (Agilent Technologies). Electrophoresis results are shown as a

virtual gel as previously described2.

RNA extraction and quantitative real-time PCR

RNA was extracted from cells by direct lysis with Trizol regent (Ambion) and subsequent

isolation using the PureLink RNA Mini Kit (Ambion) following the manufacturers protocol.

Tissues were homogenized using the TissueLyserLT (Qiagen) and RNA was extracted

using the RNeasy Fibrous Tissue Mini Kit (Qiagen). cDNA was synthesized using the

SuperScript II Reverse Transcriptase (Life technologies) according to manufacturer’s

instructions. Quantitative Real-Time PCR was carried out using the Fast SYBR Green

Master Mix (Life Technologies) in the StepOne Plus realtime PCR system (Life

Technologies) with the real-time PCR primers (sequence listed below). Measurements were

standardized to GAPDH using delta delta Ct methods.

Gene Sequence Supplier

a-SMA F: AAGCACAGAGCAAAAGAGGAAT R: ATGTCGTCCCAGTTGGTGAT

Primer design Southampton, UK

BMP2 F: GGGCATCCTCTCCACAAAAG R: CCACGTCACTGAAGTCCAC

Primer design Southampton, UK

BMP5 F: ATCCTCGTCGCATACAGTTATC R: TGTCAGCATCATTCAGAAAGTTG

Primer design Southampton, UK

BMP7 F: CCTCCATTGCTCGCCTTG R: TATGCTGCTCATGTTTCCTAATAC

Primer design Southampton, UK

CD31 F: AAGGAACAGGAGGGAGAGTATTA R: GTATTTTGCTTCTGGGGACACT

Primer design Southampton, UK

COL1A1 F: AGACAGTGATTGAATACAAAACCA R: GGAGTTTACAGGAAGCAGACA

Primer design Southampton, UK

FSP-1 F: TCTTTCTTGGTTTGATCCTGACT R: AGTTCTGACTTGTTGAGCTTGA

Primer design Southampton, UK

GAPDH undisclosed Primer design Southampton, UK

ID1 F: CTGCTCTACGACATGAACGG R: GAAGGTCCCTGATGTAGTCGAT

Eurofins MWG Operon

ID2 F: AGTCCCGTGAGGTCCGTTAG R: AGTCGTTCATGTTGTATAGCAGG

Eurofins MWG Operon

ID3 F: GAGAGGCACTCAGCTTAGCC R: TCCTTTTGTCGTTGGAGATGAC

Eurofins MWG Operon

NOGGIN F: GCCAGCACTATCTCCACATCC R: GCAGCAGCGTCTCGTTCA

Primer design Southampton, UK

SLUG F: ACTCCGAAGCCAAATGACAA R: CTCTCTCTGTGGGTGTGTGT

Primer design Southampton, UK

SMAD5 F: TCTCCAAACAGCCCTTATCCC Eurofins MWG

Page 23: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

12

R: GCAGGAGGAGGCGTATCAG Operon

SNAIL F:GGCAATTTAACAATGTCTGAAAAGG R:GAATAGTTCTGGGAGACACATCG

Primer design Southampton, UK

TGFß1 F: CACTCCCACTCCCTCTCTC R: GTCCCCTGTGCCTTGATG

Primer design Southampton, UK

TGFß2 F: TACGCCAAGGAGGTTTACAAA R: TGAAGTAGGGTCTGTAGAAAGTG

Primer design Southampton, UK

TGFß3 F: ACTATGCCAACTTCTGCTCAG R: CAGATGCTTCAGGGTTCAGA

Primer design Southampton, UK

TWIST F: CTCAAGAGGTCGTGCCAATC R: CCCAGTATTTTTATTTCTAAAGGTGTT

Primer design Southampton, UK

VE-Cadherin F: CGTGAGCATCCAGGCAGTGGTAGC R: GAGCCGCCGCCGCAGGAAG

Eurofins MWG Operon

SYBR-based amplification of methylated BMP7 promoter

For quantification of methylated DNA amplification, 5µl of eluted DNA was used for each

PCR reaction, with a ROX passive reference dye (Bio-Rad, Hercules, USA) and diluted 2x

SYBR green Supermix (Bio-Rad, Hercules, USA) in a final volume of 25 µl for each PCR

reaction. The real-time PCR reactions were performed in a 96-well plate using the

Mx3000P qPCR system (Stratagene, Santa Clara, USA). PCR reaction was manually

terminated when the fluorescent signal increased over the threshold. The electrophoresis of

PCR products was performed using a Bioanalyzer 2100 (Agilent Technologies, Santa

Clara, USA) according to the manufacturer’s protocol. Electrophoresis results are shown as

virtual gel images. Oligonucleotide sequences for methylated BMP7, BMP5, SMAD5 are

listed below.

Gene Sequence Supplier

BMP7 F: GCAGGTCTTGGAGGTCTCTG R: CCAGTGGGTTCATTCATTCC

Eurofins MWG Operon

BMP5 F: GTCCCATGCATCACTGTTTCA R: TGCTGGGAAAGAAGAGGCTT

Eurofins MWG Operon

SMAD5 F: GGATACACACCCTTGATAGGACTG R: TCTAACTACCAGCTGATTGCAGTAGCC

Eurofins MWG Operon

TGF-β/BMP qPCR Array

A human TGFβ/BMP Signaling Pathway PCR array (SABiosciences, PAHS-035Y) was

used to analyze the expression levels of BMP signaling pathway components in healthy

neonatal left ventricular control tissue and EFE tissue. Briefly, 5 g of total RNA was used

for first-strand cDNA synthesis with the RT2 first strand kit (Qiagen, Germany). The PCR

array was carried out following the manufacturer’s instructions using the ready-to-use RT2-

qPCR master mix (RT2-SYBR® Green/Fluorescein qPCR master mix, SABiosciences,

Page 24: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

13

Germany). Twenty microliters of the experimental cocktail were added into each well

containing pre-dispensed, gene-specific primer pairs and run on a StepOne Plus realtime

PCR system (Life technologies). Data analysis was performed using the web-based

standard RT PCR array suite (SABiosciences, Germany).

Bisulfite modification and Bisulfite genomic sequencing

200 ng of DNA from each sample were treated with sodium bisulfite using the “Cells- to-

CpG Bisulfite Conversion Kit” (Life technologies, UK) according to the manufacturer's

protocol. Gene specific bisulfite sequencing analysis was performed as previously

described3. In short, for sequencing of methylated BMP7 promoter region, the DNA

samples were first bisulfite converted and amplified using primers, (forward:

AAAAGGATATAGGGATTGAGGGGTAAG, reverse:

CCGCCCTCCCCCCAACTATACCCAATAAATTC). Then the PCR product was gel-

extracted, purified and then ligated into pGEM-T Easy vector system (Promega). The

sequencing experiment was performed by the Seqlab using T7 primer. The result was

analyzed with BDPC, a webpage-based program4.

High resolution melting analysis (HRM)

Methylation sensitive high resolution melting experiments were performed as previously

described5. Briefly, PCR amplification and high resolution melting analysis were carried out

using StepOne Plus realtime PCR system (Life technologies). PCR was carried out with

MeltDoctor HRM Master Mix (Life technologies) and 1 µl of bisulphite modified DNA

template. The amplification consisted of 10 min at 95°C, followed by 40 cycles of 15 s 95°C,

1min of anneal/extension. The continuous ramp mode of melting curves was added at the

end of PCR cycles. All reactions were performed in triplicate. The melting curves were

normalized by calculation of the ‘line of best fit’ in between two normalization regions. 100%

methylated DNA was mixed in different ratios with unmethylated DNA to generate MS-HRM

standard curves with 120bp amplicon containing 5 CpG dinucleotides. The primer

sequence used in MS-HRM analysis were forward: CACGGAGTAATTTAAACAGT, reverse:

CCCAAAGACCTCCAAAACCT.

In vitro BMP7 promoter methylation and luciferase assay

For human BMP7 promoter methylation analyses, a 2.1 kb promoter fragment (2271 bp to

161 bp upstream of the BMP7 transcription start site) was cloned from BAC clone into

Page 25: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

14

BamHI and HindIII sites of pCpG-Basic vector6. The construct was in vitro methylated using

M.SssI, CpG methyltransferase (NEB) according to manufacturer’s protocol, and luciferase

activity was measured by Duo-Glo luciferase assay system (Promega) as suggested by the

company’s instruction.

Regulation of VE-Cadherin promoter activity by Smad-dependent pathways.

In order to achieve optimal transfection efficiency, we immortalized HCAEC cells with

human telomerase reverse transcriptase (hTERT) delivered by lentivirus. The immortalized

endothelial cells were transfected with Lipofectamine 2000 transfection reagent(Life

Technologies) according to the manufacture’s recommendations, with combinations of the

following plasmids: human VE-Cadherin-pGL3, Renilla expression vector

pGL4.73(Promega), Smad expression vector(pcDEF3-Flag(N)-hSmad3, pcDEF3-Flag(N)-

hSmad4, pcDEF3-Flag(N)-hSmad5), constitutively active type I receptor expression vector

pcDNA3-HASL-ALK3(QD) and pcDNA3-HASL-ALK5(TD) and empty control pcDNA3

vectors (Promega) as previously described7-9. The luciferase activity was determined by

Duo-Glo luciferase assay system (Promega) and normalized luciferase activities to Renilla

luciferase activity under the control of the CMV promoter.

Page 26: Translational Research - Circulation Researchcircres.ahajournals.org/content/circresaha/116/5/857.full.pdf · HLHS is a term to describe a spectrum of congenital heart disease, ...

15

References:

1. Friehs I, Illigens B, Melnychenko I, Zhong-Hu T, Zeisberg E, Del Nido PJ. An animal model of endocardial fibroelastosis. J Surg Res. 2012.

2. Bechtel W, McGoohan S, Zeisberg EM, Muller GA, Kalbacher H, Salant DJ, Muller CA, Kalluri R, Zeisberg M. Methylation determines fibroblast activation and fibrogenesis in the kidney. Nat Med. 2010;16(5):544-550.

3. Li Y, Tollefsbol TO. DNA methylation detection: bisulfite genomic sequencing analysis. Methods Mol Biol. 2011;791:11-21.

4. Rohde C, Zhang Y, Jurkowski TP, Stamerjohanns H, Reinhardt R, Jeltsch A. Bisulfite sequencing Data Presentation and Compilation (BDPC) web server--a useful tool for DNA methylation analysis. Nucleic Acids Res. 2008;36(5):e34.

5. Wojdacz TK, Dobrovic A. Methylation-sensitive high resolution melting (MS-HRM): a new approach for sensitive and high-throughput assessment of methylation. Nucleic Acids Res. 2007;35(6):e41.

6. Klug M, Rehli M. Functional analysis of promoter CpG methylation using a CpG-free luciferase reporter vector. Epigenetics. 2006;1(3):127-130.

7. Imamura T, Takase M, Nishihara A, Oeda E, Hanai J, Kawabata M, Miyazono K. Smad6 inhibits signalling by the TGF-beta superfamily. Nature. 1997;389(6651):622-626.

8. Zeisberg M, Hanai J, Sugimoto H, Mammoto T, Charytan D, Strutz F, Kalluri R. BMP-7 counteracts TGF-beta1-induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nat Med. 2003;9(7):964-968.

9. Miyazaki H, Watabe T, Kitamura T, Miyazono K. BMP signals inhibit proliferation and in vivo tumor growth of androgen-insensitive prostate carcinoma cells. Oncogene. 2004;23(58):9326-9335.