Skip to main content

Abstract

BACKGROUND:

Distinct endothelial cell cycle states (early G1 versus late G1) provide different “windows of opportunity” to enable the differential expression of genes that regulate venous versus arterial specification, respectively. Endothelial cell cycle control and arteriovenous identities are disrupted in vascular malformations including arteriovenous shunts, the hallmark of hereditary hemorrhagic telangiectasia (HHT). To date, the mechanistic link between endothelial cell cycle regulation and the development of arteriovenous malformations (AVMs) in HHT is not known.

METHODS:

We used BMP (bone morphogenetic protein) 9/10 blocking antibodies and endothelial-specific deletion of activin A receptor like type 1 (Alk1) to induce HHT in Fucci (fluorescent ubiquitination-based cell cycle indicator) 2 mice to assess endothelial cell cycle states in AVMs. We also assessed the therapeutic potential of inducing endothelial cell cycle G1 state in HHT to prevent AVMs by repurposing the Food and Drug Administration–approved CDK (cyclin-dependent kinase) 4/6 inhibitor (CDK4/6i) palbociclib.

RESULTS:

We found that endothelial cell cycle state and associated gene expressions are dysregulated during the pathogenesis of vascular malformations in HHT. We also showed that palbociclib treatment prevented AVM development induced by BMP9/10 inhibition and Alk1 genetic deletion. Mechanistically, endothelial cell late G1 state induced by palbociclib modulates the expression of genes regulating arteriovenous identity, endothelial cell migration, metabolism, and VEGF-A (vascular endothelial growth factor A) and BMP9 signaling that collectively contribute to the prevention of vascular malformations.

CONCLUSIONS:

This study provides new insights into molecular mechanisms leading to HHT by defining how endothelial cell cycle is dysregulated in AVMs because of BMP9/10 and Alk1 signaling deficiencies, and how restoration of endothelial cell cycle control may be used to treat AVMs in patients with HHT.

Clinical Perspective

What Is New?

Endothelial cell cycle state and cell cycle regulatory genes are dysregulated in hereditary hemorrhagic telangiectasia preclinical models and in telangiectases from patients with hereditary hemorrhagic telangiectasia.
Palbociclib, a Food and Drug Administration–approved inhibitor of CDK (cyclin-dependent kinase) 4 and CDK6 (CDK4/6i), prevents vascular malformations induced by both BMP (bone morphogenetic protein) 9/10 blocking antibodies and inducible endothelial-specific loss of activin A receptor like type 1 (Alk1; Alk1ECiKO [endothelial cell inducible knock-out]) in mice.

What Are the Clinical Implications?

The present work could pave the way for repurposing drugs targeting cell cycle control, such as CDK4/6i, to prevent or regress vascular malformations in clinical studies with patients with hereditary hemorrhagic telangiectasia.
This study opens new therapeutic strategies for a larger group of diseases with angiogenic disturbances characterized by endothelial cell hyperproliferation, such as cerebral cavernous malformations and venous malformations.
Hereditary hemorrhagic telangiectasia (HHT) is a rare autosomal-dominant vascular disease characterized by telangiectases and larger vascular malformations that affects 1 in 6000 children and adults worldwide.1 The known HHT-causing mutations affect genes encoding different components of the BMP (bone morphogenic protein) 9 and BMP10 signaling pathway, namely ENG, encoding the membrane glycoprotein endoglin in HHT type 1 and ACVRL1, encoding the membrane receptor ALK1 (activin A receptor like type 1) in HHT type 2.2,3 These mutations are detected in approximately 90% of cases submitted for molecular diagnosis.
HHT causes abnormal connections to develop between arteries and veins, known as arteriovenous malformations (AVMs). The most common organs affected by vascular malformations in patients with HHT are the nose, skin, lungs, brain, and liver. In the nose and skin, the vascular malformations are referred to as telangiectases, which are abnormal direct communications between dilated arterioles and postcapillary venules disrupting the capillary bed.2 These lesions are prone to rupture and cause uncontrolled nose bleeding that can ultimately result in chronic anemia. In the lungs, brain, intestinal tract, and liver, larger AVMs are formed and can lead to life-threatening pulmonary hemorrhage, stroke, intestinal bleeding, or liver failure.1,2 Pulmonary and brain AVMs are more common in patients with HHT type 1, whereas hepatic vascular malformations are more frequent in HHT type 2.1,3 Symptoms often begin during childhood and progress in severity. However, most patients typically do not receive a definitive HHT diagnosis until >40 years of age because they do not display any warning signs before AVMs rupture and do not receive genetic counseling or testing for HHT.
Vascular endothelial cells (ECs) lining blood vessels are the primary cells affected in HHT.3 In ECs, decreased activity of BMP9/10 signaling leads to overactivation of the proangiogenic factors VEGF-A (vascular endothelial growth factor A) and ANGPT2 (angiopoietin-2),4 triggering EC hyperproliferation, as well as alterations in their permeability and migration,5 ultimately leading to vascular malformations. Preclinical models using postnatal endothelial-specific homozygous inducible deletion of any of these genes, or pharmacological inhibition of the BMP9/10 signaling pathway by blocking antibodies, induces HHT-like vascular malformations, including excessive angiogenesis, enlarged veins, and arteriovenous shunts in the murine retinal vascularization model.5 Although aberrant endothelial proliferation has been associated with loss of arteriovenous identity and AVMs,3 the mechanistic link between EC cycle control and the development of AVMs has not been investigated, and this is the focus of our study.
Cell cycle progression is highly regulated by checkpoints that maintain cells in distinct phases of the cell cycle. Cells divide during mitosis, or M phase, and proceed to the first gap or G1 phase, where there is a checkpoint that can stall cells in early G1.6,7 A later G1-S checkpoint maintains cells in late G1. Our group recently showed that during development and in adulthood, venous ECs are predominantly in early G1, whereas arterial ECs are predominantly in late G1, providing distinct “windows of opportunity” for responses to extrinsic signals that promote changes in gene expression that enable EC specification.8 We also demonstrated that the early G1 state is essential for BMP4-induced venous genes, whereas the late G1 state is essential for TGF (transforming growth factor)-β1–induced arterial gene expression.8 Because endothelial proliferation is often dysregulated in AVMs, in conjunction with loss of endothelial identity, this study investigates whether dysregulated EC cycle control contributes to AVM development and if cell cycle modulators can be used to prevent or regress them.
Herein, we show that EC cycle state and cell cycle regulatory genes are dysregulated in preclinical models of HHT induced by BMP9/10 blocking antibodies, as well as Alk1 genetic endothelial-specific inducible deletion in the Fucci (fluorescent ubiquitination-based cell cycle indicator) 2 cell cycle reporter mice.9 Furthermore, we demonstrated that palbociclib,10 a Food and Drug Administration–approved inhibitor of CDK (cyclin-dependent kinase) 4 and CDK6 currently used in clinical trials for breast cancer treatment,11 prevents vascular malformations induced by both BMP9/10 blocking antibodies and endothelial-specific loss of Alk1, referred to as Alk1ECiKO (inducible knock-out). Mechanistically, we found that CDK4/6 inhibition-induced EC cycle arrest enables the expression of genes that collectively prevent the dysregulation of arteriovenous identity, migration, and metabolism, as well as genes regulating the VEGF-A and BMP9 signaling pathways, which are known to contribute to HHT pathogenesis.

METHODS

The data and methods supporting this study’s findings are available from the corresponding authors on request. The single cell RNA sequencing (scRNAseq) data have been deposited at the Gene Expression Omnibus database. The accession number for the data set is available on request from the corresponding authors. Expanded methods are available in the Supplemental Material.

Animals

Male and female mice were used to minimize sex-related biased results. All animal protocols and procedures were reviewed and approved by the University of Virginia animal care and use committee (protocol No. 4277) and complied with all ethical regulations. The expanded methods in the Supplement Material contain a list of all mouse strains and protocols.

Data Analysis and Statistics

All continuous variables were represented as mean±SEM. The Mann-Whitney nonparametric test for unpaired samples was used to analyze continuous variables between groups. One-way ANOVA with the Holm-Šidàk multiple comparisons test was used to compare the means of continuous variables among 3 groups. The 2-way ANOVA parametric test was used to compare 2 or more groups to determine whether there was an interactive effect between 2 independent variables (control samples versus treated samples) on a continuous dependent variable (polarity or time) (IBM SPSS Statistics). All graphs and analyses were generated using Prism 8.0 software (GraphPad).

RESULTS

Cell Cycle State and RNA Expression Are Dysregulated in HHT

To analyze EC cycle state in AVMs, we first used a preclinical mouse model of HHT that uses BMP9/10 blocking antibodies to create arteriovenous shunts.12,13 BMP9/10 blocking antibodies were injected intraperitoneally at postnatal day (P) 2 and P4 in mice expressing the Fucci2 cell cycle reporter, which enables dynamic visualization and quantification among ECs residing in the different cell cycle states (Figure 1A and 1B). For example, ECs in S/G2/M phases harbor green fluorescing nuclei, whereas EC nuclei in early G1 state are reporter-negative, and EC nuclei in late G1 state are fluorescing red. Consistent with our previous findings,8 we found that in P7 control retinas, ECs forming veins and arteries predominantly reside in the early G1 and late G1 states of the cell cycle, respectively (Figure 1C). Conversely, in anti-BMP9/10–treated pups, we found a lower proportion of retinal arterial ECs in late G1, and significantly more ECs actively cycling in S/G2/M in both arteries and veins, compared with controls. In addition, among ECs forming the capillary plexi, we detected a significantly higher proportion of actively cycling cells in S/G2/M and in early G1 and, concomitantly, significantly less in late G1, compared with control (Figure 1C). In addition, we performed 5-ethynyl-2′-deoxyuridine incorporation studies to identify proliferating ECs that are undergoing DNA synthesis. In anti-BMP9/10–treated pups, we found a significantly higher number of 5-ethynyl-2′-deoxyuridine-positive retinal ECs in arterial, venous, and capillary vessels, and in distal vascular plexi above arterial and venous vessels, compared with controls. Altogether, these results show a dysregulation of cell cycle state and hyperproliferation of all endothelial subtypes in HHT (Figure S1A through S1D).
Figure 1. Endothelial cell gene expression and cell cycle state in HHT. A, Schematic of the Fucci2 reporter where cells in early G1 are reporter-negative, late G1 cell nuclei are red, and S/G2/M cell nuclei are green. B, Timeline used for treatment with BMP9/10 blocking antibodies (BMP9/10 Abs) in Fucci2 postnatal mice. C, Representative confocal images showing flat-mounted retinas, with ECs labeled with anti-ERG and IB4, from either control or BMP9/10 Abs–treated Fucci2 postnatal day (P) 7 pups. Bar graphs are quantifications of ECs (%) in early G1, late G1, and S/G2/M phases in arteries, veins, and capillaries in control vs BMP9/10 Abs–treated animals (n=6 retinas per group). Data are mean±SEM; ***P≤0.001; **P≤0.01; *P≤0.05; ns, P>0.05. Two-way ANOVA for independent measures was used. P≤0.05 was considered statistically significant. White arrowheads, AV shunts. V, vein; A, artery. D, Z score heat map showing the differential expression of genes regulating cell cycle progression in primary retinal mouse ECs of control vs BMP9/10 Abs–treated pups (n=3 different experiments/group). E, Ki67 and hematoxylin and eosin (H&E) staining of biopsies of healthy skin in resection borders from patients with melanoma (patients A and B) and skin telangiectases from patients with HHT type 2 (patients C and D). Right, Quantifications of Ki67+-ECs (n=3 patients/group). Data are mean±SEM; Mann-Whitney U test. L, vessel lumen. Red arrows point to Ki67+ ECs. Scale bar (in A)=200 mm; (in E)=20 mm. BMP indicates bone morphogenetic protein; EC, endothelial cell; ERG, ETS-related gene; Fucci, fluorescent ubiquitination-based cell cycle indicator; HHT, hereditary hemorrhagic telangiectasia; and IB4, Isolectin-B4.
To further characterize the dysregulation of EC cycle state, we performed bulk RNA sequencing of murine primary retinal ECs isolated from P7 control and anti-BMP9/10–treated pups. Gene ontology analysis revealed that mRNA expression of gene families regulating cell cycle progression and proliferation was highly enriched in the anti-BMP9/10–treated group (Figure S1E). More specifically, analysis of differentially expressed genes in retinal ECs isolated from anti-BMP9/10–treated tissues showed upregulation of several genes that promote G1-to-S cell cycle transition such as CDK4, CDK6, and Mki67, as well as genes that regulate S/G2/M phases (Figure 1D).
Finally, EC cycle status was evaluated in human dermal telangiectases from 3 patients with HHT type 2, and control normal skin biopsies in resection borders from 3 patients with melanoma. Using immunohistochemistry staining for Ki67, a protein expressed during S/G2/M phases, we found a higher number of Ki67-positive ECs in dermal telangiectases from HHT type 2 patients compared with normal skin samples (Figure 1E), which is consistent with previous studies.14 Collectively, these results demonstrate that EC cycle is dysregulated in both preclinical and clinical HHT conditions leading to EC hyperproliferation. Our data suggest that drugs that induce EC cycle arrest could be clinically relevant for the treatment of patients with HHT to prevent or regress vascular malformations.

Palbociclib Prevents AVMs Induced by BMP9/10 Blocking Antibodies by Promoting EC Cycle Late G1 State

We found that CDK4 and CDK6 are upregulated in ECs isolated from anti-BMP9/10–treated mice (Figure 1D). Thus, to test whether the use of pharmacological cell cycle modulators could prevent the development of AVMs in a murine model of HHT, we used palbociclib, a CDK4 and CDK6 inhibitor (CDK4/6i) that blocks the transition from G1 to S state.8 Pups were injected at P2 and P4 with BMP9/10 antibodies, followed by palbociclib treatment starting at P4, then at P5 and P6 (Figure 2A). At P7, we found, as expected, that mice receiving BMP9/10 antibodies exhibited arteriovenous shunts and hyperdense retinal vascular plexi compared with control tissues (Figure 2B through 2D). However, pups receiving both BMP9/10 blocking antibodies and CDK4/6i developed fewer arteriovenous shunts and had less hyperdense retinal vascular plexi, and CDK4/6i treatment normalized retinal vascular progression and venous vessel diameters but not arterial vessel diameters (Figure 2B through 2G). It is important to note that the CDK4/6i treatment by itself did not impair retinal vascular development in palbociclib-treated P7 control pups (Figure S2A and S2B).
Figure 2. Palbociclib prevents AVMs induced by BMP9/10 blocking antibodies. A, Timeline used for dosing of C57BL/6 pups treated with anti-BMP9/10 Abs and CDK4/6i. B, Representative images showing retinal vasculature, with ECs labeled with IB4, from control, BMP9/10 Abs–treated, or BMP9/10 Abs– and CDK4/6i-treated P7 animals. Red arrowheads indicate AVMs, and red asterisks show hyperproliferative plexi. C through G, Bar graphs showing quantifications of shunt number per retina (C), vascular density (D), vascular progression (E) (d, vascular coverage length; D, retinal petal length), and venous (F) and arterial (G) vessel diameter (n=8–13 retinas/group; at least 4 mice). Data are mean±SEM; 1-way ANOVA with Holm-Šidàk multiple comparison test: **P≤0.01; *P≤0.05; ns, P>0.05. H, Retinal tissue flat mounts with anti-ERG staining (white) of ECs in early G1 (reporter-negative), late G1 (red), and S/G2/M (green) in venous, arterial, and capillary vessels of control, BMP9/10 Abs–treated, and BMP9/10 Abs– and CDK4/6i-treated P7 Fucci2 animals. I, Quantifications of the percentage of ECs in early G1, late G1, and S/G2/M phases in veins, arteries, and capillaries of control, BMP9/10 Abs–treated, and BMP9/10 Abs– and CDK4/6i-treated P7 Fucci2 animals (n=6 retinas/group). Data are mean±SEM; ***P≤0.001: **P≤0.01; *P≤0.05. Two-way ANOVA for independent measures was used. P≤0.05 was considered statistically significant. J, Z score heat map showing the differential expression of genes regulating cell cycle progression from G1/S to M phases in primary retinal mouse ECs of BMP9/10 Abs–treated vs BMP9/10 Abs– and CDK4/6i-treated pups (n=3 different bulk RNA sequencing experiments/group). Scale bars: 400 mm in B and 50 mm in H. Abs indicates antibodies; AV, atrioventricular; AVM, arteriovenous malformation; BMP, bone morphogenetic protein; CDK, cyclin-dependent kinase; EC, endothelial cell; ERG, ETS-related gene; Fucci, fluorescent ubiquitination-based cell cycle indicator; IB4, Isolectin-B4; and P, postnatal day.
We then analyzed the effects of BMP9/10 antibodies and CDK4/6i treatment on lungs, brains, and intestinal tracts, which are commonly affected by vascular malformations in HHT patients. To visualize vascular anomalies in these tissues, we performed intracardiac injections of blue latex dye in P7 control and anti-BMP9/10–treated pups, with or without CDK4/6i treatment following the dosing protocol as shown in Figure 2A. In the lungs of anti-BMP9/10–treated animals, blue latex dye injections revealed vasodilated intrapulmonary arteries of first, second, and third order and perfusion of a dense network of disorganized capillaries compared with controls (Figure S2C). Anti-BMP9/10–treated animals that received CDK4/6i treatment exhibited significantly fewer vascular anomalies and intrapulmonary microvasculature vasodilation (Figure S2D). Similarly, in the intestinal tracts, we found significant vasodilation of the small capillaries perfusing the duodenum in anti-BMP9/10–treated mice that was not prevented by CDK4/6i treatment during the dosing timeframe used herein (Figure S2E and S2F). Conversely, in the brains of anti-BMP9/10–treated animals, we did not detect defects in the cerebral vasculature, notably in the middle cerebral artery and basilar artery (Figure S2G and S2H). Taken together, our data show that treatment with BMP9/10 antibodies induces significant vascular anomalies, particularly in the lungs and intestinal tracts. It is interesting that CDK4/6i treatment shows a preventive effect only in the lungs.
Next, we evaluated the effects of CDK4/6i in mice expressing the Fucci2 cell cycle reporter following the same dosing protocol outlined in Figure 2A. BMP9/10 blocking antibody treatment caused a significantly higher proportion of actively cycling ECs in S/G2/M phases in arteries, veins, and capillaries (Figure 2H and 2I), as well as in the distal plexi (Figure S3A), compared with controls, consistent with data shown in Figure 1C. In pups treated with CDK4/6i, after BMP9/10 blocking antibody treatment, vascular malformations were absent, and ECs in all vessel types were predominantly in late G1 state (Figure 2H and 2I). Because palbociclib is a known cell cycle modulator, we assessed its effects on EC cycle state in the retinal vasculature. First, bulk RNA sequencing analysis revealed that mRNA levels of key genes promoting cell cycle progression, including Ccnd2 (cyclinD2), Cdk6, and E2f5, were highly enriched in retinal ECs isolated from anti-BMP9/10–treated pups. In contrast, mRNA levels of genes that promote cell cycle arrest were highly enriched in ECs from pups receiving BMP9/10 antibodies, followed by CDK4/6i, such as Cdkn2b (P15), Trp53 (P53), and Cdkn1a (P21) (Figure 2J). Furthermore, as previously shown,15 CDK4/6i treatment of human umbilical vein ECs (HUVECs) induced hypo-phosphorylation of Rb (retinoblastoma protein) (Figure S3B), which is known to induce cell cycle arrest by inhibition of E2F transcription factor activity15 (Figure S3C). These results suggest that palbociclib prevents ECs hyperproliferation and arteriovenous shunt formation by enabling the expression of genes that collectively promote endothelial late G1 state (as schematized in Figure S3C).
Finally, we assessed the effects of palbociclib treatment on arteriovenous identity in anti-BMP9/10–treated animals following the dosing protocol shown in Figure S4A. We found that JAG1 (Jagged1) protein expression is enriched in arterial ECs in control animals and is decreased in anti-BMP9/10–treated retinal vasculature (Figure S4B and S4C). JAG1 protein expression was increased in arterial ECs in animals that received CDK4/6i, after BMP9/10 antibody treatment, compared with animals that received only BMP9/10 antibodies (Figure S4B and S4C). These results are consistent with our previous studies showing that endothelial late G1 state is required to enable arterial specification16 and, herein, prevent arterial specification defects. In addition, retinal arteries in anti-BMP9/10–treated animals exhibited reduced smooth muscle cell (SMC) coverage, as assessed by αSMA (α smooth muscle actin)–expressing cells, compared with control tissues. It is interesting that CDK4/6i treatment prevented the loss of SMC coverage on arteries (Figure S4D and S4E). Finally, ENDOMUCIN, which is enriched in veins and capillaries in control retinas, was ectopically expressed in arteries and arteriovenous shunts in the retinal vasculature of anti-BMP9/10–treated animals compared with controls (Figure S4F and S4G). However, CDK4/6 inhibition did not prevent the arterial expression of ENDOMUCIN (Figure S4F and S4G) during the short window of time between the end of treatment and the collection of retinas for analysis. Overall, these results demonstrate that endothelial late G1 cell cycle state, induced by palbociclib treatment, prevents AVM formation and loss of arteriovenous identity in HHT.

Induced Late G1 EC Cycle State Promotes Polarized Migration

Other studies reported that defective EC migration is involved in the genesis of arteriovenous shunts in HHT mouse models.17,18 Herein, we performed gene ontology analysis of bulk RNA sequencing data from retinal ECs isolated from P7 pups treated with anti-BMP9/10, with or without CDK4/6i, as in Figure 2A. We found enriched expression of several gene families that regulate cell migration in retinal ECs isolated from anti-BMP9/10– and CDK4/6i-treated animals compared with mice that received only BMP9/10 antibody treatment (Figure 3A). Further analysis of differentially expressed genes revealed enrichment of genes, such as Slit2, Kdr, Rac1, Nrp1, and Esm1, which are known to control EC migration and angiogenesis19–21 (Figure 3B), in retinal ECs isolated from animals treated with anti-BMP9/10 and CDK4/6i.
Figure 3. Induced late G1 cell cycle arrest promotes endothelial cell polarized migration. A, Gene ontology (GO) term analysis of selected gene families modified in primary retinal mouse ECs of BMP9/10 Abs– and CDK4/6i-treated vs BMP9/10 Abs–treated pups (n=3 different experiments/group). B, Z score heat map showing the differential expression of genes regulating cell migration in primary retinal mouse ECs of BMP9/10 Abs–treated vs BMP9/10 Abs– and CDK4/6i-treated pups (n=3 different experiments/group). C, The polarity axis of each cell was defined as the angle between the direction of blood flow and the cell polarity axis, defined by a vector drawn from the center of the cell nucleus to the center of the Golgi apparatus. D, EC polarization in arteries and veins of control, BMP9/10 Abs–treated, and BMP9/10 Abs– and CDK4/6i-treated animals. EC nuclei are immunostained with anti-ERG1,2,3 and Golgi apparatus with anti-GM130; ECs are labeled with IB4 (yellow arrows, direction of blood flow; green arrowheads, EC Golgi orientation towards blood flow direction; A, artery; V, vein.) E, Angular histograms showing the distribution of polarization angles of retinal ECs in arteries and veins from control, BMP9/10 Abs–treated, and BMP9/10 Abs– and CDK4/6i-treated P6 animals (n=6–8 retinas). F, Polarity index dot plots of ECs from arteries (A) and veins (V) from control, BMP9/10 Abs–treated, and BMP9/10 Abs– and CDK4/6i-treated P6 animals (n=6–8 retinas; data are mean±SEM; **P≤0.01; ns, P>0.05). Two-way ANOVA for independent measures was used. P≤0.05 was considered statistically significant. Scale bar=200 mm (D). Abs indicates antibodies; BMP, bone morphogenetic protein; CDK, cyclin-dependent kinase; EC, endothelial cell; ERG, ETS-related gene; IB4, Isolectin-B4; and P, postnatal day.
To assess flow-mediated EC polarized migration in response to anti-BMP9/10, with or without CDK4/6i, we coimmunostained P7 retinas with anti-GM130, a peripheral membrane protein located in the cis-Golgi, and IB4 and anti-ERG (ETS-related gene) 1,2,3 to label ECs. To analyze the orientation of the Golgi toward the direction of the flow in retinal vessels, we measured the angle formed by the vector nucleus/Golgi in each EC and the predicted blood flow vector in the vessel (as depicted in Figure 3C). We also quantified the EC polarization using a polarity index22 ranging from 1 (highly polarized) to 0 (random distribution). As previously described,17,22 in control retinas, both venous and arterial ECs are oriented in the opposite direction of blood flow, with the Golgi positioned in front of the nuclei (Figure 3D through 3F). In retinas from mice treated with BMP9/10 antibodies, venous ECs exhibited a nonpolarized pattern toward blood flow (Figure 3D through 3F). However, in mice treated with anti-BMP9/10 and CDK4/6i, the polarity of retinal ECs in veins and arteries was oriented similarly to controls (Figure 3D through 3F). Altogether, these results demonstrate that late G1 state induced by CDK4/6 inhibition promotes the expression of genes regulating EC migration and prevents EC migration defects caused by BMP9/10 deficiency.

Induced Late G1 EC Cycle State Prevents Pathological Metabolic Rewiring

Previous studies revealed that specific metabolic pathways control EC function and behavior.23 For example, quiescent or proliferating ECs use different metabolic pathways for energy production. A metabolic rewiring towards glycolysis and mitochondrial pathways occurs in ECs when they are activated during both physiological and pathological angiogenesis. In addition, it has been shown that activation of specific metabolic pathways is associated with cell cycle progression.24 However, such metabolic changes in ECs during the formation of AVMs have not been investigated. RNA sequencing analysis of P7 retinal ECs isolated from control and anti-BMP9/10–treated mice revealed enriched mRNA expression of genes that regulate glycolysis, mitochondrial energy production through the tricarboxylic acid or Kreb cycle, and fatty acid signaling in response to anti-BMP9/10 treatment (Figure 4A), which is consistent with increased ECs energy production/consumption associated with AVM development. It is interesting that we found that late G1 cell cycle state induced by palbociclib treatment led to decreased mRNA expression of genes controlling glycolysis, tricarboxylic acid cycle, and fatty acid pathways in retinal ECs (Figure 4B).
Figure 4. Induced-late G1 cell cycle arrest prevents pathological metabolic rewiring in ECs. Z score heat map showing the differential expression of genes regulating metabolic pathways in primary retinal mouse ECs of control vs BMP9/10 Abs–treated (A) and BMP9/10 Abs–treated vs BMP9/10 Abs– and CDK4/6i-treated (B) pups (n=3 different experiments/group). C, Representative traces of oxygen consumption rate (OCR) measured by Seahorse assay of control vs CDK4/6i-treated HUVECs in response to oligomycin (ATP synthase inhibitor), BAM15 (mitochondrial uncoupler), and antimycin A/rotenone (mitochondrial respiratory chain complex III and I inhibitors; n=6 independent experiments; data are mean±SEM; **P≤0.01; ns, P>0.05). Two-way ANOVA for independent measures with Bonferonni correction for statistical analysis. P≤0.05 was considered statistically significant. The normality and homogeneity of variances assumptions were tested using the Shapiro-Wilk test (P>0.05) and Mauchly test of sphericity, respectively. Quantification of mitochondrial activity expressed as basal OCR (D) and respiratory capacity (E) in control vs CDK4/6i-treated HUVECs (n=6 independent experiments; data are mean±SEM; Mann-Whitney U test; ***P<0.001; **P≤0.01; *P<0.05; ns, P>0.05). Abs indicates antibodies; BMP, bone morphogenetic protein; CDK, cyclin-dependent kinase; EC, endothelial cell; and HUVEC, human umbilical vein endothelial cell,
Finally, we analyzed the effect of CDK4/6 inhibition on the metabolic activity of HUVECs using the Agilent Seahorse Assay. The mitochondrial stress test was performed measuring basal oxygen consumption rate (basal respiration), and after injection of 1 mM oligomycin, 2 mM BAM15 (respiratory capacity), 10 mM antimycin A, and 1 mM rotenone (nonmitochondrial oxygen consumption, used for normalization; Figure 4C). We found that cells pretreated with palbociclib (3 mg/mL) before analysis exhibited significantly lower mitochondrial basal respiration and respiratory capacity compared with control cells (Figure 4C through 4E). These results correlate with the inhibition of genes regulating mitochondrial tricarboxylic acid cycle induced by palbociclib in ECs isolated from anti-BMP9/10–treated mice (Figure 4B). Altogether, our data suggest that the induced late G1 state prevents the upregulation of genes responsible for the pathological metabolic rewiring associated with EC hyperproliferative state because of BMP9/10 signaling deficiency.

Palbociclib Prevents Vascular Malformations in HHT Type 2

Nearly 50% of patients with HHT harbor mutations in the Acvrl1 gene,1 which encodes for the ALK1 receptor. To validate the clinical relevance of the use of palbociclib for the treatment of HHT, we crossed Cdh5CreERT2 mice with Alk1flox/flox mice (6–8 weeks old) to generate an endothelial-specific inducible deletion of Alk1 (referred to as Alk1ECiKO), which is an established preclinical mouse model of human HHT type 2.12,25 Neonatal Alk1ECiKO pups received a single intraperitoneal injection of tamoxifen at P3 to induce Alk1 gene deletion and were then treated with palbociclib by oral gavage at P4, and their retinal vasculature was analyzed at P5 (Figure 5A). Although untreated Alk1ECiKO animals developed numerous arteriovenous shunts, those that received palbociclib exhibited a significantly lower number of AVMs (Figure 5B and 5C). Palbociclib treatment also prevented the formation of hyperdense and disorganized vascular networks at the front of the retinal vascular plexi in Alk1ECiKO animals (Figure 5B and 5D) and normalized vascular progression (Figure 5B and 5E). It is interesting that, unlike in the brains of mice treated with BMP9/10 blocking antibodies (Figure S2G and S2H), the brains of Alk1ECiKO mice (Figure 5F) exhibited significant dilations of the basilar and middle cerebral arteries (Figure 5G and 5H; Figure S5C) compared with control brains. In mice treated with a single dose of palbociclib at P4, we observed a trend toward normalization of the diameter of the basilar and middle cerebral arteries (Figure 5G and 5H). Because of the relatively short survival time after tamoxifen induction in Alk1ECiKO mice (approximately 48 hours), our study is limited to a single palbociclib administration, which represents a technical challenge in exploring additional treatments. However, the number of latex-filled veins in Alk1ECiKO brains treated with palbociclib was significantly reduced when compared with untreated animals (Figure 5I). In the lungs of Alk1ECiKO, we observed strong latex blue dye extravasation across the intrapulmonary bed. In Alk1ECiKO mice treated with palbociclib, dye extravasation was reduced (Figure S5A). Finally, as previously observed,12,26 in the intestinal tracts of P7 Alk1ECiKO animals, we noticed the presence of veins filled with blue latex, demonstrating the presence of arteriovenous shunts in the gastrointestinal tract that were absent in Alk1ECiKO animals treated with CDK4/6i (Figure S5B). These results demonstrate that palbociclib prevents the development of vascular malformations in major organs affected in patients with HHT, and more robustly affects brain AVMs induced by endothelial loss of Alk1 function.
Figure 5. Palbociclib prevents vascular malformations in HHT type 2. A, Timeline used for tamoxifen (Tx) injection and CDK4/6i treatment of Alk1ECiKO pups. B, Representative images showing retinal vasculature labeled with IB4 from control, Alk1ECiKO, and Alk1ECiKO + CDK4/6i-treated P5 animals. Bar graphs showing quantification of AV shunt number per retina (C), vascular density (D), and vascular progression (E; n=8–10 retinas/group; at least 4 mice). Data are mean±SEM; 1-way ANOVA; *P≤0.05; **P≤0.01. F, Timeline used for Tx injection and CDK4/6i treatment of Alk1ECiKO pups for blue latex dye perfusion experiments. G, Blue latex dye vascular perfusion in brains from control, Alk1ECiKO, and Alk1ECiKO + CDK4/6i-treated P7 animals. Red arrows show latex-filled veins indicating the presence of AVMs. H, Quantification of left middle cerebral artery (MCA) and basilar artery (BA) in control, Alk1ECiKO, and Alk1ECiKO + CDK4/6i-treated animals. I, Quantification of the number of latex-filled veins in control, Alk1ECiKO, and Alk1ECiKO + CDK4/6i-treated animals (n=4–6 mice/group; data are mean±SEM; 1-way ANOVA with Holm-Šidàk multiple comparison test; *P≤0.05; **P≤0.01; ***P<0.001). Scale bar=4 mm (G). Alk1 indicates activin A receptor like type 1; AV, arteriovenous; AVM, arteriovenous malformation; CDK, cyclin-dependent kinase; EC, endothelial cell; HHT, hereditary hemorrhagic telangiectasia; IB4, Isolectin-B4; and P, postnatal day.

scRNAseq Analysis of Palbociclib Effects on Endothelial Arteriovenous Identity in Alk1ECiKO Mice

To gain a deeper understanding of the molecular mechanisms underlying the effects of palbociclib on endothelial identity and cell cycle state, and vascular malformations in Alk1ECiKO mice, we used an scRNAseq approach. We isolated retinal ECs from P5 Alk1fl/fl, Alk1fl/fl treated with CDK4/6i, Alk1ECiKO, and Alk1ECiKO treated with CDK4/6i animals (Figure 6A). After retinal tissue dissociation, we used fluorescence-activated cell sorting to isolate ECs (CD31+CD45-) that were used for scRNAseq analysis. After filtering, samples were processed for single-cell barcoding and downstream mRNA library preparation and sequencing (Figure 6B). Relative mRNA expression of genes known to be associated with arterial, venous, capillary, proliferative, and tip cell identities were used to annotate the 5 EC populations (Figure 6C and 6D).
Figure 6. Single cell RNA sequencing (scRNAseq) analysis of arteriovenous identity in ECs isolated from Alk1ECiKO, with or without palbociclib treatment. A, Timeline used for tamoxifen (Tx) injection and CDK4/6i treatment of Alk1ECiKO pups. B, Schematic of protocol for single cell isolation and scRNAseq of ECs isolated from P5 Alk1fl/fl, Alk1fl/fl treated with CDK4/6i, Alk1ECiKO, and Alk1ECiKO treated with CDK4/6i. C, PHATE dimensionality reduction plot with clusters of EC subtypes. D, Dot plot of cell type–specific gene expression in the EC clusters from C. E, Arterial score of individual cells in PHATE dimensionality reduction plot in the indicated groups. F, Dot-plot analysis displaying the expression of specific genes known to be enriched in arterial ECs in the indicated groups. G, Venous score of individual cells in PHATE dimensionality reduction plot in the indicated groups. H, Dot-plot analysis displaying the expression of specific genes known to be enriched in venous ECs in the indicated groups. Alk1 indicates activin A receptor like type 1; CDK, cyclin-dependent kinase; EC, endothelial cell; FACS, fluorescence-activated cell sorting; and P, postnatal day.
To investigate the effects of CDK4/6i on arteriovenous identity in HHT, we used the PHATE method (Potential of Heat-Diffusin for Affinity-Based Trajectory Embedding). This method evaluates the relative gene expression levels among clusters within scRNAseq data sets to predict lineage relationships among the populations. As previously published by our group, we used arterial- or venous-enriched gene expression data to generate arterial and venous identity module “scores” for each EC cluster in the scRNAseq data set.8 Individual EC scores were then visualized on the PHATE plots (Figure 6E and 6G). As expected, in Alk1fl/fl control ECs, cells in the arterial or venous clusters displayed a high arterial or venous score, respectively (Figure 6E and 6G). However, in retinal ECs from Alk1ECiKO, both arterial and venous clusters had low identity scoring, consistent with a loss of arteriovenous identity in HHT conditions (Figure 6E and 6G). More specifically, the mRNA expression of genes associated with arterial identity, including Efnb2, Gja4, Bmx, Jag1, and Unc5b, were downregulated in the arterial cluster (Figure 6F), whereas the mRNA expression of genes associated with venous identity was either downregulated such as Ephb4 and Nr2f2, or upregulated such as Nrp2 and Emcn, in the venous cluster (Figure 6H). In the Alk1ECiKO animals treated with CDK4/6i, the arterial cluster presented a higher arterial score (Figure 6E) and a higher mRNA expression of all genes associated with arterial identity compared with the Alk1ECiKO group (Figure 6F). On the venous identity scoring and mRNA expression of venous genes in the venous cluster, we did not observe differences between the Alk1ECiKO and Alk1ECiKO treated with CDK4/6i groups (Figure 6G and 6H). In the Alk1fl/fl animals treated with CDK4/6i, palbociclib did not affect arterial or venous scoring or the expression of arterial and venous genes in their respective clusters compared with the Alk1fl/fl group (Figure 6E through 6H). These results demonstrate that, in the Alk1ECiKO mouse model of HHT, at a single cell level, both arterial and venous identity of ECs are profoundly dysregulated. Palbociclib treatment prevents the downregulation of arterial-enriched genes in retinal ECs, whereas the expression of venous identity genes was not different within the timeframe of the experimental dosing.

Analysis of Palbociclib Effects on EC Cycle State in Alk1ECiKO Mice

Because endothelial identities and cell cycle states are closely linked,8 we further investigated the effects of palbociclib on EC cycle state using Alk1ECiKO mice expressing the Fucci2 reporter. Alk1ECiKO-Fucci2 pups received a single intraperitoneal injection of tamoxifen at P3 to induce Alk1 gene deletion, they were then treated with palbociclib by oral gavage at P4, and their retinal vasculature was analyzed at P5 (Figure 7A). In Alk1ECiKO-Fucci2 pups, we found a significant increase in ECs actively cycling in the S/G2/M phases in arteries, veins, and capillaries compared with controls (Figure 7B and 7C), consistent with the hyperproliferative state of ECs observed in patients with HHT (Figure 1E). However, in Alk1ECiKO pups treated with CDK4/6i, the percentage of ECs residing in the S/G2/M phases was significantly lower in arteries and capillaries, concomitant with a higher proportion of ECs in late G1 state; no significant effects on venous ECs were observed (Figure 7B and 7C).
Figure 7. Cell cycle state analysis of Alk1ECiKO animals treated with palbociclib. A, Timeline used for tamoxifen (Tx) injection and CDK4/6i treatment of Alk1ECiKO pups. B, Quantifications of ECs (%) in early G1, late G1, and S/G2/M phases in veins, arteries, and capillaries of indicated groups (n=6 retinas/group; data are mean±SEM; *P≤0.05). Two-way ANOVA for independent samples was used. P≤0.05 was considered statistically significant. C, Retinal tissue flat mounts with anti-ERG staining of ECs (white) in early G1 (reporter negative), late G1 (red), and S/G2/M (green) in arterial, venous, and capillary vessels of P5 Alk1fl/fl, Alk1fl/fl treated with CDK4/6i, Alk1ECiKO, and Alk1ECiKO treated with CDK4/6i animals. Insets show IB4 staining to visualize vasculature organization (V, vein; A, artery). D, Late G1 score of individual cells in PHATE dimensionality reduction plot in the indicated groups. E, Early G1 score of individual cells in PHATE dimensionality reduction plot in the indicated groups. Scale bars=100 mm (C). Alk1 indicates activin A receptor like type 1; CDK, cyclin-dependent kinase; EC, endothelial cell; ERG, ETS-related gene; and P, postnatal day.
In addition, using previously generated cell cycle state–specific gene expression data,8 we generated and applied “early G1” and “late G1” cell cycle state scores to each EC in the scRNAseq data set and visualized the results on PHATE plots (Figure 7D and 7E). In accordance with our previous studies,8 in the Alk1fl/fl group, the arterial cluster displayed a high late G1 score (Figure 7D) and the venous cluster a high early G1 score (Figure 7E). The late G1 score was lower in the arterial cluster of the Alk1ECiKO group and was higher when Alk1ECiKO animals were treated with CDK4/6i (Figure 7D). These results are consistent with a loss of arterial identity in ECs from Alk1ECiKO animals and a prevention of arterial identity loss in ECs from Alk1ECiKO treated with CDK4/6i. Similarly to the venous scoring in other analyses (Figure 6G), the early G1 score in the venous cluster did not show any obvious differences between Alk1ECiKO animals and Alk1ECiKO animals treated with CDK4/6i (Figure 7E). Similar results were also found when assessing venous EC cycle state in Alk1ECiKO-Fucci2 mice treated with CDKi4/6i (Figure 7C).
Last, because we previously showed that the early G1 state was essential for BMP-induced venous genes and late G1 state was essential for TGF-β1–induced arterial gene expression,8 we evaluated the differential expression of key genes of the BMP and TGF-β pathways in the scRNAseq data sets. Correlating with a loss of arteriovenous identity in HHT, in Alk1ECiKO ECs, we found lower expression of genes of the TGF-β pathway, such as Tgfbr1, Tgfbr2, small mothers against decapentaplegic (Smad) 6, and Tgfb2 (Figure S6A), as well as genes of the BMP pathway, including Eng (Figure S6B), compared with the control group. In ECs isolated from Alk1ECiKO mice that received palbociclib treatment, the expression of those genes was higher (Figure S6A and S6B).

Palbociclib Inhibits VEGF-A Signaling and Promotes BMP9 Signaling

To decipher the molecular mechanism(s) underlying the effects of palbociclib-induced EC cycle arrest on the prevention of AVMs, we tested the effects of CDK4/6 inhibition on VEGF-A and BMP9-mediated signaling, which are known to be dysregulated in HHT. As expected, Western blot analysis revealed that a 15-minute VEGF-A stimulation of HUVECs leads to increased pAKT and pERK1/2 (Figure 8A and 8B). It is interesting that pretreatment with palbociclib for 24 hours before VEGF-A stimulation significantly decreased pAKT and pERK1/2 activation. In addition, palbociclib pretreatment of HUVECs enhanced SMAD1/5/8 phosphorylation in response to BMP9 (Figure 8B and 8C). Next, we treated HUVECs with Alk1 small interfering RNA to silence Alk1 and test the VEGF-A–induced response, with or without palbociclib treatment, using pAKT activation as a readout. We did not detect any differences in the pAKT activation induced by VEGF-A between Alk1-silenced HUVECs treated, or not, with palbociclib (Figure S7A and S7B). Consistent with this observation, the scRNAseq data did not show any obvious differences in the expression of genes associated with the AKT pathway between retinal ECs isolated from Alk1iECKO mice treated, or not, with palbociclib (Figure S7C).
Figure 8. Palbociclib promotes BMP9 signaling and inhibits VEGF signaling in endothelial cells. A, Western blot analysis of the phosphorylation of the indicated proteins in response to VEGF-A (1.5 nM) in control vs CDK4/6i-treated HUVECs. B, Western blot analysis of the phosphorylation of the indicated proteins in response to BMP9 (10 ng/mL) in control vs CDK4/6i-treated HUVECs. C, Quantification of phosphorylation normalized to total protein levels (n=5 independent experiments; data are mean±SEM; Mann-Whitney U test: *P<0.05). D, Z score heat map showing the differential expression of genes that regulate BMP9/10 and VEGF signaling in primary retinal mouse ECs of BMP9/10 Abs–treated vs BMP9/10 Abs– and CDK4/6i-treated pups (n=3 different bulk RNA sequencing experiments/group). E, Endoglin (green) and IB4 (white) staining of retinal flat-mounts from P5 Alk1fl/fl, Alk1ECiKO, and Alk1ECiKO treated with CDK4/6i animals (A, artery; V, vein). E, Insets represent IB4-stained retinal vasculature for each panel. F, Quantification of fluorescence intensity of endoglin staining in retinal venous vessels of the indicated animals (E; n=4 retinas per group; data are mean±SEM; 1-way ANOVA with Tukey multiple comparison test: *P<0.05; ns, P>0.05). G, Timeline used for Tx injection, and CDK4/6i and FLT1 blocking antibody treatment of Alk1ECiKO pups. Representative images showing retinal vasculature labeled with IB4 from control, Alk1ECiKO, Alk1ECiKO + CDK4/6i, and Alk1ECiKO + CDK4/6i- + FLT1 Ab–treated P5 animals (A, artery; V, vein; red arrowheads, AV shunts). H, Quantification of AV shunt number per retina (n=6 retinas/group). Data are mean±SEM; ***P≤0.001; **P≤0.01. Two-way ANOVA for independent samples was used. P≤0.05 was considered statistically significant. Scale bars: 100 mm (E) and 200 mm (G). Abs indicates antibodies; Alk1, activin A receptor like type 1; AV, arteriovenous; BMP, bone morphogenetic protein; CDK, cyclin-dependent kinase; EC, endothelial cell; FLT1, FMS-like tyrosine kinase 1; HUVEC, human umbilical vein endothelial cell; IB4, Isolectin-B4; P, postnatal day; Tx, tamoxifen; and VEGF, vascular endothelial growth factor.
With bulk RNA sequencing analysis, we further investigated the effects of palbociclib on these pathways in vivo on P7 retinal ECs from control pups or those treated with anti-BMP9/10, with or without CDK4/6i, as outlined in Figure 2A. We found enhanced mRNA expression of several genes in the BMP9/10 signaling pathway, such as Eng, but also the antiangiogenic gene FMS-like tyrosine kinase 1 (Flt1), in pups treated with CDK4/6i, after anti-BMP9/10 treatment (Figure 8D). In Alk1ECiKO mice, we confirmed that palbociclib promoted the expression of ENDOGLIN in venous ECs at both mRNA (Figure S6B) and protein (Figure 8E and 8F) levels. We also investigated the potential role of FLT1, a decoy receptor for VEFG-A, in the mechanism of action of palbociclib, by injecting intraperitoneal FLT1 blocking antibodies in Alk1ECiKO mice simultaneously with CDK4/6i treatment (Figure 8G). We found that, as expected, Alk1ECiKO mice that were treated with palbociclib developed fewer arteriovenous shunts than untreated Alk1ECiKO animals; however, palbociclib-treated Alk1ECiKO mice that also received FLT1 antibodies exhibited numerous arteriovenous shunts, comparable with untreated Alk1ECiKO mice (Figure 8G and 8H). These results demonstrate that blocking FLT1 counteracts the effects of palbociclib and emphasizes a mechanism of action inhibiting VEGF-A signaling by the decoy activity of VEGFR1/FLT1. Collectively, these results suggest that the cell cycle arrest, induced by palbociclib, enables the expression of genes and proteins in the BMP9/10 signaling pathway that help to restore this defective signaling axis in HHT and prevent overactivation of proangiogenic signaling.

DISCUSSION

The present study reveals that EC cycle states are dysregulated during the pathogenesis of vascular malformations in preclinical models of HHT, as well as in human dermal telangiectases from patients with HHT type 2. We also showed the clinical relevance of palbociclib in the prevention of AVMs in a preclinical model of HHT type 2. Mechanistically, late G1 cell cycle state induced by CDK4/6 inhibition enables the expression of genes regulating VEGF-A and BMP9 signaling, EC proliferation, migration, and metabolism that collectively contribute to the prevention of vascular malformations induced by BMP9/10 immunosuppression or endothelial-specific Alk1 gene deletion.
Several studies have shown that EC hyperproliferation and loss of arteriovenous identity are key characteristics of vascular malformations in HHT models.12,27,28 However, the mechanistic link between EC cycle dysregulation and AVM development was lacking. Our group previously reported that, during normal vascular development, EC cycle state is a critical regulator of arteriovenous identity. Herein, we show that during HHT pathogenesis, venous- and arterial-specific cell cycle states are profoundly disturbed. We found that this change in EC cycle state is associated with the dysregulation of mRNA expression of genes that regulate cell cycle progression and endothelial arteriovenous identity that are likely contributing to AVM development. It is well understood that BMP9/10 signaling deficiency leads to an overactivation of proangiogenic pathways controlled by VEGF-A,4,29 resulting in impaired EC proliferation control, migration, and permeability. However, the exact molecular mechanism(s) triggering ECs to progress through the cell cycle in response to BMP9/10 signaling deficiency was unknown. Our work uncovered that Ccnd2 and Cdk6, key genes promoting cell cycle checkpoint progression, are upregulated in HHT conditions.
In the present study, repurposing the oral drug palbociclib (CDK4/6i), currently used in combination with endocrine therapy for the treatment of metastatic breast cancer,30,31 showed relevance for the prevention of AVM formation in neonatal preclinical models of HHT. Further studies investigating the clinical relevance of palbociclib in the treatment of established AVMs in human patients are needed. Although this work focused primarily on understanding the role of EC cycle regulation in AVM formation, blood vessels are also comprised of mural cells (SMCs and pericytes) that are known to be altered in Alk1ECiKO, EngECiKO, and SMAD4ECiKO HHT mouse models.12,32 We showed that CDK4/6 inhibition normalizes SMC vessel coverage in the vascular plexi of animals treated with BMP9/10 antibodies. However, whether this is caused by direct effects of palbociclib on SMCs or indirect consequences of improved vascular remodeling because of late G1 induction in ECs needs to be determined.
To date, the therapeutic options available for patients with HHT are intended to reduce the symptoms of the disease, such as epistaxis.4 However, preclinical and clinical studies using anti–VEGF-A, anti-ANGT2 molecules and PI3K (phosphoinositide 3-kinase) inhibitors are emerging to counterbalance the proangiogenic axis overactivated in HHT and, ultimately, correct AVMs to a normal vasculature.12,33–36 In addition, instead of targeting the proangiogenic signals, current studies are now aiming to restore the defective BMP9-ALK1-SMAD signaling axis in HHT, using the mTOR signaling inhibitors, such as sirolimus, that block the PI3K signaling pathway that is overactivated in HHT1 and HHT2.14,37 Another therapeutic strategy is to promote ALK1-mediated signaling with tacrolimus, leading to beneficial clinical effects on vascular malformations related to HHT.38 In the 2 preclinical models of HHT used in this study, we demonstrated that EC cycle arrest induced by palbociclib enables the expression of Eng, probably participating in the reestablishment of the deficient BMP9 signaling pathway. However, in the context of Alk1 depletion, another study showed that Eng overexpression in Alk1ECiKO mice does not prevent AVM formation.28 Therefore, further studies are necessary to fully address the impact of palbociclib on endothelial ALK1/ENDOGLIN/SMAD signaling. Nevertheless, upregulation of the antiangiogenic gene Flt1 induced by CDK4/6 inhibition also appears to counterbalance the overactivated VEGF-A pathway observed in ECs in HHT.
Furthermore, we observed phenotypic differences in the 2 postnatal mouse models of HHT used in our studies (BMP9/10 antibodies versus Alk1ECiKO mouse). Although the genetic deletion of Alk1 (Alk1ECiKO) induces vascular malformations in various organs,12,26 we found that the pharmacological approach using BMP9/10 blocking antibodies leads to vascular anomalies only in vascular beds where active vascular remodeling occurs postnatally,39,40 such as the retina and lungs. Also, the differences could be linked to the differential expression levels of BMP9 signaling in different organs. It is known that BMP9 plays an important role in regulating biological functions and tissue homeostasis of the lungs and retinas. In 2-week-old mice, BMP9 expression is higher in the lungs than in the brain.41 This observation might explain why we found a striking phenotype in the lungs of BMP9/10 antibody-treated mice but not in the brains of these animals.
Several studies suggested that defective blood flow and altered EC migration contribute to the biogenesis of vascular malformations.17,27,42 Our group previously showed that in physiological vascular development, flow shear forces regulate EC identity by the cell cycle regulator p27.16 Here, in HHT, we demonstrated that aberrant EC cycle control leads to nonpolarized EC migration, and that induced late G1 state normalizes flow-mediated EC polarization. However, in vascular malformations, whether disrupted blood flow forces are upstream of aberrant EC cycle control and migration is still unaddressed.
Endothelial metabolic perturbations have been implicated in the pathogenesis of many cardiovascular diseases.23 Herein, we found a profound dysregulation of a large spectrum of genes involved in the regulation of endothelial metabolism in HHT. Indeed, we found that BMP9/10 immunosuppression induced an overexpression of genes involved in glycolysis, tricarboxylic acid cycle, and fatty acid metabolic pathways. This observation is consistent with the idea that a metabolic rewiring happens when ECs are overactivated (ie, hyperproliferative) under pathological conditions to adapt to their new environmental conditions, such as increased flow shear forces or hypoxia. Whether theses metabolic changes associated with vascular malformations are the consequences or causes of EC cycle state dysregulation still needs to be determined. Nevertheless, endothelial late G1 cell cycle state induced by palbociclib in BMP9/10 antibody-treated mice was found to be associated with decreased expression of metabolic genes in ECs correlated with a normalization of the vasculature, suggesting that cell cycle state modulation might be upstream of metabolic rewiring.
In conclusion, this study provides new insights into molecular mechanisms leading to HHT by defining how EC cycle is dysregulated in AVMs because of BMP9/10 and Alk1 signaling deficiencies. It also shows that cell cycle modulators, such as palbociclib, may represent new options for the treatment of vascular malformations in patients with HHT. Because dysregulation of EC cycle control is certainly not restricted to HHT, the present work also opens new therapeutic strategies for a larger group of diseases characterized by EC hyperproliferation and loss of identity, such as cerebral cavernous malformations and venous malformations.

ARTICLE INFORMATION

Supplemental Material

Expanded Methods
Figures S1–S7
References 43–51

Acknowledgments

We thank Drs Mariona Graupera and Claudio Franco for their scientific advice. Author contributions: G.G., N.G., S.C., A.C., V.S., A.F., P.C., E.A.N., and A.R-M. performed experiments. U.P. and N.W.C. analyzed RNA sequencing data. S.M., D.S., and M.H. maintained mouse lines and performed mouse genotyping. G.G., N.G., and K.K.H. designed experiments, analyzed data, and wrote the article.

Footnote

Nonstandard Abbreviations and Acronyms

αSMA
α smooth muscle actin
ALK1
activin A receptor like type 1
ANGPT2
angiopoietin-2
AVM
arteriovenous malformation
BMP
bone morphogenetic protein
Ccnd2
cyclinD2
CDK
cyclin-dependent kinase
CDK4/6i
cyclin-dependent kinase 4/6 inhibitor
EC
endothelial cell
ENG
endoglin
ERG
ETS-related gene
FLT1
FMS-like tyrosine kinase 1
FUCCI
fluorescent ubiquitination-based cell cycle indicator
HHT
hereditary hemorrhagic telangiectasia
HUVEC
human umbilical vascular endothelial cell
JAG1
Jagged1
P
postnatal day
PI3K
phosphoinositide 3-kinase
Rb
retinoblastoma protein
scRNAseq
single-cell RNA sequencing
SMAD
small mothers against decapentaplegic
SMC
smooth muscle cell
TGF
transforming growth factor
VEGF-A
vascular endothelial growth factor A

Supplemental Material

File (circ_circulationaha-2022-062952_supp1.pdf)
File (circ_circulationaha-2022-062952_supp10.docx)
File (circ_circulationaha-2022-062952_supp11.docx)
File (circ_circulationaha-2022-062952_supp2.pdf)
File (circ_circulationaha-2022-062952_supp3.pdf)
File (circ_circulationaha-2022-062952_supp4.pdf)
File (circ_circulationaha-2022-062952_supp5.pdf)
File (circ_circulationaha-2022-062952_supp6.pdf)
File (circ_circulationaha-2022-062952_supp7.pdf)
File (circ_circulationaha-2022-062952_supp8.pdf)
File (circ_circulationaha-2022-062952_supp9.pdf)

REFERENCES

1.
Shovlin CL. Hereditary haemorrhagic telangiectasia: pathophysiology, diagnosis and treatment. Blood Rev. 2010;24:203–219. doi: 10.1016/j.blre.2010.07.001
2.
Uebelhoer M, Boon LM, Vikkula M. Vascular anomalies: from genetics toward models for therapeutic trials. Cold Spring Harb Perspect Med. 2012;2:a009688. doi: 10.1101/cshperspect.a009688
3.
Marziano C, Genet G, Hirschi KK. Vascular endothelial cell specification in health and disease. Angiogenesis. 2021;24:213–236. doi: 10.1007/s10456-021-09785-7
4.
Robert F, Desroches-Castan A, Bailly S, Dupuis-Girod S, Feige JJ. Future treatments for hereditary hemorrhagic telangiectasia. Orphanet J Rare Dis. 2020;15:4. doi: 10.1186/s13023-019-1281-4
5.
Tual-Chalot S, Oh SP, Arthur HM. Mouse models of hereditary hemorrhagic telangiectasia: recent advances and future challenges. Front Genet. 2015;6:25. doi: 10.3389/fgene.2015.00025
6.
Foster DA, Yellen P, Xu L, Saqcena M. Regulation of G1 cell cycle progression: distinguishing the restriction point from a nutrient-sensing cell growth checkpoint(s). Genes Cancer. 2010;1:1124–1131. doi: 10.1177/1947601910392989
7.
Saqcena M, Menon D, Patel D, Mukhopadhyay S, Chow V, Foster DA. Amino acids and mTOR mediate distinct metabolic checkpoints in mammalian G1 cell cycle. PLoS One. 2013;8:e74157. doi: 10.1371/journal.pone.0074157
8.
Chavkin NW, Genet G, Poulet M, Jeffery ED, Marziano C, Genet N, Vasavada H, Nelson EA, Acharya BR, Kour A, et al. Endothelial cell cycle state determines propensity for arterial-venous fate. Nat Commun. 2022;13:5891. doi: 10.1038/s41467-022-33324-7
9.
Sakaue-Sawano A, Kurokawa H, Morimura T, Hanyu A, Hama H, Osawa H, Kashiwagi S, Fukami K, Miyata T, Miyoshi H, et al. Visualizing spatiotemporal dynamics of multicellular cell-cycle progression. Cell. 2008;132:487–498. doi: 10.1016/j.cell.2007.12.033
10.
Fry DW, Harvey PJ, Keller PR, Elliott WL, Meade M, Trachet E, Albassam M, Zheng X, Leopold WR, Pryer NK, et al. Specific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenografts. Mol Cancer Ther. 2004;3:1427–1438.
11.
Gallanis GT, Pericas RI, Riegel AT, Pohlmann PR. An evaluation of palbociclib as a breast cancer treatment option: a current update. Expert Opin Pharmacother. 2021;22:281–290. doi: 10.1080/14656566.2020.1838485
12.
Ola R, Dubrac A, Han J, Zhang F, Fang JS, Larrivee B, Lee M, Urarte AA, Kraehling JR, Genet G, et al. PI3 kinase inhibition improves vascular malformations in mouse models of hereditary haemorrhagic telangiectasia. Nat Commun. 2016;7:13650. doi: 10.1038/ncomms13650
13.
Ruiz S, Zhao H, Chandakkar P, Chatterjee PK, Papoin J, Blanc L, Metz CN, Campagne F, Marambaud P. A mouse model of hereditary hemorrhagic telangiectasia generated by transmammary-delivered immunoblocking of BMP9 and BMP10. Sci Rep. 2016;5:37366. doi: 10.1038/srep37366
14.
Iriarte A, Figueras A, Cerda P, Mora JM, Jucgla A, Penin R, Viñals F, Riera-Mestre A. PI3K (phosphatidylinositol 3-kinase) activation and endothelial cell proliferation in patients with hemorrhagic hereditary telangiectasia type 1. Cells. 2019;8:971. doi: 10.3390/cells8090971
15.
Kim S, Leong A, Kim M, Yang HW. CDK4/6 initiates Rb inactivation and CDK2 activity coordinates cell-cycle commitment and G1/S transition. Sci Rep. 2022;12:16810. doi: 10.1038/s41598-022-20769-5
16.
Fang JS, Coon BG, Gillis N, Chen Z, Qiu J, Chittenden TW, Burt JM, Schwartz MA, Hirschi KK. Shear-induced Notch-Cx37-p27 axis arrests endothelial cell cycle to enable arterial specification. Nat Commun. 2017;8:2149. doi: 10.1038/s41467-017-01742-7
17.
Park H, Furtado J, Poulet M, Chung M, Yun S, Lee S, Sessa WC, Franco CA, Schwartz MA, Eichmann A. Defective flow-migration coupling causes arteriovenous malformations in hereditary hemorrhagic telangiectasia. Circulation. 2021;144:805–822. doi: 10.1161/CIRCULATIONAHA.120.053047
18.
Lee HW, Xu Y, He L, Choi W, Gonzalez D, Jin SW, Simons M. Role of venous endothelial cells in developmental and pathologic angiogenesis. Circulation. 2021;144:1308–1322. doi: 10.1161/CIRCULATIONAHA.121.054071
19.
Genet G, Boye K, Mathivet T, Ola R, Zhang F, Dubrac A, Li J, Genet N, Henrique Geraldo L, Benedetti L, et al. Endophilin-A2 dependent VEGFR2 endocytosis promotes sprouting angiogenesis. Nat Commun. 2019;10:2350. doi: 10.1038/s41467-019-10359-x
20.
Rama N, Dubrac A, Mathivet T, Ni Charthaigh RA, Genet G, Cristofaro B, Pibouin-Fragner L, Ma L, Eichmann A, Chedotal A. Slit2 signaling through Robo1 and Robo2 is required for retinal neovascularization. Nat Med. 2015;21:483–491. doi: 10.1038/nm.3849
21.
Dubrac A, Genet G, Ola R, Zhang F, Pibouin-Fragner L, Han J, Zhang J, Thomas JL, Chedotal A, Schwartz MA, et al. Targeting NCK-mediated endothelial cell front-rear polarity inhibits neovascularization. Circulation. 2016;133:409–421. doi: 10.1161/CIRCULATIONAHA.115.017537
22.
Franco CA, Jones ML, Bernabeu MO, Geudens I, Mathivet T, Rosa A, Lopes FM, Lima AP, Ragab A, Collins RT, et al. Dynamic endothelial cell rearrangements drive developmental vessel regression. PLoS Biol. 2015;13:e1002125. doi: 10.1371/journal.pbio.1002125
23.
Falkenberg KD, Rohlenova K, Luo Y, Carmeliet P. The metabolic engine of endothelial cells. Nat Metab. 2019;1:937–946. doi: 10.1038/s42255-019-0117-9
24.
Luo W, Garcia-Gonzalez I, Fernandez-Chacon M, Casquero-Garcia V, Sanchez-Munoz MS, Muhleder S, Garcia-Ortega L, Andrade J, Potente M, Benedito R. Arterialization requires the timely suppression of cell growth. Nature. 2021;589:437–441. doi: 10.1038/s41586-020-3018-x
25.
Tual-Chalot S, Mahmoud M, Allinson KR, Redgrave RE, Zhai Z, Oh SP, Fruttiger M, Arthur HM. Endothelial depletion of Acvrl1 in mice leads to arteriovenous malformations associated with reduced endoglin expression. PLoS One. 2014;9:e98646. doi: 10.1371/journal.pone.0098646
26.
Park SO, Wankhede M, Lee YJ, Choi EJ, Fliess N, Choe SW, Oh SH, Walter G, Raizada MK, Sorg BS, et al. Real-time imaging of de novo arteriovenous malformation in a mouse model of hereditary hemorrhagic telangiectasia. J Clin Invest. 2009;119:3487–3496. doi: 10.1172/JCI39482
27.
Ola R, Kunzel SH, Zhang F, Genet G, Chakraborty R, Pibouin-Fragner L, Martin K, Sessa W, Dubrac A, Eichmann A. SMAD4 prevents flow induced arteriovenous malformations by inhibiting casein kinase 2. Circulation. 2018;138:2379–2394. doi: 10.1161/CIRCULATIONAHA.118.033842
28.
Hwan Kim Y, Vu PN, Choe SW, Jeon CJ, Arthur HM, Vary CPH, Lee YJ, Oh SP. Overexpression of activin receptor-like kinase 1 in endothelial cells suppresses development of arteriovenous malformations in mouse models of hereditary hemorrhagic telangiectasia. Circ Res. 2020;127:1122–1137. doi: 10.1161/CIRCRESAHA.119.316267
29.
Garcia de Vinuesa A, Abdelilah-Seyfried S, Knaus P, Zwijsen A, Bailly S. BMP signaling in vascular biology and dysfunction. Cytokine Growth Factor Rev. 2016;27:65–79. doi: 10.1016/j.cytogfr.2015.12.005
30.
Serra F, Lapidari P, Quaquarini E, Tagliaferri B, Sottotetti F, Palumbo R. Palbociclib in metastatic breast cancer: current evidence and real-life data. Drugs Context. 2019;8:212579. doi: 10.7573/dic.212579
31.
Tripathy D, Blum JL, Rocque GB, Bardia A, Karuturi MS, Cappelleri JC, Liu Y, Zhang Z, Davis KL, Wang Y. POLARIS: a prospective, multicenter, noninterventional study assessing palbociclib in hormone receptor-positive advanced breast cancer. Future Oncol. 2020;16:2475–2485. doi: 10.2217/fon-2020-0573
32.
Mahmoud M, Allinson KR, Zhai Z, Oakenfull R, Ghandi P, Adams RH, Fruttiger M, Arthur HM. Pathogenesis of arteriovenous malformations in the absence of endoglin. Circ Res. 2010;106:1425–1433. doi: 10.1161/CIRCRESAHA.109.211037
33.
Flieger D, Hainke S, Fischbach W. Dramatic improvement in hereditary hemorrhagic telangiectasia after treatment with the vascular endothelial growth factor (VEGF) antagonist bevacizumab. Ann Hematol. 2006;85:631–632. doi: 10.1007/s00277-006-0147-8
34.
Kovacs-Sipos E, Holzmann D, Scherer T, Soyka MB. Nintedanib as a novel treatment option in hereditary haemorrhagic telangiectasia. BMJ Case Rep. 2017;2017:bcr2017219393. doi: 10.1136/bcr-2017-219393
35.
Faughnan ME, Gossage JR, Chakinala MM, Oh SP, Kasthuri R, Hughes CCW, McWilliams JP, Parambil JG, Vozoris N, Donaldson J, et al. Pazopanib may reduce bleeding in hereditary hemorrhagic telangiectasia. Angiogenesis. 2019;22:145–155. doi: 10.1007/s10456-018-9646-1
36.
Crist AM, Zhou X, Garai J, Lee AR, Thoele J, Ullmer C, Klein C, Zabaleta J, Meadows SM. Angiopoietin-2 inhibition rescues arteriovenous malformation in a Smad4 hereditary hemorrhagic telangiectasia mouse model. Circulation. 2019;139:2049–2063. doi: 10.1161/CIRCULATIONAHA.118.036952
37.
Iriarte A, Ochoa-Callejero L, Garcia-Sanmartin J, Cerda P, Garrido P, Narro-Iniguez J, Mora-Lujan JM, Jucgla A, Sanchez-Corral MA, Cruellas F, et al. Adrenomedullin as a potential biomarker involved in patients with hereditary hemorrhagic telangiectasia. Eur J Intern Med. 2021;88:89–95. doi: 10.1016/j.ejim.2021.03.039
38.
Ruiz S, Chandakkar P, Zhao H, Papoin J, Chatterjee PK, Christen E, Metz CN, Blanc L, Campagne F, Marambaud P. Tacrolimus rescues the signaling and gene expression signature of endothelial ALK1 loss-of-function and improves HHT vascular pathology. Hum Mol Genet. 2017;26:4786–4798. doi: 10.1093/hmg/ddx358
39.
Gao Y, Raj JU. Regulation of the pulmonary circulation in the fetus and newborn. Physiol Rev. 2010;90:1291–1335. doi: 10.1152/physrev.00032.2009
40.
Sun Y, Smith LEH. Retinal vasculature in development and diseases. Annu Rev Vis Sci. 2018;4:101–122. doi: 10.1146/annurev-vision-091517-034018
41.
Liu W, Deng Z, Zeng Z, Fan J, Feng Y, Wang X, Cao D, Zhang B, Yang L, Liu B, et al. Highly expressed BMP9/GDF2 in postnatal mouse liver and lungs may account for its pleiotropic effects on stem cell differentiation, angiogenesis, tumor growth and metabolism. Genes Dis. 2020;7:235–244. doi: 10.1016/j.gendis.2019.08.003
42.
Baeyens N, Larrivee B, Ola R, Hayward-Piatkowskyi B, Dubrac A, Huang B, Ross TD, Coon BG, Min E, Tsarfati M, et al. Defective fluid shear stress mechanotransduction mediates hereditary hemorrhagic telangiectasia. J Cell Biol. 2016;214:807–816. doi: 10.1083/jcb.201603106
43.
Sorensen I, Adams RH, Gossler A. DLL1-mediated Notch activation regulates endothelial identity in mouse fetal arteries. Blood. 2009;113:5680–5688. doi: 10.1182/blood-2008-08-174508
44.
Abe T, Sakaue-Sawano A, Kiyonari H, Shioi G, Inoue K, Horiuchi T, Nakao K, Miyawaki A, Aizawa S, Fujimori T. Visualization of cell cycle in mouse embryos with Fucci2 reporter directed by Rosa26 promoter. Development. 2013;140:237–246. doi: 10.1242/dev.084111
45.
Shovlin CL, Angus G, Manning RA, Okoli GN, Govani FS, Elderfield K, Birdsey GM, Mollet IG, Laffan MA, Mauri FA. Endothelial cell processing and alternatively spliced transcripts of factor VIII: potential implications for coagulation cascades and pulmonary hypertension. PLoS One. 2010;5:e9154. doi: 10.1371/journal.pone.0009154
46.
Chavkin NW, Cain S, Walsh K, Hirschi KK. Isolation of murine retinal endothelial cells for next-generation sequencing. J Vis Exp. 2021;176. doi: 10.3791/63133
47.
Blankenberg D, Gordon A, Von Kuster G, Coraor N, Taylor J, Nekrutenko A; Galaxy Team. Manipulation of FASTQ data with Galaxy. Bioinformatics. 2010;26:1783–1785.
48.
Blankenberg D, Von Kuster G, Coraor N, Ananda G, Lazarus R, Mangan M, Nekrutenko A, Taylor J. Galaxy: a web-based genome analysis tool for experimentalists. Curr Protoc Mol Biol. 2010;Chapter 19:Unit 19.1021. doi: 10.1002/0471142727.mb1910s89
49.
Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi: 10.1093/bioinformatics/bts635
50.
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8
51.
Hafemeister C, Satija R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 2019;20:296. doi: 10.1186/s13059-019-1874-1

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Circulation
Circulation
Pages: 944 - 962
PubMed: 38126211

Versions

You are viewing the most recent version of this article.

History

Received: 25 October 2022
Accepted: 27 November 2023
Published online: 21 December 2023
Published in print: 19 March 2024

Permissions

Request permissions for this article.

Keywords

  1. arteriovenous malformations
  2. cell cycle control
  3. endothelial cell
  4. hereditary hemorrhagic telangiectasia
  5. palbociclib

Subjects

Authors

Affiliations

Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Nicholas W. Chavkin, PhD https://orcid.org/0000-0001-9058-0245
Robert M. Berne Cardiovascular Research Center (N.W.C., V.S., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Robert M. Berne Cardiovascular Research Center (N.W.C., V.S., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Agnès Figueras, PhD
Program Against Cancer Therapeutic Resistance, Institut Catala d’Oncologia, Hospital Duran i Reynals, Barcelona, Spain (A.F.).
Oncobell Program (A.F.), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.
(P.C., A.R.-M.), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.
HHT Unit, Internal Medicine Department, Hospital Universitari Bellvitge, Barcelona, Spain (P.C., A.R.-M.).
Stephanie P. McDonnell, BS
Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Danya Sankaranarayanan, BS
Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Elizabeth A. Nelson, MS
Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
(P.C., A.R.-M.), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.
HHT Unit, Internal Medicine Department, Hospital Universitari Bellvitge, Barcelona, Spain (P.C., A.R.-M.).
Department of Clinical Science, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Spain (A.R.-M.).
Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Robert M. Berne Cardiovascular Research Center (N.W.C., V.S., K.K.H.), School of Medicine, University of Virginia, Charlottesville.
Department of Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (K.K.H.).

Notes

*
G. Genet and N. Genet contributed equally.
Supplemental Material is available at Supplemental Material.
For Sources of Funding and Disclosures, see page 961.
Circulation is available at www.ahajournals.org/journal/circ .
Correspondence to: Gael Genet, PhD, or Karen K. Hirschi, PhD, Department of Cell Biology, 1340 Jefferson Park Ave, Charlottesville, VA 22908. Email [email protected] or [email protected]

Disclosures

Disclosures None.

Sources of Funding

The authors of this work were supported by the American Heart Association career development award (938744) and the Aneurysm and AVM foundation award (2023) to G.G., National Institutes of Health grants to K.K.H. (R01 EB 016629 and HL146056, and NIH U2EB017103), National Institutes of Health grant to K.K.H. and G.G. (NIH R01 HL 171284), an American Heart Association postdoctoral fellowship and career development award to N.G. (19POST34400065 and CDA1051175), National Institutes of Health grants to N.W.C. (T32HL0072240) and S.C. (T32HL007284), and an American Heart Association predoctoral fellowship to A.C. (23PRE1026220).

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. Evaluating the Vascular Risk of PFCs: An Integrated XGBoost-Driven Structure–Activity Prediction and Experimental Validation Study, Environment & Health, (2025).https://doi.org/10.1021/envhealth.5c00014
    Crossref
  2. Thyroid Hormone Signaling in Cerebrovascular Malformations: A Regulator of Vascular Stability and Repair, Circulation Research, 136, 9, (1028-1030), (2025)./doi/10.1161/CIRCRESAHA.125.326460
    Abstract
  3. From bench to bedside: murine models of inherited and sporadic brain arteriovenous malformations, Angiogenesis, 28, 2, (2025).https://doi.org/10.1007/s10456-024-09953-5
    Crossref
  4. Human iPSCs offer an alternative for modeling vascular malformation, Cell Stem Cell, 32, 2, (177-178), (2025).https://doi.org/10.1016/j.stem.2025.01.008
    Crossref
  5. Hereditary haemorrhagic telangiectasia, Nature Reviews Disease Primers, 11, 1, (2025).https://doi.org/10.1038/s41572-024-00585-z
    Crossref
  6. Genetic and pharmacological targeting of mTORC1 in mouse models of arteriovenous malformation expose non-cell autonomous signalling in HHT, Angiogenesis, 28, 1, (2024).https://doi.org/10.1007/s10456-024-09961-5
    Crossref
  7. Alk1/Endoglin signaling restricts vein cell size increases in response to hemodynamic cues, Angiogenesis, 28, 1, (2024).https://doi.org/10.1007/s10456-024-09955-3
    Crossref
  8. Inhibition of endothelial cell proliferation as a potential therapeutic approach in hereditary hemorrhagic telangectasia, Nature Cardiovascular Research, 3, 11, (1267-1269), (2024).https://doi.org/10.1038/s44161-024-00557-2
    Crossref
  9. CDK6-mediated endothelial cell cycle acceleration drives arteriovenous malformations in hereditary hemorrhagic telangiectasia, Nature Cardiovascular Research, 3, 11, (1301-1317), (2024).https://doi.org/10.1038/s44161-024-00550-9
    Crossref
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

Induced Endothelial Cell Cycle Arrest Prevents Arteriovenous Malformations in Hereditary Hemorrhagic Telangiectasia
Circulation
  • Vol. 149
  • No. 12

Purchase access to this journal for 24 hours

Circulation
  • Vol. 149
  • No. 12
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Figures

Tables

Media

Share

Share

Share article link

Share

Comment Response