Abstract
Objective
We sought to determine how EC expression of the activating k-Ras mutation, k-RasV12, affects their ability to form lumens and tubes and interact with pericytes during capillary assembly.
Approach and Results
Using defined bioassays where human ECs undergo observable tubulogenesis, sprouting behavior, pericyte recruitment to EC-lined tubes, and pericyte-induced EC basement membrane deposition, we assessed the impact of EC k-RasV12 expression on these critical processes that are necessary for proper capillary network formation. This mutation, which is frequently seen in human ECs within brain arteriovenous malformations, was found to markedly accentuate EC lumen formation mechanisms, with strongly accelerated intracellular vacuole formation, vacuole fusion and lumen expansion, and with reduced sprouting behavior, leading to excessively widened tube networks compared to control ECs. These abnormal tubes demonstrate strong reductions in pericyte recruitment and pericyte-induced EC basement membranes compared to controls, with deficiencies in fibronectin, collagen type IV and perlecan deposition. Analyses of signaling during tube formation from these k-RasV12 ECs reveals strong enhancement of Src, Pak2, b-Raf, Erk and Akt activation and increased expression of PKCε, MT1-MMP, acetylated tubulin and CDCP1 (most are known EC lumen regulators). Pharmacologic blockade of MT1-MMP, Src, Pak, Raf, Mek kinases, Cdc42/Rac1, and Notch markedly interferes with lumen and tube formation from these ECs.
Conclusions
Overall, this novel work demonstrates that EC expression of k-RasV12 disrupts capillary assembly due to markedly excessive lumen formation coupled with strongly reduced pericyte recruitment and basement membrane deposition, which are critical pathogenic features predisposing the vasculature to develop arteriovenous malformations.
Keywords: Endothelial cell, activated k-Ras, k-RasV12, arteriovenous malformation, lumen formation, pericyte recruitment, basement membrane deposition, capillary deficiency
Graphical Abstract
INTRODUCTION
Considerable progress has occurred in defining the molecular basis for a class of vascular lesions termed vascular anomalies1–9. These anomalies are complex and encompass many different types of lesions that arise from the blood or lymphatic vasculatures. Broadly speaking, the vascular anomalies are characterized by marked vascular morphogenic lesions such as those observed in arteriovenous, venous, capillary or cavernous malformations1–3, 7, or they are characterized by more proliferative lesions such as observed in hemangiomas8–11. In recent years, it has become increasingly appreciated that these vascular anomalies are caused by genetic mutations of key molecules that primarily stimulate Ras/Map kinase and/or PI3 kinase signaling pathways. Some of the key signaling molecules that drive these vascular anomalies are activating mutations in k-Ras, b-Raf, Map2k1 (Mek1), GNAQ, PIK3CA, and Tie2, and inactivating mutations in Rasa1, a RasGAP which inactivates Ras2, 3, 5–8, 11–26. Very recently, it was found that activating mutations in k-Ras (such as k-RasV12) are observed at high frequency within endothelial cells (EC) in human brain arteriovenous malformations12.
In our studies of EC lumen and tube formation, we reported a few years ago that k-Ras was observed to be an important regulator of tube formation along with Cdc42, Rac isoforms, and Rap1b, and furthermore, that siRNA suppression of Rasa1 accelerated the lumen formation process27. Thus, it is possible that one of the key abnormalities in malformation syndromes may be due to excessive or altered EC lumen formation, a process that has not been examined in detail in this context. Over the past 25 years, considerable progress has been made investigating the mechanistic and molecular basis for EC lumen and tube assembly in vitro and in vivo28–32. Critical steps during EC lumen formation include: 1) intracellular vacuole formation and coalescence33–37; 2) vacuole/vesicle trafficking along the microtubule cytoskeleton to create and expand the apical membrane surface27, 38, 39; 3) creation of both cytoskeletal and membrane polarity that leads to basally polarized F-actin, subapically polarized acetylated tubulin (to support vacuole trafficking and the developing apical membrane surface)38–40, and apically polarized proteins such as activated Src isoforms, caveolin-1, Rasip-1, and podocalyxin27, 39, 41; and 4) tube expansion and elongation in the extracellular matrix (ECM) secondary to membrane-type 1 metalloproteinase (MT1-MMP)-dependent proteolysis of the matrix leading to generation of vascular guidance tunnels which are matrix spaces where the EC tube networks are embedded42–45. Over many years, key molecular regulators of these steps and this process have been identified and these include the small GTPases, Cdc42, Rac1, Rac2, k-Ras, and Rap1b, as well as RalA, RalB, and various Rab GTPases27, 34, 40, 41, 46. Many critical downstream effectors of these GTPases have been identified to play a role including Pak2, Pak4, IQGAP-1, MRCKβ, GIT1, βPIX, and Rasip127, 41, 46–48. Major kinases that have been found to be important for EC lumen formation include protein kinase C epsilon (PKCε), Src family kinases including Src, Fyn and Yes, as well as Pak, Raf, and Erk kinases39, 46, 47, 49. Furthermore, a key control step in EC lumen and tube formation is ECM proteolysis, which is mediated by MT1-MMP, and directly coordinated with the EC lumen signaling pathways and molecules mentioned above42, 43, 45. Other interesting regulators of EC lumen formation include the microtubule tip complex proteins, EB1, p150glued and Clasp138, or the cerebral cavernous malformation (CCM) genes, CCM1, CCM2, CCM2L, and CCM3, which interestingly suppress the activation of RhoA, an inhibitor of lumen formation50–53.
A fundamental requirement for the development of the blood vascular system are proper interactions between ECs and mural cells, such as pericytes54, which are necessary to create and maintain capillary networks with their characteristic narrow lumen diameters44, 55–58. When ECs form tube networks in the absence of pericytes in vitro, they continue to widen and become less and less elongated42. ECs without pericytes under our serum-free defined systems in either collagen or fibrin matrices are unable to form basement membranes, while with pericytes present, dramatic basement membrane deposition occurs44, 59. The dramatic widening of ECs tubes without pericytes occurs due to a continuation of morphogenesis and lack of a basement membrane, which normally suppresses morphogenesis and facilitates tube maturation30. Very recently, we investigated and identified the key EC-derived factors that regulate pericyte recruitment to EC tubes and thus, they also control capillary formation and basement membrane deposition. These EC-derived factors affecting pericyte behavior are PDGF-BB, PDGF-DD, endothelin-1 (ET-1), TGFβ1, and HB-EGF57. Individual blockade of these factors or their receptors had modest, but significant inhibitory effects on pericyte recruitment, while combined blockade of these molecules or receptors much more dramatically interfered with pericyte recruitment leading to markedly widened tubes with little to no basement membrane deposition57. In fact, under these highly inhibited conditions, the ECs formed tube networks that continued to expand in diameter and shorten in length as if the pericytes were not present.
In general terms, many vascular malformations show reduced mural cell interactions with the malformed EC morphogenic structures60. A few studies have implicated alterations in PDGF-BB levels as a potential reason for these differences61, but as mentioned above, this is likely to be much more complex due to the fact that under normal conditions multiple EC-derived factors work together to stimulate this pericyte recruitment and basement membrane assembly process57. In order to investigate these types of key questions, which are necessary to both understand the underlying pathogenesis of particular syndromes, but also to develop new therapeutic strategies to prevent or fix these abnormal vascular morphogenic or proliferative structures, there is a critical need to create definable in vitro 3D malformation or hemangioma models using human vascular cells including ECs and mural cells. This approach is important to investigate in detail the contribution of individual genetic mutations and to assess their biological impact on the key steps controlling vascular cell morphogenesis, proliferation or EC-pericyte interactions. For example, evaluation of EC lumen formation, tube elongation, sprouting behavior, pericyte recruitment to the mutant EC morphogenic structures, and the degree to which vascular basement membrane assembly occurs, are particularly relevant issues to understand the biology underlying why particular mutations lead to specific malformation syndromes.
Here, we investigate the impact of a common mutation observed in ECs within human brain arteriovenous malformations (i.e. k-RasV12), on the ability of human ECs to undergo morphogenic processes and to also assess the ability of these expressing cells to interact with pericytes to affect vascular maturation events using a defined 3D model system. Our findings demonstrate that ECs expressing activated k-RasV12 show remarkably enhanced ability to form intracellular vacuoles, lumens, vascular guidance tunnels, and tubes, which are markedly widened compared to control ECs. In contrast, the k-RasV12 ECs demonstrate reduced ability to sprout compared to control ECs, which contributes to why tubes formed by these ECs are widened and less elongated. Furthermore, the widened tubes formed by k-RasV12-expressing ECs show strongly reduced pericyte recruitment and basement membrane deposition, particularly observed with fibronectin, collagen type IV and perlecan, compared to controls. Signaling experiments during 3D morphogenesis reveal marked enhancement and activation of Src, Pak2, b-Raf, Erk, and Akt, and increased expression of PKCε, MT1-MMP, acetylated tubulin, and CDCP1 by the k-RasV12-expressing ECs. Individual pharmacologic blockade of MT1-MMP, Src family kinases, Cdc42 and Rac1, Pak kinases, Raf kinase, Mek kinase, PKCε, PI3 kinase or Notch signaling strongly inhibit the ability of k-RasV12 ECs to form lumens and tubes. Overall, k-RasV12 expression by ECs results in an inability of these cells to assemble normal capillary networks due to excessive lumen formation and tube widening combined with impaired pericyte recruitment and basement membrane deposition. These abnormalities represent key pathogenic features leading to deficiencies in the assembly of intervening capillaries, a predisposing step in the development of arteriovenous malformations.
METHODS
The authors declare that all supporting data are available within the article [and its online supplementary files].
Cell culture
The cell lines used in this paper, human umbilical vein endothelial cells (HUVECs), k-RasV12 ECs, and human brain vascular pericytes (HBVP) were cultured in our own “Supermedia,” on gelatin-coated flasks. These cells were grown in incubators containing 5% CO2 at 37°C. Our “Supermedia” consists of 20% fetal bovine serum, heparin sodium salt, bovine hypothalamus extract, amphotericin B, and gentamicin in M199 media. HUVECs were used from passage 3 to 6, k-RasV12 ECs were used from passage 6 to 12, and HBVPs were used from passage 4 to 12. The k-RasV12 expressing ECs were obtained by infecting HUVECs with a recombinant lentivirus carrying k-RasV12 (k-Ras isoform b, NP_004976) and also red fluorescent protein fused with blasticidin, which was used for selection (GenTarget). The k-RasV12 mutant gene is expressed downstream of an EF1a promoter. After infection, we waited 3 days and selected with blasticidin (10 μg/ml). The new EC line was grown up from many clones of surviving cells that were propagated into a pool of ECs expressing k-RasV12. This pool of ECs was frozen down in liquid nitrogen vials, so that they could be regrown and used in future experiments.
Vasculogenic Assay
A T75 flask of HUVECs or k-RasV12 ECs were trypsinized and resuspended in a 2.5 mg/ml type I collagen matrix62. The gel was then added to a 96-well, half area plate (28 ul/well) and placed in an incubator for 30 mins, allowing for polymerization/equilibration. After polymerization, feeding media was added on top of gels. Our feeding media consists of M199 media plus five growth factors (RSII with insulin, FGF2, SCF, IL3, and SDF)62. These cultures were fixed in 3% paraformaldehyde or 3% glutaraldehyde at predetermined timepoints for fluorescent or nonfluorescent imaging. For the drug blocking assays, the pharmacologic drugs were mixed with feeding media at the desired concentrations and were added to cultures.
EC/k-RasV12 pericyte coculture
The ECs (HUVECs or k-RasV12-expressing) and GFP-labeled pericytes were cocultured in 2.5 mg/ml type I collagen matrices in a ratio of 1:5. The GFP-pericytes were generated as previously described55. The mixture was added to the 96-well plate. After polymerization, the media with five growth factors (RSII with insulin, FGF2, SCF, IL3, and SDF) were added to these cultures. These cultures were fixed at various timepoints with paraformaldehyde and were then immunostained for CD31 to label ECs or were stained with key basement membrane matrix components. Confocal imaging was then performed.
Sprouting Assay
The 2.5 mg/ml type I collagen matrices were added to the 96-well plate. HUVECs or k-RasV12 ECs were resuspended in the five growth factor media and were added on the top of the collagen matrices after they were polymerized63. These cultures were fixed with glutaraldehyde at 16 hr and stained with 0.1% toluidine blue for imaging. To quantify the tip number of HUVECs or k-RasV12, pictures were taken using a 4X objective and ImageJ was used to count the tip number. To quantify the tip number from both cell types, each picture was taken 20 μm below the monolayer.
Culture Fluorescent Immunostaining
The cultures were fixed with 3% paraformaldehyde. After fixation, the collagen gels were plucked out and washed in Tris-Glycine for 1 hour on a shaker at 4°C. If staining for intracellular proteins, such as CD31, caveolin-1, and acetylated tubulin, gels were permeabilized with Triton TX-100 for 1 hour on the shaker. If staining for basement membrane components, for example, collagen type I or various basement membrane components such as laminin, fibronectin and collagen type IV, cells were not permeabilized. Gels were subsequentially incubated with secondary antibody-specific blocking buffer for 1 hr at 4°C. Primary antibody was added directly to the blocking buffer and the gel was incubated with the primary antibody overnight at 4°C. After several washes with wash buffer— Tris-buffered saline with Tween 20 (TBST) (if staining for intracellular proteins) or Tris-buffered saline (TBS) (if stained with basement membrane components), the same blocking buffer was added to these gels and a specific Alexa Fluor secondary antibody was added to corresponding gels and incubated for 2 hr in 4°C. After 2 hr, the secondary antibody was removed, and the gels were washed several times with TBS or TBST. Hoechst was added to the wash buffer and the gels were incubated for 1 hr. Again, TBS or TBST was used to wash gels until background has been washed out. Gels were then imaged by confocal microscopy.
Microscopy and Imaging
Images for quantification of lumen area/vacuoles number of HUVECs or k-RasV12 ECs were taken using an Olympus CKX41 microscope and imaging software (DPController/DPManager). Quantification of lumen area/vacuoles number was made by Fiji (ImageJ) and results were analyzed using Microsoft Excel. Quantitation of EC lumen and tube area was performed as described using Metamorph software (MetaMorph 7.8)64. For the time-lapse movies, pictures were taken every 10 minutes using Leica Microsystems (DMI6000B microscope plus environmental chamber) and imaging software (MetaMorph 7.8) in 72 hours period. All the pictures were taken using a monochromatic Hamamatsu ORCA-ER C4742-80 camera and 10X PH1 Leica 506507 lens. To create a movie, these pictures were stack together into a movie using MetaMorph software and stabilized using Adobe After Effects. Confocal pictures were taken using Leica SP8 3X STED Laser Confocal Microscope and image software (LAS X), all pictures were obtained with 3 different lenses (20X HC PL APO, 0.75NA, WD 0.62 mm; 40X HC PL APO, water immersion, 1.1NA, WD 0.65 mm; 63X HC PL APO, Oil immersion, 1.4NA, WD 0.14 mm). The raw pictures were made and reconstructed by Fiji (ImageJ).
Western blots
The cultures (gels) were plucked out and dissolved in a 1.5% sample buffer with 5% beta-mercaptoethanol. These samples were then loaded into a heat block (100°C) for 5 minutes. After the samples cooled down, 15 μl of sample was pipetted into a Biorad gel and ran until the ladder reached the bottom. The gel was transferred with a PVDF membrane and the membrane was blocked with 3% BSA. Specific primary antibodies were added directly into blocking buffer and incubate overnight at 4°C. After several washes, a corresponding HRP antibody in 3% milk was used to detect the protein. The results were imaged on x-ray film after developing.
Reverse transcription polymerase chain reaction (RT-PCR)
Direct-zol RNA miniprep kit was used to extract EC RNA (HUVECs or k-RasV12 ECs) based on the manufacturers’ instructions. AccuScript first strand cDNA kit was used to synthesis cDNA with 500 ng of RNA. Primer sequences that were used for targeted amplification are shown in the Major Resources Table. PCR products were run on a 1% agarose gel, and the results were visualized using a Fotodyne system (Fotodyne Inc).
Statistics
Statistical data analyses were performed using Microsoft Excel (Microsoft) or Prism 8 (Graphpad). Data were analyzed for normality and equal variances were obtained and student t-tests were used to compare means between two conditions. Statistical significance was set at a minimum of p<.05.
RESULTS
ECs expressing k-RasV12 show markedly enhanced ability to form lumen and tube networks compared to control ECs
Previously, our laboratory demonstrated that k-Ras in conjunction with other small GTPases such as Cdc42, Rac isoforms, and Rap1b control the process of EC lumen and tube formation27. Here, we examined whether EC expression of activated k-Ras (i.e. k-RasV12), a key activating mutation found in ECs within human arteriovenous malformations, would affect lumen and tube development. We directly compared lumen and tube formation using control ECs vs. ECs expressing k-RasV12 and using assays that mimic vasculogenesis. Cultures were fixed and stained with toluidine blue at different time points, and lumen/tube area and ECs with multiple intracellular vacuoles were quantified (Figures 1A and 2A,B). Co-immunostaining of fixed cultures with anti-CD31 antibodies to label ECs and anti-collagen type I antibodies to label the extracellular matrix (ECM) reveals strong acceleration of lumen formation and tube expansion from ECs carrying k-RasV12 (Figure 1B). EC lumen formation is accompanied by the creation of vascular guidance tunnels which are physical spaces in the ECM created by MT1-MMP-dependent proteolysis during this process42. ECs expressing k-RasV12 show marked expansion of EC lumens and tubes, but also vascular guidance tunnel spaces within the collagen matrices compared to control ECs (Figure 1B). In support of these conclusions, quantitative measurements of lumen area showed that ECs expressing k-RasV12 formed significantly larger lumens with markedly enhanced tube area after 24, 48, or 72 hr (Figure 3A). Detailed quantitative analysis of EC lumen formation (i.e. evaluated at 3 hr intervals) using real-time movies over 72 hr revealed the marked acceleration of lumen and tube formation that results from EC expression of k-RasV12 (Figure 3B).
Increased intracellular vacuole formation from ECs expressing k-RasV12 leading to enhanced lumen formation and apical membrane targeting of caveolin-1
Previous work from our laboratory has demonstrated a key relationship between the ability of ECs to form intracellular vacuoles and for them to rapidly form lumens and tube networks in 3D extracellular matrix environments33, 35, 36, 39, 40. Here, we addressed whether the enhanced lumen and tube formation seen following EC expression of k-RasV12 was related to their ability to form intracellular vacuoles during the lumen formation process. In fact, the ECs carrying k-RasV12 do show strong increases in the accumulation of EC intracellular vacuoles compared to control ECs during the early phases of lumen formation (Figure 2A,B). We have previously reported that caveolin-1 labels intracellular vacuoles33, 40 and can accumulate at the developing apical membrane surface as apically targeted vacuoles fuse with this surface during EC lumen formation40. Immunostaining of control vs. k-RasV12 expressing ECs with an anti-caveolin-1 antibody (and imaged using confocal microscopy) reveals strong labeling of both intracellular vacuoles, but also the developing EC apical membrane, which is markedly accentuated by the expression of k-RasV12 in ECs (Figures 4A and Suppl. Figure I). Intracellular vacuoles are decorated with caveolin-1, which is observed at 3, 6, or 9 hr of culture, and particularly evident using the k-RasV12 ECs (Figures 4A and Suppl. Figure I). The same anti-caveolin-1 immunostaining of cultures at 24, 48, and 72 hr, reveals the marked expansion of the lumen and tube spaces that occurs when ECs expressing k-RasV12 undergo morphogenesis as compared to control ECs (Figure 4B).
Real-time movies demonstrate marked accentuation of EC intracellular vacuolation as well as lumen and tube expansion from ECs expressing k-RasV12 compared to control ECs
To further evaluate the above observations during these EC lumen and tube formative events, we performed real-time movie analysis comparing control ECs (Suppl. Movies I–III) to k-RasV12 expressing ECs (Suppl. Movies IV–VII). By visualizing real-time movies over 72 hr, we observe strong increases in EC vacuolation within the first 24 hr and marked lumen formation and tube expansion in the k-RasV12 ECs compared to control ECs from 24–72 hr of culture. We selected multiple independent fields for this analysis and representative movies are shown to demonstrate these findings (Suppl. Movies I–VII). The results observed in these movies strongly support the conclusions that ECs expressing k-RasV12 markedly accelerate intracellular vacuolation, vacuole fusion, lumen formation and tube width expansion during vascular morphogenesis compared to control ECs (Figures 1–4).
ECs carrying k-RasV12 show reduced sprouting behavior compared to control ECs
Previous work from our laboratory has shown that there is an inverse relationship between EC sprouting behavior and lumen formation, whereby treatments that lead to reduced lumen formation increase EC sprouting activity and vice-versa39, 63. For example, blockade of Src family kinases, Notch signaling or both result in strong inhibition of EC lumen formation, while they concomitantly increase EC sprouting behavior. Here, we assessed the ability of k-RasV12 expressing ECs vs. control ECs to sprout in 3D matrices (Figure 5B,C). Cultures were fixed, stained, and quantified after 16 hr, using an optimized EC sprouting and invasion assay system63. A statistically significant reduction in EC tip numbers was observed for k-RasV12 ECs compared to control ECs, suggesting a strongly reduced sprouting ability for these ECs (Figure 5A). In top-down images of these cultures, control ECs displayed multiple invading EC tip cells, while k-RasV12 ECs showed many fewer invading cells (Figure 5B). Similarly, images from these cultures viewed from the side revealed the same result and conclusion. EC controls are shown to be invading (black arrowheads), whereas k-RasV12 ECs showed little invasive behavior (Figure 5C). Thus, these results are very consistent with our previous work in terms of the inverse relationship of EC sprouting behavior with the ability to form EC lumens. The marked increase in EC lumen formation by k-RasV12 expressing ECs is accompanied by strong reductions in their ability to invade as single cells to initiate EC sprouting behavior.
Identification of key signaling pathways that are activated during the marked enhancement of lumen formation and tube expansion from ECs expressing k-RasV12
In order to assess signaling pathways and identify important regulators that control k-RasV12 tubulogenesis, western blots were performed to compare both control ECs and k-RasV12 expressing ECs during 3D tube morphogenic events over time (Figure 6). Particularly striking differences between these two conditions were observed with increased phosphorylation of Src, Pak2, b-Raf, Erk, Akt, CDCP1, and GSK3β (which inactivates GSK3β) from k-Ras expressing ECs compared to controls (Figure 6A,B). Lesser increases were observed with increased phosphorylation of PKC epsilon (PKCε), and c-Raf from the k-RasV12 ECs (Figure 6A). Key previous work has demonstrated critical roles for PKCε, Src family kinases, as well as Pak, Raf, and Erk kinases on EC lumen formation using our models of EC tubulogenesis and from others work46, 47, 49, 65, while other studies demonstrated a role for GSK3β inactivation during Cdc42-dependent EC lumen formation66. In addition, increased expression of MT1-MMP, α2 integrin (a collagen-binding integrin), and acetylated tubulin were observed in the k-RasV12 expressing ECs compared to controls, and these have been shown to be important for EC lumen and tube formation33, 42, 45. Other molecules such as phospho-Pak4, Dusp6, phospho-cofilin, phospho-histone H3, and detyrosinated tubulin showed similar patterns of expression during this time course using both cell types (Figure 6). The marked downregulation of Dusp6, a key phosphatase which inactivates Erk occurred from both cell types during tube formation. In addition, the strong downregulation of phospho-histone H3 (a key marker of cell proliferation), indicates that the differences observed between the k-RasV12 and control ECs were not the result of cell proliferation (Figure 6B). Also, the real-time movies that we performed (Supplemental Movies I–VII) did not reveal any evidence of EC proliferation. Furthermore, our previous studies as well as others shows that vascular morphogenic events are accompanied by downregulation of EC proliferation58, 67, 68, and interestingly, that is also occurring with the k-RasV12 expressing ECs during their excessive lumen and tube formation (Figure 6B). Overall, these results are consistent with the conclusion that k-RasV12 expression in ECs strongly stimulates EC lumen through acceleration of intracellular vacuole formation and lumen expansion mechanisms which are driven by key signaling pathways and molecules that have been previously described by our laboratory and others.
Pharmacologic inhibitors of MT1-MMP, Cdc42/Rac1, PKCε, Src family kinase, Notch, Raf, Mek, and PI3 kinase signaling strongly inhibit the ability of k-RasV12 expressing ECs to form lumens and tubes
To address the functional role of many of the molecules and signaling pathways identified through Western blot analysis (Figure 6), we added pharmacologic inhibitors from the beginning of culture and quantified their impact on the lumen formation from the k-RasV12 expressing ECs after 72 hr (Figure 7). The marked expansion of lumens and tubes from these k-RasV12 ECs also leads to expansion of vascular guidance tunnels (Figure 1B), matrix spaces which are known to be generated by MT1-MMP-dependent proteolytic processes. Addition of the matrix metalloproteinase (MMP) inhibitor, GM6001, which inhibits the function of MT1-MMP, completely blocked the ability of these ECs to form lumens (Figure 7A,B). Addition of the Src family kinase inhibitor, PP2, markedly blocks lumen formation from k-RasV12 expressing ECs, while the Notch inhibitor, DBZ, also blocks, but to a lesser extent (Figure 7C). The combination of PP2 and DBZ, also strongly interferes with lumen formation. Interestingly, past work revealed that increased expression of activated Notch4 in ECs led to arteriovenous malformations in mice69. The Cdc42 and Rac inhibitor, MBQ167, as well as the Pak inhibitors, Frax486, and Frax597, interfere with lumen formation from these cells (Figure 7C). Our past work demonstrates the critical role of Cdc42 and Rac isoforms as well as their downstream effectors, including Pak2 and Pak4 during the EC lumen formation process. These critical molecules also play a major role in EC intracellular vacuole formation, an important regulator of EC lumen formation, and a process which is markedly accentuated by the EC expression of k-RasV12 (see Figures 2A and B, Figures 4A and Suppl. Figure I). Inhibitors of Raf kinase (PLX8394), Mek kinase (U0126) and PI3 kinase (Pictilisib) also significantly interfered with EC lumen formation from these cells, while the broad spectrum PKC inhibitor, Go6983 (an inhibitor of PKCε), also strongly blocked, while the PKCα selective inhibitor, Go6976 did not (Figure 7C,D). Importantly, both Raf kinases and PI3 kinase are direct downstream effectors of activated k-RasV12, and our findings suggest that both play roles in the ability of the k-RasV12 expressing ECs to form lumens and tubes. Other inhibitors directed to Rho kinase (Y27632), TGFβ signaling and the TGFβ receptor, Alk-5 (SB431542) had no effect on lumen formation from the k-RasV12 ECs, while the BMP signaling and BMP receptor antagonist directed to Alk-2 and Alk-3 receptors (DMH1) significantly increased lumen formation from these cells (Figure 7D). To further illustrate the effects of these agents, we show a few representative images demonstrating the strong blocking effect of GM6001, PP2, MBQ167, and U0126, while DMH1 enhances lumen formation from ECs expressing k-RasV12 (Suppl. Figure II). With these blocking agents, the ECs assumed a different morphologic or morphogenic appearance, with single cells (and no lumens) apparent following addition of GM6001, spindle-shaped cells (and no lumens) with PP2, small EC lumen cyst-like structures with MBQ167, and very small lumen structures with some branching with U0126 (Suppl. Figure II). For comparison, we added the same set of pharmacologic inhibitors to control ECs, and there were strong blocking effects of the different drugs on EC lumen formation except for Y27632, SB431542, and DMH1, which showed much less inhibitory effect (Suppl. Figure III). Overall, control ECs appear to be more sensitive to the morphogenesis-blocking effects of these signal transduction inhibitors compared to the ECs expressing k-RasV12.
Two additional experiments were performed with the k-RasV12 expressing ECs to address the effect of the pharmacologic inhibitors on the initial steps in lumen formation which was to assess their influence on vacuole formation (Suppl. Figure IV) or to assess their ability to affect pre-existing EC tubes by adding them after 48 hr of culture (Suppl. Figures V and VI). The Src family kinase inhibitor, PP2, and the Cdc42/Rac inhibitor, MBQ132, had the strongest inhibitory activity on vacuole formation from the k-RasV12 ECs, while the other drugs had less effect (Suppl. Figure IV). This is consistent with the known role of Src family kinases as well as Cdc42 and Rac in the vacuole formation process28, 34, 39, 46. When the different pharmacologic agents were added to cultures after they had formed tubes for 48 hr, strong blocking effects were observed following addition of the combination of PP2 and DBZ (blocking both Src and Notch) or MBQ132 (blocking Cdc42/Rac), while addition of DMH1 (blocking Alk2 and Alk3) led to increased lumen area (Suppl. Figure V). Representative photographs from these cultures reveal tube collapse responses seen after PP2/DBZ (collapsed lumens and appearance of spindle-shaped ECs) and MBQ132 addition, which showed many cyst-like lumens, while DMH1 treatment caused expanded lumen structures (Suppl. Figure VI). Lesser, but significant blocking effects were observed with the drugs interfering with Pak, Raf, Mek, PKCε and PI3 kinases (Suppl. Figure V). These data indicate that multiple stages of EC lumen/tube formation and maintenance are affected by these signaling molecules and pathways in the context of the abnormally accelerated lumen forming ability of ECs expressing k-RasV12.
Expression of k-RasV12 in ECs leads to deficiencies in capillary assembly due to strong reductions in pericyte recruitment and pericyte-induced EC basement membrane matrix deposition
Capillaries are composed of EC tube networks with associated pericytes, and they critically intervene between arteries and veins to keep them spatially and functionally separated. If capillary networks become deficient, this spatial and functional separation disappears leading to direct connections between arteries and veins creating arteriovenous malformations. Since EC expression of k-RasV12 is known to occur within human arteriovenous malformations, we tested whether EC expression of this activated k-Ras would affect the ability of ECs to form functional networks of capillaries with associated pericytes. Control ECs were compared to k-RasV12 expressing ECs and EC-pericyte co-cultures were established and evaluated over a 120 hr period. Cultures were fixed at 120 hr, and were immunostained with anti-CD31 antibodies to visualize the EC tube networks and determine if GFP-labeled pericytes were found to be associated with the control EC or k-RasV12 EC-lined tubes (Figure 8A). The k-RasV12 EC tubes were observed to be abnormally wide compared to control EC tubes and many fewer pericytes were associated with these tube networks compared to control ECs (Figure 8A, Suppl. Figure VII). Quantitation of these events reveals significantly fewer total pericytes, fewer pericytes elongated on tubes, and a much lower percentage of pericytes associated with ECs tubes expressing k-RasV12 (Figure 8B,C). Immunostaining of control gels with anti-collagen type I antibodies reveals narrow vascular guidance tunnels where pericytes are observed to recruit to EC tubes within the tunnel spaces (Figure 8A). In contrast, ECs with k-RasV12 show much wider and less branched tunnel spaces. Note that many of the pericytes in this instance are observed outside the tunnel spaces, and thus, did not recruit to the widened tubes (Figure 8A).
Recently, we defined key EC-derived factors that are responsible for pericyte recruitment and basement membrane deposition during capillary formation57. These EC factors are PDGF-BB, PDGF-DD, ET-1, HB-EGF, and the TGFβ isoforms, TGFβ1 and TGFβ2 (although this isoform is primarily expressed by pericytes). Blockade of these factors or their receptors in combination markedly interfered with pericyte recruitment and basement membrane deposition. Because of the deficiencies observed in pericyte recruitment to EC tubes expressing k-RasV12, we performed RT-PCR analysis to determine if there were any major observable differences between k-RasV12 ECs and control ECs (Figure 9B). The expression of k-RasV12 led to a clear upregulation of k-Ras in these ECs, while they showed very similar expression of major EC selective genes including VE-cadherin, VEGFR2, von Willebrand factor, claudin-5, Alk-1, and Erg (Figure 9B). Of the known EC-derived factors controlling pericyte recruitment, we observed decreased expression of PDGF-D and TGFβ2 by the k-RasV12 ECs compared to control ECs (Figure 9B), while the other key factors showed similar expression, so it is possible that these deficiencies could explain in part the defective pericyte recruitment to these widened tubes. However, because of the inherent complexity of this issue and process, we feel that a separate future study is necessary to address this question in detail.
A key step in pericyte-induced EC tube maturation and the development of capillary networks is the assembly of the capillary basement membrane matrix. Here, we compared the ability of control vs. k-RasV12 expressing EC tubes to deposit basement membranes in EC-pericyte co-cultures (Figure 9A, Suppl. Figure VII). Control EC-lined tubes recruit pericytes and markedly assemble the capillary basement membrane including laminins, collagen type IV, fibronectin, perlecan, nidogen 1 and nidogen 2. In contrast, strong reductions in the deposition of fibronectin, collagen type IV, and perlecan are observed from the k-RasV12 expressing EC tubes (Figure 9A, Suppl. Figure VII). In addition, reduced deposition of laminins, nidogen 1 and nidogen 2 are also observed compared to control ECs. To address if there is any apparent deficiency in the mRNA expression of these basement membrane components by the k-RasV12 ECs compared to control ECs, we performed RT-PCR (Figure 9B). Our results suggest that they express each of the key basement membrane components compared to control ECs and, also show increased expression of laminin α5, laminin β2, and nidogen 1 at higher levels compared to control ECs (Figure 9B). Thus, the deficiencies in basement membrane deposition appear to be due to failures in EC-pericyte interactions, but also may be related to the excessive lumen formation process driven by MT1-MMP-dependent proteolysis of ECM proteins, most notably by degrading collagens such as type IV collagen and the interstitial matrix protein, type I collagen. An important point is that ECM integrity is necessary to anchor the basement membrane matrix to the interstitial matrix during its assembly, which is needed to stabilize the capillary networks and their underlying basement membrane.
Another possibility is that k-RasV12 ECs might show changes in basement membrane protein secretion compared to control ECs. To address this possibility, we collected conditioned medium after 3 or 5 days from ECs forming lumens and tubes in 3D collagen matrices vs. those seeded onto two-dimensional (2D) collagen-coated plastic surfaces and we performed Western blots to measure the levels of the different basement membrane proteins. The k-RasV12 ECs show reduced secretion of fibronectin, perlecan and nidogen-1 compared to control ECs, while collagen type IV, laminin, and nidogen-2 appeared to be similar to control (Suppl. Figure VIII). In contrast, we examined other EC secreted proteins such as pro-MMP-1 and TIMP-1, which were present at higher amounts from the k-RasV12 ECs, while PAI-1 was comparable to EC controls. The reduced secretion of the key three basement membrane components by k-RasV12 expressing ECs (i.e. fibronectin, perlecan, nidogen-1) was observed from both sets of media from 3D gels or 2D cultures (Suppl. Figure VIII). This reduced secretion is consistent with the very strongly decreased deposition of fibronectin and perlecan in the k-RasV12 EC-pericyte co-cultures compared to control co-cultures (Figure 9A). However, the strong decrease in pericyte recruitment to these k-RasV12-expressing EC tubes (Figure 8) is also a major contributor to this reduced deposition of fibronectin and perlecan. The reduced collagen type IV deposition that is seen in the k-RasV12 EC co-cultures may also be due to reduced pericyte recruitment or the observed decrease in fibronectin deposition. Previously, we reported that blockade of fibronectin matrix assembly in EC-pericyte co-cultures strongly diminished collagen type IV deposition around EC tubes31, 44.
The reduced pericyte recruitment, the decreased pericyte responsiveness to the k-RasV12 EC tubes, and the strongly reduced pericyte-induced basement membrane deposition, leads to the conclusion that k-RasV12 expression in ECs results in an inability of these cells to properly communicate with pericytes, leading to capillary insufficiency (Figure 9C). These results coupled with the marked enhancement of EC lumen and tube formation mechanisms driven by k-RasV12 expression, further leads to enlarged and abnormal vessels with poor mural cell coverage (Figure 9C). Thus, our novel findings presented here provide key mechanistic reasons for the underlying pathogenesis of human arteriovenous malformations and their direct association with the k-RasV12 activating mutation.
DISCUSSION
In this study, we sought to elucidate the biological influence of activated k-RasV12 on the ability of ECs to undergo vascular morphogenesis and interact with mural cells such as pericytes compared to control ECs. Activating mutations in k-Ras are among the most common mutations observed within ECs in arteriovenous malformations. Our key findings demonstrate a dramatic increase in the ability of k-RasV12-expressing ECs to form lumens, leading to widened tube networks with reduced sprouting behavior as well as strongly reduced pericyte recruitment and deposition of major basement membrane components including collagen type IV, fibronectin and perlecan. Thus, ECs expressing an activating k-RasV12 mutation are unable to create functional capillary tube networks due to marked accentuation of the lumen and tube formation process coupled with reduced pericyte interactions and basement membrane assembly. Overall, the k-RasV12-expressing ECs appear to induce a state of capillary deficiency, a predisposing condition that could lead to arteriovenous malformations due to the loss of intervening capillary networks which keeps arteries and veins physically and functionally separated.
Our new studies reveal that the expression of k-RasV12 in ECs leads to marked enhancement of the EC lumen formation pathway, a process that has been investigated in detail over the past 25 years. Key steps in EC lumen formation are dramatically accelerated including intracellular vacuole formation, vacuole fusion events with the developing apical membrane surface, and lumen expansion mechanisms which involve MT1-MMP-dependent proteolysis and generation of vascular guidance tunnel spaces within the extracellular matrix. Each of these major steps in the EC lumen formation cascade are strongly accelerated by the expression of k-RasV12 compared to control ECs. The marked enhancement of lumen formation comes at the expense of EC sprouting behavior, which is reduced in these cells, so the tube networks are excessively wide and are not highly branched. The real-time movies that we show here demonstrates these marked differences in the vascular morphogenic response of control vs. k-RasV12-expressing ECs (Supplemental Movies I–VII).
Our past work has revealed critical roles for the small GTPases, Cdc42, Rac isoforms, k-Ras, and Rap1b as well as key downstream effectors including Pak2, Pak4, and Rasip1 in the lumen formation process. We have also identified critical functional roles for other key molecules including the kinases, PKCε, Src family kinases, Raf, Mek and Erk in these events as well as the cell surface membrane metalloproteinase, MT1-MMP. Additional regulators of EC lumen formation include molecules that target both intracellular vacuole membranes and the apical membrane surface including activated Src family kinases, Rac1, and caveolin-1 as well as the microtubule cytoskeleton (particularly acetylated tubulin which is subapically polarized) which directs and targets vacuoles to the apical membrane during lumen formation. Our studies here demonstrate that EC expression of k-RasV12 markedly enhances the activity or function of these known EC lumen formation regulators. For example, we observed strong increases in phosphorylation of Src, Pak, Raf, and Erk kinases from the k-RasV12 ECs compared to control ECs during lumen and tube formation, and pharmacologic inhibitors of Src family kinases, Pak, Raf and Mek kinases had strong blocking effects. Increased expression of PKCε (which activates Src kinases), and MT1-MMP were observed in these cells and pharmacologic inhibitors of these molecules also blocked lumen formation. Pharmacologic inhibitors of Cdc42 and Rac, which activate Pak kinases, gamma-secretase, which is necessary for Notch activation, and PI3 kinase also interfere with lumen formation from these ECs. Interestingly, the k-RasV12-expressing ECs were also observed to show increased expression of CDCP1, a Src phosphorylation target, a molecule upregulated in cells expressing activated Ras, and a molecule implicated in malignant cancer progression70–73. Finally, we observed robust targeting of caveolin-1 to intracellular vacuoles and the developing apical membrane surface during the accelerated lumen formation process from k-RasV12-expressing ECs. Overall, the accelerated lumen formation process from these cells appears to be occurring via well-described pathways, and it is quite straightforward to inhibit the formation of these lumens and tubes using pharmacologic agents.
The marked stimulation of EC lumen formation and the formation of the highly widened, but less branched tube networks from the k-RasV12 expressing ECs is not accompanied by increased pericyte recruitment or pericyte-induced tube maturation events. In fact, pericyte responsiveness to the accelerated EC lumens and tubes is strongly suppressed as reflected in significantly reduced pericyte numbers as well as reduced pericyte recruitment and elongation on tube surfaces. Since pericyte recruitment to and migration along the abluminal surface of EC tube networks is necessary to form capillary basement membranes, we demonstrate here that basement membrane deposition is strongly reduced, compared to controls, using the ECs carrying k-RasV12. Our previous work also demonstrated that narrow capillary tube width was controlled by pericyte recruitment and basement membrane formation, so this represents another reason why the k-RasV12-expressing EC tubes are abnormally wide in the presence of pericytes, due to the lack of pericyte recruitment and basement membrane assembly. However, the lumen formation process is markedly enhanced, including MT1-MMP-dependent lumen formation, and this metalloproteinase is known to have strong basement membrane degrading activities (e.g. a major target is collagen type IV). Thus, the lack of basement membrane deposition is likely the result of a combination of factors, including increased proteolysis and destruction of basement membranes as they are being deposited. Another important point relates to our finding of a dramatic lack of fibronectin deposition around the k-RasV12-expressing EC-lined tubes. Our past work showed the important role of EC-pericyte interactions and mechanical forces in controlling fibronectin matrix assembly31, 44, a process known to be regulated by RhoA-dependent mechanotransduction. Since Rho-dependent GTPases have inhibitory activity during EC lumen formation34, 74, the marked acceleration of lumen formation by the k-RasV12 may be associated with reduced Rho-dependent activities and contractile cell behavior, and thus, they may be less able to deposit fibronectin matrices due to an inability to exert mechanical forces during these processes. This possibility needs to be investigated in future work.
Based on the new mechanistic information obtained in this study, an important future direction is to try and repair the defects exhibited by the k-RasV12-expressing ECs, which would restore their ability to create functional capillary networks. For example, a potential approach would be to reduce EC lumen formation and at same time enhance pericyte recruitment and basement membrane deposition. The detailed model system that we describe would allow us to screen for pharmacologic agents, growth factors/peptides, or other factors/agents that might repair these capillary formative defects. A different strategy would be to induce regression of these abnormal vascular structures without affecting the adjacent normal vasculature. Recent work has been focused on the molecular basis for capillary regression both in terms of identifying key growth factors that induce regression, but also on ways to block regression by antagonizing specific pro-regressive factors, and pharmacologic agents that antagonize the underlying pro-regressive signaling pathways75. One important question is whether the k-RasV12-expressing EC tube structures are susceptible to pro-regressive factors like normal capillary tube networks. Finally, one of the important issues clinically with arteriovenous malformations is their tendency to bleed which can cause major problems, such as severe hemorrhage or stroke (e.g. when the lesions are present in the brain). The reasons why these abnormal vessels are prone to hemorrhage is not well understood, although the widened vessels with poor mural cell coverage and reduced basement membrane would be expected to be less stable than their normal counterparts. This is clearly a very important issue that needs to be investigated in detail in future studies. We think the model system presented here should be very useful to investigate such key questions underlying the pathogenesis and clinical issues that occur in patients with arteriovenous malformations.
Supplementary Material
HIGHLIGHTS.
Expression of the activating k-Ras mutation, k-RasV12, in human ECs leads to marked stimulation of the lumen formation process.
The accelerated lumen formation is caused by dramatic increases in EC intracellular vacuole formation and fusion as well as lumen expansion mechanisms mediated by MT1-MMP.
The k-RasV12 ECs also exhibit enhanced intracellular signaling pathways that favor lumen formation including marked increases in Src, Pak, Raf, Erk, and Akt phosphorylation, and tubulin acetylation.
The excessive lumen and tube formation is accompanied by strong decreases in pericyte recruitment and basement membrane matrix deposition.
EC expression of k-RasV12 leads to a capillary deficiency state that predisposes to the development of arteriovenous malformations.
Acknowledgments
Sources of funding: This work was supported by NIH grants HL149748 and HL126518 (GED).
ABBREVIATIONS
Non-standard Abbreviations and Acronyms
- HUVECs
Human umbilical vein endothelial cells
- HBVP
Human brain vascular pericytes
- VEGF
Vascular endothelial growth factor
- FGF
Fibroblast growth factor
- SCF
Stem cell factor
- IL-3
Interleukin-3
- SDF-1α
Stromal derived factor-1 alpha
- PDGF
Platelet derived growth factor
- HB-EGF
Heparin binding epidermal growth factor
- ECM
Extracellular matrix
- CCM
Cerebral cavernous malformation
- ET-1
Endothelin-1
- RT-PCR
Reverse transcription polymerase chain reaction
- 3D
3-Dimensional
- MT1-MMP
Membrane-type 1 matrix metalloproteinase
- TBST
Tris-buffered saline with Tween 20
- TBS
Tris-buffered saline
- GSK3β
Glycogen synthase kinase 3 beta
- Dusp6
Dual specificity phosphatase 6
- CDCP1
CUB domain containing protein 1
- Cdc42
Cell division cycle 42
- Rac1
Rac family small GTPase 1
- Pak2
P21 (RAC1) activated kinase 2
- Mek kinase
Mitogen-activated protein kinase kinase
- GNAQ
G protein subunit alpha q
- Rasa1
RAS P21 protein activator 1
- RalA
RAS Like Proto-Oncogene A
- IQGAP-1
IQ motif containing GTPase activating protein 1
- MRCKβ
Myotonic dystrophy kinase-related Cdc42-binding kinases
- GIT1
GIT ArfGAP 1
- βPIX
Rho guanine nucleotide exchange factor 7
- Rasip1
Ras interacting protein 1
- PKCε
Protein kinase C epsilon
- ALK-5
Activin receptor-like kinase 5
- EB-1
End-binding protein 1
- Clasp-1
Cytoplasmic linker protein 1
- CCM2L
CCM2-like protein
- Erk
Extracellular-signal related kinase
- Tie2
Tek receptor tyrosine kinase
- PIK3CA
Phosphatidylinositol-4,5 bisphosphate 3-kinase catalytic subunit alpha
- b-Raf
v-raf murine sarcoma viral oncogene homolog B1
- TIMP-1
Tissue inhibitor of metalloproteinases-1
- PAI-1
Plasminogen activator inhibitor-1
- Pro-MMP-1
Pro-Matrix Metalloproteinase-1
Footnotes
Disclosures: The authors declare no competing financial interests related to these studies.
REFERENCES
- 1.Wetzel-Strong SE, Detter MR, Marchuk DA. The pathobiology of vascular malformations: Insights from human and model organism genetics. J Pathol. 2017;241:281–293 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Queisser A, Seront E, Boon LM, Vikkula M. Genetic basis and therapies for vascular anomalies. Circ Res. 2021;129:155–173 [DOI] [PubMed] [Google Scholar]
- 3.Snellings DA, Hong CC, Ren AA, Lopez-Ramirez MA, Girard R, Srinath A, Marchuk DA, Ginsberg MH, Awad IA, Kahn ML. Cerebral cavernous malformation: From mechanism to therapy. Circ Res. 2021;129:195–215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cleaver O Mouse models of vascular development and disease. Curr Opin Hematol. 2021;28:179–188 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Dekeuleneer V, Seront E, Van Damme A, Boon LM, Vikkula M. Theranostic advances in vascular malformations. J Invest Dermatol. 2020;140:756–763 [DOI] [PubMed] [Google Scholar]
- 6.Bichsel C, Bischoff J. A somatic missense mutation in gnaq causes capillary malformation. Curr Opin Hematol. 2019;26:179–184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Greene AK, Goss JA. Vascular anomalies: From a clinicohistologic to a genetic framework. Plast Reconstr Surg. 2018;141:709e–717e [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ayturk UM, Couto JA, Hann S, Mulliken JB, Williams KL, Huang AY, Fishman SJ, Boyd TK, Kozakewich HPW, Bischoff J, Greene AK, Warman ML. Somatic activating mutations in gnaq and gna11 are associated with congenital hemangioma. Am J Hum Genet. 2016;98:1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Greenberger S, Bischoff J. Pathogenesis of infantile haemangioma. Br J Dermatol. 2013;169:12–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Boscolo E, Bischoff J. Vasculogenesis in infantile hemangioma. Angiogenesis. 2009;12:197–207 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Shirley MD, Tang H, Gallione CJ, Baugher JD, Frelin LP, Cohen B, North PE, Marchuk DA, Comi AM, Pevsner J. Sturge-weber syndrome and port-wine stains caused by somatic mutation in gnaq. N Engl J Med. 2013;368:1971–1979 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Nikolaev SI, Vetiska S, Bonilla X, Boudreau E, Jauhiainen S, Rezai Jahromi B, Khyzha N, DiStefano PV, Suutarinen S, Kiehl TR, Mendes Pereira V, Herman AM, Krings T, Andrade-Barazarte H, Tung T, Valiante T, Zadeh G, Tymianski M, Rauramaa T, Yla-Herttuala S, Wythe JD, Antonarakis SE, Frosen J, Fish JE, Radovanovic I. Somatic activating kras mutations in arteriovenous malformations of the brain. N Engl J Med. 2018;378:250–261 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Fish JE, Flores Suarez CP, Boudreau E, Herman AM, Gutierrez MC, Gustafson D, DiStefano PV, Cui M, Chen Z, De Ruiz KB, Schexnayder TS, Ward CS, Radovanovic I, Wythe JD. Somatic gain of kras function in the endothelium is sufficient to cause vascular malformations that require mek but not pi3k signaling. Circ Res. 2020;127:727–743 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Al-Olabi L, Polubothu S, Dowsett K, Andrews KA, Stadnik P, Joseph AP, Knox R, Pittman A, Clark G, Baird W, Bulstrode N, Glover M, Gordon K, Hargrave D, Huson SM, Jacques TS, James G, Kondolf H, Kangesu L, Keppler-Noreuil KM, Khan A, Lindhurst MJ, Lipson M, Mansour S, O’Hara J, Mahon C, Mosica A, Moss C, Murthy A, Ong J, Parker VE, Riviere JB, Sapp JC, Sebire NJ, Shah R, Sivakumar B, Thomas A, Virasami A, Waelchli R, Zeng Z, Biesecker LG, Barnacle A, Topf M, Semple RK, Patton EE, Kinsler VA. Mosaic ras/mapk variants cause sporadic vascular malformations which respond to targeted therapy. J Clin Invest. 2018;128:1496–1508 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lapinski PE, Doosti A, Salato V, North P, Burrows PE, King PD. Somatic second hit mutation of rasa1 in vascular endothelial cells in capillary malformation-arteriovenous malformation. Eur J Med Genet. 2018;61:11–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Cai Y, Schrenk S, Goines J, Davis GE, Boscolo E. Constitutive active mutant tie2 induces enlarged vascular lumen formation with loss of apico-basal polarity and pericyte recruitment. Sci Rep. 2019;9:12352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Natynki M, Kangas J, Miinalainen I, Sormunen R, Pietila R, Soblet J, Boon LM, Vikkula M, Limaye N, Eklund L. Common and specific effects of tie2 mutations causing venous malformations. Hum Mol Genet. 2015;24:6374–6389 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Limaye N, Kangas J, Mendola A, Godfraind C, Schlogel MJ, Helaers R, Eklund L, Boon LM, Vikkula M. Somatic activating pik3ca mutations cause venous malformation. Am J Hum Genet. 2015;97:914–921 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Revencu N, Boon LM, Mendola A, Cordisco MR, Dubois J, Clapuyt P, Hammer F, Amor DJ, Irvine AD, Baselga E, Dompmartin A, Syed S, Martin-Santiago A, Ades L, Collins F, Smith J, Sandaradura S, Barrio VR, Burrows PE, Blei F, Cozzolino M, Brunetti-Pierri N, Vicente A, Abramowicz M, Desir J, Vilain C, Chung WK, Wilson A, Gardiner CA, Dwight Y, Lord DJ, Fishman L, Cytrynbaum C, Chamlin S, Ghali F, Gilaberte Y, Joss S, Boente Mdel C, Leaute-Labreze C, Delrue MA, Bayliss S, Martorell L, Gonzalez-Ensenat MA, Mazereeuw-Hautier J, O’Donnell B, Bessis D, Pyeritz RE, Salhi A, Tan OT, Wargon O, Mulliken JB, Vikkula M. Rasa1 mutations and associated phenotypes in 68 families with capillary malformation-arteriovenous malformation. Hum Mutat. 2013;34:1632–1641 [DOI] [PubMed] [Google Scholar]
- 20.Burrows PE, Gonzalez-Garay ML, Rasmussen JC, Aldrich MB, Guilliod R, Maus EA, Fife CE, Kwon S, Lapinski PE, King PD, Sevick-Muraca EM. Lymphatic abnormalities are associated with rasa1 gene mutations in mouse and man. Proc Natl Acad Sci U S A. 2013;110:8621–8626 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Limaye N, Wouters V, Uebelhoer M, Tuominen M, Wirkkala R, Mulliken JB, Eklund L, Boon LM, Vikkula M. Somatic mutations in angiopoietin receptor gene tek cause solitary and multiple sporadic venous malformations. Nat Genet. 2009;41:118–124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Detter MR, Snellings DA, Marchuk DA. Cerebral cavernous malformations develop through clonal expansion of mutant endothelial cells. Circ Res. 2018;123:1143–1151 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Boscolo E, Limaye N, Huang L, Kang KT, Soblet J, Uebelhoer M, Mendola A, Natynki M, Seront E, Dupont S, Hammer J, Legrand C, Brugnara C, Eklund L, Vikkula M, Bischoff J, Boon LM. Rapamycin improves tie2-mutated venous malformation in murine model and human subjects. J Clin Invest. 2015;125:3491–3504 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Rauen KA. The rasopathies. Annu Rev Genomics Hum Genet. 2013;14:355–369 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ren AA, Snellings DA, Su YS, Hong CC, Castro M, Tang AT, Detter MR, Hobson N, Girard R, Romanos S, Lightle R, Moore T, Shenkar R, Benavides C, Beaman MM, Muller-Fielitz H, Chen M, Mericko P, Yang J, Sung DC, Lawton MT, Ruppert JM, Schwaninger M, Korbelin J, Potente M, Awad IA, Marchuk DA, Kahn ML. Pik3ca and ccm mutations fuel cavernomas through a cancer-like mechanism. Nature. 2021;594:271–276 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Boon LM, Mulliken JB, Vikkula M. Rasa1: Variable phenotype with capillary and arteriovenous malformations. Curr Opin Genet Dev. 2005;15:265–269 [DOI] [PubMed] [Google Scholar]
- 27.Norden PR, Kim DJ, Barry DM, Cleaver OB, Davis GE. Cdc42 and k-ras control endothelial tubulogenesis through apical membrane and cytoskeletal polarization: Novel stimulatory roles for gtpase effectors, the small gtpases, rac2 and rap1b, and inhibitory influence of arhgap31 and rasa1. PLoS One. 2016;11:e0147758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Davis GE, Stratman AN, Sacharidou A, Koh W. Molecular basis for endothelial lumen formation and tubulogenesis during vasculogenesis and angiogenic sprouting. Int Rev Cell Mol Biol. 2011;288:101–165 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Iruela-Arispe ML, Davis GE. Cellular and molecular mechanisms of vascular lumen formation. Dev Cell. 2009;16:222–231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Davis GE, Norden PR, Bowers SL. Molecular control of capillary morphogenesis and maturation by recognition and remodeling of the extracellular matrix: Functional roles of endothelial cells and pericytes in health and disease. Connect Tissue Res. 2015;56:392–402 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Bowers SL, Norden PR, Davis GE. Molecular signaling pathways controlling vascular tube morphogenesis and pericyte-induced tube maturation in 3d extracellular matrices. Adv Pharmacol. 2016;77:241–280 [DOI] [PubMed] [Google Scholar]
- 32.Xu K, Cleaver O. Tubulogenesis during blood vessel formation. Semin Cell Dev Biol. 2011;22:993–1004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Davis GE, Camarillo CW. An alpha 2 beta 1 integrin-dependent pinocytic mechanism involving intracellular vacuole formation and coalescence regulates capillary lumen and tube formation in three-dimensional collagen matrix. Exp Cell Res. 1996;224:39–51 [DOI] [PubMed] [Google Scholar]
- 34.Bayless KJ, Davis GE. The cdc42 and rac1 gtpases are required for capillary lumen formation in three-dimensional extracellular matrices. J Cell Sci. 2002;115:1123–1136 [DOI] [PubMed] [Google Scholar]
- 35.Davis GE, Bayless KJ, Mavila A. Molecular basis of endothelial cell morphogenesis in three-dimensional extracellular matrices. Anat Rec. 2002;268:252–275 [DOI] [PubMed] [Google Scholar]
- 36.Kamei M, Saunders WB, Bayless KJ, Dye L, Davis GE, Weinstein BM. Endothelial tubes assemble from intracellular vacuoles in vivo. Nature. 2006;442:453–456 [DOI] [PubMed] [Google Scholar]
- 37.Bayless KJ, Salazar R, Davis GE. Rgd-dependent vacuolation and lumen formation observed during endothelial cell morphogenesis in three-dimensional fibrin matrices involves the alpha(v)beta(3) and alpha(5)beta(1) integrins. Am J Pathol. 2000;156:1673–1683 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kim DJ, Martinez-Lemus LA, Davis GE. Eb1, p150glued, and clasp1 control endothelial tubulogenesis through microtubule assembly, acetylation, and apical polarization. Blood. 2013;121:3521–3530 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kim DJ, Norden PR, Salvador J, Barry DM, Bowers SLK, Cleaver O, Davis GE. Src- and fyn-dependent apical membrane trafficking events control endothelial lumen formation during vascular tube morphogenesis. PLoS One. 2017;12:e0184461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Norden PR, Sun Z, Davis GE. Control of endothelial tubulogenesis by rab and ral gtpases, and apical targeting of caveolin-1-labeled vacuoles. PLoS One. 2020;15:e0235116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Barry DM, Xu K, Meadows SM, Zheng Y, Norden PR, Davis GE, Cleaver O. Cdc42 is required for cytoskeletal support of endothelial cell adhesion during blood vessel formation in mice. Development. 2015;142:3058–3070 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Stratman AN, Saunders WB, Sacharidou A, Koh W, Fisher KE, Zawieja DC, Davis MJ, Davis GE. Endothelial cell lumen and vascular guidance tunnel formation requires mt1-mmp-dependent proteolysis in 3-dimensional collagen matrices. Blood. 2009;114:237–247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sacharidou A, Koh W, Stratman AN, Mayo AM, Fisher KE, Davis GE. Endothelial lumen signaling complexes control 3d matrix-specific tubulogenesis through interdependent cdc42- and mt1-mmp-mediated events. Blood. 2010;115:5259–5269 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Stratman AN, Malotte KM, Mahan RD, Davis MJ, Davis GE. Pericyte recruitment during vasculogenic tube assembly stimulates endothelial basement membrane matrix formation. Blood. 2009;114:5091–5101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Saunders WB, Bohnsack BL, Faske JB, Anthis NJ, Bayless KJ, Hirschi KK, Davis GE. Coregulation of vascular tube stabilization by endothelial cell timp-2 and pericyte timp-3. J Cell Biol. 2006;175:179–191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Koh W, Mahan RD, Davis GE. Cdc42- and rac1-mediated endothelial lumen formation requires pak2, pak4 and par3, and pkc-dependent signaling. J Cell Sci. 2008;121:989–1001 [DOI] [PubMed] [Google Scholar]
- 47.Koh W, Sachidanandam K, Stratman AN, Sacharidou A, Mayo AM, Murphy EA, Cheresh DA, Davis GE. Formation of endothelial lumens requires a coordinated pkcepsilon-, src-, pak- and raf-kinase-dependent signaling cascade downstream of cdc42 activation. J Cell Sci. 2009;122:1812–1822 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Xu K, Sacharidou A, Fu S, Chong DC, Skaug B, Chen ZJ, Davis GE, Cleaver O. Blood vessel tubulogenesis requires rasip1 regulation of gtpase signaling. Dev Cell. 2011;20:526–539 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Lanahan A, Zhang X, Fantin A, Zhuang Z, Rivera-Molina F, Speichinger K, Prahst C, Zhang J, Wang Y, Davis G, Toomre D, Ruhrberg C, Simons M. The neuropilin 1 cytoplasmic domain is required for vegf-a-dependent arteriogenesis. Dev Cell. 2013;25:156–168 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Whitehead KJ, Chan AC, Navankasattusas S, Koh W, London NR, Ling J, Mayo AH, Drakos SG, Jones CA, Zhu W, Marchuk DA, Davis GE, Li DY. The cerebral cavernous malformation signaling pathway promotes vascular integrity via rho gtpases. Nat Med. 2009;15:177–184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kleaveland B, Zheng X, Liu JJ, Blum Y, Tung JJ, Zou Z, Sweeney SM, Chen M, Guo L, Lu MM, Zhou D, Kitajewski J, Affolter M, Ginsberg MH, Kahn ML. Regulation of cardiovascular development and integrity by the heart of glass-cerebral cavernous malformation protein pathway. Nat Med. 2009;15:169–176 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Chan AC, Drakos SG, Ruiz OE, Smith AC, Gibson CC, Ling J, Passi SF, Stratman AN, Sacharidou A, Revelo MP, Grossmann AH, Diakos NA, Davis GE, Metzstein MM, Whitehead KJ, Li DY. Mutations in 2 distinct genetic pathways result in cerebral cavernous malformations in mice. J Clin Invest. 2011;121:1871–1881 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zheng X, Xu C, Smith AO, Stratman AN, Zou Z, Kleaveland B, Yuan L, Didiku C, Sen A, Liu X, Skuli N, Zaslavsky A, Chen M, Cheng L, Davis GE, Kahn ML. Dynamic regulation of the cerebral cavernous malformation pathway controls vascular stability and growth. Dev Cell. 2012;23:342–355 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Armulik A, Genove G, Betsholtz C. Pericytes: Developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell. 2011;21:193–215 [DOI] [PubMed] [Google Scholar]
- 55.Stratman AN, Schwindt AE, Malotte KM, Davis GE. Endothelial-derived pdgf-bb and hb-egf coordinately regulate pericyte recruitment during vasculogenic tube assembly and stabilization. Blood. 2010;116:4720–4730 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Stratman AN, Davis GE. Endothelial cell-pericyte interactions stimulate basement membrane matrix assembly: Influence on vascular tube remodeling, maturation, and stabilization. Microsc Microanal. 2012;18:68–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kemp SS, Aguera KN, Cha B, Davis GE. Defining endothelial cell-derived factors that promote pericyte recruitment and capillary network assembly. Arterioscler Thromb Vasc Biol. 2020;40:2632–2648 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bowers SLK, Kemp SS, Aguera KN, Koller GM, Forgy JC, Davis GE. Defining an upstream vegf (vascular endothelial growth factor) priming signature for downstream factor-induced endothelial cell-pericyte tube network coassembly. Arterioscler Thromb Vasc Biol. 2020;40:2891–2909 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Smith AO, Bowers SL, Stratman AN, Davis GE. Hematopoietic stem cell cytokines and fibroblast growth factor-2 stimulate human endothelial cell-pericyte tube co-assembly in 3d fibrin matrices under serum-free defined conditions. PLoS One. 2013;8:e85147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Vikkula M, Boon LM, Mulliken JB. Molecular genetics of vascular malformations. Matrix Biol. 2001;20:327–335 [DOI] [PubMed] [Google Scholar]
- 61.Uebelhoer M, Natynki M, Kangas J, Mendola A, Nguyen HL, Soblet J, Godfraind C, Boon LM, Eklund L, Limaye N, Vikkula M. Venous malformation-causative tie2 mutations mediate an akt-dependent decrease in pdgfb. Hum Mol Genet. 2013;22:3438–3448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Stratman AN, Davis MJ, Davis GE. Vegf and fgf prime vascular tube morphogenesis and sprouting directed by hematopoietic stem cell cytokines. Blood. 2011;117:3709–3719 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Salvador J, Davis GE. Evaluation and characterization of endothelial cell invasion and sprouting behavior. Methods Mol Biol. 2018;1846:249–259 [DOI] [PubMed] [Google Scholar]
- 64.Koh W, Stratman AN, Sacharidou A, Davis GE. In vitro three dimensional collagen matrix models of endothelial lumen formation during vasculogenesis and angiogenesis. Methods Enzymol. 2008;443:83–101 [DOI] [PubMed] [Google Scholar]
- 65.Alavi A, Hood JD, Frausto R, Stupack DG, Cheresh DA. Role of raf in vascular protection from distinct apoptotic stimuli. Science. 2003;301:94–96 [DOI] [PubMed] [Google Scholar]
- 66.Hoang MV, Nagy JA, Senger DR. Cdc42-mediated inhibition of gsk-3beta improves angio-architecture and lumen formation during vegf-driven pathological angiogenesis. Microvasc Res. 2011;81:34–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Bell SE, Mavila A, Salazar R, Bayless KJ, Kanagala S, Maxwell SA, Davis GE. Differential gene expression during capillary morphogenesis in 3d collagen matrices: Regulated expression of genes involved in basement membrane matrix assembly, cell cycle progression, cellular differentiation and g-protein signaling. J Cell Sci. 2001;114:2755–2773 [DOI] [PubMed] [Google Scholar]
- 68.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] [PMC free article] [PubMed] [Google Scholar]
- 69.Murphy PA, Lam MT, Wu X, Kim TN, Vartanian SM, Bollen AW, Carlson TR, Wang RA. Endothelial notch4 signaling induces hallmarks of brain arteriovenous malformations in mice. Proc Natl Acad Sci U S A. 2008;105:10901–10906 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hooper JD, Zijlstra A, Aimes RT, Liang H, Claassen GF, Tarin D, Testa JE, Quigley JP. Subtractive immunization using highly metastatic human tumor cells identifies sima135/cdcp1, a 135 kda cell surface phosphorylated glycoprotein antigen. Oncogene. 2003;22:1783–1794 [DOI] [PubMed] [Google Scholar]
- 71.Liu H, Ong SE, Badu-Nkansah K, Schindler J, White FM, Hynes RO. Cub-domain-containing protein 1 (cdcp1) activates src to promote melanoma metastasis. Proc Natl Acad Sci U S A. 2011;108:1379–1384 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Uekita T, Fujii S, Miyazawa Y, Iwakawa R, Narisawa-Saito M, Nakashima K, Tsuta K, Tsuda H, Kiyono T, Yokota J, Sakai R. Oncogenic ras/erk signaling activates cdcp1 to promote tumor invasion and metastasis. Mol Cancer Res. 2014;12:1449–1459 [DOI] [PubMed] [Google Scholar]
- 73.Martinko AJ, Truillet C, Julien O, Diaz JE, Horlbeck MA, Whiteley G, Blonder J, Weissman JS, Bandyopadhyay S, Evans MJ, Wells JA. Targeting ras-driven human cancer cells with antibodies to upregulated and essential cell-surface proteins. Elife. 2018;7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Barry DM, Koo Y, Norden PR, Wylie LA, Xu K, Wichaidit C, Azizoglu DB, Zheng Y, Cobb MH, Davis GE, Cleaver O. Rasip1-mediated rho gtpase signaling regulates blood vessel tubulogenesis via nonmuscle myosin ii. Circ Res. 2016;119:810–826 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Koller GM, Schafer C, Kemp SS, Aguera KN, Lin PK, Forgy JC, Griffin CT, Davis GE. Proinflammatory mediators, il (interleukin)-1beta, tnf (tumor necrosis factor) alpha, and thrombin directly induce capillary tube regression. Arterioscler Thromb Vasc Biol. 2020;40:365–377 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.