Phosphatidylserine (PS) asymmetry in the eukaryotic cell membrane is maintained by a group of proteins belonging to the P4-ATPase family, namely, PS flippases. The folding and transporting of P4-ATPases to their cellular destination requires a β-subunit member of the TMEM30 protein family. Loss of Tmem30a has been shown to cause multiple disease conditions. However, its roles in vascular development have not been elucidated. Here, we show that TMEM30A plays critical roles in retinal vascular angiogenesis, which is a fundamental process in vascular development. Our data indicate that knockdown of TMEM30A in primary human retinal endothelial cells led to reduced tube formation. In mice, endothelial cell (EC)-specific deletion of Tmem30a led to retarded retinal vascular development with a hyperpruned vascular network as well as blunted-end, aneurysm-like tip ECs with fewer filopodia at the vascular front and a reduced number of tip cells. Deletion of Tmem30a also impaired vessel barrier integrity. Mechanistically, deletion of TMEM30A caused reduced EC proliferation by inhibiting VEGF-induced signaling. Our findings reveal essential roles of TMEM30A in angiogenesis, providing a potential therapeutic target.

Phosphatidylserine (PS) is asymmetrically distributed in the exoplasmic and cytoplasmic leaflet of the plasma membrane (Halleck et al., 1999; Paulusma and Oude Elferink, 2005; Tang et al., 1996). Such an asymmetrical distribution of aminophospholipids is maintained by P4-ATPases called PS flippases, which are required for many biological processes, such as protein trafficking, apoptosis, neurodevelopment and sperm capacitation (Chen et al., 1999; Op den Kamp, 1979; Saito et al., 2004; Segawa et al., 2014; Seigneuret and Devaux, 1984). There are 14 P4-ATPases in the mammalian genome, and these flippases work together to maintain the phospholipid asymmetry between the outer and inner leaflets of the plasma membrane (Takatsu et al., 2017).

Mutations in several P4-ATPases have been reported to be associated with human diseases, indicating the importance of P4-ATPases. Mutations in ATP8B1 lead to progressive familial intrahepatic cholestasis and hearing loss (Bull et al., 1998; Stapelbroek et al., 2009). Mutations in ATP8B2 cause a severe neurological disorder that is characterized by cerebellar ataxia, mental retardation and disequilibrium syndrome (Cacciagli et al., 2010; Maríin-Hernández et al., 2016). Animal model experiments have provided additional evidence that P4-ATPases are critical for normal physiological function. Mice deficient in Atp11c have recently been reported to exhibit B-cell lymphopenia, anemia and intrahepatic cholestasis (Arashiki et al., 2016; Siggs et al., 2011a,b; Yabas et al., 2014; Yabas et al., 2011). Mutations in Atp8a2 result in axonal degeneration and retinal degeneration.

Proper folding and trafficking of most P4-ATPases except ATP9A and ATP9B is dependent on a β-subunit from the transmembrane 30 protein [TMEM30; also known as cell division control protein 50 (CDC50)] protein family, which includes TMEM30A, TMEM30B and TMEM30C (also known as CDC50A, CDC50B and CDC50C, respectively) (Bryde et al., 2010; Paulusma et al., 2008; Saito et al., 2004; Takatsu et al., 2011; Zhang et al., 2017). These genes encode N-glycosylated proteins with two transmembrane domains and play critical roles in the folding and function of P4-ATPases (Katoh and Katoh, 2004; van der Velden et al., 2010). Our previous studies have demonstrated the essential roles of the TMEM30A gene in retinal development, bile salt transportation and the survival of hematopoietic cells (Li et al., 2018; Liu et al., 2017; Zhang et al., 2017).

Angiogenesis is a critical process in vascular development and abnormal angiogenesis is implicated in many diseases, such as age-related macular degeneration (AMD), familial exudative vitreoretinopathy (FEVR) and cancer (Carmeliet and Jain, 2000; Folkman, 1971; Huang et al., 2016; Yang et al., 2006; Zhang et al., 2016). Angiogenesis is a complex process regulated by multiple signaling pathways. Understanding the network controlling angiogenesis is of great importance for unveiling various disease conditions (Chung and Ferrara, 2011; Potente et al., 2011). Given the essential roles of TMEM30A in organism development and no previous studies of its roles in vascular development, we set out to study the roles of TMEM30A in angiogenesis.

In the present study, we show that TMEM30A plays essential roles in the angiogenesis process. We found that knockdown of TMEM30A in primary human retinal endothelial cells (HRECs) led to impaired tube formation in vitro. Moreover, we examined retina from mice with an endothelial cell (EC)-specific deletion of mouse Tmem30a (Tmem30ai△EC) and these showed a retarded vascular extension toward the peripheral zone and deep layer, a hyperpruned vascular network with decreased tip ECs at the vascular front, and blunted-end, aneurysm-like tip ECs with fewer filopodia. By using an induced global Tmem30a knockout mouse model (Tmem30a-iKO), we also proved that deletion of Tmem30a impaired the vessel barrier integrity. We further demonstrated that TMEM30A controls EC proliferation during sprouting angiogenesis by regulating VEGF-induced signaling.

Knockdown of TMEM30A impairs vascular formation in vitro

To verify whether P4-ATPases were expressed in the vascular system, we carried out reverse transcriptional polymerase chain reaction (RT-PCR) experiments to study the expression of the 14 P4-ATPases in human HRECs obtained from Cell Systems. The results showed that several P4-ATPase-encoding genes, including ATP8B1, ATP8B2, ATP9A, ATP9B, ATP10D, ATP11A, ATP11B and ATP11C, were expressed in HRECs (Fig. 1A). The expression of multiple P4-ATPases in HRECs indicates that they could have redundant functions, making it challenging to dissect the role of each gene in angiogenesis. Proper folding of most P4-ATPases requires their β-subunit TMEM30A (Montigny et al., 2016), which is expressed in HRECs, human umbilical vein ECs (HUVECs) and human microvascular ECs (HMVECs) (Fig. 1B,C). To assess the roles of TMEM30A in HRECs, we used a lentivirus carrying TMEM30A shRNA to knockdown (KD) the expression of TMEM30A in HRECs (shTMEM30A HRECs). Analyses of TMEM30A RNA expression by RT-PCR and protein expression by western blotting showed that the expression of TMEM30A in these cells was reduced to ∼10% of that in control cells (Fig. 2A,B). Tube formation was impaired in TMEM30A-knockdown HRECs (Fig. 2C).

Fig. 1.

Expression analysis of 14 P4-ATPases and TMEM30A in human endothelial cells. (A) Analyses of the mRNA expression of 14 P4-ATPases in HRECs using an RT-PCR method. (B) RT-PCR assays revealed the expression of TMEM30A mRNA in HRECs, HUVECs and HMVECs. (C) Western blotting reveals the expression of TMEM30A protein in HRECs, HUVECs and HMVECs.

Fig. 1.

Expression analysis of 14 P4-ATPases and TMEM30A in human endothelial cells. (A) Analyses of the mRNA expression of 14 P4-ATPases in HRECs using an RT-PCR method. (B) RT-PCR assays revealed the expression of TMEM30A mRNA in HRECs, HUVECs and HMVECs. (C) Western blotting reveals the expression of TMEM30A protein in HRECs, HUVECs and HMVECs.

Fig. 2.

In vitro studies of the function of TMEM30A in tube formation. HRECs were transfected with lentivirus carrying shTMEM30A RNA (shTMEM30A HRECs) or the same amount of lentivirus carrying GFP (denoted WT controls). (A,B) RT-PCR and western blotting methods were used to detect the efficiency of shTMEM30A-mediated knockdown in HRECs. The expression level of TMEM30A in shTMEM30A HRECs was reduced to ∼10% of the level in the WT controls. Data are presented as mean±s.e.m. Sample size n=3. Data shown are from one typical experiment of three independent experiments. ***P<0.0001, two-tailed t-test. (C) Compared to WT HRECs, shTMEM30A HRECs showed reduced tube formation. Data are presented as mean±s.e.m. Data shown are from one typical experiment of three independent experiments. ***P<0.0001, two-tailed t-test.

Fig. 2.

In vitro studies of the function of TMEM30A in tube formation. HRECs were transfected with lentivirus carrying shTMEM30A RNA (shTMEM30A HRECs) or the same amount of lentivirus carrying GFP (denoted WT controls). (A,B) RT-PCR and western blotting methods were used to detect the efficiency of shTMEM30A-mediated knockdown in HRECs. The expression level of TMEM30A in shTMEM30A HRECs was reduced to ∼10% of the level in the WT controls. Data are presented as mean±s.e.m. Sample size n=3. Data shown are from one typical experiment of three independent experiments. ***P<0.0001, two-tailed t-test. (C) Compared to WT HRECs, shTMEM30A HRECs showed reduced tube formation. Data are presented as mean±s.e.m. Data shown are from one typical experiment of three independent experiments. ***P<0.0001, two-tailed t-test.

Generation of inducible vascular EC-specific deletion mice

As Tmem30a global knockout and vascular EC-specific knockout mice, when induced by Tie2-Cre (Tie2 is an endothelial cell-specific promoter), are embryonic lethal (Kisanuki et al., 2001), loss of Tmem30a might impair vasculogenesis and angiogenesis. To assess the role of Tmem30a in ECs, we crossed a Tmem30a conditional knockout allele with Pdgfb-CreER (Claxton et al., 2008) to generate an inducible vascular EC-specific knockout line. In Pdgfb-CreER mice, the expression of an improved Cre gene fused to ERT2 is controlled by the mouse Pdgfb promoter. Mice homozygous for the Tmem30a floxed allele (Tmem30aloxp/loxp) were mated to Tmem30aloxp/+ Pdgfb-CreER mice to generate Tmem30aloxp/loxp Pdgfb-CreER (named Tmem30aiΔEC) and Tmem30aloxp/loxp mice (WT), as shown in Fig. 3A. PCR methods were used to identify the genotypes of the offspring (Fig. 3B). Tmem30aiΔEC and WT mice were induced by intraperitoneal injection of tamoxifen daily starting at post-natal day (P)2 for 3 consecutive days. The specificity and efficacy of Cre-mediated recombination were assessed by means of the tdTomato reporter. To do this, we crossed ROSA26-tdTomato reporter mice with Tmem30aloxp/loxp Pdgfb-CreER mice to generate Tmem30aloxp/loxp Pdgfb-CreER Rosa-tdTomato mice. In the presence of the Cre enzyme, the stop codon before the tdTomato expression cassette was removed, and red fluorescent protein (RFP) is expressed in Cre-positive cells. As shown in Fig. 3C, after induction by tamoxifen, the expression of RFP was restricted to vascular ECs in Tmem30aiΔEC mice, here examined in the retina, and was absent in WT mice, indicating the specificity of this Cre line.

Fig. 3.

Generation of the knockout mouse models used in this study. (A) Scheme showing the targeting strategy for generating the Tmem30ai△EC and Tmem30a-iKO mice. Mice were intraperitoneally injected with a daily dosage of 25 mg/kg body weight Tamoxifen for 3 consecutive days from P2, as shown on the right. (B) Genotyping of the mouse models using PCR methods. (C) The efficiency and specificity of the induced EC-specific deletion of Tmem30a were monitored through ROSA-tdTomato expression (red). This was achieved by crossing ROSA26-tdTomato reporter mice with Tmem30aloxp/loxp Pdgfb-CreER mice to generate Tmem30aloxp/loxp Pdgfb-CreER Rosa-tdTomato mice. In the presence of the Cre enzyme, the stop codon before the tdTomato expression cassette is removed, and RFP is expressed in Cre-positive cells. As can be seen in the images, after induction by tamoxifen, the expression of RFP was restricted to vascular ECs in Tmem30aiΔEC mice, here examined in retina, indicating the specificity of this Cre line. Scale bars: 100 μm.

Fig. 3.

Generation of the knockout mouse models used in this study. (A) Scheme showing the targeting strategy for generating the Tmem30ai△EC and Tmem30a-iKO mice. Mice were intraperitoneally injected with a daily dosage of 25 mg/kg body weight Tamoxifen for 3 consecutive days from P2, as shown on the right. (B) Genotyping of the mouse models using PCR methods. (C) The efficiency and specificity of the induced EC-specific deletion of Tmem30a were monitored through ROSA-tdTomato expression (red). This was achieved by crossing ROSA26-tdTomato reporter mice with Tmem30aloxp/loxp Pdgfb-CreER mice to generate Tmem30aloxp/loxp Pdgfb-CreER Rosa-tdTomato mice. In the presence of the Cre enzyme, the stop codon before the tdTomato expression cassette is removed, and RFP is expressed in Cre-positive cells. As can be seen in the images, after induction by tamoxifen, the expression of RFP was restricted to vascular ECs in Tmem30aiΔEC mice, here examined in retina, indicating the specificity of this Cre line. Scale bars: 100 μm.

Deletion of Tmem30a impairs angiogenesis in mice retina

The fact that knockdown of TMEM30A in HRECs led to impaired tube formation in vitro (see above) promoted us to investigate the roles of Tmem30a in angiogenesis in mouse retinas. Mouse retinas are avascular at birth, and a single, superficial layer of blood vessels grows progressively from the center toward the periphery from P1 until P7. Then, the superficial blood vessels grow into the deep retinal layers. Retina whole-mount staining revealed that growth of the superficial vasculature toward the periphery zone in Tmem30aiΔEC mouse retinas was delayed at P7 and exhibited a hyperpruned vascular network compared with those of WT mice (Fig. 4A–D). These results indicate that endothelial Tmem30a promoted angiogenesis sprouting and vascular network formation in the retina during the early postnatal period. Strikingly, at the frontier of the vascular network, the number of tip ECs was reduced dramatically and these exhibited a blunt-end, aneurysm-like structure with fewer and malformed filopodia compared with those of WT mice (Fig. 4E–G). To further characterize the retinal vascular phenotypes of the Tmem30aiΔEC mice, we performed an immunofluorescence assay on frozen sections of P9 retinas. In the WT mouse retinas, the vascular branches were established in the outer plexiform layer (OPL) and inner plexiform layer (IPL) at P9, whereas vascular branches were found only in the ganglion cell layer of the Tmem30aiΔEC mouse retinas, indicating a delayed vascular development toward the deep layers in the retina (Fig. S1).

Fig. 4.

Tmem30a is essential for sprouting angiogenesis and vessel barrier integrity in mouse retinas. (A) Tmem30ai△EC, Tmem30a-iKO and WT mice were intraperitoneally injected with tamoxifen at P2 to P4 and analyzed at P7. (B–D) Images of retinal vessels stained by IB4 and comparisons of the indicated parameters in WT, Tmem30ai△EC and Tmem30a-iKO mice (n=4, each group). A quantification of vascular density and vascular progression at P9 is shown in D. Vascular density was measured as the IB4-positive retinal vessel area divided by the total measured area of the retina and is presented as a percentage by means of AngioTool (Zudaire et al., 2011). ***P<0.001, Student's t-test. (E–G) Images showing reduced number of tip ECs and filopodia in retinal vessels of Tmem30ai△EC and Tmem30a-iKO mice (n=4, each group). The number of tip cells and filopodia was counted in each genotypes and is plotted in G. (H) Red blood cells stained with Ter119 antibody show that there is vessel leakage in Tmem30a-iKO mouse retinas, but not in WT or Tmem30ai△EC mouse retinas. Data in D and G are presented as mean±s.e.m. ***P<0.0001; ns, not significant, two-tailed t-test. Scale bars: 250 μm (B); 100 μm (C); 50 μm (E); 25 μm (F,H).

Fig. 4.

Tmem30a is essential for sprouting angiogenesis and vessel barrier integrity in mouse retinas. (A) Tmem30ai△EC, Tmem30a-iKO and WT mice were intraperitoneally injected with tamoxifen at P2 to P4 and analyzed at P7. (B–D) Images of retinal vessels stained by IB4 and comparisons of the indicated parameters in WT, Tmem30ai△EC and Tmem30a-iKO mice (n=4, each group). A quantification of vascular density and vascular progression at P9 is shown in D. Vascular density was measured as the IB4-positive retinal vessel area divided by the total measured area of the retina and is presented as a percentage by means of AngioTool (Zudaire et al., 2011). ***P<0.001, Student's t-test. (E–G) Images showing reduced number of tip ECs and filopodia in retinal vessels of Tmem30ai△EC and Tmem30a-iKO mice (n=4, each group). The number of tip cells and filopodia was counted in each genotypes and is plotted in G. (H) Red blood cells stained with Ter119 antibody show that there is vessel leakage in Tmem30a-iKO mouse retinas, but not in WT or Tmem30ai△EC mouse retinas. Data in D and G are presented as mean±s.e.m. ***P<0.0001; ns, not significant, two-tailed t-test. Scale bars: 250 μm (B); 100 μm (C); 50 μm (E); 25 μm (F,H).

Our studies using the induced EC-specific deletion of Tmem30a mouse model (Tmem30aiΔEC mice) demonstrated that loss of Tmem30a in ECs impaired the retinal vascular angiogenesis process. However, other cells, such as pericytes, are also indispensable during maturation of the blood vasculature. To study the global roles of Tmem30a in angiogenesis process, we generated induced global Tmem30a knockout mice (Tmem30a-iKO) by crossing Tmem30aloxP/loxP mice with Tmem30aloxP/loxP CAG-Cre mice (Fig. 3A,B). The Tmem30a-iKO mice and their littermates without CAG-Cre were injected intraperitoneally with tamoxifen at P2 to P4 for 3 consecutive days. Retinal whole mounts were analyzed by staining with isolectin B4 (IB4) at P7. Angiogenesis in Tmem30a-iKO mice was also impaired, similar to that in Tmem30aiΔEC mice (Fig. 4A–G). Staining of red blood cells using Ter119 antibody revealed blood vessel leakage in the Tmem30a-iKO retinas (Fig. 4H). Taken together, our results demonstrate that Tmem30a mainly contributes to vascular sprouting in ECs, while in cells other than ECs, Tmem30a plays an important role in vessel barrier integrity.

The above results indicate that Tmem30a plays critical roles in the angiogenesis process, while its role in maintaining the vascular network still needs to be elucidated. To this end, we started to administer tamoxifen injections to mice when the retinal vascular was mature, specifically, from P21 for 3 consecutive days. Then, the retinal whole mounts were analyzed by IB4 staining at P30. Our results revealed that the retinal vascular density of Tmem30a-iKO mice did not change compared to that of the WT mice (Fig. 5A,B).

Fig. 5.

Deletion of Tmem30a from P21 onwards does not affect retinal vascular maintenance. (A,B) IB4 (green)-stained retinal whole mounts from P30 Tmem30a-iKO and WT mice. Scale bars: 250 μm (upper panel), 100 μm (lower panel). The vascular density (calculated as in Fig. 4D) and vascular length (radial length of retinal vessels measured as the distance from the optic nerve head to the peripheral vascular front in each leaflet of the retina). Data are presented as mean±s.e.m. Sample size n=3 for each group. Data shown is from one typical experiment of three independent experiments. ns, not significant, two-tailed t-test.

Fig. 5.

Deletion of Tmem30a from P21 onwards does not affect retinal vascular maintenance. (A,B) IB4 (green)-stained retinal whole mounts from P30 Tmem30a-iKO and WT mice. Scale bars: 250 μm (upper panel), 100 μm (lower panel). The vascular density (calculated as in Fig. 4D) and vascular length (radial length of retinal vessels measured as the distance from the optic nerve head to the peripheral vascular front in each leaflet of the retina). Data are presented as mean±s.e.m. Sample size n=3 for each group. Data shown is from one typical experiment of three independent experiments. ns, not significant, two-tailed t-test.

Deletion of Tmem30a inhibiting EC proliferation and disturbing VEGF-induced signaling

To obtain mechanical insights into how TMEM30A regulates angiogenesis, we performed a series of in vitro and in vivo analyses in HRECs and Tmem30a-iKO mice. As the tip ECs numbers and vascular density and branches in Tmem30aiΔEC mice were reduced dramatically, we first considered whether apoptosis had occurred during angiogenesis. However, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assays on the whole-mount retinas from Tmem30aiΔEC mice at P7 did not show any signs of apoptosis (Fig. S2). In addition, as shown in Fig. 5, the vascular density did not decrease when Tmem30a was removed from the mature retinal vascular, verifying that apoptosis was not ongoing in the ECs. However, the question that then arises is what causes the retarded vascular angiogenesis and reduced tip ECs upon Tmem30a deletion? Because VEGF is a key growth factor in angiogenesis, we stained retinal whole mounts from Tmem30a-iKO mice with anti-VEGFa antibody. There was no difference in the distribution of VEGFa that surrounded tip ECs between WT and Tmem30a-iKO retinas, indicating that VEGFa secretion was not disturbed when Tmem30a was removed (Fig. 6A). Furthermore, we labeled the proliferating ECs with 5-ethynyl-2'-deoxyuridine (EDU) to assess cell proliferation. The number of EDU-positive ECs in Tmem30a-iKO mouse retinas were reduced dramatically (Fig. 6B). To unveil the reasons behind this finding, we first performed RNA sequencing (RNA-seq) analysis on total RNAs extracted from the WT and HRECs shTMEM30A HRECs. Statistical analyses of the RNA-seq results revealed that mRNA expression of the genes involved in the cell cycle (CCNA1, CCNB1, CCND1, CCDN3, CDC6, DBF4 ESPL1, PCNA1 and STAG1) was significantly downregulated in the shTMEM30A HRECs (Fig. 7A). These RNA-seq results were verified by real-time quantitative PCR (RT-qPCR) (Fig. 7B). Next, we also assessed the expression of those genes in the lung of Tmem30a-iKO mice by RT-qPCR analysis. Similar to the results in HRECs, the expression of these cell-cycle-related genes was also downregulated in the lungs of Tmem30a-iKO mice at P7 (Fig. 7C). Therefore, based on the RNA expression results, we propose that the main reason for the impaired angiogenesis after deletion of Tmem30a is compromised proliferation of ECs.

Fig. 6.

Deletion of Tmem30a impairs EC proliferation without affecting VEGFa secretion. (A) Retinal whole mounts from WT and Tmem30a-iKO mice at P7 were stained with IB4 (red), VEGFa (green) and DAPI (blue). Scale bar: 25 μm. (B) EDU labeling (green) of retinal whole mounts from WT and Tmem30a-iKO mice at P7 and the corresponding statistical results. Data are presented as mean±s.e.m. Sample size n=5. Data shown are from one typical experiment of three independent experiments. *P<0.05, two-tailed t-test. Scale bar: 50 μm.

Fig. 6.

Deletion of Tmem30a impairs EC proliferation without affecting VEGFa secretion. (A) Retinal whole mounts from WT and Tmem30a-iKO mice at P7 were stained with IB4 (red), VEGFa (green) and DAPI (blue). Scale bar: 25 μm. (B) EDU labeling (green) of retinal whole mounts from WT and Tmem30a-iKO mice at P7 and the corresponding statistical results. Data are presented as mean±s.e.m. Sample size n=5. Data shown are from one typical experiment of three independent experiments. *P<0.05, two-tailed t-test. Scale bar: 50 μm.

Fig. 7.

RNA expression analysis shows the expression of several genes involved in cell proliferation is downregulated when TMEM30A was removed. (A) RNA-seq results showing that several cell-proliferation-related genes are downregulated when TMEM30A expression was knocked down in HRECs. (B) RT-qPCR verified the RNA-seq results in HRECs for the indicated genes. Data are presented as mean±s.e.m. Sample size n=4. Data shown was from one typical experiment of three independent experiments. ***P<0.0001, two-tailed t-test. (C) RT-qPCR verified that the expression of several cell-proliferation-related genes was downregulated in the lungs of Tmem30a-iKO mice. Data are presented as mean±s.e.m. Sample size n=3. Data shown is from one typical experiment of three independent experiments. ***P<0.0001; **P<0.001; ns, not significant, two-tailed t-test.

Fig. 7.

RNA expression analysis shows the expression of several genes involved in cell proliferation is downregulated when TMEM30A was removed. (A) RNA-seq results showing that several cell-proliferation-related genes are downregulated when TMEM30A expression was knocked down in HRECs. (B) RT-qPCR verified the RNA-seq results in HRECs for the indicated genes. Data are presented as mean±s.e.m. Sample size n=4. Data shown was from one typical experiment of three independent experiments. ***P<0.0001, two-tailed t-test. (C) RT-qPCR verified that the expression of several cell-proliferation-related genes was downregulated in the lungs of Tmem30a-iKO mice. Data are presented as mean±s.e.m. Sample size n=3. Data shown is from one typical experiment of three independent experiments. ***P<0.0001; **P<0.001; ns, not significant, two-tailed t-test.

These findings prompted us to ask another question: how does Tmem30a regulate EC proliferation? VEGF is a key player governing sprouting angiogenesis. VEGF ligands bind to VEGFR2 receptors to stimulate endothelial stalk cells to differentiate to endothelial tip cells, which is a critical initial step in angiogenesis. Based on the fact that the number of retinal endothelial tip cells was greatly reduced in Tmem30a deletion mice, we inferred that the VEGF-induced signaling was impaired. We performed western blot analyses on the phosphorylation status of VEGFR2, an indicator of VEGF signaling pathway activity. In both the shTMEM30A HRECs and the lungs of Tmem30a-iKO mice (at P7), the Tyr1175 phosphorylation level of VEGFR2 was reduced compared to that of the WT controls (Fig. 8A,B). We further examined the phosphorylation of other proteins involved in VEGF induced signaling. Phosphorylation of PLCγ1, SRC, ERK1 and ERK2 (ERK1/2, also known as MAPK3 and MAPK1, respectively), p38 MAPK family proteins (α, β and γ forms; also known as MAPK14, MAPK11 and MAPK12, respectively) and MEK1 and MEK2 (MEK1/2, also known as MAP2K1 and MAP2K2, respectively) were also downregulated in shTMEM30A HRECs and in the lung tissue of Tmem30a-iKO mice (Fig. 8A,B). The phosphorylation of these proteins play critical roles in the angiogenesis process, such as cell proliferation and cell migration (Olsson et al., 2006). However, the phosphorylation level of AKT1 (hereafter AKT), which is responsible for ECs survival, seemed to be unchanged when Tmem30a was removed (Fig. S2) (Koch et al., 2011). Consistent with this observation, no obvious apoptosis was observed in Tmem30aiΔEC mice (Fig. S3A). An ER stress assay using anti-CHOP antibody (CHOP is also known as DDIT3) also did not reveal any difference between WT and Tmem30a-iKO samples (Fig. S3B). Taken together, we demonstrated that deletion of TMEM30A led to an extensive disturbance on the activity of VEGF-induced signaling and further impaired the angiogenesis process.

Fig. 8.

Western blot analysis on the phosphorylation status of several proteins involved in the VEGF-induced signaling. (A) The phosphorylation level of VEGFR2, PLCγ1, SRC, MEK1/2, ERK1/2 and p38 MAPK proteins was decreased when TMEM30A was removed from HRECs. GAPDH was used as the loading control. Data are presented as mean±s.e.m. Sample size n=3. Data shown are from one typical experiment of three independent experiments. ***P<0.0001; **P<0.001; ns, not significant, two-tailed t-test. (B) Western blot analysis showing that the phosphorylation of VEGFR2, PLCγ1, SRC, MEK1/2, ERK1/2 and p38 MAPK proteins was downregulated in the lungs from Tmem30a-iKO mice. GAPDH was used as the loading control. Data are presented as mean±s.e.m. Sample size n=3. Data shown are from one typical experiment of three independent experiments. ***P<0.0001; **P<0.001; *P<0.05; ns, not significant, two-tailed t-test.

Fig. 8.

Western blot analysis on the phosphorylation status of several proteins involved in the VEGF-induced signaling. (A) The phosphorylation level of VEGFR2, PLCγ1, SRC, MEK1/2, ERK1/2 and p38 MAPK proteins was decreased when TMEM30A was removed from HRECs. GAPDH was used as the loading control. Data are presented as mean±s.e.m. Sample size n=3. Data shown are from one typical experiment of three independent experiments. ***P<0.0001; **P<0.001; ns, not significant, two-tailed t-test. (B) Western blot analysis showing that the phosphorylation of VEGFR2, PLCγ1, SRC, MEK1/2, ERK1/2 and p38 MAPK proteins was downregulated in the lungs from Tmem30a-iKO mice. GAPDH was used as the loading control. Data are presented as mean±s.e.m. Sample size n=3. Data shown are from one typical experiment of three independent experiments. ***P<0.0001; **P<0.001; *P<0.05; ns, not significant, two-tailed t-test.

In this study, we uncovered the important roles of TMEM30A, the β-subunit of P4-ATPases, in the sprouting angiogenesis process. P4-ATPases are members of a family of proteins that transport aminophospholipids from the exoplasmic to the cytoplasmic leaflet of cell membranes by utilizing ATP, thereby maintaining the aminophospholipid asymmetry in the cellular membrane (Halleck et al., 1999; Holthuis and Levine, 2005). Several P4-ATPases have been reported to have important functions in physiological and pathological conditions (Bull et al., 1998; Cacciagli et al., 2010; Darland-Ransom et al., 2008; Wang et al., 2004; Liu et al., 2017; Zhang et al., 2017). By using RT-PCR, we demonstrated that eight of 14 P4-ATPases are expressed in HRECs (Fig. 1A), indicating that they might have redundant roles, which would make it difficult to study their functions. To this end, we focused on the β-subunit of these P4-ATPases, namely TMEM30A. TMEM30A is expressed in three primary EC lines, as revealed by RT-PCR and western blotting methods (Fig. 1B,C). Defective tube formation of shTMEM30A-treated HRECs in vitro prompted us to assess what would happen when Tmem30a was deleted from ECs in a genetically modified mouse model. As constant deletion of Tmem30a in ECs when induced by Tie2-Cre is prenatally lethal (Table S2), we generated a tamoxifen-inducible endothelial cell-specific deletion model (Tmem30ai△EC) using PDGFb-Cre. Deletion of Tmem30a in ECs resulted in retarded vascular sprouting, a greatly reduced number of tip ECs, and blunt-end, aneurysm-like structures with fewer and malformed filopodia in the retinal vessels (Fig. 4). EDU labeling also revealed that there was reduced EC proliferation in this model system (Fig. 6).

To uncover the mechanisms underlying these phenotypes, we carried out a series of RNA-seq, RT-qPCR and western blotting experiments. Our RNA-seq and RT-qPCR analysis showed that mRNA expression of those genes involved in the cell cycle (CCNA1, CCNB1, CCND1, CCDN3, CDC6, DBF4, ESPL1, PCNA1 and STAG1) was reduced in TMEM30A-knockdown HRECs and Tmem30a-iKO lung tissues (Fig. 7). Further western blot analyses of proteins involved in VEGF-induced signaling revealed significantly reduced Tyr1175 phosphorylation level of VEGFR2 in both the shTMEM30A HRECs and the lungs of Tmem30a-iKO mice (at P7), indicating compromised formation of the VEGFA–VEGFR2 complex in the absence of TMEM30A. The phosphorylation levels of other proteins involved in VEGF signaling, including PLCγ1, SRC, p38 MAPK family proteins and ERK1/2, were also decreased (Fig. 8). Our data demonstrate that TMEM30A governs the angiogenesis sprouting process by regulating the VEGF-induced signaling. In addition, an apoptosis assay (TUNEL) revealed no obvious apoptosis (Fig. S3A). Consistent with this, the phosphorylation level of AKT, which involved in VEGF-mediated EC survival, was not reduced (Fig. S1). One possible explanation is that the phosphorylation of AKT is maintained via feedback-mediated upregulation of another growth factor whose signaling is not affected by Tmem30a. Indeed, lack of upregulation of the stress-induced CHOP staining confirmed no apoptosis due to cellular stress (Fig. S2B). Therefore, we propose the following mechanism for the role of Tmem30a in angiogenesis: loss of Tmem30a leads to reduced phosphorylation on Tyr1175 of VEGFR2, which in turn leads to reduced PLCγ1 phosphorylation, which, via PKC, causes reduced phosphorylation of p38 MAPK and ERK1/2 proteins (Takahashi et al., 2001). This ultimately leads to decreased cell proliferation and retarded angiogenesis. Further work is warranted to dissect the relationship bewteen PS distribution and VEGF signaling.

Angiogenesis is a fundamental process in neovascularization, which plays important roles in multiple pathological conditions, such as AMD and cancer (Carmeliet and Jain, 2000). Interventions into the angiogenesis process by chemical or antibody-based drugs have been used to treat certain diseases, such as AMD and cancer (Bizzaro et al., 2018; Calugaru and Calugaru, 2018; Feng et al., 2018; Gorsi et al., 2018; Hutton-Smith et al., 2018; Martini et al., 2018). Our findings in this research have unveiled indispensable roles for TMEM30A, the β-subunit of P4-ATPases, in angiogenesis, which could provide new targets for the treatment of these diseases.

In summary, our study has delineated the essential roles of TMEM30A for sprouting angiogenesis and blood vessel barrier integrity. We further demonstrated that deletion of Tmem30a inhibited angiogenesis by disturbing the VEGF-induced signaling. Our results also show that TMEM30A is a potential target for treatment of pathological angiogenesis.

Cell culture and knockdown of TMEM30A

HRECs (obtained from Cell Systems, Seattle, WA) were cultured in EGM™-2 media (Lonza, Rochester, NY) at 37°C in a 5% CO2 incubator. HRECs at passages 3–7 were transfected with a lentivirus carrying shRNA targeting TMEM30A (5′-TACAATTACCCTGTACATT-3′, Genechem, Shanghai, China) or negative control shRNA (5′-TTCTCCGAACGTGTCACGT-3′) according to the manufacturer's protocol.

RT-PCR

Total RNA of HRECs or retinal tissues was extracted. The cDNA was synthesized by a Superscript cDNA Synthesis Kit (Invitrogen, Waltham, MA, USA) and then used as template for PCR with different primers. The primers used for RT-PCR are listed in Table S1.

RNA-seq analysis

After extraction of total RNA from HRECs using RNeasy Mini kits (QIAGEN), the RNA purity was checked using a K5500 spectrophotometer (Kaiao, Beijing, China). RNA integrity and concentration were assessed using an RNA Nano 6000 Assay Kit of a Bioanalyzer 2100 system (Agilent Technologies, CA). For library preparation, a total of 2 μg RNA per sample was used as input material for the RNA sample preparations. Sequencing libraries were generated using NEB Next, Ultra RNA Library Prep Kit for Illumina® (Cat# E7530L, NEB, USA), following the manufacturer's recommendations, and index codes were added to attribute sequences to each sample. Briefly, mRNA was purified from total RNA using poly-T oligo-attached magnetic beads. Fragmentation was carried out using divalent cations under elevated temperature conditions in NEBNext First Strand Synthesis Reaction Buffer (5×). First-strand cDNA was synthesized using random hexamer primers and RNase H. Second-strand cDNA synthesis was subsequently performed using dNTPs, DNA polymerase I and RNase H. The library fragments were purified with QiaQuick PCR kits and elution with EB buffer, and then terminal repair, A-tailing and addition of an adapter were implemented. The RNA concentration of the library was measured using a Qubit RNA Assay Kit in Qubit 3.0 to preliminarily quantify and then dilute to 1 ng/μl. Insert size was assessed using the Agilent Bioanalyzer 2100 system (Agilent Technologies), and the qualified insert size was accurately quantified using a StepOnePlus™ Real-Time PCR System (library valid concentration>10 nM). The clustering of the index-coded samples was performed on a cBot cluster generation system using a HiSeq PE Cluster Kit v4-cBot-HS (Illumina, CA, USA) according to the manufacturer's instructions. After cluster generation, the libraries were sequenced on an Illumina HiSeq 2500 platform, and 150 bp paired-end reads were generated. All gene expression values from RNA-seq were changed to a log2 value and analyzed further. Then, those gene sets that had less than a 0.05 nominal P value were called.

RT-qPCR

Total RNA from HRECs or lung tissues was extracted with RNeasy Mini kits (QIAGEN), and 1 μg total RNA was reverse transcribed with EasyScript One-Step RT-PCR SuperMix (TransGen Biotech, China) following the manufacturer's instructions. The cDNAs were amplified using TransStart Tip Green qPCR SuperMix (TransGen Biotech, China) in a 7500 Fast Real-Time PCR System (Applied Biosystems, CA). The primers used for RT-qPCR are listed in Table S1.

Western blotting

Cells or tissues from mice were lysed in SDS lysis buffer (2% SDS and 62.5 mM Tris-HCl pH 6.8, containing protease inhibitor cocktail tablets ordered from Roche) and sonicated three times for 5 s. Equal amounts of protein (20 μg) were loaded onto a 10% polyacrylamide gel and analyzed by immunoblotting. The antibodies used for western blotting were against VEGFR2 [Cat# 2479S, Cell Signaling Technology (CST), Danvers, MA,, 1:2000 dilution], phosphorylated (p)-VEGFR2 (Cat# 2478S, CST, 1:2000 dilution), ERK1/2 (Cat# 16443-1-AP, Proteintech, Wuhan, China, 1:3000 dilution), p-ERK1/2 (Cat# 4370S, CST, 1:2000 dilution), MEK1 (Cat# 2352S, CST, 1:2000 dilution), p-MEK1/2 (Cat# 2338S, CST, 1:2000 dilution), p-PLCγ (Cat# 8713T, CST, 1:2000 dilution), SRC (Cat# 2123T, CST, 1:2000 dilution), p-SRC (Cat# 6943T, CST, 1:2000 dilution), p38 MAPK family proteins (Cat# 8690T, CST, 1:2000 dilution), p-p38 MAPK family proteins (Cat# 4511T, CST, 1:2000 dilution), AKT (Cat# 10176-2-AP, Proteintech, 1:2000 dilution), p-AKT (Cat# 4060S, CST, 1:2000 dilution) and GAPDH (Cat# 60004-1-Ig, Proteintech, 1:3000 dilution). HRP-conjugated goat anti-rabbit-IgG secondary antibody (Cat# 7074, CST, 1:5000 dilution) and goat anti-mouse-IgG secondary antibody (Cat# 7076, CST, 1:5000 dilution) were used for immunoblotting.

In vitro Matrigel tube formation assays

The in vitro Matrigel tube formation assays were carried out according to a previously reported method (Shao et al., 2009), with a few modifications. Specifically, 10 μl of liquid Matrigel (Corning, Cat# 354234) was carefully added to each inner well of a μ-Slide Angiogenesis plate (Ibidi, Germany, Cat# 81506) and then incubated at 37°C for half an hour. After the Matrigel was solidified, HRECs were trypsinized, and the cell number was determined using an automated cell counter (Inno-Alliance Biotech). The HRECs were then diluted to 1×106 cells/ml, and 50 μl was added to each well of the μ-Slide angiogenesis plate. After 3 or 6 h, the images of controls or shTMEM30As were captured under an anatomical lens (Carl Zeiss, Germany).

Mouse strains and genotyping

All animal study protocols were approved by the Animal Care and Use Committee of Sichuan Provincial People's Hospital. All experimental procedures and methods were carried out in accordance with the approved study protocols and relevant regulations. Mice were raised in cyclic lighting conditions with a 12-h-light and 12-h-dark cycle.

Tmem30aloxp/+ mice were mated to Pdgfb-CreER mice to generate Tmem30aloxp/+Pdgfb-iCre mice (Claxton et al., 2008). Tmem30aloxp/+Pdgfb-CreER mice were crossed with Tmem30aloxp/loxp homozygous mice to generate an inducible EC deletion of Tmem30a (Tmem30aloxp/loxp Pdgfb-iCre). Tmem30aloxp/+mice were crossed with broadly expressed inducible CRE CAG-Cre-ER mice (Hayashi and McMahon, 2002) [stock Tg(CAG-cre/Esr1*) 5Amc/J, https://www.jax.org/strain/004453] to generate inducible adult knockout mice. To monitor the efficiency of Cre-mediated deletion of the floxed exon, a tdTomato reporter was used [strain name, B6.Cg-Gt(ROSA)26Sortm14(CAG-tdTomato)Hze/J, also named Ai14D, http://jaxmice.jax.org/strain/007914.html]. The reporter contains a loxP-flanked STOP cassette that prevents transcription of the downstream CAG promoter-driven red fluorescent protein variant tdTomato. In the presence of Cre recombinase, reporter mice will have the STOP cassette removed in the Cre-expressing tissue(s) and will express tdTomato. Because this CAG promoter-driven reporter construct was inserted into the Gt(ROSA)26Sor locus, tdTomato expression is determined by the tissue(s) that expressed Cre recombinase.

Tamoxifen treatment

Tamoxifen salt (100 mg; Sigma, St Louis, MO) was dissolved in 10 ml of ethanol as a stock solution. On the day of injection, a 1 mg/ml working solution was prepared by mixing the 10 mg/ml stock solution with corn oil (Sigma) at a ratio of 1:9 and mixed well. Mice were intraperitoneally injected with a daily dosage of 25 mg/kg body weight for 3 consecutive days.

Immunohistochemistry and EDU labeling of retinal ECs

Retinal dissection was carried out as described previously, and whole-mounted retinas were preserved in 0.4% paraformaldehyde (PFA). Enucleated eyes were fixed with 4% PFA and embedded in Tissue-Tek optimal cutting-temperature compound (OCT, Sakura Finetek). Before immunostaining, whole-mounted retinas and cryosections (12 μm, Leica CM1950) were rinsed in PBS three times (5 min/rinse) and blocked in PBS containing 5% fetal bovine serum and 0.2% Triton X-100 for 30 min at room temperature, followed by incubation with primary antibodies at 4°C overnight. Primary antibodies were diluted in blocking buffer as follows: isolectin GS-IB4 (1:200; Thermo Fischer, Waltham, MA; CAT# I21411), rat anti-mouse Ter-119 (1:20 dilution; 553670; BD Biosciences, San Jose, CA), and goat anti-mouse Vegf164 (1:100 dilution; AF-493; R&D Systems, Minneapolis, MN), and C/EBP homologous protein (CHOP; 1:500 dilution, 15204-1-AP; Proteintech, Chicago, IL). Then, the sections were washed with PBS three times and labeled for 1–4 h with Alexa Fluor™-488- or Alexa Fluor™-594-labeled goat anti-rat- or anti-rabbit-IgG or donkey anti-goat-IgG secondary antibody (1:500; Thermo Fischer). To detect EC proliferation in the retinas, 200 μg EDU (Thermo Fischer) per pup was injected intraperitoneally 3 h before animals were euthanized. EDU-positive cells were visible by subsequent staining with a Click-iT EDU Alexa Fluor-488 Imaging Kit (C10337; Thermo Fischer).

TUNEL assay

Retinal dissection was carried out as described previously, and whole-mounted retinas were fixed in 4% PFA and preserved in 0.4% PFA. Whole-mounted retinas were rinsed in PBS three times (5 min/rinse). Retinas were incubated in -ermeabilisation solution (0.1% Triton X-100 in 0.1% sodium citrate, freshly prepared) for 8–10 min. Then the samples were rinsed twice with PBS. After the slides were dried, 50 μl of TUNEL reaction mixture (Roche, In Situ Cell Death Detection Kit, POD Cat# 11684817910) was added to the specimen, and left to react at 37°C for 1 h in a dark humid chamber. For the positive controls, recombinant DNase I (3 U/ml in 50 mM Tris-HCl pH 7.5, 10 mM MgCl2 and 1 mg/ml BSA) was added and incubated for 10 min at 25°C to induce DNA strand breaks, prior to labeling procedures. Slides were rinsed three times with PBS. Images were captured via fluorescence microscopy after slides were sealed with anti-fluorescence quenching liquid.

Author contributions

Conceptualization: L.Z., X.Z.; Methodology: S.Z., W.L., L.Z., X.Z.; Validation: S.Z., Y.Y., X.Z.; Investigation: S.Z., W.L., Y.Y., K.S., S.L., H.X., M.Y., L.Z., X.Z.; Resources: X.Z.; Data curation: S.Z., L.Z., X.Z.; Writing - original draft: L.Z., X.Z.; Writing - review & editing: X.Z.; Supervision: X.Z.; Project administration: L.Z., X.Z.; Funding acquisition: X.Z., L.Z.

Funding

This study was supported by grants from the National Natural Science Foundation of China (http://www.nsfc.gov.cn/; 81470668 and 81770950 to X.Z.; 81700876 to L.Z.), the National Key Scientific Research Program (www.most.gov.cn, 2015CB554100), and the Department of Science and Technology of Sichuan Province (www.scst.gov.cn; 2016TD0009, 2015SZ0060 and 2017TJPT0010 to X.Z.; 2018YSZH0020 to L.Z.) as well as by a grant from the China Postdoctoral Science Foundation (http://jj.chinapostdoctor.org.cn, 2016M600734) to L.Z. The funders had no role in the study design, data collection and analysis, or preparation of the manuscript.

Data availability

The raw sequence data reported in this paper have been deposited in the Genome Sequence Archive in BIG Data Center, Beijing Institute of Genomics (BIG), Chinese Academy of Sciences, under accession numbers CRA001482 that are publicly accessible at http://bigd.big.ac.cn/gsa/browse/CRA001482.

Arashiki
,
N.
,
Takakuwa
,
Y.
,
Mohandas
,
N.
,
Hale
,
J.
,
Yoshida
,
K.
,
Ogura
,
H.
,
Utsugisawa
,
T.
,
Ohga
,
S.
,
Miyano
,
S.
,
Ogawa
,
S.
, et al. 
(
2016
).
ATP11C is a major flippase in human erythrocytes and its defect causes congenital hemolytic anemia
.
Haematologica
101
,
559
-
565
.
Bizzaro
,
F.
,
Falcetta
,
F.
,
D'Agostini
,
E.
,
Decio
,
A.
,
Minoli
,
L.
,
Erba
,
E.
,
Peccatori
,
F. A.
,
Scanziani
,
E.
,
Colombo
,
N.
,
Zucchetti
,
M.
, et al. 
(
2018
).
Tumor progression and metastatic dissemination in ovarian cancer after dose-dense or conventional paclitaxel and cisplatin plus bevacizumab
.
Int. J. Cancer
143
,
2187
-
2199
.
Bryde
,
S.
,
Hennrich
,
H.
,
Verhulst
,
P. M.
,
Devaux
,
P. F.
,
Lenoir
,
G.
and
Holthuis
,
J. C.
(
2010
).
CDC50 proteins are critical components of the human class-1 P4-ATPase transport machinery
.
J. Biol. Chem.
285
,
40562
-
40572
.
Bull
,
L. N.
,
van Eijk
,
M. J. T.
,
Pawlikowska
,
L.
,
DeYoung
,
J. A.
,
Juijn
,
J. A.
,
Liao
,
M.
,
Klomp
,
L. W.
,
Lomri
,
N.
,
Berger
,
R.
,
Scharschmidt
,
B. F.
, et al. 
(
1998
).
A gene encoding a P-type ATPase mutated in two forms of hereditary cholestasis
.
Nat. Genet.
18
,
219
-
224
.
Cacciagli
,
P.
,
Haddad
,
M.-R.
,
Mignon-Ravix
,
C.
,
El-Waly
,
B.
,
Moncla
,
A.
,
Missirian
,
C.
,
Chabrol
,
B.
and
Villard
,
L.
(
2010
).
Disruption of the ATP8A2 gene in a patient with a t(10;13) de novo balanced translocation and a severe neurological phenotype
.
Eur. J. Hum. Genet.
18
,
1360
-
1363
.
Calugaru
,
D.
and
Calugaru
,
M.
(
2018
).
The fate of eyes with wet AMD beyond 4 years of anti-VEGF therapy
.
Graefes Arch. Clin. Exp. Ophthalmol.
256
,
1551
-
1552
.
Carmeliet
,
P.
and
Jain
,
R. K.
(
2000
).
Angiogenesis in cancer and other diseases
.
Nature
407
,
249
-
257
.
Chen
,
C.-Y.
,
Ingram
,
M. F.
,
Rosal
,
P. H.
and
Graham
,
T. R.
(
1999
).
Role for Drs2p, a P-type ATPase and potential aminophospholipid translocase, in yeast late Golgi function
.
J. Cell Biol.
147
,
1223
-
1236
.
Chung
,
A. S.
and
Ferrara
,
N.
(
2011
).
Developmental and pathological angiogenesis
.
Annu. Rev. Cell Dev. Biol.
27
,
563
-
584
.
Claxton
,
S.
,
Kostourou
,
V.
,
Jadeja
,
S.
,
Chambon
,
P.
,
Hodivala-Dilke
,
K.
and
Fruttiger
,
M.
(
2008
).
Efficient, inducible Cre-recombinase activation in vascular endothelium
.
Genesis
46
,
74
-
80
.
Darland-Ransom
,
M.
,
Wang
,
X.
,
Sun
,
C. L.
,
Mapes
,
J.
,
Gengyo-Ando
,
K.
,
Mitani
,
S.
and
Xue
,
D.
(
2008
).
Role of C. elegans TAT-1 protein in maintaining plasma membrane phosphatidylserine asymmetry
.
Science
320
,
528
-
531
.
Feng
,
L.
,
Hu
,
J.-H.
,
Chen
,
J.
and
Xie
,
X.
(
2018
).
An efficacy analysis of anti-vascular endothelial growth factor therapy for choroidal neovascularization secondary to multifocal choroiditis and comparison with wet age-related macular degeneration
.
J. Zhejiang Univ. Sci. B
19
,
327
-
332
.
Folkman
,
J.
(
1971
).
Tumor angiogenesis: therapeutic implications
.
N. Engl. J. Med.
285
,
1182
-
1186
.
Gorsi
,
H. S.
,
Khanna
,
P. C.
,
Tumblin
,
M.
,
Yeh-Nayre
,
L.
,
Milburn
,
M.
,
Elster
,
J. D.
and
Crawford
,
J. R.
(
2018
).
Single-agent bevacizumab in the treatment of recurrent or refractory pediatric low-grade glioma: a single institutional experience
.
Pediatr. Blood Cancer
65
,
e27234
.
Halleck
,
M. S.
,
Lawler
,
J. J.
,
Blackshaw
,
S.
,
Gao
,
L.
,
Nagarajan
,
P.
,
Hacker
,
C.
,
Pyle
,
S.
,
Newman
,
J. T.
,
Nakanishi
,
Y.
,
Ando
,
H.
, et al. 
(
1999
).
Differential expression of putative transbilayer amphipath transporters
.
Physiol. Genomics
1
,
139
-
150
.
Hayashi
,
S.
and
McMahon
,
A. P.
(
2002
).
Efficient recombination in diverse tissues by a tamoxifen-inducible form of Cre: a tool for temporally regulated gene activation/inactivation in the mouse
.
Dev. Biol.
244
,
305
-
318
.
Holthuis
,
J. C. M.
and
Levine
,
T. P.
(
2005
).
Lipid traffic: floppy drives and a superhighway
.
Nat. Rev. Mol. Cell Biol.
6
,
209
-
220
.
Huang
,
L.
,
Zhang
,
H.
,
Cheng
,
C.-Y.
,
Wen
,
F.
,
Tam
,
P. O. S.
,
Zhao
,
P.
,
Chen
,
H.
,
Li
,
Z.
,
Chen
,
L.
,
Tai
,
Z.
, et al. 
(
2016
).
A missense variant in FGD6 confers increased risk of polypoidal choroidal vasculopathy
.
Nat. Genet.
48
,
640
-
647
.
Hutton-Smith
,
L. A.
,
Gaffney
,
E. A.
,
Byrne
,
H. M.
,
Caruso
,
A.
,
Maini
,
P. K.
and
Mazer
,
N. A.
(
2018
).
Theoretical insights into the retinal dynamics of VEGF in patients treated with ranibizumab, based on an ocular pharmacokinetic/pharmacodynamic model
.
Mol. Pharm.
15
,
2270
-
2784
.
Katoh
,
Y.
and
Katoh
,
M.
(
2004
).
Identification and characterization of CDC50A, CDC50B and CDC50C genes in silico
.
Oncol. Rep.
12
,
939
-
943
.
Kisanuki
,
Y. Y.
,
Hammer
,
R. E.
,
Miyazaki
,
J.
,
Williams
,
S. C.
,
Richardson
,
J. A.
and
Yanagisawa
,
M.
(
2001
).
Tie2-cre transgenic mice: a new model for endothelial cell-lineage analysis in vivo
.
Dev Biol.
230
,
230
-
242
.
Koch
,
S.
,
Tugues
,
S.
,
Li
,
X.
,
Gualandi
,
L.
and
Claesson-Welsh
,
L.
(
2011
).
Signal transduction by vascular endothelial growth factor receptors
.
Biochem. J.
437
,
169
-
183
.
Li
,
N.
,
Yang
,
Y.
,
Liang
,
C.
,
Qiu
,
Q.
,
Pan
,
C.
,
Li
,
M.
,
Yang
,
S.
,
Chen
,
L.
,
Zhu
,
X.
and
Hu
,
Y.
(
2018
).
Tmem30a plays critical roles in ensuring the survival of hematopoietic cells and leukemia cells in mice
.
Am. J. Pathol.
188
,
1457
-
1468
.
Liu
,
L.
,
Zhang
,
L.
,
Zhang
,
L.
,
Yang
,
F.
,
Zhu
,
X.
,
Lu
,
Z.
,
Yang
,
Y.
,
Lu
,
H.
,
Feng
,
L.
,
Wang
,
Z.
, et al. 
(
2017
).
Hepatic Tmem30a Deficiency Causes Intrahepatic Cholestasis by Impairing Expression and Localization of Bile Salt Transporters
.
Am. J. Pathol.
187
,
2775
-
2787
.
Martín-Hernández
,
E.
,
Rodríguez-García
,
M. E.
,
Camacho
,
A.
,
Matilla-Dueñas
,
A.
,
Garcia-Silva
,
M. T.
,
Quijada-Fraile
,
P.
,
Corral-Juan
,
M.
,
Tejada-Palacios
,
P.
,
de Las Heras
,
R. S.
,
Arenas
,
J.
, et al. 
(
2016
).
New ATP8A2 gene mutations associated with a novel syndrome: encephalopathy, intellectual disability, severe hypotonia, chorea and optic atrophy
.
Neurogenetics
17
,
259
-
263
.
Martini
,
M.
,
de Pascalis
,
I.
,
D'Alessandris
,
Q. G.
,
Fiorentino
,
V.
,
Pierconti
,
F.
,
Marei
,
H. E.-S.
,
Ricci-Vitiani
,
L.
,
Pallini
,
R.
and
Larocca
,
L. M.
(
2018
).
VEGF-121 plasma level as biomarker for response to anti-angiogenetic therapy in recurrent glioblastoma
.
BMC Cancer
18
,
553
.
Montigny
,
C.
,
Lyons
,
C.
,
Champeil
,
P.
,
Nissen
,
P.
and
Lenoir
,
G.
(
2016
).
On the molecular mechanism of flippase- and scramblase-mediated phospholipid transport
.
Biochim. Biophys. Acta.
1861
,
767
-
783
.
Olsson
,
A.-K.
,
Dimberg
,
A.
,
Kreuger
,
J.
and
Claesson-Welsh
,
L.
(
2006
).
VEGF receptor signalling - in control of vascular function
.
Nat. Rev. Mol. Cell Biol.
7
,
359
-
371
.
Op den Kamp
,
J. A. F.
(
1979
).
Lipid asymmetry in membranes
.
Annu. Rev. Biochem.
48
,
47
-
71
.
Paulusma
,
C. C.
and
Oude Elferink
,
R. P. J.
(
2005
).
The type 4 subfamily of P-type ATPases, putative aminophospholipid translocases with a role in human disease
.
Biochim. Biophys. Acta
1741
,
11
-
24
.
Paulusma
,
C. C.
,
Folmer
,
D. E.
,
Ho-Mok
,
K. S.
,
de Waart
,
D. R.
,
Hilarius
,
P. M.
,
Verhoeven
,
A. J.
and
Oude Elferink
,
R. P. J.
(
2008
).
ATP8B1 requires an accessory protein for endoplasmic reticulum exit and plasma membrane lipid flippase activity
.
Hepatology
47
,
268
-
278
.
Potente
,
M.
,
Gerhardt
,
H.
and
Carmeliet
,
P.
(
2011
).
Basic and therapeutic aspects of angiogenesis
.
Cell
146
,
873
-
887
.
Saito
,
K.
,
Fujimura-Kamada
,
K.
,
Furuta
,
N.
,
Kato
,
U.
,
Umeda
,
M.
and
Tanaka
,
K.
(
2004
).
Cdc50p, a protein required for polarized growth, associates with the Drs2p P-type ATPase implicated in phospholipid translocation in Saccharomyces cerevisiae
.
Mol. Biol. Cell
15
,
3418
-
3432
.
Segawa
,
K.
,
Kurata
,
S.
,
Yanagihashi
,
Y.
,
Brummelkamp
,
T. R.
,
Matsuda
,
F.
and
Nagata
,
S.
(
2014
).
Caspase-mediated cleavage of phospholipid flippase for apoptotic phosphatidylserine exposure
.
Science
344
,
1164
-
1168
.
Seigneuret
,
M.
and
Devaux
,
P. F.
(
1984
).
ATP-dependent asymmetric distribution of spin-labeled phospholipids in the erythrocyte membrane: relation to shape changes
.
Proc. Natl. Acad. Sci. USA
81
,
3751
-
3755
.
Shao
,
R.
,
Hamel
,
K.
,
Petersen
,
L.
,
Cao
,
Q. J.
,
Arenas
,
R. B.
,
Bigelow
,
C.
,
Bentley
,
B.
and
Yan
,
W.
(
2009
).
Ykl-40, a secreted glycoprotein, promotes tumor angiogenesis
.
Oncogene
28
,
4456
-
4468
.
Siggs
,
O. M.
,
Arnold
,
C. N.
,
Huber
,
C.
,
Pirie
,
E.
,
Xia
,
Y.
,
Lin
,
P.
,
Nemazee
,
D.
and
Beutler
,
B.
(
2011a
).
The P4-type ATPase ATP11C is essential for B lymphopoiesis in adult bone marrow
.
Nat. Immunol.
12
,
434
-
440
.
Siggs
,
O. M.
,
Schnabl
,
B.
,
Webb
,
B.
and
Beutler
,
B.
(
2011b
).
X-linked cholestasis in mouse due to mutations of the P4-ATPase ATP11C
.
Proc. Natl. Acad. Sci. USA
108
,
7890
-
7895
.
Stapelbroek
,
J. M.
,
Peters
,
T. A.
,
van Beurden
,
D. H. A.
,
Curfs
,
J. H. A. J.
,
Joosten
,
A.
,
Beynon
,
A. J.
,
van Leeuwen
,
B. M.
,
van der Velden
,
L. M.
,
Bull
,
L.
,
Oude Elferink
,
R. P.
, et al. 
(
2009
).
ATP8B1 is essential for maintaining normal hearing
.
Proc. Natl Acad. Sci. USA
106
,
9709
-
9714
.
Takahashi
,
T.
,
Yamaguchi
,
S.
,
Chida
,
K.
and
Shibuya
,
M.
(
2001
).
A single autophosphorylation site on KDR/Flk-1 is essential for VEGF-A-dependent activation of PLC-gamma and DNA synthesis in vascular endothelial cells
.
EMBO J.
20
,
2768
-
2678
.
Takatsu
,
H.
,
Baba
,
K.
,
Shima
,
T.
,
Umino
,
H.
,
Kato
,
U.
,
Umeda
,
M.
,
Nakayama
,
K.
and
Shin
,
H.-W.
(
2011
).
ATP9B, a P4-ATPase (a putative aminophospholipid translocase), localizes to the trans-Golgi network in a CDC50 protein-independent manner
.
J. Biol. Chem.
286
,
38159
-
38167
.
Takatsu
,
H.
,
Takayama
,
M.
,
Naito
,
T.
,
Takada
,
N.
,
Tsumagari
,
K.
,
Ishihama
,
Y.
,
Nakayama
,
K.
and
Shin
,
H. W.
(
2017
).
Phospholipid flippase ATP11C is endocytosed and downregulated following Ca(2+)-mediated protein kinase C activation
.
Nat. Commun.
8
,
1423
.
Tang
,
X.
,
Halleck
,
M. S.
,
Schlegel
,
R. A.
and
Williamson
,
P.
(
1996
).
A subfamily of P-type ATPases with aminophospholipid transporting activity
.
Science
272
,
1495
-
1497
.
van der Velden
,
L. M.
,
Wichers
,
C. G. K.
,
van Breevoort
,
A. E. D.
,
Coleman
,
J. A.
,
Molday
,
R. S.
,
Berger
,
R.
,
Klomp
,
L. W. J.
and
van de Graaf
,
S. F. J.
(
2010
).
Heteromeric interactions required for abundance and subcellular localization of human CDC50 proteins and class 1 P4-ATPases
.
J. Biol. Chem.
285
,
40088
-
40096
.
Wang
,
L.
,
Beserra
,
C.
and
Garbers
,
D. L.
(
2004
).
A novel aminophospholipid transporter exclusively expressed in spermatozoa is required for membrane lipid asymmetry and normal fertilization
.
Dev. Biol.
267
,
203
-
215
.
Yabas
,
M.
,
Teh
,
C. E.
,
Frankenreiter
,
S.
,
Lal
,
D.
,
Roots
,
C. M.
,
Whittle
,
B.
,
Andrews
,
D. T.
,
Zhang
,
Y.
,
Teoh
,
N. C.
,
Sprent
,
J.
, et al. 
(
2011
).
ATP11C is critical for the internalization of phosphatidylserine and differentiation of B lymphocytes
.
Nat. Immunol.
12
,
441
-
449
.
Yabas
,
M.
,
Coupland
,
L. A.
,
Cromer
,
D.
,
Winterberg
,
M.
,
Teoh
,
N. C.
,
D'Rozario
,
J.
,
Kirk
,
K.
,
Bröer
,
S.
,
Parish
,
C. R.
and
Enders
,
A.
(
2014
).
Mice deficient in the putative phospholipid flippase ATP11C exhibit altered erythrocyte shape, anemia, and reduced erythrocyte life span
.
J. Biol. Chem.
289
,
19531
-
19537
.
Yang
,
Z.
,
Camp
,
N. J.
,
Sun
,
H.
,
Tong
,
Z.
,
Gibbs
,
D.
,
Cameron
,
D. J.
,
Chen
,
H.
,
Zhao
,
Y.
,
Pearson
,
E.
,
Li
,
X.
, et al. 
(
2006
).
A variant of the HTRA1 gene increases susceptibility to age-related macular degeneration
.
Science
314
,
992
-
993
.
Zhang
,
L.
,
Yang
,
Y.
,
Li
,
S.
,
Tai
,
Z.
,
Huang
,
L.
,
Liu
,
Y.
,
Zhu
,
X.
,
Di
,
Y.
,
Qu
,
C.
,
Jiang
,
Z.
, et al. 
(
2016
).
Whole Exome Sequencing Analysis Identifies Mutations in LRP5 in Indian Families with Familial Exudative Vitreoretinopathy
.
Genet. Test Mol Biomarkers
20
,
346
-
351
.
Zhang
,
L.
,
Yang
,
Y.
,
Li
,
S.
,
Zhang
,
S.
,
Zhu
,
X.
,
Tai
,
Z.
,
Yang
,
M.
,
Liu
,
Y.
,
Guo
,
X.
,
Chen
,
B.
, et al. 
(
2017
).
Loss of Tmem30a leads to photoreceptor degeneration
.
Sci. Rep.
7
,
9296
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information