Regulation of angiogenic behaviors by oxytocin receptor through Gli1-indcued transcription of HIF-1α in human umbilical vein endothelial cells

Regulation of angiogenic behaviors by oxytocin receptor through Gli1-indcued transcription of HIF-1α in human umbilical vein endothelial cells

Biomedicine & Pharmacotherapy 90 (2017) 928–934 Available online at ScienceDirect www.sciencedirect.com Original article Regulation of angiogenic ...

1MB Sizes 0 Downloads 32 Views

Biomedicine & Pharmacotherapy 90 (2017) 928–934

Available online at

ScienceDirect www.sciencedirect.com

Original article

Regulation of angiogenic behaviors by oxytocin receptor through Gli1-indcued transcription of HIF-1a in human umbilical vein endothelial cells Ji Zhua , Huiyan Wangb , Xiaoyan Zhanga , Yin Xiec,* a b c

Department of Pharmacy, Changzhou Maternal and Child Health Care Hospital, Changzhou 213000, China Department of Obstetrics, Changzhou Maternal and Child Health Care Hospital, Changzhou 213000, China Department of Pharmacy, The First People's Hospital of Changzhou, Changzhou 213000, China

A R T I C L E I N F O

Article history: Received 24 January 2017 Received in revised form 30 March 2017 Accepted 10 April 2017 Keywords: Oxytocin receptor Angiogenesis Hypoxia-inducible factor-1a Gli1 Human umbilical vein endothelial cells

A B S T R A C T

Angiogenesis is a dynamic hypoxia-stimulated process playing a key role in tissue growth and repair under various pathophysiological circumstances. Abnormal angiogenesis contributes to the pathogenesis of many human diseases. Oxytocin receptor is a classical G-protein-coupled receptor expressed on endothelial cells. The present study was aimed to investigate how oxytocin receptor regulated the angiogenic behaviors of human umbilical vein endothelial cells (HUVECs). We found that oxytocin at 0.1 mM significantly increased cell proliferation, upregulated the mRNA and protein expression of CD31 and vWF (two important endothelial markers), and enhanced the tuber formation capacity in HUVECs. However, oxytocin receptor inhibitor atosiban at 10 mM significantly suppressed these angiogenic properties of HUVECs. Additionally, hypoxia-inducible factor-1a (HIF-1a) inhibitor PX-478 at 20 mM also remarkably inhibited the angiogenic properties of HUVECs. We further found that atosiban at 10 mM significantly repressed the promoter activity of HIF-1a and reduced the mRNA and protein expression of HIF-1a in HUVECs. Moreover, pharmacological inhibition of HIF-1a by PX-478 at 20 mM abolished oxytocin-enhanced angiogenic properties of HUVECs. Finally, transcription factor Gli1 inhibitor GANT-58 at 5 mM significantly abolished oxytocin-induced mRNA and protein expression of HIF-1a, while the nuclear abundance of Gli1 was significantly reduced by atosiban at 10 mM, but was increased by oxytocin at 0.1 mM in HUVECs. GANT-58 at 5 mM also significantly abolished oxytocin-enhanced angiogenic properties of HUVECs. Altogether, these discoveries suggested that oxytocin receptor signaling promoted the angiogenic behaviors of HUVECs via Gli1-indcued transcription of HIF-1a. We provided novel molecular insights into endothelial cell-mediated angiogenesis. © 2017 Elsevier Masson SAS. All rights reserved.

1. Introduction Angiogenesis represents a dynamic hypoxia-stimulated process leading to the formation of new vessels from pre-existing blood vessels. It occurs in nearly all tissues and organs, and is thought to be a key step for tissue growth and repair under various physiological circumstances [1]. However, dysregulation of angiogenesis contributes to the pathogenesis of many human diseases,

Abbreviations: DMSO, dimethylsulfoxide; FBS, bovine serum; GAPDH, glyceraldehyde phosphate dehydrogenase; GPCR, G-protein-coupled receptor; HIF-1a, hypoxia-inducible factor-1a; HUVECs, human umbilical vein endothelial cells; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide; NF-kB, nuclear factor-kB. * Corresponding author. E-mail address: [email protected] (Y. Xie). http://dx.doi.org/10.1016/j.biopha.2017.04.021 0753-3322/© 2017 Elsevier Masson SAS. All rights reserved.

historically including cancer, arthritis, psoriasis and blindness. In addition, many common disorders such as obesity and atherosclerosis and several congenital diseases are also caused by abnormal vascular remodeling [2]. On the other hand, insufficient vessel growth or abnormal vessel regression can lead to heart and brain ischemia, hypertension, neurodegeneration, osteoporosis and other disorders [3]. Endothelial cells are elongated and thin cells playing a pivotal role during angiogenesis. They are responsible for building channels that efficiently distribute blood to each parts of the body. When stimulated or activated, they are able to rapidly send out sprouts in a directional and coordinated manner to form nascent vascular bed, which matures into a system of stable vessels [4]. Endothelial cells can also sense changes in blood flow and pressure, and dynamically communicate with cells inside and outside the vessel lumen [4]. Hypoxia is an important stimulus for

J. Zhu et al. / Biomedicine & Pharmacotherapy 90 (2017) 928–934

expansion of vascular bed by signaling through hypoxia-inducible factor-1a (HIF-1a). HIF-1a drives transcription of a variety of angiogenic genes, leading to physiological and pathological angiogenesis [5]. Consistent with the key role of hypoxia in the overall process, many phenotypic processes of endothelial cells are modulated by HIF-1a during angiogenesis [5]. Although HIF-1a activation under hypoxia is mostly obtained by post-translational mechanisms, there are scenarios in which HIF-1a mRNA transcription can be increased by cytokines, growth factors, oncogenes, metabolic stress and reactive oxygen species [6]. Currently, despite the increasing understanding of angiogenesis, the molecular mechanisms and their possible implications for medicine remain to be defined. Oxytocin receptor is a classical G-protein-coupled receptor (GPCR) found to be expressed on endothelial cells [7]. Through oxytocin receptor, oxytocin could stimulate the migration of human dermal microvascular endothelial cells [8]. A recent study also showed that oxytocin stimulated migration and invasion in human umbilical vein endothelial cells (HUVECs) via activation of oxytocin receptor [9]. Given these discoveries, we here hypothesized that oxytocin receptor could regulate the angiogenic properties of endothelial cells with important pathophysiological consequences in angiogenesis-associated diseases. We examined the effects of oxytocin and atosiban, a specific antagonist of oxytocin receptor, on the angiogenic behaviors of HUVECs and investigate the underlying mechanism. 2. Materials and methods 2.1. Chemicals and antibodies Recombinant human oxytocin was obtained from Calbiochem (Darmstadt, Germany). The following compounds were used: atosiban, PX-478 and PDTC (Selleck Chemicals, Houston, TX, USA); XAV-939 and GANT-98 (Cayman Chemical, Ann Arbor, MI, USA). They were dissolved in dimethylsulfoxide (DMSO) for experiments. Treatment with DMSO alone was used as vehicle control throughout the current study. The following primary antibodies were used: CD31, vWF, and HIF-1a (Proteintech Group, Chicago, IL, USA); Gli1, Lamin B1 and GAPDH (Cell Signaling Technology, Danvers, MA, USA). 2.2. Cell culture HUVECs were obtained from the Cell Bank of Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (Shanghai, China). HUVECs were cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS), 100 mg/ml penicillin, and 100 mg/ml streptomycin, and maintained in an incubator with a humidified atmosphere of 5% CO2 at 37  C. 2.3. Cell proliferation assay HUVECs cells were seeded in 96-well plates and cultured in RPMI 1640 medium supplemented with 10% FBS for 24 h, and then were treated with DMSO or different reagents at indicated concentrations for 24 h. Then the medium was replaced with 100 ml phosphate buffered saline containing 0.5 mg/ml 3-(4,5dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and then was incubated at 37  C for 4 h. Next, the crystals were dissolved with 200 ml DMSO. The spectrophotometric absorbance at 490 nm was measured by a SPECTRAmaxTM microplate spectrophotometer (Molecular Devices, Sunnyvale, CA, USA). Results were from three independent experiments and each experiment had six replicates.

929

2.4. Real-time PCR Total RNAwas prepared from treated HUVECs using Trizol reagent (Sigma, Saint Louis, MO, USA) and then was subjected to reverse transcription to cDNA using the kits provided by TaKaRa Biotechnology Co., Ltd. (Dalian, China) according to the protocol. Real-time PCR was performed using the SYBR Green Master Mix (Vazyme Biotech Co., Ltd., Nanjing China) according to the protocol provided by the manufacture. Fold changes in the mRNA levels of target genes related to the invariant control glyceraldehyde phosphate dehydrogenase (GAPDH) were calculated as suggested [10]. The primers of genes (GenScript, Nanjing, China) were as follows: CD31: (forward) 50 GACAGCCAAGGCAGATGCAC-30 , (reverse) 50 -ATTGGATGGCTTGGCCTGAA-30 ; vWF: (forward) 50 -GCGTGGCAGTGGTAGAGTA-30 , (reverse) 50 -GGAGATAGCGGGTGAAAT-30 ; GAPDH: (forward) 50 -TGACAACAGCCTCAAGAT-30 , (reverse) 50 -GAGTCCTTCCACGATACC-30 . Results were from triplicate experiments. 2.5. Western blot analyses Whole cell protein extracts were prepared from treated HUVECs with RIPA buffer containing protease inhibitor. In certain experiments, nuclear proteins were separated using a Bioepitope Nuclear and Cytoplasmic Extraction Kit (Bioworld Technology, Saint Louis Park, MN, USA) according to the protocol. Proteins (50 mg/well) were separated by SDS-polyacrylamide gel, transferred to a PVDF membrane (Millipore, Burlington, MA, USA), blocked with 5% skim milk in Tris-buffered saline containing 0.1% Tween 20. Target proteins were detected by corresponding primary antibodies, and subsequently by horseradish peroxidase-conjugated secondary antibodies. Protein bands were visualized using chemiluminescence reagent (Millipore, Burlington, MA, USA) by Bio-Rad Universal Hood II DOC Electrophoresis Imaging Cabinet. GAPDH was used as an invariant control for equal loading of total proteins, and Lamin B1 was for nuclear proteins. Representative blots were shown. 2.6. Tubulogenesis assay HUVECs were seeded on growth factor-reduced Matrigel (BD Biosciences, Bedford, MA) after 30 min of preincubation at 37  C in 24-well plates. HUVECs were treated with DMSO or different reagents at indicated concentrations for 24 h. Tubulogenesis was visualized at random fields under a microscope (Leica, Germany). Tubulogenesis was assessed by counting the number of closed intercellular compartments (closed rings or pro-angiogenic structures) according to reported methods [11]. Representative views were shown. 2.7. Dual-luciferase reporter assay HUVECs were seeded in 96-well plates and transfected with pHIF-1a-pGL3-Basic-Luc provided by Zoonbio Biotechnology Co., Ltd. (Nanjing, China) using X-tremeGENE 9 DNA Transfection Reagent (Roche, Swiss) in antibiotic free medium for 24 h. Then, cells were grown in refreshed medium and treated with DMSO or different reagents for 24 h. Transfection efficiency was normalized by co-transfection of renilla luciferase reporter plasmid pRL-TK Vector (Roche, Swiss). Luciferase activities were measured using a dual-luciferase reporter system (Promega, Madison, WI, USA) and presented in arbitrary units after normalization to renilla luciferase activities. 2.8. Statistical analyses Data were presented as mean  SD and analyzed using SPSS16.0 software. The significance of difference was determined by one-

930

J. Zhu et al. / Biomedicine & Pharmacotherapy 90 (2017) 928–934

way ANOVA with the post hoc Tukey's test. Values of P < 0.05 were considered to be statistically significant. 3. Results 3.1. Oxytocin receptor regulates angiogenic behaviors of HUVECs Proliferation of endothelial cells is a key step of angiogenesis. We initially examined the role of oxytocin receptor in proliferation of HUVECs. The data showed that pharmacological inhibition of oxytocin receptor by atosiban at 10 mM significantly decreased HUVEC proliferation, whereas oxytocin at 0.1 mM significantly increased HUVEC proliferation (Fig. 1A). Examinations of endothelial markers demonstrated that atosiban significantly downregulated the mRNA and protein expression of CD31 and vWF; however, their expression at both mRNA and protein levels was significantly upregulated by oxytocin (Fig. 1B and C). Additionally, HUVECmediated angiogenesis was evaluated by tubulogenesis assay on Matrigel. The results showed that HUVEC-mediated tuber formation was significantly inhibited by atosiban at 10 mM, but was significantly enhanced by oxytocin at 0.1 mM (Fig. 1D). Taken together, these findings indicated that oxytocin receptor regulated angiogenic behaviors of HUVECs. 3.2. HIF-1a also regulates angiogenic behaviors of HUVECs We next postulated that HIF-1a could be involved in regulation of angiogenic properties of HUVECs, and thus used HIF-1a specific inhibitor PX-478 to testify this postulation. As expected, PX-478 at 20 mM significantly repressed proliferation of HUVECs (Fig. 2A). PX-478 at 20 mM also remarkably reduced the mRNA and protein expression of CD31 and vWF in HUVECs (Fig. 2B, C). Furthermore, HUVEC-mediated angiogenesis on Matrigel was significantly repressed by PX-478 inhibition of HIF-1a (Fig. 2D). Collectively, these date suggested that the angiogenic behaviors of HUVECs were also controlled by HIF-1a.

3.3. Oxytocin receptor transcriptionally controls the expression of HIF1a, which mediates oxytocin receptor regulation of angiogenic behaviors of HUVECs We subsequently attempted to explore the molecular link of HIF-1a to oxytocin receptor regulation of angiogenic behaviors of HUVECs. Luciferase assays showed that the promoter activity of HIF-1a was significantly repressed by atosiban at 10 mM, but was significantly enhanced by oxytocin at 0.1 mM in HUVECs (Fig. 3A). Consistently, the mRNA and protein expression of HIF-1a was also significantly reduced by atosiban at 10 mM, but was increased by oxytocin at 0.1 mM in HUVECs (Fig. 3B and C). The above data collectively indicated that oxytocin receptor regulated the transcription of HIF-1a. Furthermore, pharmacological inhibition of HIF-1a by PX-478 at 20 mM abolished oxytocin-enhanced proliferation of HUVECs (Fig. 4A). Oxytocin upregulation of expression of HIF-1a was also significantly diminished by PX478 at both mRNA and protein levels (Fig. 4B and C). Similarly, PX478 at 20 mM significantly abrogated oxytocin-induced tuber formation of HUVECs on Matrigel (Fig. 4D). Altogether, these data revealed that transcription of HIF-1a mediated oxytocin receptor regulation of angiogenic behaviors of HUVECs. 3.4. Crosstalk with Gli1 mediates oxytocin receptor regulation of HIF1a and angiogenic behaviors of HUVECs We finally investigated how oxytocin receptor transcriptionally regulated HIF-1a expression in HUVECs. We used the nuclear factor-kB (NF-kB) inhibitor PDTC, b-catenin inhibitor XAV-939, and Gli1 inhibitor GANT-58 to screen the potential transcription factor mediating oxytocin receptor regulation of HIF-1a expression. The results showed that GANT-58 at 5 mM significantly abolished oxytocin-induced HIF-1a mRNA expression, whereas PDTC at 5 mM and XAV-939 at 10 mM did not apparently affect oxytocin-induced HIF-1a mRNA expression (Fig. 5A). Western blot analyses showed similar results at the protein level (Fig. 5B). These data suggested that the transcription factor Gli1 could be involved

Fig. 1. Oxytocin receptor regulates angiogenic behaviors of HUVECs. HUVECs were treated with vehicle, atociban (10 mM), or oxytocin (0.1 mM) for 24 h. (A) Cell proliferation was determined using MTT assay. Significance: *P < 0.05 versus control. (B) Real-time PCR analyses of mRNA expression of CD31 and vWF. Significance: *P < 0.05 versus control. (C) Western blot analyses of protein expression of CD31 and vWF. (D) Tubulogenesis assay with quantification of number of closed intercellular compartments (100 magnification). Significance: *P < 0.05 versus control.

J. Zhu et al. / Biomedicine & Pharmacotherapy 90 (2017) 928–934

931

Fig. 2. HIF-1a regulates angiogenic behaviors of HUVECs. HUVECs were treated with vehicle or PX-478 (20 mM) for 24 h. (A) Cell proliferation was determined using MTT assay. Significance: *P < 0.05 versus control. (B) Real-time PCR analyses of mRNA expression of CD31 and vWF. Significance: *P < 0.05 versus control. (C) Western blot analyses of protein expression of CD31 and vWF. (D) Tubulogenesis assay with quantification of number of closed intercellular compartments (100 magnification). Significance: *P < 0.05 versus control.

Fig. 3. Oxytocin receptor transcriptionally controls the expression of HIF-1a in HUVECs. HUVECs were treated with vehicle, atociban (10 mM), or oxytocin (0.1 mM) for 24 h. (A) Luciferase activities of HIF-1a promoter expressed as arbitrary units. Significance: *P < 0.05 versus control. (B) Real-time PCR analyses of mRNA expression of HIF-1a. Significance: *P < 0.05 versus control. (C) Western blot analyses of protein expression of HIF-1a.

in current context. Indeed, subsequent Western blot examinations of nuclear Gli1 showed that atosiban at 10 mM reduced nuclear abundance of Gli1 but oxytocin at 0.1 mM increased nuclear abundance of Gli1 in HUVECs (Fig. 5C). Furthermore, GANT-58 at 5 mM significantly abolished oxytocin-enhanced proliferation (Fig. 5D) and tuber formation (Fig. 5E) in HUVECs. Altogether, these discoveries suggested that oxytocin receptor regulated HIF1a transcription and angiogenic behaviors via crosstalk with Gli1 in HUVECs. 4. Discussion The past decade has witnessed much progress in understanding how the behaviors of endothelial cells are coordinated to generate an interconnected and functional vascular network. Angiogenesis is considered a multistep branching morphogenesis process including the sprouting of new vessels from pre-existing ones, the formation of vascular loops through anastomosis of sprouts, and the optimization of the vascular network by vessel pruning [12]. During these processes, endothelial cells exhibit numerous, sometimes unique, cellular behaviors. In current study, HUVECs

were cultured for experiments, which are derived from the endothelium of veins from the umbilical cord exhibiting a cobblestone phenotype when lining vessel walls, and are usually used as a laboratory model system for the study of the function and pathology of endothelial cells and angiogenesis [13]. Using this classical model, we focused on examining cell proliferation, endothelial marker expression and tuber formation capacity on Matrigel, because these characteristic behaviors of endothelial cells are used primarily to investigate the cellular and molecular basis of angiogenesis. Recent understanding of oxytocin biology has come to disclose how the dynamic assembly of endothelial cells into functional vascular networks is controlled and executed at the molecular level. The classical action of oxytocin is, through oxytocin receptor, to stimulate uterine smooth muscle contraction during labor and milk ejection [14]. Oxytocin is also expressed outside the reproductive system, and in particular that it is produced by some cancer tissues [15]. In the same way, oxytocin receptor is expressed in various physiological and pathological tissues. For example, the expression of oxytocin receptor has been demonstrated in human vascular endothelial cells, on which oxytocin induced a

932

J. Zhu et al. / Biomedicine & Pharmacotherapy 90 (2017) 928–934

Fig. 4. HIF-1a mediates oxytocin receptor regulation of angiogenic behaviors of HUVECs. HUVECs were treated with vehicle, oxytocin (0.1 mM), and/or PX-478 (20 mM) for 24 h. (A) Cell proliferation was determined using MTT assay. Significance: *P < 0.05 versus control, #P < 0.05 versus oxytocin. (B) Real-time PCR analyses of mRNA expression of CD31 and vWF. Significance: *P < 0.05 versus control, #P < 0.05 versus oxytocin. (C) Western blot analyses of protein expression of CD31 and vWF. (D) Tubulogenesis assay with quantification of number of closed intercellular compartments (100 magnification). Significance: *P < 0.05 versus control, #P < 0.05 versus oxytocin.

Fig. 5. Crosstalk with Gli1 mediates oxytocin receptor regulation of HIF-1a and angiogenic behaviors of HUVECs. HUVECs were treated with vehicle, atosiban (10 mM), oxytocin (0.1 mM), and/or PDTC (5 mM), XAV-939 (10 mM), GANT-58 (5 mM) for 24 h. (A) Real-time PCR analyses of mRNA expression of HIF-1a. Significance: *P < 0.05 versus control, #P < 0.05 versus oxytocin. (B) Western blot analyses of protein expression of HIF-1a. (C) Western blot analyses of nuclear abundance of Gli1. (D) Cell proliferation was determined using MTT assay. Significance: *P < 0.05 versus control, #P < 0.05 versus oxytocin. (E) Tubulogenesis assay with quantification of number of closed intercellular compartments (100 magnification). Significance: *P < 0.05 versus control, #P < 0.05 versus oxytocin.

proliferative response [9]. Bases on these discoveries, we here used pharmacological approaches to investigate the role of oxytocin receptor in regulation of angiogenic behaviors of endothelial cells

implicated in therapeutics of angiogenesis-related diseases. In addition to using oxytocin for activating the receptor, the specific inhibitor atosiban was used in parallel to antagonize the receptor

J. Zhu et al. / Biomedicine & Pharmacotherapy 90 (2017) 928–934

and disrupt its intracellular signaling [16]. We found that the angiogenic behaviors of HUVECs were all enhanced by oxytocin but repressed by atosiban, indicating that oxytocin receptor controlled endothelial cell-involved angiogenesis. Consistent with our data, some other aspects of endothelial cell behaviors such as migration and invasion were also increased by oxytocin in HUVECs [17]. Similarly, oxytocin receptor was also found to participate in growth and migration in immortalized human dermal microvascular endothelial cells and human breast tumor-derived endothelial cells [8]. We further discovered that HIF-1a was involved in this context, because pharmacological inhibition of HIF-1a significantly suppressed the angiogenic behaviors of HUVECs. This observation was reasonable, because of the biological nature of HIF-1a as a master transcription regulator in angiogenesis network [18]. The question that how oxytocin receptor was linked to HIF-1a in this context led us to found that activation or inhibition of oxytocin receptor affected the transcription of HIF-1a in HUVECs, suggesting that HIF-1a could be a target gene of oxytocin receptor-transmitted signaling cascade. In addition, pharmacological repression of HIF1a significantly abolished oxytocin-enhanced angiogenic behaviors, confirming that transcription of HIF-1a was required for oxytocin receptor to regulate the angiogenic behaviors of HUVECs. This was a newly identified molecular link between oxytocin signaling and HIF-1a in the signaling network controlling angiogenesis. Subsequently, we attempted to uncover which transcription factor mediating oxytocin-induced HIF-1a transcription in HUVECs. We screened several transcription factors that might be involved in controlling transcription of HIF-1a using pharmacological approaches. Interestingly, we found that Gli1 could be involved in this context, because Gli1 inhibitor GANT-58 significantly abrogated oxytocin-induced mRNA and protein expression of HIF-1a, but NF-kB inhibitor PDTC and b-catenin inhibitor XAV939 failed to do so. In agreement, we observed that the nuclear translocation of Gli1 was increased by ligand activation of oxytocin receptor in HUVECs. Classically, Gli1 is a well-established transcription factor of hedgehog signaling, a conserved morphogenic cascade that is propagated by a family of ligands, which bind to the membrane receptor Patched and de-repress the activity of Patched on Smoothened, regulating the expression of hedgehog target genes [19]. Our current data indicated an interaction between oxytocin signaling and hedgehog pathway in modulation of angiogenic properties of endothelial cells. These molecular discoveries could be supported by the recognition that many of the cellular responses mediated by GPCRs do not involve the sole stimulation of conventional second-messenger-generating systems, but instead, result from the functional integration of an intricate network of intracellular signaling pathways [20]. Effectors for GPCRs that are independent of G proteins have now been identified widely, thus changing the conventional view of the GPCR-heterotrimeric-G-protein-associated effector [20]. Meanwhile, Gli1, in addition to mediating canonical hedgehog pathway, has been found to crosstalk with multiple signal pathways such as epidermal growth factor receptor signaling in human medulloblastoma cells [21], and NF-kB pathway in diffuse large B-cell lymphoma [22]. In the present study, crosstalk between oxytocin signaling and hedgehog pathway might play an important role in the complex molecular network governing endothelial cell-mediated angiogenesis. However, whether this regulation is present in other types of cells merits more investigations. Finally, our additional experiments showed that pharmacological suppression of Gli1 eliminated oxytocin-enhanced angiogenic properties in HUVECs, strengthening the proangiogenic role for Gli1. Consistently, there were also studies showing that Gli1 signaling contributed to angiogenesis in gliomas

933

Fig. 6. Schematic diagram illustrating the mechanisms by which oxytocin receptor regulated the angiogenic behaviors of HUVECs.

[23] and regulated hepatic stellate cell-mediated angiogenesis in liver fibrosis [24]. In conclusion, oxytocin receptor regulated the angiogenic behaviors of HUVECs through Gli1-induced transcription of HIF1a (illustrated in Fig. 6). Our discoveries suggested a proangiogenic role for oxytocin and its receptor, and uncovered a novel molecular mechanism underlying endothelial cell-mediated angiogenesis. Conflict of interest The authors declare no conflicts of interest. Acknowledgements This work was supported by the Science and Technology Plan Project of Changzhou (Grant No. 2016411). References [1] L.A. DiPietro, Angiogenesis and wound repair: when enough is enough, J. Leukoc. Biol. 100 (2016) 979–984. [2] Y. Cao, Angiogenesis as a therapeutic target for obesity and metabolic diseases, Chem. Immunol. Allergy 99 (2014) 170–179. [3] P. Carmeliet, R.K. Jain, Molecular mechanisms and clinical applications of angiogenesis, Nature 473 (2011) 298–307. [4] C. Betz, A. Lenard, H.G. Belting, M. Affolter, Cell behaviors and dynamics during angiogenesis, Development 143 (2016) 2249–2260. [5] J. Brocato, Y. Chervona, M. Costa, Molecular responses to hypoxia-inducible factor 1alpha and beyond, Mol. Pharmacol. 85 (2014) 651–657. [6] C. Brahimi-Horn, N. Mazure, J. Pouyssegur, Signalling via the hypoxiainducible factor-1alpha requires multiple posttranslational modifications, Cell Signal. 17 (2005) 1–9. [7] T. Kimura, F. Saji, K. Nishimori, K. Ogita, H. Nakamura, M. Koyama, et al., Molecular regulation of the oxytocin receptor in peripheral organs, J. Mol. Endocrinol. 30 (2003) 109–115. [8] P. Cassoni, T. Marrocco, B. Bussolati, E. Allia, L. Munaron, A. Sapino, et al., Oxytocin induces proliferation and migration in immortalized human dermal microvascular endothelial cells and human breast tumor-derived endothelial cells, Mol. Cancer Res. 4 (2006) 351–359. [9] M. Thibonnier, D.M. Conarty, J.A. Preston, C.L. Plesnicher, R.A. Dweik, S.C. Erzurum, Human vascular endothelial cells express oxytocin receptors, Endocrinology 140 (1999) 1301–1309. [10] T.D. Schmittgen, B.A. Zakrajsek, A.G. Mills, V. Gorn, M.J. Singer, M.W. Reed, Quantitative reverse transcription-polymerase chain reaction to study mRNA decay: comparison of endpoint and real-time methods, Anal. Biochem. 285 (2000) 194–204. [11] C. Caliceti, G. Aquila, M. Pannella, M.B. Morelli, C. Fortini, P. Pinton, et al., 17beta-estradiol enhances signalling mediated by VEGF-A-delta-like ligand 4notch1 axis in human endothelial cells, PLOS ONE 8 (2013) e71440. [12] F. Binet, P. Sapieha, ER stress and angiogenesis, Cell Metab. 22 (2015) 560–575. [13] H.J. Park, Y. Zhang, S.P. Georgescu, K.L. Johnson, D. Kong, J.B. Galper, Human umbilical vein endothelial cells and human dermal microvascular endothelial cells offer new insights into the relationship between lipid metabolism and angiogenesis, Stem Cell Rev. 2 (2006) 93–102. [14] G. Gimpl, F. Fahrenholz, The oxytocin receptor system: structure, function, and regulation, Physiol. Rev. 81 (2001) 629–683. [15] P. Cassoni, A. Sapino, T. Marrocco, B. Chini, G. Bussolati, Oxytocin and oxytocin receptors in cancer cells and proliferation, J. Neuroendocrinol. 16 (2004) 362– 364. [16] S.H. Kim, D.A. MacIntyre, A.C. Hanyaloglu, A.M. Blanks, S. Thornton, P.R. Bennett, et al., The oxytocin receptor antagonist, atosiban, activates pro-

934

[17] [18]

[19] [20]

J. Zhu et al. / Biomedicine & Pharmacotherapy 90 (2017) 928–934 inflammatory pathways in human amnion via G(alphai) signalling, Mol. Cell. Endocrinol. 420 (2016) 11–23. M.G. Cattaneo, B. Chini, L.M. Vicentini, Oxytocin stimulates migration and invasion in human endothelial cells, Br. J. Pharmacol. 153 (2008) 728–736. J.A. Diaz-Gonzalez, J. Russell, A. Rouzaut, I. Gil-Bazo, L. Montuenga, Targeting hypoxia and angiogenesis through HIF-1alpha inhibition, Cancer Biol. Ther. 4 (2005) 1055–1062. T. Gorojankina, Hedgehog signaling pathway: a novel model and molecular mechanisms of signal transduction, Cell. Mol. Life Sci. 73 (2016) 1317–1332. M.J. Marinissen, J.S. Gutkind, G-protein-coupled receptors and signaling networks: emerging paradigms, Trends Pharmacol. Sci. 22 (2001) 368–376.

[21] F. Gotschel, D. Berg, W. Gruber, C. Bender, M. Eberl, M. Friedel, et al., Synergism between Hedgehog-GLI and EGFR signaling in Hedgehog-responsive human medulloblastoma cells induces downregulation of canonical Hedgehog-target genes and stabilized expression of GLI1, PLOS ONE 8 (2013) e65403. [22] N.K. Agarwal, C.H. Kim, K. Kunkalla, H. Konno, Y. Tjendra, D. Kwon, et al., Active IKKbeta promotes the stability of GLI1 oncogene in diffuse large B-cell lymphoma, Blood 127 (2016) 605–615. [23] D. Cui, X. Chen, J. Yin, W. Wang, M. Lou, S. Gu, Aberrant activation of Hedgehog/ Gli1 pathway on angiogenesis in gliomas, Neurol. India 60 (2012) 589–596. [24] F. Zhang, M. Hao, H. Jin, Z. Yao, N. Lian, L. Wu, et al., Canonical hedgehog signaling regulates hepatic stellate cell-mediated angiogenesis in liver fibrosis, Br. J. Pharmacol. (2017).