Open Access

Recombinant chromosome 6 open reading frame 120 protein promotes angiogenesis and endothelial‑to‑mesenchymal transition in human umbilical vein endothelial cells via the PI3K/Akt signaling pathway

  • Authors:
    • Yingying Lin
    • Xin Wang
    • Yanyan Li
    • Xinyu Cui
    • Na Zhu
    • Xin Li
  • View Affiliations

  • Published online on: June 17, 2025     https://doi.org/10.3892/mmr.2025.13596
  • Article Number: 231
  • Copyright: © Lin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The vascular endothelium plays a pivotal role in modulating various physiological processes and its dysfunction is fundamental to the development of numerous vascular and non‑vascular diseases. Chromosome 6 open reading frame 120 (C6ORF120) has been implicated in cellular processes such as apoptosis, inflammation, immunomodulation and fibrosis. However, the specific effects of C6ORF120 on endothelial cell function remain unclear. The present study aimed to explore the potential role of C6ORF120 in endothelial dysfunction and its underlying molecular mechanisms. It synthesized recombinant C6ORF120 protein (rC6ORF120) and assessed its effects on human umbilical vein endothelial cells (HUVECs) through various functional assays, including the CCK‑8 assay for proliferation, scratch assay for migration and tube formation assay for angiogenesis. Additionally, immunofluorescence (IF) and western blotting (WB) were employed to evaluate endothelial‑mesenchymal transition (EndMT). The present study also quantified the expression of key proteins within the PI3K/Akt signaling pathway to elucidate its role in mediating the effects of rC6ORF120 on HUVECs. Treatment with rC6ORF120 significantly enhanced HUVEC proliferation (200 ng/ml vs. control at 72 h, 1.14±0.01 vs. 1.05±0.02; t=8.15; P<0.001) and induced phenotypic changes. In migration and angiogenesis assays, rC6ORF120‑treated HUVECs exhibited increased wound closure (37.69±2.74% vs. 66.16±6.13%; t=7.35; P=0.002) and angiogenesis assays showed significant improvements in tube formation parameters such as total tubule length (77,199.67±4,684.88 µm vs. 96,203.00±3,354.89 µm; t=5.71; P=0.002). WB and IF analyses both indicated that rC6ORF120 promotes EndMT in HUVECs. Furthermore, rC6ORF120 treatment increased PI3K/Akt phosphorylation significantly compared with controls (p‑PI3K; 1.57±0.18 vs. 1.00±0.00; t=5.64; P=0.005). LY294002 significantly reversed these effects on EndMT and angiogenesis (P<0.05), while the effect on cell migration was less pronounced (P=0.565). Our study highlights the critical role of C6ORF120 in HUVECs, promoting proliferation, migration, angiogenesis and EndMT, which are mediated, at least in part, by the PI3K/Akt pathway. 

Introduction

The human endothelial layer, composed of endothelial cells, is one of the largest organ systems in the human body, playing a critical role in maintaining physiological homeostasis through immune modulation, signal transduction, redox equilibrium and regulation of vascular tone (1). When endothelial integrity is compromised, endothelial cells undergo a series of pathological changes termed endothelial reprogramming, primarily characterized by endothelial-to-mesenchymal transition (EndMT) and pathological angiogenesis (2,3). During endothelial reprogramming, endothelial cells transition from a quiescent phenotype to an activated state, accompanied by enhanced migration and angiogenic capacities (3,4). Such endothelial dysfunction markedly contributes to the pathogenesis of various diseases, including inflammation, atherosclerosis, thrombosis, hypertension and fibrosis (5).

EndMT was first identified during endocardial differentiation (4) and has since been recognized as a pivotal mechanism involved in developmental anomalies, tumor progression and various fibrotic diseases (4,6,7). For instance, in liver fibrosis, EndMT is considered an important source of fibroblasts and contributes to hepatic stellate cell activation (8). Additionally, EndMT is closely associated with disease progression, pathological angiogenesis and drug resistance in conditions such as atherosclerosis and cancer (6,7). During the EndMT process, cells acquire an intermediate phenotype characterized by markedly enhanced angiogenic, migratory and proliferative capabilities (9,10). Thus, elucidating the mechanisms underlying endothelial dysfunction and reprogramming could provide valuable insights into the pathogenesis of these diseases and potentially identify novel therapeutic targets.

Chromosome 6 open reading frame 120 (C6ORF120) is a recently identified secreted protein implicated in multiple physiological and pathological processes. Previous studies have demonstrated its association with various diseases, including AIDS (11), diabetes (12), autoimmune hepatitis (1315) and liver fibrosis (16). Our prior studies revealed that C6ORF120 could regulate physiological and pathological processes such as apoptosis, immune response and fibrogenesis through its effects on hepatocytes (17), CD4 T lymphocytes (15) and hepatic stellate cells (16). However, the role of C6ORF120 in endothelial cells has not yet been explored. The present study aimed to investigate its potential involvement in endothelial cell biology, as understanding the function of C6ORF120 in endothelial cells could provide new insights into not only liver diseases but also vascular and fibrosis-related conditions beyond the liver. It was hypothesized that C6ORF120 may play a previously unexplored role in endothelial dysfunction, which could have implications for a wide range of vascular and fibrotic diseases.

Human umbilical vein endothelial cells (HUVECs), due to their representative and accessible nature, are extensively utilized as a classical endothelial cell model in vascular biology research (18,19). Therefore, the present study aimed to investigate the effects of C6ORF120 on endothelial cell functions (particularly EndMT, angiogenesis and migration) in HUVECs and to further elucidate the underlying molecular mechanisms, thereby providing insights into the role of C6ORF120 in endothelial dysfunction.

Materials and methods

Cell culture

The HUVECs used in the present study were purchased from American Type Culture Collection (cat. no. CRL-1730) through the Institute of Infectious Diseases, Ditan Hospital, Beijing, China (20). These cells were obtained at passage 2 and used between passages 2 and 5 in all experiments (21). Cells were cultured in Dulbecco's Modified Eagle's Medium (DMEM; cat. no. A4192101; Gibco; Thermo Fisher Scientific, Inc.), supplemented with 1% penicillin/streptomycin (cat. no. 15140-122; Gibco; Thermo Fisher Scientific, Inc.) and 10% fetal bovine serum (FBS; cat. no. A5670701; Gibco; Thermo Fisher Scientific, Inc.) and incubated in a humidified atmosphere containing 5% CO2 at 37°C. HUVECs were passaged when reaching 80% confluence.

Cell treatments

HUVECs in the logarithmic growth phase were seeded in 6-well plates and serum-starved in serum-free DMEM for 24 h before treatment. Cells were subsequently treated with 2 ml of medium containing recombinant C6ORF120 protein (rC6ORF120; Cusabio Technology, LLC) at indicated concentrations (15,16), TGF-β1 (10 ng/ml; BioLegend, Inc.) as a positive control (22), or the PI3K inhibitor LY294002 (10 µmol; MedChemExpress) for mechanistic studies (23,24). The control group was cultured with normal medium Each experiment was independently repeated at least three times and cells were harvested at predetermined time points for subsequent analysis.

CCK-8 assay

HUVECs (5,000 cells/well) were seeded into 96-well plates and treated with rC6ORF120 at concentrations of 0, 100, 200 and 400 ng/ml in a volume of 100 µl per well. After incubation for 24, 48 and 72 h, cell viability was evaluated using a Cell Counting Kit-8 (CCK-8; Shanghai Topscience Co., Ltd.) according to the manufacturer's instructions (25). Specifically, 10 µl of CCK-8 solution was added to each well, followed by incubation at 37°C for 2 h. Absorbance was measured at 450 nm using a microplate reader. Each condition was tested in triplicate and the experiment was independently repeated three times.

Cell morphology assay

HUVECs were seeded in 6-well plates at a density of 2×105 cells per well and treated with rC6ORF120 at concentrations of 0, 100, 200 and 400 ng/ml. Morphological changes were observed under an inverted phase-contrast microscope after 48 h of treatment and after continuous stimulation for five passages (26). Changes in cell shape (such as elongation, loss of cobblestone appearance and spindle-like transformation) were recorded. Representative images were captured from three randomly selected fields in each well and the experiments were independently repeated three times.

Western blotting

Cells were lysed in RIPA buffer (Beyotime Institute of Biotechnology) supplemented with phosphatase inhibitors and PMSF on ice for 1 h, followed by centrifugation at 12,000 × g for 15 min at 4°C. Protein concentrations were determined using a BCA protein assay kit (Beyotime Institute of Biotechnology). Equal amounts of protein (30 µg) were separated by 10% SDS-PAGE and transferred onto PVDF membranes (Millipore; Merck KGaA). After blocking with 5% skimmed milk for 1 h at room temperature, membranes were incubated overnight at 4°C with primary antibodies against CD31 (cat. no. 28083-1-AP; 1:1,000; Proteintech Group, Inc.), VE-cadherin (cat. no. 66804-1-IG; 1:1,000; Proteintech Group, Inc.), Vimentin (cat. no. 10366-1-AP; 1:5,000; Proteintech Group, Inc.), α-smooth muscle actin (α-SMA; cat. no. 67735-1-IG; 1:5,000; Proteintech Group, Inc.), AKT (cat. no. 60203-2-Ig; 1:1,000; Proteintech Group, Inc.), phosphorylated (p-)AKT (cat. no. 66444-1-Ig; 1:1,000; Proteintech Group, Inc.), PI3K (cat. no. 60225-1-Ig; 1:1,000; Proteintech Group, Inc.), p-PI3K (cat. no. ab182651; 1:1,000; Abcam) and GAPDH (cat. no. 60004-1-Ig; 1:5,000; Proteintech Group, Inc.). Membranes were washed three times with tris-buffered saline containing 0.1% Tween-20 (TBST) and incubated with HRP-conjugated secondary antibodies (anti-rabbit: cat. no. SA00001-2; 1:5,000; anti-mouse: cat. no. SA00001-1; 1:5,000; both Proteintech Group, Inc.) for 1 h at room temperature. Bands were visualized using chemiluminescence detection as previously described (27). Experiments were independently repeated three times.

Immunofluorescence (IF) staining assay

HUVECs were seeded on sterile glass coverslips (1×105 cells/ml) were fixed with 4% paraformaldehyde for 20 min, permeabilized with 0.05% Triton X-100 for 15 min and blocked with 5% bovine serum albumin (BSA) for 1 h all at 37°C. Cells were then incubated with primary antibodies against CD31 (cat. no. 28083-1-AP; 1:500; Proteintech Group, Inc.) and α-SMA (cat. no. 14395-1-AP; 1:500; Proteintech Group, Inc.) overnight at 4°C. After washing with PBS, cells were incubated with Alexa Fluor 555-conjugated secondary antibodies (cat. no. ab150078; 1:200; Abcam) for 1 h at room temperature in the dark. Nuclei were counterstained with DAPI (Beijing Solarbio Science & Technology Co., Ltd.) for 10 min at room temperature (28). Fluorescence signals were visualized and imaged using a fluorescence microscope. ImageJ software (version 1.54; National Institutes of Health) was used for quantitative analysis of fluorescence intensity. Each condition was tested in triplicate and three independent experiments were conducted.

Wound healing assay

Before cell seeding, the reverse side of 6-well plates was marked with horizontal lines using a black marker to ensure consistent scratch locations. HUVECs were cultured in 6-well plates and grown to 95% confluence. A linear scratch was generated using a 10 µl pipette tip. Cells were washed gently with PBS to remove detached cells and subsequently cultured with serum-free medium corresponding to each treatment group. Wound healing was monitored at 0 and 48 h using a light microscope. Images were analyzed using ImageJ software (version 1.54; National Institutes of Health) to quantify wound closure percentage (29). Experiments were independently repeated three times.

Tube formation assay

Matrigel (Corning, Inc.) was evenly applied onto 24-well plates at 100 µl per well and incubated at 37°C for 1 h to solidify. HUVECs suspended in medium were seeded at a density of 1.5×105 cells per well in a volume of 250 µl. After 8 h of incubation, tube-like structures were visualized using an inverted microscope and tube formation parameters, including the total vessel length and number of junctions, were quantified by ImageJ software (version 1.54) (30). Each experiment was independently repeated three times.

Statistical analyses

Data analysis was conducted using GraphPad Prism 6 software (Dotmatics). Results were expressed as mean ± standard deviation. Statistical comparisons between two groups were performed using unpaired Student's t-tests and multiple group comparisons were conducted using one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

rC6ORF120 affects the viability and morphology of HUVECs

HUVEC viability significantly increased in a concentration-dependent manner after treatment with rC6ORF120 at 24, 48 and 72 h (Fig. 1A). Specifically, at 72 h, treatment with 200 ng/ml (200 ng/ml vs. control: 1.14±0.01 vs. 1.05±0.02; t=8.15; P<0.001) and 400 ng/ml (400 ng/ml vs. control: 1.16±0.02 vs. 1.05±0.02; t=6.83; P<0.001) rC6ORF120 markedly enhanced cell viability compared to the control. Due to comparable viability at 200 and 400 ng/ml (t=1.05, P=0.353), 200 ng/ml was selected for subsequent experiments.

Morphologically, control HUVECs exhibited a typical cobblestone-like morphology, whereas those exposed to rC6ORF120 underwent a time- and concentration-dependent morphological shift towards a spindle-like, fibroblast-like appearance (Fig. 1B). This transformation was more pronounced after long-term exposure compared to short-term treatment, implying a potential role of rC6ORF120 in modulating endothelial cell shape and phenotype.

rC6ORF120 induces EndMT in HUVECs

The morphological observations of HUVECs following rC6ORF120 treatment prompted the hypothesis that these cells might undergo EndMT. To confirm EndMT induction by rC6ORF120, relevant protein markers were assessed by western blotting (WB) using TGF-β1 as a positive control (4,31). Compared with controls, rC6ORF120-treated HUVECs showed significantly reduced endothelial markers CD31 (0.60±0.08 vs. control 1.00; t=8.32; P<0.001) and VE-cadherin (0.56±0.07 vs. control 1.00; t=11.39; P<0.001), along with significantly increased mesenchymal markers α-SMA (1.50±0.16 vs. control 1.00; t=5.48, P=0.005) and Vimentin (1.35±0.05 vs. control 1.00; t=11.98, P=0.007; Fig. 2A-E).

IF staining further validated these findings, indicating significantly decreased fluorescence intensity for CD31 (0.77±0.04 vs. 1.00±0.01; t=9.10; P<0.001) and increased intensity for α-SMA (1.33±0.01 vs. 1.00±0.08; t=6.57, P=0.003) in the rC6ORF120-treated group compared to control (Fig. 3). Collectively, the results indicated that rC6ORF120 induced EndMT in HUVECs, as evidenced by phenotypic changes and alterations in marker expression.

rC6ORF120 promotes migration and angiogenesis in HUVECs

Mesenchymal cells are recognized for their higher migratory capacity relative to endothelial cells, a characteristic that is often accentuated during EndMT (1). Cell migration analysis via wound-healing assay demonstrated significantly increased wound closure in rC6ORF120-treated HUVECs compared with control (66.16±6.13 vs. 37.69±2.74%; t=7.35, P=0.002), indicating enhanced migratory capability (Fig. 4A and B).

Considering that angiogenesis is frequently associated with cell migration and EndMT, the present study further investigated the influence of rC6ORF120 on HUVEC tube-forming ability. Tube formation assays further revealed that rC6ORF120 significantly promoted angiogenesis, reflected by increased numbers of vascular junctions (990.00±29.87 vs. 766.00±76.37; t=4.73, P=0.009) and greater total vessel length (96203.00±3354.89 µm vs. 77199.67±4684.88 µm; t=5.71, P=0.005) compared with control (Fig. 4C-E).

rC6ORF120 activates the PI3K/Akt signaling pathway in HUVECs

Building on our previous research that suggested C6ORF120 could affect the PI3K/Akt signaling pathway (16). This led us to hypothesize that rC6ORF120 might similarly regulate this pathway in HUVECs. WB results indicated that rC6ORF120 significantly increased phosphorylation of PI3K (1.57±0.18 vs. control 1.00; t=5.64, P=0.005) and Akt (1.55±0.09 vs. control 1.00; t=11.12; P<0.001), confirming activation of the PI3K/Akt signaling pathway in HUVECs (Fig. 5).

rC6ORF120 promotes EndMT in HUVECs via the PI3K/Akt signaling pathway

To further explore the mechanism underlying rC6ORF120-induced EndMT, cells were pretreated with the PI3K inhibitor LY294002 prior to rC6ORF120 exposure. WB analysis demonstrated significant reversal of EndMT marker alterations: CD31 (1.18±0.12 vs. 0.65±0.06; t=7.00; P<0.001) and VE-cadherin (1.48±0.21 vs. 0.74±0.07; t=5.92, P=0.004) expression increased, while α-SMA (0.83±0.14 vs. 1.37±0.15; t=4.52, P=0.011) and Vimentin (0.82±0.17 vs. 1.35±0.12; t=4.50, P=0.013) expression decreased in LY294002 + rC6ORF120-treated cells compared with cells treated with rC6ORF120 alone (Fig. 6). These results were further confirmed by immunofluorescence staining (Fig. 7), indicating PI3K/Akt pathway as a critical mediator of rC6ORF120-induced EndMT.

Differential role of PI3K/Akt pathway in rC6ORF120-mediated migration and angiogenesis

The present study further investigated the mechanisms underlying the influence of rC6ORF120 on HUVEC migration and angiogenesis. Notably, LY294002 did not significantly affect the migratory capacity (63.66±3.23% vs. 66.16±6.13%; t=0.63, P=0.565) (Fig. 8A and B) although LY294002 significantly reversed the angiogenic effects of rC6ORF120 (number of junctions: 709.67±55.64 vs. 990.00±29.87; t=7.69, P=0.002; total tubule length: 73579.33±4339.87 µm vs. 96203.00±3354.89 µm; t=4.76, P=0.009) (Fig. 8C-E). These findings indicated that while angiogenesis induced by rC6ORF120 depends heavily on PI3K/Akt signaling, migration enhancement probably involves alternative or redundant signaling pathways.

Discussion

Endothelial dysfunction plays a pivotal role in various pathological conditions and identifying key molecules and regulatory pathways involved in endothelial growth factor signaling may uncover novel therapeutic strategies for related diseases (32). Endothelial cells not only maintain vascular integrity as a mechanical barrier but also regulate critical physiological processes, including homeostasis, immune response and vascular tone (1). Endothelial dysfunction is increasingly recognized as a central pathological event underlying cardiovascular, metabolic and fibrotic disorders. The present study, for the first time to the best of the authors' knowledge, identified novel biological functions of the secreted protein C6ORF120 in endothelial cell biology, demonstrating that rC6ORF120 promotes proliferation, migration, angiogenesis and EndMT in HUVECs, at least partly through activating the PI3K/Akt signaling pathway. This work is significant in directly establishing a link between C6ORF120 and endothelial dysfunction, thus broadening the functional repertoire of this protein.

The present study demonstrated that rC6ORF120 induced typical features of EndMT in HUVECs, including loss of endothelial markers (VE-cadherin and CD31), gain of mesenchymal markers (α-SMA and Vimentin), cytoskeletal remodeling and increased extracellular matrix production (33,34). EndMT is considered a form of cellular reprogramming, partial or complete, that enhances endothelial cell migratory and proliferative capacity (35,36). In line with this, it was also observed that rC6ORF120 significantly promoted migration and tube formation in HUVECs, highlighting its role in remodeling endothelial phenotype.

Mechanistically, our previous work suggested that C6ORF120 can modulate the PI3K/Akt pathway (16), which has also been implicated in regulating endothelial dysfunction in studies by Zhou et al (37) and He et al (38). The findings of the present study further identified rC6ORF120 as a novel upstream activator of PI3K/Akt signaling in HUVECs. Notably, pharmacological inhibition of this pathway with LY294002 effectively reversed rC6ORF120-induced EndMT and angiogenesis. However, the enhanced migratory capacity of HUVECs persisted, suggesting that rC6ORF120-mediated migration may be driven by PI3K/Akt-independent mechanisms.

Although endothelial migration and angiogenesis are functionally related, they are regulated by partially distinct signaling programs. Angiogenesis is a complex multistep process involving matrix degradation, cell proliferation, migration, lumen formation and vessel maturation (39). Migration alone cannot complete this process; thus, distinct signaling controls each phase (40). The inability of LY294002 to inhibit rC6ORF120-induced migration suggests compensatory signaling involvement. Indeed, studies have shown that endothelial behavior is shaped by a network of partially redundant pathways, including MAPK, VEGF/VEGFR, Notch and Rho-GTPases (4144). Moreover, in other cell types such as HepG2, the PI3K/Akt pathway modulates diverse processes (including autophagy, metabolism and oxidative stress) with function-specific variation in sensitivity (45,46). This could explain the functional divergence observed in our system.

As a secreted protein, rC6ORF120 may interact with multiple membrane receptors and initiate crosstalk across downstream signaling cascades, collectively influencing endothelial phenotype (47). For instance, class III semaphorins such as Sema3d modulate endothelial migration through distinct and parallel signaling routes involving PlxnD1 and Nrp1 (48). This supports the hypothesis that rC6ORF120 probably exerts its effects via multiple converging and compensatory signaling pathways. Future studies are needed to identify the key downstream effectors mediating rC6ORF120-induced migration, which may help clarify its broader regulatory network in endothelial dysfunction.

The present study offers two major innovations: Firstly, it provided the first evidence that rC6ORF120 regulates endothelial phenotype through both PI3K/Akt-dependent- and -independent mechanisms; and secondly it uncovered a previously unrecognized function of C6ORF120 in vascular biology beyond its known roles in immune modulation and fibrosis, suggesting its potential as a therapeutic target for vascular-related diseases. These findings contribute to a deeper understanding of the mechanisms driving endothelial dysfunction and may inform the development of new strategies for disease intervention.

Despite these strengths, several limitations should be noted. First, the present study is limited to in vitro experiments using HUVECs, which may not fully replicate the in vivo endothelial microenvironment. Second, while PI3K/Akt was identified as a major signaling mediator, other potential pathways such as MAPK, Rho/ROCK and Notch were not examined. Third, the specific receptors or upstream binding partners of rC6ORF120 remain unknown, which restricts comprehensive pathway mapping. Lastly, the long-term effects of rC6ORF120-induced EndMT and angiogenesis were not evaluated in animal disease models.

The present study provided novel insights into the functional role of C6ORF120 in endothelial biology and highlight its potential translational relevance. The ability of rC6ORF120 to induce EndMT and promote angiogenesis suggests its involvement in pathological vascular remodeling, which underlies a broad spectrum of diseases, including atherosclerosis, hypertension, fibrosis and tumor progression. Given that C6ORF120 is a secreted protein, it holds promise not only as a biomarker for endothelial plasticity but also as a therapeutic target for modulating vascular dysfunction. In particular, targeting C6ORF120 or its downstream PI3K/Akt signaling axis may offer novel strategies for preventing or reversing endothelial maladaptation in cardiovascular and fibrotic disorders. Future studies evaluating circulating C6ORF120 levels and functional inhibition in in vivo disease models are warranted to further define its clinical utility.

In conclusion, the present study identified rC6ORF120 as a novel regulator of endothelial dysfunction, capable of promoting EndMT, angiogenesis and migration through both PI3K/Akt-dependent and independent pathways. These findings expanded the functional spectrum of C6ORF120 and provided new insights into endothelial regulation with potential therapeutic implications, thereby opening new avenues for research in vascular biology and translational medicine.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Special Fund of Capital Health Research and Development (grant no. 2024-1-1203), the Dengfeng Talent Support Program of Beijing Municipal Administration of Hospitals (grant no. DFL20221601) and the High-level Public Health Technical Personnel Construction Project (grant no. Subject leaders-03-21).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

YiL, XW and XL conceived the study, designed the methodology and performed the formal analysis. YaL, XC and NZ conducted the investigation. YiL and XW prepared the original draft, while YaL, XC, NZ and XL reviewed and edited the manuscript. XL managed the project. XL secured funding. YiL, XW and XL confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Xu Y and Kovacic JC: Endothelial to mesenchymal transition in health and disease. Annu Rev Physiol. 85:245–267. 2023. View Article : Google Scholar : PubMed/NCBI

2 

Zhang L, Wu X and Hong L: Endothelial reprogramming in atherosclerosis. Bioengineering (Basel). 11:3252024. View Article : Google Scholar : PubMed/NCBI

3 

Liu Y, Wu Z, Li Y, Chen Y, Zhao X, Wu M and Xia Y: Metabolic reprogramming and interventions in angiogenesis. J Adv Res. 70:323–338. 2024. View Article : Google Scholar : PubMed/NCBI

4 

Piera-Velazquez S and Jimenez SA: Endothelial to mesenchymal transition: Role in physiology and in the pathogenesis of human diseases. Physiol Rev. 99:1281–1324. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Naderi-Meshkin H and Setyaningsih WAW: Endothelial cell dysfunction: Onset, progression, and consequences. Front Biosci (Landmark Ed). 29:2232024. View Article : Google Scholar : PubMed/NCBI

6 

Singh A, Bhatt KS, Nguyen HC, Frisbee JC and Singh KK: Endothelial-to-mesenchymal transition in cardiovascular pathophysiology. Int J Mol Sci. 25:61802024. View Article : Google Scholar : PubMed/NCBI

7 

Clere N, Renault S and Corre I: Endothelial-to-mesenchymal transition in cancer. Front Cell Dev Biol. 8:7472020. View Article : Google Scholar : PubMed/NCBI

8 

Poisson J, Lemoinne S, Boulanger C, Durand F, Moreau R, Valla D and Rautou PE: Liver sinusoidal endothelial cells: Physiology and role in liver diseases. J Hepatol. 66:212–227. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Medici D and Kalluri R: Endothelial-mesenchymal transition and its contribution to the emergence of stem cell phenotype. Semin Cancer Biol. 22:379–384. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Man S, Duffhues GS, Dijke PT and Baker D: The therapeutic potential of targeting the endothelial-to-mesenchymal transition. Angiogenesis. 22:3–13. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Li X, Qiao Y, Chang LS, Xiao F, Lu LH, Hao XH, Zhang RW, Wu H and Wei HS: Role of C6ORF120, an N-glycosylated protein, is implicated in apoptosis of CD4+ T lymphocytes. Chin Med J (Engl). 124:3560–3567. 2011.PubMed/NCBI

12 

Bradfield JP, Qu HQ, Wang K, Zhang H, Sleiman PM, Kim CE, Mentch FD, Qiu H, Glessner JT, Thomas KA, et al: A genome-wide meta-analysis of six type 1 diabetes cohorts identifies multiple associated loci. PLoS Genet. 7:e10022932011. View Article : Google Scholar : PubMed/NCBI

13 

Wu Y, Zhang R, Song X, Han X, Zhang J and Li X: C6orf120 gene knockout in rats mitigates concanavalin A-induced autoimmune hepatitis via regulating NKT cells. Cell Immunol. 371:1044672022. View Article : Google Scholar : PubMed/NCBI

14 

Zhang M, Ma H, Zhang J, Song XC, Ye XH, Li YF, Zhang YF, He LL, Wei HS and Li X: Deletion of the C6orf120 gene with unknown function ameliorates autoimmune hepatitis induced by concanavalin A. Cell Immunol. 331:9–15. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Liu H, Wang X, Wang P, Wang YQ, Yi YY and Li X: Novel protein C6ORF120 promotes apoptosis through mitochondria-dependent pathway in CD4 +T lymphocytes. Biomed Environ Sci. 36:639–643. 2023.PubMed/NCBI

16 

Wang X, Liu H, Wang Y, Wang P, Yi Y, Lin Y and Li X: Novel protein C6ORF120 promotes liver fibrosis by activating hepatic stellate cells through the PI3K/Akt/mTOR pathway. J Gastroenterol Hepatol. 39:1422–1430. 2024. View Article : Google Scholar : PubMed/NCBI

17 

Zhang J, Zhang M, Ma H, Song XC, Wu YN, Zhang R, He LL, Ye XH, Gao MX and Li X: C6orf120 gene deficiency may be vulnerable to carbon tetra-chloride induced acute hepatic injury in rats. Arch Med Sci. 18:1626–1637. 2020.PubMed/NCBI

18 

Jin X, Fu W, Zhou J, Shuai N, Yang Y and Wang B: Oxymatrine attenuates oxidized low-density lipoprotein-induced HUVEC injury by inhibiting NLRP3 inflammasome-mediated pyrop-tosis via the activation of the SIRT1/Nrf2 signaling pathway. Int J Mol Med. 48:1872021. View Article : Google Scholar : PubMed/NCBI

19 

Arakelian L, Lion J, Churlaud G, Bargui R, Thierry B, Mutabazi E, Bruneval P, Alberdi AJ, Doliger C, Veyssiere M, et al: 20-Endothelial CD34 expression and regulation of immune cell response in-vitro. Sci Rep. 13:135122023. View Article : Google Scholar : PubMed/NCBI

20 

Duranova H, Kuzelova L, Borotova P, Simora V and Fialkova V: Human umbilical vein endothelial cells as a versatile cellular model system in diverse experimental paradigms: An ultrastructural perspective. Microsc Microanal. 30:419–439. 2024. View Article : Google Scholar : PubMed/NCBI

21 

Gong L, Lei Y, Liu Y, Tan F, Li S, Wang X, Xu M, Cai W, Du B, Xu F, et al: Vaccarin prevents ox-LDL-induced HUVEC EndMT, inflammation and apoptosis by suppressing ROS/p38 MAPK signaling. Am J Transl Res. 11:2140–2154. 2019.PubMed/NCBI

22 

Zhang Y, Liu J, Zou T, Qi Y, Yi B, Dissanayaka WL and Zhang C: DPSCs treated by TGF-β1 regulate angiogenic sprouting of three-dimensionally co-cultured HUVECs and DPSCs through VEGF-Ang-Tie2 signaling. Stem Cell Res Ther. 12:2812021. View Article : Google Scholar : PubMed/NCBI

23 

Wu X, Pu L, Chen W, Zhao Q, Wu G, Li D and Zhu H: LY294002 attenuates inflammatory response in endotoxin-induced uveitis by downregulating JAK3 and inactivating the PI3K/Akt signaling. Immunopharmacol Immunotoxicol. 44:510–518. 2022. View Article : Google Scholar : PubMed/NCBI

24 

Eskiler GG and Ozturk M: Therapeutic potential of the PI3K inhibitor LY294002 and PARP inhibitor talazoparib combination in BRCA-deficient triple negative breast cancer cells. Cell Signal. 91:1102292022. View Article : Google Scholar : PubMed/NCBI

25 

Ma Q, Yang F, Huang B, Pan X, Li W, Yu T, Wang X, Ran L, Qian K, Li H, et al: CircARID1A binds to IGF2BP3 in gastric cancer and promotes cancer proliferation by forming a circARID1A-IGF2BP3-SLC7A5 RNA-protein ternary complex. J Exp Clin Cancer Res. 41:2512022. View Article : Google Scholar : PubMed/NCBI

26 

Zeng Z, Inoue K, Sun H, Leng T, Feng X, Zhu L and Xiong ZG: TRPM7 regulates vascular endothelial cell adhesion and tube formation. Am J Physiol Cell Physiol. 308:C308–C318. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Wang W, Li Y and Zhang Y, Ye T, Wang K, Li S and Zhang Y: SIRT1 mediates the inhibitory ef-fect of Dapagliflozin on EndMT by inhibiting the acetylation of endothelium notch1. Cardiovasc Diabetol. 22:3312023. View Article : Google Scholar : PubMed/NCBI

28 

Zhang Z, Guo Q, Ma C, Zhao Z, Shi Q, Yu H, Rao L and Li M: USF1 transcriptionally activates USP14 to drive atherosclerosis by promoting EndMT through NLRC5/Smad2/3 axis. Mol Med. 30:322024. View Article : Google Scholar : PubMed/NCBI

29 

Xie X, Qu P, Wu H, Liu P, Luo J, Chi J, Liu X, Chen X and Xu C: Circulating exosomal miR-21 mediates HUVEC proliferation and migration through PTEN/PI3K/AKT in Crohn's disease. Ann Transl Med. 10:2582022. View Article : Google Scholar : PubMed/NCBI

30 

Wang W, Li H, Qian Y, Li M, Deng M, Bi D and Zou J: ALKBH5 regulates corneal neovascularization by Mediating FOXM1 M6A demethylation. Invest Ophthalmol Vis Sci. 65:342024. View Article : Google Scholar

31 

Sobierajska K, Wawro ME, Ciszewski WM and Niewiarowska J: Transforming growth Factor-β receptor internalization via caveolae is regulated by tubulin-β2 and tubulin-β3 during endothelial-mesenchymal transition. Am J Pathol. 189:2531–2546. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Zhang L, Ge T and Cui J: FLI-1-driven regulation of endothelial cells in human diseases. J Transl Med. 22:7402024. View Article : Google Scholar : PubMed/NCBI

33 

Ciszewski WM, Woźniak LA and Sobierajska K: Diverse roles of SARS-CoV-2 spike and nucleocapsid proteins in EndMT stimulation through the TGF-β-MRTF axis inhibited by aspirin. Cell Commun Signal. 22:2962024. View Article : Google Scholar : PubMed/NCBI

34 

Wu X, Du X, Yang Y, Liu X, Liu X, Zhang N, Li Y, Jiang X, Jiang Y and Yang Z: Inhibition of miR-122 reduced atherosclerotic lesion formation by regulating NPAS3-mediated endothelial to mesenchymal transition. Life Sci. 265:1188162021. View Article : Google Scholar : PubMed/NCBI

35 

Hu Z, Wang J, Pan T, Li X, Tao C, Wu Y, Wang X, Zhang Z, Liu Y, Zhang W, et al: The exosome-transmitted lncRNA LOC100132249 induces endothelial dysfunction in diabetic retinopathy. Diabetes. 72:1307–1319. 2023. View Article : Google Scholar : PubMed/NCBI

36 

Bischoff J: Endothelial to mesenchymal transition-purposeful versus maladaptive differentiation. Circ Res. 124:1163–1165. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Zhou WW, Dai C, Liu WZ, Zhang C, Zhang Y, Yang GS, Guo QH, Li S, Yang HX and Li AY: Gentianella acuta improves TAC-induced cardiac remodel-ling by regulating the notch and PI3K/Akt/FOXO1/3 pathways. Biomed Pharmacother. 154:1135642022. View Article : Google Scholar : PubMed/NCBI

38 

He Y, Dan Y, Gao X, Huang L, Lv H and Chen J: DNMT1-mediated lncRNA MEG3 methylation accelerates endothelial-mesenchymal transition in diabetic retinopathy through the PI3K/Akt/mTOR signaling pathway. Am J Physiol Endocrinol Metab. 320:E598–E608. 2021. View Article : Google Scholar : PubMed/NCBI

39 

La Mendola D, Trincavelli ML and Martini C: Angiogenesis in disease. Int J Mol Sci. 23:109622022. View Article : Google Scholar : PubMed/NCBI

40 

Gentile MT, Pastorino O, Bifulco M and Colucci-D'Amato L: HUVEC tube-formation assay to evaluate the impact of natural products on angiogenesis. J Vis Exp. 24:e585912019.

41 

Yang DR, Wang MY, Zhang CL and Wang Y: Endothelial dysfunction in vascular complications of diabetes: A comprehensive review of mechanisms and implications. Front Endocrinol (Lausanne). 15:13592552024. View Article : Google Scholar : PubMed/NCBI

42 

Hall IF, Kishta F, Xu Y, Baker AH and Kovacic JC: Endothelial to mesenchymal transition: At the axis of cardiovascular health and disease. Cardiovasc Res. 120:223–236. 2024. View Article : Google Scholar : PubMed/NCBI

43 

Romano E, Rosa I, Fioretto BS and Manetti M: The contribution of endothelial cells to tissue fibrosis. Curr Opin Rheumatol. 36:52–60. 2024. View Article : Google Scholar : PubMed/NCBI

44 

Ciszewski WM, Wawro ME, Sacewicz-Hofman I and Sobierajska K: Cytoskeleton reorganization in EndMT-the role in cancer and fibrotic diseases. Int J Mol Sci. 22:116072021. View Article : Google Scholar : PubMed/NCBI

45 

Song C, Wang Z, Cao J, Dong Y and Chen Y: Hesperetin alleviates aflatoxin B1 induced liver toxicity in mice: Modulating lipid peroxidation and ferritin autophagy. Ecotoxicol Environ Saf. 284:1168542024. View Article : Google Scholar : PubMed/NCBI

46 

Zhou K, Xiao S, Cao S, Zhao C, Zhang M and Fu Y: Improvement of glucolipid metabolism and oxidative stress via modulating PI3K/Akt pathway in insulin resistance HepG2 cells by chickpea flavonoids. Food Chem X. 23:1016302024. View Article : Google Scholar : PubMed/NCBI

47 

Cao Y: 34-Lack of basic rationale in epithelial-mesenchymal transition and its related concepts. Cell Biosci. 14:1042024. View Article : Google Scholar : PubMed/NCBI

48 

Hamm MJ, Kirchmaier BC and Herzog W: Sema3d controls collective endothelial cell migration by distinct mechanisms via Nrp1 and PlxnD1. J Cell Biol. 215:415–430. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2025
Volume 32 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin Y, Wang X, Li Y, Cui X, Zhu N and Li X: Recombinant chromosome 6 open reading frame 120 protein promotes angiogenesis and endothelial‑to‑mesenchymal transition in human umbilical vein endothelial cells via the PI3K/Akt signaling pathway. Mol Med Rep 32: 231, 2025.
APA
Lin, Y., Wang, X., Li, Y., Cui, X., Zhu, N., & Li, X. (2025). Recombinant chromosome 6 open reading frame 120 protein promotes angiogenesis and endothelial‑to‑mesenchymal transition in human umbilical vein endothelial cells via the PI3K/Akt signaling pathway. Molecular Medicine Reports, 32, 231. https://doi.org/10.3892/mmr.2025.13596
MLA
Lin, Y., Wang, X., Li, Y., Cui, X., Zhu, N., Li, X."Recombinant chromosome 6 open reading frame 120 protein promotes angiogenesis and endothelial‑to‑mesenchymal transition in human umbilical vein endothelial cells via the PI3K/Akt signaling pathway". Molecular Medicine Reports 32.3 (2025): 231.
Chicago
Lin, Y., Wang, X., Li, Y., Cui, X., Zhu, N., Li, X."Recombinant chromosome 6 open reading frame 120 protein promotes angiogenesis and endothelial‑to‑mesenchymal transition in human umbilical vein endothelial cells via the PI3K/Akt signaling pathway". Molecular Medicine Reports 32, no. 3 (2025): 231. https://doi.org/10.3892/mmr.2025.13596