Open Access

Roles of endothelial cell specific molecule‑1 in tumor angiogenesis (Review)

  • Authors:
    • Jie Zhou
    • Ping Zhou
    • Jinfang Wang
    • Jie Song
  • View Affiliations

  • Published online on: February 1, 2024     https://doi.org/10.3892/ol.2024.14270
  • Article Number: 137
  • Copyright: © Zhou 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

Angiogenesis plays a crucial role in tumor growth and metastasis, and is heavily influenced by the tumor microenvironment (TME). Endothelial cell dysfunction is a key factor in tumor angiogenesis and is characterized by the aberrant expression of pro‑angiogenic factors. Endothelial cell specific molecule‑1 (ESM1), also known as endocan, is a marker of endothelial cell dysfunction. Although ESM1 is primarily expressed in normal endothelial cells, dysregulated ESM1 expression has been observed in human tumors and animal tumor models, and implicated in tumor growth, metastasis and angiogenesis. The precise role of ESM1 in tumor angiogenesis and its potential regulatory mechanisms are not yet conclusively defined. However, the aim of the present review was to explore the involvement of ESM1 in the process of tumor angiogenesis in the TME and the characteristics of neovascularization. In addition, the present review discusses the interaction between ESM1 and angiogenic factors, as well as the mechanisms through which ESM1 contributes to tumor angiogenesis. Furthermore, the reciprocal regulation between ESM1 and the TME is explored. Finally, the potential of targeting ESM1 as a therapeutic strategy for tumor angiogenesis is presented.

Introduction

Tumors pose a serious threat to human health, and their occurrence, progression and metastasis are influenced by several factors. These include angiogenesis, which plays a crucial role in tumor growth. The formation of new blood vessels leads to the supply oxygen and nutrients to the tumor, contributing to its growth and metastasis (1). Consequently, inhibition of angiogenesis has emerged as a promising approach for cancer treatment. At present, a number of anti-angiogenic drugs, including bevacizumab and sorafenib are available for clinical use (2). However, the limited effectiveness and development of resistance in certain patients have restricted the application of these drugs (2). Therefore, novel targets are being sought to identify an alternative strategy for the development of more effective anti-angiogenic drugs.

Endothelial cell-specific molecule-1 (ESM1), also known as endocan, is a proteoglycan that is released from cells and circulates in the bloodstream (3). Under normal physiological conditions, ESM1 is primarily expressed in the bronchial epithelium, as well as in vascular endothelial cells in the lung and kidneys (4). However, under pathological conditions, ESM1 is aberrantly expressed and interacts with its substrate proteins to promote abnormal cell adhesion, proliferation and angiogenesis (5). During tumor development, ESM1 is sporadically expressed in tumor cells and upregulated in tumor vascular endothelial cells, thereby modulating angiogenesis (610).

The present study is a comprehensive review of the importance of ESM1 in tumor angiogenesis, which aims to summarize the underlying mechanisms. By elucidating the role of ESM1 in tumor angiogenesis, it is hoped that valuable ideas and directions will be provided for the future development of more effective anti-angiogenic therapies.

Structure and expression of ESM1

Structure of ESM1

ESM1 was initially isolated from a human umbilical vein endothelial cell (HUVEC) cDNA library constructed by Lassalle et al (11) in 1996. It is a secreted proteoglycan composed of an 165-amino-acid polypeptide covalently linked to a sulfated dermal polysaccharide chain (11,12). The gene encoding human ESM1 is located on chromosome 5 at the q11.2 gene locus (13). This gene consists of three exons, exons 1–3. Exon 1 encodes an N-terminal cysteine-rich region that contains an epidermal growth factor (EGF)-like domain, while exon 2 encodes a phenylalanine-rich region that is considered to be involved in ESM1 function. Exon 3 encodes a cysteine-free C-terminal region of 33 amino acids, including a unique O-glycosylation site at serine 137 (13,14). The protein structure of ESM1 is illustrated in Fig. 1. The ESM1 protein detected in human serum and HUVEC supernatants has a molecular weight of 50 kDa. However, purified mature ESM1 obtained from the 293-ESM1 cell line is a 165-amino-acid protein with a predicted molecular weight of only 20 kDa, which indicates that post-translational modifications occur when ESM1 is secreted (15). The modification site has been identified to be serine 137, where a single dermatan sulfate polysaccharide chain is bound (15).

Expression and regulation of ESM1

ESM1 is physiologically expressed in various tissues with proliferative activity in the human body, including gastrointestinal glandular, bronchial, pulmonary capillary, glomerular and renal tubular tissues. Conversely, it is not expressed in tissues without proliferative properties, including large blood vessels and the spleen (5). However, expression and secretion of ESM1 can become abnormal under pathological conditions. In diseases associated with the endothelium, such as pneumonia, hypertension and coronary heart disease, the serum levels of ESM1 can be dysregulated; hence, ESM1 has been established as a potential marker for endothelial dysfunction (4,16,17). Furthermore, studies have revealed an association between aberrant ESM1 expression and the development of various tumors. For example, clinical investigations have observed increased ESM1 levels in secretions or serum from patients with nasopharyngeal, endometrial and ovarian carcinoma (18,19). In addition, elevated ESM1 expression has been observed in cancer cells, including cervical cancer, squamous cell cancer of the head and neck, esophageal cancer and hepatocellular cancer cells (6,8,20,21). Notably, ESM1 expression has been demonstrated to be increased in the vascular endothelial cells of prolactinoma and gastric cancer, indicating its involvement in tumor angiogenesis (22,23).

The expression of ESM1 is regulated by multiple factors. Inflammatory factors such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and lipopolysaccharide (LPS), as well as hypoxia-inducible factor (HIF), vascular endothelial growth factor (VEGF), high glucose, high salt and lead, can upregulate ESM1 (2426). By contrast, interferon-g, platelet-derived growth factor, angiotensin 2, endothelin 1 and insulin have inhibitory effects on ESM1 expression (27,28).

Processes of tumor angiogenesis

Regulation and major processes of tumor angiogenesis

Angiogenesis in tumors is regulated by a balance of pro- and anti-angiogenic factors. Neovascularization occurs when the levels of pro-angiogenic factors exceed those of anti-angiogenic factors (29).

Among pro-angiogenic factors, the VEGF family and its receptors have been extensively studied. The VEGF family includes VEGF subtypes A-E and placental growth factor, among which VEGF-A plays a major role in the regulation of angiogenesis (30). VEGF-A interacts with its receptors, primarily VEGF receptors 1 and 2 (VEGFR1/2), with a stronger affinity for VEGFR1 (31). However, the binding of VEGF-A to VEGFR-2 promotes mitosis and the permeability of vascular endothelial cells, leading to angiogenesis, and serves as the principal regulatory mechanism in angiogenesis (31). In the context of tumors, endothelial cells have been found to exhibit autocrine production of VEGF-A, while tumor cells exhibit paracrine secretion of VEGF-A (32). VEGF-A has been identified to be one of the most influential factors in tumor angiogenesis (32).

In addition to the VEGF/VEGFR-2 pathway, angiopoietin-2 (Ang2) and its receptor, which is known as Ang1 receptor or tyrosine-protein kinase receptor TEK, are also involved in tumor angiogenesis. Ang2 expression is normally low in dormant endothelial cells but can be upregulated by hypoxia, endothelial cell activation and VEGF-A (33). The upregulation of Ang2 expression in tumor endothelial cells increases vascular endothelial cell permeability, leading to tissue hypoxia and the subsequent upregulation of VEGF-A, thereby promoting angiogenesis (33).

Furthermore, tumor cells have been shown to secrete fibroblast growth factor (FGF) to stimulate endothelial cell proliferation (34). In addition, tumor-associated macrophages and fibroblasts secrete matrix metalloproteinases (MMPs) that degrade the extracellular matrix (ECM) and thereby facilitate tumor angiogenesis (35). These factors collectively contribute to the complex regulation of angiogenesis in tumors.

There are four main steps in the formation of tumor vessels. First, angiogenesis is initiated in response to stimulation from hypoxia, inflammation and reactive oxygen species in the tumor microenvironment (TME). Tumor cells release factors such as VEGF and FGF to induce angiogenesis through paracrine signaling (32). Endothelial cells, in turn, respond by autocrine signaling with increased VEGF production and Ang2 upregulation (33). Inflammatory, endothelial and stromal cells, among others, secrete MMPs, specifically MMP-2, −9 and −14 (36).

Secondly, vascular sprouting occurs as MMPs degrade the basement membrane (BM) and ECM. This weakens the connections between pericytes and endothelial cells, allowing endothelial cells in dormant vessels to sense angiogenic signals (36). In response, the endothelial cells at the leading edge of capillary growth are converted into tip cells, which are highly proliferative and can migrate towards the angiogenic signals (36,37).

Thirdly, vascular lumen formation occurs. Stalk cells, located behind the tip cells, proliferate and modify their morphology to establish adherent and tight junctions with neighboring endothelial cells. This process results in the formation of the vascular lumen (38,39).

Finally, vascular maturation occurs. Once the vascular lumen is formed, neovascularization occurs through a series of signaling events that recruit pericytes and facilitate the deposition of BMs. These processes contribute to the maturation of the newly formed vasculature. The various processes by which angiogenesis occurs in the TME are shown in Fig. 2.

Characteristics of tumor neovascularization

The distribution, morphology, structure and function of neovascularization in tumor tissues differ significantly from those of normal physiological blood vessels. In tumors, the distribution of blood vessels is not uniform, as a higher micro-vessel density is generally observed in the central region compared with the surrounding and normal tissues outside the tumor (40). However, certain studies have indicated that the blood vessel density within the tumor is lower than that in the surrounding tissue due to the production of anti-angiogenic factors by tumor cells and excessive matrix deposition, leading to hypoxia (40,41).

In addition to being unevenly distributed within the tumor, tumor blood vessels have distinct characteristics. They often exhibit a curved morphology, disorganized vascular networks and increased vascular permeability. This increased permeability is attributed to a reduction in pericytes, disruptions in intercellular adherent junctions and tight junctions, and abnormalities in BM structure, including uneven thicknesses and perforations (4245). These features provide favorable conditions for tumor metastasis, as shown in Fig. 2.

Regulation of ESM1 in tumor angiogenesis

Interaction of ESM1 with tumor angiogenesis factors

The role of ESM1 in angiogenesis has been established in various studies. For example, it has been revealed that ESM1 is specifically expressed in retinal endothelial tip cells during mouse development (46). In experiments with ESM1 knockout mice, it was observed that these cells had fewer filopodia and reduced phosphorylation of extracellular signal-regulated protein kinase (ERK) compared with those in wild-type mice, leading to delayed vascular development (46). Furthermore, ESM1 expression was found to be upregulated in mouse models of hypoxia-induced retinal vascular neovascularization and laser-induced choroidal vascular neovascularization (47). In addition, the administration of an intravitreal injection of ESM1 neutralizing antibody to the mice resulted in the significant downregulation of several vascular neovascularization-associated factors, including VEGFR1, VEGFR2, placental growth factor and MMP-2 (47).

The role of ESM1 in tumor angiogenesis has been confirmed through in vivo and in vitro experiments. For instance, a study showed that lung adenocarcinoma tumors subcutaneous transplanted in ESM1 knockout mice exhibited reduced neovascularization compared with those in wild-type mice (46). Similarly, the volume of subcutaneously transplanted tumors formed by ESM1 knockout ovarian cancer A2780 cells was smaller compared with these formed by control A2780 cells (48). In in vitro experiments, the tube formation ability of HUVECs was observed to decrease when they were incubated with supernatants from ESM1 knockdown A2780 cells (48). These findings suggested that ESM1 promotes angiogenesis in vivo and in vitro.

Further investigations into the mechanism of action of ESM1 have shown its involvement in the regulation of several angiogenic factors and receptors. For example, the knockdown of ESM1 in GH3 and MMQ rat prolactinoma cell lines led to the reduced expression of genes associated with angiogenesis, such as VEGFR2, von Willebrand factor and EGF receptor (EGFR) (22). Similar results were obtained when ESM1 was silenced in AN3CA and RL95-2 endometrial cancer cells, which resulted in the downregulation of the angiogenic factor receptors VEGFR1, VEGFR2 and EGFR, ultimately leading to reduced angiogenesis, while ESM1 overexpression had the opposite effects (49). ESM1 has been shown to influence the major angiogenic factor VEGF, as the knockdown of ESM1 decreased VEGF expression in human cervical cancer squamous cell carcinoma cells (50). Conversely, VEGF is able to regulate ESM1; in vivo experiments with VEGF-A knockout mice or mice injected with VEGFR2 blockers showed a reduction in the ESM1 expression of the mice (47). Studies have revealed that the binding of VEGF-A to VEGFR2 leads to the phosphorylation of VEGFR2 and the subsequent activation of downstream signaling pathways that regulate ESM1 expression (47,51). The interactions of ESM1 with factors associated with tumor angiogenesis are shown in Table I and Fig. 3.

Figure 3.

Roles and mechanisms of ESM1 in tumor angiogenesis. Solid lines indicate demonstrated mechanisms and dashed lines indicate possible mechanisms. ESM1 activates the NF-κB pathway in two ways: Via the upregulation of TNF-α and IL-8, and via promotion of binding between VEGF and VEGFR2. The activated NF-κB upregulates VEGF via the transcription factor HIF-1α, thereby promoting angiogenesis. ESM1 also activates the PI3K/AKT/mTOR pathway to upregulate MMPs, VEGF and HIF-1α by facilitating the binding of VEGF with VEGFR2. Regarding EGF and its receptor EGFR, ESM1 not only directly binds to EGFR but also facilitates the binding of EGF to EGFR, leading to activation of the EGFR signaling pathway. The activated EGFR, in turn, increases ESM1 expression through the JAK/STAT3 and ERK/ELK pathways, establishing a regulatory loop between ESM1 and EGFR. Increased ESM1 also upregulates angiogenesis-related factors such as VEGFR1, VEGFR2, EGFR and VWF, thereby promoting angiogenesis. In addition, activated EGFR can upregulate HIF-1α and MMPs through the PI3K/AKT/mTOR pathway. In the MAPK pathway, ESM1 increases the expression of VEGF through ERK, JNK and p38. As for the Notch pathway, the ligand DLL4 is upregulated by ESM1, thereby inhibiting VEGF expression. ESM1, endothelial cell specific molecule-1; NF-κB, nuclear factor-κB; TNF-α, tumor necrosis factor-α; IL-8, interleukin-8; VEGF, vascular endothelial growth factor; VEGFR1/2, VEGF receptor 1/2; PI3K, phosphoinositide 3-kinase; mTOR, mammalian target of rapamycin; MMPs, matrix metalloproteinases; HIF-1α, hypoxia-inducible factor-1α; EGF, epidermal growth factor; EGFR, EGF receptor; JAK, Janus kinase; STAT3, signal transducer and activator of transcription 3; ERK, extracellular signal-regulated kinase; ELK, ETS-like kinase; JNK, c-Jun N-terminal kinase; VWF, von Willebrand factor; DLL4, d-like ligand 4; P, phosphorylated; TAB1-3, TAK1-binding protein 1–3; TAK1, transforming growth factor-b-activated kinase 1; RAS, rat sarcoma; RAF, rapidly accelerated fibrosarcoma; MEK/MKK, mitogen-activated protein kinase kinase; NICD, Notch intracellular domain.

Table I.

Tumor angiogenesis factors and signaling pathways regulated by ESM1.

Table I.

Tumor angiogenesis factors and signaling pathways regulated by ESM1.

First author/s, yearIn vivo or in vitroESM1 expressionBiological importanceESM1-regulated genes/proteins/pathway(Refs.)
Cai et al, 2016In vitroDownInhibits the viability of GH3 and MMQ rat prolactinoma cell linesVEGFR2; VWF(22)
Rocha et al, 2014In vivo and in vitroDownInhibits retinal vascular outgrowthVEGF; ERK(46)
Su et al, 2018In vitroUpPromotes retinal neovascularizationVEGFR1, VEGFR2, PIGF, MMP-2; ERK; P38(47)
Li et al, 2023In vivo and in vitroDownInhibits ovarian cancer angiogenesisVEGF; PI3K/Akt/mTOR(48)
He et al, 2022In vitroUpPromotes endometrial cancer angiogenesisVEGFR1, VEGFR2, EGFR(49)
Li, 2023In vitroDownInhibits proliferation, invasion and apoptosis of SiHa and ME-180 cervical squamous cell carcinoma cell linesVEGF(50)
Yang et al, 2023In vivo and in vitroUpPromotes colorectal cancer angiogenesisMMP-2, MMP-3, MMP-9, VEGF, COX-2, HIF-1α; PI3K/Akt/mTOR(53)
Yang et al, 2020In vitro and in vivoUpPromotes non-small cell lung cancer growthEGF; EGFR; JAK/STAT3; ERK/ELK(55)
Lee et al, 2014In vitro and in vivoUpPromotes vascular cell permeabilityVEGFR1, VEGFR2, MAPK (ERK, JNK, P38)(58)
Kang et al, 2022In vivo and n vitroUp/downPromotes/inhibits breast cancer angiogenesisVEGF; DLL4(63)
Huang et al, 2021In vitroDownInhibits SW13 human adrenocortical carcinoma cell line growthDLL4(64)
Kumar and Mani, 2021In vitroUpPromotes NO and ROS in endothelial cellsNF-κB(68)

[i] ESM1, endothelial cell specific molecule-1; VEGF, vascular endothelial growth factor; VEGFR2, VEGF receptor 2; VWF, von Willebrand factor; PIGF, placental growth factor; MMP, matrix metalloproteinase; ERK, extracellular signal-regulated protein kinase; PI3K, phosphoinositide 3-kinase; mTOR, mammalian target of rapamycin; EGF, epidermal growth factor; EGFR, EGF receptor; COX-2, cyclooxygenase 2; HIF-1α, hypoxia-inducible factor-1α; JAK, Janus kinase; STAT3, signal transducer and activator of transcription 3; ELK, ERK/ETS-like-1 kinase; MAPK, mitogen-activated protein kinase; DLL4, d-like ligand 4; NO, nitric oxide; ROS, reactive oxygen species; NF-κB, nuclear factor-κB.

In summary, ESM1 promotes angiogenesis via the regulation of angiogenic factors, and there is a reciprocal relationship between ESM1 and angiogenic factors, which form a positive feedback loop.

Role of ESM1 in the signaling pathway of tumor angiogenesis

Limited research has been conducted on the specific signaling pathways by which ESM1 regulates tumor angiogenesis. However, existing studies suggest the involvement of several signaling pathways, including the phosphoinositide 3-kinase (PI3K)/AKT, mitogen-activated protein kinase (MAPK), Notch and nuclear factor-κB (NF-κB) pathways.

PI3K/AKT signaling pathway

PI3K and AKT play a critical role in various processes, including tumor growth, metabolism and angiogenesis (52). ESM1 has been found to act through the PI3K/AKT pathway during angiogenesis. For example, it was found that when HUVECs were cultured with the supernatants of SW480 and SW620 colorectal cancer cells transfected with ESM1 mimic, their tube-forming ability was increased (53). Further experiments demonstrated that the transfection of these cancer cells with ESM1 mimic upregulated the expression of MMP-2, MMP-3, MMP-9, VEGF, cyclooxygenase-2 and HIF-1α through the PI3K/AKT/mammalian target of rapamycin (mTOR) signaling pathway (53). Considering that ESM1 increases the binding of VEGF to VEGFR2 (46), and PI3K/AKT/mTOR is downstream of the VEGF/VEGFR2 pathway (54), this suggests that ESM1 activates the PI3K/AKT/mTOR pathway via the phosphorylation of VEGFR2, consequently upregulating the aforementioned angiogenesis-related factors. In a study of non-small cell lung cancer cells, it was observed that recombinant ESM1 directly bound to EGFR and facilitated the binding of EGF to EGFR, leading to activation of the EGFR signaling pathway (55). The activated EGFR, in turn, was indicated to upregulate ESM1 via Janus kinase/signal transducer and activator of transcription 3 signaling and ERK/ETS-like kinase, thereby establishing a regulatory loop between ESM1 and EGFR (55). In addition, another study revealed that the activated EGFR pathway sustains angiogenesis in solid tumors through the downstream PI3K/AKT/mTOR signaling pathway (56). Summaries of the mechanism by which ESM1 regulates tumor angiogenesis via the PI3K/AKT signaling pathway are shown in Table I and Fig. 3.

MAPK signaling pathway

The MAPK pathway, which includes the ERK, c-Jun N-terminal kinase (JNK) and p38 signaling pathways, plays a crucial role in a number of cellular processes, including cell proliferation, differentiation and apoptosis, angiogenesis and tumor metastasis (57). In vivo and in vitro experiments have confirmed that ESM1 regulates angiogenesis through the MAPK pathway. For example, in vivo experiments conducted using ESM1 knockout mice revealed a reduction in phosphorylated ERK in the retina, which led to delayed vascular development (46). In addition, in vitro experiments involving the stimulation of human retinal endothelial cells and HUVECs with recombinant ESM1 demonstrated the activation of MAPK (ERK, JNK and p38) signaling pathways associated with angiogenesis (47,58), which further confirms the relationship between ESM1 and the MAPK signaling pathway. Phosphorylated ERK, JNK and p38 can translocate into the nucleus and induce VEGF production, thereby regulating angiogenesis (57,59,60). Furthermore, since ESM1 maintains tumor angiogenesis through regulation of the EGFR signaling pathway, and rat sarcoma/rapidly accelerated fibrosarcoma/mitogen-activated protein kinase kinase/ERK and PI3K/AKT are the main signaling pathways downstream of EGFR (55,61), ESM1 can also regulate these pathways. Summaries of how ESM1 regulates tumor angiogenesis through the MAPK signaling pathway are shown in Table I and Fig. 3.

Notch signaling pathway

The Notch signaling pathway plays a critical role in normal cell differentiation, proliferation, apoptosis and angiogenesis under physiological conditions (62). In pathological conditions, it also regulates angiogenesis. For instance, ESM1 overexpression in the MDA-MB-231 human breast cancer cell line and mice enhances angiogenesis via the upregulation of VEGF and d-like ligand 4 (DLL4, a ligand of Notch1 in the Notch signaling) (63). In addition, ESM1 activates the DLL4/Notch signaling pathway in adrenocortical carcinoma cells (64) (Table I and Fig. 3). It is worth noting that activation of the DLL4/Notch pathway inhibits VEGF-induced neovascularization (65). This may appear contradictory to the angiogenic role of ESM1; however, it has been suggested that activation of the DLL4/Notch pathway contributes to vessel sprouting (66). Since the number of studies investigating the regulatory mechanisms of ESM1 in the DLL4/Notch pathway is limited, the precise role of the DLL4/Notch pathway in ESM1-mediated angiogenesis regulation remains unclear, emphasizing that further research is necessary.

NF-κB signaling pathway

The NF-κB pathway is crucial in tumor angiogenesis (67). ESM1 has been shown to regulate tumor angiogenesis through the NF-κB pathway. Recombinant ESM1 has been shown to activate NF-κB in HUVECs (68), and it is known that NF-κB is downstream of PI3K/AKT (69). Therefore, it is speculated that ESM1 activates NF-κB through the PI3K/AKT pathway. Furthermore, ESM1 upregulates inflammatory factors such as TNF-α and IL-8, both of which can activate NF-κB (58,70,71). Furthermore, NF-κB upregulates VEGF via HIF-1α, leading to angiogenesis (72). The regulation of tumor angiogenesis by ESM1 through the NF-κB signaling pathway is summarized in Table I and Fig. 3.

Interaction of ESM1 with the TME

Mutual reinforcement between hypoxia and ESM1

In addition to tumor cells, the TME comprises various components, including fibroblasts, endothelial cells, immune cells and ECM (73). Rapid and uncontrolled tumor growth, coupled with inadequate blood supply, leads to the TME typically being hypoxic (74). Hypoxia is mainly characterized by the production of HIF (75). Among the HIF family, HIF-1α is the primary transcription factor associated with hypoxia (76). Studies have confirmed the existence of a regulatory relationship between HIF-1α and ESM1. For example, when HUVECs were exposed to intermittent hypoxia, ESM1 expression was significantly increased in a time-dependent manner. However, the knockdown of HIF-1α expression led to a reduction in ESM1 levels under these conditions (77). The regulation of ESM1 expression by HIF-1α was shown to be mediated VEGF (77), as shown in Fig. 4. In addition, in human umbilical artery endothelial cells, HIF was found to facilitate the translocation of the transcription factor Forkhead box O1 (FoxO1) from the cytoplasm to the nucleus in response to hypoxia, thereby upregulating ESM1 levels (78), as shown in Fig. 4. These findings indicate that HIF-1α indirectly regulates ESM1 through VEGF and FoxO1. Moreover, ESM1 has been found to regulate HIF-1α. In vitro experiments demonstrated that ESM1 promoted drug resistance, proliferation, migration, invasion and epithelial-mesenchymal transition through HIF-1α in A549 and PC-9 non-small cell lung cancer cells following intermittent hypoxia (79). Similar results were observed in a mouse model of lung cancer in which the mice were exposed to chronic intermittent hypoxia and a small interfering RNA targeting ESM1 was delivered using a lentivirus (79), as shown in Fig. 4. It is evident that HIF-1α and ESM1 mutually regulate each other, forming a positive feedback loop. In summary, hypoxia induces the expression of ESM1 in the TME, and the elevation of ESM1 further exacerbates tissue hypoxia, ultimately promoting angiogenesis.

Mutual reinforcement between inflammation and ESM1

Inflammation is a prominent feature of the TME. Within the TME, inflammatory cells play a pivotal role in tumor development, growth and metastasis through the secretion of chemokines, inflammatory factors and growth factors (80,81). A link between ESM1 and inflammation has been shown in several studies. Clinical investigations have identified that ESM1 levels are elevated in the plasma or tissue of patients afflicted with various inflammatory diseases, including acute respiratory distress syndrome, sepsis and rheumatoid arthritis (8284). In addition, experimental research has substantiated the association between high ESM1 levels and inflammatory responses, since in vitro assays using HUVECs and in vivo experiments using mice revealed increased levels of ESM1 following treatment with LPS (58).

ESM1 levels have also been found to increase in HUVECs following stimulation with IL-8, monocyte chemotactic protein-1 (MCP-1) and TNF-α (58), as depicted in Fig. 4. As aforementioned and illustrated in Fig. 3, ESM1 activates HUVECs through the MAPK pathways, namely ERK, JNK and p38, and the NF-κB pathway (58). Furthermore, the treatment of HUVECs with IL-1β results in the induction of ESM1 expression (11), as shown in Fig. 4. It has also been shown that ESM1 can influence the expression of inflammatory factors, chemokines and adhesion molecules. Upon stimulation with LPS and IL-1β, HUVECs and trophoblast cells exhibited significantly increased expression of IL-6, IL-8 and MCP-1, but when ESM1 was knocked down using small interfering RNA, the stimulatory effect of IL-6, IL-8 and MCP-1 expression was diminished in both cell types (85), as depicted in Fig. 4.

In another study, the culture of HUVECs with ESM1 for 3 h led to the upregulation of IL-8, MCP-1 and TNF-α expression (58). Furthermore, ESM1 also upregulated vascular cell adhesion molecule-1 (VCAM-1) and intercellular cell adhesion molecule-1 (ICAM-1) (58), as shown in Fig. 4. Conversely, ESM1 knockdown reduced VCAM-1 and ICAM-1 protein expression in mouse aortic vascular smooth muscle cells (26). These findings indicate that a reciprocal regulatory relationship exists between specific inflammatory factors and ESM1; inflammatory factors upregulate ESM1 expression, while ESM1 induces the expression of factors associated with inflammation, thereby exacerbating inflammation within the TME and promoting angiogenesis.

Value of ESM1 as a future therapeutic target for cancer

Current research progress of ESM1

As a novel marker of cancer, ESM1 has been studied extensively. Research has shown that ESM1 is involved in the process of tumor development in two ways: Firstly, it is abnormally expressed in cancer cells and participates in tumor cell proliferation, migration and invasion (6,8,20,21); secondly, its expression is increased in vascular endothelial cells, which is associated with the promotion of cancer angiogenesis (22,23). Most of these findings have been obtained from experiments using cells or animals. However, only the abnormal expression of ESM1 in tumor tissues has been confirmed in clinical trials (18,19). Although ESM1 has been shown to be closely associated with tumorigenesis, no interventional agents targeting ESM1 are currently being used in the clinic.

Based on the association between ESM1 and tumors, and the lack of clarity regarding the mechanism of ESM1 in the development of cancer, it is necessary to further clarify its mechanism through various studies in the future, particularly in clinical research. Following this, the development of novel antitumor drugs that target ESM1 may be possible.

Value of ESM1 as a prognostic and predictive indicator of cancer

The relationship of ESM1 with tumor growth, metastasis and patient prognosis has garnered significant attention within the medical community. Clinical investigations have consistently confirmed the abnormal expression of ESM1 in the blood or tumor tissues of patients with diverse cancers. Notably, elevated ESM1 levels have been observed in the circulatory systems of patients diagnosed with endometrial, cervical, renal, breast and lung cancer, multiple myeloma and hepatocellular carcinoma (19,8690). Furthermore, upregulated ESM1 expression has been detected in tumor samples collected from patients with pancreatic neuroendocrine tumors and glioblastoma (91,92).

A meta-analysis exploring the association between ESM1 expression and the prognosis of patients with cancer has unveiled a significant negative association between ESM1 levels and patient survival rate (93). Consequently, ESM1 has emerged as a promising novel tumor marker for the evaluation of treatment efficacy, monitoring of disease progression and assessment of prognosis in patients with cancer. The practicality of using ESM1 in clinical settings is bolstered by its status as a secreted protein readily detectable in blood samples. In fact, commercial enzyme-linked immunosorbent assay kits designed for the detection of ESM1 have already entered the market, which further substantiates the feasibility of using ESM1 as a cancer marker in future clinical practice.

Therapeutic strategies and drug development for ESM1

Given the pivotal role of ESM1 in tumor development and metastasis, it has emerged as a promising therapeutic target for cancer. Future therapeutic strategies for ESM1 may include the following: i) Inhibiting the synthesis and secretion of ESM1; ii) neutralizing the already-produced ESM1, and iii) blocking the downstream signaling pathways activated by ESM1.

Research has clearly shown that factors such as inflammation and hypoxia can stimulate the expression and release of ESM1 (58,77). Consequently, the use of anti-inflammatory drugs and inhibitors targeting HIF-1α is a viable approach for impeding the synthesis and secretion of ESM1. Monoclonal antibodies designed for the neutralization of ESM1 have not yet been developed; however, the search for ESM1-neutralizing antibodies is a promising direction for future drug development efforts. Furthermore, ESM1 has been shown to activate downstream signaling pathways, including the PI3K/AKT, MAPK and NF-κB pathways (53,58,68). Inhibitors targeting these pathways may serve as an additional means for blocking the downstream effects mediated by ESM1.

Conclusions and prospects

The present review reveals the multifaceted roles of ESM1 and the underlying mechanisms by which ESM1 promotes tumor angiogenesis. It also comprehensively summarizes the regulatory mechanisms governing ESM1 within the TME. In summary, it describes how ESM1 expression is increased in response to hypoxic and inflammatory conditions within the TME. This heightened ESM1 expression, in turn, orchestrates the expression of pro-angiogenic factors through the PI3K/AKT, MAPK, NF-κB and Notch signaling pathways, thereby inducing angiogenesis.

Furthermore, the review introduces the concept that ESM1 actively engages with the TME. ESM1 expression is increased within the TME and, notably, ESM1 reciprocally promotes the progression and maintenance of the TME via the regulation of inflammatory factors, thereby establishing a positive feedback loop between ESM1 and the TME.

As an emerging tumor marker, ESM1 plays a pivotal role in tumor growth, metastasis, invasion and angiogenesis. Consequently, it is a promising biomarker for disease monitoring and a potential therapeutic target for clinical practice in the future. However, it is worth noting that the research on the role of ESM1 in angiogenesis remains limited, and the precise mechanisms by which ESM1 regulates angiogenesis require further exploration in future studies.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

JZ and PZ were responsible for drafting and revising the manuscript, and creating the figures. JW and JS revised the manuscript. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.

Ethical 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 

Folkman J: What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst. 82:4–6. 1990. View Article : Google Scholar : PubMed/NCBI

2 

Qi S, Deng S, Lian Z and Yu K: Novel drugs with high efficacy against tumor angiogenesis. Int J Mol Sci. 23:69342022. View Article : Google Scholar : PubMed/NCBI

3 

Pan KF, Yang YC, Lee WJ, Hua KT and Chien MH: Proteoglycan endocan: A multifaceted therapeutic target in Cancer. Biochim Biophys Acta Rev Cancer. 1877:1886722022. View Article : Google Scholar : PubMed/NCBI

4 

Chen J, Jiang L, Yu XH, Hu M, Zhang YK, Liu X, He P and Ouyang X: Endocan: A key player of cardiovascular disease. Front Cardiovasc Med. 8:7986992022. View Article : Google Scholar : PubMed/NCBI

5 

Zhang SM, Zuo L, Zhou Q, Gui SY, Shi R, Wu Q, Wei W and Wang Y: Expression and distribution of endocan in human tissues. Biotech Histochem. 87:172–178. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Lu J, Liu Q, Zhu L, Liu Y, Zhu X, Peng S, Chen M and Li P: Endothelial cell-specific molecule 1 drives cervical cancer progression. Cell Death Dis. 13:10432022. View Article : Google Scholar : PubMed/NCBI

7 

Liu W, Yang Y, He B, Ma F, Sun F, Guo M, Zhang M and Dong Z: ESM1 promotes triple-negative breast cancer cell proliferation through activating AKT/NF-κB/Cyclin D1 pathway. Ann Transl Med. 9:5332021. View Article : Google Scholar : PubMed/NCBI

8 

Li J, Yang D, Zhang C, Wei S, Zhao R, Dai S and Shan B: ESM1 is a promising therapeutic target and prognostic indicator for esophageal Carcinogenesis/Esophageal squamous cell carcinoma. Biomed Res Int. 2022:53281922022.PubMed/NCBI

9 

Abid MR, Yi X, Yano K, Shih SC and Aird WC: Vascular endocan is preferentially expressed in tumor endothelium. Microvasc Res. 72:136–145. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Chen LY, Liu X, Wang SL and Qin CY: Over-expression of the Endocan gene in endothelial cells from hepatocellular carcinoma is associated with angiogenesis and tumour invasion. J Int Med Res. 38:498–510. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Lassalle P, Molet S, Janin A, Heyden JV, Tavernier J, Fiers W, Devos R and Tonnel AB: ESM-1 is a novel human endothelial cell-specific molecule expressed in lung and regulated by cytokines. J Biol Chem. 271:20458–20464. 1996. View Article : Google Scholar : PubMed/NCBI

12 

Kali A and Shetty KS: Endocan: A novel circulating proteoglycan. Indian J Pharmacol. 46:579–583. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Sarrazin S, Adam E, Lyon M, Depontieu F, Motte V, Landolfi C, Lortat-Jacob H, Bechard D, Lassalle P and Delehedde M: Endocan or endothelial cell specific molecule-1 (ESM-1): A potential novel endothelial cell marker and a new target for cancer therapy. Biochim Biophys Acta. 1765:25–37. 2006.PubMed/NCBI

14 

Delehedde M, Devenyns L, Maurage CA and Vivès RR: Endocan in cancers: A lesson from a circulating dermatan sulfate proteoglycan. Int J Cell Biol. 2013:7050272013. View Article : Google Scholar : PubMed/NCBI

15 

Béchard D, Gentina T, Delehedde M, Scherpereel A, Lyon M, Aumercier M, Vazeux R, Richet C, Degand P, Jude B, et al: Endocan is a novel chondroitin sulfate/dermatan sulfate proteoglycan that promotes hepatocyte growth factor/scatter factor mitogenic activity. J Biol Chem. 276:48341–48349. 2001. View Article : Google Scholar : PubMed/NCBI

16 

De Freitas Caires N, Gaudet A, Portier L, Tsicopoulos A, Mathieu D and Lassalle P: Endocan, sepsis, pneumonia, and acute respiratory distress syndrome. Crit Care. 22:2802018. View Article : Google Scholar : PubMed/NCBI

17 

Leite AR, Borges-Canha M, Cardoso R, Neves JS, Castro-Ferreira R and Leite-Moreira A: Novel biomarkers for evaluation of endothelial dysfunction. Angiology. 71:397–410. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Yu PH, Chou SF, Chen CL, Hung H, Lai CY, Yang PM, Jeng YM, Liaw SF, Kuo HH, Hsu HC, et al: Upregulation of endocan by Epstein-Barr virus latent membrane protein 1 and its clinical significance in nasopharyngeal carcinoma. PLoS One. 8:e822542013. View Article : Google Scholar : PubMed/NCBI

19 

Laloglu E, Kumtepe Y, Aksoy H and Topdagi Yilmaz EP: Serum endocan levels in endometrial and ovarian cancers. J Clin Lab Anal. 31:e220792017. View Article : Google Scholar : PubMed/NCBI

20 

Xu H, Chen X and Huang Z: Identification of ESM1 overexpressed in head and neck squamous cell carcinoma. Cancer Cell Int. 19:1182019. View Article : Google Scholar : PubMed/NCBI

21 

Baghy K, Tátrai P, Regős E and Kovalszky I: Proteoglycans in liver cancer. World J Gastroenterol. 22:379–393. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Cai L, Leng ZG, Guo YH, Lin SJ, Wu ZR, Su ZP, Lu JL, Wei LF, Zhuge QC, Jin K and Wu ZB: Dopamine agonist resistance-related endocan promotes angiogenesis and cells viability of prolactinomas. Endocrine. 52:641–651. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Chang Y, Niu W, Lian PL, Wang XQ, Meng ZX, Liu Y and Zhao R: Endocan-expressing microvessel density as a prognostic factor for survival in human gastric cancer. World J Gastroenterol. 22:5422–5429. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Zhang H, Shen YW, Zhang LJ, Chen JJ, Bian HT, Gu WJ, Zhang H, Chen HZ, Zhang WD and Luan X: Targeting endothelial cell-specific molecule 1 protein in cancer: A promising therapeutic approach. Front Oncol. 11:6871202021. View Article : Google Scholar : PubMed/NCBI

25 

Zolali E, Rezabakhsh A, Nabat E, Jaberi H, Rahbarghazi R and Garjani A: Metformin effect on Endocan biogenesis in human endothelial cells under diabetic condition. Arch Med Res. 50:304–314. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Zeng M, Xie Z, Zhang J, Li S, Wu Y and Yan X: Arctigenin attenuates vascular inflammation induced by high salt through TMEM16A/ESM1/VCAM-1 pathway. Biomedicines. 10:27602022. View Article : Google Scholar : PubMed/NCBI

27 

Hortal J, Giannella M, Pérez MJ, Barrio JM, Desco M, Bouza E and Muñoz P: Incidence and risk factors for ventilator-associated pneumonia after major heart surgery. Intensive Care Med. 35:1518–1525. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Daly C, Pasnikowski E, Burova E, Wong V, Aldrich TH, Griffiths J, Ioffe E, Daly TJ, Fandl JP, Papadopoulos N, et al: Angiopoietin-2 functions as an autocrine protective factor in stressed endothelial cells. Proc Natl Acad Sci USA. 103:15491–15496. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Baeriswyl V and Christofori G: The angiogenic switch in carcinogenesis. Semin Cancer Biol. 19:329–337. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Apte RS, Chen DS and Ferrara N: VEGF in signaling and disease: Beyond discovery and development. Cell. 176:1248–1264. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Ellis LM and Hicklin DJ: VEGF-targeted therapy: Mechanisms of anti-tumour activity. Nat Rev Cancer. 8:579–591. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Ferrara N and Henzel WJ: Pituitary follicular cells secrete a novel heparin-binding growth factor specific for vascular endothelial cells. Biochem Biophys Res Commun. 161:851–858. 1989. View Article : Google Scholar : PubMed/NCBI

33 

Duran CL, Borriello L, Karagiannis GS, Entenberg D, Oktay MH and Condeelis JS: Targeting Tie2 in the tumor microenvironment: From Angiogenesis to Dissemination. Cancers (Basel). 13:57302021. View Article : Google Scholar : PubMed/NCBI

34 

Bremnes RM, Camps C and Sirera R: Angiogenesis in non-small cell lung cancer: The prognostic impact of neoangiogenesis and the cytokines VEGF and bFGF in tumours and blood. Lung Cancer. 51:143–158. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Huang H: Matrix metalloproteinase-9 (MMP-9) as a cancer biomarker and MMP-9 biosensors: Recent advances. Sensors (Basel). 18:32492018. View Article : Google Scholar : PubMed/NCBI

36 

Kessenbrock K, Plaks V and Werb Z: Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell. 141:52–67. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Rust R, Gantner C and Schwab ME: Pro- and antiangiogenic therapies: Current status and clinical implications. FASEB J. 33:34–48. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Viallard C and Larrivée B: Tumor angiogenesis and vascular normalization: Alternative therapeutic targets. Angiogenesis. 20:409–426. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Phng LK, Stanchi F and Gerhardt H: Filopodia are dispensable for endothelial tip cell guidance. Development. 140:4031–4040. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Barău A, Ruiz-Sauri A, Valencia G, Gómez-Mateo Mdel C, Sabater L, Ferrandez A and Llombart-Bosch A: High microvessel density in pancreatic ductal adenocarcinoma is associated with high grade. Virchows Arch. 462:541–546. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Di Maggio F, Arumugam P, Delvecchio FR, Batista S, Lechertier T, Hodivala-Dilke K and Kocher HM: Pancreatic stellate cells regulate blood vessel density in the stroma of pancreatic ductal adenocarcinoma. Pancreatology. 16:995–1004. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Ramjiawan RR, Griffioen AW and Duda DG: Anti-angiogenesis for cancer revisited: Is there a role for combinations with immunotherapy? Angiogenesis. 20:185–204. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Morikawa S, Baluk P, Kaidoh T, Haskell A, Jain RK and McDonald DM: Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors. Am J Pathol. 160:985–1000. 2002. View Article : Google Scholar : PubMed/NCBI

44 

Dejana E, Tournier-Lasserve E and Weinstein BM: The control of vascular integrity by endothelial cell junctions: Molecular basis and pathological implications. Dev Cell. 16:209–221. 2009. View Article : Google Scholar : PubMed/NCBI

45 

Baluk P, Morikawa S, Haskell A, Mancuso M and McDonald DM: Abnormalities of basement membrane on blood vessels and endothelial sprouts in tumors. Am J Pathol. 163:1801–1815. 2003. View Article : Google Scholar : PubMed/NCBI

46 

Rocha SF, Schiller M, Jing D, Li H, Butz S, Vestweber D, Biljes D, Drexler HC, Nieminen-Kelhä M, Vajkoczy P, et al: Esm1 modulates endothelial tip cell behavior and vascular permeability by enhancing VEGF bioavailability. Circ Res. 115:581–590. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Su T, Zhong Y, Demetriades AM, Shen J, Sui A, Yao Y, Gao Y, Zhu Y, Shen X and Xie B: Endocan blockade suppresses experimental ocular neovascularization in mice. Invest Ophthalmol Vis Sci. 59:930–939. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Li YK, Zeng T, Guan Y, Liu J, Liao NC, Wang MJ, Chen KX, Luo XY, Chen CY, Quan FF, et al: Validation of ESM1 related to ovarian cancer and the biological function and prognostic significance. Int J Biol Sci. 19:258–280. 2023. View Article : Google Scholar : PubMed/NCBI

49 

He Y, Lin L, Ou Y, Hu X, Xu C and Wang C: Endothelial cell-specific molecule 1 (ESM1) promoted by transcription factor SPI1 acts as an oncogene to modulate the malignant phenotype of endometrial cancer. Open Med (Wars). 17:1376–1389. 2022. View Article : Google Scholar : PubMed/NCBI

50 

Li D, Su X, Xue S, Yao L, Yu D, Tang X and Huang Y: Targeting ESM1/VEGFα signaling axis: A promising therapeutic avenue for angiogenesis in cervical squamous cell carcinoma. J Cancer. 14:1725–1735. 2023. View Article : Google Scholar : PubMed/NCBI

51 

Roudnicky F, Poyet C, Wild P, Krampitz S, Negrini F, Huggenberger R, Rogler A, Stöhr R, Hartmann A, Provenzano M, et al: Endocan is upregulated on tumor vessels in invasive bladder cancer where it mediates VEGF-A-induced angiogenesis. Cancer Res. 73:1097–1106. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Shahcheraghi SH, Tchokonte-Nana V, Lotfi M, Lotfi M, Ghorbani A and Sadeghnia HR: Wnt/beta-catenin and PI3K/Akt/mTOR signaling pathways in Glioblastoma: Two main targets for drug design: A Review. Curr Pharm Des. 26:1729–1741. 2020. View Article : Google Scholar : PubMed/NCBI

53 

Yang L, Dong Z, Li S and Chen T: ESM1 promotes angiogenesis in colorectal cancer by activating PI3K/Akt/mTOR pathway, thus accelerating tumor progression. Aging (Albany NY). 15:2920–2936. 2023.PubMed/NCBI

54 

Namjoo M, Ghafouri H, Assareh E, Aref AR, Mostafavi E, Hamrahi Mohsen A, Balalaie S, Broussy S and Asghari SM: A VEGFB-based peptidomimetic inhibits VEGFR2-Mediated PI3K/Akt/mTOR and PLCγ/ERK signaling and elicits apoptotic, antiangiogenic, and antitumor activities. Pharmaceuticals (Basel). 16:9062023. View Article : Google Scholar : PubMed/NCBI

55 

Yang YC, Pan KF, Lee WJ, Chang JH, Tan P, Gu CC, Chang WM, Yang SF, Hsiao M, Hua KT and Chien MH: Circulating proteoglycan endocan mediates EGFR-Driven progression of non-small cell lung cancer. Cancer Res. 80:3292–3304. 2020. View Article : Google Scholar : PubMed/NCBI

56 

Rao L, Giannico D, Leone P, Solimando AG, Maiorano E, Caporusso C, Duda L, Tamma R, Mallamaci R, Susca N, et al: HB-EGF-EGFR signaling in bone marrow endothelial cells mediates angiogenesis associated with multiple myeloma. Cancers (Basel). 12:1732020. View Article : Google Scholar : PubMed/NCBI

57 

Guo YJ, Pan WW, Liu SB, Shen ZF, Xu Y and Hu LL: ERK/MAPK signalling pathway and tumorigenesis. Exp Ther Med. 19:1997–2007. 2020.PubMed/NCBI

58 

Lee W, Ku SK, Kim SW and Bae JS: Endocan elicits severe vascular inflammatory responses in vitro and in vivo. J Cell Physiol. 229:620–630. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Guma M, Rius J, Duong-Polk KX, Haddad GG, Lindsey JD and Karin M: Genetic and pharmacological inhibition of JNK ameliorates hypoxia-induced retinopathy through interference with VEGF expression. Proc Natl Acad Sci USA. 106:8760–8765. 2009. View Article : Google Scholar : PubMed/NCBI

60 

Volpi G, Facchinetti F, Moretto N, Civelli M and Patacchini R: Cigarette smoke and α,β-unsaturated aldehydes elicit VEGF release through the p38 MAPK pathway in human airway smooth muscle cells and lung fibroblasts. Br J Pharmacol. 163:649–661. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Zhou J, Ji Q and Li Q: Resistance to anti-EGFR therapies in metastatic colorectal cancer: Underlying mechanisms and reversal strategies. J Exp Clin Cancer Res. 40:3282021. View Article : Google Scholar : PubMed/NCBI

62 

Jiang N, Hu Y, Wang M, Zhao Z and Li M: The notch signaling pathway contributes to angiogenesis and tumor immunity in breast cancer. Breast Cancer (Dove Med Press). 14:291–309. 2022.PubMed/NCBI

63 

Kang N, Liang X, Fan B, Zhao C, Shen B, Ji X and Liu Y: Endothelial-Specific Molecule 1 inhibition lessens productive angiogenesis and tumor metastasis to overcome bevacizumab resistance. Cancers (Basel). 14:56812022. View Article : Google Scholar : PubMed/NCBI

64 

Huang YG, Wang Y, Zhu RJ, Tang K, Tang XB and Su XM: EMS1/DLL4-Notch signaling axis augments cell cycle-mediated tumorigenesis and progress in human adrenocortical carcinoma. Front Oncol. 11:7715792021. View Article : Google Scholar : PubMed/NCBI

65 

Yan M and Plowman GD: Delta-like 4/Notch signaling and its therapeutic implications. Clin Cancer Res. 13:7243–7246. 2007. View Article : Google Scholar : PubMed/NCBI

66 

Pitulescu ME, Schmidt I, Giaimo BD, Antoine T, Berkenfeld F, Ferrante F, Park H, Ehling M, Biljes D, Rocha SF, et al: Dll4 and Notch signalling couples sprouting angiogenesis and artery formation. Nat Cell Biol. 19:915–927. 2017. View Article : Google Scholar : PubMed/NCBI

67 

Gilmore TD: NF-κB and human cancer: What have we learned over the Past 35 Years? Biomedicines. 9:8892021. View Article : Google Scholar : PubMed/NCBI

68 

Kumar SK and Mani KP: Endocan alters nitric oxide production in endothelial cells by targeting AKT/eNOS and NFkB/iNOS signaling. Nitric Oxide. 117:26–33. 2021. View Article : Google Scholar : PubMed/NCBI

69 

Yu M, Qi B, Xiaoxiang W, Xu J and Liu X: Baicalein increases cisplatin sensitivity of A549 lung adenocarcinoma cells via PI3K/Akt/NF-κB pathway. Biomed Pharmacother. 90:677–685. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Zhang D, Li H, Luo X, Liu D, Wei Q and Ye X: Integrated 16S rDNA, metabolomics, and TNF-α/NF-κB signaling pathway analyses to explain the modulatory effect of Poria cocos aqueous extract on anxiety-like behavior. Phytomedicine. 104:1543002022. View Article : Google Scholar : PubMed/NCBI

71 

Xiong H, Ye J, Xie K, Hu W, Xu N and Yang H: Exosomal IL-8 derived from Lung Cancer and Colon Cancer cells induced adipocyte atrophy via NF-κB signaling pathway. Lipids Health Dis. 21:1472022. View Article : Google Scholar : PubMed/NCBI

72 

Szade A, Grochot-Przeczek A, Florczyk U, Jozkowicz A and Dulak J: Cellular and molecular mechanisms of inflammation-induced angiogenesis. IUBMB Life. 67:145–159. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Xiao Y and Yu D: Tumor microenvironment as a therapeutic target in cancer. Pharmacol Ther. 221:1077532021. View Article : Google Scholar : PubMed/NCBI

74 

Shao C, Yang F, Miao S, Liu W, Wang C, Shu Y and Shen H: Role of hypoxia-induced exosomes in tumor biology. Mol Cancer. 17:1202018. View Article : Google Scholar : PubMed/NCBI

75 

Forsythe JA, Jiang BH, Iyer NV, Agani F, Leung SW, Koos RD and Semenza GL: Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol. 16:4604–4613. 1996. View Article : Google Scholar : PubMed/NCBI

76 

Lv X, Li J, Zhang C, Hu T, Li S, He S, Yan H, Tan Y, Lei M, Wen M and Zuo J: The role of hypoxia-inducible factors in tumor angiogenesis and cell metabolism. Genes Dis. 4:19–24. 2017. View Article : Google Scholar : PubMed/NCBI

77 

Sun H, Zhang H, Li K, Wu H, Zhan X, Fang F, Qin Y and Wei Y: ESM-1 promotes adhesion between monocytes and endothelial cells under intermittent hypoxia. J Cell Physiol. 234:1512–1521. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Fukumoto M, Kondo K, Uni K, Ishiguro T, Hayashi M, Ueda S, Mori I, Niimi K, Tashiro F, Miyazaki S, et al: Tip-cell behavior is regulated by transcription factor FoxO1 under hypoxic conditions in developing mouse retinas. Angiogenesis. 21:203–214. 2018. View Article : Google Scholar : PubMed/NCBI

79 

Gu X, Zhang J, Shi Y, Shen H, Li Y, Chen Y and Liang L: ESM1/HIF-1α pathway modulates chronic intermittent hypoxia-induced non-small-cell lung cancer proliferation, stemness and epithelial-mesenchymal transition. Oncol Rep. 45:1226–1234. 2021. View Article : Google Scholar : PubMed/NCBI

80 

Wigner P, Grębowski R, Bijak M, Saluk-Bijak J and Szemraj J: The interplay between oxidative stress, inflammation and angiogenesis in bladder cancer development. Int J Mol Sci. 22:44832021. View Article : Google Scholar : PubMed/NCBI

81 

Wu L, Saxena S and Singh RK: Neutrophils in the tumor microenvironment. Adv Exp Med Biol. 1224:1–20. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Tang L, Zhao Y, Wang D, Deng W, Li C, Li Q, Huang S and Shu C: Endocan levels in peripheral blood predict outcomes of acute respiratory distress syndrome. Mediators Inflamm. 2014:6251802014. View Article : Google Scholar : PubMed/NCBI

83 

Reinhart K, Meisner M and Brunkhorst FM: Markers for sepsis diagnosis: What is useful? Crit Care Clin. 22:503–519. ix–x. 2006. View Article : Google Scholar : PubMed/NCBI

84 

Kim KS, Lee YA, Ji HI, Song R, Kim JY, Lee SH, Hong SJ, Yoo MC and Yang HI: Increased expression of endocan in arthritic synovial tissues: Effects of adiponectin on the expression of endocan in fibroblast-like synoviocytes. Mol Med Rep. 11:2695–2702. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Murthi P, Sarkis R, Lim R, Nguyen-Ngo C, Pratt A, Liong S and Lappas M: Endocan expression is increased in the placenta from obese women with gestational diabetes mellitus. Placenta. 48:38–48. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Leroy X, Aubert S, Zini L, Franquet H, Kervoaze G, Villers A, Delehedde M, Copin MC and Lassalle P: Vascular endocan (ESM-1) is markedly overexpressed in clear cell renal cell carcinoma. Histopathology. 56:180–187. 2010. View Article : Google Scholar : PubMed/NCBI

87 

Basim P and Argun D: A comparison of the circulating endocan levels between the inflammatory and malignant diseases of the same organ: The breast. J Invest Surg. 34:1207–1213. 2021. View Article : Google Scholar : PubMed/NCBI

88 

Scherpereel A, Gentina T, Grigoriu B, Sénéchal S, Janin A, Tsicopoulos A, Plénat F, Béchard D, Tonnel AB and Lassalle P: Overexpression of endocan induces tumor formation. Cancer Res. 63:6084–6089. 2003.PubMed/NCBI

89 

Steiner N, Hajek R, Sevcikova S, Borjan B, Untergasser G, Göbel G and Gunsilius E: The plasma levels of the angiogenic cytokine endocan are elevated in patients with multiple myeloma. Anticancer Res. 38:5087–5092. 2018. View Article : Google Scholar : PubMed/NCBI

90 

Ozaki K, Toshikuni N, George J, Minato T, Matsue Y, Arisawa T and Tsutsumi M: Serum endocan as a novel prognostic biomarker in patients with hepatocellular carcinoma. J Cancer. 5:221–230. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Lin LY, Yeh YC, Chu CH, Won JGS, Shyr YM, Chao Y, Li CP, Wang SE and Chen MH: Endocan expression is correlated with poor progression-free survival in patients with pancreatic neuroendocrine tumors. Medicine (Baltimore). 96:e82622017. View Article : Google Scholar : PubMed/NCBI

92 

Maurage CA, Adam E, Minéo JF, Sarrazin S, Debunne M, Siminski RM, Baroncini M, Lassalle P, Blond S and Delehedde M: Endocan expression and localization in human glioblastomas. J Neuropathol Exp Neurol. 68:633–641. 2009. View Article : Google Scholar : PubMed/NCBI

93 

Huang X, Chen C, Wang X, Zhang JY, Ren BH, Ma DW, Xia L, Xu XY and Xu L: Prognostic value of endocan expression in cancers: Evidence from meta-analysis. Onco Targets Ther. 9:6297–6304. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2024
Volume 27 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou J, Zhou P, Wang J and Song J: Roles of endothelial cell specific molecule‑1 in tumor angiogenesis (Review). Oncol Lett 27: 137, 2024
APA
Zhou, J., Zhou, P., Wang, J., & Song, J. (2024). Roles of endothelial cell specific molecule‑1 in tumor angiogenesis (Review). Oncology Letters, 27, 137. https://doi.org/10.3892/ol.2024.14270
MLA
Zhou, J., Zhou, P., Wang, J., Song, J."Roles of endothelial cell specific molecule‑1 in tumor angiogenesis (Review)". Oncology Letters 27.3 (2024): 137.
Chicago
Zhou, J., Zhou, P., Wang, J., Song, J."Roles of endothelial cell specific molecule‑1 in tumor angiogenesis (Review)". Oncology Letters 27, no. 3 (2024): 137. https://doi.org/10.3892/ol.2024.14270