Emerging roles of angiopoietin‑like 4 in human tumors (Review)
- Authors:
- Published online on: December 19, 2024 https://doi.org/10.3892/ijo.2024.5715
- Article Number: 9
-
Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Cancer remains a major global public health burden, with incidence and mortality rates rising rapidly. According to Global Cancer Statistics, nearly 20 million new cases of cancer and 9.7 million cancer-related deaths worldwide were reported in 2022 (1). It is estimated that ~20% of individuals will develop cancer during their lifetime. Lung cancer has the highest incidence rate (12.4%), followed by female breast cancer (BRC) (11.6%), colorectal cancer (CRC) (9.6%), prostate cancer (7.3%) and gastric cancer (GC) (4.9%) (1). Generally, the initiation and progression of tumors are caused by genetic or epigenetic alterations caused by both internal and external factors, which activate or inhibit specific signaling pathways (2,3). Therefore, investigating the mechanisms underlying tumorigenesis is essential for improving the diagnosis, prognosis and the development of targeted therapies.
The angiopoietin family consists of growth factors that regulate vascular development, maintenance and remodeling, playing a pivotal role in angiogenesis. Currently, the angiopoietin family includes angiopoietin (ANGPT) 1-4 and angiopoietin-like (ANGPTL) 1-8. ANGPTs act as ligands for the endothelial cell receptors TIE1 and TIE2 and are crucial in regulating tumor angiogenesis, inflammation, lymphatic angiogenesis and the cardiovascular system (4,5). Although ANGPTLs share structural similarities with ANGPTs, they do not bind to TIE receptors to mediate their biological functions. The homologous receptors of ANGPTLs remain unidentified, and they are classified as orphan ligands (6-9). ANGPTLs, particularly ANGPTL3, ANGPTL4 and ANGPTL8, have been extensively studied for their roles in lipid metabolism (10,11). Additionally, ANGPTLs regulate both acute and chronic inflammation (12) and atherosclerosis (13) through various mechanisms. ANGPTL4, a member of the angiopoietin family, was initially identified for its key roles in lipid metabolism (14), inflammatory response (15) and angiogenesis (6,7). As research has progressed, increasing evidence has demonstrated that ANGPTL4 is involved in various stages of tumor progression (8,9).
In the present review, the multifaceted roles of ANGPTL4 in tumor development and its underlying mechanisms of action are discussed. While earlier reviews (8,9) provided a foundational understanding of ANGPTL4, the current article integrates the latest research and findings, offering a comprehensive and updated perspective on its functions in cancer biology.
Biological characteristics of ANGPTL4
In 2000, three research teams successively identified a new member of the ANGPT family. Kim et al (16) isolated a new sequence from human and mouse embryonic cDNA using degeneracy PCR, which was named hepatic fibrinogen/angiopoietin-related protein (HFARP). Yoon et al (17) described the isolation and characterization of a novel target gene induced by peroxisome proliferator-activated receptor (PPAR) γ ligands, termed PGAR (for PPAR γ angiopoietin-related); Kersten et al (18) identified a novel PPARα target gene called FIAF (fasting-induced adipose factor) through subtractive hybridization. Subsequently, the gene encoding this protein was collectively referred to as ANGPTL4 by the HUGO Gene Nomenclature Committee.
ANGPTL4 is located at 19p13.2, with an mRNA length of ~2,000 bp and an open reading frame of 1,218 bp. It encodes a secreted glycoprotein composed of 406 amino acids with a relative molecular weight of ~45-65 kDa. Structurally similar to other ANGPT family proteins, ANGPTL4 contains a highly hydrophobic signal peptide, an N-terminal helical domain with three glycosylation sites, and a larger C-terminal fibrinogen-like domain, with a small connecting peptide between these domains. The natural full-length ANGPTL4 (fANGPTL4) exists as either a dimer or tetramer. It can be cleaved by furin-like proprotein convertase to yield an N-terminal coiled-coil fragment (nANGPTL4), containing amino acids 1 to 170, and a C-terminal fibrinogen-like domain monomer fragment (cANGPTL4), consisting of amino acids 171 to 406. The cleavage of fANGPTL4 at the -RXR-site by proprotein convertase depends on the tissue in which ANGPTL4 is synthesized and the physiological or pathological conditions (19-21).
In humans, ANGPTL4 exhibits widespread expression, with notable prominence in the heart, liver, small intestine, adipose tissue, plasma and placenta, as reported in multiple studies (22-26). Its expression is regulated by PPARs (9), glucocorticoid receptors (27,28), hypoxia-inducible factor-1α (HIF-1α) (28), transforming growth factor-β (TGF-β) (29-31) and other regulatory factors.
Roles of ANGPTL4 in cancer
Previous findings suggest that ANGPTL4 is commonly dysregulated in various malignancies, and plays important dual roles, functioning as either an oncogene or a tumor suppressor (Fig. 1; Tables I and II). Altered ANGPTL4 expression impacts diverse cellular phenotypes via multiple signaling pathways, affecting tumor growth, invasion, metastasis, angiogenesis, programmed cell death, cell metabolism and treatment resistance (Fig. 2).
ANGPTL4 and tumor growth
ANGPTL4 exhibits varied regulatory effects across different types of tumors. Ding et al (32) demonstrated that the recombinant ANGPTL4 protein reduces CD8+ T cell infiltration and activation through metabolic reprogramming, thereby diminishing immune surveillance during tumor progression and facilitating tumor growth in vivo. In breast tumors of obese mice, ANGPTL4 expression has been shown upregulated, and the suppression of ANGPTL4 leads to a significant reduction in obesity-induced tumor growth (33). Interleukin-1β (IL-1β) in primary adipocytes stimulates ANGPTL4 expression through the activation of nuclear factor-κ B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways, with hypoxia further enhancing IL-1β expression (33). This indicates that ANGPTL4 mediates the crosstalk between obesity-associated inflammation and BRC progression. Avalle et al (34) found that cancer-associated fibroblasts (CAFs) promote tumor growth in BRC, with signal transducer and activator of transcription 3 (STAT3) amplifying the effects of CAFs via ANGPTL4. In ovarian cancer (OVC), ANGPTL4 was significantly upregulated in clinical samples and correlated with poor prognosis (35). ANGPTL4 promotes OVC progression by activating the Janus kinase 2 (JAK2)/STAT3 pathway (35). Xu et al (36) further demonstrated that ANGPTL4 enhances ovarian tumor cell proliferation through the extracellular signal-related kinase (ERK) pathway, and its inhibition can suppress OVC progression via phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR2) (37). Additionally, several studies have shown that ANGPTL4 accelerates the proliferation and progression of numerous types of malignancies, including CRC (38), GC (39), hepatocellular carcinoma (HCC) (40,41), lung adenocarcinoma (42,43), osteosarcoma (OS) (44), thyroid cancer (45) and melanoma (46).
However, some studies suggest that ANGPTL4 may function as a tumor suppressor. Hsieh et al (47) reported the dual roles of ANGPTL4 in urothelial carcinoma (UC), showing that ANGPTL4 mRNA expression is decreased in UC cells and tumor tissues compared with adjacent normal bladder epithelial cells. Overexpression of ANGPTL4 inhibits UC cell proliferation both in vivo and in vitro (47). Hui et al (48) found that long non-coding RNA (lncRNA) AGAP2-AS1 downregulates ANGPTL4 expression through its interaction with the enhancer of zeste homolog 2 (EZH2), thereby promoting the proliferation and metastasis of pancreatic cancer. A negative correlation between the expression of ANGPTL4 and OS progression has also been observed (49). Knocking out ANGPTL4 in OS cells leads to the accumulation of branched-chain amino acids (BCAAs), which activates the mechanistic target of rapamycin (mTOR) signaling pathway and enhances OS cell proliferation (49). In addition, homeobox transcription factor 1α (LMX1A) has been shown to suppress tumor growth by activating ANGPTL4 to hinder c-Myc in GC (50). The negative regulatory effects of ANGPTL4 on tumor growth have also been reported in renal clear cell carcinoma (51) and CRC (52).
ANGPTL4 and tumor invasion and metastasis
Metastasis refers to the spread of malignant cells to distant organs, and it is often the primary cause of mortality in most cancers, as metastatic cancer cells typically exhibit high invasiveness and resistance to anticancer therapies. It has been indicated that ANGPTL4 plays a crucial role in cancer invasion and metastasis processes.
Hübers et al (53) identified cANGPTL4 and nANGPTL4 as pro- and antitumor contributors, respectively, in the bidirectional communication between primary tumors and distant metastases. It was observed that cANGPTL4 promotes tumor growth and metastasis, while nANGPTL4 inhibits metastasis and improves overall survival by suppressing Wnt signaling and reducing vascular distribution at metastasis sites. These findings suggested that cANGPTL4 could serve as a potential biomarker for tumor progression and a target for anti-meta-static therapy.
In BRC, HIF-2 induces the expression of the lncRNA RAB11B-AS1, which facilitates brain metastasis through the promotion of ANGPTL4 (54). Silencing ANGPTL4 in triple-negative BRC (TNBC) cells has been shown to reduce the metastatic growth of brain tumors in vivo (31). Tumor cells secrete IL-1β and tumor necrosis factor-α (TNF-α), which communicate with astrocytes, leading to increased expression of transforming growth factor-β2 (TGF-β2) and promoting the brain metastasis via the TGF-β2/ANGPTL4 axis (31). These findings provide a theoretical basis for targeting ANGPTL4 in the treatment of BRC metastasis.
In CRC, ANGPTL4 plays a role in metastasis through various mechanisms. Shen et al (55) found that NADPH oxidase 4 (NOX4)/reactive oxygen species (ROS) axis is crucial for CRC metastasis induced by oleic acid (OA). Downregulation of ANGPTL4 leads to the suppression of NOX4, ROS, matrix metalloproteinase-1 (MMP-1) and MMP-9, thus inhibiting OA-induced CRC metastasis (55). Furthermore, the metastasis in KRAS/p53 mutant CRC has been shown to depend on the activation of the ANGPTL4/IL-8/NOX4 axis (56), underscoring the importance of the ANGPTL4/NOX4 signaling axis in dyslipidemia-related and KRAS/p53 mutant CRC metastasis. Zhu et al (30) focused on the peritoneal metastasis of CRC. They reported that adipose-derived stem cells (ADSCs) secrete TGF-β1 to activate Smad3 in CRC cells, which enhances ANGPTL4 transcription. The upregulation of ANGPTL4 increases the anoikis resistance, facilitating CRC cell survival in the peritoneum and promoting metastatic foci formation (30).
Bajwa et al (57) reported that ANGPTL4, derived from cancer-associated mesothelial cells, promotes the early-stage OVC metastasis through the interactions between mesothelial cells and the tumor microenvironment (TME). Additionally, Hefni et al (58) found that ANGPTL4 induces the migration of head and neck squamous cell carcinoma (HNSCC) cells via the neuropilin1/ABL1/paxillin pathway. Similarly, ANGPTL4 has been implicated in the invasion and metastasis of lung cancer (42,59), with studies showing that modified Bu Fei decoction inhibits the metastasis of non-small cell lung cancer (NSCLC) by suppressing ANGPTL4 expression in endothelial cells (59).
Previous studies have highlighted the pivotal role of the cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) pathway in promoting metastasis through ANGPTL4. In GC, leptin induces the phosphorylation of ANGPTL4 at the serine 30 residue, reducing its binding affinity to lipoprotein lipase (LPL), thus enhancing lipid uptake and intracellular arachidonic acid levels. This accumulation subsequently activates the COX-2/PGE2 pathway, promoting lymph node metastasis (60). Moreover, Chiang et al (61) demonstrated that the COX-2/PGE2/ANGPTL4 axis, activated by epidermal growth factor (EGF), enhances metastasis in HNSCC, with PGE2 promoting ANGPTL4 expression through the ERK pathway.
Despite the general role of ANGPTL4 in promoting metastasis, some studies have identified its inhibitory effects. Overexpression of ANGPTL4 has been found to inhibit the adhesion, migration and invasion of TNBC cells in vitro, with positive correlations to favorable outcomes in patients with TNBC (62). Moreover, lncRNA AGAP2-AS1 epigenetically inhibits ANKRD1 and ANGPTL4 expression by recruiting EZH2, promoting pancreatic cancer metastasis (48). In CRC, DNA methylation-mediated downregulation of ANGPTL4 activates CAFs in the TME, promoting epithelial-mesenchymal transition (EMT) through the ERK signaling pathway, which leads to metastasis (63). In vivo experiments also revealed that overexpression of ANGPTL4 inhibits lung metastasis in CRC models (63). Additionally, inhibitory effects of ANGPTL4 on tumor metastasis have been also observed in OS (64).
ANGPTL4 and tumor angiogenesis
Angiogenesis and increased vascular permeability are ubiquitous characteristics of all solid tumors. Therefore, research on the role of ANGPTL4 in cancer angiogenesis holds significant potential to profoundly enhance our comprehension of cancer pathogenesis and inform the development of therapeutic strategies.
Wu et al (37) found that the pro-angiogenic effects of ANGPTL4 in OVC are mediated via its association with VEGF2. Similarly, Li et al (35) discovered that ANGPTL4 enhances angiogenesis in ovarian serous cystadenocarcinoma by activating the JAK2/STAT3 signaling pathway. The mechanism is potentially due to the interaction among ANGPTL4, endothelial cell specific molecule 1 and the TME (35). ANGPTL4 has also been reported to exert pro-angiogenic effects in malignant glioma cells (65). Knockdown of ANGPTL4 significantly reduces microvascular density in xenograft tumors and inhibits tumor growth (65). Additionally, EGF receptor (EGFR) variant III induces ANGPTL4 expression through the ERK/c-Myc pathway to regulate angiogenesis (65). In Kaposi's sarcoma, the upregulation of ANGPTL4 by viral G protein-coupled receptors has been shown to enhance tumor angiogenesis and vascular permeability through the Rho/Rho-associated kinase pathway (66). The complementary effect on VEGF, a potent angiogenic factor, has also been confirmed (66). Furthermore, ANGPTL4 has exhibited a stimulatory effect on tumor angiogenesis in various cancer types, including BRC (33), uveal melanoma (67), NSCLC (68) and OS (69).
The inhibitory effects of ANGPTL4 on angiogenesis are partially attributed to its suppression of the ERK signaling pathway and post-translational modifications. Tumor-derived ANGPTL4 has been found to inhibit the angiogenesis and proliferation of umbilical endothelial cells by suppressing ERK signaling (70). The inactivation of ANGPTL4 through genetic and epigenetic mechanisms, such as hypermethylation of CpG islands in the promoter region, causes an increase in tumor growth and angiogenesis in GC (70). Yang et al (71) reported that N-glycosylated cANGPTL4 exerts an inhibitory effect on the Raf/MEK/ERK signaling cascade in endothelial cells, which suppresses the angiogenesis induced by alkaline fibroblast growth factor and VEGF. Furthermore, The inhibitory effects of ANGPTL4 on angiogenesis have also been observed in CRC (52) and HCC (72), reinforcing its potential as a modulator of angiogenesis across various cancer types.
ANGPTL4 and programmed cell death (PCD)
PCD is a widespread mechanism in living organisms, essential for maintaining cellular homeostasis, development, immunity and stress response (73). PCD is regulated by numerous evolutionarily conserved pathways and well-characterized mechanisms of action (74). Based on specific morphological, immunological and genetic characteristics, PCD can be classified into several forms, including apoptosis, ferroptosis, necroptosis, pyroptosis and autophagy-dependent cell death (75,76). PCD is crucial in tumor suppression through its involvement in anticancer therapies. In the present review, the interactions between various forms of PCD and ANGPTL4 in cancer are discussed.
ANGPTL4 and apoptosis
Apoptosis, the most extensively studied form of PCD, was first described by Kerr et al in 1972 (77). Apoptosis relies on a cascade of caspase proteases and can be initiated via extrinsic or intrinsic pathways (78). As early as the 1970s, studies demonstrated that apoptosis is crucial for eliminating potentially malignant cells, controlling hyperplasia, and inhibiting tumor progression (79). The induction of apoptosis is fundamental in cancer therapy. The dual role of ANGPTL4 in regulating tumor cell apoptosis has been reported.
Lim et al (80) reported the anti-apoptotic effect of ANGPTL4 and observed that inhibiting ANGPTL4 leads to the accumulation of chemotherapy drugs in cells, thereby inducing apoptosis. The mechanism involves reduced energy production and accumulation in cancer cells and the weakened drug efflux during EMT. In HCC (81), ANGPTL4 knockout cell lines exhibited significantly higher levels of apoptosis. Bai et al (41) further investigated the role of different ANGPTL4 transcripts and found a notable increase in ANGPTL4-Transcript 3 expression in HCC tissues. The overexpression of ANGPTL4-Transcript 3 significantly confers apoptosis resistance to HCC cells, whereas Transcript 1 has no such effects (41). Fang et al (43) conducted a study on the role of ANGPTL4 in regulating gefitinib resistance in NSCLC cells. The aforementioned study demonstrated that ANGPTL4 is crucial in inhibiting cell apoptosis by suppressing the NOD-like receptor family, pyrin domain containing 3 (NLRP3)/apoptosis-associated Speck-like protein (ASC)/caspase-8 pathway, thereby enhancing resistance to gefitinib (43). ANGPTL4 also enhances the apoptosis resistance in CRC (38,82). Knocking down bone morphogenetic protein 7 (BMP7) reverses the anti-apoptotic effect of ANGPTL4 overexpression, suggesting that ANGPTL4 may inhibit apoptosis in CRC cells by upregulating BMP7 (82).
Most studies to date have highlighted the anti-apoptotic effect of ANGPTL4 in tumors. However, Hsieh et al (83) reported the pro-apoptotic effect of ANGPTL4 in UC. Cyproheptadine was found to upregulate ANGPTL4 expression and activate apoptosis-related proteins such as caspase-3 and poly (ADP-ribose) polymerase (PARP), thereby promoting apoptosis and inhibiting the growth of UC cells. This process might involve the regulation of glycogen synthase kinase 3β (GSK3β)/tuberous sclerosis complex subunit 2 (TSC2)/mTOR and GSK3β/β-catenin signaling pathways.
ANGPTL4 and anoikis
Anoikis is a specialized form of apoptosis that occurs when cells detach from the surrounding cellular or extracellular matrix (ECM) (84,85). However, tumor cells can develop resistance to anoikis, enabling them to evade cell death and continue proliferating after detachment. Anoikis resistance facilitates immune evasion, alters the TME, and ultimately contributes to the invasion and metastasis (85,86).
In HNSCC, EGF induces ANGPTL4 expression, significantly enhancing anoikis resistance, and promoting migration and invasion (87). This effect is mediated through the expression of MMPs, particularly MMP-1, as regulated by ANGPTL4 (87). Shen et al (88) discovered that OA induces ANGPTL4 expression in HNSCC, which significantly enhances the anoikis resistance by upregulating Ras-related C3 botulinum toxin substrate 1 (Rac1)/cell division control protein 42 (Cdc42) and MMP-9 pathways, thus promoting tumor metastasis.
Zhu et al (89) found that ANGPTL4 secreted by tumors specifically binds to integrins β1 and β5, leading to the activation of focal adhesion kinase (FAK) and Rac1. This activation increases the O2-: H2O2 ratio, subsequently activating the Src, which triggers the phosphatidylinositol 3-kinase (PI3K)/AKT and extracellular signal-regulated kinase (ERK) pathways, resulting in the resistance to anoikis (89). Similarly, in GC, ANGPTL4 inhibits the occurrence of anoikis by regulating the focal FAK/Src/PI3K-AKT/ERK pathway and suppressing caspases-3, -8, and -9 (90).
During the peritoneal metastasis of CRC, ADSCs enhance the anoikis resistance of CRC cells through the TGF-β1/Smad3/ANGPTL4 axis (30). Additionally, Hao et al (59) demonstrated that the downregulation of ANGPTL4 weakens the endothelial barrier disruption and promotes anoikis in lung cancer. Furthermore, the overexpression of ANGPTL4 is closely associated with anoikis resistance in cholangiocarcinoma (91), HCC (92) and uveal melanoma (93).
These findings provide new insights into the mechanisms of metastasis, suggesting that ANGPTL4 may serve as a promising therapeutic target for intervening in anoikis and tumor metastasis. The FAK/Src/PI3K-AKT/ERK pathway, along with MMPs, is the key mediator of anoikis regulated by ANGPTL4.
ANGPTL4 and ferroptosis
Ferroptosis, an iron-dependent form of cell death, is characterized by excessive iron accumulation, lipid peroxidation and cell membrane rupture (94,95). It can be triggered via the inhibition of intracellular antioxidant enzymes such as glutathione peroxidase 4 (GPX4) (96,97). Ferroptosis has been identified as a key mechanism in tumor development and radiation respons (98-100).
Zhang et al (101) uncovered the molecular function of ANGPTL4 in hypoxic TME and proposed that hypoxia-induced ANGPTL4 contributes to radiotherapy resistance in NSCLC by regulating ferroptosis. Under hypoxic conditions, the expression of ANGPTL4 is significantly upregulated in NSCLC cells and can be enriched in extracellular vesicles, which can be transferred to adjacent normoxic cells (101). Both in vivo and in vitro experiments have confirmed that ANGPTL4 inhibits ferroptosis by regulating radiation-induced lipid peroxidation and the expression of hallmark ferroptosis proteins, such as GPX4 and ferritin heavy chain 1 (101).
By contrast, Yang et al (102) reported the opposite effect in OVC. The study identified ANGPTL4 as a direct target gene of transcriptional coactivator with PDZ binding motif (TAZ) through the integrated genomic analysis (102). The upregulation of ANGPTL4 activated NADPH oxidase 2 (NOX2) in vitro, thereby increasing the sensitivity to ferroptosis (102).
ANGPTL4 and pyroptosis
Pyroptosis is a previously identified form of PCD characterized by immune responses and inflammation (103,104). As a critical innate immune response, pyroptosis induces immune phagocytosis to counter infections and endogenous threats (105-107). Pyroptosis is triggered by caspase-1, -4, -5 and -11, and activated by inflammasomes such as NLRP3 (108-110).
The role of ANGPTL4 in pyroptosis has been reported in sepsis-induced acute lung injury, where its knockdown was shown to inhibit macrophage M1 polarization and pyroptosis, thereby providing a protective effect (111). In a study by Fang et al (43), ANGPTL4 was found to be highly expressed in lung adenocarcinoma cells, and its knockdown leads to increased pyroptosis via the NLRP3/apoptosis-associated speck-like protein containing a CARD (ASC)/Caspase-8 signaling pathway (43). Currently, the research on the regulation of tumor pyroptosis by ANGPTL4 is limited, and further studies are needed to clarify its precise role in the pyroptosis of tumor cells.
ANGPTL4 and tumor metabolism
Metabolism is a fundamental biological activity intrinsic to all organisms, reflecting the processes of matter and energy transformation. The rapid proliferation and high energy demand of tumor cells lead to the significant metabolic alterations, which provide a biochemical basis and directly promote tumorigenicity and malignancy (112-116). The metabolic reprogramming of glucose, lipids and amino acids, three major functional biomolecules, enables tumor cells to acquire energy through various pathways, supporting their uncontrolled proliferation and survival. Targeting these metabolic pathways has become a promising anticancer strategy.
ANGPTL4 and glucose metabolic reprogramming
Glucose is the most critical energy source for living organisms, and its metabolic pathways include glycolysis, the pentose phosphate pathway and oxidative phosphorylation. Even in the presence of oxygen, tumor cells predominantly produce ATP through glycolysis, a phenomenon known as aerobic glycolysis or the Warburg effect (117,118). Aerobic glycolysis is crucial for the proliferation, growth, invasion and treatment of cancer (119).
In recent years, an increasing number of studies have shown that ANGPTL4 plays a significant role in tumor aerobic glycolysis. Zheng et al (120) found that in Fusobacterium nucleatum-infected CRC cells, increased acetylation of histone H3 lysine 27 upregulates the expression of ANGPTL4. ANGPTL4 promotes glucose uptake and aerobic glycolysis in CRC cells both in vitro and in vivo, which in turn enhances Fusobacterium nucleatum colonization. This effect is mediated by ANGPTL4's regulation of glucose transporter-1 (GLUT-1), thereby promoting the development, metastasis and chemoresistance of CRC. Similarly, in a study by Mizuno et al (121), it has been shown that ANGPTL4 affects the expression of GLUT-1 and GLUT-3 in CRC, and is associated with glucose metabolism activity and cancer progression. These studies indicate that GLUTs, particularly GLUT-1, is a key molecule in ANGPTL4-mediated regulation of aerobic glycolysis. Additionally, ADSCs derived from fat tissues have been shown to promote glycolysis in CRC cells through the TGF-β1/Smad3/ANGPTL4 axis, ultimately facilitating peritoneal metastasis (30). Overall, ANGPTL4 serves as a key target for regulating aerobic glycolysis and tumor progression in CRC.
At present, most research on the regulation of glucose metabolic reprogramming by ANGPTL4 has focused on CRC, and its regulatory roles and associated molecular mechanisms in other tumors require further investigations.
A NGPTL 4 and lipid metabolic reprogramming
Reprogramming of lipid metabolism is a newly recognized hallmark of malignancy (122,123). Increased lipid uptake, storage and lipogenesis are observed in various cancers and contribute to accelerated tumor growth (124-127). ANGPTL4, as a lipid regulatory factor, has been widely reported for its role in lipid metabolism, particularly its effects on LPL (7,14,128-131).
Numerous studies have demonstrated the close association between ANGPTL4 and lipid metabolism in tumors. In GC, Xiao et al (60) discovered that leptin induces the phosphorylation of the serine 30 residue of ANGPTL4, thereby reducing its binding affinity with LPL. This reduction promotes LPL-mediated lipid uptake and increases intracellular arachidonic acid levels, disrupting cellular lipid homeostasis, and triggering the COX-2/PGE2 pathway. Consequently, this process promotes tumor lymphangiogenesis and lymph node metastasis in GC. Xiao et al (132) reported that ANGPTL4 significantly affects fatty acid oxidation and promotes energy generation in NSCLC cells, which may be achieved through carnitine palmitoyl-transferase 1 (CPT1). Hu et al (42) identified ANGPTL4 as a direct target of miR-133a-3p, with its expression significantly accelerating lipid metabolism in lung adenocarcinoma.
ANGPTL4 also mediates the metabolic crosstalk between tumor cells and adipocytes. Blücher et al (133) demonstrated that adipose tissue secretory factors reprogram tumor lipid metabolism and induce motility by regulating PPAR α/ANGPTL4 and FAK in TNBC cells, with ANGPTL4 identified as the key factor in lipid metabolism regulation. In pancreatic cancer, adipocytes activate the hypoxia signaling pathway, leading to increased expression of ANGPTL4 (134). This upregulation enhances β-oxidation in cancer cells and activates the STAT3 signaling pathway, promoting lipid metabolic reprogramming and metastasis of pancreatic cancer (134).
ANGPTL4 and amino acid metabolic reprogramming
Amino acids, the building blocks of proteins, generate metabolites that fuel biosynthetic pathways, produce energy, and support cancer cell survival. Due to their heightened proliferative needs, cancer cells often cannot synthesize sufficient quantities of amino acids. The reprogramming of amino acid metabolism fulfills this increased demand (135).
Currently, limited research exists on the relationship between ANGPTL4 and the reprogramming of amino acid metabolism in cancer. Xiao et al (132) utilized tracer technology and Seahorse XF technology to explore the metabolic effects of ANGPTL4 in NSCLC. Their findings revealed that ANGPTL4 promotes glutamine consumption and fatty acid oxidation in NSCLC, both in vitro and in vivo, while having no significant effect on glycolysis. ANGPTL4 regulates glutaminase and CPT1, thereby facilitating glutamine metabolism and fatty acid oxidation, which in turn supports NSCLC cell proliferation (132). Lin et al (49) identified a negative correlation between ANGPTL4 and BCAA metabolism in OS samples and cell lines. The downregulation of ANGPTL4 in OS cells leads to the accumulation of BCAAs and the activation of mTOR signaling, which promotes OS cell proliferation (49).
ANGPTL4, chemoresistance and radioresistance
Chemotherapy and radiotherapy are essential treatments for unresectable tumors and serve as neoadjuvant therapies. However, their clinical efficacy is often hindered by the development of resistance. Chemoresistance and radioresistance also contribute to tumor recurrence and metastasis, posing major challenges to patient prognosis (136,137). Research has increasingly focused on how ANGPTL4, a multifunctional molecule in tumor cell survival, proliferation and apoptosis, influences the resistance to chemotherapy and radiotherapy (Table III).
Chemoresistance promoted by ANGPTL4 is closely tied to its metabolic reprogramming functions. It has been shown that ANGPTL4 enhances cellular ATP through the c-Myc and NF-κ B signaling pathways during EMT (80). The increased ATP provides fuel for multiple ATP-binding cassette (ABC) transporters to upregulate their expression, ensuring sufficient cellular energy for drug efflux, thereby conferring chemoresistance in tumors (80). In HCC, ANGPTL4 induces the upregulation of pyruvate dehydrogenase kinase 4, an inhibitor of mitochondrial pyruvate dehydrogenase, enhancing the resistance to sorafenib and cisplatin in stem cells (138). These findings suggest that targeting the metabolic function of ANGPTL4 and combining the restoration of mitochondrial metabolic activity with chemotherapy present attractive therapeutic options in cancer treatment.
In OVC, TAZ has been shown to promote the resistance to cisplatin via the ANGPTL4/sex-determining region Y-box2 (SOX2) axis (139). Similarly, Zhou et al (140) reported that adipocyte-derived ANGPTL4 induces carboplatin resistance in OVC. ANGPTL4 activates the c-Myc/NF-κ B pathway, which subsequently stimulates the expression of the anti-apoptotic proteins and ABC transporter family members. However, Xu et al (102) came to the opposite conclusion. They found that TAZ activates the ANGPTL4/NOX2 axis, making OVC cells sensitive to ferroptosis and chemotherapy. TAZ levels were lower in chemotherapy-resistant recurrent OVC cells.
In GBM, where temozolomide (TMZ) combined with radiotherapy is the standard therapy, glioma stem-like cells (GSCs) are considered to be the primary cause of drug resistance. In the study of Tsai et al' (141), ANGPTL4 expression was significantly increased in GSCs. The overexpression of ANGPTL4 activates the PI3K/AKT, EGFR and ERK signaling pathways, enriching GSCs and resulting in TMZ resistance (141). Additionally, Gordon et al (142) showed that ANGPTL4 contributes to gemcitabine resistance in pancreatic cancer and shortens patient survival by regulating the expression of apolipoprotein L1 and integrin β4. In cholangiocarcinoma, Curcumin has been shown to enhance chemotherapy efficacy via the inhibition of ANGPTL4 (91). Furthermore, ANGPTL4 inhibits pyroptosis and apoptosis of lung adenocarcinoma cells through the NLRP3/ASC/Caspase-8 signaling pathway, contributing to the resistance against gefitinib (43).
The role of ANGPTL4 in radioresistance has also been recently explored. Hypoxia-induced ANGPTL4 expression has been positively correlated with radiotherapy resistance in NSCLC cells and xenograft tumors (101). ANGPTL4 promotes resistance through two mechanisms: intracellular ANGPTL4 and exosomal ANGPTL4. One pathway involves the upregulation of GPX4, which inhibits ferroptosis and lipid peroxidation (101).
The aforementioned studies suggest that ANGPTL4 could serve as a potential therapeutic target to enhance the efficacy of radiotherapy and chemotherapy. However, research on the relationship between ANGPTL4 and radioresistance remains limited, and further investigations are required to improve understanding of its effects, and underlying mechanisms in chemoresistance and radioresistance.
Regulation of ANGPTL4 expression and function in cancer
The expression of ANGPTL4 is quite common in cancer cells, but the molecular mechanisms of ANGPTL4 in cancer are quite complex. HIF-1, TGF-β and PPARs are critical upstream regulators of ANGPTL4 (Table I; Fig. 3). HIF-1, a heterodimeric protein composed of HIF-1α and HIF-1β subunits, activates the transcription of numerous genes involved in numerous aspects of cancer biology. Studies in scirrhous GC (90), HCC (143), uveal melanoma (67), and OS (44) have shown that the expression of ANGPTL4 is directly regulated by HIF-1α, subsequently promoting malignant processes, including tumor growth and metastasis.
TGF-β is a multifunctional cytokine that plays important roles in cell proliferation, differentiation, immune regulation, apoptosis, and tissue repair. The Smad pathway and PPARs are essential for the expression of ANGPTL4 induced by TGF-β. In BRC, TGF-β promotes ANGPTL4 expression via the Smad pathway, mediating the lung metastasis and brain metastasis (31,127). Similarly, TGF-β1 activates Smad3, which binds to the ANGPTL4 promoter and promotes the transcription in CRC (30). Adhikary et al (144) found that TGF-β can also induce ANGPTL4 expression in a PPAR β/δ-dependent manner.
PPARs are a class of ligand-dependent transcription factors in the nuclear receptor superfamily. PPARs regulate the expression of ANGPTL4 in several cancers, including BRC and HNSCC. In BRC, PPAR α, PPAR β, and PPAR γ target ANGPTL4 to promote lipid metabolism, angiogenesis, tumor growth and invasion (133,144,145). In HNSCC, PPARs are induced by OA and target ANGPTL4, promoting anoikis resistance and metastasis (88).
Multiple signaling pathways are associated with the regulation of tumor development by ANGPTL4, with the STATs, PI3K/AKT, and COX-2/PGE2 pathways being particularly prominent (Table I; Fig. 3). The STAT family is the transcription factors, which have been implicated in cancer development, metastasis and resistance to treatments. To date, seven STAT genes have been identified: STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b and STAT6. Specifically, the functions of ANGPTL4 are closely linked to the STAT1, STAT2 and STAT3 pathways. ANGPTL4 activates the JAK2/STAT3 pathway, which promotes the malignant progression of OVC (35). In pancreatic cancer, STAT3 activated by ANGPTL4 also drives metabolic reprogramming, invasion and metastasis (134). Additionally, ANGPTL4 enhances CRC cell proliferation through the activation of STAT1 (146). STAT2 induces the linc02231/hnRNPA1/ANGPTL4 axis, which decreases the expression of ANGPTL4 and promotes the tumorigenesis and angiogenesis in CRC (52).
The PI3K/AKT pathway is another critical signaling cascade in the network of ANGPTL4. PI3K is a lipid kinase responsible for propagating intracellular signaling cascades and regulating numerous cellular processes (147). AKT, a major downstream effector of PI3K, influences multiple vital pathways for tumor growth, apoptosis and cellular metabolism (147). ANGPTL4 has been found to activate FAK and Rac1, and subsequently trigger Src. This cascade leads to the activation of PI3K/AKT and ERK pathways, contributing to the anoikis resistance (89,90). The PI3K/AKT and ERK signaling pathways activated by ANGPTL4 also result in GSCs enrichment and the resistance to TMZ (141). Moreover, ANGPTL4 regulates the expression of GLUTs via the PI3K/AKT pathway, which promotes the aerobic glycolysis in CRC (121).
The COX-2/PGE2 pathway, which plays a significant role in inflammation, is also implicated in tumor progression. COX enzymes convert arachidonic acid into five prostaglandins, with PGE2 being the most abundant. In the present review, the COX-2/PGE2 pathway is highlighted in the ANGPTL4-mediated tumor metastasis and lipid metabolic reprogramming. This indicates that ANGPTL4 may facilitate the crosstalk between tumorigenesis and inflammation. In GC, leptin reduces the binding affinity of ANGPTL4 to LPL, enhancing LPL-mediated lipid uptake, which increases intracellular arachidonic acid levels (60). Arachidonic acid then activates the COX-2/PGE2 pathway, promoting lymph node metastasis (60). Additionally, Chiang et al (61) demonstrated that the COX-2/PGE2/ANGPTL4 axis, activated by EGF, enhances metastasis in HNSCC.
ANGPTL4 in the TME
The TME consists of various components, including tumor cells, stromal cells, blood vessels, immune cells and ECM. These elements affect the biological characteristics of tumors through complex interactions and are crucial in tumor initiation and progression (148). The critical involvement of ANGPTL4 in the TME has been increasingly highlighted. For example, ANGPTL4 has been shown to activate the JAK2/STAT3 pathway, enhancing tumorigenesis and angiogenesis in the TME (35). Zhu et al (30) emphasized that ADSCs in the TME promote tumor glycolysis and metastasis via ANGPTL4 in CRC. Immune cells are an important component of TME, and new evidence links ANGPTL4 with immune cell dynamics in the TME. In patients with CRC, ANGPTL4 suppresses the activation of CD8+ T cells through metabolic reprogramming, leading to diminished immune surveillance (32). Recombinant ANGPTL4 has also been reported to induce regulatory T (Treg) cells and M2 macrophages in mice, which may contribute to the tumor progression (149). Furthermore, spatial transcriptomics analyses have revealed that bladder cancer cells in the stressed or hypoxic state interact with plasma cells via the ANGPTL4/Syndecan-1 (SDC1) axis, which is associated with ineffective responses to immunotherapy and poor survival (150).
Sialylation, a common glycosylation modification of attaching sialic acid to the ends of sugar chains, is widely present in cell membranes, secreted proteins and serum proteins (151,152). This process significantly influences the function, stability, immune recognition and intercellular communication of cells and proteins in the TME (151). Recent studies have identified the distinct roles of ANGPTL4 in podocytopathies due to varying degrees of sialylation. Hyposialylated ANGPTL4 with a high isoelectric point (high-pI) is upregulated in patients with minimal change disease and correlates with increased proteinuria. By contrast, normal sialylated ANGPTL4 with a neutral isoelectric point (neutral-pI) reduces proteinuria by binding to β5 integrin (153,154).
In tumors, high-pI ANGPTL4, with fewer negative charges, may enhance tumor cell adhesion and migration by binding more effectively to collagen and fibronectin in the ECM. Its high affinity for vascular endothelial cells may also enable it to play a more direct role in angiogenesis. Conversely, neutral-pI ANGPTL4 demonstrates greater stability in body fluids and may support the sustained regulation of angiogenesis. Moreover, neutral-pI ANGPTL4 can directly bind to sialic acid-binding immunoglobulin-like lectin (Siglec) receptors, which may inhibit the activity of natural killer (NK) cells and macrophages, thereby facilitating tumor immune evasion (155,156). Jin et al (51) found that nANGPTL4 exerts antitumor effects in clear cell renal cell carcinoma (ccRCC) by regulating the lysosomal acid lipase activity. However, no studies have explored whether high-pI and neutral-pI ANGPTL4 exhibit different impacts in renal and other tumors, as they do in podocytopathies. Future research could focus on the sialylation of ANGPTL4 in the TME to clarify its roles and associated mechanisms.
Discussion
A substantial body of research has demonstrated that ANGPTL4 plays critical biological roles, including the regulation of tumor growth, metastasis and angiogenesis (Fig. 2). It is also involved in the regulatory processes of programmed cell death, metabolic reprogramming and drug resistance (Fig. 2). Taken together, all the evidence cited in the present review indicates that ANGPTL4 is an important molecule implicated in various aspects of cancer progression.
ANGPTL4 is overexpressed in various cancers and its expression is closely linked to clinicopathological features, such as BRC, cholangiocarcinoma, cervical, esophageal and gallbladder cancer (157-161). In the majority of cancers, such as TNBC, GBM and NSCLC, the overexpression of ANGPTL4 significantly enhances tumor progression (31,42,141). Additionally, ANGPTL4 has been found to promote the resistance to treatments (Table III).
However, the role of ANGPTL4 in tumors remains complex and controversial due to its dual effects. ANGPTL4 can both promote and inhibit tumorigenesis (31,34,62) (Fig. 1). It not only serves as a biomarker for poor prognosis (157,158,162,163), but also indicates favorable prognosis (164), and even exhibits the opposing roles within the same type of cancers (47). It is hypothesized that these contradictory effects are related to the structural and functional characteristics of ANGPTL4. Firstly, ANGPTL4 undergoes proteolytic cleavage, producing different functional fragments. The hydrolysis-generated nANGPTL4 and cANGPTL4 fragments may exert distinct biological effects compared with fANGPTL4. The nANGPTL4 inhibits LPL activity in both blood and adipocytes, leading to reduced circulating triglyceride levels (165-170). The nANGPTL4 also inhibits metastasis and improves overall survival by inhibiting WNT signaling and reducing vascularity at the metastatic site (53). Meanwhile, the cANGPTL4 fragment plays a role in angiogenesis, increases vascular permeability, causes endothelial damage, and promotes tumor growth and metastasis in multiple tumor models (47,53,171,172). Furthermore, fANGPTL4 exists in tumor tissues, whereas nANGPTL4 is more prevalent in systemic circulation (53). Secondly, as a protein capable of entering the nucleus, ANGPTL4 can directly or indirectly influence the expression of various genes, thereby performing multiple functions. It may also be secreted into the extracellular space or transported through exosomes and extracellular vesicles, potentially contributing to the intercellular and the inter-organ communication (68,101). This diversity in function may help explain its complex roles. Finally, differences in the TME and tissue-specific conditions may contribute to the diverse functions of ANGPTL4. For instance, while ANGPTL4 expression is low in UC cell lines and tissue samples, the elevated circulating levels are observed in patient samples (47).
The strong association between ANGPTL4 and the tumor progression highlights its potential as a promising biomarker and therapeutic target. Its expression is closely associated with prognosis in various cancers, supporting the rationale for developing ANGPTL4 as a biomarker (157,158,162-164). Additionally, the differential expression and functions of ANGPTL4 across cancer types suggest that targeting ANGPTL4 enables offering personalized treatment strategies tailored to specific tumor characteristics (31,34,62) (Fig. 1). Jin et al (51) identified a subset of patients with no ANGPTL4 increase in ccRCC, who had a poorer prognosis than those with high ANGPTL4 expression. This finding highlights the potential of stratifying patients based on ANGPTL4 expression levels, which may not only provide the more accurate predictions of survival outcomes but also optimize therapeutic interventions. Furthermore, as ANGPTL4 is closely connected with chemoresistance and radioresistance (Table III), the development of ANGPTL4-targeted drugs or antibodies for combination therapy holds significant promise to enhance treatment efficacy and patient outcomes. ANGPTL4 also acts as a potent pro-angiogenic factor in certain cancers (35,37,65,66). Targeting ANGPTL4 may therefore provide a novel anti-angiogenic approach. In addition, as aforementioned, ANGPTL4 regulates various immune cells in the TME, including CD8+T cells, NK cells, macrophages and plasma cells (32,164,165,173). Antibodies against ANGPTL4 may help restore immune surveillance and enhance antitumor immunity. Combining ANGPTL4-targeted antibodies with existing immunotherapies, such as programmed cell death protein 1 and programmed death-ligand 1 inhibitors, offers new perspectives for the combination immunotherapy.
Despite current advancements in research, numerous key gaps remain in fully understanding ANGPTL4. Firstly, ANGPTL4 exhibits both tumor-promoting and tumor-inhibiting effects in the progression of different cancers, yet its underlying molecular mechanisms have not been insufficiently elucidated. Secondly, although ANGPTL4 can be expressed in the nucleus or secreted, the specific form in which it exists across various organs and diseases is unclear. Whether the ANGPTL4 protein is modified post-secretion during this process remains unknown. Thirdly, ANGPTL4 can undergo glycosylation modifications structurally, but it is unclear how different modifications such as sialylation affect the role and mechanism of ANGPTL4 in tumors. Finally, although some inhibitors targeting the ANGPT family have been explored, including MEDI-3617 and nesvacumab (targeting ANGPT2), and trebananib and AMG780 (targeting both ANGPT1 and ANGPT2), no inhibitors or drugs targeting ANGPTL4 have been developed to date (173). The clinical application of ANGPTL4 as a biomarker for cancer and the development of novel antitumor drugs targeting ANGPTL4 still require extensive experimental research and exploration.
The present review updates the understanding of ANGPTL4 in tumors by systematically analyzing its roles in the six hallmarks of cancer and its interactions with the TME. It also addresses the dual effects of ANGPTL4 across different cancer types and highlights its potential in precision medicine. These contributions provide a comprehensive, in-depth and innovative perspective on ANGPTL4 in malignancies.
In conclusion, ANGPTL4 predominantly exerts pro-tumor effects, yet its antitumor functions should not be overlooked. Further research is necessary to fully understand its roles, including the functions of different ANGPTL4 fragments, and the effects and interactions of ANGPTL4 in the TME. Exploring the diverse mechanisms of ANGPTL4 in human cancers and assessing its clinical value will be crucial for future studies. With a deeper understanding of its structure, function and drug response, supported by comprehensive preclinical analyses, ANGPTL4 holds significant potential for future application in clinical prediction and therapy.
Availability of data and materials
Not applicable.
Authors' contributions
ZW and YC conceived the study. RL, MF and PC wrote the original draft of the manuscript. RL, YL, XS and PZ wrote, reviewed and edited the manuscript. RL, MF and WH illustrated the figures. ZW and YC supervised the study. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
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.
Acknowledgements
Not applicable.
Funding
The present study was supported by the National Natural Science Foundation of China (grant no. 82172664) and the Natural Science Foundation of Shandong (grant no. ZR2022MH074).
References
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI | |
Gilbertson RJ: Mapping cancer origins. Cell. 145:25–29. 2011. View Article : Google Scholar : PubMed/NCBI | |
Dhanasekaran R, Deutzmann A, Mahauad-Fernandez WD, Hansen AS, Gouw AM and Felsher DW: The MYC oncogene - the grand orchestrator of cancer growth and immune evasion. Nat Rev Clin Oncol. 19:23–36. 2022. View Article : Google Scholar | |
Huang H, Bhat A, Woodnutt G and Lappe R: Targeting the ANGPT-TIE2 pathway in malignancy. Nat Rev Cancer. 10:575–585. 2010. View Article : Google Scholar : PubMed/NCBI | |
Eklund L, Kangas J and Saharinen P: Angiopoietin-Tie signalling in the cardiovascular and lymphatic systems. Clin Sci (Lond). 131:87–103. 2017. View Article : Google Scholar | |
Guo L, Li SY, Ji FY, Zhao YF, Zhong Y, Lv XJ, Wu XL and Qian GS: Role of Angptl4 in vascular permeability and inflammation. Inflamm Res. 63:13–22. 2014. View Article : Google Scholar | |
Aryal B, Price NL, Suarez Y and Fernández-Hernando C: ANGPTL4 in Metabolic and Cardiovascular Disease. Trends Mol Med. 25:723–734. 2019. View Article : Google Scholar : PubMed/NCBI | |
Tan MJ, Teo Z, Sng MK, Zhu P and Tan NS: Emerging roles of angiopoietin-like 4 in human cancer. Mol Cancer Res. 10:677–688. 2012. View Article : Google Scholar : PubMed/NCBI | |
La Paglia L, Listì A, Caruso S, Amodeo V, Passiglia F, Bazan V and Fanale D: Potential role of ANGPTL4 in the cross talk between metabolism and cancer through PPAR signaling pathway. PPAR Res. 2017:81872352017. View Article : Google Scholar : PubMed/NCBI | |
Kersten S: Angiopoietin-like 3 in lipoprotein metabolism. Nat Rev Endocrinol. 13:731–739. 2017. View Article : Google Scholar : PubMed/NCBI | |
Sylvers-Davie KL and Davies BSJ: Regulation of lipoprotein metabolism by ANGPTL3, ANGPTL4, and ANGPTL8. Am J Physiol Endocrinol Metab. 321:E493–E508. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yang J, Song QY, Niu SX, Chen HJ, Petersen RB, Zhang Y and Huang K: Emerging roles of angiopoietin-like proteins in inflammation: Mechanisms and potential as pharmacological targets. J Cell Physiol. 237:98–117. 2022. View Article : Google Scholar | |
Thorin E, Labbé P, Lambert M, Mury P, Dagher O, Miquel G and Thorin-Trescases N: Angiopoietin-like proteins: Cardiovascular biology and therapeutic targeting for the prevention of cardiovascular diseases. Can J Cardiol. 39:1736–1756. 2023. View Article : Google Scholar : PubMed/NCBI | |
Kersten S: Role and mechanism of the action of angiopoietin-like protein ANGPTL4 in plasma lipid metabolism. J Lipid Res. 62:1001502021. View Article : Google Scholar : PubMed/NCBI | |
Zuo Y, He Z, Chen Y and Dai L: Dual role of ANGPTL4 in inflammation. Inflamm Res. 72:1303–1313. 2023. View Article : Google Scholar : PubMed/NCBI | |
Kim I, Kim HG, Kim H, Kim HH, Park SK, Uhm CS, Lee ZH and Koh GY: Hepatic expression, synthesis and secretion of a novel fibrinogen/angiopoietin-related protein that prevents endothelial-cell apoptosis. Biochem J. 346:603–610. 2000. View Article : Google Scholar : PubMed/NCBI | |
Yoon JC, Chickering TW, Rosen ED, Dussault B, Qin Y, Soukas A, Friedman JM, Holmes WE and Spiegelman BM: Peroxisome proliferator-activated receptor gamma target gene encoding a novel angiopoietin-related protein associated with adipose differentiation. Mol Cell Biol. 20:5343–5349. 2000. View Article : Google Scholar : PubMed/NCBI | |
Kersten S, Mandard S, Tan NS, Escher P, Metzger D, Chambon P, Gonzalez FJ, Desvergne B and Wahli W: Characterization of the fasting-induced adipose factor FIAF, a novel peroxisome proliferator-activated receptor target gene. J Biol Chem. 275:28488–28493. 2000. View Article : Google Scholar : PubMed/NCBI | |
Górecka M, Krzemiński K, Buraczewska M, Kozacz A, Dąbrowski J and Ziemba AW: Effect of mountain ultra-marathon running on plasma angiopoietin-like protein 4 and lipid profile in healthy trained men. Eur J Appl Physiol. 120:117–125. 2020. View Article : Google Scholar : | |
Li L, Foo BJW, Kwok KW, Sakamoto N, Mukae H, Izumikawa K, Mandard S, Quenot JP, Lagrost L, The WK, et al: Antibody treatment against angiopoietin-like 4 reduces pulmonary edema and injury in secondary pneumococcal pneumonia. mBio. 10:e024692019. View Article : Google Scholar : PubMed/NCBI | |
Sodhi A, Ma T, Menon D, Deshpande M, Jee K, Dinabandhu A, Vancel J, Lu D and Montaner S: Angiopoietin-like 4 binds neuropilins and cooperates with VEGF to induce diabetic macular edema. J Clin Invest. 129:4593–4608. 2019. View Article : Google Scholar : PubMed/NCBI | |
Kersten S, Lichtenstein L, Steenbergen E, Mudde K, Hendriks HF, Hesselink MK, Schrauwen P and Müller M: Caloric restriction and exercise increase plasma ANGPTL4 levels in humans via elevated free fatty acids. Arterioscler Thromb Vasc Biol. 29:969–974. 2009. View Article : Google Scholar : PubMed/NCBI | |
Singh AK, Aryal B, Chaube B, Rotllan N, Varela L, Horvath TL, Suárez Y and Fernández-Hernando C: Brown adipose tissue derived ANGPTL4 controls glucose and lipid metabolism and regulates thermogenesis. Mol Metab. 11:59–69. 2018. View Article : Google Scholar : PubMed/NCBI | |
Shu L, Wang C, Ding Z, Tang J, Zhu Y, Wu L, Wang Z, Zhang T, Wang T, Xu Y and Sun L: A novel regulated network mediated by downregulation HIF1A-AS2 lncRNA impairs placental angiogenesis by promoting ANGPTL4 expression in preeclampsia. Front Cell Dev Biol. 10:8370002022. View Article : Google Scholar : PubMed/NCBI | |
Spitler KM, Shetty SK, Cushing EM, Sylvers-Davie KL and Davies BSJ: Chronic high-fat feeding and prolonged fasting in liver-specific ANGPTL4 knockout mice. Am J Physiol Endocrinol Metab. 321:E464–E478. 2021. View Article : Google Scholar : PubMed/NCBI | |
Alex S, Lichtenstein L, Dijk W, Mensink RP, Tan NS and Kersten S: ANGPTL4 is produced by entero-endocrine cells in the human intestinal tract. Histochem Cell Biol. 141:383–391. 2014. View Article : Google Scholar | |
Kuo T, Chen TC, Yan S, Foo F, Ching C, McQueen A and Wang JC: Repression of glucocorticoid-stimulated angiopoietin-like 4 gene transcription by insulin. J Lipid Res. 55:919–928. 2014. View Article : Google Scholar : PubMed/NCBI | |
Inoue T, Kohro T, Tanaka T, Kanki Y, Li G, Poh HM, Mimura I, Kobayashi M, Taguchi A, Maejima T, et al: Cross-enhancement of ANGPTL4 transcription by HIF1 alpha and PPAR beta/delta is the result of the conformational proximity of two response elements. Genome Biol. 15:R632014. View Article : Google Scholar : PubMed/NCBI | |
Kaddatz K, Adhikary T, Finkernagel F, Meissner W, Müller-Brüsselbach S and Müller R: Transcriptional profiling identifies functional interactions of TGF β and PPAR β/δ signaling: Synergistic induction of ANGPTL4 transcription. J Biol Chem. 285:29469–29479. 2010. View Article : Google Scholar : PubMed/NCBI | |
Zhu C, Teng L, Lai Y, Yao X, Fang Y, Wang Z, Lin S, Zhang H, Li Q, Li Y, et al: Adipose-derived stem cells promote glycolysis and peritoneal metastasis via TGF-β1/SMAD3/ANGPTL4 axis in colorectal cancer. Cell Mol Life Sci. 81:1892024. View Article : Google Scholar | |
Gong X, Hou Z, Endsley MP, Gronseth EI, Rarick KR, Jorns JM, Yang Q, Du Z, Yan K, Bordas ML, et al: Interaction of tumor cells and astrocytes promotes breast cancer brain metastases through TGF-β2/ANGPTL4 axes. NPJ Precis Oncol. 3:242019. View Article : Google Scholar | |
Ding S, Lin Z, Zhang X, Jia X, Li H, Fu Y, Wang X, Zhu G, Lu G, Xiao W and Gong W: Deficiency of angiopoietin-like 4 enhances CD8(+) T cell bioactivity via metabolic reprogramming for impairing tumour progression. Immunology. 170:28–46. 2023. View Article : Google Scholar : PubMed/NCBI | |
Kolb R, Kluz P, Tan ZW, Borcherding N, Bormann N, Vishwakarma A, Balcziak L, Zhu P, Davies BS, Gourronc F, et al: Obesity-associated inflammation promotes angiogenesis and breast cancer via angiopoietin-like 4. Oncogene. 38:2351–2363. 2019. View Article : Google Scholar : | |
Avalle L, Raggi L, Monteleone E, Savino A, Viavattene D, Statello L, Camperi A, Stabile SA, Salemme V, De Marzo N, et al: STAT3 induces breast cancer growth via ANGPTL4, MMP13 and STC1 secretion by cancer associated fibroblasts. Oncogene. 41:1456–1467. 2022. View Article : Google Scholar : PubMed/NCBI | |
Li YK, Gao AB, Zeng T, Liu D, Zhang QF, Ran XM, Tang ZZ, Li Y, Liu J, Zhang T, et al: ANGPTL4 accelerates ovarian serous cystadenocarcinoma carcinogenesis and angiogenesis in the tumor microenvironment by activating the JAK2/STAT3 pathway and interacting with ESM1. J Transl Med. 22:462024. View Article : Google Scholar : PubMed/NCBI | |
Xu J, Wu F, Zhu Y, Wu T, Cao T, Gao W, Liu M, Qian W, Feng G, Xi X and Hou S: ANGPTL4 regulates ovarian cancer progression by activating the ERK1/2 pathway. Cancer Cell Int. 24:542024. View Article : Google Scholar : PubMed/NCBI | |
Wu Y, Gao J and Liu X: Deregulation of angiopoietin-like 4 slows ovarian cancer progression through vascular endothelial growth factor receptor 2 phosphorylation. Cancer Cell Int. 21:1712021. View Article : Google Scholar : PubMed/NCBI | |
Wen L, Zhang Y, Yang B, Han F, Ebadi AG and Toughani M: Knockdown of angiopoietin-like protein 4 suppresses the development of colorectal cancer. Cell Mol Biol (Noisy-le-grand). 66:117–124. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chen JW, Luo YJ, Yang ZF, Wen LQ and Huang L: Knockdown of angiopoietin-like 4 inhibits the development of human gastric cancer. Oncol Rep. 39:1739–1746. 2018.PubMed/NCBI | |
Bai Y, Cui G, Sun X, Wei M, Liu Y, Guo J and Yang Y: ANGPTL4 stabilizes bone morphogenetic protein 7 through deubiquitination and promotes HCC proliferation via the SMAD/MAPK pathway. DNA Cell Biol. 43:395–400. 2024. View Article : Google Scholar : PubMed/NCBI | |
Bai Y, Cui G, Sun X, Wei M, Liu Y, Guo J and Yang Y: Angiopoietin-related protein 4-Transcript 3 increases the proliferation, invasion, and migration of hepatocellular carcinoma cells and inhibits apoptosis. DNA Cell Biol. 43:175–184. 2024. View Article : Google Scholar : PubMed/NCBI | |
Hu Q, Chen S, Li Y, Hu T, Hu J, Wang C, Yang F, Yang X, Zhou F, Liu Z, et al: ANGPTL4, a direct target of hsa-miR-133a-3p, accelerates lung adenocarcinoma lipid metabolism, proliferation and invasion. Aging (Albany NY). 16:8348–8360. 2023. | |
Fang Y, Li X, Cheng H, Zhang L and Hao J: ANGPTL4 regulates lung adenocarcinoma pyroptosis and apoptosis via NLRP3\ASC\ Caspase 8 signaling pathway to promote resistance to gefitinib. J Oncol. 2022:36235702022. View Article : Google Scholar | |
Zhang T, Kastrenopoulou A, Larrouture Q, Athanasou NA and Knowles HJ: Angiopoietin-like 4 promotes osteosarcoma cell proliferation and migration and stimulates osteoclastogenesis. BMC Cancer. 18:5362018. View Article : Google Scholar : PubMed/NCBI | |
Yang L, Wang Y, Sun R, Zhang Y, Fu Y, Zheng Z, Ji Z and Zhao D: ANGPTL4 promotes the proliferation of papillary thyroid cancer via AKT pathway. Onco Targets Ther. 13:2299–2309. 2020. View Article : Google Scholar : PubMed/NCBI | |
Izraely S, Ben-Menachem S, Sagi-Assif O, Meshel T, Marzese DM, Ohe S, Zubrilov I, Pasmanik-Chor M, Hoon DSB and Witz IP: ANGPTL4 promotes the progression of cutaneous melanoma to brain metastasis. Oncotarget. 8:75778–75796. 2017. View Article : Google Scholar : PubMed/NCBI | |
Hsieh HY, Jou YC, Tung CL, Tsai YS, Wang YH, Chi CL, Lin RI, Hung SK, Chuang YM, Wu SF, et al: Epigenetic silencing of the dual-role signal mediator, ANGPTL4 in tumor tissues and its overexpression in the urothelial carcinoma microenvironment. Oncogene. 37:673–686. 2018. View Article : Google Scholar | |
Hui B, Ji H, Xu Y, Wang J, Ma Z, Zhang C, Wang K and Zhou Y: RREB1-induced upregulation of the lncRNA AGAP2-AS1 regulates the proliferation and migration of pancreatic cancer partly through suppressing ANKRD1 and ANGPTL4. Cell Death Dis. 10:2072019. View Article : Google Scholar : PubMed/NCBI | |
Lin S, Miao Y, Zheng X, Dong Y, Yang Q, Yang Q, Du S, Xu J, Zhou S and Yuan T: ANGPTL4 negatively regulates the progression of osteosarcoma by remodeling branched-chain amino acid metabolism. Cell Death Discov. 8:2252022. View Article : Google Scholar : PubMed/NCBI | |
Qian P, Li J, Zhang X, Li F, Bei S, Li H, Sun Q and Feng L: LMX1A inhibits C-Myc expression through ANGPTL4 to exert tumor suppressive role in gastric cancer. PLoS One. 14:e02216402019. View Article : Google Scholar : PubMed/NCBI | |
Jin Z, De U, Tithi TI, Kleberg J, Nataraj A, Jolley E, Carelock ME, Davies BS, Zhang W and Kolb R: ANGPTL4 suppresses clear cell renal cell carcinoma via inhibition of lysosomal acid lipase. Cancer Res Commun. 4:2242–2254. 2024. View Article : Google Scholar : PubMed/NCBI | |
Xu S, Fang Y, Chang L, Bian Y, Wang Y, Ding J, Wang Y, Zhang Y, Pu J and Wang K: STAT2-induced linc02231 promotes tumorigenesis and angiogenesis through modulation of hnRNPA1/ANGPTL4 in colorectal cancer. J Gene Med. 25:e35062023. View Article : Google Scholar : PubMed/NCBI | |
Hübers C, Abdul Pari AA, Grieshober D, Petkov M, Schmidt A, Messmer T, Heyer CM, Schölch S, Kapel SS, Gengenbacher N, et al: Primary tumor-derived systemic nANGPTL4 inhibits metastasis. J Exp Med. 220:e202025952023. View Article : Google Scholar | |
Niu Y, Bao L, Chen Y, Wang C, Luo M, Zhang B, Zhou M, Wang JE, Fang YV, Kumar A, et al: HIF2-induced long noncoding RNA RAB11B-AS1 promotes hypoxia-mediated angiogenesis and breast cancer metastasis. Cancer Res. 80:964–975. 2020. View Article : Google Scholar : PubMed/NCBI | |
Shen CJ, Chang KY, Lin BW, Lin WT, Su CM, Tsai JP, Liao YH, Hung LY, Chang WC and Chen BK: Oleic acid-induced NOX4 is dependent on ANGPTL4 expression to promote human colorectal cancer metastasis. Theranostics. 10:7083–7099. 2020. View Article : Google Scholar : PubMed/NCBI | |
Shen CJ, Chan RH, Lin BW, Li NC, Huang YH, Chang WC and Chen BK: Oleic acid-induced metastasis of KRAS/p53-mutant colorectal cancer relies on concurrent KRAS activation and IL-8 expression bypassing EGFR activation. Theranostics. 13:4650–4666. 2023. View Article : Google Scholar : PubMed/NCBI | |
Bajwa P, Kordylewicz K, Bilecz A, Lastra RR, Wroblewski K, Rinkevich Y, Lengyel E and Kenny HA: Cancer-associated mesothelial cell-derived ANGPTL4 and STC1 promote the early steps of ovarian cancer metastasis. JCI Insight. 8:e1630192023. View Article : Google Scholar : PubMed/NCBI | |
Hefni E, Menon D, Ma T, Asiedu EB, Sultan A, Meiller T, Schneider A, Sodhi A and Montaner S: Angiopoietin-like 4 induces head and neck squamous cell carcinoma cell migration through the NRP1/ABL1/PXN pathway. Cell Signal. 108:1106972023. View Article : Google Scholar : PubMed/NCBI | |
Hao H, Guo Z, Li Z, Li J, Jiang S, Fu J, Jiao Y, Deng X, Han S and Li P: Modified Bu-Fei decoction inhibits lung metastasis via suppressing angiopoietin-like 4. Phytomedicine. 106:1544092022. View Article : Google Scholar : PubMed/NCBI | |
Xiao J, Cao S, Wang J, Li P, Cheng Q, Zhou X, Dong J, Li Y, Zhao X, Xu Z and Yang L: Leptin-mediated suppression of lipoprotein lipase cleavage enhances lipid uptake and facilitates lymph node metastasis in gastric cancer. Cancer Commun (Lond). 44:855–878. 2024. View Article : Google Scholar : PubMed/NCBI | |
Chiang KH, Shieh JM, Shen CJ, Chang TW, Wu PT, Hsu JY, Tsai JP, Chang WC and Chen BK: Epidermal growth factor-induced COX-2 regulates metastasis of head and neck squamous cell carcinoma through upregulation of angiopoietin-like 4. Cancer Sci. 111:2004–2015. 2020. View Article : Google Scholar : PubMed/NCBI | |
Cai YC, Yang H, Wang KF, Chen TH, Jiang WQ and Shi YX: ANGPTL4 overexpression inhibits tumor cell adhesion and migration and predicts favorable prognosis of triple-negative breast cancer. BMC Cancer. 20:8782020. View Article : Google Scholar : PubMed/NCBI | |
Zhang K, Zhai Z, Yu S and Tao Y: DNA methylation mediated down-regulation of ANGPTL4 promotes colorectal cancer metastasis by activating the ERK pathway. J Cancer. 12:5473–5485. 2021. View Article : Google Scholar : PubMed/NCBI | |
Jiang F, Wang GJ, Huang P, Chen S, Xiao H, Zhang L and Zou H: Geiparvarin inhibits OS metastasis through upregulation of ANGPTL4 expression by inhibiting miRNA-3912-3p expression. Evid Based Complement Alternat Med. 2022:46636842022.PubMed/NCBI | |
Katanasaka Y, Kodera Y, Kitamura Y, Morimoto T, Tamura T and Koizumi F: Epidermal growth factor receptor variant type III markedly accelerates angiogenesis and tumor growth via inducing c-myc mediated angiopoietin-like 4 expression in malignant glioma. Mol Cancer. 12:312013. View Article : Google Scholar : PubMed/NCBI | |
Xiao W, Tan D, Xiong X, Liu J and Xu J: Large volume collapse observed in the phase transition in cubic PbCrO3 perovskite. Proc Natl Acad Sci USA. 107:14026–14029. 2010. View Article : Google Scholar : PubMed/NCBI | |
Hu K, Babapoor-Farrokhran S, Rodrigues M, Deshpande M, Puchner B, Kashiwabuchi F, Hassan SJ, Asnaghi L, Handa JT, Merbs S, et al: Hypoxia-inducible factor 1 upregulation of both VEGF and ANGPTL4 is required to promote the angiogenic phenotype in uveal melanoma. Oncotarget. 7:7816–7828. 2016. View Article : Google Scholar : PubMed/NCBI | |
Mo F, Xu Y and Zhang J, Zhu L, Wang C, Chu X, Pan Y, Bai Y, Shao C and Zhang J: Effects of hypoxia and radiation-induced exosomes on migration of lung cancer cells and angiogenesis of umbilical vein endothelial Cells. Radiat Res. 194:71–80. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chen S, Yang M and Chang S: LncRNA CCAL Promotes Angiogenesis Through Regulating the MiR-29b/ANGPTL4 Axis in Osteosarcoma. Cancer Manag Res. 12:10521–10530. 2020. View Article : Google Scholar : PubMed/NCBI | |
Okochi-Takada E, Hattori N, Tsukamoto T, Miyamoto K, Ando T, Ito S, Yamamura Y, Wakabayashi M, Nobeyama Y and Ushijima T: ANGPTL4 is a secreted tumor suppressor that inhibits angiogenesis. Oncogene. 33:2273–2278. 2014. View Article : Google Scholar | |
Yang YH, Wang Y, Lam KS, Yau MH, Cheng KK, Zhang J, Zhu W, Wu D and Xu A: Suppression of the Raf/MEK/ERK signaling cascade and inhibition of angiogenesis by the carboxyl terminus of angiopoietin-like protein 4. Arterioscler Thromb Vasc Biol. 28:835–840. 2008. View Article : Google Scholar : PubMed/NCBI | |
Ng KT, Xu A, Cheng Q, Guo DY, Lim ZX, Sun CK, Fung JH, Poon RT, Fan ST, Lo CM and Man K: Clinical relevance and therapeutic potential of angiopoietin-like protein 4 in hepatocellular carcinoma. Mol Cancer. 13:1962014. View Article : Google Scholar : PubMed/NCBI | |
Gong L, Huang D, Shi Y, Liang Z and Bu H: Regulated cell death in cancer: From pathogenesis to treatment. Chin Med J (Engl). 136:653–665. 2023. View Article : Google Scholar | |
Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al: Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 25:486–541. 2018. View Article : Google Scholar : PubMed/NCBI | |
Moujalled D, Strasser A and Liddell JR: Molecular mechanisms of cell death in neurological diseases. Cell Death Differ. 28:2029–2044. 2021. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Ding W, Wang J, Ao X and Xue J: Non-coding RNA-mediated modulation of ferroptosis in cardiovascular diseases. Biomed Pharmacother. 164:1149932023. View Article : Google Scholar : PubMed/NCBI | |
Kerr JF, Wyllie AH and Currie AR: Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 26:239–257. 1972. View Article : Google Scholar : PubMed/NCBI | |
Nuñez G, Benedict MA, Hu Y and Inohara N: Caspases: the proteases of the apoptotic pathway. Oncogene. 17:3237–3245. 1998. View Article : Google Scholar | |
Saraste A and Pulkki K: Morphologic and biochemical hallmarks of apoptosis. Cardiovasc Res. 45:528–537. 2000. View Article : Google Scholar : PubMed/NCBI | |
Lim MMK, Wee JWK, Soong JC, Chua D, Tan WR, Lizwan M, Li Y, Teo Z, Goh WWB, Zhu P and Tan NS: Targeting metabolic flexibility via angiopoietin-like 4 protein sensitizes metastatic cancer cells to chemotherapy drugs. Mol Cancer. 17:1522018. View Article : Google Scholar : PubMed/NCBI | |
Bai Y, Cui G, Sun X, Wei M, Liu Y, Guo J and Yang Y: Effect of deletion of ANGPTL4 gene on viability, migration and invasion ability and apoptosis of hepatocellular carcinoma cells. Discov Med. 36:173–181. 2024. View Article : Google Scholar : PubMed/NCBI | |
Li X, Chen T, Shi Q, Li J, Cai S, Zhou P, Zhong Y and Yao L: Angiopoietin-like 4 enhances metastasis and inhibits apoptosis via inducing bone morphogenetic protein 7 in colorectal cancer cells. Biochem Biophys Res Commun. 467:128–134. 2015. View Article : Google Scholar : PubMed/NCBI | |
Hsieh HY, Shen CH, Lin RI, Feng YM, Huang SY, Wang YH, Wu SF, Hsu CD and Chan MW: Cyproheptadine exhibits antitumor activity in urothelial carcinoma cells by targeting GSK3β to suppress mTOR and β-catenin signaling pathways. Cancer Lett. 370:56–65. 2016. View Article : Google Scholar | |
Sattari Fard F, Jalilzadeh N, Mehdizadeh A, Sajjadian F and Velaei K: Understanding and targeting anoikis in metastasis for cancer therapies. Cell Biol Int. 47:683–698. 2023. View Article : Google Scholar | |
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S and Xiao M: Extracellular matrix remodeling in tumor progression and immune escape: From mechanisms to treatments. Mol Cancer. 22:482023. View Article : Google Scholar : PubMed/NCBI | |
Chaojun L, Pengping L, Yanjun L, Fangyuan Z, Yaning H, Yingbo S, Qi C and Hui L: TJP3 promotes T cell immunity escape and chemoresistance in breast cancer: A comprehensive analysis of anoikis-based prognosis prediction and drug sensitivity stratification. Aging (Albany NY). 15:12890–12906. 2023. View Article : Google Scholar : PubMed/NCBI | |
Liao YH, Chiang KH, Shieh JM, Huang CR, Shen CJ, Huang WC and Chen BK: Epidermal growth factor-induced ANGPTL4 enhances anoikis resistance and tumour metastasis in head and neck squamous cell carcinoma. Oncogene. 36:2228–2242. 2017. View Article : Google Scholar : | |
Shen CJ, Chan SH, Lee CT, Huang WC, Tsai JP and Chen BK: Oleic acid-induced ANGPTL4 enhances head and neck squamous cell carcinoma anoikis resistance and metastasis via up-regulation of fibronectin. Cancer Lett. 386:110–122. 2017. View Article : Google Scholar | |
Zhu P, Tan MJ, Huang RL, Tan CK, Chong HC, Pal M, Lam CR, Boukamp P, Pan JY, Tan SH, et al: Angiopoietin-like 4 protein elevates the prosurvival intracellular O2(-):H2O2 ratio and confers anoikis resistance to tumors. Cancer Cell. 19:401–415. 2011. View Article : Google Scholar : PubMed/NCBI | |
Baba K, Kitajima Y, Miyake S, Nakamura J, Wakiyama K, Sato H, Okuyama K, Kitagawa H, Tanaka T, Hiraki M, et al: Hypoxia-induced ANGPTL4 sustains tumour growth and anoikis resistance through different mechanisms in scirrhous gastric cancer cell lines. Sci Rep. 7:111272017. View Article : Google Scholar : PubMed/NCBI | |
San TT, Khaenam P, Prachayasittikul V, Sripa B, Kunkeaw N and Chan-On W: Curcumin enhances chemotherapeutic effects and suppresses ANGPTL4 in anoikis-resistant cholangiocarcinoma cells. Heliyon. 6:e032552020. View Article : Google Scholar : PubMed/NCBI | |
Zhang Z, Cao L, Li J, Liang X, Liu Y, Liu H, Du J, Qu Z, Cui M, Liu S, et al: Acquisition of anoikis resistance reveals a synoikis-like survival style in BEL7402 hepatoma cells. Cancer Lett. 267:106–115. 2008. View Article : Google Scholar : PubMed/NCBI | |
Ness C, Garred Ø, Eide NA, Kumar T, Olstad OK, Bærland TP, Petrovski G, Moe MC and Noer A: Multicellular tumor spheroids of human uveal melanoma induce genes associated with anoikis resistance, lipogenesis, and SSXs. Mol Vis. 23:680–694. 2017.PubMed/NCBI | |
Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, et al: Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 171:273–285. 2017. View Article : Google Scholar : PubMed/NCBI | |
Agmon E, Solon J, Bassereau P and Stockwell BR: Modeling the effects of lipid peroxidation during ferroptosis on membrane properties. Sci Rep. 8:51552018. View Article : Google Scholar : PubMed/NCBI | |
Tang D and Kroemer G: Ferroptosis. Curr Biol. 30:R1292–R1297. 2020. View Article : Google Scholar : PubMed/NCBI | |
Tang D, Chen X, Kang R and Kroemer G: Ferroptosis: Molecular mechanisms and health implications. Cell Res. 31:107–125. 2021. View Article : Google Scholar : | |
Li D and Li Y: The interaction between ferroptosis and lipid metabolism in cancer. Signal Transduct Target Ther. 5:1082020. View Article : Google Scholar : PubMed/NCBI | |
Pope LE and Dixon SJ: Regulation of ferroptosis by lipid metabolism. Trends Cell Biol. 33:1077–1087. 2023. View Article : Google Scholar : PubMed/NCBI | |
Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Liu X, Zeng L, Zhao X, Chen Q, Pan Y, Bai Y, Shao C and Zhang J: Exosomal protein angiopoietin-like 4 mediated radioresistance of lung cancer by inhibiting ferroptosis under hypoxic microenvironment. Br J Cancer. 127:1760–1772. 2022. View Article : Google Scholar : PubMed/NCBI | |
Yang WH, Huang Z, Wu J, Ding CC, Murphy SK and Chi JT: A TAZ-ANGPTL4-NOX2 axis regulates ferroptotic cell death and chemoresistance in epithelial ovarian cancer. Mol Cancer Res. 18:79–90. 2020. View Article : Google Scholar : | |
Broz P, Pelegrín P and Shao F: The gasdermins, a protein family executing cell death and inflammation. Nat Rev Immunol. 20:143–157. 2020. View Article : Google Scholar | |
Wei X, Xie F, Zhou X, Wu Y, Yan H, Liu T, Huang J, Wang F, Zhou F and Zhang L: Role of pyroptosis in inflammation and cancer. Cell Mol Immunol. 19:971–992. 2022. View Article : Google Scholar : PubMed/NCBI | |
Strowig T, Henao-Mejia J, Elinav E and Flavell R: Inflammasomes in health and disease. Nature. 481:278–286. 2012. View Article : Google Scholar : PubMed/NCBI | |
Fang Y, Tian S, Pan Y, Li W, Wang Q, Tang Y, Yu T, Wu X, Shi Y, Ma P and Shu Y: Pyroptosis: A new frontier in cancer. Biomed Pharmacother. 121:1095952020. View Article : Google Scholar | |
Man SM, Karki R and Kanneganti TD: Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunol Rev. 277:61–75. 2017. View Article : Google Scholar : PubMed/NCBI | |
Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y, Cai T, Wang F and Shao F: Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 526:660–665. 2015. View Article : Google Scholar : PubMed/NCBI | |
Huang Y, Xu W and Zhou R: NLRP3 inflammasome activation and cell death. Cell Mol Immunol. 18:2114–2127. 2021. View Article : Google Scholar : PubMed/NCBI | |
Sharma BR and Kanneganti TD: NLRP3 inflammasome in cancer and metabolic diseases. Nat Immunol. 22:550–559. 2021. View Article : Google Scholar : PubMed/NCBI | |
Sun B, Bai L, Li Q, Sun Y, Li M, Wang J, Shi X and Zhao M: Knockdown of angiopoietin-like 4 suppresses sepsis-induced acute lung injury by blocking the NF-κB pathway activation and hindering macrophage M1 polarization and pyroptosis. Toxicol In Vitro. 94:1057092024. View Article : Google Scholar | |
DeBerardinis RJ, Lum JJ, Hatzivassiliou G and Thompson CB: The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 7:11–20. 2008. View Article : Google Scholar : PubMed/NCBI | |
Kaelin WG Jr and Thompson CB: Q&A: Cancer: Clues from cell metabolism. Nature. 465:562–564. 2010. View Article : Google Scholar : PubMed/NCBI | |
Martínez-Reyes I and Chandel NS: Cancer metabolism: Looking forward. Nat Rev Cancer. 21:669–680. 2021. View Article : Google Scholar : PubMed/NCBI | |
Xia L, Oyang L, Lin J, Tan S, Han Y, Wu N, Yi P, Tang L, Pan Q, Rao S, et al: The cancer metabolic reprogramming and immune response. Mol Cancer. 20:282021. View Article : Google Scholar : PubMed/NCBI | |
Pan C, Li B and Simon MC: Moonlighting functions of metabolic enzymes and metabolites in cancer. Mol Cell. 81:3760–3774. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hsu PP and Sabatini DM: Cancer cell metabolism: Warburg and beyond. Cell. 134:703–707. 2008. View Article : Google Scholar : PubMed/NCBI | |
Fendt SM: 100 years of the Warburg effect: A cancer metabolism endeavor. Cell. 187:3824–3828. 2024. View Article : Google Scholar : PubMed/NCBI | |
Paul S, Ghosh S and Kumar S: Tumor glycolysis, an essential sweet tooth of tumor cells. Semin Cancer Biol. 86(Pt 3): 1216–1230. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zheng X, Liu R, Zhou C, Yu H, Luo W, Zhu J, Liu J, Zhang Z, Xie N, Peng X, et al: ANGPTL4-mediated promotion of glycolysis facilitates the colonization of fusobacterium nucleatum in colorectal cancer. Cancer Res. 81:6157–6170. 2021. View Article : Google Scholar : PubMed/NCBI | |
Mizuno S, Seishima R, Yamasaki J, Hattori K, Ogiri M, Matsui S, Shigeta K, Okabayashi K, Nagano O, Li L and Kitagawa Y: Angiopoietin-like 4 promotes glucose metabolism by regulating glucose transporter expression in colorectal cancer. J Cancer Res Clin Oncol. 148:1351–1361. 2022. View Article : Google Scholar : PubMed/NCBI | |
Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI | |
Hanahan D: Hallmarks of cancer: New dimensions. Cancer Discov. 12:31–46. 2022. View Article : Google Scholar : PubMed/NCBI | |
Currie E, Schulze A, Zechner R, Walther TC and Farese RV Jr: Cellular fatty acid metabolism and cancer. Cell Metab. 18:153–161. 2013. View Article : Google Scholar : PubMed/NCBI | |
Jin Z, Chai YD and Hu S: Fatty acid metabolism and cancer. Adv Exp Med Biol. 1280:231–241. 2021. View Article : Google Scholar : PubMed/NCBI | |
Lee JY, Nam M, Son HY, Hyun K, Jang SY, Kim JW, Kim MW, Jung Y, Jang E, Yoon SJ, et al: Polyunsaturated fatty acid biosynthesis pathway determines ferroptosis sensitivity in gastric cancer. Proc Natl Acad Sci USA. 117:32433–32442. 2020. View Article : Google Scholar : PubMed/NCBI | |
Pham DV and Park PH: Adiponectin triggers breast cancer cell death via fatty acid metabolic reprogramming. J Exp Clin Cancer Res. 41:92022. View Article : Google Scholar : PubMed/NCBI | |
Dijk W and Kersten S: Regulation of lipoprotein lipase by Angptl4. Trends Endocrinol Metab. 25:146–155. 2014. View Article : Google Scholar : PubMed/NCBI | |
Georgiadi A, Lichtenstein L, Degenhardt T, Boekschoten MV, van Bilsen M, Desvergne B, Müller M and Kersten S: Induction of cardiac Angptl4 by dietary fatty acids is mediated by peroxisome proliferator-activated receptor beta/delta and protects against fatty acid-induced oxidative stress. Circ Res. 106:1712–1721. 2010. View Article : Google Scholar : PubMed/NCBI | |
Zhang R and Zhang K: An updated ANGPTL3-4-8 model as a mechanism of triglyceride partitioning between fat and oxidative tissues. Prog Lipid Res. 85:1011402022. View Article : Google Scholar : | |
Lichtenstein L, Mattijssen F, de Wit NJ, Georgiadi A, Hooiveld GJ, van der Meer R, He Y, Qi L, Köster A, Tamsma JT, et al: Angptl4 protects against severe proinflammatory effects of saturated fat by inhibiting fatty acid uptake into mesenteric lymph node macrophages. Cell Metab. 12:580–592. 2010. View Article : Google Scholar : PubMed/NCBI | |
Xiao S, Nai-Dong W, Jin-Xiang Y, Long T, Xiu-Rong L, Hong G, Jie-Cheng Y and Fei Z: ANGPTL4 regulate glutamine metabolism and fatty acid oxidation in nonsmall cell lung cancer cells. J Cell Mol Med. 26:1876–1885. 2022. View Article : Google Scholar : PubMed/NCBI | |
Blücher C, Iberl S, Schwagarus N, Müller S, Liebisch G, Höring M, Hidrobo MS, Ecker J, Spindler N, Dietrich A, et al: Secreted factors from adipose tissue reprogram tumor lipid metabolism and induce motility by modulating PPARα/ANGPTL4 and FAK. Mol Cancer Res. 18:1849–1862. 2020. View Article : Google Scholar | |
Cai Z, Li Y, Ma M, Wang L, Wang H, Liu M and Jiang C: Adipocytes promote pancreatic cancer migration and invasion through fatty acid metabolic reprogramming. Oncol Rep. 50:1412023. View Article : Google Scholar : PubMed/NCBI | |
Lieu EL, Nguyen T, Rhyne S and Kim J: Amino acids in cancer. Exp Mol Med. 52:15–30. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lippert TH, Ruoff HJ and Volm M: Intrinsic and acquired drug resistance in malignant tumors. The main reason for therapeutic failure. Arzneimittelforschung. 58:261–264. 2008.PubMed/NCBI | |
Olivares-Urbano MA, Griñán-Lisón C, Marchal JA and Núñez MI: CSC Radioresistance: A therapeutic challenge to improve radiotherapy effectiveness in cancer. Cells. 9:16512020. View Article : Google Scholar : PubMed/NCBI | |
Fekir K, Dubois-Pot-Schneider H, Désert R, Daniel Y, Glaise D, Rauch C, Morel F, Fromenty B, Musso O, Cabillic F and Corlu A: Retrodifferentiation of Human Tumor Hepatocytes to Stem Cells Leads to Metabolic Reprogramming and Chemoresistance. Cancer Res. 79:1869–1883. 2019. View Article : Google Scholar : PubMed/NCBI | |
Li C, Wang Q, Luo Y and Xiang J: TAZ regulates the cisplatin resistance of epithelial ovarian cancer cells via the ANGPTL4/SOX2 axis. Anal Cell Pathol (Amst). 2022:56321642022.PubMed/NCBI | |
Zhou S, Wang R and Xiao H: Adipocytes induce the resistance of ovarian cancer to carboplatin through ANGPTL4. Oncol Rep. 44:927–938. 2020. View Article : Google Scholar : PubMed/NCBI | |
Tsai YT, Wu AC, Yang WB, Kao TJ, Chuang JY, Chang WC and Hsu TI: ANGPTL4 induces TMZ resistance of glioblastoma by promoting cancer stemness enrichment via the EGFR/AKT/4E-BP1 cascade. Int J Mol Sci. 20:56252019. View Article : Google Scholar : PubMed/NCBI | |
Gordon ER, Wright CA, James M and Cooper SJ: Transcriptomic and functional analysis of ANGPTL4 overexpression in pancreatic cancer nominates targets that reverse chemoresistance. BMC Cancer. 23:5242023. View Article : Google Scholar : PubMed/NCBI | |
Li H, Ge C, Zhao F, Yan M, Hu C, Jia D, Tian H, Zhu M, Chen T, Jiang G, et al: Hypoxia-inducible factor 1 alpha-activated angiopoietin-like protein 4 contributes to tumor metastasis via vascular cell adhesion molecule-1/integrin β1 signaling in human hepatocellular carcinoma. Hepatology. 54:910–919. 2011. View Article : Google Scholar : PubMed/NCBI | |
Adhikary T, Brandt DT, Kaddatz K, Stockert J, Naruhn S, Meissner W, Finkernagel F, Obert J, Lieber S, Scharfe M, et al: Inverse PPARβ/δ agonists suppress oncogenic signaling to the ANGPTL4 gene and inhibit cancer cell invasion. Oncogene. 32:5241–5252. 2013. View Article : Google Scholar | |
Tian L, Zhou J, Casimiro MC, Liang B, Ojeifo JO, Wang M, Hyslop T, Wang C and Pestell RG: Activating peroxisome proliferator-activated receptor gamma mutant promotes tumor growth in vivo by enhancing angiogenesis. Cancer Res. 69:9236–9244. 2009. View Article : Google Scholar : PubMed/NCBI | |
Kim SH, Park YY, Kim SW, Lee JS, Wang D and DuBois RN: ANGPTL4 induction by prostaglandin E2 under hypoxic conditions promotes colorectal cancer progression. Cancer Res. 71:7010–7020. 2011. View Article : Google Scholar : PubMed/NCBI | |
Fresno Vara JA, Casado E, de Castro J, Cejas P, Belda-Iniesta C and González-Barón M: PI3K/Akt signalling pathway and cancer. Cancer Treat Rev. 30:193–204. 2004. View Article : Google Scholar : PubMed/NCBI | |
Pitt JM, Marabelle A, Eggermont A, Soria JC, Kroemer G and Zitvogel L: Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann Oncol. 27:1482–1492. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zhou S, Tu J, Ding S, Lu G, Lin Z, Ding Y, Deng B, Zhang Y, Xiao W and Gong W: High expression of angiopoietin-like protein 4 in advanced colorectal cancer and its association with regulatory T Cells and M2 macrophages. Pathol Oncol Res. 26:1269–1278. 2020. View Article : Google Scholar | |
Long F, Wang W, Li S, Wang B, Hu X, Wang J, Xu Y, Liu M, Zhou J, Si H, et al: The potential crosstalk between tumor and plasma cells and its association with clinical outcome and immunotherapy response in bladder cancer. J Transl Med. 21:2982023. View Article : Google Scholar : PubMed/NCBI | |
Li F and Ding J: Sialylation is involved in cell fate decision during development, reprogramming and cancer progression. Protein Cell. 10:550–565. 2019. View Article : Google Scholar : | |
Ma X, Li M, Wang X, Qi G, Wei L and Zhang D: Sialylation in the gut: From mucosal protection to disease pathogenesis. Carbohydr Polym. 343:1224712024. View Article : Google Scholar : PubMed/NCBI | |
Calabrese V, Zirino F, Vienna FG, Siligato R, Cernaro V and Santoro D: Insight into the role of angiopoietin-like protein 4 in podocypopathies (Review). World Acad Sci J. 6:292024. View Article : Google Scholar | |
Chugh SS and Clement LC: 'Idiopathic' minimal change nephrotic syndrome: A podocyte mystery nears the end. Am J Physiol Renal Physiol. 325:F685–F694. 2023. View Article : Google Scholar | |
Smith BAH, Deutzmann A, Correa KM, Delaveris CS, Dhanasekaran R, Dove CG, Sullivan DK, Wisnovsky S, Stark JC, Pluvinage JV, et al: MYC-driven synthesis of siglec ligands is a glycoimmune checkpoint. Proc Natl Acad Sci USA. 120:e22153761202023. View Article : Google Scholar : PubMed/NCBI | |
Stanczak MA and Läubli H: Siglec receptors as new immune checkpoints in cancer. Mol Aspects Med. 90:1011122023. View Article : Google Scholar | |
Zhao J, Liu J, Wu N, Zhang H, Zhang S, Li L and Wang M: ANGPTL4 overexpression is associated with progression and poor prognosis in breast cancer. Oncol Lett. 20:2499–2505. 2020. View Article : Google Scholar : PubMed/NCBI | |
Nie D, Zheng Q, Liu L, Mao X and Li Z: Up-regulated of angiopoietin-like protein 4 predicts poor prognosis in cervical cancer. J Cancer. 10:1896–1901. 2019. View Article : Google Scholar : PubMed/NCBI | |
Aung TM, Ciin MN, Silsirivanit A, Jusakul A, Lert-Itthiporn W, Proungvitaya T, Roytrakul S and Proungvitaya S: Serum angiopoietin-like protein 4: A potential prognostic biomarker for prediction of vascular invasion and lymph node metastasis in cholangiocarcinoma patients. Front Public Health. 10:8369852022. View Article : Google Scholar : PubMed/NCBI | |
Wang FT, Li XP, Pan MS, Hassan M, Sun W and Fan YZ: Identification of the prognostic value of elevated ANGPTL4 expression in gallbladder cancer-associated fibroblasts. Cancer Med. 10:6035–6047. 2021. View Article : Google Scholar : PubMed/NCBI | |
Shibata K, Nakayama T, Hirakawa H, Hidaka S and Nagayasu T: Clinicopathological significance of angiopoietin-like protein 4 expression in oesophageal squamous cell carcinoma. J Clin Pathol. 63:1054–1058. 2010. View Article : Google Scholar : PubMed/NCBI | |
Tanaka T, Imamura T, Irie A, Yoneda M, Imamura R, Kikuchi K, Kitagawa S, Kubo T, Ogi H and Nakayama H: Association of high cellular expression and plasma concentration of angiopoietin-like 4 with tongue cancer lung metastasis and poor prognosis. Oncol Lett. 24:2992022. View Article : Google Scholar : PubMed/NCBI | |
Dong D, Jia L, Zhou Y, Ren L, Li J and Zhang J: Serum level of ANGPTL4 as a potential biomarker in renal cell carcinoma. Urol Oncol. 35:279–285. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kubo H, Kitajima Y, Kai K, Nakamura J, Miyake S, Yanagihara K, Morito K, Tanaka T, Shida M and Noshiro H: Regulation and clinical significance of the hypoxia-induced expression of ANGPTL4 in gastric cancer. Oncol Lett. 11:1026–1034. 2016. View Article : Google Scholar : PubMed/NCBI | |
Sukonina V, Lookene A, Olivecrona T and Olivecrona G: Angiopoietin-like protein 4 converts lipoprotein lipase to inactive monomers and modulates lipase activity in adipose tissue. Proc Natl Acad Sci USA. 103:17450–17455. 2006. View Article : Google Scholar : PubMed/NCBI | |
Mysling S, Kristensen KK, Larsson M, Kovrov O, Bensadouen A, Jørgensen TJ, Olivecrona G, Young SG and Ploug M: The angiopoietin-like protein ANGPTL4 catalyzes unfolding of the hydrolase domain in lipoprotein lipase and the endothelial membrane protein GPIHBP1 counteracts this unfolding. Elife. 5:e209582016. View Article : Google Scholar : PubMed/NCBI | |
Yoshida K, Shimizugawa T, Ono M and Furukawa H: Angiopoietin-like protein 4 is a potent hyperlipidemia-inducing factor in mice and inhibitor of lipoprotein lipase. J Lipid Res. 43:1770–1772. 2002. View Article : Google Scholar : PubMed/NCBI | |
Dijk W, Beigneux AP, Larsson M, Bensadoun A, Young SG and Kersten S: Angiopoietin-like 4 promotes intracellular degradation of lipoprotein lipase in adipocytes. J Lipid Res. 57:1670–1683. 2016. View Article : Google Scholar : PubMed/NCBI | |
Reimund M, Kovrov O, Olivecrona G and Lookene A: Lipoprotein lipase activity and interactions studied in human plasma by isothermal titration calorimetry. J Lipid Res. 58:279–288. 2017. View Article : Google Scholar : | |
Kersten S: New insights into angiopoietin-like proteins in lipid metabolism and cardiovascular disease risk. Curr Opin Lipidol. 30:205–211. 2019. View Article : Google Scholar : PubMed/NCBI | |
Huang RL, Teo Z, Chong HC, Zhu P, Tan MJ, Tan CK, Lam CR, Sng MK, Leong DT, Tan SM, et al: ANGPTL4 modulates vascular junction integrity by integrin signaling and disruption of intercellular VE-cadherin and claudin-5 clusters. Blood. 118:3990–4002. 2011. View Article : Google Scholar : PubMed/NCBI | |
Cazes A, Galaup A, Chomel C, Bignon M, Bréchot N, Le Jan S, Weber H, Corvol P, Muller L, Germain S and Monnot C: Extracellular matrix-bound angiopoietin-like 4 inhibits endothelial cell adhesion, migration, and sprouting and alters actin cytoskeleton. Circ Res. 99:1207–1215. 2006. View Article : Google Scholar : PubMed/NCBI | |
Sheridan C: Amgen's angiopoietin blocker fails in ovarian cancer. Nat Biotechnol. 33:5–6. 2015. View Article : Google Scholar : PubMed/NCBI | |
Padua D, Zhang XH, Wang Q, Nadal C, Gerald WL, Gomis RR and Massagué J: TGFbeta primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell. 133:66–77. 2008. View Article : Google Scholar : PubMed/NCBI | |
Simeon J, Thrush J and Bailey TA: Angiopoietin-like protein 4 is a chromatin-bound protein that enhances mammosphere formation in vitro and experimental triple-negative breast cancer brain and liver metastases in vivo. J Carcinog. 20:82021. View Article : Google Scholar : PubMed/NCBI | |
Nakayama T, Hirakawa H, Shibata K, Nazneen A, Abe K, Nagayasu T and Taguchi T: Expression of angiopoietin-like 4 (ANGPTL4) in human colorectal cancer: ANGPTL4 promotes venous invasion and distant metastasis. Oncol Rep. 25:929–935. 2011. View Article : Google Scholar : PubMed/NCBI | |
Yi J, Pan BZ, Xiong L and Song HZ: Clinical significance of angiopoietin-like protein 4 expression in tissue and serum of esophageal squamous cell carcinoma patients. Med Oncol. 30:6802013. View Article : Google Scholar : PubMed/NCBI | |
Nakayama T, Hirakawa H, Shibata K, Abe K, Nagayasu T and Taguchi T: Expression of angiopoietin-like 4 in human gastric cancer: ANGPTL4 promotes venous invasion. Oncol Rep. 24:599–606. 2010. View Article : Google Scholar : PubMed/NCBI | |
Ma T, Jham BC, Hu J, Friedman ER, Basile JR, Molinolo A, Sodhi A and Montaner S: Viral G protein-coupled receptor up-regulates Angiopoietin-like 4 promoting angiogenesis and vascular permeability in Kaposi's sarcoma. Proc Natl Acad Sci USA. 107:14363–14368. 2010. View Article : Google Scholar : PubMed/NCBI | |
Yang W, Khoury E, Guo Q, Prabhu SA, Emond A, Huang F, Gonçalves C, Zhan Y, Plourde D, Nichol JN, et al: MNK1 signaling induces an ANGPTL4-mediated gene signature to drive melanoma progression. Oncogene. 39:3650–3665. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zhu X, Guo X, Wu S and Wei L: ANGPTL4 correlates with NSCLC progression and regulates epithelial-mesenchymal transition via ERK pathway. Lung. 194:637–646. 2016. View Article : Google Scholar : PubMed/NCBI | |
Lou H, Lin X, Wei G, Wu Z and Xiao Y: Construction of an Anoikis-related gene prognostic signature and identification of ANGPTL4 as a key oncogene in lung adenocarcinoma. Mol Biotechnol. 66:1290–1302. 2024. View Article : Google Scholar : PubMed/NCBI | |
Al-Kadash A, Alshaer W, Mahmoud IS, Wehaibi S and Zihlif M: Enhancing chemosensitivity of PANC1 pancreatic cancer cells to gemcitabine using ANGTPL4, Notch1 and NF-κβ1 siRNAs. Future Sci OA. 10:FSO9182024. View Article : Google Scholar | |
Hata S, Nomura T, Iwasaki K, Sato R, Yamasaki M, Sato F and Mimata H: Hypoxia-induced angiopoietin-like protein 4 as a clinical biomarker and treatment target for human prostate cancer. Oncol Rep. 38:120–128. 2017. View Article : Google Scholar : PubMed/NCBI | |
Tanaka T, Imamura T, Yoneda M, Irie A, Ogi H, Nagata M, Yoshida R, Fukuma D, Kawahara K, Shinohara M and Nakayama H: Enhancement of active MMP release and invasive activity of lymph node metastatic tongue cancer cells by elevated signaling via the TNF-α-TNFR1-NF-κB pathway and a possible involvement of angiopoietin-like 4 in lung metastasis. Int J Oncol. 49:1377–1384. 2016. View Article : Google Scholar : PubMed/NCBI | |
Yu Y, Zhang M, Liu J, Xu B, Yang J, Wang N, Yan S, Wang F, He X, Ji G, et al: Long Non-coding RNA PVT1 promotes cell proliferation and migration by silencing ANGPTL4 expression in cholangiocarcinoma. Mol Ther Nucleic Acids. 13:503–513. 2018. View Article : Google Scholar : PubMed/NCBI |