Annexin A1 affects tumor metastasis through epithelial-mesenchymal transition: a narrative review
Review Article

Annexin A1 affects tumor metastasis through epithelial-mesenchymal transition: a narrative review

Lulu Zheng1, Lanxin Li1, Baiqi Wang2, Shanshan Zhang1, Zhuqiong Fu1, Ailan Cheng1, Xiaoqiu Liang1

1Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China; 2The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China

Contributions: (I) Conception and design: L Zheng, X Liang, A Cheng; (II) Administrative support: X Liang, A Cheng; (III) Provision of study materials or patients: B Wang, S Zhang, Z Fu; (IV) Collection and assembly of data: L Zheng, L Li; (V) Data analysis and interpretation: L Zheng, X Liang, A Cheng; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Prof. Xiaoqiu Liang. Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang 421001, China. Email: 1996001889@usc.edu.cn; Prof. Ailan Cheng. Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang 421001, China. Email: 375355191@qq.com.

Background and Objective: Annexin A1 (annexin I, ANXA1), the first discovered member of the annexin superfamily, plays important roles in tumor development, invasion, metastasis, apoptosis and drug resistance based on tumor type-specific patterns of expression. The acquisition of the epithelial-mesenchymal transition (EMT) characteristics is an essential mechanism of metastasis because they increase the mobility and invasiveness of cancer cells. Cancer invasion and metastasis remain major health problems worldwide. Elucidating the role and mechanism of ANXA1 in the occurrence of EMT will help advance the development of novel therapeutic strategies. Hence, this review aims to attract everyone’s attention to the important role of ANXA1 in tumors and provide new ideas for clinical tumor treatment.

Methods: The PubMed database was mainly used to search for various English research papers and reviews related to the role of ANXA1 in tumors and EMT published from November 1994 to April 2022. The search terms used mainly include ANXA1, EMT, tumor, cancer, carcinoma, and mechanism.

Key Content and Findings: This article mainly provides a summary of the roles of ANXA1 and EMT in tumor metastasis as well as the various mechanisms via which ANXA1 facilitates the occurrence of EMT, thereby affecting tumor metastasis. In addition, the expression of ANXA1 in different metastatic tumor cell lines and its roles in tumorigenesis and development are also elaborated. This article has found many tumorous therapeutic targets related to ANXA1 and EMT, further confirming that ANXA1 has a huge potential for the diagnosis, treatment and prognosis of certain cancers.

Conclusions: Both the abnormal expression of ANXA1 and the occurrence of EMT are closely related to the invasion and metastasis of tumors, and more interestingly, ANXA1 can impact EMT directly or indirectly by mediating signaling pathways and adhesion among cells. We need more studies to elucidate the effects of ANXA1 on tumor invasion, migration and metastasis through EMT in vitro and in vivo clearly, and ultimately in patients to identify more therapeutic targets.

Keywords: Annexin A1; epithelial-mesenchymal transition (EMT); tumor metastasis; molecular mechanism


Submitted May 31, 2022. Accepted for publication Nov 12, 2022.

doi: 10.21037/tcr-22-1544


Introduction

There have been many reviews on the role of Annexin A1 (annexin I, ANXA1) in different tumors and the occurrence, development and mechanism of EMT in tumors. ANXA1, a calcium-dependent phospholipid-binding protein, is widely expressed in various bodily tissues and cells and exerts wide-ranging biological effects on processes such as inflammatory regulation, cell signal transduction, cell proliferation, differentiation and apoptosis (1). Increasing evidence supports that the dysregulation of ANXA1 plays an important role in the occurrence of the epithelial-mesenchymal transition (EMT) and may affect the metastasis of various tumors, and the relationship between ANXA1 and EMT is thus worthy of further study.

However, there is no report that summarizes the relationship between ANXA1 and EMT and how ANXA1 affects tumor invasion and metastasis through EMT. Therefore, in this review, we not only describe the role of ANXA1 in the occurrence and development of certain cancers but also summarize the various mechanisms by which ANXA1 acts on EMT and affects tumor metastasis. ANXA1 has potential as a biomarker for the diagnosis, treatment and prognosis of certain cancers. We present the following article in accordance with the Narrative Review reporting checklist (available at https://tcr.amegroups.com/article/view/10.21037/tcr-22-1544/rc).


Methods

Information used to write this paper was collected from the sources listed in Table 1.

Table 1

The search strategy summary

Items Specification
Date of search May 2022
Databases and other sources searched PubMed
Search terms used See Table S1
Timeframe November 1994 to April 2022
Inclusion and exclusion criteria Inclusion criteria: English research papers and review
Selection process Lulu Zheng and Lanxin Li initially screened out the literature related to the theme, and then Xiaoqiu Liang and Ailan Cheng discussed the final literature to use

ANXA1

The Annexin family

Annexins are a superfamily of calcium-dependent phospholipid-binding proteins (2) that are involved in a wide range of biological processes, such as the formation of the cell membrane and cytoskeleton; the regulation of cell signal transduction and inflammation; cell proliferation; apoptosis; autophagy; and the regulation of extracellular matrix integrity (3,4). Structurally, annexins are hallmarked by a highly alpha-helical and tightly packed protein core domain, consisting of four similar repeats approximately 70 amino acids long (5), that is considered a Ca2+-regulated membrane-binding module. However, studies have shown that annexins A1, A2, A5 and B12 have significantly different Ca2+-dependent membrane-binding properties due to their ability to form Ca2+-dependent membrane-bound trimers (2,6). This feature may be associated with their second principal domains, which are responsible for the distinct localizations and specialized functions of the proteins through posttranslational modification and binding to other proteins (6). A recent study suggested that more than one thousand annexin superfamily proteins have been identified, mainly in only eukaryotic phyla and not in yeasts or prokaryotes (5). The human annexin superfamily comprises 13 members, the majority of which are frequently dysregulated in cancer (7).

The structure and function of ANXA1

ANXA1, a 37 kDa protein, was the first member of the annexin superfamily to be discovered (6,8) and has been widely studied only in the context of inflammation resolution at the outset. An increasing number of studies have reported ANXA1 deregulation in numerous cancers, and further in-depth studies have suggested that it regulates the occurrence and metastasis of cancer at different levels (9). ANXA1 consists of 346 amino acids and has two distinct structural regions: a single N-terminal domain, also known as the tail, and a C-terminal domain, called the core domain. The C-terminus has Ca2+ and membrane-binding sites that regulate the release and exposure of the N-terminus, similar to a separate conformational switch. Then, the exposed N-terminus allows ANXA1 to interact with different ligands, enabling the modification (e.g., phosphorylation, glycosylation, acetylation, and lipidation) of many potential sites. This unique structure endows ANXA1 with a variety of biological functions (10). ANXA1, a Ca2+-dependent phospholipid-binding protein, plays a regulatory role in not only adhesion, apoptosis, cytoskeletal protein reorganization, angiogenesis, differentiation, inflammation, and immunity but also cancer cell proliferation, invasion and metastasis (3,9,11-13).

Expression and role of ANXA1 in tumors

Numerous studies have reported that ANXA1 expression is dysregulated in various common malignant tumors, including nonmetastatic gliomas (14-17) and metastatic tumors. Among the metastatic tumors, ANXA1 is mainly upregulated in pancreatic cancer (PC) (18-21), multiple myeloma (MM) (22), nasopharyngeal carcinoma (NPC) (11,12), oral squamous cell carcinoma (OSCC) (23), papillary thyroid carcinoma (PTC) (24-26), lung cancer (27-30), and colorectal cancer (31) and downregulated in melanoma (32,33) and esophageal and bile duct cancers (34); moreover, its expression in gastric cancer (GC) (34,35) and breast cancer (BC) (36-39) remains controversial among researchers (Table 2). Tumor development and metastasis are closely associated with the differential expression of ANXA1 in normal and tumor tissue samples (8). Increasing evidence has indicated that ANXA1 dysregulation and subcellular localization are involved in the development, invasion, and metastasis of various cancers in tumor type-specific patterns (4).

Table 2

Expression and roles of ANXA1 in the various common malignant tumors

Tumor types Species Tissue types Cell lines Expression Role References
Metastatic tumors
   Breast cancer Human Adenocarcinoma MCF-7, T-47D, SK-Br-3, ZR-75-1/all TNBC cell lines −/+++ Promotes invasion, prognostic biomarkers (36-39)
   Gastric cancer Human mice Adenocarcinoma AGS, N87, MKN45 −/+ Inhibits cell growth, promotes invasiveness (34,35)
   Lung cancer Human Adenocarcinoma H1975, H1650, A549, H1299 +++ Decrease chemosensitivity to Osimertinib; promote the tumorigenesis, invasion and migration (27-30)
   Melanoma Human Connective tissue SK-MEL-5 Induces invasion and metastasis (32,33)
   Multiple myeloma Human Connective tissue NCI-H929, RPMI-8226 +++ Its knockdown enhances chemosensitivity (22)
   Nasopharyngeal carcinoma Human Squamous cell carcinoma 5-8F, 6-10B +++, ++ Promotes metastasis (11,12)
   Oral squamous cell carcinoma Human Squamous cell carcinoma Tca-8113 SCC-9 + Inhibits cell proliferation, invasion; reverses TGF-β1/EGF-induced EMT (23)
   Pancreatic carcinoma Human Adenocarcinoma MIA PaCa-2 +++ Maintains a malignant phenotype (18-21)
   Papillary thyroid carcinoma Human Papillary adenocarcinoma BCPAP +++ Expedites the growth and metastasis (24-26)
Non-metastatic tumor
   Glioma Human Connective tissue U87, U251, U118, A172 +++ Inhibits apoptosis; promotes invasion and migration, prognostic biomarkers (14-17)

−: Negative; + to +++: low to high. ANXA1, annexin A1, annexin I; TNBC, triple negative breast cancer; TGF-β1 transforming growth factor-β1; EMT, epithelial-mesenchymal transition.

The role of ANXA1 in tumor development

ANXA1 may play a role in the angiogenesis of tumors. In BC cells, ANXA1 can reportedly directly activate nuclear factor-κB (NF-κB) expression by regulating miR-26b and miR-562, thereby promoting tumor-induced endothelial cell tube formation (40). In addition, a recent study showed that Ac2-26, an N-terminal ANXA1 mimetic peptide, promotes the activation of endothelial cells and affects the blood vessel formation of PC cells in vitro. Most importantly, these phenomena are interrupted by the interaction between heparan sulfate and ANXA1 (20). ANXA1 may contribute to the growth of some cancers (24,28,41). One recent report indicates that ANXA1 may be regulated by MYC to promote PTC proliferation (26). One study also reported that ANXA1 knockdown directly suppressed the proliferation of non-small cell lung cancer (NSCLC) cells (28). Another study demonstrated that ANXA1 was a direct target gene of miR-196a and that its knockdown reversed the inhibitory effect of miR-196a on the proliferation of EC109 esophageal squamous cell carcinoma (ESCC) cells (41). In addition, ANXA1 has been reported to promote the proliferation of other cancer cells, such as PC (42), BC (43), gastrointestinal cancer (44), and glioma (16,17) cells. The mechanisms include directly binding to formyl peptide receptor (FPR) to stimulate mitogen-activated proliferation (42), affecting G1 phase cell cycle arrest, targeting erythropoietin-producing hepatocellular receptor tyrosine kinase subtype A2 (EphA2) degradation (44), targeting the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway (16), and promoting alternative macrophage polarization in the tumor microenvironment (43). However, ANXA1 may also exert antiproliferative effects, as studies have shown that it is related to the sustained activation of the mitogen-activated protein kinase/extracellular signal regulated protein kinase (MAPK/ERK) signaling pathway, resulting in the destruction of the actin cytoskeleton and the inhibition of cyclin D1, thereby reducing the proliferation of A549 lung cancer cells and RAW264.7 macrophages (45). ANXA1 also inhibits the proliferation of OSCC (23), human laryngeal squamous cell carcinoma (46), cervical cancer (47), BC (48), and GC (34) cells. Its antitumor effect is associated with the activation of FPRs and inhibition of miR-196a in a negative feedback loop through NF-kB and c-Myc (46,48).

The role of ANXA1 in tumor migration and invasion

ANXA1 promotes both the invasion and migration of most tumors, such as the pancreatic ductal adenocarcinoma (PDAC) (42), ESCC (41,49), and NSCLC (27,28), and it facilitates only the invasion of BC (38,39,43) and GC (35) cells. In addition, ANXA1 inhibits the invasion of OSCC cells in vitro (23) and silencing ANXA1 reduces the migration, invasion, and proliferation of glioma cells (14).

The mechanisms by which ANXA1 regulates tumor invasion and migration are diverse. One recent study showed that ANXA1 was targeted and regulated by miR-196a to promote the invasion and migration of ESCC cells (41,49). The ANXA1-NF-κB-miR26a regulatory pathway also plays a role in the invasion and migration of NSCLC (27). ANXA1 can also increase the migration and invasion of PDAC cells by upregulating the expression and activity of matrix metalloproteinase-9 (MMP-9) (42). Additionally, the presence of secreted forms of the ANXA1 protein is thought to be responsible for its differential invasive behavior in various PC cells, as the secreted forms may be able to induce the migration and invasion of PC in vitro (19). ANXA1 can promote migration by activating EGFR signaling in bladder cancer and is a reliable clinical predictor for the prognosis of bladder cancer (50).

The role of ANXA1 in tumor metastasis

Metastasis is an unsalvageable fatal stage of malignant tumors. Although multiple studies have shown that changes in ANXA1 protein expression are closely related to the metastasis of various tumors, the results of these studies are contradictory. High ANXA1 expression is positively correlated with tumor metastasis, which is mainly manifested in PC (18,19), melanoma (32), NPC (11,12), PTC (25), and positive BC (51-53). However, the downregulation of ANXA1 has been confirmed to be associated with the distant metastasis of NSCLC (54) as well as the metastatic mouse and human BC cell lines (37,55,56). At present, the expression and functional roles of ANXA1 in BC remain controversial (53,57).

In different locations, ANXA1 affects tumor metastasis in different forms by many mechanisms in cancer cells. Extracellular ANXA1 can activate FPRs to affect metastasis by favoring cell migration and invasion, which is consistent with the finding that Ac2-26 significantly increases the cell migration/invasion rate by inducing the release of intracellular calcium (19). In addition, intracellularly, the ANXA1 protein can regulate metastasis by preserving cytoskeletal integrity and maintaining a malignant phenotype in vitro, which is independent of the FPR pathway (18). Another study reported that ANXA1 may increase metastatic potential via the constitutive activation of NF-κB, caused by its interaction with IKKγ, which can recruit RIP1 to the IκB kinase complex (58).

ANXA1 can also decrease its own expression through various mechanisms, such as allelic loss (59), gene methylation (60), p53 mutation causing reduced promoter activity (61), and mRNA degradation acceleration by endogenous microRNAs (miRNAs) (62,63), which affects tumor metastasis.

Clinical application of ANXA1

ANXA1 was originally identified as a regulator of inflammation and immunity. In recent years, its clinical application in the oncology field has gradually been explored, and it has been shown to play roles in tumor prevention, diagnosis, therapy, therapeutic efficacy evaluation and prognosis evaluation. The ANXA1 molecule is pluripotent and of great interest due to its strong potential clinical implications. However, ANXA1 cannot be classified simply as a tumor suppressor or promoter, as its expression levels and effects in the same tumors remain controversial. These differential effects are the main factor limiting its clinical application.

The role of ANXA1 in tumor apoptosis

Increasing evidence suggests that apoptosis, a form of programmed cell death, plays an important role in regulating tumor growth and responses to various cancer treatments, including radiotherapy and chemotherapy, and the regulation of apoptosis may be an effective way to improve the efficacy of tumor treatments (64). In recent decades, promoting the effective elimination of cancer cells by apoptosis has been a mainstay and goal of clinical cancer therapies (65). ANXA1 is closely related to tumor apoptosis, and the elucidation of this relationship contributes to the design and development of therapeutic strategies based on rational molecular approaches that aim to modulate apoptotic pathways.

ANXA1 is involved in the apoptosis of cancer cells induced by many chemicals. ANXA1 promotes the apoptosis of not only lung epithelial cells infected with influenza type A viruses and myelomonocytic lineage cells but also tumor cells through the intrinsic apoptotic pathway (66-69). ANXA1 has differential effects on the apoptosis of cells treated with different doses of FR235222, a novel histone deacetylase inhibitor in human promyelocytic leukemia U937 cells, human chronic myelogenous leukemia K562 cells and human T-cell leukemia Jurkat cells (70). However, in prostate cancer cells, the inhibition of ANXA1 expression is related to reduced levels of apoptosis induced by FR235222 regardless of the dose (71). In addition, a recent study showed that the modulation of ANXA1 expression is correlated with the punicalagin-induced apoptosis of colorectal cancer cells (72). In various cancers, the effects of ANXA1 expression on apoptosis also differ. The overexpression of ANXA1 in human histiocytic lymphoma cells and bronchoalveolar epithelial cells is reported to promote apoptosis, which is associated with the activation of Caspase-3 and transient intracellular calcium influx (66,68,73). However, in other cancers, antagonizing ANXA1 expression promotes cell apoptosis (14,16,30,74).

The role of ANXA1 in tumor drug resistance

Chemotherapy is an important cancer treatment modality, and its effectiveness is challenging (75). Inherent or adventitious drug resistance to chemotherapy can result in poor treatment outcomes and tumor relapse, a major cause of treatment failure (76). The expression of ANXA1 plays a key role in chemotherapy resistance, and regulating ANXA1 expression represents a possible strategy for overcoming resistance. ANXA1 can potentially predict resistance to antitumor drugs, including trastuzumab, gemcitabine, 5-fluorouracil, cisplatin, docetaxel, and doxorubicin, and serves as a novel target for improving the chemosensitivity of cancers, such as oral cancer, MM, NSCLC, esophageal carcinoma, GC, PC, renal cell carcinoma and colorectal cancer.

Currently, chemotherapy plus trastuzumab remains a standard therapeutic strategy for patients with human epidermal growth factor receptor 2-BC (77). Trastuzumab is remarkably effective, and reducing its resistance can benefit more patients. Concerning trastuzumab resistance, the results of the Fin-her phase III randomized trial indicated that the ANXA1 metagene is a novel predictive biomarker of resistance to adjuvant trastuzumab (78). However, the latest research shows that ANXA1 protein expression is not predictive of trastuzumab resistance but is associated with BC mortality and recurrence (52). Gemcitabine, 5-fluorouracil, and doxorubicin are the main antitumoral agents used in clinical practice. In primary PC cell lines, ANXA1 expression is related to drug responses; for example, high levels are positively associated with sensitivity to gemcitabine and doxorubicin and negatively associated with sensitivity to 5-fluorouracil (79). In PDAC cells, the downregulation of ANXA1 promotes resistance to gemcitabine and 5-fluorouracil through the protein kinase C/c-Jun N-terminal kinase/P-glycoprotein (PKC/JNK/P-GP) signaling pathway (80). Nevertheless, in ANXA1 knockout (KO) MIA PaCa-2 PC cells, ANXA1 was not shown to be involved in the apoptosis process mediated by gemcitabine (18). Additionally, in oral cancer cells, reduced ANXA1 expression promotes chemosensitivity to cisplatin, docetaxel, and 5-fluorouracil chemotherapy by enhancing caspase-dependent apoptosis (74). Arsenic trioxide, which functions by inducing apoptosis, was originally used as a treatment for patients with acute promyelocytic leukemia. Arsenic trioxide was later found to have an inhibitory effect on neuroblastoma, head and neck, esophageal, gastric, and cervical cancer cells by disrupting the cell cycle and has thus become an antitumor agent. One study reported that the specific silencing of ANXA1 enhanced the sensitivity of cancer cells to a low concentration of arsenic trioxide by inhibiting MAPK/ERK activation (81). The knockdown of ANXA1 enhances chemosensitivity to bortezomib, one of the most frequently used drugs in the treatment of MM (22). In NSCLC, regardless of whether epidermal growth factor receptor mutations are present, ANXA1 is correlated with chemotherapeutic resistance to osimertinib and cisplatin (29,30). Stabilization of ANXA1 by RRM2 can activate the AKT pathway, thereby promoting resistance to sunitinib in renal cell carcinoma (82).

The role of ANXA1 in tumor-targeted therapy

In tumors, ANXA1 is a valuable novel target for research on therapeutic drugs targeting tumor growth, migration, and invasion (10). Its peptide mimetics have been widely researched for use in a variety of diseases, such as neuroinflammatory diseases (83), eye diseases (84,85), cardiovascular disease (86,87), and gastroenteritis (88), and have shown great potential in therapies targeted at certain tumors, such as NPC (11), gastric and colon cancer (44), and melanoma (32). NPC patients receiving chemoradiotherapy have an extremely poor quality of life and severe side effects such as bone suppression. A11, an ANXA1-derived peptide, occupies the binding sites of ANXA1 and EphA2, thereby blocking the binding of the two proteins, and significantly inhibiting the proliferation, invasion, and metastasis of NPC (11). This targeted therapy blocks signal transmission by targeting specific molecules related to tumor progression in and around NPC cells, thereby inhibiting the occurrence and development of NPC, can accurately identify and treat NPC cells with low toxicity and side effects and has broad prospects in the clinical treatment of NPC (89).

The role of ANXA1 in tumor prevention, diagnosis, and prognostic evaluation

Numerous studies have shown that ANXA1 not only regulates inflammation and immunity (90-92) but also has potential as a novel marker for the screening (93,94), diagnosis (95,96), treatment efficacy prediction (97) and prognostic prediction (15,98,99) of tumors. ANXA1 can enhance the function of Treg cells and reduce the survival rate of patients with BC (100). In addition, ANXA1 plays a role in cancer prevention, as monoubiquitinated nuclear ANXA1 was shown to inhibit chemical-induced mutagenesis potentially by preventing DNA damage-induced gene mutations, ultimately conferring cancer chemoprevention (101).


EMT

Classification of EMT

EMT, a reversible conversion process characterized by the loss of epithelial cell features and the gain of mesenchymal phenotypes, can confer stem cell characteristics, reduce apoptosis and senescence, and promote immunosuppression (102,103). EMT occurs in three distinct conditions and is thus divided into three types based on its mechanisms of induction and progression, which vary dramatically among conditions. The first type is related to implantation, embryogenesis, and organ development, which are driven by the evolutionary need to remodel and diversify tissue to enable proper morphogenesis and generate a functional organism. The second type is associated with tissue regeneration and fibrosis, which depends on inflammation-inducing injuries for its initiation and continued occurrence. In the case of injury, the tissue is repaired by generating activated mesenchymal cells, notably myofibroblasts that produce excessive amounts of the collagen-rich extracellular matrix. Type 3 EMT occurs in the context of tumor growth and cancer progression, which is facilitated by the genomic alterations acquired by cancer cells and this type generates cells with invasive properties that enable them to move into the bloodstream and spread systemically to other organs (104-107). Therefore, EMT not only participates in embryonic development, wound healing and fibrosis but also plays a key role in cancer metastasis (102,103). For transition and transition-primed tumor epithelial cells, the occurrence of type 3 EMT is an important mechanism for movement, invasion and metastasis.

Common biological characteristics of EMT

Normally, sharp changes in the expression of selected epithelial and mesenchymal markers are regarded as evidence of EMT. Among them, the most common markers of epithelial traits are E-cadherin, occludins, and cytokeratins, while N-cadherin and vimentin are more common mesenchymal markers. These cell molecular markers are associated with the epithelial and mesenchymal state, affecting the disassembly of epithelial cell–cell junctions and the dissolution of apical-basal cell polarity (102,105). Pathologists usually regard the detection of these EMT-related protein markers as highly specific indicators of high-grade malignant tumors (106). Interestingly, the clinical prognosis depends on the co-expression of epithelial and mesenchymal markers, rather than on the fully epithelial or mesenchymal phenotype (108).

EMT-inducing transcription factors (EMT-TFs)

Upon the expression of EMT-TFs induced by signals derived from the tumor-associated reactive stroma that act on carcinoma cells, EMT is activated in carcinoma cells. EMT-TFs can orchestrate the expression of various components of the EMT program, such as ZEB, SNAIL and TWIST, which inhibit the expression of molecular markers associated with the epithelial state and concomitantly activate the expression of molecular markers associated with the mesenchymal state. Many variations of the EMT program play roles in normal tissues and neoplastic growth depending on the diverse expression of EMT-TFs (104). The considerable variability and tissue specificity of the roles and functions of different EMT-TFs (109), which determine the complexity of the mechanism of EMT, need to be further studied.

The relationship between EMT and tumor metastasis

EMT, as a complex biological transformation process, is considered to be important for cancer invasion and metastasis (110-115). During EMT, epithelial cells lose their apical-basal polarity and acquire a more mesenchymal and motile phenotype followed by cell-to-cell contact disintegration (113). These mesenchymal phenotype cells have a stronger ability to infiltrate and metastasize than epithelial cells (104). Neoplastic cells are in an epithelial-like state in early-stage carcinomas, and more mesenchymal characteristics are gradually acquired as tumor progression proceeds (104). Increasing the mobility and invasiveness of cancer cells by acquiring EMT characteristics is an important mechanism of metastasis (18,112).

The latest research shows that EMT occurs through distinct intermediate states rather than via a binary process, and multiple tumor subpopulations are associated with different degrees of EMT in various cancers. The plasticity, invasiveness and metastatic potential of these subpopulations differ (116). In different stages of EMT (from epithelial to completely mesenchymal states, passing through intermediate hybrid states), the hybrid EMT state is the most correlated with increased metastatic potential (108) (Figure 1).

Figure 1 Different EMT transition states. The different EMT transition states represent multiple tumor subpopulations ranging from epithelial to completely mesenchymal states, passing through intermediate hybrid states. The hybrid EMT state is the most correlated with increased metastatic potential. EMT, epithelial-mesenchymal transition; MET, mesenchymal-epithelial transition.

ANXA1 acts on EMT through different mechanisms to affect tumor metastasis

During the process of tumor progression, EMT enables tumor cells to acquire a more aggressive and metastatic mesenchymal phenotype, thereby promoting tumor metastasis. However, ANXA1 can promote the occurrence of EMT in different forms through different pathways to affect tumor metastasis (Figure 2). In most cases, ANXA1 does not act on EMT directly but rather affects the expression of EMT-TFs after interacting with certain factors (such as FPRs and miRNAs) or activating cell signaling pathways through certain physiological processes (such as autophagy), thereby affecting the occurrence of EMT (Figure 3). We can regard these factor inhibitors or signaling pathway inhibitors as potential therapeutic targets affecting tumor metastasis since they can block the effect of ANXA1 on EMT, thereby further prolonging the lives of cancer patients (Table 3).

Figure 2 Relevant pathological biomarkers of EMT. MMP, matrix metalloproteinase; ANXA1, annexin A1, annexin I; EMT, epithelial-mesenchymal transition.
Figure 3 ANXA1 regulates the occurrence of EMT via different mechanisms to thereby affect tumor metastasis. ANXA1 regulates the expression of EMT-TFs or activates cell signaling pathways after interacting with a substance to regulate EMT via different mechanisms, thereby influencing tumor metastasis. ANXA1, annexin A1, annexin I; TGF-β, transforming growth factor-β; PI3K, phosphoinositide 3-kinase; IKK, the IkappaB kinase; NF-κB, nuclear factor-κB; MMP, matrix metalloproteinase; IL, interleukin; JAK2, Janus kinase 2; STAT3, signal transducers and activators of transcription 3; FPRs, formyl peptide receptors; MAPK, mitogen-activated protein kinase; EVs, extracellular vesicles; MiRNA, microRNA; TF, transcription factor.

Table 3

ANXA1 promotes EMT through different mechanisms

Mechanisms Inhibitor Ref
Signaling pathways
   The TGF-β signaling pathway (36,37,117)
   The PI3K/AKT/mTOR signaling pathway (12)
   The NF-κB signaling pathway (118,119)
   The IL-6/JAK2/STAT3 signaling pathway (25)
Interacts with FPRs CHIPS, CsA, ICT12035, CsH (32,51,120-122)
A component of tumor-derived EVs (123)
Accepts transcriptional regulation by miRNA196a (124)
Induced by butyrate (33)

ANXA1, annexin A1, annexin I; EMT, epithelial-mesenchymal transition; TGF-β, transforming growth factor-β; PI3K, phosphoinositide 3-kinase; mTOR, the mechanistic target of rapamycin; NF-κB, nuclear factor-κB; IL-6, interleukin 6; JAK2, Janus kinase 2; STAT3, signal transducers and activators of transcription 3; FPRs, formyl peptide receptors; CHIPS, chemotaxis inhibitory protein of S. aureus; CsA, cyclosporin A; CsH, cyclosporin H; EVs, extracellular vesicles.

ANXA1 promotes EMT through different signaling pathways

Several activated intracellular signaling events can induce EMT when epithelial cells encounter specific signals during normal development, wound healing and carcinoma progression (104,125). These pathways include the transforming growth factor-β (TGF-β), wnt signaling, Notch, PI3K/AKT, MAPK/ERK, p38 MAPK and c-Jun N-terminal kinase (JNK) pathways.

Multiple studies have shown that EMT activated by ANXA1 through the TGF-β signaling pathway plays a key role in the invasion and migration of BC. ANXA1 is similar to an EMT switch, which promotes the formation of basal BC cell metastasis by enhancing TGF-β/Smad signaling and actin reorganization (36,126). ANXA1 can also specifically bind directly and closely to the DC-STAMP domain containing 1-antisense 1 to alter its own expression, which promotes TGF-β-induced EMT processes (117). Paradoxically, another study revealed that ANXA1 could efficiently suppress TGF-β-independent EMT to inhibit the metastasis of BC (37). In addition, in BC cells, ANXA1 induces the constitutive activation of NF-κB by interacting with the IκB kinase complex. This phenomenon further significantly enhances the gene expression of MMP-9, which is closely related to EMT (118), to induce tumor invasion and migration (58,119). Due to the heterogeneity of BC, the effects of the relationship between ANXA1 and EMT on BC cell behavior remain controversial (57).

ANXA1 can also activate the PI3K/AKT pathway, a classic pathway of oncogenic transformation (127), to promote the growth of colorectal cancer and the invasion and metastasis of NPC (12,31). Specifically, as a membrane protein, ANXA1 is a component of the lysosomal membranes and is involved in the development of autophagosomes. It can activate the PI3K/AKT signaling pathway, leading to Beclin-1- and ATG5-dependent autophagy inhibition, and further inducing EMT-like alterations, thereby affecting the migration, invasion and metastasis of NPC (12).

In addition, the mechanisms underlying the Janus kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT3) pathway have been extensively studied, and most are known to play roles in the invasion and metastasis of tumors by affecting EMT processes, including the mechanism by which ANXA1 affects PTC (128,129). ANXA1 can regulate malignant PTC phenotypes by regulating the interleukin 6 (IL-6)/JAK2/STAT3 signaling network and thereby influencing the dynamic properties of EMT (25).

ANXA1 interacts with FPRs to promote EMT

FPRs, which are a seven-transmembrane, G protein-coupled receptors, consist of the three proteins: PPR1, FPR2 (formerly known as ALX or formyl peptide receptor like-1) and FPR3 (formerly known as formyl peptide receptor like-2) (120). By interacting with ANXA1, FPRs play an important role in tumor proliferation, invasion and metastasis (51,120,130,131) and activate components of the tumor microenvironment (132). Recently, ANXA1 was reported to polarize macrophages to an alternatively activated subtype (M2). These cells then bind to FPR2, triggering the G protein-coupled receptor-mediated signaling cascade to thereby modulate cytokine signaling and the tumor microenvironment and promote the growth and migration of BC cells (43). In addition, FPRs can be activated by the annexin 1 peptide, derived from the unique N-terminal domain of ANXA1 (133). The interaction between ANXA1 and FPRs can lead to the occurrence of EMT via different mechanisms. In melanoma, the activation of matrix metalloproteinase-2 (MMP-2), followed by EMT, occurs through the MAPK and STAT3 pathways after Ac2-26 interacts with FPRs (134). It is worth noting that MMP-2 can efficiently degrade collagen IV and laminin and thus help melanoma cells to cross the basement membrane, which is crucial for EMT (135). Another study showed that ANXA1 can regulate EMT in cE1 cells, a prostate cancer cell line from a mouse model, and that the ANXA1/FPR interaction confers a stem cell-like phenotype to cancer cells (136). Inhibiting the activation of FPR1 by FPR1 inhibitors, such as a chemotaxis inhibitory protein of S. aureus (121), cyclosporin A (51), ICT12035 (120), and cyclosporin H (122), which blocks the effect of ANXA1 on EMT, is a potential strategy for reducing cell motility and tumor cell activation and inhibiting cell growth and invasion. These phenomena were confirmed by a study on the use of cyclosporin A to disrupt the activation of the ANXA1/FPR1 autocrine axis and thereby reduce MDA-MB-231 BC cell growth and aggressiveness in vitro and in vivo and might be a therapeutic target for triple-negative BC (51). Notably, some researchers have proposed that FPR2 agonists can inhibit the continuous inflammatory process to favor reparative and regenerative processes within the patients themselves (137). Given the subtle relationship between inflammation and tumors, more studies are required to clarify the effects of FPR2 agonists on tumors.

ANXA1, a component of tumor-derived extracellular vesicles (EVs), modulates the EMT-like phenotypic switch

EVs are a group of heterogeneous cell-derived membrane structures that contain various types of biomolecules, including proteins, RNA, DNA, lipids and metabolites, and participate in a variety of physiological and pathological processes (138,139). According to their forms in biological fluids, they are mainly divided into exosomes (40–200 nm), microvesicles (0.2–2 µm), apoptotic bodies (0.5–2 µm), high-density lipoprotein particles (7–13 nm), and low-density lipoprotein particles (21–27 nm) (140). Among them, exosomes have been a research hotspot in recent years and play important roles in physiological and pathological processes such as intercellular communication, immune surveillance, inflammation, and tumor development. In PC, the ANXA1 protein could be regarded as an oncogenic factor due to its overexpression and special identity as a component of tumor-derived EVs, which can nourish the tumor microenvironment (132). The detection of EVs isolated from wild-type and ANXA1 KO PC cells in an in vitro MIA PaCa-2 model verified that the autocrine effect of ANXA1-containing EVs on PC MIA PaCa-2 cells can induce the progression of PC. In addition, ANXA1 plays a key role in the effects of EVs (123). One recent study also shows that ANXA1 contained in EVs can regulate macrophage polarization in tumor microenvironment and promotes PC progression and metastasis (21). Notably, extracellular ANXA1 plays an important role in the acquisition of a more aggressive PC phenotype, similar to a modulator of the EMT-like phenotypic switch (132,141). ANXA1 KO cells recover their metastatic potential only when treated with wild-type EVs, as they undergo EMT and become significantly more mobile. Therefore, the ANXA1 protein in EVs can trigger EMT in ANXA1 KO MIA PaCa-2 cells, leading to a more aggressive phenotype (123). The acquisition of a mesenchymal phenotype is essential for the metastasis of PC (18), and ANXA1 may promote this aggressive phenotype, suggesting that the protein can serve as a PC diagnostic/prognostic marker. In addition, the suppression of EMT induces PC chemoresistance to antiproliferative drugs, such as gemcitabine, suggesting that targeting EMT will enhance chemotherapeutic efficacy (110). Additionally, one recent study has shown that thyroid cancer SW579 cell-derived exosomal ANXA1 can promote tumor development and the thyroid follicular epithelial Nthy-ori3-1 cells’ malignant transformation (142).

ANXA1 is transcriptionally regulated by miR-196a to promote EMT

MiRNAs are an abundant family of endogenous small noncoding RNAs (approximately 22 nucleotides) that play roles in biological processes such as cellular differentiation, proliferation, apoptosis, and synaptic plasticity by regulating target gene expression (143,144). MiRNAs significantly affect EMT and mesenchymal-epithelial transition processes by regulating key genes, such as zeb1, zeb2, snail, and twist, which are related to stem cell pluripotency and tumor progression (145). In the context of cancer, these miRNAs function as tumor suppressors or promoters. Some miRNAs, including miR-1271 (146), the miRNA-200 family (miR-200a, miR-200b, miR-200c, miR-141 and miR-429) and miR-205 (147), exert tumor suppressor functions by regulating EMT. For example, miR-1271 suppresses EMT by targeting zeb1 and twist1, thereby affecting the proliferation, invasion and migration of PC (146). MiR-200a suppresses EMT by targeting zeb1 and zeb2, delaying the growth of GC (148). However, other miRNAs play a tumor-promoting role, including miR-196 (124) , by affecting EMT. In one study, the aggressive phenotype of ANXA1 KO PC cells was restored by miR-196, a well-known oncogenic factor. MiR-196 not only triggers EMT but also mediates dynamics and other protein functions, promoting the migration and invasion of PC cells (124). Both ANXA1 and miR-196a participate in the induction of mesenchymal and more aggressive phenotypes jointly or individually (18,124,149). In addition, another study reported that in ESCC, miR-196a may have directly targeted ANXA1, which potentially further regulated the expression levels of COX2, MMP-2, snail and E-cadherin, thereby affecting the proliferation, invasion and migration of ESCC cells (41,49). MiR-196a can also suppress ANXA1 to thereby exert an oncogenic effect on head and neck cancer cells (62). The miR-196a/ANXA1 axis may represent a therapeutic target for ESCC, head and neck cancer, BC and endometrial cancer (41,62,150). In conclusion, various miRNAs are practical therapeutic targets, and they could be targeted to inhibit tumor invasion and metastasis, potentially representing a new therapeutic strategy.

ANXA1 promotes EMT after induction by butyrate

In melanoma, multiple signaling pathways promote EMT activation, including RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, and Wnt/β-catenin (125). One recent study proposed that the upregulation of ANXA1 expression induced by butyrate in a time- and dose-dependent manner regulates the expression of E-cadherin in human melanoma cells, thereby promoting their invasion via activation of the EMT signaling pathway (33). The loss of the cell–cell adhesion molecule E-cadherin can indeed lead to the uncontrolled growth and invasion of transformed melanocytes in the progression of melanoma (151). Inducing the expression of ANXA1 by butyrate, thereby blocking the occurrence of EMT, can restrict the invasion and metastasis of melanoma. This finding is consistent with the potential strategy of using synthetic and phytochemical agents to regulate the EMT signaling pathway and thereby reduce the aggressive progression of metastatic melanoma (125).


Conclusions

ANXA1 is differentially expressed in various tumors and performs numerous functions as either a suppressor or promoter of neoplastic development according to the cancer type. EMT is a highly dynamic process controlling the transdifferentiation of epithelial cells into motile mesenchymal cells (with stem cell-like properties and increased mobility and invasive ability). ANXA1 promotes the occurrence of EMT through different mechanisms. Among them, ANXA1 is known to effect EMT via distinct signaling pathways, but it remains unclear whether signaling crosstalk exists. While ANXA1 and FPR are known to interact in various tumors, which is a classical mechanism, the role of FPR inhibitors in tumors has not been fully elucidated. In addition, ANXA1 can be an EV component, be transcriptionally regulated by miR-196, and be induced by chemical drugs to promote the occurrence of EMT. ANXA1 promotes EMT by “external forces”, such as miR-196a and butyrate, suggesting its important role in tumor metastasis. However, these “external forces” that can affect the ANXA1 expression in tumor cells still need to be further studied and supplemented.

Since ANXA1 is potentially a new target in the study of therapeutic drugs that combat tumor growth, migration and invasion, elucidating the mechanisms by which it affects EMT will further identify more promising cancer therapeutic targets. Overall, cancer is a serious disease that imposes a heavy burden on the health of patients, and tumor metastasis remains the most challenging aspect of cancer treatment. Future studies should elucidate the effects of ANXA1 on tumor invasion, migration and metastasis through EMT in vitro, in vivo, and ultimately in patients to identify more therapeutic targets.


Acknowledgments

Funding: This work was supported by the National Natural Science Foundation of China (Grant No. 81072198 and No. 81372894), the Natural Science Foundation of Hunan Province (No. 2021JJ30589), and the Provincial Technological Innovation Guidance Project of Hunan Province (No. 2018SK51505).


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tcr.amegroups.com/article/view/10.21037/tcr-22-1544/rc

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-22-1544/coif). BW reports funding from the Provincial Technological Innovation Guidance Project of Hunan Province (No. 2018SK51505). AC reports funding from the National Natural Science Foundation of China (Grant No. 81072198 and No. 81372894), the Natural Science Foundation of Hunan Province (No. 2021JJ30589). The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Han PF, Che XD, Li HZ, et al. Annexin A1 involved in the regulation of inflammation and cell signaling pathways. Chin J Traumatol 2020;23:96-101. [Crossref] [PubMed]
  2. Patel DR, Isas JM, Ladokhin AS, et al. The conserved core domains of annexins A1, A2, A5, and B12 can be divided into two groups with different Ca2+-dependent membrane-binding properties. Biochemistry 2005;44:2833-44. [Crossref] [PubMed]
  3. Xi Y, Ju R, Wang Y. Roles of Annexin A protein family in autophagy regulation and therapy. Biomed Pharmacother 2020;130:110591. [Crossref] [PubMed]
  4. Guo C, Liu S, Sun MZ. Potential role of Anxa1 in cancer. Future Oncol 2013;9:1773-93. [Crossref] [PubMed]
  5. Moss SE, Morgan RO. The annexins. Genome Biol 2004;5:219. [Crossref] [PubMed]
  6. Gerke V, Moss SE. Annexins: from structure to function. Physiol Rev 2002;82:331-71. [Crossref] [PubMed]
  7. Zhuang C, Wang P, Sun T, et al. Expression levels and prognostic values of annexins in liver cancer. Oncol Lett 2019;18:6657-69. [Crossref] [PubMed]
  8. Foo SL, Yap G, Cui J, et al. Annexin-A1 - A Blessing or a Curse in Cancer? Trends Mol Med 2019;25:315-27. [Crossref] [PubMed]
  9. Boudhraa Z, Bouchon B, Viallard C, et al. Annexin A1 localization and its relevance to cancer. Clin Sci (Lond) 2016;130:205-20. [Crossref] [PubMed]
  10. Fu Z, Zhang S, Wang B, et al. Annexin A1: A double-edged sword as novel cancer biomarker. Clin Chim Acta 2020;504:36-42. [Crossref] [PubMed]
  11. Feng J, Lu SS, Xiao T, et al. ANXA1 Binds and Stabilizes EphA2 to Promote Nasopharyngeal Carcinoma Growth and Metastasis. Cancer Res 2020;80:4386-98. [Crossref] [PubMed]
  12. Zhu JF, Huang W, Yi HM, et al. Annexin A1-suppressed autophagy promotes nasopharyngeal carcinoma cell invasion and metastasis by PI3K/AKT signaling activation. Cell Death Dis 2018;9:1154. [Crossref] [PubMed]
  13. Sheikh MH, Solito E. Annexin A1: Uncovering the Many Talents of an Old Protein. Int J Mol Sci 2018; [Crossref] [PubMed]
  14. Lin Z, Wen M, Yu E, et al. ANXA1 as a Prognostic and Immune Microenvironmental Marker for Gliomas Based on Transcriptomic Analysis and Experimental Validation. Front Cell Dev Biol 2021;9:659080. [Crossref] [PubMed]
  15. Wang W, Li J, Lin F, et al. Expression and prognostic value of mRNAs in lower grade glioma with MGMT promoter methylated. J Clin Neurosci 2020;75:45-51. [Crossref] [PubMed]
  16. Wei L, Li L, Liu L, et al. Knockdown of Annexin-A1 Inhibits Growth, Migration and Invasion of Glioma Cells by Suppressing the PI3K/Akt Signaling Pathway. ASN Neuro 2021;13:17590914211001218. [Crossref] [PubMed]
  17. Zhu X, Shi G, Lu J, et al. Potential regulatory mechanism of TNF-α/TNFR1/ANXA1 in glioma cells and its role in glioma cell proliferation. Open Life Sci 2022;17:208-220. [Crossref] [PubMed]
  18. Belvedere R, Bizzarro V, Forte G, et al. Annexin A1 contributes to pancreatic cancer cell phenotype, behaviour and metastatic potential independently of Formyl Peptide Receptor pathway. Sci Rep 2016;6:29660. [Crossref] [PubMed]
  19. Belvedere R, Bizzarro V, Popolo A, et al. Role of intracellular and extracellular annexin A1 in migration and invasion of human pancreatic carcinoma cells. BMC Cancer 2014;14:961. [Crossref] [PubMed]
  20. Belvedere R, Novizio N, Pessolano E, et al. Heparan sulfate binds the extracellular Annexin A1 and blocks its effects on pancreatic cancer cells. Biochem Pharmacol 2020;182:114252. [Crossref] [PubMed]
  21. Novizio N, Belvedere R, Pessolano E, et al. ANXA1 Contained in EVs Regulates Macrophage Polarization in Tumor Microenvironment and Promotes Pancreatic Cancer Progression and Metastasis. Int J Mol Sci 2021; [Crossref] [PubMed]
  22. Jia C, Kong D, Guo Y, et al. Enhanced antitumor effect of combination of annexin A1 knockdown and bortezomib treatment in multiple myeloma in vitro and in vivo. Biochem Biophys Res Commun 2018;505:720-5. [Crossref] [PubMed]
  23. Wan YM, Tian J, Qi L, et al. ANXA1 affects cell proliferation, invasion and epithelial-mesenchymal transition of oral squamous cell carcinoma. Exp Ther Med 2017;14:5214-8. [Crossref] [PubMed]
  24. Petrella A, Festa M, Ercolino SF, et al. Annexin-1 downregulation in thyroid cancer correlates to the degree of tumor differentiation. Cancer Biol Ther 2006;5:643-7. [Crossref] [PubMed]
  25. Zhao X, Ma W, Li X, et al. ANXA1 enhances tumor proliferation and migration by regulating epithelial-mesenchymal transition and IL-6/JAK2/STAT3 pathway in papillary thyroid carcinoma. J Cancer 2021;12:1295-306. [Crossref] [PubMed]
  26. Ying X, Chen L, Xie J, et al. ANXA1 (Annexin A1) regulated by MYC (MYC proto-oncogene) promotes the growth of papillary thyroid carcinoma. Bioengineered 2021;12:9251-65. [Crossref] [PubMed]
  27. Guan X, Fang Y, Long J, et al. Annexin 1-nuclear factor-κB-microRNA-26a regulatory pathway in the metastasis of non-small cell lung cancer. Thorac Cancer 2019;10:665-75. [Crossref] [PubMed]
  28. Fang Y, Guan X, Cai T, et al. Knockdown of ANXA1 suppresses the biological behavior of human NSCLC cells in vitro. Mol Med Rep 2016;13:3858-66. [Crossref] [PubMed]
  29. Wang C, Xiao Q, Li YW, et al. Regulatory mechanisms of annexin-induced chemotherapy resistance in cisplatin resistant lung adenocarcinoma. Asian Pac J Cancer Prev 2014;15:3191-4. [Crossref] [PubMed]
  30. Chuang MC, Lung JH, Chen YC, et al. The Association of Annexin A1 and Chemosensitivity to Osimertinib in Lung Cancer Cells. Cancers (Basel) 2021.
  31. Hagihara T, Kondo J, Endo H, et al. Hydrodynamic stress stimulates growth of cell clusters via the ANXA1/PI3K/AKT axis in colorectal cancer. Sci Rep 2019;9:20027. [Crossref] [PubMed]
  32. Boudhraa Z, Rondepierre F, Ouchchane L, et al. Annexin A1 in primary tumors promotes melanoma dissemination. Clin Exp Metastasis 2014;31:749-60. [Crossref] [PubMed]
  33. Shin J, Song IS, Pak JH, et al. Upregulation of annexin A1 expression by butyrate in human melanoma cells induces invasion by inhibiting E-cadherin expression. Tumour Biol 2016;37:14577-84. [Crossref] [PubMed]
  34. Gao Y, Chen Y, Xu D, et al. Differential expression of ANXA1 in benign human gastrointestinal tissues and cancers. BMC Cancer 2014;14:520. [Crossref] [PubMed]
  35. Cheng TY, Wu MS, Lin JT, et al. Annexin A1 is associated with gastric cancer survival and promotes gastric cancer cell invasiveness through the formyl peptide receptor/extracellular signal-regulated kinase/integrin beta-1-binding protein 1 pathway. Cancer 2012;118:5757-67. [Crossref] [PubMed]
  36. de Graauw M, van Miltenburg MH, Schmidt MK, et al. Annexin A1 regulates TGF-beta signaling and promotes metastasis formation of basal-like breast cancer cells. Proc Natl Acad Sci U S A 2010;107:6340-5. [Crossref] [PubMed]
  37. Maschler S, Gebeshuber CA, Wiedemann EM, et al. Annexin A1 attenuates EMT and metastatic potential in breast cancer. EMBO Mol Med 2010;2:401-14. [Crossref] [PubMed]
  38. Bhardwaj A, Ganesan N, Tachibana K, et al. Annexin A1 Preferentially Predicts Poor Prognosis of Basal-Like Breast Cancer Patients by Activating mTOR-S6 Signaling. PLoS One 2015;10:e0127678. [Crossref] [PubMed]
  39. Okano M, Kumamoto K, Saito M, et al. Upregulated Annexin A1 promotes cellular invasion in triple-negative breast cancer. Oncol Rep 2015;33:1064-70. [Crossref] [PubMed]
  40. Anbalagan D, Yap G, Yuan Y, et al. Annexin-A1 regulates microRNA-26b* and microRNA-562 to directly target NF-κB and angiogenesis in breast cancer cells. PLoS One 2014;9:e114507. [Crossref] [PubMed]
  41. Hu C, Peng J, Lv L, et al. miR-196a regulates the proliferation, invasion and migration of esophageal squamous carcinoma cells by targeting ANXA1. Oncol Lett 2019;17:5201-9. [Crossref] [PubMed]
  42. Liu QH, Shi ML, Bai J, et al. Identification of ANXA1 as a lymphatic metastasis and poor prognostic factor in pancreatic ductal adenocarcinoma. Asian Pac J Cancer Prev 2015;16:2719-24. [Crossref] [PubMed]
  43. Moraes LA, Kar S, Foo SL, et al. Annexin-A1 enhances breast cancer growth and migration by promoting alternative macrophage polarization in the tumour microenvironment. Sci Rep 2017;7:17925. [Crossref] [PubMed]
  44. Feng J, Xiao T, Lu SS, et al. ANXA1-derived peptides suppress gastric and colon cancer cell growth by targeting EphA2 degradation. Int J Oncol 2020;57:1203-13. [PubMed]
  45. Alldridge LC, Bryant CE. Annexin 1 regulates cell proliferation by disruption of cell morphology and inhibition of cyclin D1 expression through sustained activation of the ERK1/2 MAPK signal. Exp Cell Res 2003;290:93-107. [Crossref] [PubMed]
  46. Gastardelo TS, Cunha BR, Raposo LS, et al. Inflammation and cancer: role of annexin A1 and FPR2/ALX in proliferation and metastasis in human laryngeal squamous cell carcinoma. PLoS One 2014;9:e111317. [Crossref] [PubMed]
  47. Prates J, Moreli JB, Gimenes AD, et al. Cisplatin treatment modulates Annexin A1 and inhibitor of differentiation to DNA 1 expression in cervical cancer cells. Biomed Pharmacother 2020;129:110331. [Crossref] [PubMed]
  48. Yuan Y, Anbalagan D, Lee LH, et al. ANXA1 inhibits miRNA-196a in a negative feedback loop through NF-kB and c-Myc to reduce breast cancer proliferation. Oncotarget 2016;7:27007-20. [Crossref] [PubMed]
  49. Han G, Lu K, Huang J, et al. Effect of Annexin A1 gene on the proliferation and invasion of esophageal squamous cell carcinoma cells and its regulatory mechanisms. Int J Mol Med 2017;39:357-63. [Crossref] [PubMed]
  50. Li P, Li L, Li Z, et al. Annexin A1 promotes the progression of bladder cancer via regulating EGFR signaling pathway. Cancer Cell Int 2022;22:7. [Crossref] [PubMed]
  51. Vecchi L, Alves Pereira Zóia M, Goss Santos T, et al. Inhibition of the AnxA1/FPR1 autocrine axis reduces MDA-MB-231 breast cancer cell growth and aggressiveness in vitro and in vivo. Biochim Biophys Acta Mol Cell Res 2018;1865:1368-82. [Crossref] [PubMed]
  52. Silva-Oliveira R, Pereira FF, Petronilho S, et al. Clinical Significance of ARID1A and ANXA1 in HER-2 Positive Breast Cancer. J Clin Med 2020;9:3911. [Crossref] [PubMed]
  53. Sobral-Leite M, Wesseling J, Smit VT, et al. Annexin A1 expression in a pooled breast cancer series: association with tumor subtypes and prognosis. BMC Med 2015;13:156. [Crossref] [PubMed]
  54. Deng C, Liu X, Zhang C, et al. ANXA1-GSK3β interaction and its involvement in NSCLC metastasis. Acta Biochim Biophys Sin (Shanghai) 2021;53:912-24. [Crossref] [PubMed]
  55. Yom CK, Han W, Kim SW, et al. Clinical significance of annexin A1 expression in breast cancer. J Breast Cancer 2011;14:262-8. [Crossref] [PubMed]
  56. Alli-Shaik A, Wee S, Lim LHK, et al. Phosphoproteomics reveals network rewiring to a pro-adhesion state in annexin-1-deficient mammary epithelial cells. Breast Cancer Res 2017;19:132. [Crossref] [PubMed]
  57. Tu Y, Johnstone CN, Stewart AG. Annexin A1 influences in breast cancer: Controversies on contributions to tumour, host and immunoediting processes. Pharmacol Res 2017;119:278-88. [Crossref] [PubMed]
  58. Bist P, Leow SC, Phua QH, et al. Annexin-1 interacts with NEMO and RIP1 to constitutively activate IKK complex and NF-κB: implication in breast cancer metastasis. Oncogene 2011;30:3174-85. [Crossref] [PubMed]
  59. Hu N, Flaig MJ, Su H, et al. Comprehensive characterization of annexin I alterations in esophageal squamous cell carcinoma. Clin Cancer Res 2004;10:6013-22. [Crossref] [PubMed]
  60. Vishwanatha JK, Salazar E, Gopalakrishnan VK. Absence of annexin I expression in B-cell non-Hodgkin's lymphomas and cell lines. BMC Cancer 2004;4:8. [Crossref] [PubMed]
  61. Lecona E, Barrasa JI, Olmo N, et al. Upregulation of annexin A1 expression by butyrate in human colon adenocarcinoma cells: role of p53, NF-Y, and p38 mitogen-activated protein kinase. Mol Cell Biol 2008;28:4665-74. [Crossref] [PubMed]
  62. Suh YE, Raulf N, Gäken J, et al. MicroRNA-196a promotes an oncogenic effect in head and neck cancer cells by suppressing annexin A1 and enhancing radioresistance. Int J Cancer 2015;137:1021-34. [Crossref] [PubMed]
  63. Chuang TD, Khorram O. Expression Profiling of lncRNAs, miRNAs, and mRNAs and Their Differential Expression in Leiomyoma Using Next-Generation RNA Sequencing. Reprod Sci 2018;25:246-55. [Crossref] [PubMed]
  64. Milas L, Stephens LC, Meyn RE. Relation of apoptosis to cancer therapy. In Vivo 1994;8:665-73. [PubMed]
  65. Carneiro BA, El-Deiry WS. Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol 2020;17:395-417. [Crossref] [PubMed]
  66. Debret R, El Btaouri H, Duca L, et al. Annexin A1 processing is associated with caspase-dependent apoptosis in BZR cells. FEBS Lett 2003;546:195-202. [Crossref] [PubMed]
  67. Canaider S, Solito E, de Coupade C, et al. Increased apoptosis in U937 cells over-expressing lipocortin 1 (annexin I). Life Sci 2000;66:PL265-70. [Crossref] [PubMed]
  68. Solito E, de Coupade C, Canaider S, et al. Transfection of annexin 1 in monocytic cells produces a high degree of spontaneous and stimulated apoptosis associated with caspase-3 activation. Br J Pharmacol 2001;133:217-28. [Crossref] [PubMed]
  69. Yap GLR, Sachaphibulkij K, Foo SL, et al. Annexin-A1 promotes RIG-I-dependent signaling and apoptosis via regulation of the IRF3-IFNAR-STAT1-IFIT1 pathway in A549 lung epithelial cells. Cell Death Dis 2020;11:463. [Crossref] [PubMed]
  70. Petrella A, D'Acunto CW, Rodriquez M, et al. Effects of FR235222, a novel HDAC inhibitor, in proliferation and apoptosis of human leukaemia cell lines: role of annexin A1. Eur J Cancer 2008;44:740-9. [Crossref] [PubMed]
  71. D'Acunto CW, Fontanella B, Rodriquez M, et al. Histone deacetylase inhibitor FR235222 sensitizes human prostate adenocarcinoma cells to apoptosis through up-regulation of Annexin A1. Cancer Lett 2010;295:85-91. [Crossref] [PubMed]
  72. Ganesan T, Sinniah A, Chik Z, et al. Punicalagin Regulates Apoptosis-Autophagy Switch via Modulation of Annexin A1 in Colorectal Cancer. Nutrients 2020; [Crossref] [PubMed]
  73. Solito E, Romero IA, Marullo S, et al. Annexin 1 binds to U937 monocytic cells and inhibits their adhesion to microvascular endothelium: involvement of the alpha 4 beta 1 integrin. J Immunol 2000;165:1573-81. [Crossref] [PubMed]
  74. Sun W, Zhao T, Aladelusi TO, et al. Decreased Annexin A1 expression enhances sensitivity to docetaxel, cisplatin and 5-fluorouracil combination induction chemotherapy in oral squamous cell carcinoma. J Oral Pathol Med 2021;50:795-802. [Crossref] [PubMed]
  75. Kachalaki S, Ebrahimi M, Mohamed Khosroshahi L, et al. Cancer chemoresistance; biochemical and molecular aspects: a brief overview. Eur J Pharm Sci 2016;89:20-30. [Crossref] [PubMed]
  76. Liu T, Xia R, Li C, et al. mRNA expression level of CDH2, LEP, POSTN, TIMP1 and VEGFC modulates 5-fluorouracil resistance in colon cancer cells. Exp Ther Med 2021;22:1023. [Crossref] [PubMed]
  77. Blaes AH, Dang C. Trastuzumab: Weighing the Benefits and the Risks. J Natl Cancer Inst 2020;112:1181-2. [Crossref] [PubMed]
  78. Sonnenblick A, Brohée S, Fumagalli D, et al. Integrative proteomic and gene expression analysis identify potential biomarkers for adjuvant trastuzumab resistance: analysis from the Fin-her phase III randomized trial. Oncotarget 2015;6:30306-16. [Crossref] [PubMed]
  79. Oshi M, Tokumaru Y, Mukhopadhyay S, et al. Annexin A1 Expression Is Associated with Epithelial-Mesenchymal Transition (EMT), Cell Proliferation, Prognosis, and Drug Response in Pancreatic Cancer. Cells 2021.
  80. Liu QH, Yong HM, Zhuang QX, et al. Reduced expression of annexin A1 promotes gemcitabine and 5-fluorouracil drug resistance of human pancreatic cancer. Invest New Drugs 2020;38:350-9. [Crossref] [PubMed]
  81. Zhang X, Li X, Li X, et al. ANXA1 silencing increases the sensitivity of cancer cells to low-concentration arsenic trioxide treatment by inhibiting ERK MAPK activation. Tumori 2015;101:360-7. [Crossref] [PubMed]
  82. Xiong W, Zhang B, Yu H, et al. RRM2 Regulates Sensitivity to Sunitinib and PD-1 Blockade in Renal Cancer by Stabilizing ANXA1 and Activating the AKT Pathway. Adv Sci (Weinh) 2021;8:e2100881. [Crossref] [PubMed]
  83. Solito E, McArthur S, Christian H, et al. Annexin A1 in the brain--undiscovered roles? Trends Pharmacol Sci 2008;29:135-42. [Crossref] [PubMed]
  84. Cardin LT, Sonehara NM, Mimura KK, et al. ANXA1(Ac2-26) peptide, a possible therapeutic approach in inflammatory ocular diseases. Gene 2017;614:26-36. [Crossref] [PubMed]
  85. Yu C, Chen H, Qi X, et al. Annexin A1 mimetic peptide Ac2-26 attenuates mechanical injury induced corneal scarring and inflammation. Biochem Biophys Res Commun 2019;519:396-401. [Crossref] [PubMed]
  86. Li X, Zheng L, Xia Q, et al. A novel cell-penetrating peptide protects against neuron apoptosis after cerebral ischemia by inhibiting the nuclear translocation of annexin A1. Cell Death Differ 2019;26:260-75. [Crossref] [PubMed]
  87. Zhang L, Zheng YL, Hu RH, et al. Annexin A1 Mimetic Peptide AC2-26 Inhibits Sepsis-induced Cardiomyocyte Apoptosis through LXA4/PI3K/AKT Signaling Pathway. Curr Med Sci 2018;38:997-1004. [Crossref] [PubMed]
  88. Reischl S, Lee JH, Miltschitzky JRE, et al. Ac2-26-Nanoparticles Induce Resolution of Intestinal Inflammation and Anastomotic Healing via Inhibition of NF-κB Signaling in a Model of Perioperative Colitis. Inflamm Bowel Dis 2021;27:1379-93. [Crossref] [PubMed]
  89. Kang Y, He W, Ren C, et al. Advances in targeted therapy mainly based on signal pathways for nasopharyngeal carcinoma. Signal Transduct Target Ther 2020;5:245. [Crossref] [PubMed]
  90. Perretti M, D'Acquisto F. Annexin A1 and glucocorticoids as effectors of the resolution of inflammation. Nat Rev Immunol 2009;9:62-70. [Crossref] [PubMed]
  91. Galvão I, de Carvalho RVH, Vago JP, et al. The role of annexin A1 in the modulation of the NLRP3 inflammasome. Immunology 2020;160:78-89. [Crossref] [PubMed]
  92. Williams SL, Milne IR, Bagley CJ, et al. A proinflammatory role for proteolytically cleaved annexin A1 in neutrophil transendothelial migration. J Immunol 2010;185:3057-63. [Crossref] [PubMed]
  93. Wu W, Wan C, Xie Q, et al. Expression and Clinical Significance of ANXA1 and DICER1 in Myelodysplastic Syndromes. J Coll Physicians Surg Pak 2020;30:1291-6. [Crossref] [PubMed]
  94. Chen S, Liu M, Liang B, et al. Identification of human peripheral blood monocyte gene markers for early screening of solid tumors. PLoS One 2020;15:e0230905. [Crossref] [PubMed]
  95. Adel FW, Rikhi A, Wan SH, et al. Annexin A1 is a Potential Novel Biomarker of Congestion in Acute Heart Failure. J Card Fail 2020;26:727-32. [Crossref] [PubMed]
  96. Ciregia F, Giusti L, Molinaro A, et al. Proteomic analysis of fine-needle aspiration in differential diagnosis of thyroid nodules. Transl Res 2016;176:81-94. [Crossref] [PubMed]
  97. Zeng GQ, Cheng AL, Tang J, et al. Annexin A1: a new biomarker for predicting nasopharyngeal carcinoma response to radiotherapy. Med Hypotheses 2013;81:68-70. [Crossref] [PubMed]
  98. Lin Y, Lin G, Fang W, et al. Increased expression of annexin A1 predicts poor prognosis in human hepatocellular carcinoma and enhances cell malignant phenotype. Med Oncol 2014;31:327. [Crossref] [PubMed]
  99. Liang Z, Li X. Identification of ANXA1 as a potential prognostic biomarker and correlating with immune infiltrates in colorectal cancer. Autoimmunity 2021;54:76-87. [Crossref] [PubMed]
  100. Bai F, Zhang P, Fu Y, et al. Targeting ANXA1 abrogates Treg-mediated immune suppression in triple-negative breast cancer. J Immunother Cancer 2020; [Crossref] [PubMed]
  101. Hirata F, Harada T, Corcoran GB, et al. Dietary flavonoids bind to mono-ubiquitinated annexin A1 in nuclei, and inhibit chemical induced mutagenesis. Mutat Res 2014;759:29-36. [Crossref] [PubMed]
  102. Nieto MA, Huang RY, Jackson RA, et al. EMT: 2016. Cell 2016;166:21-45. [Crossref] [PubMed]
  103. Dalla Pozza E, Forciniti S, Palmieri M, et al. Secreted molecules inducing epithelial-to-mesenchymal transition in cancer development. Semin Cell Dev Biol 2018;78:62-72. [Crossref] [PubMed]
  104. Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol 2019;20:69-84. [Crossref] [PubMed]
  105. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest 2009;119:1420-8. [Crossref] [PubMed]
  106. Kalluri R. EMT: when epithelial cells decide to become mesenchymal-like cells. J Clin Invest 2009;119:1417-9. [Crossref] [PubMed]
  107. Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 2014;15:178-96. [Crossref] [PubMed]
  108. Pastushenko I, Blanpain C. EMT Transition States during Tumor Progression and Metastasis. Trends Cell Biol 2019;29:212-26. [Crossref] [PubMed]
  109. Krebs AM, Mitschke J, Lasierra Losada M, et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat Cell Biol 2017;19:518-29. [Crossref] [PubMed]
  110. Zheng X, Carstens JL, Kim J, et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015;527:525-30. [Crossref] [PubMed]
  111. Satoh K, Hamada S, Shimosegawa T. Involvement of epithelial to mesenchymal transition in the development of pancreatic ductal adenocarcinoma. J Gastroenterol 2015;50:140-6. [Crossref] [PubMed]
  112. Puisieux A, Brabletz T, Caramel J. Oncogenic roles of EMT-inducing transcription factors. Nat Cell Biol 2014;16:488-94. [Crossref] [PubMed]
  113. Virtakoivu R, Mai A, Mattila E, et al. Vimentin-ERK Signaling Uncouples Slug Gene Regulatory Function. Cancer Res 2015;75:2349-62. [Crossref] [PubMed]
  114. Brabletz T, Kalluri R, Nieto MA, et al. EMT in cancer. Nat Rev Cancer 2018;18:128-34. [Crossref] [PubMed]
  115. Shibue T, Weinberg RA. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat Rev Clin Oncol 2017;14:611-29. [Crossref] [PubMed]
  116. Pastushenko I, Brisebarre A, Sifrim A, et al. Identification of the tumour transition states occurring during EMT. Nature 2018;556:463-8. [Crossref] [PubMed]
  117. Tang L, Chen Y, Chen H, et al. DCST1-AS1 Promotes TGF-β-Induced Epithelial-Mesenchymal Transition and Enhances Chemoresistance in Triple-Negative Breast Cancer Cells via ANXA1. Front Oncol 2020;10:280. [Crossref] [PubMed]
  118. Liu L, Ye Y, Zhu X. MMP-9 secreted by tumor associated macrophages promoted gastric cancer metastasis through a PI3K/AKT/Snail pathway. Biomed Pharmacother 2019;117:109096. [Crossref] [PubMed]
  119. Kang H, Ko J, Jang SW. The role of annexin A1 in expression of matrix metalloproteinase-9 and invasion of breast cancer cells. Biochem Biophys Res Commun 2012;423:188-94. [Crossref] [PubMed]
  120. Ahmet DS, Basheer HA, Salem A, et al. Application of small molecule FPR1 antagonists in the treatment of cancers. Sci Rep 2020;10:17249. [Crossref] [PubMed]
  121. Boer JC, Domanska UM, Timmer-Bosscha H, et al. Inhibition of formyl peptide receptor in high-grade astrocytoma by CHemotaxis Inhibitory Protein of S. aureus. Br J Cancer 2013;108:587-96. [Crossref] [PubMed]
  122. Snapkov I, Öqvist CO, Figenschau Y, et al. The role of formyl peptide receptor 1 (FPR1) in neuroblastoma tumorigenesis. BMC Cancer 2016;16:490. [Crossref] [PubMed]
  123. Pessolano E, Belvedere R, Bizzarro V, et al. Annexin A1 May Induce Pancreatic Cancer Progression as a Key Player of Extracellular Vesicles Effects as Evidenced in the In Vitro MIA PaCa-2 Model System. Int J Mol Sci 2018;19:3878. [Crossref] [PubMed]
  124. Belvedere R, Saggese P, Pessolano E, et al. miR-196a Is Able to Restore the Aggressive Phenotype of Annexin A1 Knock-Out in Pancreatic Cancer Cells by CRISPR/Cas9 Genome Editing. Int J Mol Sci 2018;19:1967. [Crossref] [PubMed]
  125. Pearlman RL, Montes de Oca MK, Pal HC, et al. Potential therapeutic targets of epithelial-mesenchymal transition in melanoma. Cancer Lett 2017;391:125-40. [Crossref] [PubMed]
  126. Okano M, Oshi M, Butash AL, et al. Triple-Negative Breast Cancer with High Levels of Annexin A1 Expression Is Associated with Mast Cell Infiltration, Inflammation, and Angiogenesis. Int J Mol Sci 2019; [Crossref] [PubMed]
  127. Aoki M, Fujishita T. Oncogenic Roles of the PI3K/AKT/mTOR Axis. Curr Top Microbiol Immunol 2017;407:153-89. [Crossref] [PubMed]
  128. Bi CL, Zhang YQ, Li B, et al. MicroRNA-520a-3p suppresses epithelial-mesenchymal transition, invasion, and migration of papillary thyroid carcinoma cells via the JAK1-mediated JAK/STAT signaling pathway. J Cell Physiol 2019;234:4054-67. [Crossref] [PubMed]
  129. Pan XM, He XY, Yang YL, et al. MiR-630 inhibits papillary thyroid carcinoma cell growth, metastasis, and epithelial-mesenchymal transition by suppressing JAK2/STAT3 signaling pathway. Eur Rev Med Pharmacol Sci 2019;23:2453-60. [PubMed]
  130. Khau T, Langenbach SY, Schuliga M, et al. Annexin-1 signals mitogen-stimulated breast tumor cell proliferation by activation of the formyl peptide receptors (FPRs) 1 and 2. FASEB J 2011;25:483-96. [Crossref] [PubMed]
  131. Foo SL, Sachaphibulkij K, Lee CLY, et al. Breast cancer metastasis to brain results in recruitment and activation of microglia through annexin-A1/formyl peptide receptor signaling. Breast Cancer Res 2022;24:25. [Crossref] [PubMed]
  132. Novizio N, Belvedere R, Pessolano E, et al. Annexin A1 Released in Extracellular Vesicles by Pancreatic Cancer Cells Activates Components of the Tumor Microenvironment, through Interaction with the Formyl-Peptide Receptors. Cells 2020; [Crossref] [PubMed]
  133. Ernst S, Lange C, Wilbers A, et al. An annexin 1 N-terminal peptide activates leukocytes by triggering different members of the formyl peptide receptor family. J Immunol 2004;172:7669-76. [Crossref] [PubMed]
  134. Boudhraa Z, Merle C, Mazzocut D, et al. Characterization of pro-invasive mechanisms and N-terminal cleavage of ANXA1 in melanoma. Arch Dermatol Res 2014;306:903-14. [Crossref] [PubMed]
  135. Kreiseder B, Orel L, Bujnow C, et al. α-Catulin downregulates E-cadherin and promotes melanoma progression and invasion. Int J Cancer 2013;132:521-30. [Crossref] [PubMed]
  136. Geary LA, Nash KA, Adisetiyo H, et al. CAF-secreted annexin A1 induces prostate cancer cells to gain stem cell-like features. Mol Cancer Res 2014;12:607-21. [Crossref] [PubMed]
  137. Perretti M, Godson C. Formyl peptide receptor type 2 agonists to kick-start resolution pharmacology. Br J Pharmacol 2020;177:4595-600. [Crossref] [PubMed]
  138. Teng F, Fussenegger M. Shedding Light on Extracellular Vesicle Biogenesis and Bioengineering. Adv Sci (Weinh) 2020;8:2003505. [Crossref] [PubMed]
  139. van Niel G, D'Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol 2018;19:213-28. [Crossref] [PubMed]
  140. Shao H, Im H, Castro CM, et al. New Technologies for Analysis of Extracellular Vesicles. Chem Rev 2018;118:1917-50. [Crossref] [PubMed]
  141. Bizzarro V, Belvedere R, Milone MR, et al. Annexin A1 is involved in the acquisition and maintenance of a stem cell-like/aggressive phenotype in prostate cancer cells with acquired resistance to zoledronic acid. Oncotarget 2015;6:25076-92. [Crossref] [PubMed]
  142. Li Q, Liu W, Wang Z, et al. Exosomal ANXA1 derived from thyroid cancer cells is associated with malignant transformation of human thyroid follicular epithelial cells by promoting cell proliferation. Int J Oncol 2021;59:104. [Crossref] [PubMed]
  143. Pu M, Chen J, Tao Z, et al. Regulatory network of miRNA on its target: coordination between transcriptional and post-transcriptional regulation of gene expression. Cell Mol Life Sci 2019;76:441-51. [Crossref] [PubMed]
  144. Carroll AP, Goodall GJ, Liu B. Understanding principles of miRNA target recognition and function through integrated biological and bioinformatics approaches. Wiley Interdiscip Rev RNA 2014;5:361-79. [Crossref] [PubMed]
  145. Lamouille S, Subramanyam D, Blelloch R, et al. Regulation of epithelial-mesenchymal and mesenchymal-epithelial transitions by microRNAs. Curr Opin Cell Biol 2013;25:200-7. [Crossref] [PubMed]
  146. Liu H, Wang H, Liu X, et al. miR-1271 inhibits migration, invasion and epithelial-mesenchymal transition by targeting ZEB1 and TWIST1 in pancreatic cancer cells. Biochem Biophys Res Commun 2016;472:346-52. [Crossref] [PubMed]
  147. Gregory PA, Bert AG, Paterson EL, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol 2008;10:593-601. [Crossref] [PubMed]
  148. Cong N, Du P, Zhang A, et al. Downregulated microRNA-200a promotes EMT and tumor growth through the wnt/β-catenin pathway by targeting the E-cadherin repressors ZEB1/ZEB2 in gastric adenocarcinoma. Oncol Rep 2013;29:1579-87. [Crossref] [PubMed]
  149. Szafranska AE, Davison TS, John J, et al. MicroRNA expression alterations are linked to tumorigenesis and non-neoplastic processes in pancreatic ductal adenocarcinoma. Oncogene 2007;26:4442-52. [Crossref] [PubMed]
  150. Luthra R, Singh RR, Luthra MG, et al. MicroRNA-196a targets annexin A1: a microRNA-mediated mechanism of annexin A1 downregulation in cancers. Oncogene 2008;27:6667-78. [Crossref] [PubMed]
  151. Fenouille N, Tichet M, Dufies M, et al. The epithelial-mesenchymal transition (EMT) regulatory factor SLUG (SNAI2) is a downstream target of SPARC and AKT in promoting melanoma cell invasion. PLoS One 2012;7:e40378. [Crossref] [PubMed]
Cite this article as: Zheng L, Li L, Wang B, Zhang S, Fu Z, Cheng A, Liang X. Annexin A1 affects tumor metastasis through epithelial-mesenchymal transition: a narrative review. Transl Cancer Res 2022;11(12):4416-4433. doi: 10.21037/tcr-22-1544

Download Citation