Association of annexin A2 with cancer development (Review)

  • Authors:
    • Xiao-Heng Xu
    • Wei Pan
    • Li-Hua Kang
    • Hui Feng
    • Yan-Qiu Song
  • View Affiliations

  • Published online on: March 5, 2015     https://doi.org/10.3892/or.2015.3837
  • Pages: 2121-2128
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Annexin A2 (ANXA2) is a well-known calcium-dependent phospholipid binding protein widely distributed in the nucleus, cytoplasm and extracellular surface of various eukaryotic cells. It has been recognized as a pleiotropic protein affecting a wide range of molecular and cellular processes. Dysregulation and abnormal expression of ANXA2 are linked to a large number of prevalent diseases, including autoimmune and neurodegenerative disease, antiphospholipid syndrome, inflammation, diabetes mellitus and a series of cancers. Accumulating data suggest that ANXA2 is aberrantly expressed in a wide spectrum of cancers, and exerts profound effects on tumor cell adhesion, proliferation, apoptosis, invasion and metastasis as well as tumor neovascularization via different modes of action. However, despite significant research, our knowledge of the mechanism by which ANXA2 participates in cancer development remains fragmented. The present review systematically summarizes the effects of ANXA2 on tumor progression, in an attempt to gain an improved understanding of the underlying mechanisms and to provide a potential effective target for cancer therapy.

1. Introduction

Cancer remains a major public health issue due to the limitations of current therapy. Despite improvements in surgical techniques, radiation therapy and chemotherapy, there has been no major improvement in the effective blockage of tumor progression. Tumor development is a complex stepwise process involving the accumulation of abnormalities in miscellaneous molecules that drive tumor growth and progression by coordinating critical interactions between tumor cells and the host microenvironment, including a variety of normal stromal cell types, the extracellular matrix (ECM), proteases and cytokines (1). Thus, an improvement in the prognosis of cancer will require the successful development of more effective molecular-targeted therapies. Annexin A2 (ANXA2) is one of the most important molecules that are aberrantly expressed in a wide range of cancers and participate in tumor cell adhesion, proliferation, invasion, metastasis and tumor neovascularization, thereby playing a crucial role in cancer growth and progression (24). In this review, we summarize the contribution of ANXA2 to cancer development and the underlying mechanisms, in an attempt to highlight the effects of ANXA2 on tumor cell adhesion, proliferation, invasion, metastasis and tumor neovascularization, and to provide a therapeutic target for molecular-based strategies.

2. Properties of annexin A2

ANXA2 (also called p36, calpactin I heavy chain and lipocortin II) is a 36-kDa protein belonging to the calcium-dependent phospholipid binding proteins (57). It is structurally highly conserved and is expressed in nearly all eukaryotes (710), where ANXA2 is distributed mainly in the plasma membrane (11) and cytoplasm with a small proportion in the nucleus (12). Similar to other annexins, the basic structure of ANXA2 consists of a homologous C-terminal core composed of four repeats, and a highly variable N-terminal tail (13,14). The C-terminus of ANXA2 harbors the binding sites of calcium, phospholipids and F-actin which are necessary for the membrane-associated activities of ANXA2 (1521). This domain also interacts directly with various molecules such as heparin and RNA, thus endowing ANXA2 with more regulating actions (2225). The N-terminal domain of ANXA2 contains a nuclear export signal (NES) (12) as well as multiple phosphorylation sites such as Tyr23, Ser11 and Ser25, which can be phosphorylated by Src kinase and protein kinase C, respectively (2628). The phosphorylation of ANXA2 affects its intracellular localization and regulating actions in specific cell types (2938). Moreover, ANXA2 has been identified as a cellular redox regulatory protein, and this action is related to a reactive cysteine residue (Cys-8) in the N-terminus. These domains dictate its functional and regulatory specificity distinct from other annexin members (39).

ANXA2 exists as a monomer or as a heterotetramer (AIIt). AIIt consists of 2 molecules of ANXA2 and a dimer of S100A10 (P11), and possesses a spectrum of biological behavior (4042). The most well-documented function of ANXA2 is the interaction with tissue plasminogen activator (tPA) as well as its substrate, plasminogen, and promotes the conversion of plasminogen into plasmin (10,4244) by which it regulates the fibrinolytic process (45), facilitates tissue remodeling, degrades ECM and participates in angiogenesis (4648) when it locates on the extracellular surface. The expression of ANXA2 in the cytoplasm and plasma membrane is involved in the regulation of actin cytoskeleton dynamics (6,19,21,49), endocytosis and exocytosis (31,5052), cell-cell adhesion (5,53,54), cell polarity (55) and endosome formation (56,57). ANXA2 has also been shown to play an important role in DNA synthesis and mRNA transport and translation after identification of a small population existing in the nucleus and a NES in the N-terminus of this protein that regulates its nuclear export (12,58). ANXA2 acting as a part of the primer recognition protein complex and as a DNA-binding protein regulates DNA polymerase α activity, DNA synthesis, cell proliferation and cell cycle progression (5966). Other studies have revealed that ANXA2 directly binds to ribonucleotide homopolymers, cytoskeleton-bound polysomes and is involved in association of mRNA with the cytoskeleton and perinuclear localization (25). The nuclear accumulation of the ANXA2 monomer plays a role in protecting the cells from DNA damage during oxidative stress (67). More recently, increasing data have shown that ANXA2 is implicated in a wide range of biological action such as facilitating the cell cycle partly through a p53-dependent mechanism (68), regulating signal transducer and activator of transcription 6 (STAT6) activity (69) and participating in multiple redox cycles (39). Post-translation modification of ANXA2 such as acetylation and phosphorylation regulates its localization, NES, as well as binding to S100A10 and plasminogen, which determines its biological activation (29,3135,37,38). Phosphorylation of ANXA2, at Tyr23 for example, was found to induce actin reorganization and cell scattering in MDCK cells (30); meanwhile, Tyr23 phosphorylation is required for cell-surface localization of ANXA2 involved in pancreatic ductal adenocarcinoma invasion (36). ANXA2 is aberrantly expressed in a wide spectrum of tumors, and this abnormal expression of ANXA2 plays a crucial role in tumor growth and progression.

3. Aberrant expression of annexin A2 in cancers

ANXA2 is one of the most common proteins that are overexpressed in a series of cancers and are implicated in the multistep processes of tumor development. The first association between ANXA2 and tumorigenesis was described in hepatocellular carcinoma (HCC) in 1990, in which an abundance of ANXA2 was detected (70). Recently, a number of studies have found the increased expression of ANXA2 at both the protein and mRNA levels in many types of malignancies such as colorectal (71,72), breast (7376) and lung cancer (77), HCC (78), gastric carcinoma (79) and pancreatic cancer (80), particularly in the more aggressive or poorer prognosis phenotype of these cancers (8184). As a secretory protein, the serum level of ANXA2 was also found to be elevated in patients with cancers, including HCC (78,85,86) and invasive breast cancer (87).

There is increasing evidence to suggest that overexpression of ANXA2 is closely associated with the differentiation status, histological type, lymph node metastasis and distant metastasis in non-small cell lung cancer (NSCLC) (68,88,89), colorectal (71,90,91) and gastric cancer (79,84). Statistical analysis has also shown that ANXA2 overexpression is correlated with a reduced survival time and a higher risk of recurrence in colorectal (71), pancreatic (80,92), gastric (79), clear-cell renal cell carcinoma (83) as well as NSCLC (88,89). These studies indicate the involvement of ANXA2 in tumor progression. In contrast, ANXA2 was also found to have an inverse correlation with esophageal carcinomas (93,94) as well as head and neck squamous cell carcinoma (95). In other words, the expression of ANXA2 was found to be significantly lower in tumor tissues compared to its paired adjacent normal tissues in these cancers, and the downregulation of ANXA2 was significantly correlated with advanced clinical stage, more frequent recurrence and regional lymph node and distant metastasis. The different experimental techniques and the difference between primary tumors and metastatic lesions may be responsible for these contradictory findings.

Taken together, these data indicate that aberrant expression of ANXA2 is an important prognostic factor in a number of tumor types, and exerts profound effects on tumor progression.

4. Annexin A2 and tumor cell adhesion

High ANXA2 expression in cancer cells and tumor stroma has been implicated in tumor cell adhesion. Initial evidence for the involvement of ANXA2 in tumor adhesion was discovered in RAW117 large cell lymphoma cells (96). This study demonstrated that the binding of RAW117 tumor cells to endothelial cells (ECs) was mediated by ANXA2 expressed on the surface of the RAW117 tumor cells, and this binding was inhibited by antibodies of ANXA2, indicating the association of ANXA2 with tumor cell adhesion. In the secretome of co-cultured cells, ANXA2 siRNA significantly inhibited ovarian cancer cell adhesion to peritoneal cells, supporting the role of ANXA2 in cell adhesion (97). Similarly, the ability of prostate cancer PC-3 cells to bind to human bone marrow ECs and osteoblasts was significantly blocked by an antibody to ANXA2 or the N-terminal competing peptide of this protein. The adhesive capacity of PC-3 cells to osteoblasts derived from Anxa2+/+ mice was significantly increased compared to those from Anxa2−/− mice, further supporting the involvement of ANXA2 in tumor adhesion (98). However, the underlying mechanism responsible for the actions of ANXA2 in tumor adhesion remains unclear. Recently studies have demonstrated that the adhesion between breast cancer cells and ECs is mediated by interactions between ANXA2 and S100A10. ANXA2 expressed on the surface of breast cancer cells interacts with S100A10 located on microvascular ECs, facilitating the process by which cancer cells form cell-cell contact with microvascular ECs (99).

5. Annexin A2 and tumor cell proliferation

ANXA2 is a key contributor to the stimulation of tumor cell proliferation and promotion of cancer growth under multiple regulatory modes. A relatively early study demonstrated a higher expression of ANXA2 in pancreatic carcinoma cell lines compared with cells of the normal pancreas, and an inverse relationship was noted between the levels of ANXA2 and the doubling time of the culture cells (100). As one of the most highly expressed genes in primary multiple myeloma (MM) cells (101), ANXA2 was found to increase cell proliferation and inhibit cell apoptosis (2). Downregulation of ANXA2 expression by siRNA in lung cancer A549 cells or breast cancer MDA-MB-231 and JIMT-1 cells significantly decreased the cell proliferative capacity (74,102). Similarly, in human HCC cells, silencing of ANXA2 suppressed cell proliferation and led to abnormal apoptosis, and the percentage of cells in the S phase was markedly decreased (103). When ANXA2 was suppressed by RNA interference (RNAi) in breast cancer cells, the treated cells were found to accumulate in the G0/G1 phase accompanied by a decrease in the S/G2+M phase population and a reduction in cell proliferation (104). ANXA2 facilitates proliferation and inhibits apoptosis via different pathways in a wide range of cancer cell types. ANXA2 was suggested as a part of the primer recognition protein complex and DNA binding protein involved in DNA replication (61,62,64,65). ANXA2 was also identified as an RNA-binding protein interacting with specific mRNAs such as its cognate mRNA and c-myc mRNA that are involved in the transport and/or anchorage of specific mRNAs (25,105107). In human HeLa, 293 and 293T cells, for example, downregulation of ANXA2 protein levels reduced DNA synthesis and inhibited cell division and proliferation (66). The interaction of ANXA2 and c-myc mRNA was found to lead to the increased expression levels of c-myc protein which is involved in cell proliferation, differentiation and apoptosis (108). It is well known that p53, as a tumor suppressor, plays a critical role in cell cycle regulation and apoptosis in different cancer cells. ANXA2 has also been proposed to be involved in p53-mediated apoptosis based on a study that overexpression of p53 induced apoptosis of lung cancer cells concomitantly with downregulation of ANXA2. In addition, ANXA2 knockdown increased the levels of p53 and its downstream gene expression, and caused p53 translocation from the cytoplasm to the nucleus (109). In vivo and in vitro studies showed that ANXA2 facilitates cell cycle progression and cell proliferation in part mediated by inhibition of p53 expression (68). Furthermore, ANXA2 was reported as a receptor mediating proliferative and anti-apoptotic effects of progastrin/gastrin on target cells such as colon cancer and pancreatic cancer (110,111).

6. Annexin A2 in tumor neovascularization

A considerable body of research has documented that angiogenesis is one hallmark of cancer (112), and is required for tumor growth, migration and metastasis (113,114). This process is initiated by the activation of proangiogenic factors such as vascular EC growth factor (VEGF), basic fibroblast growth factor (bFGF), plasminogen, followed by degradation of the ECM and proliferation and migration of ECs, as well as the synthesis of new matrix components (115118). Increased ANXA2 expression in tumors has been recognized as a key contributor to cancer angiogenesis in vivo and in vitro (47,119). Studies in human breast tumor xenograft models have demonstrated that neoangiogenesis in the tumor microenvironment can be markedly inhibited by the ANXA2 antibody, indicating the involvement of ANXA2 in new vessel formation in cancer (120). Clinical specimens also showed that the accumulation of tPA and ANXA2 on the surface of invasive human breast cancer was correlated with tumor neovascularization (48). The plasminogen/plasmin system activated matrix metalloproteinases (MMPs) into active protease which is required for the degradation of the ECM during the sprouting of new blood vessels (121). ANXA2 plays an important role in the plasminogen activation system and acts as a tPA receptor on the cell surface of endothelial and cancer cells, which mediates the conversion of plasminogen into plasmin (44,48). In addition, ANXA2 also participates in VEGF-mediated neovascularization. ANXA2 was found to be increased in a murine model of ischemic retinopathy through a VEGF/VEGF-R2/PKCβ pathway (122). Silencing of the ANXA2 gene by siRNA inhibited the expression of proangiogenic molecules, including VEGF, leading to the inhibition of neovascularization (2). Simultaneously, addition of purified domains I and IV of ANXA2 partly inhibited VEGF-dependent formation of capillary-like networks in a dose-dependent manner (123). Furthermore, ANXA2 was demonstrated to interact directly with the vascular endothelial cadherin (VE-cad)-based complex which is required to maintain VE-cad-dependent cell-cell junctions responsible for the maintenance of vascular endothelium integrity (124). The domains I and IV of ANXA2 compete with endogenous ANXA2 for interaction with VE-cad, leading to the disruption of the capillary-like network by affecting endothelial cell-cell contacts (123). Studies also showed that under the stimulation of sphingosine 1 and angiogenic growth factors, ANXA2 regulated Akt activation in sprouting angiogenesis, and depletion of ANXA2 attenuated Akt activation during EC invasion which was associated with increased phosphorylation of VE-cad and endothelial barrier leakage (125).

Importantly, increased vasculogenesis which occurs via mature ECs from proliferation and differentiation of bone marrow-derived endothelial progenitor cells (EPCs) has been recognized to contribute to tumor development (126128). In addition to the direct cellular contribution to new vessel formation, EPCs secrete a spectrum of proangiogenic cytokines that promote not only angiogenesis but vasculogenesis by different modes, thus playing a crucial role in neovascularization during neonatal growth and tumor progression (126128). However, EPCs mobilized from the bone marrow into the peripheral circulation, migrating and adhering to the sites of new vessel formation is a complex process dependent on cell active mobility. Cytoskeleton remodeling plays crucial roles in cell mobility. Various cell activities, including migration, morphological change and polarity formation are regulated by actin filament dynamics, including actin filament disassembly, severing and reorganization (129,130). Studies have shown that the dysfunction of actin leads to the impairment in EPC functions, including tube formation (131,132). ANXA2 plays a crucial role in regulating actin cytoskeletal rearrangements by binding the regions of free-barbed ends (19). Thus, ANXA2 may be involved in the neovascularization of EPCs by interacting with actin, yet this theory requires more supporting evidence.

These findings indicate that ANXA2 plays a crucial role in tumor progression by enhancing neovascularization. Thus, ANXA2 may be a potential target for the therapeutic management of cancer via blockage of ANXA2-mediated neovascularization.

7. Annexin A2 in tumor invasion and metastasis

Tumor invasion and metastasis is responsible for the majority of deaths among cancer patients. This complex process includes adhesion of tumor cells to ECM proteins, proteolysis of ECM proteins and remodeling of ECM. Through these mechanisms, tumor cells create intercellular spaces for migration, an event that requires membrane synthesis and cytoskeletal rearrangements. The contribution of ANXA2 to tumor invasion and metastasis by interacting with its potential invasion-associated protease proteins as well as the actin cytoskeleton has been reported in many advanced human tumors. In breast cancer, for example, ANXA2 was found to be overexpressed in the highly invasive cell line MDA-MB-231 compared with a poorly invasive cell line MCF-7 (75). In MCF-7/ADR cells, the administration of adriamycin increased the expression of ANXA2 consistent with the enhancement in cell proliferation and invasion, suggesting the involvement of ANXA2 in cancer cell invasion (133). In two head and neck squamous cell carcinoma cell lines, respectively, ANXA2 was found to be upregulated in metastatic lymph node compare with the primary tumor of the same patient (134). Analogously, the elevated expression of ANXA2 was also detected in lymph node metastatic tissues of lung cancers (88). ANXA2 was also differentially expressed in a pair of canine glioma subclones that exhibited different invasive phenotypes in rat brains yet had similar genetic backgrounds (135). In addition, the reduction in ANXA2 expression by siRNA or neutralizing antibodies significantly inhibited the motility and invasion of a number of cancer types such as ovarian cancer, human glioma and HCC (97,103,136), further supporting the contribution of ANXA2 in tumor invasion. Accumulating data suggest that the plasminogen activation system plays a crucial role in various processes of tumor development, including activation of MMPs, degradation of ECM and switch of growth factors, which together facilitate cellular migration and invasion (137142). The convertion of inactive enzyme plasminogen to active serine protease plasmin is a key event in this process, which is mediated by plasminogen activators, t-PA and urokinase plasminogen activator (uPA). ANXA2 catalyzes the conversion via the interaction with tPA, thus efficiently enforcing the effects of plasmin on tumor angiogenesis and tissue remodeling, MMPs and latent growth factor activation, and ECM degration, leading to tumor progression and metastasis (44,45,143) (Fig. 1).

Moreover, ANXA2 has also been shown to regulate migration and invasion of tumor cells by interaction with other proteins. In invasive human breast cancer cell lines which overexpress ANXA2, the invasive capacity of the cancer cells was decreased by the siRNA of ANXA2 via inhibition of c-myc expression (104). In contrast, upregulation of ANXA2 in the noninvasive breast cancer cell line MCF-7 was correlated with enhanced migration and invasion ability of cells both in vitro and in vivo by increasing expression of c-myc and cyclin D1 via activation of the Erk1/2 signaling pathways (144). In HCC, the interaction between ANXA2 and CD147 was found to regulate the trafficking of CD147-harboring membrane microvesicles thereby promoting the production of MMP-2 by tumor stoma fibroblasts, and suppressing the migration and invasion of tumor cells (145,146). ANXA2 was also confirmed to promote pancreatic cancer cell motility by interaction with S100A6 (147). Studies also showed that the location of ANXA2 on the cell surface promoted TGFβ-Rho-mediated epithelial-mesenchymal transition (EMT) in pancreatic ductal adenocarcinoma (36) which is an important process for the invasion and metastasis of this cancer (148).

These results indicate that ANXA2 may be a vital component in the regulation of tumor invasion and metastasis, and understanding of the mechanisms of ANXA2-mediated tumor development is crucial.

8. Conclusion

Development of a tumor involves a complex process, and multiple pathological events are considered to mediate and drive tumor cell growth and development. ANXA2 is an important molecule involved in regulating tumor cell adhesion, proliferation, invasion, metastasis and tumor neovasculogenesis, thus playing a crucial role in tumor development. The cellular and molecular mechanisms of the effects of ANXA2 on tumor development require further elucidation, and may provide a potential efficient therapeutic target for molecular-based strategies for tumor treatment.

Abbreviations:

tPA

tissue plasminogen activator

ECM

extracellular matrix

NES

nuclear export signal

STAT6

signal transducer and activator of transcription 6

HCC

hepatocellular carcinoma

NSCLC

non-small cell lung cancer

VEGF

vascular endothelial cell growth factor

bFGF

basic fibroblast growth factor

EC

endothelial cell

MMPs

matrix metalloproteinases

VE-cad

vascular endothelial cadherin

EPCs

endothelial progenitor cells

References

1 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Bao H, Jiang M, Zhu M, Sheng F, Ruan J and Ruan C: Overexpression of Annexin II affects the proliferation, apoptosis, invasion and production of proangiogenic factors in multiple myeloma. Int J Hematol. 90:177–185. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Sharma MC and Sharma M: The role of annexin II in angiogenesis and tumor progression: A potential therapeutic target. Curr Pharm Des. 13:3568–3575. 2007. View Article : Google Scholar

4 

Lokman NA, Ween MP, Oehler MK and Ricciardelli C: The role of annexin A2 in tumorigenesis and cancer progression. Cancer Microenviron. 4:199–208. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Gerke V and Moss SE: Annexins: From structure to function. Physiol Rev. 82:331–371. 2002.PubMed/NCBI

6 

Rescher U and Gerke V: Annexins - unique membrane binding proteins with diverse functions. J Cell Sci. 117:2631–2639. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Hajjar KA, Guevara CA, Lev E, Dowling K and Chacko J: Interaction of the fibrinolytic receptor, annexin II, with the endothelial cell surface. Essential role of endonexin repeat 2. J Biol Chem. 271:21652–21659. 1996. View Article : Google Scholar : PubMed/NCBI

8 

Mai J, Finley RL Jr, Waisman DM and Sloane BF: Human procathepsin B interacts with the annexin II tetramer on the surface of tumor cells. J Biol Chem. 275:12806–12812. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Madureira PA, Surette AP, Phipps KD, Taboski MA, Miller VA and Waisman DM: The role of the annexin A2 heterotetramer in vascular fibrinolysis. Blood. 118:4789–4797. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Hajjar KA, Jacovina AT and Chacko J: An endothelial cell receptor for plasminogen/tissue plasminogen activator. I. Identity with annexin II. J Biol Chem. 269:21191–21197. 1994.PubMed/NCBI

11 

Courtneidge S, Ralston R, Alitalo K and Bishop JM: Subcellular location of an abundant substrate (p36) for tyrosine-specific protein kinases. Mol Cell Biol. 3:340–350. 1983.PubMed/NCBI

12 

Eberhard DA, Karns LR, VandenBerg SR and Creutz CE: Control of the nuclear-cytoplasmic partitioning of annexin II by a nuclear export signal and by p11 binding. J Cell Sci. 114:3155–3166. 2001.PubMed/NCBI

13 

Burger A, Berendes R, Liemann S, et al: The crystal structure and ion channel activity of human annexin II, a peripheral membrane protein. J Mol Biol. 257:839–847. 1996. View Article : Google Scholar : PubMed/NCBI

14 

Camors E, Monceau V and Charlemagne D: Annexins and Ca2+ handling in the heart. Cardiovasc Res. 65:793–802. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Jost M, Thiel C, Weber K and Gerke V: Mapping of three unique Ca2+-binding sites in human annexin II. Eur J Biochem. 207:923–930. 1992. View Article : Google Scholar : PubMed/NCBI

16 

Jost M, Weber K and Gerke V: Annexin II contains two types of Ca2+-binding sites. Biochem J. 298:553–559. 1994.

17 

Filipenko NR, Kang HM and Waisman DM: Characterization of the Ca2+-binding sites of annexin II tetramer. J Biol Chem. 275:38877–38884. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Filipenko NR and Waisman DM: The C terminus of annexin II mediates binding to F-actin. J Biol Chem. 276:5310–5315. 2001. View Article : Google Scholar

19 

Hayes MJ, Shao D, Bailly M and Moss SE: Regulation of actin dynamics by annexin 2. EMBO J. 25:1816–1826. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Jones PG, Moore GJ and Waisman DM: A nonapeptide to the putative F-actin binding site of annexin-II tetramer inhibits its calcium-dependent activation of actin filament bundling. J Biol Chem. 267:13993–13997. 1992.PubMed/NCBI

21 

Rescher U, Ruhe D, Ludwig C, Zobiack N and Gerke V: Annexin 2 is a phosphatidylinositol (4,5)-bisphosphate binding protein recruited to actin assembly sites at cellular membranes. J Cell Sci. 117:3473–3480. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Su M, Shi JJ, Yang YP, Li J, Zhang YL, Chen J, Hu LF and Liu CF: HDAC6 regulates aggresome-autophagy degradation pathway of α-synuclein in response to MPP+-induced stress. J Neurochem. 117:112–120. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Aukrust I, Hollås H, Strand E, Evensen L, Travé G, Flatmark T and Vedeler A: The mRNA-binding site of annexin A2 resides in helices C-D of its domain IV. J Mol Biol. 368:1367–1378. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Kassam G, Manro A, Braat CE, Louie P, Fitzpatrick SL and Waisman DM: Characterization of the heparin binding properties of annexin II tetramer. J Biol Chem. 272:15093–15100. 1997. View Article : Google Scholar : PubMed/NCBI

25 

Vedeler A and Hollås H: Annexin II is associated with mRNAs which may constitute a distinct subpopulation. Biochem J. 348:565–572. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Gould KL, Woodgett JR, Isacke CM and Hunter T: The protein-tyrosine kinase substrate p36 is also a substrate for protein kinase C in vitro and in vivo. Mol Cell Biol. 6:2738–2744. 1986.PubMed/NCBI

27 

Jost M and Gerke V: Mapping of a regulatory important site for protein kinase C phosphorylation in the N-terminal domain of annexin II. Biochim Biophys Acta. 1313:283–289. 1996. View Article : Google Scholar : PubMed/NCBI

28 

Glenney JR Jr and Tack BF: Amino-terminal sequence of p36 and associated p10: Identification of the site of tyrosine phosphorylation and homology with S-100. Proc Natl Acad Sci USA. 82:7884–7888. 1985. View Article : Google Scholar : PubMed/NCBI

29 

Luo W, Yan G, Li L, et al: Epstein-Barr virus latent membrane protein 1 mediates serine 25 phosphorylation and nuclear entry of annexin A2 via PI-PLC-PKCalpha/PKCbeta pathway. Mol Carcinog. 47:934–946. 2008. View Article : Google Scholar : PubMed/NCBI

30 

de Graauw M, Tijdens I, Smeets MB, Hensbergen PJ, Deelder AM and van de Water B: Annexin A2 phosphorylation mediates cell scattering and branching morphogenesis via cofilin activation. Mol Cell Biol. 28:1029–1040. 2008. View Article : Google Scholar :

31 

Morel E and Gruenberg J: Annexin A2 binding to endosomes and functions in endosomal transport are regulated by tyrosine 23 phosphorylation. J Biol Chem. 284:1604–1611. 2009. View Article : Google Scholar

32 

Hubaishy I, Jones PG, Bjorge J, Bellagamba C, Fitzpatrick S, Fujita DJ and Waisman DM: Modulation of annexin II tetramer by tyrosine phosphorylation. Biochemistry. 34:14527–14534. 1995. View Article : Google Scholar : PubMed/NCBI

33 

Regnouf F, Sagot I, Delouche B, Devilliers G, Cartaud J, Henry JP and Pradel LA: ‘In vitro’ phosphorylation of annexin 2 heterotetramer by protein kinase C. Comparative properties of the unphosphorylated and phosphorylated annexin 2 on the aggregation and fusion of chromaffin granule membranes. J Biol Chem. 270:27143–27150. 1995. View Article : Google Scholar : PubMed/NCBI

34 

Johnstone SA, Hubaishy I and Waisman DM: Phosphorylation of annexin II tetramer by protein kinase C inhibits aggregation of lipid vesicles by the protein. J Biol Chem. 267:25976–25981. 1992.PubMed/NCBI

35 

Deora AB, Kreitzer G, Jacovina AT and Hajjar KA: An annexin 2 phosphorylation switch mediates p11-dependent translocation of annexin 2 to the cell surface. J Biol Chem. 279:43411–43418. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Zheng L, Foley K, Huang L, et al: Tyrosine 23 phosphorylation-dependent cell-surface localization of annexin A2 is required for invasion and metastases of pancreatic cancer. PLoS One. 6:e193902011. View Article : Google Scholar : PubMed/NCBI

37 

Yan G, Luo W, Lu Z, Luo X, Li L, Liu S, Liu Y, Tang M, Dong Z and Cao Y: Epstein-Barr virus latent membrane protein 1 mediates phosphorylation and nuclear translocation of annexin A2 by activating PKC pathway. Cell Signal. 19:341–348. 2007. View Article : Google Scholar

38 

Liu J, Rothermund CA, Ayala-Sanmartin J and Vishwanatha JK: Nuclear annexin II negatively regulates growth of LNCaP cells and substitution of ser 11 and 25 to glu prevents nucleocytoplasmic shuttling of annexin II. BMC Biochem. 4:102003. View Article : Google Scholar

39 

Madureira PA, Hill R, Miller VA, Giacomantonio C, Lee PW and Waisman DM: Annexin A2 is a novel cellular redox regulatory protein involved in tumorigenesis. Oncotarget. 2:1075–1093. 2011.PubMed/NCBI

40 

Johnsson N, Marriott G and Weber K: p36, the major cytoplasmic substrate of src tyrosine protein kinase, binds to its p11 regulatory subunit via a short amino-terminal amphiphatic helix. EMBO J. 7:2435–2442. 1988.PubMed/NCBI

41 

Réty S, Sopkova J, Renouard M, Osterloh D, Gerke V, Tabaries S, Russo-Marie F and Lewit-Bentley A: The crystal structure of a complex of p11 with the annexin II N-terminal peptide. Nat Struct Biol. 6:89–95. 1999. View Article : Google Scholar : PubMed/NCBI

42 

Waisman DM: Annexin II tetramer: Structure and function. Mol Cell Biochem. 149–150:301–322. 1995. View Article : Google Scholar

43 

Kassam G, Choi KS, Ghuman J, Kang HM, Fitzpatrick SL, Zackson T, Zackson S, Toba M, Shinomiya A and Waisman DM: The role of annexin II tetramer in the activation of plasminogen. J Biol Chem. 273:4790–4799. 1998. View Article : Google Scholar : PubMed/NCBI

44 

Cesarman GM, Guevara CA and Hajjar KA: An endothelial cell receptor for plasminogen/tissue plasminogen activator (t-PA). II. Annexin II-mediated enhancement of t-PA-dependent plasminogen activation. J Biol Chem. 269:21198–21203. 1994.PubMed/NCBI

45 

Hajjar KA and Krishnan S: Annexin II: A mediator of the plasmin/plasminogen activator system. Trends Cardiovasc Med. 9:128–138. 1999. View Article : Google Scholar

46 

Jacovina AT, Deora AB, Ling Q, Broekman MJ, Almeida D, Greenberg CB, Marcus AJ, Smith JD and Hajjar KA: Homocysteine inhibits neoangiogenesis in mice through blockade of annexin A2-dependent fibrinolysis. J Clin Invest. 119:3384–3394. 2009.PubMed/NCBI

47 

Valapala M, Thamake SI and Vishwanatha JK: A competitive hexapeptide inhibitor of annexin A2 prevents hypoxia-induced angiogenic events. J Cell Sci. 124:1453–1464. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Sharma M, Ownbey RT and Sharma MC: Breast cancer cell surface annexin II induces cell migration and neoangiogenesis via tPA dependent plasmin generation. Exp Mol Pathol. 88:278–286. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Gerke V, Creutz CE and Moss SE: Annexins: Linking Ca2+ signalling to membrane dynamics. Nat Rev Mol Cell Biol. 6:449–461. 2005. View Article : Google Scholar : PubMed/NCBI

50 

Ali SM, Geisow MJ and Burgoyne RD: A role for calpactin in calcium-dependent exocytosis in adrenal chromaffin cells. Nature. 340:313–315. 1989. View Article : Google Scholar : PubMed/NCBI

51 

Sarafian T, Pradel LA, Henry JP, Aunis D and Bader MF: The participation of annexin II (calpactin I) in calcium-evoked exocytosis requires protein kinase C. J Cell Biol. 114:1135–1147. 1991. View Article : Google Scholar : PubMed/NCBI

52 

Morel E, Parton RG and Gruenberg J: Annexin A2-dependent polymerization of actin mediates endosome biogenesis. Dev Cell. 16:445–457. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Yamada A, Irie K, Hirota T, Ooshio T, Fukuhara A and Takai Y: Involvement of the annexin II-S100A10 complex in the formation of E-cadherin-based adherens junctions in Madin-Darby canine kidney cells. J Biol Chem. 280:6016–6027. 2005. View Article : Google Scholar

54 

Mai J, Waisman DM and Sloane BF: Cell surface complex of cathepsin B/annexin II tetramer in malignant progression. Biochim Biophys Acta. 1477:215–230. 2000. View Article : Google Scholar : PubMed/NCBI

55 

Grieve AG, Moss SE and Hayes MJ: Annexin A2 at the interface of actin and membrane dynamics: A focus on its roles in endocytosis and cell polarization. Int J Cell Biol. 2012:8524302012. View Article : Google Scholar : PubMed/NCBI

56 

Jost M, Zeuschner D, Seemann J, Weber K and Gerke V: Identification and characterization of a novel type of annexin-membrane interaction: Ca2+ is not required for the association of annexin II with early endosomes. J Cell Sci. 110:221–228. 1997.

57 

König J and Gerke V: Modes of annexin-membrane interactions analyzed by employing chimeric annexin proteins. Biochim Biophys Acta. 1498:174–180. 2000. View Article : Google Scholar : PubMed/NCBI

58 

Liu J and Vishwanatha JK: Regulation of nucleo-cytoplasmic shuttling of human annexin A2: A proposed mechanism. Mol Cell Biochem. 303:211–220. 2007. View Article : Google Scholar : PubMed/NCBI

59 

Jindal HK and Vishwanatha JK: Purification and characterization of primer recognition proteins from HeLa cells. Biochemistry. 29:4767–4773. 1990. View Article : Google Scholar : PubMed/NCBI

60 

Kumble KD and Vishwanatha JK: Immunoelectron microscopic analysis of the intracellular distribution of primer recognition proteins, annexin 2 and phosphoglycerate kinase, in normal and transformed cells. J Cell Sci. 99:751–758. 1991.PubMed/NCBI

61 

Vishwanatha JK, Jindal HK and Davis RG: The role of primer recognition proteins in DNA replication: Association with nuclear matrix in HeLa cells. J Cell Sci. 101:25–34. 1992.PubMed/NCBI

62 

Boyko V, Mudrak O, Svetlova M, Negishi Y, Ariga H and Tomilin N: A major cellular substrate for protein kinases, annexin II, is a DNA-binding protein. FEBS Lett. 345:139–142. 1994. View Article : Google Scholar : PubMed/NCBI

63 

Krutilina RI, Babich VS, Kropotov AV, Mudrak OS, Chesnokov IN, Turoverova LV, Konstantinova IM and Tomilin NV: The DNA-binding activity of the membrane protein annexin II and its interaction with antibodies to the chromatin nuclear ribonucleoprotein (alpha-RNP). Tsitologiia. 38:1106–1114. 1996.In Russian.

64 

Vishwanatha JK and Kumble S: Involvement of annexin II in DNA replication: Evidence from cell-free extracts of Xenopus eggs. J Cell Sci. 105:533–540. 1993.PubMed/NCBI

65 

Kumble KD, Iversen PL and Vishwanatha JK: The role of primer recognition proteins in DNA replication: Inhibition of cellular proliferation by antisense oligodeoxyribonucleotides. J Cell Sci. 101:35–41. 1992.PubMed/NCBI

66 

Chiang Y, Rizzino A, Sibenaller ZA, Wold MS and Vishwanatha JK: Specific down-regulation of annexin II expression in human cells interferes with cell proliferation. Mol Cell Biochem. 199:139–147. 1999. View Article : Google Scholar : PubMed/NCBI

67 

Madureira PA, Hill R, Lee PW and Waisman DM: Genotoxic agents promote the nuclear accumulation of annexin A2: Role of annexin A2 in mitigating DNA damage. PLoS One. 7:e505912012. View Article : Google Scholar : PubMed/NCBI

68 

Wang CY, Chen CL, Tseng YL, Fang YT, Lin YS, Su WC, Chen CC, Chang KC, Wang YC and Lin CF: Annexin A2 silencing induces G2 arrest of non-small cell lung cancer cells through p53-dependent and -independent mechanisms. J Biol Chem. 287:32512–32524. 2012. View Article : Google Scholar : PubMed/NCBI

69 

Das S, Shetty P, Valapala M, Dasgupta S, Gryczynski Z and Vishwanatha JK: Signal transducer and activator of transcription 6 (STAT6) is a novel interactor of annexin A2 in prostate cancer cells. Biochemistry. 49:2216–2226. 2010. View Article : Google Scholar : PubMed/NCBI

70 

Frohlich M, Motté P, Galvin K, Takahashi H, Wands J and Ozturk M: Enhanced expression of the protein kinase substrate p36 in human hepatocellular carcinoma. Mol Cell Biol. 10:3216–3223. 1990.PubMed/NCBI

71 

Yang T, Peng H, Wang J, Yang J, Nice EC, Xie K and Huang C: Prognostic and diagnostic significance of annexin A2 in colorectal cancer. Colorectal Dis. 15:e373–e381. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Duncan R, Carpenter B, Main LC, Telfer C and Murray GI: Characterisation and protein expression profiling of annexins in colorectal cancer. Br J Cancer. 98:426–433. 2008. View Article : Google Scholar

73 

Deng S, Wang J, Hou L, Li J, Chen G, Jing B, Zhang X and Yang Z: Annexin A1, A2, A4 and A5 play important roles in breast cancer, pancreatic cancer and laryngeal carcinoma, alone and/or synergistically. Oncol Lett. 5:107–112. 2013.

74 

Shetty PK, Thamake SI, Biswas S, Johansson SL and Vishwanatha JK: Reciprocal regulation of annexin A2 and EGFR with Her-2 in Her-2 negative and herceptin-resistant breast cancer. PLoS One. 7:e442992012. View Article : Google Scholar : PubMed/NCBI

75 

Sharma MR, Koltowski L, Ownbey RT, Tuszynski GP and Sharma MC: Angiogenesis-associated protein annexin II in breast cancer: Selective expression in invasive breast cancer and contribution to tumor invasion and progression. Exp Mol Pathol. 81:146–156. 2006. View Article : Google Scholar : PubMed/NCBI

76 

Chuthapisith S, Bean BE, Cowley G, et al: Annexins in human breast cancer: Possible predictors of pathological response to neoadjuvant chemotherapy. Eur J Cancer. 45:1274–1281. 2009. View Article : Google Scholar : PubMed/NCBI

77 

Yao H, Zhang Z, Xiao Z, et al: Identification of metastasis associated proteins in human lung squamous carcinoma using two-dimensional difference gel electrophoresis and laser capture microdissection. Lung Cancer. 65:41–48. 2009. View Article : Google Scholar

78 

Zhang HJ, Yao DF, Yao M, Huang H, Wu W, Yan MJ, Yan XD and Chen J: Expression characteristics and diagnostic value of annexin A2 in hepatocellular carcinoma. World J Gastroenterol. 18:5897–5904. 2012. View Article : Google Scholar : PubMed/NCBI

79 

Zhang Q, Ye Z, Yang Q, He X, Wang H and Zhao Z: Upregulated expression of annexin II is a prognostic marker for patients with gastric cancer. World J Surg Oncol. 10:1032012. View Article : Google Scholar : PubMed/NCBI

80 

Takano S, Togawa A, Yoshitomi H, et al: Annexin II overexpression predicts rapid recurrence after surgery in pancreatic cancer patients undergoing gemcitabine-adjuvant chemotherapy. Ann Surg Oncol. 15:3157–3168. 2008. View Article : Google Scholar : PubMed/NCBI

81 

Yang SF, Hsu HL, Chao TK, Hsiao CJ, Lin YF and Cheng CW: Annexin A2 in renal cell carcinoma: Expression, function, and prognostic significance. Urol Oncol. 33:22.e11–22.e21. 2014. View Article : Google Scholar

82 

Ma RL, Shen LY and Chen KN: Coexpression of ANXA2, SOD2 and HOXA13 predicts poor prognosis of esophageal squamous cell carcinoma. Oncol Rep. 31:2157–2164. 2014.PubMed/NCBI

83 

Ohno Y, Izumi M, Kawamura T, Nishimura T, Mukai K and Tachibana M: Annexin II represents metastatic potential in clear-cell renal cell carcinoma. Br J Cancer. 101:287–294. 2009. View Article : Google Scholar : PubMed/NCBI

84 

Emoto K, Sawada H, Yamada Y, et al: Annexin II overexpression is correlated with poor prognosis in human gastric carcinoma. Anticancer Res. 21:1339–1345. 2001.PubMed/NCBI

85 

Liu Z, Ling Q, Wang J, Xie H, Xu X and Zheng S: Annexin A2 is not a good biomarker for hepatocellular carcinoma in cirrhosis. Oncol Lett. 6:125–129. 2013.PubMed/NCBI

86 

Ji NY, Park MY, Kang YH, et al: Evaluation of annexin II as a potential serum marker for hepatocellular carcinoma using a developed sandwich ELISA method. Int J Mol Med. 24:765–771. 2009.PubMed/NCBI

87 

Jeon YR, Kim SY, Lee EJ, Kim YN, Noh DY, Park SY and Moon A: Identification of annexin II as a novel secretory biomarker for breast cancer. Proteomics. 13:3145–3156. 2013. View Article : Google Scholar : PubMed/NCBI

88 

Luo CH, Liu QQ, Zhang PF, Li MY, Chen ZC and Liu YF: Prognostic significance of annexin II expression in non-small cell lung cancer. Clin Transl Oncol. 15:938–946. 2013. View Article : Google Scholar : PubMed/NCBI

89 

Jia JW, Li KL, Wu JX and Guo SL: Clinical significance of annexin II expression in human non-small cell lung cancer. Tumour Biol. 34:1767–1771. 2013. View Article : Google Scholar : PubMed/NCBI

90 

Pei H, Zhu H, Zeng S, et al: Proteome analysis and tissue microarray for profiling protein markers associated with lymph node metastasis in colorectal cancer. J Proteome Res. 6:2495–2501. 2007. View Article : Google Scholar : PubMed/NCBI

91 

Emoto K, Yamada Y, Sawada H, Fujimoto H, Ueno M, Takayama T, Kamada K, Naito A, Hirao S and Nakajima Y: Annexin II overexpression correlates with stromal tenascin-C overexpression: A prognostic marker in colorectal carcinoma. Cancer. 92:1419–1426. 2001. View Article : Google Scholar : PubMed/NCBI

92 

Kagawa S, Takano S, Yoshitomi H, et al: Akt/mTOR signaling pathway is crucial for gemcitabine resistance induced by annexin II in pancreatic cancer cells. J Surg Res. 178:758–767. 2012. View Article : Google Scholar : PubMed/NCBI

93 

Liu Z, Feng JG, Tuersun A, et al: Proteomic identification of differentially-expressed proteins in esophageal cancer in three ethnic groups in Xinjiang. Mol Biol Rep. 38:3261–3269. 2011. View Article : Google Scholar

94 

Zhi H, Zhang J, Hu G, Lu J, Wang X, Zhou C, Wu M and Liu Z: The deregulation of arachidonic acid metabolism-related genes in human esophageal squamous cell carcinoma. Int J Cancer. 106:327–333. 2003. View Article : Google Scholar : PubMed/NCBI

95 

Pena-Alonso E, Rodrigo JP, Parra IC, Pedrero JM, Meana MV, Nieto CS, Fresno MF, Morgan RO and Fernandez MP: Annexin A2 localizes to the basal epithelial layer and is down-regulated in dysplasia and head and neck squamous cell carcinoma. Cancer Lett. 263:89–98. 2008. View Article : Google Scholar : PubMed/NCBI

96 

Tressler RJ, Updyke TV, Yeatman T and Nicolson GL: Extracellular annexin II is associated with divalent cation-dependent tumor cell-endothelial cell adhesion of metastatic RAW117 large-cell lymphoma cells. J Cell Biochem. 53:265–276. 1993. View Article : Google Scholar : PubMed/NCBI

97 

Lokman NA, Elder AS, Ween MP, Pyragius CE, Hoffmann P, Oehler MK and Ricciardelli C: Annexin A2 is regulated by ovarian cancer-peritoneal cell interactions and promotes metastasis. Oncotarget. 4:1199–1211. 2013.PubMed/NCBI

98 

Shiozawa Y, Havens AM, Jung Y, et al: Annexin II/annexin II receptor axis regulates adhesion, migration, homing, and growth of prostate cancer. J Cell Biochem. 105:370–380. 2008. View Article : Google Scholar : PubMed/NCBI

99 

Myrvang HK, Guo X, Li C and Dekker LV: Protein interactions between surface annexin A2 and S100A10 mediate adhesion of breast cancer cells to microvascular endothelial cells. FEBS Lett. 587:3210–3215. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Kumble KD, Hirota M, Pour PM and Vishwanatha JK: Enhanced levels of annexins in pancreatic carcinoma cells of Syrian hamsters and their intrapancreatic allografts. Cancer Res. 52:163–167. 1992.PubMed/NCBI

101 

Claudio JO, Masih-Khan E, Tang H, et al: A molecular compendium of genes expressed in multiple myeloma. Blood. 100:2175–2186. 2002. View Article : Google Scholar : PubMed/NCBI

102 

Wang YX, Lv H, Li ZX, Li C and Wu XY: Effect of shRNA mediated down-regulation of annexin A2 on biological behavior of human lung adencarcinoma cells A549. Pathol Oncol Res. 18:183–190. 2012. View Article : Google Scholar

103 

Zhang HJ, Yao DF, Yao M, Huang H, Wang L, Yan MJ, Yan XD, Gu X, Wu W and Lu SL: Annexin A2 silencing inhibits invasion, migration, and tumorigenic potential of hepatoma cells. World J Gastroenterol. 19:3792–3801. 2013. View Article : Google Scholar : PubMed/NCBI

104 

Zhang J, Guo B, Zhang Y, Cao J and Chen T: Silencing of the annexin II gene down-regulates the levels of S100A10, c-Myc, and plasmin and inhibits breast cancer cell proliferation and invasion. Saudi Med J. 31:374–381. 2010.PubMed/NCBI

105 

Filipenko NR, MacLeod TJ, Yoon CS and Waisman DM: Annexin A2 is a novel RNA-binding protein. J Biol Chem. 279:8723–8731. 2004. View Article : Google Scholar

106 

Mickleburgh I, Burtle B, Hollås H, Campbell G, Chrzanowska-Lightowlers Z, Vedeler A and Hesketh J: Annexin A2 binds to the localization signal in the 3′ untranslated region of c-myc mRNA. FEBS J. 272:413–421. 2005. View Article : Google Scholar : PubMed/NCBI

107 

Hollås H, Aukrust I, Grimmer S, Strand E, Flatmark T and Vedeler A: Annexin A2 recognises a specific region in the 3′-UTR of its cognate messenger RNA. Biochim Biophys Acta. 1763:1325–1334. 2006. View Article : Google Scholar

108 

Pelengaris S and Khan M: The c-MYC oncoprotein as a treatment target in cancer and other disorders of cell growth. Expert Opin Ther Targets. 7:623–642. 2003. View Article : Google Scholar : PubMed/NCBI

109 

Huang Y, Jin Y, Yan CH, Yu Y, Bai J, Chen F, Zhao YZ and Fu SB: Involvement of annexin A2 in p53 induced apoptosis in lung cancer. Mol Cell Biochem. 309:117–123. 2008. View Article : Google Scholar

110 

Rengifo-Cam W, Umar S, Sarkar S and Singh P: Antiapoptotic effects of progastrin on pancreatic cancer cells are mediated by sustained activation of nuclear factor-κB. Cancer Res. 67:7266–7274. 2007. View Article : Google Scholar : PubMed/NCBI

111 

Sarkar S, Swiercz R, Kantara C, Hajjar KA and Singh P: Annexin A2 mediates up-regulation of NF-κB, β-catenin, and stem cell in response to progastrin in mice and HEK-293 cells. Gastroenterology. 140:583.e4–595.e4. 2011. View Article : Google Scholar

112 

Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI

113 

Folkman J: Tumor angiogenesis: Therapeutic implications. N Engl J Med. 285:1182–1186. 1971. View Article : Google Scholar : PubMed/NCBI

114 

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

115 

Hanahan D and Folkman J: Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 86:353–364. 1996. View Article : Google Scholar : PubMed/NCBI

116 

Carmeliet P: Angiogenesis in health and disease. Nat Med. 9:653–660. 2003. View Article : Google Scholar : PubMed/NCBI

117 

Risau W: Mechanisms of angiogenesis. Nature. 386:671–674. 1997. View Article : Google Scholar : PubMed/NCBI

118 

Bergers G and Benjamin LE: Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 3:401–410. 2003. View Article : Google Scholar : PubMed/NCBI

119 

Lima e Silva R, Shen J, Gong YY, Seidel CP, Hackett SF, Kesavan K, Jacoby DB and Campochiaro PA: Agents that bind annexin A2 suppress ocular neovascularization. J Cell Physiol. 225:855–864. 2010. View Article : Google Scholar : PubMed/NCBI

120 

Sharma M, Blackman MR and Sharma MC: Antibody-directed neutralization of annexin II (ANX II) inhibits neoangiogenesis and human breast tumor growth in a xenograft model. Exp Mol Pathol. 92:175–184. 2012. View Article : Google Scholar

121 

Carmeliet P: Mechanisms of angiogenesis and arteriogenesis. Nat Med. 6:389–395. 2000. View Article : Google Scholar : PubMed/NCBI

122 

Zhao S, Huang L, Wu J, Zhang Y, Pan D and Liu X: Vascular endothelial growth factor upregulates expression of annexin A2 in vitro and in a mouse model of ischemic retinopathy. Mol Vis. 15:1231–1242. 2009.PubMed/NCBI

123 

Raddum AM, Evensen L, Hollås H, Grindheim AK, Lorens JB and Vedeler A: Domains I and IV of annexin A2 affect the formation and integrity of in vitro capillary-like networks. PLoS One. 8:e602812013. View Article : Google Scholar : PubMed/NCBI

124 

Dejana E, Orsenigo F and Lampugnani MG: The role of adherens junctions and VE-cadherin in the control of vascular permeability. J Cell Sci. 121:2115–2122. 2008. View Article : Google Scholar : PubMed/NCBI

125 

Su SC, Maxwell SA and Bayless KJ: Annexin 2 regulates endothelial morphogenesis by controlling AKT activation and junctional integrity. J Biol Chem. 285:40624–40634. 2010. View Article : Google Scholar : PubMed/NCBI

126 

Sun XT, Yuan XW, Zhu HT, Deng ZM, Yu DC, Zhou X and Ding YT: Endothelial precursor cells promote angiogenesis in hepatocellular carcinoma. World J Gastroenterol. 18:4925–4933. 2012. View Article : Google Scholar : PubMed/NCBI

127 

Yu DC, Chen J, Sun XT, Zhuang LY, Jiang CP and Ding YT: Mechanism of endothelial progenitor cell recruitment into neo-vessels in adjacent non-tumor tissues in hepatocellular carcinoma. BMC Cancer. 10:4352010. View Article : Google Scholar : PubMed/NCBI

128 

Finney MR, Greco NJ, Haynesworth SE, et al: Direct comparison of umbilical cord blood versus bone marrow-derived endothelial precursor cells in mediating neovascularization in response to vascular ischemia. Biol Blood Marrow Transplant. 12:585–593. 2006. View Article : Google Scholar : PubMed/NCBI

129 

Pollard TD and Borisy GG: Cellular motility driven by assembly and disassembly of actin filaments. Cell. 112:453–465. 2003. View Article : Google Scholar : PubMed/NCBI

130 

Revenu C, Athman R, Robine S and Louvard D: The co-workers of actin filaments: From cell structures to signals. Nat Rev Mol Cell Biol. 5:635–646. 2004. View Article : Google Scholar : PubMed/NCBI

131 

Wei J, Liu Y, Chang M, Sun CL, Li DW, Liu ZQ and Hu LS: Proteomic analysis of oxidative modification in endothelial colony-forming cells treated by hydrogen peroxide. Int J Mol Med. 29:1099–1105. 2012.PubMed/NCBI

132 

Zhang X, Cui X, Cheng L, Guan X, Li H, Li X and Cheng M: Actin stabilization by jasplakinolide affects the function of bone marrow-derived late endothelial progenitor cells. PLoS One. 7:e508992012. View Article : Google Scholar : PubMed/NCBI

133 

Zhang F, Zhang L, Zhang B, Wei X, Yang Y, Qi RZ, Ying G, Zhang N and Niu R: Anxa2 plays a critical role in enhanced invasiveness of the multidrug resistant human breast cancer cells. J Proteome Res. 8:5041–5047. 2009. View Article : Google Scholar : PubMed/NCBI

134 

Wu W, Tang X, Hu W, Lotan R, Hong WK and Mao L: Identification and validation of metastasis-associated proteins in head and neck cancer cell lines by two-dimensional electrophoresis and mass spectrometry. Clin Exp Metastasis. 19:319–326. 2002. View Article : Google Scholar : PubMed/NCBI

135 

Maruo T, Ichikawa T, Kanzaki H, et al: Proteomics-based analysis of invasion-related proteins in malignant gliomas. Neuropathology. 33:264–275. 2013. View Article : Google Scholar

136 

Tatenhorst L, Rescher U, Gerke V and Paulus W: Knockdown of annexin 2 decreases migration of human glioma cells in vitro. Neuropathol Appl Neurobiol. 32:271–277. 2006. View Article : Google Scholar : PubMed/NCBI

137 

Mignatti P and Rifkin DB: Biology and biochemistry of proteinases in tumor invasion. Physiol Rev. 73:161–195. 1993.PubMed/NCBI

138 

McColl BK, Baldwin ME, Roufail S, Freeman C, Moritz RL, Simpson RJ, Alitalo K, Stacker SA and Achen MG: Plasmin activates the lymphangiogenic growth factors VEGF-C and VEGF-D. J Exp Med. 198:863–868. 2003. View Article : Google Scholar : PubMed/NCBI

139 

Bajou K, Masson V, Gerard RD, et al: The plasminogen activator inhibitor PAI-1 controls in vivo tumor vascularization by interaction with proteases, not vitronectin. Implications for antiangiogenic strategies. J Cell Biol. 152:777–784. 2001. View Article : Google Scholar : PubMed/NCBI

140 

Díaz VM, Planaguma J, Thomson TM, Reventós J and Paciucci R: Tissue plasminogen activator is required for the growth, invasion, and angiogenesis of pancreatic tumor cells. Gastroenterology. 122:806–819. 2002. View Article : Google Scholar : PubMed/NCBI

141 

Pepper MS: Extracellular proteolysis and angiogenesis. Thromb Haemost. 86:346–355. 2001.PubMed/NCBI

142 

Tarui T, Majumdar M, Miles LA, Ruf W and Takada Y: Plasmin-induced migration of endothelial cells. A potential target for the anti-angiogenic action of angiostatin. J Biol Chem. 277:33564–33570. 2002. View Article : Google Scholar : PubMed/NCBI

143 

Ranson M and Andronicos NM: Plasminogen binding and cancer: Promises and pitfalls. Front Biosci. 8:s294–s304. 2003. View Article : Google Scholar : PubMed/NCBI

144 

Wu B, Zhang F, Yu M, Zhao P, Ji W, Zhang H, Han J and Niu R: Up-regulation of Anxa2 gene promotes proliferation and invasion of breast cancer MCF-7 cells. Cell Prolif. 45:189–198. 2012. View Article : Google Scholar : PubMed/NCBI

145 

Zhang W, Zhao P, Xu XL, Cai L, Song ZS, Cao DY, Tao KS, Zhou WP, Chen ZN and Dou KF: Annexin A2 promotes the migration and invasion of human hepatocellular carcinoma cells in vitro by regulating the shedding of CD147-harboring microvesicles from tumor cells. PLoS One. 8:e672682013. View Article : Google Scholar : PubMed/NCBI

146 

Zhao P, Zhang W, Tang J, Ma XK, Dai JY, Li Y, Jiang JL, Zhang SH and Chen ZN: Annexin II promotes invasion and migration of human hepatocellular carcinoma cells in vitro via its interaction with HAb18G/CD147. Cancer Sci. 101:387–395. 2010. View Article : Google Scholar : PubMed/NCBI

147 

Nedjadi T, Kitteringham N, Campbell F, Jenkins RE, Park BK, Navarro P, Ashcroft F, Tepikin A, Neoptolemos JP and Costello E: S100A6 binds to annexin 2 in pancreatic cancer cells and promotes pancreatic cancer cell motility. Br J Cancer. 101:1145–1154. 2009. View Article : Google Scholar : PubMed/NCBI

148 

Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2:442–454. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2015
Volume 33 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu X, Pan W, Kang L, Feng H and Song Y: Association of annexin A2 with cancer development (Review). Oncol Rep 33: 2121-2128, 2015
APA
Xu, X., Pan, W., Kang, L., Feng, H., & Song, Y. (2015). Association of annexin A2 with cancer development (Review). Oncology Reports, 33, 2121-2128. https://doi.org/10.3892/or.2015.3837
MLA
Xu, X., Pan, W., Kang, L., Feng, H., Song, Y."Association of annexin A2 with cancer development (Review)". Oncology Reports 33.5 (2015): 2121-2128.
Chicago
Xu, X., Pan, W., Kang, L., Feng, H., Song, Y."Association of annexin A2 with cancer development (Review)". Oncology Reports 33, no. 5 (2015): 2121-2128. https://doi.org/10.3892/or.2015.3837