5
Annexin A11 in disease Jiasheng Wang a , Chunmei Guo a , Shuqing Liu b , Houbao Qi a , Yuling Yin b , Rui Liang c , Ming-Zhong Sun a, , Frederick T. Greenaway d a Department of Biotechnology, Dalian Medical University, Dalian 116044, China b Department of Biochemistry, Dalian Medical University, Dalian 116044, China c Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian 116027, China d Carlson School of Chemistry and Biochemistry, Clark University, Worcester, MA 01610, USA abstract article info Article history: Received 27 November 2013 Received in revised form 16 January 2014 Accepted 18 January 2014 Available online 6 February 2014 Keywords: Anxa11 Function Disease Cancer Ubiquitously expressed in many cell types, annexin A11 (Anxa11) is a member of the multigene family of Ca 2+ - regulated phospholipid-dependent and membrane-binding annexin proteins. Studies have shown that Anxa11 plays an important role in cell division, Ca 2+ signaling, vesicle trafcking and apoptosis. The deregulation and mutation of Anxa11 are involved in systemic autoimmune diseases, sarcoidosis and the development, chemoresistance and recurrence of cancers. Malfunction of Anxa11 may lead to or enhance the metastasis, inva- sion and drug resistance of cancers through the platelet-derived growth factor receptor (PDGFR) pathway and/or the mitogen-activated protein kinase (MAPK)/p53 pathway. In a variety of diseases, Anxa11 is most commonly reported to function through interactions with apoptosis-linked gene-2 protein (ALG-2) and/or calcyclin (S100A6). Although it has been little studied, Anxa11 is a promising biomarker for the diagnosis, treatment and prognosis of certain diseases. In this review, the associations of Anxa11 with Ca 2+ -regulated exocytosis, cy- tokinesis, sex differentiation, autoimmune diseases, thrombolysis and cancers are summarized and interpreted. © 2014 Elsevier B.V. All rights reserved. 1. Introduction 1.1. The features of annexins The annexins are Ca 2+ -regulated phospholipid-binding proteins widely expressed in all eukaryotic cells except yeasts [1,2]. The annexins have been classied into ve groups. Group A includes 12 members (annexins A1A11 and A13; their chromosome locations are summa- rized in Table 1) found in vertebrates (mammalian); Group B, C, D and E annexins are found in non-vertebrates, fungi/molds, plants and pro- tists, respectively [3]. The annexins are cytosolic proteins distributed both in the cellular cytoplasm and in the nuclear membranes. They ex- hibit diverse biological functions by mediating the interactions between proteins in cell membranes with other proteins in cells, in the nuclear membrane, and in the extracellular matrix [47]. The annexins are composed of a conserved C-terminal core and a variable N-terminal domain [7]. The C-terminal core contains four ho- mologous repeat domains except for annexin A6, which has eight. Each annexin repeat contains about 70 amino acid residues, and these repeats pack into a highly α-helical disk with a slight curvature. Ca 2+ fa- cilitates the binding of membrane phospholipids to the disk region of the annexins and the removal of Ca 2+ leads to the unbinding of the phospholipids. The N-terminal domain is located on the concave side of annexin, on the opposite side to the Ca 2+ -binding sites [7], and is more exible. Different annexins vary considerably in length, amino acid sequence and hydrophobicity of the N-terminal domain, which plays an important role in mediating the interaction of annexins with cytoplasmic proteins and membranes. Its diversity is the principal crite- rion for distinguishing the different annexin subfamilies [7,8]. Various reports indicate that annexins play signicant roles in autoimmune dis- eases, cardiovascular disease and cancers [914] and as a consequence, more attention has been recently paid to the relationship between annexins and diseases. 1.2. The structural features of Anxa11 Annexin A11 (Anxa11), also named Annexin XI, is a member of the group A annexins. The Anxa11 gene is located on human chromosome 10q22q23 and is composed of 15 exons and 14 introns without the 5anking region [15]. The N-terminal domain and C-terminal tetrad core repeat region of Anxa11 are encoded by exons 25 and 615, re- spectively [16]. Anxa11 is encoded by 504 amino acids and has a molec- ular weight of 56 kDa [17]. Anxa11 has three mRNA isoforms a, b and c due to alternative splicing; the a isoform is the most common one. Al- though all three isoforms of Anxa11 exist in humans, only one Anxa11 product is expressed [16]. Anxa11 is also a structural prototype and fun- damental functional model for other members based on the cladogram assay of annexin family. Clinica Chimica Acta 431 (2014) 164168 Corresponding author at: Department of Biotechnology, Dalian Medical University, 9 West Section Lvshun Southern Road, Dalian 116044, China. E-mail address: [email protected] (M.-Z. Sun). http://dx.doi.org/10.1016/j.cca.2014.01.031 0009-8981/© 2014 Elsevier B.V. All rights reserved. Contents lists available at ScienceDirect Clinica Chimica Acta journal homepage: www.elsevier.com/locate/clinchim

Annexin A11 in disease

Embed Size (px)

Citation preview

Page 1: Annexin A11 in disease

Clinica Chimica Acta 431 (2014) 164–168

Contents lists available at ScienceDirect

Clinica Chimica Acta

j ourna l homepage: www.e lsev ie r .com/ locate /c l inch im

Annexin A11 in disease

Jiasheng Wang a, Chunmei Guo a, Shuqing Liu b, Houbao Qi a, Yuling Yin b, Rui Liang c,Ming-Zhong Sun a,⁎, Frederick T. Greenaway d

a Department of Biotechnology, Dalian Medical University, Dalian 116044, Chinab Department of Biochemistry, Dalian Medical University, Dalian 116044, Chinac Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian 116027, Chinad Carlson School of Chemistry and Biochemistry, Clark University, Worcester, MA 01610, USA

⁎ Corresponding author at: Department of Biotechnol9 West Section Lvshun Southern Road, Dalian 116044, Ch

E-mail address: [email protected] (M.-Z. Sun).

http://dx.doi.org/10.1016/j.cca.2014.01.0310009-8981/© 2014 Elsevier B.V. All rights reserved.

a b s t r a c t

a r t i c l e i n f o

Article history:Received 27 November 2013Received in revised form 16 January 2014Accepted 18 January 2014Available online 6 February 2014

Keywords:Anxa11FunctionDiseaseCancer

Ubiquitously expressed in many cell types, annexin A11 (Anxa11) is a member of the multigene family of Ca2+-regulated phospholipid-dependent and membrane-binding annexin proteins. Studies have shown that Anxa11plays an important role in cell division, Ca2+ signaling, vesicle trafficking and apoptosis. The deregulation andmutation of Anxa11 are involved in systemic autoimmune diseases, sarcoidosis and the development,chemoresistance and recurrence of cancers. Malfunction of Anxa11may lead to or enhance the metastasis, inva-sion and drug resistance of cancers through the platelet-derived growth factor receptor (PDGFR) pathway and/orthe mitogen-activated protein kinase (MAPK)/p53 pathway. In a variety of diseases, Anxa11 is most commonlyreported to function through interactions with apoptosis-linked gene-2 protein (ALG-2) and/or calcyclin(S100A6). Although it has been little studied, Anxa11 is a promising biomarker for the diagnosis, treatmentand prognosis of certain diseases. In this review, the associations of Anxa11 with Ca2+-regulated exocytosis, cy-tokinesis, sex differentiation, autoimmune diseases, thrombolysis and cancers are summarized and interpreted.

© 2014 Elsevier B.V. All rights reserved.

1. Introduction

1.1. The features of annexins

The annexins are Ca2+-regulated phospholipid-binding proteinswidely expressed in all eukaryotic cells except yeasts [1,2]. The annexinshave been classified into five groups. Group A includes 12 members(annexins A1–A11 and A13; their chromosome locations are summa-rized in Table 1) found in vertebrates (mammalian); Group B, C, D andE annexins are found in non-vertebrates, fungi/molds, plants and pro-tists, respectively [3]. The annexins are cytosolic proteins distributedboth in the cellular cytoplasm and in the nuclear membranes. They ex-hibit diverse biological functions bymediating the interactions betweenproteins in cell membranes with other proteins in cells, in the nuclearmembrane, and in the extracellular matrix [4–7].

The annexins are composed of a conserved C-terminal core and avariable N-terminal domain [7]. The C-terminal core contains four ho-mologous repeat domains except for annexin A6, which has eight.Each annexin repeat contains about 70 amino acid residues, and theserepeats pack into a highlyα-helical diskwith a slight curvature. Ca2+ fa-cilitates the binding of membrane phospholipids to the disk region ofthe annexins and the removal of Ca2+ leads to the unbinding of the

ogy, Dalian Medical University,ina.

phospholipids. The N-terminal domain is located on the concave sideof annexin, on the opposite side to the Ca2+-binding sites [7], and ismore flexible. Different annexins vary considerably in length, aminoacid sequence and hydrophobicity of the N-terminal domain, whichplays an important role in mediating the interaction of annexins withcytoplasmic proteins andmembranes. Its diversity is the principal crite-rion for distinguishing the different annexin subfamilies [7,8]. Variousreports indicate that annexins play significant roles in autoimmune dis-eases, cardiovascular disease and cancers [9–14] and as a consequence,more attention has been recently paid to the relationship betweenannexins and diseases.

1.2. The structural features of Anxa11

Annexin A11 (Anxa11), also named Annexin XI, is a member of thegroup A annexins. The Anxa11 gene is located on human chromosome10q22–q23 and is composed of 15 exons and 14 introns without the5′ flanking region [15]. The N-terminal domain and C-terminal tetradcore repeat region of Anxa11 are encoded by exons 2–5 and 6–15, re-spectively [16]. Anxa11 is encoded by 504 amino acids and has amolec-ular weight of 56 kDa [17]. Anxa11 has three mRNA isoforms a, b and cdue to alternative splicing; the a isoform is the most common one. Al-though all three isoforms of Anxa11 exist in humans, only one Anxa11product is expressed [16]. Anxa11 is also a structural prototype and fun-damental functional model for other members based on the cladogramassay of annexin family.

Page 2: Annexin A11 in disease

Table 1The chromosomal locations of human Annexins.

Annexin Chromosome

Annexin A1 (Anxa1) 9q12–q21.2Annexin A2 (Anxa2) 15q21–q22Annexin A3 (Anxa3) 4q13–q22Annexin A4 (Anxa4) 2p13Annexin A5 (Anxa5) 4q28–q32Annexin A6 (Anxa6) 5q32–q34Annexin A7 (Anxa7) 10q21.1–q21.2Annexin A8 (Anxa8) 10q11.22Annexin A9 (Anxa9) 1q21Annexin A10 (Anxa10) 4q33Annexin A11 (Anxa11) 10q22-q23Annexin A13 (Anxa13) 8q24.13

165J. Wang et al. / Clinica Chimica Acta 431 (2014) 164–168

The Anxa11 N-terminal tail, which is important for the nuclear loca-tion and degradation of Anxa11 [18,19], is composed of 219 amino acidresidues and is rich in glycine, tyrosine and proline residues [20]. The C-terminus of Anxa11 contains the homologous tetrad annexin repeatcore and Ca2+ binding sites. The presence of Ca2+ is required for mem-brane binding, thermostability and the tertiary structure of Anxa11.Anxa11 binding with phosphatidylethanolamine, phosphatidylserineandphosphatidic acid is Ca2+-dependent. Binding of Anxa11 to Ca2+ in-creases its melting temperature by ~14 °C and α-helical content by 9%.The presence of Ca2+ affects the tertiary structure of Anxa11 by drivingfour tyrosine residues into a more hydrophobic environment [8]. TheAnxa11 N-terminal tail is critical for interaction with apoptosis-linkedgene-2 protein (ALG-2) and with calcyclin (S100A6) [21,22]. Anxa11can be phosphorylated by mitogen-activated protein kinase (MAPK)and platelet-derived growth factor (PDGF) although the phosphorylat-ed amino acid residues and the reaction mechanisms are not known[23,24]. In this review, we summarize the biological functions ofAnxa11, and its roles and potential action mechanisms of action indiseases.

Fig. 1. The potential biological actionmechanisms of Anxa11.(1) Anxa11may be a factor downsmay bind to S100A6 or ALG-2 to affect exocytosis and differentiation. (3) The interaction betwebalance among Anxa11, S100A6 and p53 may be essential and Anxa11 may act as an inhibitoactivity of p53, and results in cell survival, growth, apoptosis and differentiation and developm

2. The biological functions of Anxa11

2.1. Anxa11 and Ca2+-regulated exocytosis

Ca2+-regulated exocytosis regulates the release of hormones, diges-tive enzymes, immune modulators and neurotransmitters [25–27] andAnxa11 is an important factor in this process.

Several members of the annexin family have been found to beinvolved in vesicle trafficking and exocytosis [28–30]. Sjölin et al. firstdetected Anxa11 fragments in neutrophil-specific granules [31]. Frag-mental or intact Anxa11 protein was also detected in active granulesor insulin granules in a Ca2+-dependent manner by proteomics andIHC assays [32–34]. Although anti-Anxa11 antibody could not alter in-sulin release induced by calcyclin (S100A6) [35] in pancreatic β cells,Iino et al. have demonstrated that anti-Anxa11 antibody can inhibitCa2+-induced insulin release [34] in pancreatic β cells, as shown inFig. 1.

2.2. Anxa11 and cytokinesis

Cytokinesis is the final stage of the cell cycle occurring at the end ofnuclear division [36]. Themidbodyplays an important role in the abscis-sion step of cytokinesis [37]. Anxa11 has been reported to be involved incytokinesis by interrupting the formation of the midbody.

Tomas found Anxa11 to be involved in cell cycle progression [38].During progression of cell cycle, Anxa11 was co-localized with S100A6in the nuclear envelope during the prophase but not in other phasesfor A431 (epidermoid carcinoma) cells, and was finally translocated tothe midbody during cytokinesis [2]. Depletion of Anxa11 inhibitsmidbody formation in A431 cells, which causes the daughter cells tolink together and leads to cell apoptosis due to cytokinesis failure. In ad-dition, Anxa11 co-localizes with mitotic kinesin-like protein CHO1 incytokinesis [2]. How does Anxa11 influence cytokinesis? A mechanismhas been suggested whereby Ca2+-activated tyrosine kinase activatesAnxa11 and the resulting activated-Anxa11 interacts with S100A6

tream fromMAPK that exerts its biological functions as a phosphorylated form. (2) Anxa11en Anxa11 and ALG-2 is possibly involved in or interferes with the caspase pathway. (4) Ar that interferes with interactions between S100A6 and p53, mediates the transcriptionalent of cancer cells.

Page 3: Annexin A11 in disease

166 J. Wang et al. / Clinica Chimica Acta 431 (2014) 164–168

leading to microtubule-induced folds that serve an initiating role in nu-clear envelope breakdown [38]. Anxa11 co-localizes with CHO1 in cyto-kinesis [2], suggesting that these two proteins, both indispensable formidbody formation, are in some way functionally linked. Both the sep-arate and joint molecular functions of Anxa11 and CHO1 in cytokinesisare worthy of further study.

2.3. Anxa11 and sex differentiation

Sex differentiation is a complex process of developing a functionaltestis or ovary from a bipotential vertebrate gonad. Sry is the Y-chromosomal gene pivotal in mammalian sex determination [39,40].Anxa11 participates in the sex differentiation processmainly by interac-tion with S100A6.

Anxa11mRNA levels in the developing gonadhave been analyzed bythe whole-mount in situ hybridization (WISH) technique. Sry only ex-presses during 10.5 to 12.5 days post-coitum (dpc), and the expressionof SrymRNA reaches the highest levels at 11.5 dpc [39,41]. Interestingly,following high Sry expression Anxa11 is upregulated inmales but not infemales until 13 dpc, and after 11.5 dpc the differential expression ofAnxa11 between females and males becomes increasingly significant.It becomes increasingly abundant in the developing testis, while it isprogressively lost in the female. IHC revealed that Anxa11 was morehighly expressed in testis cords but not at all in the ovary at 12.5 dpc. In-terestingly, S100A6 expressionwas also detected in proliferating cells ofembryonic testis, supporting a possible interaction of S100A6 withAnxa11 in vivo [39]. The functional mechanism of Anxa11 in sex differ-entiation is illustrated in Fig. 1.

3. The association of Anxa11 with disease

3.1. Anxa11 is associated with autoimmune diseases

Autoimmune diseases (ADs) result from abnormal B/T lymphocyterecognition of self-reactive antigens, which leads to high levels of auto-antibodies in patients causing inflammation and irreversible structuraland functional damage [42,43]. Anxa11 is reported to be associatedwith ADs [44–47].

Using Hela or Raji cell extracts as substrates, anti-Anxa11 antibodieswere detected in the serum of patients with SLE (systemic lupus erythe-matosus), undifferentiated connective tissue disease, rheumatoid ar-thritis and APS (anti-phospholipid togliere antibodies syndrome)[44–47], which indicates the implication of anti-Anxa11 antibodies inADs. But why are anti-Anxa11 antibodies present in the sera of patientswith AD? As yet there is no direct evidence indicating the regulationmechanisms of Anxa11 in AD. Apoptosis has been hypothesized as animportant process for AD, so based on the previously summarized re-sults that Anxa11 can bind to ALG-2 and S100A6, two key factors impli-cated in the stimulation of apoptosis process [48,49], we propose thatapoptosis bodies in the lesions captured by APCs (antigen-presentingcells) produce Anxa11 autoantigens.

Anxa11 has been identified as a new susceptibility locus for sarcoid-osis, a rare genetic AD [50–54]. A common non-synonymous SNP,rs10449550, has been found to be closely associated with the biologicalfunctions of Anxa11. An R230C mutation (Anxa11R230C) located at thefirst annexin repeat of Anxa11 strongly correlated with sarcoidosis.The effect of the mutation of the conserved R230 of Anxa11 in sarcoid-osis was interpreted using annexin V as a study model. Mutation of theconserved Arg 45 localized in the first annexin repeat of annexin V, as inAnxa11, led to greater sensitivity to proteolytic digestion and lowerphospholipid affinity in comparisonwith thewild type [18,55], suggest-ing the likelihood of a similar effect of the conserved Arg residue onAnxa11 function in sarcoidosis. In addition, themRNAandprotein levelsof Anxa11 were significantly downregulated when activating stimuliwere given in two immune cells important in sarcoidosis, CD8+ T andCD19+ B cells [54]. This suggests that genetic instability and mutation

of a key residue in Anxa11 might contribute to the development andprogression of sarcoidosis through immunocytes.

3.2. Anxa11 is associated with thrombolysis

Acute myocardial infarction and ischemic strokes remain two lead-ing causes of death throughout the world. Thrombolytic therapy is animportant treatment for them but certain disadvantages reduce thetherapeutic efficacy of available thrombolytic agents [56,57]. Anxa11has proved to have potential as an adjuvant therapeutic promotingthrombolytic efficiency.

SAK (staphylokinase) is a promising blood clot dissolving agent.However, clinical trials have already proved that it is a good fibrin-specific agent and it has been used as a clotting component in chimeras[56,58]. Chiou et al. found that the fusion protein SAK–Anxa11 pos-sessed a similar efficiency in plasma clot lysis as SAK, but a higherefficiency in retarding clot formation. SAK–Anxa11 dissolved bothplatelet rich plasma (PRP) clots and platelet poor plasma (PPP) clotswith an efficiency similar to SAK, but SAK–Anxa11 showed a strongereffect in dose-dependent extension of clotting time. It has been sug-gested that the long N-terminal tail of Anxa11 probably serves as a nat-ural linker to enable twomoieties to function properly in a complicatedmicroenvironmentwhen they are still linked [14]. Anxa11 could bind toan important pro-coagulant, phosphatidyl-L-serine (PS) [59,60]. Thus,the SAK–Anxa11 chimera could suppress the accumulation of coagula-tion enzyme complexes on platelets and enhance the clot resolvingefficiency.

4. Anxa11 and cancers

4.1. Anxa11 is associated with ovarian cancer recurrence anddrug resistance

Ovarian cancer is the leading cause of death due to gynecological tu-mors [61]. The combination of surgery and chemotherapy is the basictreatment for ovarian cancer, but chemoresistance is always a consider-able treatment problem during chemotherapy [62,63].

Anxa11 is associated with recurrence and chemoresistance of ovari-an cancer. Antibody microarray and immunoblotting results have indi-cated that Anxa11 is down-regulated 3–8 folds in cisplatin-resistantovarian cancer cell lines 2008/C13*5.25, HEY C2 and A2780cis com-pared to cisplatin-sensitive 2008, HEY and A2780 lines, respectively.Consistent with these results, the resistance of 2008 cells to cisplatin in-creased 2.6-fold after Anxa11 knockdown (P b 0.01) [11]. HigherAnxa11 expression levels seemed to prolong the disease-free intervalof patients. IHC analysis showed that Anxa11 levels in recurrent ovariancancers were much lower than in primary ovarian cancers and its levelwas inversely correlated with the recurrence and cisplatin resistance ofovarian cancer [11,63]. These results implicate Anxa11 down-regulationas a characteristic for cisplatin-resistant ovarian cancer cells and a con-tributor to tumor recurrence. Thus Anxa11 may be a potential markerfor chemo-resistance and earlier recurrence in ovarian cancer patients.

P53 is a common transcriptional factor associatedwith cancer devel-opment and chemo-resistance [64]. Anxa11 might influence ovariancancer development and drug resistance by way of p53. Anxa11 canbind to S100A6 in the presence of Ca2+, and S100A6 can bind to p53in a Ca2+-dependent manner, which inhibits the ubiquitylation of p53byMDM2 (p53 tumor suppressor) and enhances p53 transcriptional ac-tivity [65]. Although no direct relationship has yet been reported be-tween advanced ovarian cancer and chemoresistance, both p53 andS100A6 are up-regulated in advanced ovarian cancers [66–68]. There-fore, the dynamic balance among Anxa11, S100A6 and p53 should bestrictly controlled. Anxa11 may act as an inhibitor interfering with theinteraction of S100A6 and p53. Down-regulation of Anxa11 probably in-creases the binding capacity of S100A6 to p53, resulting in enhanced

Page 4: Annexin A11 in disease

Fig. 2. The relationship between Anxa11 and cancers.

167J. Wang et al. / Clinica Chimica Acta 431 (2014) 164–168

p53 transcriptional activation and leading to increased chemoresistanceof ovarian cancers.

4.2. Anxa11 up-regulation enhances the progression of breast cancer

Breast cancer (BC) is the most common cancer in women. It affectsover one million women globally, accounting for over 400,000 deathsannually [69].

Anxa11 up-regulation is positively correlated with the malignancyof BC. Using T7 phage cDNA library assay, Tang et al. revealed that theN-terminal 41–74 amino acid sequence of Anxa11 was positivelyexpressed in 60% of sera of patients with DCIS (ductal carcinoma insitu, the earliest formof BC) and 7% of sera of patientswith IDC (invasiveductal carcinoma) [70]. Furthermore, Anxa11 protein was up-regulatedtwo fold in malignant BC tissues compared with non-malignant BC tis-sues. Interestingly, p53 was also up-regulated 2.15-fold in malignantBC tissues. The basis of why Anxa11 affects BC progression might beconnected with p53 [71]. Anxa11 can be phosphorylated by MAPKand the downstream effectors of MAPK kinase 3 (MKK3) and MAPK ki-nase 7 (MKK7) were also found to be over-expressed in malignant BC[23,72]. Considering the fact that the MKK3/p38 and MKK7/JNK path-ways can regulate p53 transcription activity in BC, the over-expressionof Anxa11 is likely to contribute to p53 activity.

Butwhy can Anxa11 autoantibody or Anxa11 fragments be detectedin sera of invasive BC patients? It has been proposed that overexpres-sion of tumor-associated antigens (TAAs) in specific tissues may leadto a loss of self-tolerance, resulting in the genesis of autoantibodies orfragments [73]. So far no studies of Anxa11 and TAAs have been report-ed, and the role of Anxa11 in BC progression definitely deserves furtherattention.

4.3. Anxa11 is associated with metastasis and drug resistance of colorectalcancer

Colorectal cancer (CRC) is the fourth most common cancer world-wide [74] and is an increasing clinical and public health challenge.CRC metastasis has a great effect on patients' survival. Anxa11 hasbeen reported to be associatedwith CRCmetastasis [75–77], suggestingthat it is of potential use as a diagnostic and prognostic indicator for CRC.

Comparative proteomics and IHC have shown that Anxa11 isoverexpressed in tissues of primary CRC patients compared to normaltissues (P b 0.001). The up-regulation of Anxa11 significantly correlatedwith the presence of advanced tumors but, interestingly, Anxa11 wassignificantly decreased in CRC tissues with lymph node metastasis(LNM) compared to CRC tissues without LNM (P = 0.01). It is clearthat Anxa11 plays an important role in the development andmetastasisof CRC [76] and may have potential as a therapeutic target for CRC.

Mutation of Anxa11 is associatedwith drug-resistance of CRC.Muta-tion of the basic residue, 230Arg, to the polar residue, Cys (Anxa11R230C),was made with SNP rs1049550 resulting in an increase in the overall re-sponse rate of metastatic CRC patients with bevacizuma (an angiogene-sis inhibitor) treatment by ~200% compared with patients with wildtype Anxa11. It has been proposed that Anxa11R230C plays an importantrole in angiogenesis by influencing its chemosensitivity [12,77], and thatits level is associated with CRC chemoresistance. This might be associat-edwith an altered ability of cancer cells to break through the ECM. Com-pared to wild type Anxa11-transfected PKO CRC cells (without Anxa11expression), Anxa11R230C-transfected PKO CRC cells showed reducedMMP-9 expression following treatment with a combined regimen of5-FU, leucovorin, irinotecna and bevacizumab [12]. Like its effect ofAnxa11R230C in sarcoidosis [55], the mutation of Arg230 of Anxa11might exert this action in CRC cells by altering its sensitivity to proteolyt-ic digestion and its phospholipid binding affinity. Thus, although there isno direct evidence so far, we postulate that Anxa11 may interfere withdrug sensitivity in CRC metastasis by way of the MAPK pathway,S100A6 or ALG-2.

5. Conclusion and perspectives

Anxa11 is a Ca2+-regulated phospholipid-binding protein. It playsimportant roles in cytokinesis, apoptosis, sex differentiation, exocytosisand cancers. The dysregulation and mutation of Anxa11 are involved insystemic autoimmune diseases and sarcoidosis, and are associated withthe development, chemoresistance and recurrence of cancers. The evi-dence for and potential mechanisms of action of Anxa11 in diseasehave been summarized and are illustrated in Figs. 1 and 2. In summary,results indicate that Anxa11 is involved in the metastasis, invasion anddrug resistance of cancers through the PDGFR andMAPK/p53 pathways.ALG-2 and/or S100A6 are the molecules most frequently reported to beassociated with Anxa11 in diseases. We suggest that Anxa11 may be auseful indicator for the diagnosis, treatment and prognosis of certaindiseases.

Acknowledgements

This work was supported by grants from the National NaturalScience Foundation of China (81171957; 81272186; 81050010), theDistinguished Young Scholars of Liaoning College and University(LJQ2011094), the Liaoning BaiQianWan Talent Project (2012921015)and the Key Laboratory of the Department of Education of Liaoning(LS2010050).

References

[1] Gerke V, Creutz CE, Moss SE. Annexins: linking Ca2+ signalling to membranedynamics. Nat Rev Mol Cell Biol 2005;6(6):449–61.

[2] Tomas A, Futter C, Moss SE. Annexin 11 is required for midbody formation andcompletion of the terminal phase of cytokinesis. J Cell Biol 2004;165(6):813–22.

[3] Mussunoor S, Murray GI. The role of annexins in tumor development and progres-sion. J Pathol 2008;216(2):131–40.

[4] Moss SE, Morgan RO. The annexins. Genome Biol 2004;5(4):219.[5] Perron B, Lewit-Bentley A, Geny B, et al. Can enzymatic activity, or otherwise, be in-

ferred from structural studies of annexin III? J Biol Chem 1997;272(25):11321–6.[6] Rosenbaum S, Kreft S, Etich J, et al. Identification of novel binding partners

(annexins) for the cell death signal phosphatidylserine and definition of their recog-nition motif. J Biol Chem 2011;286(7):5708–16.

[7] Gerke V, Moss SE. Annexins: from structure to function. Physiol Rev2002;82(2):331–71.

[8] Lecona E, Turnay J, Olmo N, et al. Structural and functional characterization of re-combinant mouse annexin A11: influence of calcium binding. Biochem J2003;373(Pt 2):437–49.

[9] Bastian BC. Annexins in cancer and autoimmune diseases. Cell Mol Life Sci1997;53(6):554–6.

Page 5: Annexin A11 in disease

168 J. Wang et al. / Clinica Chimica Acta 431 (2014) 164–168

[10] Zhao SH, Pan DY, Zhang Y, et al. Annexin A2 promotes choroidal neovascularizationby increasing vascular endothelial growth factor expression in a rat model of argonlaser coagulation-induced choroidal neovascularization. Chin Med J (Engl) 2010;123(6):713–21.

[11] Song J, Shih leM, Salani R, et al. Annexin XI is associatedwith cisplatin resistance andrelated to tumor recurrence in ovarian cancer patients. Clin Cancer Res 2007;13(22):6842–9.

[12] Kim JC, Kim SY, Cho DH, et al. Novel chemosensitive single-nucleotide polymor-phism markers to targeted regimens in metastatic colorectal cancer. Clin CancerRes 2011;17(5):1200–9.

[13] Hayes MJ, Moss SE. Annexins and disease. Biochem Biophys Res Commun2004;322(4):1166–70.

[14] Chiou JF, Woon MD, Cheng SN, et al. Staphylokinase-annexin X1 chimera exhibitedefficient in vitro thrombolytic activities. Biosci Biotechnol Biochem 2007;71(5):1122–9.

[15] Morgan RO, Bell DW, Testa JR, et al. Genomic locations of ANX11 and ANX13 and theevolutionary genetics of human annexins. Genomics 1998;48(1):100–10.

[16] Bances P, Fernandez MR, Rodriguez-Garcia MI, et al. Annexin A11 (ANXA11) genestructure as the progenitor of paralogous annexins and source of orthologouscDNA isoforms. Genomics 2000;69(1):95–103.

[17] Misaki Y, Pruijn GJ, van der Kemp AW, et al. The 56K autoantigen is identical tohuman annexin XI. J Biol Chem 1994;269(6):4240–6.

[18] Mizutani A, Watanabe N, Kitao T, et al. The long amino-terminal tail domain ofannexin XI is necessary for its nuclear localization. Arch Biochem Biophys1995;318(1):157–65.

[19] Barnes JA, Gomes AV. Proteolytic signals in the primary structure of annexins. MolCell Biochem 2002;231(1–2):1–7.

[20] Fatimathas L, Moss SE. Characterization of the sarcoidosis-associated variant ofannexin A11. Gen Physiol Biophys 2009;28:F29–38.

[21] Sudo T, Hidaka H. Regulation of calcyclin (S100A6) binding by alternative splicingin the N-terminal regulatory domain of annexin XI isoforms. J Biochem 1998;273(11):6351–7.

[22] Satoh H, Nakano Y, Shibata H, et al. The penta-EF-hand domain of ALG-2 interactswith amino-terminal domains of both annexin VII and annexin XI in a Ca2+-dependent manner. Biochim Biophys Acta 2002;1600(1–2):61–7.

[23] Mizutani A, Tokumitsu H, Kobayashi R, et al. Phosphorylation of annexin XI (CAP-50) in SR-3Y1 cells. J Biol Chem 1993;268(21):15517–22.

[24] Furge LL, Chen K, Cohen S. Annexin VII and annexin XI are tyrosine phosphorylatedin peroxovanadate-treated dogs and in platelet-derived growth factor-treated ratvascular smooth muscle cells. J Biol Chem 1999;274(47):33504–9.

[25] Neher E, Sakaba T. Multiple roles of calcium ions in the regulation of neurotransmit-ter release. Neuron 2008;59(6):861–72.

[26] Bergmeier W, Weidinger C, Zee I, et al. Emerging role of store-operated Ca(2+)entry through STIM and ORAI protein in immunity, hemostasis and cancer. Channels2013;7(5):1–13.

[27] Gandini MA, Sandoval A, González-Ramírez R, et al. Functional coupling of Rab3-interacting molecule 1 (RIM1) and L-type Ca2+ channels in insulin release. J BiolChem 2011;286(18):15757–65.

[28] Creutz CE, Pazoles CJ, Pollard HB. Identification and purification of an adrenal med-ullary protein (synexin) that causes calcium-dependent aggregation of isolatedchromaffin granules. J Biol Chem 1978;253(8):2858–66.

[29] Zaks WJ, Creutz CE. Ca(2+)-dependent annexin self-association on membrane sur-faces. Biochemistry 1991;30(40):9607–15.

[30] Meers P, Bentz J, Alford D, et al. Synexin enhances the aggregation rate but not thefusion rate liposomes. Biochemistry 1988;27(12):4430–9.

[31] Sjölin C, Dahlgren C. Isolation by calcium-dependent translation to neutrophil-specific granules of a 42-kD cytosolic protein, identified as being a fragment ofannexin XI. Blood 1996;87(11):4817–23.

[32] Sjölin C, Movitz C, Lundqvist H, et al. Translocation of annexin XI to neutrophil sub-cellular organelles. Biochim Biophys Acta 1997;1326(1):149–56.

[33] BoussacM, Garin J. Calcium-dependent secretion in human neutrophils: a proteomicapproach. Electrophoresis 2000;21(3):665–72.

[34] Iino S, Sudo T, Niwa T, et al. Annexin XI may be involved in Ca2+- or GTP-γS-inducedinsulin secretion in the pancreatic β-cell. FEBS Lett 2000;479(1–2):46–50.

[35] Okazaki K, Niki I, Iino S, et al. A role of calcyclin, a Ca2+-binding protein, on the Ca2+-dependent insulin release from the pancreatic β-cell. J Biol Chem 1994;269(8):6149–52.

[36] Sagona AP, Stenmark H. Cytokinesis and cancer. FEBS Lett 2010;584(12):2652–61.[37] McCollum D. Cytokinesis: breaking the ties that bind. Curr Biol 2005;15(24):

R998-1000.[38] Tomas A,Moss SE. Calcium- and cell cycle-dependent association of annexin 11with

the nuclear envelope. J Biol Chem 2003;278(22):20210–6.[39] Williams LH,McClive PJ, VanDen Bergen JA, et al. Annexin XI co-localiseswith calcyclin

in proliferating cells of the embryonic mouse testis. Dev Dyn 2005;234(2):432–7.[40] Haugen T, Almeida FF, Andersson E, et al. Sex differentiation in Atlantic cod (Gadus

morhua L.): morphological and gene expression studies. Reprod Biol Endocrinol2012;10:47.

[41] Hacker A, Capel B, Goodfellow P, et al. Expression of Sry, the mouse sex determininggene. Development 1995;121(6):1603–14.

[42] Salinas GF, Braza F, Brouard S, et al. The role of B lymphocytes in the progressionfrom autoimmunity to autoimmune disease. Clin Immunol 2013;146(1):34–45.

[43] Elkon K, Casali P. Nature and functions of autoantibodies. Nat Clin Pract Rheumatol2008;4(9):491–8.

[44] van Venrooij WJ, Wodzig KW, Habets WJ, et al. Anti-56K: a novel, frequently occur-ring autoantibody specificity in connective tissue diseases. Clin Exp Rheumatol1989;7(3):277–82.

[45] Jorgensen CS, Levantino G, Houen G, et al. Determination of autoantibodies toannexin XI in systemic autoimmune diseases. Lupus 2000;9(7):515–20.

[46] Misaki Y, Van Venrooij WJ, Pruijn GJ. Prevalence and characteristic of anti-56K/annexin XI autoantibodies in systemic autoimmune diseases. J Rheumatol1995;22(1):97–102.

[47] Iaccarino L, Ghirardello A, Canova M, et al. Anti-annexins autoantibodies: their roleas biomarkers of autoimmune diseases. Autoimmun Rev 2011;10(9):553–8.

[48] Rao RV, Poksay KS, Castro-Obregon S, et al. Molecular components of a cell deathpathway activated by endoplasmic reticulum stress. J Biol Chem 2004;279(1):177–87.

[49] Joo JH, Yoon SY, Kim JH, et al. S100A6 (calcyclin) enhances the sensitivity to apopto-sis via the upregulation of caspase-3 activity in Hep3B cells. J Cell Biochem2008;103(4):1183–97.

[50] Levin AM, Iannuzzi MC, Montgomery CG, et al. Association of ANXA11 genetic vari-ation with sarcoidosis in African Americans and European Americans. Genes Immun2013;14(1):13–8.

[51] Adrianto I, Lin CP, Hale JJ, et al. Genome-wide association study of African andEuropean Americans implicates multiple shared and ethnic specific loci in sarcoido-sis susceptibility. PLoS One 2012;7(8):e43907.

[52] Li Y, Pabst S, Kubisch C, et al. First independent replication study confirms the stronggenetic association of ANXA11 with sarcoidosis. Thorax 2010;65(10):939–40.

[53] Page M. Nursing care and management of patients with sarcoidosis. Br J Nurs2008;17(4):252–7.

[54] Hofmann S, Franke A, Fischer A, et al. Genome-wide association study identifiesANXA11 as a new susceptibility locus for sarcoidosis. Nat Genet 2008;40(9):1103–6.

[55] Campos B, Wang S, Retzinger GS, et al. Mutation of highly conserved arginine resi-dues disrupts the structure and function of annexin V. Arch Med Res 1999;30(5):360–7.

[56] Wang M, Wang Y, Wang J, et al. Construction and characterization of a novelstaphylokinase variant with thrombin-inhibitory activity. Biotechnol Lett2009;31(12):1923–7.

[57] Jariwala P, Chandra S. Diagnosis and management of failed thrombolytic therapy foracute myocardial infarction. Indian Heart J 2010;62(1):21–8.

[58] Collen D, Vanderschueren S, Van de Werf F. Fibrin-selective thrombolytic therapywith recombinant staphylokinase. Haemostasis 1996;26(Suppl. 4):294–300.

[59] Raynal P, Pollard HB. Annexins: the problem of assessing the biological role for agene family of multifunctional calcium- and phospholipid-binding proteins. BiochimBiophys Acta 1994;1197:63–93.

[60] Chen HH, Vicente CP, He L, et al. Fusion proteins comprising annexin V and Kunitzprotease inhibitor are highly potent thrombogenic site-directed anticoagulants.Blood 2005;105(10):3902–9.

[61] Eltabbakh GH, Awtrey CS. Current treatment for ovarian cancer. Expert OpinPharmacother 2001;2(1):109–24.

[62] Kim A, Ueda Y, Naka T, et al. Therapeutic strategies in epithelial ovarian cancer. J ExpClin Cancer Res 2012;31:14.

[63] Song J, Shih I, Chan DW, et al. Suppression of annexin A11 in ovarian cancer: impli-cations in chemoresistance. Neoplasia 2009;11(6):605–14.

[64] Levine AJ, Oren M. The first 30 years of p53: growing ever more complex. Nat RevCancer 2009;9(10):749–58.

[65] Słomnicki ŁP, Nawrot B, Leśniak W. S100A6 binds p53 and affects its activity. Int JBiochem Cell Biol 2009;41(4):784–90.

[66] Godoy H, Mhawech-Fauceglia P, Beck A, et al. Expression of poly (adenosinediphosphate- ribose) polymerase and p53 in epithelial ovarian cancer andtheir role in prognosis and disease outcome. Int J Gynecol Pathol 2011;30(2):139–44.

[67] Wei BR, Hoover SB, Ross MM, et al. Serum S100A6 concentration predicts peritonealtumor burden in mice with epithelial ovarian cancer and is associated with ad-vanced stage in patients. PLoS One 2009;4(10):e7670.

[68] Itamochi H, Kigawa J. Clinical trials and future potential of targeted therapy for ovar-ian cancer. Int J Clin Oncol 2012;17(5):430–40.

[69] Iorio MV, Casalini P, Piovan C, et al. Breast cancer and microRNAs: therapeuticimpact. Breast 2011;20(Suppl. 3):S63–70.

[70] Fernández-Madrid F, Tang N, Alansari H, et al. Autoantibodies to annexin XI-A andother autoantigens in the diagnosis of breast cancer. Cancer Res 2004;64(15):5089–96.

[71] Hudelist G, Pacher-Zavisin M, Singer CF, et al. Use of high-throughput protein arrayfor profiling of differentially expressed proteins in normal and malignant breast tis-sue. Breast Cancer Res Treat 2004;86(3):281–91.

[72] Kim ES, Jeong JB, Kim S, et al. The G12 family proteins upregulate matrixmetalloproteinase-2 via p53 leading to human breast cell invasion. Breast CancerRes Treat 2010;124(1):49–61.

[73] Nolen BM, Lokshin AE. Autoantibodies for cancer detection: still cause for excite-ment? Cancer Biomark 2010;6(5–6):229–45.

[74] Feng S, Xiong L, Ji Z, et al. Correlation between increased copy number of mitochon-drial DNA and clinicopathological stage in colorectal cancer. Oncol Lett 2011;2(5):899–903.

[75] Rahbari NN, Bork U, Motschall E, et al. Molecular detection of tumor cells in regionallymph nodes is associated with disease recurrence and poor survival in node-negative colorectal cancer: a systematic review and meta-analysis. J Clin Oncol2012;30(1):60–70.

[76] Duncan R, Carpenter B, Main LC, et al. Characterisation and protein expression pro-filing of annexins in colorectal cancer. Br J Cancer 2008;98(2):426–33.

[77] Kim JC, Ha YJ, Roh SA, et al. Feasibility of proposed single-nucleotide polymorphismsas predictive markers for targeted regimens in metastatic colorectal cancer. Br JCancer 2013;108(9):1862–9.