Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Dec 20.
Published in final edited form as: Semin Thromb Hemost. 2013 Mar 12;39(4):10.1055/s-0033-1334143. doi: 10.1055/s-0033-1334143

Annexin A2 System in Human Biology: Cell Surface and Beyond

Min Luo 1, Katherine A Hajjar 1
PMCID: PMC3869233  NIHMSID: NIHMS537707  PMID: 23483454

Abstract

Annexin A2 (A2) is a multicompartmental, multifunctional protein that orchestrates a growing spectrum of biologic processes. At the endothelial cell surface, A2 and S100A10 (p11) form a heterotetramer, which accelerates tissue plasminogen activator–dependent activation of the fibrinolytic protease, plasmin. In antiphospholipid syndrome, anti-A2 antibodies are associated with clinical thrombosis, whereas overexpression of A2 in acute promyelocytic leukemia promotes hyperfibrinolytic bleeding. A2 is upregulated in hypoxia, and mice deficient in A2 are resistant to oxygen-induced retinal neovascularization, suggesting a role for A2 in human retinal vascular proliferation. In solid malignancies, the (A2•p11)2 tetramer may promote cancer cell invasion, whereas in multiple myeloma A2 enables malignant plasmacyte growth and predicts prognosis. In the central nervous system, the p11 enables membrane insertion of serotonin receptors that govern mood. In the peripheral nervous system, p11 directs sodium channels to the plasma membrane, enabling pain perception. In cerebral cortex neurons, A2 stabilizes the microtubule-associated tau protein, which, when mutated, is associated with frontotemporal dementia. In inflammatory dendritic cells, A2 maintains late endosomal/lysosomal membrane integrity, thus modulating inflammasome activation and cytokine secretion in a model of aseptic arthritis. Together, these findings suggest an emerging, multifaceted role for A2 in human health and disease.

Keywords: annexin A2, S100A2/protein p11, fibrinolysis, inflammation

Cell Biology of Annexin A2

Annexin Family of Proteins

The annexins are an evolutionarily ancient and conserved family of Ca2+-regulated phospholipid-binding proteins.1 Annexin proteins, which have existed for over 500 million years, typically possess two main structural domains. The core domain, usually 30 to 35 kDa in mass, contains four α-helical “annexin” repeats that bind Ca2+ and form a convex face that can associate with membrane phospholipid. The more hydrophilic amino-terminal “tail” or “interaction” domains are essentially unique to each family member and allow the annexins to partner with a wide variety of protein ligands. The term “annexinopathy,” first coined in 19992 and expanded in more recent reviews,3-7 describes the mechanistic role played by the annexins in human disease.

Of the 12 annexins (annexins A1 to A11 and A13) expressed in humans, annexin A2 (A2) is among the most extensively studied, especially with respect to mammalian biology and human disease.8 A 36-kDa protein produced by endothelial cells, monocytes, macrophages, dendritic cells, trophoblast cells, epithelial cells, tumor cells, and many others, A2 can exist as a free monomer in the cytoplasm, in association with intracellular membranes, or tethered to the external face of the plasma membrane.9,10 Human A2 is the product of the ANXA2 gene, composed of 13 exons distributed over 40 kb of genomic DNA on chromosome 15 (15q21).11 Versions of A2 protein among mammalian species are ~98% identical at the amino acid level.

Protein S100A10/p11 and (A2•p11)2 Complex Formation

Protein S100A10, often designated p11, is a recognized binding partner of A2.12,13 p11 belongs to the S100 family of proteins by virtue of its solubility in 100% ammonium sulfate at neutral pH and its Ca2+-binding helix-loop-helix motifs. p11 endows A2 with increased phospholipid binding affinity. Although most S100 proteins, in response to Ca2+, undergo a conformational change that allows them to associate with target proteins, p11 exists in a permanent “calcium-on” state, due to two key amino acid substitutions at positions E65 and D56 within its Ca2+-binding domain.14,15 Within the (A2•p11)2 heterotetramer, p11 exists as a noncovalently linked homodimer; together the N-terminal HI and C-terminal HIV helices of adjacent p11 molecules form a groove, which is occupied by the α-helical N-terminal 14 amino acids of A2.15 p11 appears to be stabilized by its interaction with partner proteins such as A2.

Subcellular Localization of Annexin A2

A2 is a multicompartmental protein, which, like other family members, fulfills a spectrum of membrane organizing functions. The available evidence indicates that A2 consolidates membrane microdomains, recruits specialized membrane proteins, regulates membrane fusion events, and participates in membrane repair.9 Although heterotrameric (A2•p11)2 assembles fibrinolytic proteases on the extracellular face of the plasma membrane, monomeric A2 remains soluble in the cytoplasm of cells at resting Ca2+ concentration. The subcellular and organellar localization of A2 appears to be governed by a multitude of factors that include Ca2+ concentration, pH, membrane phospholipid composition, post-translational modifications, and the availability of other proteins.

Like many annexins, A2 orchestrates a wide range of membrane trafficking events.9 A2 promotes Ca2+-regulated exocytosis, including Ca2+-induced release of Weibel-Palade body proteins,16 chromaffin granules,17 and surfactant-containing lamellar bodies.18 A2 possesses an endosome targeting sequence, and its depletion within cells leads to abnormal morphology of recycling endosomes.19 A2 appears to be involved in the biogenesis of multivesicular bodies and was the fifth most frequently cited protein among 140 proteins identified in 19 different proteomic studies of exosomes.20-22 A2 possesses a nuclear export signal within residues 3 to 12, which overlap with the p11 binding site; when A2 is overexpressed, its translocation from nucleus to cytoplasm is subject to inhibition by leptomycin B.20 Because A2 binds to specific messenger RNAs via its fourth core domain repeat, it may serve to escort specific RNAs from the nucleus to specific subcellular locations for localized protein synthesis.21 The actin and microfilament interactions of A2 are complex and appear to be connected with many of these functions.22 How these multiple activities may relate to human health and disease, however, is largely unknown.

Annexin A2 and p11 Dynamics at the Cell Surface

The cell surface is a discrete compartment for expression of both A2 and protein p11.23-25 Here, especially on vascular endothelial cells, the (A2•p11)2 tetramer serves as an assembly site for plasminogen and tissue plasminogen activator (tPA), an endothelial cell secretory product. Cleavage of the R560-V561 peptide bond of plasminogen gives rise to the active, primary fibrinolytic protease, plasmin.26-28 This membrane-oriented reaction accelerates the catalytic efficiency of tPA-dependent plasminogen activation by up to 2 log orders of magnitude. In classic fibrinolysis, on the other hand, activation of plasminogen by tPA is even more dramatically accelerated in the presence of fibrin, which serves as a cofactor for its own destruction.26 Thus, the A2-based fibrinolytic system may serve as a protective, surveillance mechanism for fibrin clearance, whereas the more potent classic fibrin-based system may be more important in clearance of already formed fibrin thrombi.

Cell surface expression of the (A2•p11)2 tetramer is a dynamic process, and translocation of the complex from the cytoplasm to the outer leaflet of the endothelial cell’s plasma membrane appears to be a key regulatory step in fibrinolysis.23,24 In nonendothelial cells, the cell surface appearance of A2 has been linked to plasma membrane fusion of multivesicular bodies in NIH 3T3 fibroblasts,29 to exocytosis of secretory granules in chromaffin cells,30 to fusion of secretory vesicles with the plasma membrane in porcine intestinal epithelial cells,31 and to caspase-1 activation in stimulated macrophages or ultraviolet-irradiated keratinocytes.32 Still, the exact mechanism by which endothelial cells regulate surface (A2•p11)2 remains unclear.

Endothelial cell translocation of A2 to the cell surface is initiated by several factors, including heat stress, thrombin stimulation, and hypoxia, and can occur within minutes.33-35 A2 was originally identified as an src kinase substrate,36 and its translocation is driven by phosphorylation at Y23 by a pp60src-like kinase.34 In the endothelial cell, translocation also requires expression of p11, which is stabilized by A2.34 Upon binding to p11, A2 masks a critical “degron,” or polyubiquitination site on p11, so that in the absence of sufficient A2, p11 is polyubiquitinated and targeted to the proteasome for degradation.37 In AnxA2−/− mice, which demonstrate low to nondetectable p11 expression in most cells, treatment with bortezomib, a proteasome inhibitor, restored p11 expression in lung tissue, thus verifying that p11 is regulated via polyubiquitination in vivo.

Formation of the (A2•p11)2 heterotetramer appears to be regulated by protein kinase C.38,39 Protein kinase C phosphorylates S11 or S25 residues within the N-terminal tail domain of intracellular A2, thereby dissociating the complex and preventing further translocation to the cell surface. Dissociation of p11 from the complex allows it to be polyubiquitinated and destroyed in the proteasome.38 Serine phosphorylation of A2 by protein kinase C appears to be initiated by plasmin, which, once generated at the cell surface, cleaves A2, activates toll-like receptor 4, and signals conventional protein kinase C–mediated phosphorylation of intracellular A2. Through this negative feedback mechanism, plasmin may regulate its own formation.

Annexin A2 at the Cell Surface

Thrombosis and Vascular Occlusion

Increasing evidence shows that the A2 system functions at the cell surface to regulate hemostasis in vivo (►Fig. 1A). This concept evolved from findings that the (A2•p11)2 complex binds both tPA and plasminogen40,41 and accelerates plasmin generation. Specific binding sites reported for plasminogen include lysine (K) 308 of A224 or the C-terminal K of p11,42 or both.43 tPA binding to A2 appears to require a linear sequence within the tail domain (LCKLSL).44 The Ca2+-dependent sequence KGLGT…D in core domain 2 facilitates membrane phospholipid interaction on the endothelial cell.45

Fig. 1.

Fig. 1

Some examples of cell surface and intracellular A2 system functions that relate to human health and disease. (A) Thrombosis and hemostasis. 1. In the vasculature, the endothelial cell surface A2 tetramer [(A2•p11)2] tetramer binds plasminogen (Plg) and tissue plasminogen activator (tPA), accelerating the catalytic generation of plasmin (PN). Plasmin hydrolyzes insoluble fibrin (FN) into soluble fibrin degradation products (FDPs), thus maintaining blood vessel patency. In the absence of A2, fibrinolytic surveillance system is impaired, and fibrin accumulates. 2. In antiphospholipid syndrome and related autoimmune disorders, anti-A2 antibodies lead to fibrin deposition, either by inhibiting cell surface fibrinolysis or by activating endothelial cells and inducing a loss of cell surface thromboresistance. 3. Overexpression of (A2•p11)2, as occurs in acute promyelocytic leukemia (APL), may lead to excessive plasmin generation, depletion of plasmin inhibitors, and induction of a hemorrhagic state (arrow). (B) Angiogenesis. Endothelial cell surface (A2•p11)2, which is upregulated in hypoxia, promotes neovascularization of underperfused tissues, most likely through direct profibrinolytic activity. A2-generated plasmin may also activate downstream matrix metalloproteinases (MMPs), which further remodel the extracellular matrix (ECM). (C) Neurodegenerative disease. Within neurons, A2, presumably in complex with p11, enables the microtubule (MT)-associated protein, tau, to associate with the plasma membrane within axonal growth cones. In familial frontotemporal dementia, the mutated R406W tau fails to associate with plasma membrane-associated A2, leading to axonal instability, and is thought to lead neurodegeneration. (D) Aseptic arthritis. When dendritic cells are exposed to nonbiodegradable wear debris particles (WDP) derived from joint prostheses, the particles are endocytosed and traffic to lysosomes (L). The ensuing late endosomal/ lysosomal membrane damage is thought to be normally repaired through a (A2•p11)2-dependent mechanism. In the absence of A2, escape of lysosomal cathepsin B (CatB) and hydrogen ions (H+) leads to rapid activation of the NLRP3 inflammasome, with activation of caspase-1, and cleavage of pro-interluekin-1 (pro-IL-1β) into its mature, secretable form. Release of IL-1β initiates a potent inflammatory response that exacerbates osteolysis and joint damage.

The AnxA2−/− mouse has been informative in investigating the role of the A2 system in vascular homeostasis in vivo. Although A2-deficient mice display uncompromised development, fertility, and lifespan, fibrin accumulation is a key phenotypic hallmark. Fibrin can be observed in both intra- and extravascular locations within the lungs, spleen, small intestine, liver, and kidney.46 In addition, injury to the carotid artery through application of FeCl3 at the adventitial surface led to a significant decline in carotid blood flow and increased frequency of complete thrombotic occlusion in the AnxA2−/− versus AnxA2+/+ mouse. Cultured microvascular endothelial cells isolated from neonatal AnxA2−/− mice, moreover, failed to support tPA-dependent plasmin generation. S100A10−/− mice also display increased vascular fibrin, reduced clearance of thrombi, and impaired neovascularization of Matrigel implants,47 reflecting either the loss of S100A10 (as a binding site for Plg), loss of p11 “chaperone” function in directing A2 to the plasma membrane surface, or both.

Interestingly, mice with diet-induced hyperhomocysteinemia, like AnxA2−/− mice, also display fibrin accumulation in a wide range of tissues.48 The thiol-containing amino acid, homocysteine, forms as a metabolic intermediate in vivo when methionine is converted to cysteine.49 Hyperhomocysteinemia, which occurs in several inborn errors of metabolism or as a consequence of folate or B vitamin deficiency, has been implicated in thrombotic and atherosclerotic vascular disease,50 although therapies that lower plasma homocysteine may not fully reverse the risk of recurrent cardiovascular disease.51 Pretreatment of endothelial cells with homocysteine largely inhibited both tPA binding and tPA-dependent plasmin formation at the cell surface.52 Incubation of purified A2 with homocysteine also blocked its ability to bind tPA, and A2 extracted from hyperhomocysteinemic mice failed to support tPA binding or tPA-dependent plasmin generation.44 Hyperhomocysteinemia has emerged as an independent risk factor for cerebral small vessel disease, which leads to lacunar infarction and approximately one fourth of all ischemic strokes.53,54 The cell surface A2 pathway may be central to understanding this process.

Animal models of thrombosis have illustrated the theoretical clinical utility of recombinant A2 in ischemic stroke. Compared with those treated with tPA alone, rats treated with tPA plus supplemental rA2 at 2 or 4 hours after the initiation of focal embolic stroke developed significantly smaller infarcts and showed improved cerebral blood flow.55,56 In a follow-up study, labeled embolized thrombi were imaged by magnetic resonance and found, in tPA plus A2 treated rats, to be significantly smaller than those in animals treated with tPA alone.57 Similarly, when carotid artery thrombosis was induced by adventitial application of FeCl3 in the rat, administration of recombinant full-length, but not tail-less, A2 was associated with improved cerebral blood flow and reduced thrombus size in comparison with untreated control animals.58 Because rA2 appears to improve the lytic efficacy of tPA,59 these findings could reduce the small, but significant, risk of cerebral hemorrhage associated with the use of tPA for thromboembolic stroke in humans by lowering the required intensity of tPA treatment.60-62

Beyond these rodent models, there is strong evidence that the A2 system has important implications for human vasoocclusive disease. In antiphospholipid syndrome (APS), a well-known cause of acquired thrombophilia, A2 has been identified as one of many autoantibody targets.63,64 High titer autoantibodies to A2 were detected specifically in patients with APS and severe thrombosis and/or pregnancy morbidity.65,66 Anti-A2 antibodies isolated from APS patients impaired endothelial surface tPA-dependent plasmin generation and incited them to express elevated levels of procoagulant tissue factor.65 Among a cohort of 40 consecutive patients with cerebral venous thrombosis studied 2 to 6 months after the index thrombotic event, moreover, 12.5% were found to have high titer anti-A2 antibodies compared with 2.1% in healthy subjects.67 Although this finding has been confirmed in APS and extended to other autoimmune disorders, such as rheumatoid arthritis, the nature of the target epitopes remains undefined.68-70 In lupus nephritis, finally, autoantibodies directed against A2 have been found to react with renal mesangial cells.71 Together, these data suggest that cell surface A2 represents a prominent autoantibody target and that high titer anti-A2 in the vasculature is associated with a thrombotic diathesis.72

In children with sickle cell disease, stroke is a potentially devastating complication that occurs in 6 to 8% of patients.73 Two studies linked single nucleotide polymorphisms in the ANXA2 gene with increased risk of stroke (odds ratio > 2.7),74,75 whereas additional ANXA2 single nucleotide polymorphisms have been associated with elevated risk of avascular necrosis of bone (osteonecrosis)76 and, possibly, pulmonary hypertension.77 These data suggest A2 may be a significant modifier gene, which, when modified, reduces blood fluidity and exacerbates the natural history of sickle hemoglobinopathy.

Hyperfibrinolysis and Hemorrhage

Acute promyelocytic leukemia (APL) is commonly associated with life-threatening hemorrhage at the time of presentation.78 The presence of the [t(15;17)(q22–24;q12–21)] chromosomal translocation gives rise to the transcriptionally active promyelocytic leukemia-retinoic acid receptor α (PML-RARα) fusion protein. At diagnosis, coagulopathy appears to reflect a combination of disseminated intravascular coagulation and hyperfibrinolysis due to consumption of α2-antiplasmin and excessive plasmin generation. Blast cells isolated from newly diagnosed APL patients expressed unusually high levels of A2, and all patients had evidence of hyperfibrinolysis, with elevated circulating fibrin degradation products, including D-dimer, and depletion of plasma fibrinogen.79 NB4 cells, which carry the t(15;17) translocation and express the PML-RARα fusion protein, displayed steady-state A2 messenger RNA levels that were ~10-fold higher than those found on leukemia cells that lacked the fusion protein. Treatment of NB4 cells with the retinoic acid receptor ligand, all trans-retinoic acid, attenuated A2 expression in a time frame associated with clinical resolution of bleeding.79

In a more recent prospective study, 26 APL patients were found at diagnosis to have enhanced fibrinolysis, despite normal tPA levels and increased plasminogen activator inhibitor-1; harvested APL cells expressed threefold higher levels of A2 and supported elevated tPA-dependent plasmin generation.80 Elevated expression of p11 in NB4 cells has also been reported and appears to respond, like overexpression of A2, to all trans-retinoic acid treatment.81 These studies confirm the likely role of the A2 system in hyperfibrinolytic bleeding in APL, especially in the central nervous system where microvascular expression of A2 may be relatively high.78 Interestingly, induction of diet-induced hyperhomocysteinemia in a mouse model of APL (hCG-PML-RARα mice) reversed hyperfibrinolysis, thus reinforcing the relevance of A2 to APL in vivo and suggesting a potentially novel the rapeutic approach to APL.82

Retinal Angiogenesis

In at least three different models of postnatal angiogenesis, including Matrigel implant, corneal pocket, and oxygen-induced retinopathy, AnxA2−/− mice show a diminished capacity to form new blood vessels (►Fig. 1B).46 In mice with diet-induced hyperhomocysteinemia, which renders A2 unable to bind tPA effectively, corneal neoangiogenesis is also reduced and can be restored to normal upon intravenous treatment with recombinant A2.48 These data suggest that either absence of A2 or its modification by homocysteine deprives the endothelial cell of plasmin-generating capacity, leading most likely to accumulation of fibrin as well as diminished activation of downstream matrix metalloproteases. Unable to adequately remodel extracellular matrix, these cells are unable to mount an appropriate angiogenic response.

In the oxygen-induced retinopathy (OIR) model, newborn mice are maintained with their nursing mothers in a 75% oxygen environment for 5 days and then abruptly transferred to room air; the ensuing relative hypoxia initiates a robust retinal vascular proliferative reaction.83 Interestingly, the in vivo neoangiogenic response is reduced by about 50% in AnxA2−/− pups, and synthesis of A2 is upregulated by the relative hypoxia induced in this model and also by true hypoxia in vitro.35,84,85 Further experiments revealed that this reflects the direct action of hypoxia-inducible factor-1, a master regulator of oxygen-sensitive genes, upon the hypoxia responsive element–containing A2 promoter.35 Electrophoretic mobility shift experiments, chromatin immunoprecipitation studies, and luciferase promoter reporter assays all demonstrate a functional interaction between hypoxia-inducible factor-1α and hypoxia-inducible factor-1β and the human A2 promoter. Gene expression profiling of retinal tissue in murine OIR, furthermore, revealed a 4.3-fold increase in A2 expression at P17, 5 days after the mice were returned to room air.86 A2 was also found to be upregulated and to promote neovascularization after laser rupture of Bruch’s membrane in a model of choroidal angiogenesis.87 In addition, an agent (TM601) that binds directly to A2 appears to suppress both choroidal and retinal neovascularization in the mouse,88 and an A2 N-terminal peptide specifically blocked neoangiogenesis in the chick chorioallantoic membrane model and the mouse Matrigel plug assay.84 These data suggest that A2 is likely to be upregulated in other tissues in response to hypoxia and may be central to the angiogenic response in many pathologic settings.

From a mechanistic point of view, it appears that the blockade to normal angiogenesis in the AnxA2−/− mouse in OIR reflects an inability of endothelial cells to traverse an extracellular fibrin barrier. Hypoxic angiogenesis, which is initiated by vascular endothelial growth factor, involves loss of interendothelial cell junctions and leakage of plasma into the extracellular space with fibrin formation. Neovascularization during OIR in the AnxA2−/− retina can be normalized upon systemic pretreatment of the mice with ancrod, an agent that depletes fibrinogen, thus preventing fibrin formation.35 Similarly, the robust angiogenic response that occurs in the wild-type mouse can be tempered by pretreatment with tranexamic acid, a global fibrinolytic inhibitor.35 These results link fibrinolysis with neoangiogenesis, suggesting new therapeutic approaches for proliferative retinal vascular disorders, such as retinopathy of prematurity or diabetic retinopathy.

Cancer Progression

Glioblastomas are highly aggressive and treatment-refractory malignancies that typically exhibit widespread infiltration of surrounding structures early in the development of the disease.89 Gliomas produce proteases, including plasminogen activators and matrix metalloproteinases, both of which are thought to promote malignant cell invasion.89,90 In proteomic analyses, high expression of A2 has been associated with more aggressive phenotypes,91 and A2 appears to localize to pseudopodia of invasive glioma cells.92 In both rat and mouse models, stable knockdown of A2 significantly inhibited glioblastoma invasion, proliferation, and angiogenesis after stereotactic implantation in the brain; interestingly, these effects were independent of p11 expression.93 High expression of A2 has also been documented in human glioblastoma, where it correlates positively with histologic grade and central nervous system dissemination.93-96 These data suggest that A2-directed treatment might offer a new therapeutic modality for human glioblastoma.

In a xenograft model of highly invasive breast cancer, similarly, malignant human cells were implanted into nude mice, and both tumor growth and vascular density was blocked by administration of anti-A2 monoclonal antibody.97 The tumor cells used in this experiment expressed abundant A2, strongly supported tPA binding and tPA-dependent plasmin generation, and exhibited plasmin- and A2-dependent cellular matrix invasion.98,99 In another study, growth of Lewis lung and T241 sarcoma tumors implanted into S100A10−/− mice was reduced compared with wild-type controls.100 Impaired tumor growth was correlated with diminished macrophage density within the tumors, and clodronate-mediated depletion of macrophages in wild-type mice led to a similar reduction in tumor size, suggesting an effect of p11 on macrophage recruitment.42 Finally, soluble (A2•p11)2 tetramer has been reported to activate monocyte-derived macrophages via toll-like receptor 4, thus modulating cytokine production.101

In multiple myeloma, A2 is a candidate prognostic marker. It is expressed in both normal and malignant plasma cells, but elevated expression in myeloma is associated with reduced event-free and overall survival.102 High-level expression of A2 in human myeloma cells has been confirmed in transcriptional profiling studies.103 Primary multiple myeloma cells harvested from a cohort of patients displayed 10-fold higher cell surface A2 expression than that observed on normal plasma cells; silencing of A2 in related cell lines suppressed expression of pro-angiogenic genes.104 Mechanistically, A2 and a related A2-binding “receptor” appear to promote myeloma cell malignant plasma cell growth and adhesion to stromal cells within the bone marrow microenvironment.105 Interestingly, prostate cancer cells also appear to use A2 in homing to the bone marrow and in establishing bone metastases.106 Together, these studies suggest that A2 may contribute to cancer progression through multiple mechanisms.

Neuropsychiatric Disease

Protein S100A10 appears to orchestrate several aspects of neuropsychiatric function.107 p11 binds to two serotonin receptors (5-HT1B and 5-HT4), both involved in mood regulation.108 S100A10−/− mice exhibit increased immobility in the tail suspension test, increased thigmotaxis, and decreased responsiveness to a sucrose reward, all thought to be indicative of depression-like behavior. In wild-type mice, suppression of p11 expression within the nucleus accumbens resulted in depression-like behavior, similar to that observed in globally deficient p11 mice; exogenous administration of p11 within the nucleus accumbens of p11-deleted mice seems to restore normal behavior.109 In humans suffering from depression, p11 levels in the nucleus accumbens were reduced, as were messenger RNA levels in peripheral blood mononuclear cells, suggesting a novel biomarker for patients at high risk for suicide.110 Interestingly, frequently used antiinflammatory agents that attenuate the antidepressive effects of serotonin reuptake inhibitors may do so by inhibiting the effects of interferon-γ, a known inducer of p11.111,112 Thus, some forms of human depression may be amenable to augmentation of p11 expression; it is not yet known whether this p11 function is A2 dependent.113

The sensory neuron tetrodotoxin-resistant sodium channel (Nav1.8/SNS) is the primary pain perception receptor and is expressed in 85% of neurons emanating from the dorsal route ganglia. p11 promotes translocation of to the Nav1.8/SNS channel to the plasma membrane by binding to its amino terminus,114 but it is not yet clear whether this interaction requires the participation of A2.115 Deletion of p11 in primary nociceptor sensory neurons results in a severe reduction in coding of painful stimuli in dorsal root sensory neurons.116 These data raise the possibility that p11 targeting could provide useful treatment for refractory pain disorders.

Annexin A2 in the Cytoplasm

Frontotemporal Dementia

Neurodegenerative processes are increasing in frequency as the world’s population ages. In frontotemporal lobar degeneration with ubiquitinated inclusions, A2 was found to be overexpressed in expression profiling of postmortem frontal lobe tissue.117 In familial frontotemporal dementia, another neurodegenerative process, mutations in the microtubule-associated protein, tau, have been identified.118 A study revealed that A2 enables wild-type tau to interact with the neuronal growth cone plasma membrane, thus stabilizing process outgrowth by bridging microtubules to the membrane (►Fig. 1C).119 A2 is, however, unable to interact with the R406W, disease-associated form of tau, possibly leading to its intracellular accumulation and neuronal cell death.

Limb-Girdle Muscular Dystrophy

Limb-girdle muscular dystrophy type 2B, a disorder characterized by progressive weakness and atrophy of proximal muscles and elevated serum creatine kinase levels, reflects mutation of the gene encoding dysferlin, a member of the dystrophin-associated complex of muscle proteins.120,121 Gene expression profiling has revealed an approximately twofold overexpression of A2 transcript levels in biopsied muscle tissue from these patients compared with healthy control subjects,122 and the level of expression of A2, as well as A1, in biopsied muscle appears to correlate with clinical severity.123 Of note, both A2 and A1 have been shown to interact with dysferlin by co-immunoprecipitation, to colocalize with dysferlin in normal muscle biopsies, and to promote sarcolemmal wound healing in an in vivo model.124 In response to myotube injury, A2 appears to aggregate intracellular vesicles that fuse to form a patch that breaches the plasma membrane defect.

Aseptic Wear Debris Arthritis

Although several million arthroplastic joint replacements are performed each year, 10 to 15% of these procedures fail due to aseptic osteolysis.125 In this process, wear debris particles, generated by frictional forces exerted on artificial joint surfaces, are endocytosed by inflammatory cells, such as macrophages and dendritic cells. Eight to 12 carbon alkanes, derived from wear debris particles, can bind directly to toll-like receptors 1 and 2, thereby activating downstream inflammatory pathways.126 In addition, wear debris particles can induce endosomal membrane damage, which is associated with dramatic recruitment of cytoplasmic A2 to the endosomal membrane. In the absence of A2, endo/lysosomal disruption leads to leakage of lysosomal cathepsins and H+ ions into the cytosol with subsequent activation of the nucleotide-binding, leucine-rich, pyrin-containing-3 (NLRP3) inflammasome, secretion of interleukin-1β, and an accelerated inflammatory response (►Fig. 1D).127

Summary

The A2 system fulfills a range of biologic functions both on the plasma membrane and within multiple intracellular compartments. At the cell surface, the A2 system promotes fibrinolysis, angiogenesis, and cell migration. Within the cell, it aids in organelle membrane organization, fusion, and repair. Although we are beginning to understand the physiologic orchestration of these events, key questions remain about the regulation of the system and its precise role in human health and disease. The next few years in A2 biology should prove exciting and illuminating.

Acknowledgments

Supported by National Institutes of Health grants HL42493, HL046403, and HL090895 and by a grant from the March of Dimes (6-FY12-356). In addition, we apologize to anyone whose work we have omitted in the interest of space.

References

  • 1.Moss SE, Morgan RO. The annexins. Genome Biol. 2004;5:1–8. doi: 10.1186/gb-2004-5-4-219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Rand JH. “Annexinopathies”—a new class of diseases. N Engl J Med. 1999;340(13):1035–1036. doi: 10.1056/NEJM199904013401310. [DOI] [PubMed] [Google Scholar]
  • 3.Rand JH. The annexinopathies: a new category of diseases. Biochim Biophys Acta. 2000;1498(2-3):169–173. doi: 10.1016/s0167-4889(00)00093-8. [DOI] [PubMed] [Google Scholar]
  • 4.Hayes MJ, Moss SE. Annexins and disease. Biochem Biophys Res Commun. 2004;322(4):1166–1170. doi: 10.1016/j.bbrc.2004.07.124. [DOI] [PubMed] [Google Scholar]
  • 5.Hayes MJ, Longbottom RE, Evans MA, Moss SE. Annexinopathies. Subcell Biochem. 2007;45:1–28. doi: 10.1007/978-1-4020-6191-2_1. [DOI] [PubMed] [Google Scholar]
  • 6.Fatimathas L, Moss SE. Annexins as disease modifiers. Histol Histopathol. 2010;25(4):527–532. doi: 10.14670/HH-25.527. [DOI] [PubMed] [Google Scholar]
  • 7.Heizmann CW, Ackermann GE, Galichet A. Pathologies involving the S100 proteins and RAGE. Subcell Biochem. 2007;45:93–138. doi: 10.1007/978-1-4020-6191-2_5. [DOI] [PubMed] [Google Scholar]
  • 8.Hedhli N, Falcone DJ, Huang B, et al. The annexin A2/S100A10 system in health and disease: emerging paradigms. J Biomed Biotechnol. 2012;2012:406273. doi: 10.1155/2012/406273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gerke V, Creutz CE, Moss SE. Annexins: linking Ca2+ signalling to membrane dynamics. Nat Rev Mol Cell Biol. 2005;6(6):449–461. doi: 10.1038/nrm1661. [DOI] [PubMed] [Google Scholar]
  • 10.Gerke V, Moss SE. Annexins: from structure to function. Physiol Rev. 2002;82(2):331–371. doi: 10.1152/physrev.00030.2001. [DOI] [PubMed] [Google Scholar]
  • 11.Spano F, Raugei G, Palla E, Colella C, Melli M. Characterization of the human lipocortin-2-encoding multigene family: its structure suggests the existence of a short amino acid unit undergoing duplication. Gene. 1990;95(2):243–251. doi: 10.1016/0378-1119(90)90367-z. [DOI] [PubMed] [Google Scholar]
  • 12.Waisman DM. Annexin II tetramer: structure and function. Mol Cell Biochem. 1995;149-150:301–322. doi: 10.1007/BF01076592. [DOI] [PubMed] [Google Scholar]
  • 13.Rescher U, Gerke V. S100A10/p11: Family, friends, and functions. Pflugers Arch Eur J Physiol. 2007;455:575–582. doi: 10.1007/s00424-007-0313-4. [DOI] [PubMed] [Google Scholar]
  • 14.Donato R. S100: a multigenic family of calcium-modulated proteins of the EF-hand type with intracellular and extracellular functional roles. Int J Biochem Cell Biol. 2001;33(7):637–668. doi: 10.1016/s1357-2725(01)00046-2. [DOI] [PubMed] [Google Scholar]
  • 15.Réty S, Sopkova J, Renouard M, et al. The crystal structure of a complex of p11 with the annexin II N-terminal peptide. Nat Struct Biol. 1999;6(1):89–95. doi: 10.1038/4965. [DOI] [PubMed] [Google Scholar]
  • 16.Knop M, Aareskjold E, Bode G, Gerke V. Rab3D and annexin A2 play a role in regulated secretion of vWF, but not tPA, from endothelial cells. EMBO J. 2004;23(15):2982–2992. doi: 10.1038/sj.emboj.7600319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Umbrecht-Jenck E, Demais V, Calco V, Bailly Y, Bader MF, Chasserot-Golaz S. S100A10-mediated translocation of annexin-A2 to SNARE proteins in adrenergic chromaffin cells undergoing exocytosis. Traffic. 2010;11(7):958–971. doi: 10.1111/j.1600-0854.2010.01065.x. [DOI] [PubMed] [Google Scholar]
  • 18.Wang P, Chintagari NR, Gou D, Su L, Liu L. Physical and functional interactions of SNAP-23 with annexin A2. Am J Respir Cell Mol Biol. 2007;37(4):467–476. doi: 10.1165/rcmb.2006-0447OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zobiack N, Rescher U, Ludwig C, Zeuschner D, Gerke V. The annexin 2/S100A10 complex controls the distribution of transferrin receptor-containing recycling endosomes. Mol Biol Cell. 2003;14(12):4896–4908. doi: 10.1091/mbc.E03-06-0387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Eberhard DA, Karns LR, VandenBerg SR, Creutz CE. Control of the nuclear-cytoplasmic partitioning of annexin II by a nuclear export signal and by p11 binding. J Cell Sci. 2001;114(Pt 17):3155–3166. doi: 10.1242/jcs.114.17.3155. [DOI] [PubMed] [Google Scholar]
  • 21.Vedeler A, Hollås H, Grindheim AK, Raddum AM. Multiple roles of annexin A2 in post-transcriptional regulation of gene expression. Curr Protein Pept Sci. 2012;13(4):401–412. doi: 10.2174/138920312801619402. [DOI] [PubMed] [Google Scholar]
  • 22.Grieve AG, Moss SE, 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;2012:852430. doi: 10.1155/2012/852430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Flood EC, Hajjar KA. The annexin A2 system and vascular homeostasis. Vascul Pharmacol. 2011;54(3-6):59–67. doi: 10.1016/j.vph.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dassah M, Deora AB, He K, Hajjar KA. The endothelial cell annexin A2 system and vascular fibrinolysis. Gen Physiol Biophys. 2009;28:F20–F28. Spec No Focus. [PMC free article] [PubMed] [Google Scholar]
  • 25.Madureira PA, Surette AP, Phipps KD, Taboski MAS, Miller VA, Waisman DM. The role of the annexin A2 heterotetramer in vascular fibrinolysis. Blood. 2011;118(18):4789–4797. doi: 10.1182/blood-2011-06-334672. [DOI] [PubMed] [Google Scholar]
  • 26.Cesarman-Maus G, Hajjar KA. Molecular mechanisms of fibrinolysis. Br J Haematol. 2005;129(3):307–321. doi: 10.1111/j.1365-2141.2005.05444.x. [DOI] [PubMed] [Google Scholar]
  • 27.Hajjar KA. The molecular basis of fibrinolysis. In: Orkin SH, Nathan DG, Ginsburg D, Look AT, Fisher DE, Lux SE, editors. Nathan and Oski’s Hematology of Infancy and Childhood. 7th ed. Saunders Elsevier; Philadelphia: 2009. pp. 1425–1447. [Google Scholar]
  • 28.Hajjar KA, Ruan J. Fibrinolysis and thrombolysis. In: Kaushansky K, Lichtman MA, Beutler E, Kipps TJ, Seligsohn U, Prchal JT, editors. Williams Hematology. 8th ed. McGraw-Hill; New York: 2010. pp. 2219–2246. [Google Scholar]
  • 29.Valapala M, Vishwanatha JK. Lipid raft endocytosis and exosomal transport facilitate extracellular trafficking of annexin A2. J Biol Chem. 2011;286(35):30911–30925. doi: 10.1074/jbc.M111.271155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Faure AV, Migné C, Devilliers G, Ayala-Sanmartin J. Annexin 2 “secretion” accompanying exocytosis of chromaffin cells: possible mechanisms of annexin release. Exp Cell Res. 2002;276(1):79–89. doi: 10.1006/excr.2002.5512. [DOI] [PubMed] [Google Scholar]
  • 31.Danielsen EM, van Deurs B, Hansen GH. “Nonclassical” secretion of annexin A2 to the lumenal side of the enterocyte brush border membrane. Biochemistry. 2003;42(49):14670–14676. doi: 10.1021/bi0355239. [DOI] [PubMed] [Google Scholar]
  • 32.Keller M, Rüegg A, Werner S, Beer HD. Active caspase-1 is a regulator of unconventional protein secretion. Cell. 2008;132(5):818–831. doi: 10.1016/j.cell.2007.12.040. [DOI] [PubMed] [Google Scholar]
  • 33.Peterson EA, Sutherland MR, Nesheim ME, Pryzdial EL. Thrombin induces endothelial cell-surface exposure of the plasminogen receptor annexin 2. J Cell Sci. 2003;116(Pt 12):2399–2408. doi: 10.1242/jcs.00434. [DOI] [PubMed] [Google Scholar]
  • 34.Deora AB, Kreitzer G, Jacovina AT, Hajjar KA. An annexin 2 phosphorylation switch mediates p11-dependent translocation of annexin 2 to the cell surface. J Biol Chem. 2004;279(42):43411–43418. doi: 10.1074/jbc.M408078200. [DOI] [PubMed] [Google Scholar]
  • 35.Huang B, Deora AB, He KL, et al. Hypoxia-inducible factor-1 drives annexin A2 system-mediated perivascular fibrin clearance in oxygen-induced retinopathy in mice. Blood. 2011;118(10):2918–2929. doi: 10.1182/blood-2011-03-341214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Erikson E, Erikson RL. Identification of a cellular protein substrate phosphorylated by the avian sarcoma virus-transforming gene product. Cell. 1980;21(3):829–836. doi: 10.1016/0092-8674(80)90446-8. [DOI] [PubMed] [Google Scholar]
  • 37.He KL, Deora AB, Xiong H, et al. Endothelial cell annexin A2 regulates polyubiquitination and degradation of its binding partner S100A10/p11. J Biol Chem. 2008;283(28):19192–19200. doi: 10.1074/jbc.M800100200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.He KL, Sui G, Xiong H, et al. Feedback regulation of endothelial cell surface plasmin generation by PKC-dependent phosphorylation of annexin A2. J Biol Chem. 2011;286(17):15428–15439. doi: 10.1074/jbc.M110.185058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Jost M, Gerke V. Mapping of a regulatory important site for protein kinase C phosphorylation in the N-terminal domain of annexin II. Biochim Biophys Acta. 1996;1313(3):283–289. doi: 10.1016/0167-4889(96)00101-2. [DOI] [PubMed] [Google Scholar]
  • 40.Hajjar KA, Jacovina AT, Chacko J. An endothelial cell receptor for plasminogen/tissue plasminogen activator. I. Identity with annexin II. J Biol Chem. 1994;269(33):21191–21197. [PubMed] [Google Scholar]
  • 41.Cesarman GM, Guevara CA, 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. 1994;269(33):21198–21203. [PubMed] [Google Scholar]
  • 42.O’Connell PA, Surette AP, Liwski RS, Svenningsson P, Waisman DM. S100A10 regulates plasminogen-dependent macrophage invasion. Blood. 2010;116(7):1136–1146. doi: 10.1182/blood-2010-01-264754. [DOI] [PubMed] [Google Scholar]
  • 43.Das R, Burke T, Plow EF. Histone H2B as a functionally important plasminogen receptor on macrophages. Blood. 2007;110(10):3763–3772. doi: 10.1182/blood-2007-03-079392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hajjar KA, Mauri L, Jacovina AT, et al. Tissue plasminogen activator binding to the annexin II tail domain. Direct modulation by homocysteine. J Biol Chem. 1998;273(16):9987–9993. doi: 10.1074/jbc.273.16.9987. [DOI] [PubMed] [Google Scholar]
  • 45.Hajjar KA, Guevara CA, Lev E, Dowling K, Chacko J. Interaction of the fibrinolytic receptor, annexin II, with the endothelial cell surface. Essential role of endonexin repeat 2. J Biol Chem. 1996;271(35):21652–21659. doi: 10.1074/jbc.271.35.21652. [DOI] [PubMed] [Google Scholar]
  • 46.Ling Q, Jacovina AT, Deora AB, et al. Annexin II regulates fibrin homeostasis and neoangiogenesis in vivo. J Clin Invest. 2004;113(1):38–48. doi: 10.1172/JCI200419684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Surette AP, Madureira PA, Phipps KD, Miller VA, Svenningsson P, Waisman DM. Regulation of fibrinolysis by S100A10 in vivo. Blood. 2011;118(11):3172–3181. doi: 10.1182/blood-2011-05-353482. [DOI] [PubMed] [Google Scholar]
  • 48.Jacovina AT, Deora AB, Ling Q, et al. Homocysteine inhibits neoangiogenesis in mice through blockade of annexin A2-dependent fibrinolysis. J Clin Invest. 2009;119(11):3384–3394. doi: 10.1172/JCI39591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Selhub J. Homocysteine metabolism. Annu Rev Nutr. 1999;19:217–246. doi: 10.1146/annurev.nutr.19.1.217. [DOI] [PubMed] [Google Scholar]
  • 50.Humphrey LL, Fu R, Rogers K, Freeman M, Helfand M. Homocysteine level and coronary heart disease incidence: a systematic review and meta-analysis. Mayo Clin Proc. 2008;83(11):1203–1212. doi: 10.4065/83.11.1203. [DOI] [PubMed] [Google Scholar]
  • 51.Bønaa KH, Njølstad I, Ueland PM, et al. NORVIT Trial Investigators Homocysteine lowering and cardiovascular events after acute myocardial infarction. N Engl J Med. 2006;354(15):1578–1588. doi: 10.1056/NEJMoa055227. [DOI] [PubMed] [Google Scholar]
  • 52.Hajjar KA. Homocysteine-induced modulation of tissue plasminogen activator binding to its endothelial cell membrane receptor. J Clin Invest. 1993;91(6):2873–2879. doi: 10.1172/JCI116532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Markus HS. Genes, endothelial function and cerebral small vessel disease in man. Exp Physiol. 2008;93(1):121–127. doi: 10.1113/expphysiol.2007.038752. [DOI] [PubMed] [Google Scholar]
  • 54.Wong A, Mok V, Fan YH, Lam WWM, Liang KS, Wong KS. Hyperhomocysteinemia is associated with volumetric white matter change in patients with small vessel disease. J Neurol. 2006;253(4):441–447. doi: 10.1007/s00415-005-0022-x. [DOI] [PubMed] [Google Scholar]
  • 55.Zhu H, Fan X, Yu Z, et al. Annexin A2 combined with low-dose tPA improves thrombolytic therapy in a rat model of focal embolic stroke. J Cereb Blood Flow Metab. 2010;30(6):1137–1146. doi: 10.1038/jcbfm.2009.279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Tanaka Y, Ishii H, Hiraoka M, et al. Efficacy of recombinant annexin 2 for fibrinolytic therapy in a rat embolic stroke model: a magnetic resonance imaging study. Brain Res. 2007;1165:135–143. doi: 10.1016/j.brainres.2007.06.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Walvick RP, Bråtane BT, Henninger N, et al. Visualization of clot lysis in a rat embolic stroke model: application to comparative lytic efficacy. Stroke. 2011;42(4):1110–1115. doi: 10.1161/STROKEAHA.110.602102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ishii H, Yoshida M, Hiraoka M, et al. Recombinant annexin II modulates impaired fibrinolytic activity in vitro and in rat carotid artery. Circ Res. 2001;89(12):1240–1245. doi: 10.1161/hh2401.101066. [DOI] [PubMed] [Google Scholar]
  • 59.Fan X, Yu Z, Liu J, et al. Annexin A2: a tissue plasminogen activator amplifier for thrombolytic stroke therapy. Stroke. 2010;41(10, Suppl):S54–S58. doi: 10.1161/STROKEAHA.110.596106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Yepes M, Sandkvist M, Moore EG, Bugge TH, Strickland DK, Lawrence DA. Tissue-type plasminogen activator induces opening of the blood-brain barrier via the LDL receptor-related protein. J Clin Invest. 2003;112(10):1533–1540. doi: 10.1172/JCI19212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wang X, Tsuji K, Lee SR, et al. Mechanisms of hemorrhagic transformation after tissue plasminogen activator reperfusion therapy for ischemic stroke. Stroke. 2004;35(11, Suppl 1):2726–2730. doi: 10.1161/01.STR.0000143219.16695.af. [DOI] [PubMed] [Google Scholar]
  • 62.Kaur J, Zhao Z, Klein GM, Lo EH, Buchan AM. The neurotoxicity of tissue plasminogen activator? J Cereb Blood Flow Metab. 2004;24(9):945–963. doi: 10.1097/01.WCB.0000137868.50767.E8. [DOI] [PubMed] [Google Scholar]
  • 63.Cockrell E, Espinola RG, McCrae KR. Annexin A2: biology and relevance to the antiphospholipid syndrome. Lupus. 2008;17(10):943–951. doi: 10.1177/0961203308095329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Cohen D, Berger SP, Steup-Beekman GM, Bloemenkamp KWM, Bajema IM. Diagnosis and management of the antiphosphlipid syndrome. BMJ. 2010;340:1125–1132. doi: 10.1136/bmj.c2541. [DOI] [PubMed] [Google Scholar]
  • 65.Cesarman-Maus G, Ríos-Luna NP, Deora AB, et al. Autoantibodies against the fibrinolytic receptor, annexin 2, in antiphospholipid syndrome. Blood. 2006;107(11):4375–4382. doi: 10.1182/blood-2005-07-2636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ao W, Zheng H, Chen XW, Shen Y, Yang CD. Anti-annexin II antibody is associated with thrombosis and/or pregnancy morbidity in antiphospholipid syndrome and systemic lupus erythematosus with thrombosis. Rheumatol Int. 2011;31(7):865–869. doi: 10.1007/s00296-010-1379-4. [DOI] [PubMed] [Google Scholar]
  • 67.Cesarman-Maus G, Cantú-Brito C, Barinagarrementeria F, et al. Autoantibodies against the fibrinolytic receptor, annexin A2, in cerebral venous thrombosis. Stroke. 2011;42(2):501–503. doi: 10.1161/STROKEAHA.110.592121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Salle V, Mazière JC, Brulé A, et al. Antibodies against the N-terminal domain of annexin A2 in antiphospholipid syndrome. Eur J Intern Med. 2012;23(7):665–668. doi: 10.1016/j.ejim.2012.04.006. [DOI] [PubMed] [Google Scholar]
  • 69.Salle V, Mazière JC, Smail A, et al. Anti-annexin II antibodies in systemic autoimmune diseases and antiphospholipid syndrome. J Clin Immunol. 2008;28(4):291–297. doi: 10.1007/s10875-008-9188-1. [DOI] [PubMed] [Google Scholar]
  • 70.Ao W, Zheng H, Chen XW, Shen Y, Yang CD. Anti-annexin II antibody is associated with thrombosis and/or pregnancy morbidity in antiphospholipid syndrome and systemic lupus erythematosus with thrombosis. Rheumatol Int. 2011;31(7):865–869. doi: 10.1007/s00296-010-1379-4. [DOI] [PubMed] [Google Scholar]
  • 71.Yung S, Cheung KF, Zhang QY, Chan TM. Anti-dsDNA antibodies bind to mesangial annexin II in lupus nephritis. J Am Soc Nephrol. 2010;21(11):1912–1927. doi: 10.1681/ASN.2009080805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Krone KA, Allen KL, McCrae KR. Impaired fibrinolysis in the antiphospholipid syndrome. Curr Rheumatol Rep. 2010;12(1):53–57. doi: 10.1007/s11926-009-0075-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Platt OS. Preventing stroke in sickle cell anemia. N Engl J Med. 2005;353(26):2743–2745. doi: 10.1056/NEJMp058274. [DOI] [PubMed] [Google Scholar]
  • 74.Sebastiani P, Ramoni MF, Nolan V, Baldwin CT, Steinberg MH. Genetic dissection and prognostic modeling of overt stroke in sickle cell anemia. Nat Genet. 2005;37(4):435–440. doi: 10.1038/ng1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Flanagan JM, Frohlich DM, Howard TA, et al. Genetic predictors for stroke in children with sickle cell anemia. Blood. 2011;117(24):6681–6684. doi: 10.1182/blood-2011-01-332205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Baldwin CT, Nolan VG, Wyszynski DF, et al. Association of klotho, bone morphogenic protein 6, and annexin A2 polymorphisms with sickle cell osteonecrosis. Blood. 2005;106(1):372–375. doi: 10.1182/blood-2005-02-0548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ashley-Koch AE, Elliott L, Kail ME, et al. Identification of genetic polymorphisms associated with risk for pulmonary hypertension in sickle cell disease. Blood. 2008;111(12):5721–5726. doi: 10.1182/blood-2007-02-074849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Stein E, McMahon B, Kwaan H, Altman JK, Frankfurt O, Tallman MS. The coagulopathy of acute promyelocytic leukaemia revisited. Best Pract Res Clin Haematol. 2009;22(1):153–163. doi: 10.1016/j.beha.2008.12.007. [DOI] [PubMed] [Google Scholar]
  • 79.Menell JS, Cesarman GM, Jacovina AT, McLaughlin MA, Lev EA, Hajjar KA. Annexin II and bleeding in acute promyelocytic leukemia. N Engl J Med. 1999;340(13):994–1004. doi: 10.1056/NEJM199904013401303. [DOI] [PubMed] [Google Scholar]
  • 80.Liu Y, Wang Z, Jiang M, et al. The expression of annexin II and its role in the fibrinolytic activity in acute promyelocytic leukemia. Leuk Res. 2011;35(7):879–884. doi: 10.1016/j.leukres.2010.11.008. [DOI] [PubMed] [Google Scholar]
  • 81.O’Connell PA, Madureira PA, Berman JN, Liwski RS, Waisman DM. Regulation of S100A10 by the PML-RAR-α oncoprotein. Blood. 2011;117(15):4095–4105. doi: 10.1182/blood-2010-07-298851. [DOI] [PubMed] [Google Scholar]
  • 82.Jácomo RH, Santana-Lemos BA, Lima ASG, et al. Methionine-induced hyperhomocysteinemia reverts fibrinolytic pathway activation in a murine model of acute promyelocytic leukemia. Blood. 2012;120(1):207–213. doi: 10.1182/blood-2011-04-347187. [DOI] [PubMed] [Google Scholar]
  • 83.Smith LE, Wesolowski E, McLellan A, et al. Oxygen-induced retinopathy in the mouse. Invest Ophthalmol Vis Sci. 1994;35(1):101–111. [PubMed] [Google Scholar]
  • 84.Valapala M, Thamake SI, Vishwanatha JK. A competitive hexapeptide inhibitor of annexin A2 prevents hypoxia-induced angiogenic events. J Cell Sci. 2011;124(Pt 9):1453–1464. doi: 10.1242/jcs.079236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Zhao S, Huang L, Wu J, Zhang Y, Pan D, Liu X. Vascular endothelial growth factor upregulates expression of annexin A2 in vitro and in a mouse model of ischemic retinopathy. Mol Vis. 2009;15:1231–1242. [PMC free article] [PubMed] [Google Scholar]
  • 86.Sato T, Kusaka S, Hashida N, Saishin Y, Fujikado T, Tano Y. Comprehensive gene-expression profile in murine oxygen-induced retinopathy. Br J Ophthalmol. 2009;93(1):96–103. doi: 10.1136/bjo.2008.142646. [DOI] [PubMed] [Google Scholar]
  • 87.Zhao SH, Pan DY, Zhang Y, Wu JH, Liu X, Xu Y. Annexin A2 promotes choroidal neovascularization by increasing vascular endothelial growth factor expression in a rat model of argon laser coagulation-induced choroidal neovascularization. Chin Med J (Engl) 2010;123(6):713–721. [PubMed] [Google Scholar]
  • 88.Lima e Silva R, Shen J, Gong YY, et al. Agents that bind annexin A2 suppress ocular neovascularization. J Cell Physiol. 2010;225(3):855–864. doi: 10.1002/jcp.22296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Rao JS. Molecular mechanisms of glioma invasiveness: the role of proteases. Nat Rev Cancer. 2003;3(7):489–501. doi: 10.1038/nrc1121. [DOI] [PubMed] [Google Scholar]
  • 90.Tsatas D, Kaye AH. The role of the plasminogen activation cascade in glioma cell invasion: a review. J Clin Neurosci. 2003;10(2):139–145. doi: 10.1016/s0967-5868(02)00328-4. [DOI] [PubMed] [Google Scholar]
  • 91.Maruo T, Ichikawa T, Kanzaki H, et al. Proteomics-based analysis of invasion-related proteins in malignant gliomas. Neuropathology. 2012 doi: 10.1111/j.1440-1789.2012.01361.x. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Beckner ME, Chen X, An J, Day BW, Pollack IF. Proteomic characterization of harvested pseudopodia with differential gel electrophoresis and specific antibodies. Lab Invest. 2005;85(3):316–327. doi: 10.1038/labinvest.3700239. [DOI] [PubMed] [Google Scholar]
  • 93.Zhai H, Acharya S, Gravanis I, et al. Annexin A2 promotes tumor progression in glioma cells. J Neurosci. 2011;31(40):14346–14360. doi: 10.1523/JNEUROSCI.3299-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Reeves SA, Chavez-Kappel C, Davis R, Rosenblum M, Israel MA. Developmental regulation of annexin II (lipocortin 2) in human brain and expression in high grade glioma. Cancer Res. 1992;52(24):6871–6876. [PubMed] [Google Scholar]
  • 95.Nygaard SJ, Haugland HK, Kristoffersen EK, Lund-Johansen M, Laerum OD, Tysnes OB. Expression of annexin II in glioma cell lines and in brain tumor biopsies. J Neurooncol. 1998;38(1):11–18. doi: 10.1023/a:1005953000523. [DOI] [PubMed] [Google Scholar]
  • 96.Roseman BJ, Bollen A, Hsu J, Lamborn K, Israel MA. Annexin II marks astrocytic brain tumors of high histologic grade. Oncol Res. 1994;6(12):561–567. [PubMed] [Google Scholar]
  • 97.Sharma M, Blackman MR, Sharma MC. Antibody-directed neutralization of annexin II (ANX II) inhibits neoangiogenesis and human breast tumor growth in a xenograft model. Exp Mol Pathol. 2012;92(1):175–184. doi: 10.1016/j.yexmp.2011.10.003. [DOI] [PubMed] [Google Scholar]
  • 98.Sharma MR, Koltowski L, Ownbey RT, Tuszynski GP, 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. 2006;81(2):146–156. doi: 10.1016/j.yexmp.2006.03.003. [DOI] [PubMed] [Google Scholar]
  • 99.Sharma M, Ownbey RT, Sharma MC. Breast cancer cell surface annexin II induces cell migration and neoangiogenesis via tPA dependent plasmin generation. Exp Mol Pathol. 2010;88(2):278–286. doi: 10.1016/j.yexmp.2010.01.001. [DOI] [PubMed] [Google Scholar]
  • 100.Phipps KD, Surette AP, O’Connell PA, Waisman DM. Plasmingen receptor S100A10 is essential for the migration of tumor-promoting macrophages into tumor sites. Cancer Res. 2011;7:6676–6683. doi: 10.1158/0008-5472.CAN-11-1748. [DOI] [PubMed] [Google Scholar]
  • 101.Swisher JFA, Burton N, Bacot SM, Vogel SN, Feldman GM. Annexin A2 tetramer activates human and murine macrophages through TLR4. Blood. 2010;115(3):549–558. doi: 10.1182/blood-2009-06-226944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Seckinger A, Meissner T, Moreaux J, et al. Clinical and prognostic role of annexin A2 in multiple myleoma. Blood. 2012;120:10871094. doi: 10.1182/blood-2012-03-415588. [DOI] [PubMed] [Google Scholar]
  • 103.Claudio JO, Masih-Khan E, Tang H, et al. A molecular compendium of genes expressed in multiple myeloma. Blood. 2002;100(6):2175–2186. doi: 10.1182/blood-2002-01-0008. [DOI] [PubMed] [Google Scholar]
  • 104.Bao H, Jiang M, Zhu M, Sheng F, Ruan J, Ruan C. Overexpression of Annexin II affects the proliferation, apoptosis, invasion and production of proangiogenic factors in multiple myeloma. Int J Hematol. 2009;90(2):177–185. doi: 10.1007/s12185-009-0356-8. [DOI] [PubMed] [Google Scholar]
  • 105.D’Souza S, Kurihara N, Shiozawa Y, et al. Annexin II interactions with the annexin II receptor enhance multiple myeloma cell adhesion and growth in the bone marrow microenvironment. Blood. 2012;119(8):1888–1896. doi: 10.1182/blood-2011-11-393348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.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. 2008;105(2):370–380. doi: 10.1002/jcb.21835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Svenningsson P, Greengard P. p11 (S100A10)—an inducible adaptor protein that modulates neuronal functions. Curr Opin Pharmacol. 2007;7(1):27–32. doi: 10.1016/j.coph.2006.10.001. [DOI] [PubMed] [Google Scholar]
  • 108.Svenningsson P, Chergui K, Rachleff I, et al. Alterations in 5-HT1B receptor function by p11 in depression-like states. Science. 2006;311(5757):77–80. doi: 10.1126/science.1117571. [DOI] [PubMed] [Google Scholar]
  • 109.Alexander B, Warner-Schmidt JL, Eriksson T, et al. Reversal of depressed behaviors in mice by p11 gene therapy in the nucleus accumbens. Sci Transl Med. 2010;2(54):54ra76. doi: 10.1126/scitranslmed.3001079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Zhang L, Su TP, Choi K, et al. P11 (S100A10) as a potential biomarker of psychiatric patients at risk of suicide. J Psychiatr Res. 2011;45(4):435–441. doi: 10.1016/j.jpsychires.2010.08.012. [DOI] [PubMed] [Google Scholar]
  • 111.Huang XL, Pawliczak R, Yao XL, et al. Interferon-gamma induces p11 gene and protein expression in human epithelial cells through interferon-gamma-activated sequences in the p11 promoter. J Biol Chem. 2003;278(11):9298–9308. doi: 10.1074/jbc.m212704200. [DOI] [PubMed] [Google Scholar]
  • 112.Warner-Schmidt JL, Vanover KE, Chen EY, Marshall JJ, Greengard P. Antidepressant effects of selective serotonin reuptake inhibitors (SSRIs) are attenuated by antiinflammatory drugs in mice and humans. Proc Natl Acad Sci USA. 2011;108(22):9262–9267. doi: 10.1073/pnas.1104836108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Chen G, Twyman R, Manji HK. p11 and gene therapy for severe psychiatric disorders: a practical goal? Sci Transl Med. 2010;2(54):54ps51. doi: 10.1126/scitranslmed.3001754. [DOI] [PubMed] [Google Scholar]
  • 114.Okuse K, Malik-Hall M, Baker MD, et al. Annexin II light chain regulates sensory neuron-specific sodium channel expression. Nature. 2002;417(6889):653–656. doi: 10.1038/nature00781. [DOI] [PubMed] [Google Scholar]
  • 115.Rezvanpour A, Shaw GS. Unique S100 target protein interactions. Gen Physiol Biophys. 2009;28:F39–F46. Spec No Focus. [PubMed] [Google Scholar]
  • 116.Foulkes T, Nassar MA, Lane T, et al. Deletion of annexin 2 light chain p11 in nociceptors causes deficits in somatosensory coding and pain behavior. J Neurosci. 2006;26(41):10499–10507. doi: 10.1523/JNEUROSCI.1997-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Mishra M, Paunesku T, Woloschak GE, et al. Gene expression analysis of frontotemporal lobar degeneration of the motor neuron disease type with ubiquitinated inclusions. Acta Neuropathol. 2007;114(1):81–94. doi: 10.1007/s00401-007-0240-7. [DOI] [PubMed] [Google Scholar]
  • 118.Paulson HL, Igo I. Genetics of dementia. Semin Neurol. 2011;31(5):449–460. doi: 10.1055/s-0031-1299784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Gauthier-Kemper A, Weissmann C, Golovyashkina N, et al. The frontotemporal dementia mutation R406W blocks tau’s interaction with the membrane in an annexin A2-dependent manner. J Cell Biol. 2011;192(4):647–661. doi: 10.1083/jcb.201007161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Bashir R, Britton S, Strachan T, et al. A gene related to Caenorhabditis elegans spermatogenesis factor fer-1 is mutated in limbgirdle muscular dystrophy type 2B. Nat Genet. 1998;20(1):37–42. doi: 10.1038/1689. [DOI] [PubMed] [Google Scholar]
  • 121.Liu J, Aoki M, Illa I, et al. Dysferlin, a novel skeletal muscle gene, is mutated in Miyoshi myopathy and limb girdle muscular dystrophy. Nat Genet. 1998;20(1):31–36. doi: 10.1038/1682. [DOI] [PubMed] [Google Scholar]
  • 122.Campanaro S, Romualdi C, Fanin M, et al. Gene expression profiling in dysferlinopathies using a dedicated muscle microarray. Hum Mol Genet. 2002;11(26):3283–3298. doi: 10.1093/hmg/11.26.3283. [DOI] [PubMed] [Google Scholar]
  • 123.Cagliani R, Magri F, Toscano A, et al. Mutation finding in patients with dysferlin deficiency and role of the dysferlin interacting proteins annexin A1 and A2 in muscular dystrophies. Hum Mutat. 2005;26(3):283. doi: 10.1002/humu.9364. [DOI] [PubMed] [Google Scholar]
  • 124.Lennon NJ, Kho A, Bacskai BJ, Perlmutter SL, Hyman BT, Brown RH., Jr. Dysferlin interacts with annexins A1 and A2 and mediates sarcolemmal wound-healing. J Biol Chem. 2003;278(50):50466–50473. doi: 10.1074/jbc.M307247200. [DOI] [PubMed] [Google Scholar]
  • 125.Cobelli N, Scharf B, Crisi GM, Hardin J, Santambrogio L. Mediators of the inflammatory response to joint replacement devices. Nat Rev Rheumatol. 2011;7(10):600–608. doi: 10.1038/nrrheum.2011.128. [DOI] [PubMed] [Google Scholar]
  • 126.Maitra R, Clement CC, Scharf B, et al. Endosomal damage and TLR2 mediated inflammasome activation by alkane particles in the generation of aseptic osteolysis. Mol Immunol. 2009;47(2-3):175–184. doi: 10.1016/j.molimm.2009.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Scharf B, Clement CC, Wu XX, et al. Annexin A2 binds to endosomes following organelle destabilization by particulate wear debris. Nat Commun. 2012;3:755–765. doi: 10.1038/ncomms1754. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES