2,381
Views
51
CrossRef citations to date
0
Altmetric
Extra View - Commissioned

S100 and annexin proteins identify cell membrane damage as the Achilles heel of metastatic cancer cells

&
Pages 502-509 | Received 24 Nov 2014, Accepted 02 Dec 2014, Published online: 16 Mar 2015

Abstract

Mechanical activity of cells and the stress imposed on them by extracellular environment is a constant source of injury to the plasma membrane (PM). In invasive tumor cells, increased motility together with the harsh environment of the tumor stroma further increases the risk of PM injury. The impact of these stresses on tumor cell plasma membrane and mechanism by which tumor cells repair the PM damage are poorly understood. Ca2+ entry through the injured PM initiates repair of the PM. Depending on the cell type, different organelles and proteins respond to this Ca2+ entry and facilitate repair of the damaged plasma membrane. We recently identified that proteins expressed in various metastatic cancers including Ca2+-binding EF hand protein S100A11 and its binding partner annexin A2 are used by tumor cells for plasma membrane repair (PMR). Here we will discuss the involvement of S100, annexin proteins and their regulation of actin cytoskeleton, leading to PMR. Additionally, we will show that another S100 member – S100A4 accumulates at the injured PM. These findings reveal a new role for the S100 and annexin protein up regulation in metastatic cancers and identify these proteins and PMR as targets for treating metastatic cancers.

Introduction

The phospholipid bilayer of the plasma membrane surrounds and physically separates the interior structures of the cell from the extracellular environment. It is selectively permeable to ions and organic molecules and yet maintains an essential osmotic barrier to the outside. Loss of this barrier function due to PM injury poses critical threat to single as well as multicellular organisms and thus cells rapidly repair PM lesions. Even in the shielding tissue environment where the tissue architecture provides the protective environment to restrict the injury by damaging forces, PM disruptions are frequent. Prevention of damage can be achieved by dynamic adaptations at a single cell or tissue level to limit the level of imposed stress, e.g., by actively modulating the epithelial layer to relieve mechanical forces. Still, many cells experience plasma membrane ruptures on a recurring basis that they need to cope with to maintain cell and tissue integrity. This is particularly evident in cells from, e.g., skin, lungs, gastrointestinal tract, skeletal and heart muscles, which reside in areas with high mechanical activity and hence increased damage frequency. Muscle and lung cells offer a good example of this as they are routinely wounded as a result of exercise and over stretching.Citation1,2 Defect in the muscle cell's ability to repair has been shown to result in muscular dystrophy,Citation3-Citation6 where poor repair of injured muscle cell membrane leads to cell death and tissue inflammation. Poor plasma membrane repair (PMR) is also associated with Niemann-Pick type A,Citation7 diabetes,Citation8 and Chediak-HigashiCitation9 syndrome and therapies targeting PMR have been shown to be effective in treating muscle and lung injuries.Citation10-Citation12 While much of the focus has been on disease resulting from poor PMR, we have recently identified that improved PMR is an important contributor for cancer metastasis.Citation13 Human breast cancer cells that turn metastatic through up-regulation of truncated EGF receptor (ErbB2) also need to up-regulate their PMR machinery, and absence of enhanced PMR compromises their invasive ability. Here we will briefly discuss the mechanisms involved in PMR, with a focus on the involvement of annexins, actin and S100 proteins. Additionally, we will describe the involvement of this process in cancer metastasis and discuss the potential for harnessing this novel aspect of tumor metastasis for developing new therapeutic approaches to target metastatic cancers.

The Membrane Repair Machinery

Studies over the past 2 decades have revealed that plasma membrane repair is a complex and active process that require membrane replacements, fusion events and cytoskeletal reorganization.Citation14 PMR is triggered by Ca2+ influx at the injury site due to over a thousand-fold gradient of calcium that exists across the plasma membrane.Citation15 The calcium influx triggers a versatile repair system that involves replacing or patching the injured membrane.Citation16 Fusion of intracellular vesicles around the wound perimeter to form a patch was revealed using the sea urchin egg.Citation17 This formed the basis for the patch model, according to which Ca2+ entry at the wound site triggers recruitment and homotypic fusion of vesicles at the wound site which then fuses with the plasma membrane to seal the wound.Citation17,18 In the sea urchin eggs these vesicles were identified as the yolk granules. In case of mammalian cells identification of lysosomes as the vesicles that undergo Ca2+-triggered fusion with the injured membrane led to the proposal that these are the patch forming vesicles in the mammalian cells.Citation19,20 Additionally, injury-triggered fusion of non-secretory vesicles called enlargeosomes,Citation21 and accumulation of mitochondria at the site of PM injuryCitation22 have also been found to be required for PMR.

In cells wounded by small pore forming toxins, PMR progresses by the replacement of the damaged cell membrane, such that the injured membrane area is physically removed by endocytosis,Citation23 blebbing,Citation24 or ectocytosis.Citation25 Shedding and endocytosis of damaged cell membrane by vesicles has also been shown for lesions formed by the membrane attack complex (MAC).Citation26,27 Lysosome exocytosis has been shown to be important for removal of injured membrane through the secretion of Acid Sphingomyelinase (ASMase).Citation23 Secreted ASMase hydrolyses the plasma membrane sphingomyelin to ceramide causing endocytosis of the pores in the plasma membrane formed by the pore forming toxins.Citation23 Cells from NPA patients, lacking ASMase protein, or Limb Girdle Muscular dystrophy 2B (LGMD2B) patients (lacking dysferlin protein), who show slow and poor injury-triggered ASMase secretion both show compromised PMR.Citation4,23 The PM repair deficit in these patient cells can be rescued by providing sphingomyelinase.Citation4 ASMase is also known to trigger cell membrane shedding and could contribute to the shedding of membrane by ectocytosis.Citation25,28,29 Thus, ASMase secreted by lysosome exocytosis may be involved in all the 3 processes (exocytosis, endocytosis, and ectocytosis) involved in PMR.

Recently, it was identified that vesicular shedding of damaged plasma membrane is not limited to pore forming toxins, but small focal injury can also trigger Endosomal Sorting Complex Required for Transport (ESCRT) III-mediated shedding of the injured PM.Citation30 We find that shedding of damaged membrane is required even following larger focal injury. This process is initiated by the EF hand calcium binding protein Apoptosis linked gene (ALG)-2, which accumulates at the injured PM and through binding its partner ALIX (ALG-2 interacting protein X) it facilitates accumulation of ESCRT III complex, resulting in cutting and shedding of damaged cell membrane.Citation31 Annexins are another group of calcium-binding protein that are implicated in sensing PM injury and formation of membrane blebs to shed the membrane damaged by pore forming toxin.Citation32 The members of the annexin protein family interact with the S100 proteins and anionic phospholipids to promote membrane segregation, vesicle trafficking, and vesicle fusion in a Ca2+-dependent manner.Citation33,34 Annexin and S100 proteins are also known regulators of actin cytoskeleton.Citation35,36

Actin cytoskeleton associated with the plasma membrane is another key regulator of PM repair. The cortical actin causes membrane tension, which prevents spontaneous resealing upon injury.Citation37 Ca2+ influx at the injury site triggers depolymerization of cortical actin as observed in Xenopus oocytes and Drosophila Embryos,Citation38 which is followed by resynthesis of the cortical actin. This progresses in way of a contractile actomyosin ring that forms a “purse string” - a circular structure and constricts circumferentially, in a manner coincident with the recruitment of filamentous actin (F-actin) and myosin-II at the wound borders.Citation39 Disruption of the actin cytoskeleton by either Cytochalasin D or Latrunculin B prevents actomyosin ring assembly and impairs wound closure.Citation39 PMR in Drosophila embryos also involves formation of actomyosin complex and a plasma membrane plug that is rapidly recruited from the surrounding edges of the membrane.Citation38 Additionally, intracellular vesicles are recruited to the wound perimeter enabling formation of a membrane patch within the actin ring to seal the wound.Citation14,38

Annexin and S100 Proteins in Membrane Repair

Annexin protein family, believed to have originated a billion years ago, has members in all major eukaryotic phyla.Citation40 Annexins have evolved extensively and independently in several eukaryotic lineages into a varied family.Citation40 This family of proteins appear to be instrumental in dealing with membrane stress - plant annexins are up-regulated during abiotic stress response and help to cope with it.Citation41,42 In humans there are 12 different annexin proteins with orthologs in most vertebrates.Citation40,33 They contain a unique COOH-terminal core domain that consists of 4 preserved structural repeats on which type-2 Ca2+ binding domains are located. Upon binding Ca2+, annexins bind the negatively charged phospholipids of the membrane to form a ternary complex bridging adjacent membranes.Citation43 The NH2-terminal region of annexins vary in length and sequence between family members and enables the individual protein to interact with distinct cytoplasmic partners such as the calcium binding S100 proteins.Citation44

S100 proteins are small (10-14 kDa), EF-hand-type Ca2+-binding proteins that upon Ca2+ activation exert both intracellular and extracellular functions. S100 genes are exclusively found in vertebrates and are clustered on chromosome 1q21 in humans (S100A1–S100A16).Citation45 The majority of the protein family members form symmetric noncovalent homodimers - a unique feature of the S100 proteins among the EF-hand protein family.Citation46 These proteins undergo a change in conformation upon binding calcium, which exposes a hydrophobic domain that can interact with the NH2-terminal region of specific annexins.Citation44 This interaction facilitates close apposition of adjacent phospholipid membranes and promotes membrane fusion.Citation33 Additionally, several pair of S100-annexin complexes such as S100A10 and annexin A2 (S100A10–ANXA2) can bind cytoskeletal components and have been associated with intracellular vesicle fusion.Citation47 S100A10 and ANXA2 are known to exist as a heterotetrameric complex where an S100A10 dimer resides in the center of the complex, interconnecting 2 Annexin A2 molecules.Citation44 Similarly, annexin A1 and S100A11 (also called S100C or calgizzarin) interact in a temporal Ca2+-dependent manner.Citation48 Several other pairs of annexin and S100 proteins have been discovered and it seems plausible that some S100 proteins can bind several annexins to exert their biological roles.Citation49

Annexins were first implicated in the process of PMR by gene expression analysis of muscle from LGMD2B mouse model, which identified injury-dependent interaction of ANXA1 and ANXA2 proteins with dysferlin.Citation50 Dysferlin is the muscle protein, lack of which results in poor repair of muscle fibers.Citation3 Due to the known role of annexins in aggregating membranes it was proposed that interaction of dysferlin and annexins may facilitate PMR through aggregation and fusion of intracellular vesicles.Citation50 However, work using zebrafish suggests that these dysferlin vesicles may be derived from the PM.Citation51 The first demonstration of a role of annexins in PMR in human cells was offered for ANXA1.Citation52 ANXA1 was shown to accumulate at the wound perimeter and use of ANXA1 inhibiting antibody, peptide, or dominant-negative mutant inhibited PMR.Citation52 Subsequently role of ANXA1 was identified in repair of plasma membrane injured by pore forming toxins, here ANXA1 was shown to facilitate shedding of the injured plasma membrane through the formation of blebs containing the membrane pores.Citation24 Annexins A5 and A6 have also been implicated in the process of PMR.Citation51,53 The ability of ANXA5 to assemble into 2-dimensional arrays at the sites of membrane injury in response to Ca2+ has been implicated in preventing wound expansion by keeping the membrane edges together. This is prevented in cells lacking ANXA5 causing PMR defect. Addition of exogenous recombinant ANXA5 protein rescues this PMR defect.Citation53 ANXA6 on the other hand forms a structure termed as “repair cap” at the site of injury, which facilitates healing of injured muscle fibers.Citation54 ANXA6 was also shown to be involved in shedding of microvesicles containing portions of the PM with the lesions formed by streptolysin O. Here, ANXA6 was shown to be activated and recruited to the site of injury at lower Ca2+ concentration as compared to ANXA1 suggesting a potential for sequential recruitment of annexins to facilitate PMR.Citation55

Enhanced PMR is Needed for Tumor metastasis - Role of Annexin and S100 Proteins

Cellular membranes of cancer cells are known to be destabilized – we have previously shown lysosomal membrane of tumor cells are more fragileCitation56,57 and that death induced by lysosomal membrane pemebilization is prominent in cancer cells.Citation56 This fragility has been harnessed for the development of novel therapeutics.Citation58,59 Similar to lysosomal membrane, PM of cancer cells are more unstable and have reduced stiffness, which is caused by an increase in saturated phospholipids.Citation60 The PM stiffness is inversely correlated with the ability of the cancer cell to migrate and invade 3-dimensional matrix.Citation60 The greater motility of metastatic cells and increase in mechanical stress due to invasion of the tissue matrix can increase PM damage. We recently identified this to be the case, as PM of invasive cancer cells suffered greater damage.Citation13 In response, we find these invasive cancer cells enhance their PMR by up regulating expression of S100A11 and annexin A2 proteins.Citation13 Annexins and S100 proteins are commonly up regulated in variety of cancers.Citation61,62 Specifically, S100A11 is overexpressed in several tumors, where it is associated with metastasis and poor disease prognosis.Citation63-Citation65 S100A11 is enriched in pseudopodia of metastatic cancer cells and is required for forming actin-dependent pseudopodial protrusions, which facilitates migration of the tumor cells.Citation66

To study the involvement of PM damage and repair in cancer cells as they enhance their motility and invasiveness, we made use of the MCF7 human breast cancer cell model. Here invasiveness was modulated by ectopically expressing a truncated ErbB2 oncogene (p95ErbB2).Citation67 The p95ErbB2 oncogene mimics constitutively active cleaved form of ErbB2 oncoprotein commonly found in aggressive breast cancers.Citation68 Increased invasiveness of these cells resulted in greater damage to the PM of these cells. Signaling through p95ErbB2 caused increase in the expression of the S100A11, a protein that we find enhances repair of the injured PM. Upon injury, S100A11 co-accumulates with ANXA2 at the site of PM repair. Co-accumulation of these proteins at the repair site is mutually dependent and is independent of ANXA1 - the other binding partner of S100A11. While ANXA1 is also recruited to the injured cell membrane, it localizes away from S100A11-ANXA2 complex. Greater Ca2+-dependent binding of S100A11 with ANXA2 as compared with ANXA1Citation69 could be responsible for this differential response of the 2 annexins. In resting cell, ANXA1, ANXA2 and S100A11 are all predominantly cytosolic and the PM is supported by a layer of cortical actin. The PM-associated cortical F-actin is necessary to support the plasma membrane, but it also creates tension that can inhibit passive resealing of the PM after injury.Citation70 Following injury, cortical actin depolymerizes at the site of injury, arguably by actin severing proteins. This reduces cortical tension and avoids further damaging the injured PM. Also, by the collapse of the neighboring wounded edges these membranes are brought together to facilitate their eventual fusion. As such, removal of cortical actin can facilitate repair of the wounded membrane.Citation71 We find that PM injury-triggered ANXA2 and S100A11 accumulation is followed by a rapid buildup of F-actin at the site of repair.Citation13 S100A11–ANXA2 can bind F-actin and decrease the depolymerization rate of preformed actin filaments.Citation72,73 Thus, these proteins preserve existing F-actin and allow buildup of new F-actin around the injury site (). Absence of either of these proteins prevents F-actin buildup following injury and prevents PMR, similar to the drugs that alter actin polymerization or depolymerization.Citation13,74 Cortical F-actin deploymerization around the injury site is followed by the recruitment of ANXA1 at the damaged PM and excision of this damaged membrane. Loss of ANXA2 and S100A11 and pharmacological inhibition of F-actin buildup prevent PMR by blocking excision of the damaged part of the PM marked by ANXA1. Thus, actin remodeling is required for the tumor cells to excise the injured part of the PM indicating that ANXA2 and S100A11 facilitate PMR, in part, by regulating F-actin buildup and excision of the damaged part of the PM at the site of injury.Citation13

Figure 1. Mechanism and need of plasma membrane repair in metastatic tumor cells. In the process of invading tumor stroma the metastatic tumor cells experience increased damage to their PM which is facilitated by the increased expression of proteins such as Annexin A2 and S100A11. These proteins regulate Ca2+-dependent F-actin growth, which provides support to the repairing plasma membrane and potentially facilitates actin based accumulation of repair vesicles derived from e.g. PM or endosomes providing membrane to repair the damaged membrane. Lack of annexin A2 or S100A11 proteins compromises these activity and result in poor PM repair of the invading cell. This triggers calcium overload and leakage of the cytoplasmic content eventually leading to the death of the invading cell. Thus regulating these and other proteins involved in PM repair in metastatic cells could provide an avenue to target metastatic potential of the tumor cell. ECM: Extra cellular matrix.

Figure 1. Mechanism and need of plasma membrane repair in metastatic tumor cells. In the process of invading tumor stroma the metastatic tumor cells experience increased damage to their PM which is facilitated by the increased expression of proteins such as Annexin A2 and S100A11. These proteins regulate Ca2+-dependent F-actin growth, which provides support to the repairing plasma membrane and potentially facilitates actin based accumulation of repair vesicles derived from e.g. PM or endosomes providing membrane to repair the damaged membrane. Lack of annexin A2 or S100A11 proteins compromises these activity and result in poor PM repair of the invading cell. This triggers calcium overload and leakage of the cytoplasmic content eventually leading to the death of the invading cell. Thus regulating these and other proteins involved in PM repair in metastatic cells could provide an avenue to target metastatic potential of the tumor cell. ECM: Extra cellular matrix.

ANXA2 has Ca2+ dependent phospholipid binding ability, which enables aggregation of endosomes and other vesicles.Citation75 We found that upregulation of S100A11 and ANXA2 does not enhance injury triggered fusion of the endosomes/lysosomes.Citation13 However, in light of the ability of these proteins to nucleate polymerization of cortical F-actin at the endosomesCitation76 they may facilitate accumulation of endosomal vesicles at the site of injury. The buildup of the cortical F-actin together with presence of vesicular endomembrane at the wounded edges of the plasma membrane will facilitate PMR at the repair site marked by the S100A11–ANXA2 complex and excise the damaged part of the PM marked by ANXA1 (). The buildup of cortical actin is analogous to F-actin drawstring formation during healing of injured Xenopus oocyte cell membrane.Citation39 Thus, by analogy we believe F-actin buildup may pull the wounded membrane edges together during excision and vesicle aggregation and F-actin assembly mediated by the S100A11–ANXA2 complex may help with PMR by facilitating vesicle fusion and cortical actin buildup. Additionally, it may help with vesicle fusion, as Ca2+-regulated F-actin dynamics at the cell membrane facilitates vesicle fusion.Citation77 These findings demonstrate that invasive cancer cells are dependent on efficient PMR system to cope with elevated rate of cell injury and that they rely on the S100A11–ANXA2 complex to facilitate plasma membrane repair.

PMR as Target for Therapeutic Intervention

While the diseases linked to altered PMR are caused primarily due to a defect in this process,Citation37 tumor metastasis offers a counter example to such diseases.Citation13 This presents an interesting therapeutic scenario since lessons learned from most of the PMR associated diseases could be applied to control tumor metastasis. In one such recent study of LGMD 2B, we identified that dysferlin protein facilitates injury-triggered secretion of the lysosomal enzyme Acid Sphingomyelinase (ASMase).Citation4 This leads to poor repair of LGMD2B patient muscle cells, a defect that is rescued by providing this enzyme function exogenously.Citation4 Such a role of ASMase suggests that inhibiting ASMase would be an attractive therapeutic target against cancers. Thus, it is interesting that in an independent study we identified acid sphingomyelinase (ASM) inhibition as a target for cancer therapy.Citation58 While in the use of ASMase inhibitors we focused our attention on destabilization of the lysosomal membrane, our findings with the LGMD2B patient cells suggest that such an inhibition may have additional unrealized benefit in the cancer cells by inhibiting PMR. Targeting this and other regulators of PMR is a novel therapeutic approach to control tumor metastasis, one whose potential is yet to be properly explored. In order to realize this potential it would be valuable to identify specific molecular regulators of PMR that tumor cells rely upon. Our results that inhibiting PMR by depletion of S100A11 in the metastatic cells makes them unable to invade 3D tissue matrix offers support for the utility of such a targeted strategy.Citation13

S100 and annexins proteins are among the most frequently dysregulated proteins in neoplasia and are overexpressed in various cancers.Citation61 Thus members of these proteins families are attractive candidates to compromise PMR in tumor cells. As discussed above members of the annexin family - annexin A1, A2, A5 and A6 have already been implicated in PMR and over expression of some of these annexins is directly correlated with aggressive clinical stage in colorectal, pancreatic and brain tumors and linked to metastatic progression.Citation78 Further, changes in annexin A3 and A4 expression has been associated with chemo resistance in ovarian cancer cells.Citation79,80 These findings make a case for a potential therapeutic approach against tumor metastasis that could involve targeting PMR by regulating expression of specific annexins. However, in view of the sequence homology of the annexins and use of multiple annexins in PMR, suggests the likelihood of functional redundancy between the family members. In line with this, changes in the expression of one annexin can profoundly affect the expression levels of another suggesting a strict functional fail-safe mechanism.Citation81 Thus, efficient pharmacological strategies to target annexins may require that several family members are inhibited simultaneously, e.g. by targeting the conserved annexin core domain. Alternatively, annexin function can be compromised by restricting the interaction with its S100 protein binding partners, e.g., by blocking the interaction between ANXA2 and S100A11. Additionally, in light of our identification of actin as a key mediator of PMR through the action of S100A11– ANXA2 complex proteins, regulation of actin polymerization offers another set of targets. Such targets have an additional advantage of inhibiting metastasis by inhibiting the motility and thus invasiveness of the cancer cells.

Aside from annexins, S100 proteins offer another potential target. These proteins are implicated in multiple stages of cancer and are commonly upregulated and associated with tumor progression in various cancers.Citation62 However, with S100A11 as an exceptionCitation13 their direct regulatory role in PMR has not yet been characterized. Other S100 family members may also function as Ca2+ triggered switches that can regulate PMR through interaction with other annexins at the plasma membrane. To this end, we looked at the response of S100A4, which is a well-documented metastasis-promoting protein that is overexpressed in various types of cancers and exerts its function both intra- and extracellularly.Citation82,83 Our analysis of S100A4 by live cell imaging of PMR in HeLa cells show that injury triggers S100A4 co-accumulation with S100A11 at the injury site (). In a manner similar to S100A11, S100A4 accumulation precedes the accumulation of F-actin at the site of repair (). It remains to be established whether S100A4 can facilitate F-actin accumulation or PMR in invasive cancer cells. If so, in view of the prevalence of metastatic cancers associated with S100A4 overexpression, S100A4 would be an attractive candidate to broadly target inhibition of tumor metastasis by regulating PMR.

Figure 2. S100A4 and S100A11 are recruited to the site of injury before actin build-up. HeLa cells were transiently transfected to express (A) S100A4-GFP and S100A11-RFP or (B) S100A4-GFP and the F-actin reporter protein Utrophin-mCherry. The cells were injured focally by a pulsed laser (white arrow) and the response of the proteins was monitored live as the cells underwent repair in the same way as we described previously.Citation13 The plot shows the kinetics of accumulation of individual proteins at the repair site (blue arrow) marked by the white box.

Figure 2. S100A4 and S100A11 are recruited to the site of injury before actin build-up. HeLa cells were transiently transfected to express (A) S100A4-GFP and S100A11-RFP or (B) S100A4-GFP and the F-actin reporter protein Utrophin-mCherry. The cells were injured focally by a pulsed laser (white arrow) and the response of the proteins was monitored live as the cells underwent repair in the same way as we described previously.Citation13 The plot shows the kinetics of accumulation of individual proteins at the repair site (blue arrow) marked by the white box.

A common feature of the S100 proteins is the presence of evolutionarily primitive Ca2+-binding EF-hand domains. Calpains are another member of the EF -hand protein family implicated in cancer progression and PMR.Citation84,85 We recently identified yet another member of the EF -hand protein family (Apoptosis linked gene 2 - ALG-2) that can regulate PMR.Citation31 While Calpains regulate various aspects of tumor progression,Citation84 their involvement in PMR in cancer cells has not been elucidated. In case of ALG-2 we found that it facilitates PMR by enabling shedding of damaged cell membrane through the Ca2+-triggered accumulation of the ESCRT III complex at the injured cell membrane.Citation31 The ESCRT machinery is involved in membrane curvature and cleavage.Citation86 While the role of ESCRT machinery in cancer is disputed,Citation87 metastatic tumor cells are long known to make use of membrane cleavage to shed their PM.Citation88,89 Shedding of PM by tumor cells is gaining wider recognition due to their role in the process of cell-cell communication through the use of extracellular vesicles.Citation90 Thus, our finding that PMR of metastatic cells involves excision and shedding of PM may extend beyond cell survival to extracellular communication that facilitates tumor growth and metastasis. We hope this new aspect of tumor metastasis and its potential involvement in tumor cell invasion and signaling will attract strong translational interest leading to the development of new cancer therapeutic strategy targeting this Achille's heel of cancers.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgment

We thank Birgitte Grum-Schwensen and Noona Ambartsumian for the S100A4-GFP plasmid, University of Copenhagen.

References

  • McNeil PL, Khakee R. Disruptions of muscle fiber plasma membranes. Role in exercise-induced damage. Am J Pathol 1992; 140:1097-109; PMID:1374591
  • Gajic O, Lee J, Doerr CH, Berrios JC, Myers JL, Hubmayr RD. Ventilator-induced cell wounding and repair in the intact lung. Am J Respir Crit Care Med 2003; 167:1057-63; PMID:12480613; http://dx.doi.org/10.1164/rccm.200208-889OC
  • Bansal D, Miyake K, Vogel SS, Groh S, Chen CC, Williamson R, McNeil PL, Campbell KP. Defective membrane repair in dysferlin-deficient muscular dystrophy. Nature 2003; 423:168-72; PMID:12736685; http://dx.doi.org/10.1038/nature01573
  • Defour A, Van der Meulen JH, Bhat R, Bigot A, Bashir R, Nagaraju K, Jaiswal JK. Dysferlin regulates cell membrane repair by facilitating injury-triggered acid sphingomyelinase secretion. Cell Death Dis 2014; 5:e1306; PMID:24967968; http://dx.doi.org/10.1038/cddis.2014.272
  • Jaiswal JK, Marlow G, Summerill G, Mahjneh I, Mueller S, Hill M, Miyake K, Haase H, Anderson LV, Richard I, et al. Patients with a non-dysferlin Miyoshi myopathy have a novel membrane repair defect. Traffic 2007; 8:77-88; PMID:17132147; http://dx.doi.org/10.1111/j.1600-0854.2006.00505.x
  • Cheng X, Zhang X, Gao Q, Ali Samie M, Azar M, Tsang WL, Dong L, Sahoo N, Li X, Zhuo Y, et al. The intracellular Ca(2+) channel MCOLN1 is required for sarcolemma repair to prevent muscular dystrophy. Nat Med 2014; 20:1187-92; PMID:25216637; http://dx.doi.org/10.1038/nm.3611
  • Tam C, Idone V, Devlin C, Fernandes MC, Flannery A, He X, Schuchman E, Tabas I, Andrews NW. Exocytosis of acid sphingomyelinase by wounded cells promotes endocytosis and plasma membrane repair. J Cell Biol 2010; 189:1027-38; PMID:20530211; http://dx.doi.org/10.1083/jcb.201003053
  • Howard AC, McNeil AK, Xiong F, Xiong WC, McNeil PL. A novel cellular defect in diabetes: membrane repair failure. Diabetes 2011; 60:3034-43; PMID:21940783; http://dx.doi.org/10.2337/db11-0851
  • Huynh C, Roth D, Ward DM, Kaplan J, Andrews NW. Defective lysosomal exocytosis and plasma membrane repair in Chediak-Higashi/beige cells. Proc Natl Acad Sci USA 2004; 101:16795-800; PMID:15557559; http://dx.doi.org/10.1073/pnas.0405905101
  • Yasuda S, Townsend D, Michele DE, Favre EG, Day SM, Metzger JM. Dystrophic heart failure blocked by membrane sealant poloxamer. Nature 2005; 436:1025-9; PMID:16025101; http://dx.doi.org/10.1038/nature03844
  • Weisleder N, Takizawa N, Lin P, Wang X, Cao C, Zhang Y, Tan T, Ferrante C, Zhu H, Chen PJ, et al. Recombinant MG53 protein modulates therapeutic cell membrane repair in treatment of muscular dystrophy. Sci Transl Med 2012; 4:139ra85; PMID:22723464; http://dx.doi.org/10.1126/scitranslmed.3003921
  • Jia Y, Chen K, Lin P, Lieber G, Nishi M, Yan R, Wang Z, Yao Y, Li Y, Whitson BA, et al. Treatment of acute lung injury by targeting MG53-mediated cell membrane repair. Nat Commun 2014; 5:4387; PMID:25034454
  • Jaiswal JK, Lauritzen SP, Scheffer L, Sakaguchi M, Bunkenborg J, Simon SM, Kallunki T, Jaattela M, Nylandsted J. S100A11 is required for efficient plasma membrane repair and survival of invasive cancer cells. Nat Commun 2014; 5:3795; PMID:24806074; http://dx.doi.org/10.1038/ncomms4795
  • Abreu-Blanco MT, Verboon JM, Parkhurst SM. Single cell wound repair: dealing with life's little traumas. Bioarchitecture 2011; 1:114-21; PMID:21922041; http://dx.doi.org/10.4161/bioa.1.3.17091
  • Jaiswal JK. Calcium - how and why? J Biosci 2001; 26:357-63; PMID:11568481; http://dx.doi.org/10.1007/BF02703745
  • McNeil PL, Vogel SS, Miyake K, Terasaki M. Patching plasma membrane disruptions with cytoplasmic membrane. J Cell Sci 2000; 113(Pt 11):1891-902; PMID:10806100
  • Terasaki M, Miyake K, McNeil PL. Large plasma membrane disruptions are rapidly resealed by Ca2+-dependent vesicle-vesicle fusion events. J Cell Biol 1997; 139:63-74; PMID:9314529; http://dx.doi.org/10.1083/jcb.139.1.63
  • Steinhardt RA, Bi G, Alderton JM. Cell membrane resealing by a vesicular mechanism similar to neurotransmitter release. Science 1994; 263:390-3; PMID:7904084; http://dx.doi.org/10.1126/science.7904084
  • Reddy A, Caler EV, Andrews NW. Plasma membrane repair is mediated by Ca(2+)-regulated exocytosis of lysosomes. Cell 2001; 106:157-69; PMID:11511344; http://dx.doi.org/10.1016/S0092-8674(01)00421-4
  • McNeil PL. Repairing a torn cell surface: make way, lysosomes to the rescue. J Cell Sci 2002; 115:873-9; PMID:11870206
  • Lorusso A, Covino C, Priori G, Bachi A, Meldolesi J, Chieregatti E. Annexin2 coating the surface of enlargeosomes is needed for their regulated exocytosis. EMBO J 2006; 25:5443-56; PMID:17082761; http://dx.doi.org/10.1038/sj.emboj.7601419
  • Sharma N, Medikayala S, Defour A, Rayavarapu S, Brown KJ, Hathout Y, Jaiswal JK. Use of quantitative membrane proteomics identifies a novel role of mitochondria in healing injured muscles. J Biol Chem 2012; 287:30455-67; PMID:22778268; http://dx.doi.org/10.1074/jbc.M112.354415
  • Idone V, Tam C, Goss JW, Toomre D, Pypaert M, Andrews NW. Repair of injured plasma membrane by rapid Ca2+-dependent endocytosis. J Cell Biol 2008; 180:905-14; PMID:18316410; http://dx.doi.org/10.1083/jcb.200708010
  • Babiychuk EB, Monastyrskaya K, Potez S, Draeger A. Blebbing confers resistance against cell lysis. Cell Death Differ 2011; 18:80-9; PMID:20596076; http://dx.doi.org/10.1038/cdd.2010.81
  • Keyel PA, Loultcheva L, Roth R, Salter RD, Watkins SC, Yokoyama WM, Heuser JE. Streptolysin O clearance through sequestration into blebs that bud passively from the plasma membrane. J Cell Sci 2011; 124:2414-23; PMID:21693578; http://dx.doi.org/10.1242/jcs.076182
  • Morgan BP. Complement membrane attack on nucleated cells: resistance, recovery and non-lethal effects. Biochem J 1989; 264:1-14; PMID:2690818
  • Moskovich O, Herzog LO, Ehrlich M, Fishelson Z. Caveolin-1 and dynamin-2 are essential for removal of the complement C5b-9 complex via endocytosis. J Biol Chem 2012; 287:19904-15; PMID:22528500; http://dx.doi.org/10.1074/jbc.M111.333039
  • Bianco F, Perrotta C, Novellino L, Francolini M, Riganti L, Menna E, Saglietti L, Schuchman EH, Furlan R, Clementi E, et al. Acid sphingomyelinase activity triggers microparticle release from glial cells. EMBO J 2009; 28:1043-54; PMID:19300439; http://dx.doi.org/10.1038/emboj.2009.45
  • Gills JJ, Zhang C, Abu-Asab MS, Castillo SS, Marceau C, LoPiccolo J, Kozikowski AP, Tsokos M, Goldkorn T, Dennis PA. Ceramide mediates nanovesicle shedding and cell death in response to phosphatidylinositol ether lipid analogs and perifosine. Cell Death Dis 2012; 3:e340; PMID:22764099; http://dx.doi.org/10.1038/cddis.2012.72
  • Jimenez AJ, Maiuri P, Lafaurie-Janvore J, Divoux S, Piel M, Perez F. ESCRT machinery is required for plasma membrane repair. Science 2014; 343:1247136; PMID:24482116; http://dx.doi.org/10.1126/science.1247136
  • Scheffer LSSC, Sharma N, Medikayala S, Brown KJ, Defour A, Jaiswal JK. Mechanism of Ca2+-triggered ESCRT assembly and regulation of cell membrane repair. Nat Commun 2014; 5:5646; PMID: 25534348; http://dx.doi.org/10.1038/ncomms6646
  • Draeger A, Monastyrskaya K, Babiychuk EB. Plasma membrane repair and cellular damage control: the annexin survival kit. Biochem Pharmacol 2011; 81:703-12; PMID:21219882; http://dx.doi.org/10.1016/j.bcp.2010.12.027
  • Gerke V, Moss SE. Annexins: from structure to function. Physiol Rev 2002; 82:331-71; PMID:11917092
  • Miwa N, Uebi T, Kawamura S. S100-annexin complexes–biology of conditional association. FEBS J 2008; 275:4945-55; PMID:18795952; http://dx.doi.org/10.1111/j.1742-4658.2008.06653.x
  • Zhao XQ, Naka M, Muneyuki M, Tanaka T. Ca(2+)-dependent inhibition of actin-activated myosin ATPase activity by S100C (S100A11), a novel member of the S100 protein family. Biochem Biophys Res Commun 2000; 267:77-9; PMID:10623577; http://dx.doi.org/10.1006/bbrc.1999.1918
  • Hayes MJ, Rescher U, Gerke V, Moss SE. Annexin-actin interactions. Traffic 2004; 5:571-6; PMID:15260827; http://dx.doi.org/10.1111/j.1600-0854.2004.00210.x
  • McNeil PL, Steinhardt RA. Plasma membrane disruption: repair, prevention, adaptation. Annu Rev Cell Dev Biol 2003; 19:697-731; PMID:14570587; http://dx.doi.org/10.1146/annurev.cellbio.19.111301.140101
  • Abreu-Blanco MT, Verboon JM, Parkhurst SM. Cell wound repair in Drosophila occurs through three distinct phases of membrane and cytoskeletal remodeling. J Cell Biol 2011; 193:455-64; PMID:21518790; http://dx.doi.org/10.1083/jcb.201011018
  • Bement WM, Mandato CA, Kirsch MN. Wound-induced assembly and closure of an actomyosin purse string in Xenopus oocytes. Curr Biol 1999; 9:579-87; PMID:10359696; http://dx.doi.org/10.1016/S0960-9822(99)80261-9
  • Moss SE, Morgan RO. The annexins. Genome Biol 2004; 5:219; PMID:15059252; http://dx.doi.org/10.1186/gb-2004-5-4-219
  • Konopka-Postupolska D, Clark G, Goch G, Debski J, Floras K, Cantero A, Fijolek B, Roux S, Hennig J. The role of annexin 1 in drought stress in Arabidopsis. Plant Physiol 2009; 150:1394-410; PMID:19482919; http://dx.doi.org/10.1104/pp.109.135228
  • Clark G, Konopka-Postupolska D, Hennig J, Roux S. Is annexin 1 a multifunctional protein during stress responses? Plant Signal Behav 2010; 5:303-7; PMID:20215861; http://dx.doi.org/10.4161/psb.5.3.10835
  • Swairjo MA, Concha NO, Kaetzel MA, Dedman JR, Seaton BA. Ca(2+)-bridging mechanism and phospholipid head group recognition in the membrane-binding protein annexin V. Nat Struct Biol 1995; 2:968-74; PMID:7583670; http://dx.doi.org/10.1038/nsb1195-968
  • Rintala-Dempsey AC, Rezvanpour A, Shaw GS. S100-annexin complexes–structural insights. FEBS J 2008; 275:4956-66; PMID:18795951; http://dx.doi.org/10.1111/j.1742-4658.2008.06654.x
  • Marenholz I, Heizmann CW, Fritz G. S100 proteins in mouse and man: from evolution to function and pathology (including an update of the nomenclature). Biochem Biophys Res Commun 2004; 322:1111-22; PMID:15336958; http://dx.doi.org/10.1016/j.bbrc.2004.07.096
  • Santamaria-Kisiel L, Rintala-Dempsey AC, Shaw GS. Calcium-dependent and -independent interactions of the S100 protein family. Biochem J 2006; 396:201-14; PMID:16683912; http://dx.doi.org/10.1042/BJ20060195
  • Osborn M, Johnsson N, Wehland J, Weber K. The submembranous location of p11 and its interaction with the p36 substrate of pp60 src kinase in situ. Exp Cell Res 1988; 175:81-96; PMID:3126079; http://dx.doi.org/10.1016/0014-4827(88)90257-1
  • Seemann J, Weber K, Gerke V. Annexin I targets S100C to early endosomes. FEBS Lett 1997; 413:185-90; PMID:9287141; http://dx.doi.org/10.1016/S0014-5793(97)00911-3
  • Liu Y, Myrvang HK, Dekker LV. Annexin A2 complexes with S100 proteins: structure, function and pharmacological manipulation. Br J Pharmacol 2014; PMID:25303710
  • 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; 278:50466-73; PMID:14506282; http://dx.doi.org/10.1074/jbc.M307247200
  • Roostalu U, Strahle U. In vivo imaging of molecular interactions at damaged sarcolemma. Dev Cell 2012; 22:515-29; PMID:22421042; http://dx.doi.org/10.1016/j.devcel.2011.12.008
  • McNeil AK, Rescher U, Gerke V, McNeil PL. Requirement for annexin A1 in plasma membrane repair. J Biol Chem 2006; 281:35202-7; PMID:16984915; http://dx.doi.org/10.1074/jbc.M606406200
  • Bouter A, Gounou C, Berat R, Tan S, Gallois B, Granier T, d'Estaintot BL, Poschl E, Brachvogel B, Brisson AR. Annexin-A5 assembled into two-dimensional arrays promotes cell membrane repair. Nat Commun 2011; 2:270; PMID:21468022; http://dx.doi.org/10.1038/ncomms1270
  • Swaggart KA, Demonbreun AR, Vo AH, Swanson KE, Kim EY, Fahrenbach JP, Holley-Cuthrell J, Eskin A, Chen Z, Squire K, et al. Annexin A6 modifies muscular dystrophy by mediating sarcolemmal repair. Proc Natl Acad Sci U S A 2014; 111:6004-9; PMID:24717843; http://dx.doi.org/10.1073/pnas.1324242111
  • Potez S, Luginbuhl M, Monastyrskaya K, Hostettler A, Draeger A, Babiychuk EB. Tailored protection against plasmalemmal injury by annexins with different Ca2+ sensitivities. J Biol Chem 2011; 286:17982-91; PMID:21454475; http://dx.doi.org/10.1074/jbc.M110.187625
  • Kallunki T, Olsen OD, Jaattela M. Cancer-associated lysosomal changes: friends or foes? Oncogene 2013; 32:1995-2004; PMID:22777359; http://dx.doi.org/10.1038/onc.2012.292
  • Nylandsted J, Gyrd-Hansen M, Danielewicz A, Fehrenbacher N, Lademann U, Hoyer-Hansen M, Weber E, Multhoff G, Rohde M, Jaattela M. Heat shock protein 70 promotes cell survival by inhibiting lysosomal membrane permeabilization. J Exp Med 2004; 200:425-35; PMID:15314073; http://dx.doi.org/10.1084/jem.20040531
  • Petersen NH, Olsen OD, Groth-Pedersen L, Ellegaard AM, Bilgin M, Redmer S, Ostenfeld MS, Ulanet D, Dovmark TH, Lonborg A, et al. Transformation-associated changes in sphingolipid metabolism sensitize cells to lysosomal cell death induced by inhibitors of acid sphingomyelinase. Cancer Cell 2013; 24:379-93; PMID:24029234; http://dx.doi.org/10.1016/j.ccr.2013.08.003
  • Ellegaard AM, Groth-Pedersen L, Oorschot V, Klumperman J, Kirkegaard T, Nylandsted J, Jaattela M. Sunitinib and SU11652 inhibit acid sphingomyelinase, destabilize lysosomes, and inhibit multidrug resistance. Mol Cancer Ther 2013; 12:2018-30; PMID:23920274; http://dx.doi.org/10.1158/1535-7163.MCT-13-0084
  • Swaminathan V, Mythreye K, O'Brien ET, Berchuck A, Blobe GC, Superfine R. Mechanical stiffness grades metastatic potential in patient tumor cells and in cancer cell lines. Cancer Res 2011; 71:5075-80; PMID:21642375; http://dx.doi.org/10.1158/0008-5472.CAN-11-0247
  • Mussunoor S, Murray GI. The role of annexins in tumour development and progression. J Pathol 2008; 216:131-40; PMID:18698663; http://dx.doi.org/10.1002/path.2400
  • Salama I, Malone PS, Mihaimeed F, Jones JL. A review of the S100 proteins in cancer. Eur J Surg Oncol 2008; 34:357-64; PMID:17566693; http://dx.doi.org/10.1016/j.ejso.2007.04.009
  • Melle C, Ernst G, Schimmel B, Bleul A, Von EF. Colon-derived liver metastasis, colorectal carcinoma, and hepatocellular carcinoma can be discriminated by the Ca(2+)-binding proteins S100A6 and S100A11. PLoS One 2008; 3:e3767; PMID:19048101; http://dx.doi.org/10.1371/journal.pone.0003767
  • Rehman I, Azzouzi AR, Cross SS, Deloulme JC, Catto JW, Wylde N, Larre S, Champigneuille J, Hamdy FC. Dysregulated expression of S100A11 (calgizzarin) in prostate cancer and precursor lesions. Hum Pathol 2004; 35:1385-91; PMID:15668896; http://dx.doi.org/10.1016/j.humpath.2004.07.015
  • Xiao MB, Jiang F, Ni WK, Chen BY, Lu CH, Li XY, Ni RZ. High expression of S100A11 in pancreatic adenocarcinoma is an unfavorable prognostic marker. Med Oncol 2011; 29(3):1886-91; PMID:21912994; http://dx.doi.org/10.1007/s12032-011-0058-y
  • Shankar J, Messenberg A, Chan J, Underhill TM, Foster LJ, Nabi IR. Pseudopodial actin dynamics control epithelial-mesenchymal transition in metastatic cancer cells. Cancer Res 2010; 70:3780-90; PMID:20388789; http://dx.doi.org/10.1158/0008-5472.CAN-09-4439
  • Egeblad M, Mortensen OH, Jaattela M. Truncated ErbB2 receptor enhances ErbB1 signaling and induces reversible, ERK-independent loss of epithelial morphology. Int J Cancer 2001; 94:185-91; PMID:11668496; http://dx.doi.org/10.1002/ijc.1459
  • Rafn B, Nielsen CF, Andersen SH, Szyniarowski P, Corcelle-Termeau E, Valo E, Fehrenbacher N, Olsen CJ, Daugaard M, Egebjerg C, et al. ErbB2-driven breast cancer cell invasion depends on a complex signaling network activating myeloid zinc finger-1-dependent cathepsin B expression. Mol Cell 2012; 45:764-76; PMID:22464443; http://dx.doi.org/10.1016/j.molcel.2012.01.029
  • Rintala-Dempsey AC, Santamaria-Kisiel L, Liao Y, Lajoie G, Shaw GS. Insights into S100 target specificity examined by a new interaction between S100A11 and annexin A2. Biochemistry 2006; 45:14695-705; PMID:17144662; http://dx.doi.org/10.1021/bi061754e
  • Togo T, Krasieva TB, Steinhardt RA. A decrease in membrane tension precedes successful cell-membrane repair. Mol Biol Cell 2000; 11:4339-46; PMID:11102527; http://dx.doi.org/10.1091/mbc.11.12.4339
  • Miyake K, McNeil PL, Suzuki K, Tsunoda R, Sugai N. An actin barrier to resealing. J Cell Sci 2001; 114:3487-94; PMID:11682608
  • Hayes MJ, Shao D, Bailly M, Moss SE. Regulation of actin dynamics by annexin 2. EMBO J 2006; 25:1816-26; PMID:16601677; http://dx.doi.org/10.1038/sj.emboj.7601078
  • Sakaguchi M, Miyazaki M, Inoue Y, Tsuji T, Kouchi H, Tanaka T, Yamada H, Namba M. Relationship between contact inhibition and intranuclear S100C of normal human fibroblasts. J Cell Biol 2000; 149:1193-206; PMID:10851017; http://dx.doi.org/10.1083/jcb.149.6.1193
  • Burkel BM, Benink HA, Vaughan EM, von DG, Bement WM. A Rho GTPase signal treadmill backs a contractile array. Dev Cell 2012; 23:384-96; PMID:22819338; http://dx.doi.org/10.1016/j.devcel.2012.05.025
  • Glenney JR, Jr., Tack B, Powell MA. Calpactins: two distinct Ca++-regulated phospholipid- and actin-binding proteins isolated from lung and placenta. J Cell Biol 1987; 104:503-11; PMID:2950118; http://dx.doi.org/10.1083/jcb.104.3.503
  • Morel E, Parton RG, Gruenberg J. Annexin A2-dependent polymerization of actin mediates endosome biogenesis. Dev Cell 2009; 16:445-57; PMID:19289089; http://dx.doi.org/10.1016/j.devcel.2009.01.007
  • Wollman R, Meyer T. Coordinated oscillations in cortical actin and Ca2+ correlate with cycles of vesicle secretion. Nat Cell Biol 2012; 14:1261-9; PMID:23143397; http://dx.doi.org/10.1038/ncb2614
  • Lokman NA, Ween MP, Oehler MK, Ricciardelli C. The role of annexin A2 in tumorigenesis and cancer progression. Cancer Microenviron 2011; 4:199-208; PMID:21909879; http://dx.doi.org/10.1007/s12307-011-0064-9
  • Yan X, Yin J, Yao H, Mao N, Yang Y, Pan L. Increased expression of annexin A3 is a mechanism of platinum resistance in ovarian cancer. Cancer Res 2010; 70:1616-24; PMID:20103635; http://dx.doi.org/10.1158/0008-5472.CAN-09-3215
  • Kim A, Serada S, Enomoto T, Naka T. Targeting annexin A4 to counteract chemoresistance in clear cell carcinoma of the ovary. Expert Opin Ther Targets 2010; 14:963-71; PMID:20673185; http://dx.doi.org/10.1517/14728222.2010.511180
  • Hayes MJ, Moss SE. Annexins and disease. Biochem Biophys Res Commun 2004; 322:1166-70; PMID:15336964; http://dx.doi.org/10.1016/j.bbrc.2004.07.124
  • Grigorian M, Ambartsumian N, Lukanidin E. Metastasis-inducing S100A4 protein: implication in non-malignant human pathologies. Curr Mol Med 2008; 8:492-6; PMID:18781956; http://dx.doi.org/10.2174/156652408785747942
  • Grum-Schwensen B, Klingelhofer J, Berg CH, El-Naaman C, Grigorian M, Lukanidin E, Ambartsumian N. Suppression of tumor development and metastasis formation in mice lacking the S100A4(mts1) gene. Cancer Res 2005; 65:3772-80; PMID:15867373; http://dx.doi.org/10.1158/0008-5472.CAN-04-4510
  • Storr SJ, Carragher NO, Frame MC, Parr T, Martin SG. The calpain system and cancer. Nat Rev Cancer 2011; 11:364-74; PMID:21508973; http://dx.doi.org/10.1038/nrc3050
  • Mellgren RL, Zhang W, Miyake K, McNeil PL. Calpain is required for the rapid, calcium-dependent repair of wounded plasma membrane. J Biol Chem 2007; 282:2567-75; PMID:17121849; http://dx.doi.org/10.1074/jbc.M604560200
  • McCullough J, Colf LA, Sundquist WI. Membrane fission reactions of the mammalian ESCRT pathway. Ann Rev Biochem 2013; 82:663-92; PMID:23527693; http://dx.doi.org/10.1146/annurev-biochem-072909-101058
  • Mattissek C, Teis D. The role of the endosomal sorting complexes required for transport (ESCRT) in tumorigenesis. Mol Mem Biol 2014; 31:111-9; PMID:24641493; http://dx.doi.org/10.3109/09687688.2014.894210
  • Taylor DD, Taylor CG, Jiang CG, Black PH. Characterization of plasma membrane shedding from murine melanoma cells. Int J Cancer 1988; 41:629-35; PMID:3356493; http://dx.doi.org/10.1002/ijc.2910410425
  • Mayer C, Maaser K, Daryab N, Zanker KS, Brocker EB, Friedl P. Release of cell fragments by invading melanoma cells. Eur J Cell Biol 2004; 83:709-15; PMID:15679115; http://dx.doi.org/10.1078/0171-9335-00394
  • Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Redondo M, Williams C, Garcia-Santos G, Ghajar C, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 2012; 18:883-91; PMID:22635005; http://dx.doi.org/10.1038/nm.2753

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.