This article was downloaded by: [Tulane University] On: 13 January 2015, At: 00:31 Publisher: Taylor & Francis Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK

Cell Cycle Publication details, including instructions for authors and subscription information: http://www.tandfonline.com/loi/kccy20

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

Jyoti K. Jaiswal & Jesper Nylandsted

b

a

Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC, 20010-2970. b

Membrane Integrity Group, Unit for Cell Death and Metabolism, Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark. Accepted author version posted online: 07 Jan 2015.

Click for updates To cite this article: Jyoti K. Jaiswal & Jesper Nylandsted (2015): S100 and annexin proteins identify cell membrane damage as the Achilles heel of metastatic cancer cells, Cell Cycle To link to this article: http://dx.doi.org/10.1080/15384101.2014.995495

Disclaimer: This is a version of an unedited manuscript that has been accepted for publication. As a service to authors and researchers we are providing this version of the accepted manuscript (AM). Copyediting, typesetting, and review of the resulting proof will be undertaken on this manuscript before final publication of the Version of Record (VoR). During production and pre-press, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal relate to this version also.

PLEASE SCROLL DOWN FOR ARTICLE Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and should be independently verified with primary sources of information. Taylor and Francis shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of the Content. This article may be used for research, teaching, and private study purposes. Any substantial or systematic reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http:// www.tandfonline.com/page/terms-and-conditions

S100 and annexin proteins identify cell membrane damage as the Achilles heel of metastatic cancer cells Authors: Jyoti K. Jaiswal1*and Jesper Nylandsted2*. 1

Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan

Membrane Integrity Group, Unit for Cell Death and Metabolism, Danish Cancer Society

us

*Correspondence: JKJ: [email protected]; JN: [email protected]

A2; Metastasis; Breast Cancer; Actin.

M

Abstract

an

Key words: Plasma Membrane Repair; Cancer; Annexins; S100 proteins ; S100A11; Annexin

ed

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

pt

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

ce

tumor cells repair the PM damage are poorly understood. Ca2+ entry through the injured PM initiates repair of the injured PMR. Depending on the cell type, different organelles and proteins respond to this Ca2+ entry and facilitate repair the damaged plasma membrane. We recently

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

Research Center, DK-2100 Copenhagen, Denmark.

cr

2

ip t

Avenue, NW, Washington, DC 20010-2970.

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 PMR. Here we will discuss the involvement of S100, annexins and its regulation of actin cytoskeleton leading to PMR. Additionally, we will show that another S100 member – S100A4 accumulates at the

1

injured PM. These findings reveal a new role for the S100 and annexin protein up regulation in

cr us an M ed pt ce Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

ip t

metastatic cancers and identify these proteins and PMR as targets for treating metastatic cancers.

2

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

ip t

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

cr

us

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

an

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

M

ruptures on a recurring basis that they need to cope with to maintain cell and tissue integrity.

ed

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

pt

frequency. Muscle and lung cells offer a good example of this as they are routinely wounded as a result of exercise and over stretching1, 2. Defect in the muscle cell’s ability to repair has been

ce

shown to result in muscular dystrophy3-6, 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 A7, diabetes8, and Chediak-Higashi9 syndrome and therapies

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

and thus cells rapidly repair PM lesions. Even in the shielding tissue environment where the

targeting PMR have been shown to be effective in treating muscle and lung injuries10-12. 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 metastasis13. Human breast cancer cells that turn metastatic through up-regulation of truncated EGF receptor (ErbB2) also need to up-

3

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

ip t

novel aspect of tumor metastasis for developing new therapeutic approaches to target metastatic cancers.

cr

us

Studies over the past two decades have revealed that plasma membrane repair is a complex and active process that require membrane replacements, fusion events and cytoskeletal

an

reorganization14. PMR is triggered by Ca2+ influx at the injury site due to over a thousand-fold gradient of calcium that exists across the plasma membrane15. The calcium influx triggers a

M

versatile repair system that involves replacing or patching the injured membrane16. Fusion of intracellular vesicles around the wound perimeter to form a patch was revealed using the sea

ed

urchin egg17. This formed the basis for the patch model, according to which Ca2+ entry at the

pt

wound site triggers recruitment and homotypic fusion of vesicles at the wound site which then fuses with the plasma membrane to seal the wound17, 18. In the sea urchin eggs these vesicles

ce

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 cells19, 20. Additionally, injury-

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

The membrane repair machinery

triggered fusion of non-secretory vesicles called enlargeosomes21, and accumulation of mitochondria at the site of PM injury22 have also been found to be required for PMR.

In cells wounded by small pore forming toxins, PMR progresses by the replacement of the

4

damaged cell membrane, such that the injured membrane area is physically removed by endocytosis23, blebbing24, or ectocytosis25. Shedding and endocytosis of damaged cell membrane by vesicles has also been shown for lesions formed by the membrane attack complex (MAC)26, 27

. Lysosome exocytosis has been shown to be important for removal of injured membrane

ip t

through the secretion of Acid Sphingomyelinase (ASMase)23. Secreted ASMase hydrolyses the plasma membrane sphingomyelin to ceramide causing endocytosis of the pores in the plasma

cr

us

expression, or Limb Girdle Muscular dystrophy 2B (LGMD2B) patients (lacking dysferlin protein), who show slow and poor injury-triggered ASMase secretion both show compromised

an

PMR4, 23. The PM repair deficit in these patient cells can be rescued by providing sphingomyelinase4. ASMase is also known to trigger cell membrane shedding and could

M

contribute to the shedding of membrane by ectocytosis25, 28, 29. Thus, ASMase secreted by

ed

lysosome exocytosis may be involved in all the three processes (exocytosis, endocytosis, ectocytosis) involved in PMR.

pt

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

ce

Complex Required for Transport (ESCRT) III-mediated shedding of the injured PM30. We find that shedding of damaged membrane is required even following larger focal injury. This process

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

membrane formed by the pore forming toxins23. Cells from NPA patients, lacking ASMase

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 membrane31. Annexins are another group of calcium-binding protein that are implicated in sensing PM injury and formation of membrane blebs to shed the membrane

5

damaged by pore forming toxin32. 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 manner33, 34. Annexin and S100 proteins are known regulators of actin cytoskeleton35, 36.

ip t

Actin cytoskeleton associated with the plasma membrane is another key regulator

of PM repair. The cortical actin causes membrane tension, which prevents spontaneous resealing

cr

us

observed in Xenopus oocytes and Drosophila Embryos38, which is followed by resynthesis of the cortical actin. This progresses in way of a contractile actomyosin ring that forms a “purse string”

an

- a circular structure and constricts circumferentially, in a manner coincident with the recruitment of filamentous actin (F-actin) and myosin-II at the wound borders39. Disruption of the actin

M

cytoskeleton by either Cytochalasin D or Latrunculin B prevents actomyosin ring assembly and

ed

impairs wound closure39. 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

pt

membrane38. Additionally, intracellular vesicles are recruited to the wound perimeter enabling formation of a membrane patch within the actin ring to seal the wound14, 38.

ce

Annexin and S100 proteins in membrane repair Annexin protein family, believed to have originated a billion years ago, has members in all major eukaryotic phyla40. Annexins have evolved extensively and independently in several eukaryotic

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

upon injury37. Ca2+ influx at the injury site triggers depolymerization of cortical actin as

lineages into a varied family40. 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 it41, 42. In humans there are 12 different annexin proteins with orthologs in most vertebrates40,33. They contain a unique COOH-terminal core domain that consists of four

6

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 43. The NH2-terminal region of annexins is variable in length and sequence between family members and enables the individual protein to interact with

ip t

distinct cytoplasmic partners such as the calcium binding S100 proteins44.

S100 proteins are small (10-14 kDa), EF-hand-type Ca2+-binding proteins that upon

cr

us

found in vertebrates and are clustered on chromosome 1q21 in humans (S100A1–

S100A16)45. The majority of the protein family members form symmetric noncovalent

an

homodimers - a unique feature of the S100 proteins amongst the EF-hand protein family46. These

M

proteins undergo a change in conformation upon binding calcium, which exposes a hydrophobic domain that can interact with the NH2-terminal region of specific annexins44. This interaction

ed

facilitates close apposition of adjacent phospholipid membranes and promotes membrane fusion33. Additionally, several pair of S100-annexin complexes such as S100A10 and annexin

pt

A2 (S100A10–ANXA2) can bind cytoskeletal components and have been associated with

ce

intracellular vesicle fusion47. S100A10 and ANXA2 are known to exist as a heterotetrameric complex where an S100A10 dimer resides in the center of the complex, interconnecting two Annexin A2 molecules44. Similarly, annexin A1 and S100A11 (also called S100C or calgizzarin)

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

Ca2+ activation exert both intracellular and extracellular functions. S100 genes are exclusively

interact in a temporal Ca2+-dependent manner48. 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 roles49. 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 7

ANXA1 and ANXA2 proteins with dysferlin50. Dysferlin is the muscle protein, lack of which results in poor repair of muscle fibers3. 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 vesicles50. However, work using zebrafish

ip t

suggests that these dysferlin vesicles may be derived from the PM51. The first demonstration of a role of annexins in PMR in human cells was offered for ANXA152. ANXA1 was shown to

cr

us

dominant-negative mutant inhibited PMR52. Subsequently role of ANXA1 was identified in repair of plasma membrane injured by pore forming toxins, here ANXA1 was shown to facilitate

an

shedding of the injured plasma membrane through the formation of blebs containing the membrane pores24. Annexins A5 and A6 have also been implicated in the process of PMR51, 53.

M

The ability of ANXA5 to assemble into two-dimensional arrays at the sites of membrane injury

ed

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.

pt

Addition of exogenous recombinant ANXA5 protein rescues this PMR defect53. ANXA6 on the other hand forms a structure termed as “repair cap” at the site of injury, which facilitates healing

ce

of injured muscle fibers54. 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

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

accumulate at the wound perimeter and use of ANXA1 inhibiting antibody, peptide, or

to ANXA1 suggesting a potential for sequential recruitment of annexins to facilitate PMR55. 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 for tumor cells are more fragile56, 57 and that death induced by lysosomal

8

membrane pemebilization is prominent in cancer cells56. This fragility has been harnessed for the development of novel therapeutics58, 59. Similar to lysosomal membrane, PM of cancer cells are more unstable and have reduced stiffness, which is caused by an increase in saturated phospholipids60. The PM stiffness is inversely correlated with the ability of the cancer cell to

ip t

migrate and invade three-dimensional matrix60. The greater motility of metastatic cells and

increase in mechanical stress due to invasion of the tissue matrix can increase PM damage. We

cr

us

response, we find these invasive cancer cells enhance their PMR by up regulating expression of S100A11 and annexin A2 proteins13. Annexins and S100 proteins are commonly up regulated in

an

variety of cancers61, 62. Specifically, S100A11 is overexpressed in several tumors, where it is associated with metastasis and poor disease prognosis63-65. S100A11 is enriched in pseudopodia

M

of metastatic cancer cells and is required for forming actin-dependent pseudopodial protrusions,

ed

which facilitates migration of the tumor cells66.

To study the involvement of PM damage and repair in cancer cells as they enhance

pt

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

ce

(p95ErbB2)67. The p95ErbB2 oncogene mimics constitutively active cleaved form of ErbB2 oncoprotein commonly found in aggressive breast cancers68. Increased invasiveness of these

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

recently identified this to be the case as PM of invasive cancer cells suffered greater damage13. In

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. Coaccumulation 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

9

cell membrane, it localizes away from S100A11-ANXA2 complex. Greater Ca2+-dependent binding of S100A11 with ANXA2 as compared with ANXA169 could be responsible for this differential response of the two 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

ip t

cortical F-actin is necessary to support the plasma membrane, but it also creates tension that can inhibit passive resealing of the PM after injury70. Following injury, cortical actin depolymerizes

cr

us

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

an

actin can facilitate repair of the wounded membrane71. We find that PM injury-triggered ANXA2 and S100A11 accumulation is followed by a rapid buildup of F-actin at the site of repair13.

M

S100A11–ANXA2 can bind F-actin and decrease the depolymerization rate of preformed actin

ed

filaments72, 73. Thus, these proteins preserve existing F-actin and allow buildup of new F-actin around the injury site (Fig. 1). Absence of either of these proteins prevents F-actin buildup

pt

following injury and prevents PMR, similar to the drugs that alter actin polymerization or depolymerization13, 74. Cortical F-actin deploymerization around the injury site is followed by the

ce

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

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

at the site of injury, arguably by actin severing proteins. This reduces cortical tension and avoids

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 injury13. ANXA2 has Ca2+ dependent phospholipid binding ability, which enables

10

aggregation of endosomes and other vesicles75. We found that up-regulation of S100A11 and ANXA2 does not enhance injury triggered fusion of the endosomes/lysosomes13. However, in light of the ability of these proteins to nucleate polymerization of cortical F-actin at the endosomes76 they may facilitate accumulation of endosomal vesicles at the site of injury. The

ip t

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–

cr

us

buildup of cortical actin is analogous to F-actin drawstring formation during healing of injured Xenopus oocyte cell membrane39. Thus, by analogy we believe F-actin buildup may pull the

an

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

M

vesicle fusion and cortical actin buildup. Additionally, it may help with vesicle fusion as Ca2+-

ed

regulated F-actin dynamics at the cell membrane facilitates vesicle fusion77. These findings demonstrate that invasive cancer cells are dependent on efficient PMR system to cope with

pt

elevated rate of cell injury and that they rely on the S100A11–ANXA2 complex to facilitate plasma membrane repair.

ce

PMR as target for therapeutic intervention While the diseases linked to altered PMR are caused primarily due to a defect in this process37, tumor metastasis offers a counter example to such diseases13. This presents an interesting

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

ANXA2 complex and excise the damaged part of the PM marked by ANXA1 (Fig. 1). The

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)4. This leads to poor repair of LGMD2B patient muscle cells, a

11

defect that is rescued by providing this enzyme exogenously4. 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 therapy58. While in the use of ASMase inhibitors we focused our attention on

ip t

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

cr

us

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

an

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 strategy13.

M

S100 and annexins proteins are amongst the most frequently dysregulated proteins

ed

in neoplasia and are overexpressed in various cancers61. Thus members of these proteins families are attractive candidates to compromise PMR in tumor cells. As discussed above members of the

pt

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

ce

colorectal, pancreatic and brain tumors and linked to metastatic progression78. Further, changes in annexin A3 and A4 expression has been associated with chemo resistance in ovarian cancer cells79, 80. These findings make a case for a potential therapeutic approach against tumor

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

PMR. Targeting this and other regulators of PMR is a novel therapeutic approach to control

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

12

another suggesting a strict functional fail-safe mechanism81. Thus, efficient pharmacologically 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

ip t

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

cr

us

an additional advantage of inhibiting metastasis by inhibiting the motility and thus invasiveness of the cancer cells.

an

Aside from annexins, S100 proteins offer another potential target. These proteins are implicated in multiple stages of cancer and are commonly up-regulated and associated with

M

tumor progression in various cancers62. However, with S100A11 as an exception13 their direct regulatory role in PMR has not yet been characterized. Other S100 family members may also

ed

function as Ca2+ triggered switches that can regulate PMR through interaction with other

pt

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

ce

and exerts its function both intra- and extracellular82, 83. Our analysis of S100A4 by live cell imaging of PMR in HeLa cells show that injury triggers S100A4 co-accumulation with S100A11

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

complex proteins regulating actin polymerization offer another set of targets. Such targets have

at the injury site (Fig. 2A). In a manner similar to S100A11, S100A4 accumulation precedes the accumulation of F-actin at the site of repair (Fig. 2B). 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 over expression, S100A4 would be an attractive candidate to broadly target inhibition of tumor metastasis by regulating PMR.

13

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 PMR84, 85. We recently identified yet another member of the EF -hand protein family (Apoptosis linked gene 2 - ALG-2) that can regulate PMR31. While

ip t

Calpains regulate various aspects of tumor progression84, their involvement in PMR in cancer cells has not been elucidated. In case of ALG-2 we found that it facilitates PMR by enabling

cr

us

complex at the injured cell membrane31. The ESCRT machinery is involved in membrane curvature and cleavage86. While the role of ESCRT machinery in cancer is disputed87, metastatic

an

tumor cells are long known to make use of membrane cleavage to shed their PM88, 89. Shedding of PM by tumor cells is gaining wider recognition due to their role in the process of cell-cell

M

communication through the use of extracellular vesicles90. Thus, our finding that PMR of

ed

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

pt

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

ce

targeting this Achille’s heel of cancers. Acknowledgement

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

shedding of damaged cell membrane through the Ca2+-triggered accumulation of the ESCRT III

We thank Birgitte Grum-Schwensen and Noona Ambartsumian for the S100A4-GFP plasmid.

14

References 1.

McNeil PL, Khakee R. Disruptions of muscle fiber plasma membranes. Role in

exercise-induced damage. The American journal of pathology 1992; 140:1097-109. Gajic O, Lee J, Doerr CH, Berrios JC, Myers JL, Hubmayr RD. Ventilator-induced

ip t

2.

cell wounding and repair in the intact lung. American journal of respiratory and critical care

cr

Bansal D, Miyake K, Vogel SS, Groh S, Chen CC, Williamson R, McNeil PL,

us

3.

Campbell KP. Defective membrane repair in dysferlin-deficient muscular dystrophy. Nature

4.

an

2003; 423:168-72.

Defour A, Van der Meulen JH, Bhat R, Bigot A, Bashir R, Nagaraju K, Jaiswal JK.

M

Dysferlin regulates cell membrane repair by facilitating injury-triggered acid sphingomyelinase

5.

ed

secretion. Cell death & disease 2014; 5:e1306.

Jaiswal JK, Marlow G, Summerill G, Mahjneh I, Mueller S, Hill M, Miyake K,

pt

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. Cheng X, Zhang X, Gao Q, Ali Samie M, Azar M, Tsang WL, Dong L, Sahoo N,

ce

6.

Li X, Zhuo Y, et al. The intracellular Ca(2+) channel MCOLN1 is required for sarcolemma

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

medicine 2003; 167:1057-63.

repair to prevent muscular dystrophy. Nature medicine 2014; 20:1187-92. 7.

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. The Journal of cell biology 2010; 189:1027-38. 8.

Howard AC, McNeil AK, Xiong F, Xiong WC, McNeil PL. A novel cellular defect

15

in diabetes: membrane repair failure. Diabetes 2011; 60:3034-43. 9.

Huynh C, Roth D, Ward DM, Kaplan J, Andrews NW. Defective lysosomal

exocytosis and plasma membrane repair in Chediak-Higashi/beige cells. ProcNatlAcadSciUSA 2004; 101:16795-800. Yasuda S, Townsend D, Michele DE, Favre EG, Day SM, Metzger JM. Dystrophic

ip t

10.

heart failure blocked by membrane sealant poloxamer. Nature 2005; 436:1025-9.

cr

Weisleder N, Takizawa N, Lin P, Wang X, Cao C, Zhang Y, Tan T, Ferrante C,

us

Zhu H, Chen PJ, et al. Recombinant MG53 protein modulates therapeutic cell membrane repair in treatment of muscular dystrophy. Science translational medicine 2012; 4:139ra85. Jia Y, Chen K, Lin P, Lieber G, Nishi M, Yan R, Wang Z, Yao Y, Li Y, Whitson

an

12.

BA, et al. Treatment of acute lung injury by targeting MG53-mediated cell membrane repair.

Jaiswal JK, Lauritzen SP, Scheffer L, Sakaguchi M, Bunkenborg J, Simon SM,

ed

13.

M

Nature communications 2014; 5:4387.

Kallunki T, Jaattela M, Nylandsted J. S100A11 is required for efficient plasma membrane repair

14.

pt

and survival of invasive cancer cells. Nature communications 2014; 5:3795. Abreu-Blanco MT, Verboon JM, Parkhurst SM. Single cell wound repair: Dealing

15.

ce

with life's little traumas. Bioarchitecture 2011; 1:114-21. Jaiswal JK. Calcium - how and why? Journal of biosciences 2001; 26:357-63.

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

11.

16.

McNeil PL, Vogel SS, Miyake K, Terasaki M. Patching plasma membrane

disruptions with cytoplasmic membrane. JCell Sci 2000; 113 ( Pt 11):1891-902. 17.

Terasaki M, Miyake K, McNeil PL. Large plasma membrane disruptions are

rapidly resealed by Ca2+-dependent vesicle-vesicle fusion events. The Journal of cell biology 1997; 139:63-74.

16

18.

Steinhardt RA, Bi G, Alderton JM. Cell membrane resealing by a vesicular

mechanism similar to neurotransmitter release. Science 1994; 263:390-3. 19.

Reddy A, Caler EV, Andrews NW. Plasma membrane repair is mediated by

Ca(2+)-regulated exocytosis of lysosomes. Cell 2001; 106:157-69. McNeil PL. Repairing a torn cell surface: make way, lysosomes to the rescue. JCell

ip t

20.

Sci 2002; 115:873-9.

cr

Lorusso A, Covino C, Priori G, Bachi A, Meldolesi J, Chieregatti E. Annexin2

us

coating the surface of enlargeosomes is needed for their regulated exocytosis. The EMBO journal 2006; 25:5443-56.

Sharma N, Medikayala S, Defour A, Rayavarapu S, Brown KJ, Hathout Y, Jaiswal

an

22.

JK. Use of quantitative membrane proteomics identifies a novel role of mitochondria in healing

M

injured muscles. The Journal of biological chemistry 2012; 287:30455-67. Idone V, Tam C, Goss JW, Toomre D, Pypaert M, Andrews NW. Repair of injured

ed

23.

plasma membrane by rapid Ca2+-dependent endocytosis. The Journal of cell biology 2008;

24.

pt

180:905-14.

Babiychuk EB, Monastyrskaya K, Potez S, Draeger A. Blebbing confers resistance

25.

ce

against cell lysis. Cell DeathDiffer 2011; 18:80-9. Keyel PA, Loultcheva L, Roth R, Salter RD, Watkins SC, Yokoyama WM, Heuser

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

21.

JE. Streptolysin O clearance through sequestration into blebs that bud passively from the plasma membrane. Journal of cell science 2011; 124:2414-23. 26.

Morgan BP. Complement membrane attack on nucleated cells: resistance, recovery

and non-lethal effects. The Biochemical journal 1989; 264:1-14. 27.

Moskovich O, Herzog LO, Ehrlich M, Fishelson Z. Caveolin-1 and dynamin-2 are

17

essential for removal of the complement C5b-9 complex via endocytosis. The Journal of biological chemistry 2012; 287:19904-15. 28.

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

ip t

microparticle release from glial cells. The EMBO journal 2009; 28:1043-54. 29.

Gills JJ, Zhang C, Abu-Asab MS, Castillo SS, Marceau C, LoPiccolo J,

cr

us

and cell death in response to phosphatidylinositol ether lipid analogs and perifosine. Cell death & disease 2012; 3:e340.

Jimenez AJ, Maiuri P, Lafaurie-Janvore J, Divoux S, Piel M, Perez F. ESCRT

an

30.

machinery is required for plasma membrane repair. Science 2014; 343:1247136. Scheffer L SSC, Sharma N, Medikayala S, Brown K.J, Defour A, Jaiswal J.K

M

31.

Commun 2014.

Draeger A, Monastyrskaya K, Babiychuk EB. Plasma membrane repair and cellular

pt

32.

ed

Mechanism of Ca2+-triggered ESCRT assembly and regulation of cell membrane repair. Nat

damage control: the annexin survival kit. BiochemPharmacol 2011; 81:703-12. Gerke V, Moss SE. Annexins: from structure to function. Physiol Rev 2002;

ce

33.

82:331-71.

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

Kozikowski AP, Tsokos M, Goldkorn T, Dennis PA. Ceramide mediates nanovesicle shedding

34.

Miwa N, Uebi T, Kawamura S. S100-annexin complexes--biology of conditional

association. FEBS J 2008; 275:4945-55. 35.

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. BiochemBiophysResCommun 2000; 267:77-9.

18

36.

Hayes MJ, Rescher U, Gerke V, Moss SE. Annexin-actin interactions. Traffic

2004; 5:571-6. 37.

McNeil PL, Steinhardt RA. Plasma membrane disruption: repair, prevention,

adaptation. AnnuRevCell DevBiol 2003; 19:697-731. Abreu-Blanco MT, Verboon JM, Parkhurst SM. Cell wound repair in Drosophila

ip t

38.

occurs through three distinct phases of membrane and cytoskeletal remodeling. JCell Biol 2011;

cr

Bement WM, Mandato CA, Kirsch MN. Wound-induced assembly and closure of

us

39.

an actomyosin purse string in Xenopus oocytes. CurrBiol 1999; 9:579-87.

Moss SE, Morgan RO. The annexins. Genome biology 2004; 5:219.

41.

Konopka-Postupolska D, Clark G, Goch G, Debski J, Floras K, Cantero A, Fijolek

an

40.

M

B, Roux S, Hennig J. The role of annexin 1 in drought stress in Arabidopsis. Plant physiology

42.

ed

2009; 150:1394-410.

Clark G, Konopka-Postupolska D, Hennig J, Roux S. Is annexin 1 a multifunctional

43.

pt

protein during stress responses? Plant signaling & behavior 2010; 5:303-7. Swairjo MA, Concha NO, Kaetzel MA, Dedman JR, Seaton BA. Ca(2+)-bridging

ce

mechanism and phospholipid head group recognition in the membrane-binding protein annexin V. Nature structural biology 1995; 2:968-74.

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

193:455-64.

44.

Rintala-Dempsey AC, Rezvanpour A, Shaw GS. S100-annexin complexes--

structural insights. FEBS J 2008; 275:4956-66. 45.

Marenholz I, Heizmann CW, Fritz G. S100 proteins in mouse and man: from

evolution to function and pathology (including an update of the nomenclature). Biochemical and biophysical research communications 2004; 322:1111-22.

19

46.

Santamaria-Kisiel L, Rintala-Dempsey AC, Shaw GS. Calcium-dependent and -

independent interactions of the S100 protein family. The Biochemical journal 2006; 396:201-14. 47.

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. Experimental cell research

48.

ip t

1988; 175:81-96.

Seemann J, Weber K, Gerke V. Annexin I targets S100C to early endosomes.

cr

Liu Y, Myrvang HK, Dekker LV. Annexin A2 complexes with S100 proteins:

us

49.

structure, function and pharmacological manipulation. British journal of pharmacology 2014. Lennon NJ, Kho A, Bacskai BJ, Perlmutter SL, Hyman BT, Brown RH, Jr.

an

50.

JBiolChem 2003; 278:50466-73.

Roostalu U, Strahle U. In vivo imaging of molecular interactions at damaged

ed

51.

M

Dysferlin interacts with annexins A1 and A2 and mediates sarcolemmal wound-healing.

sarcolemma. Developmental cell 2012; 22:515-29. McNeil AK, Rescher U, Gerke V, McNeil PL. Requirement for annexin A1 in

pt

52.

plasma membrane repair. JBiolChem 2006; 281:35202-7. Bouter A, Gounou C, Berat R, Tan S, Gallois B, Granier T, d'Estaintot BL, Poschl

ce

53.

E, Brachvogel B, Brisson AR. Annexin-A5 assembled into two-dimensional arrays promotes cell

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

FEBS letters 1997; 413:185-90.

membrane repair. NatCommun 2011; 2:270. 54.

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. Proceedings of the National Academy of Sciences of the United States of America 2014; 111:6004-9.

20

55.

Potez S, Luginbuhl M, Monastyrskaya K, Hostettler A, Draeger A, Babiychuk EB.

Tailored protection against plasmalemmal injury by annexins with different Ca2+ sensitivities. JBiolChem 2011; 286:17982-91. 56.

Kallunki T, Olsen OD, Jaattela M. Cancer-associated lysosomal changes: friends or

57.

ip t

foes? Oncogene 2013; 32:1995-2004.

Nylandsted J, Gyrd-Hansen M, Danielewicz A, Fehrenbacher N, Lademann U,

cr

us

cell survival by inhibiting lysosomal membrane permeabilization. The Journal of experimental medicine 2004; 200:425-35.

Petersen NH, Olsen OD, Groth-Pedersen L, Ellegaard AM, Bilgin M, Redmer S,

an

58.

Ostenfeld MS, Ulanet D, Dovmark TH, Lonborg A, et al. Transformation-associated changes in

M

sphingolipid metabolism sensitize cells to lysosomal cell death induced by inhibitors of acid

59.

ed

sphingomyelinase. Cancer cell 2013; 24:379-93.

Ellegaard AM, Groth-Pedersen L, Oorschot V, Klumperman J, Kirkegaard T,

pt

Nylandsted J, Jaattela M. Sunitinib and SU11652 inhibit acid sphingomyelinase, destabilize lysosomes, and inhibit multidrug resistance. Molecular cancer therapeutics 2013; 12:2018-30. Swaminathan V, Mythreye K, O'Brien ET, Berchuck A, Blobe GC, Superfine R.

ce

60.

Mechanical stiffness grades metastatic potential in patient tumor cells and in cancer cell lines.

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

Hoyer-Hansen M, Weber E, Multhoff G, Rohde M, Jaattela M. Heat shock protein 70 promotes

Cancer Res 2011; 71:5075-80. 61.

Mussunoor S, Murray GI. The role of annexins in tumour development and

progression. The Journal of pathology 2008; 216:131-40. 62.

Salama I, Malone PS, Mihaimeed F, Jones JL. A review of the S100 proteins in

cancer. European journal of surgical oncology : the journal of the European Society of Surgical

21

Oncology and the British Association of Surgical Oncology 2008; 34:357-64. 63.

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. PLoSOne 2008; 3:e3767. Rehman I, Azzouzi AR, Cross SS, Deloulme JC, Catto JW, Wylde N, Larre S,

ip t

64.

Xiao MB, Jiang F, Ni WK, Chen BY, Lu CH, Li XY, Ni RZ. High expression of

us

65.

S100A11 in pancreatic adenocarcinoma is an unfavorable prognostic marker. MedOncol 2011. Shankar J, Messenberg A, Chan J, Underhill TM, Foster LJ, Nabi IR. Pseudopodial

an

66.

actin dynamics control epithelial-mesenchymal transition in metastatic cancer cells. Cancer Res

Egeblad M, Mortensen OH, Jaattela M. Truncated ErbB2 receptor enhances ErbB1

ed

67.

M

2010; 70:3780-90.

signaling and induces reversible, ERK-independent loss of epithelial morphology. IntJCancer

68.

pt

2001; 94:185-91.

Rafn B, Nielsen CF, Andersen SH, Szyniarowski P, Corcelle-Termeau E, Valo E,

ce

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

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

cancer and precursor lesions. HumPathol 2004; 35:1385-91.

cr

Champigneuille J, Hamdy FC. Dysregulated expression of S100A11 (calgizzarin) in prostate

cathepsin B expression. MolCell 2012; 45:764-76. 69.

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. 70.

Togo T, Krasieva TB, Steinhardt RA. A decrease in membrane tension precedes

22

successful cell-membrane repair. MolBiolCell 2000; 11:4339-46. 71.

Miyake K, McNeil PL, Suzuki K, Tsunoda R, Sugai N. An actin barrier to

resealing. JCell Sci 2001; 114:3487-94. 72.

Hayes MJ, Shao D, Bailly M, Moss SE. Regulation of actin dynamics by annexin

73.

ip t

2. EMBO J 2006; 25:1816-26.

Sakaguchi M, Miyazaki M, Inoue Y, Tsuji T, Kouchi H, Tanaka T, Yamada H,

cr

us

fibroblasts. JCell Biol 2000; 149:1193-206. 74.

Burkel BM, Benink HA, Vaughan EM, von DG, Bement WM. A Rho GTPase

an

signal treadmill backs a contractile array. DevCell 2012; 23:384-96. 75.

Glenney JR, Jr., Tack B, Powell MA. Calpactins: two distinct Ca++-regulated

M

phospholipid- and actin-binding proteins isolated from lung and placenta. JCell Biol 1987;

76.

ed

104:503-11.

Morel E, Parton RG, Gruenberg J. Annexin A2-dependent polymerization of actin

77.

pt

mediates endosome biogenesis. DevCell 2009; 16:445-57. Wollman R, Meyer T. Coordinated oscillations in cortical actin and Ca2+ correlate

78.

ce

with cycles of vesicle secretion. NatCell Biol 2012; 14:1261-9. Lokman NA, Ween MP, Oehler MK, Ricciardelli C. The role of annexin A2 in

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

Namba M. Relationship between contact inhibition and intranuclear S100C of normal human

tumorigenesis and cancer progression. Cancer microenvironment : official journal of the International Cancer Microenvironment Society 2011; 4:199-208. 79.

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 research 2010; 70:1616-24. 80.

Kim A, Serada S, Enomoto T, Naka T. Targeting annexin A4 to counteract

23

chemoresistance in clear cell carcinoma of the ovary. Expert opinion on therapeutic targets 2010; 14:963-71. 81.

Hayes MJ, Moss SE. Annexins and disease. Biochemical and biophysical research

communications 2004; 322:1166-70. Grigorian M, Ambartsumian N, Lukanidin E. Metastasis-inducing S100A4 protein:

ip t

82.

implication in non-malignant human pathologies. Current molecular medicine 2008; 8:492-6.

cr

Grum-Schwensen B, Klingelhofer J, Berg CH, El-Naaman C, Grigorian M,

us

Lukanidin E, Ambartsumian N. Suppression of tumor development and metastasis formation in mice lacking the S100A4(mts1) gene. Cancer research 2005; 65:3772-80.

Storr SJ, Carragher NO, Frame MC, Parr T, Martin SG. The calpain system and

an

84.

cancer. Nature reviews Cancer 2011; 11:364-74.

Mellgren RL, Zhang W, Miyake K, McNeil PL. Calpain is required for the rapid,

M

85.

2007; 282:2567-75.

McCullough J, Colf LA, Sundquist WI. Membrane fission reactions of the

pt

86.

ed

calcium-dependent repair of wounded plasma membrane. The Journal of biological chemistry

mammalian ESCRT pathway. Annual review of biochemistry 2013; 82:663-92. Mattissek C, Teis D. The role of the endosomal sorting complexes required for

ce

87.

transport (ESCRT) in tumorigenesis. Molecular membrane biology 2014; 31:111-9.

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

83.

88.

Taylor DD, Taylor CG, Jiang CG, Black PH. Characterization of plasma membrane

shedding from murine melanoma cells. International journal of cancer Journal international du cancer 1988; 41:629-35. 89.

Mayer C, Maaser K, Daryab N, Zanker KS, Brocker EB, Friedl P. Release of cell

fragments by invading melanoma cells. European journal of cell biology 2004; 83:709-15.

24

90.

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. Nature

cr us an M ed pt ce Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

ip t

medicine 2012; 18:883-91.

25

Figure Legends 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

ip t

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

cr

us

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

an

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

ce

pt

ed

M

the tumor cell. ECM: Extra cellular matrix.

Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

derived from e.g. PM or endosomes providing membrane to repair the damaged membrane. Lack

26

27

pt

ce

Ac ed ip t

cr

us

an

M

Downloaded by [Tulane University] at 00:31 13 January 2015

Figure 2: S100A4 and S100A11 are recruited to the site of injury before actin build-up. (A) HeLa cells 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

ip t

underwent repair in the same way as we described previously13. The plot shows the kinetics of

cr us an M ed pt ce Ac

Downloaded by [Tulane University] at 00:31 13 January 2015

accumulation of individual proteins at the repair site (blue arrow) marked by the white box.

28

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

Mechanical activity of cells and the stress imposed on them by extracellular environment is a constant source of injury to the plasma membrane (PM). I...
810KB Sizes 0 Downloads 4 Views