endocarditis pathogen promotes vegetation formation by ... · colonization and biofilm formation on...

33
DOI: 10.1161/CIRCULATIONAHA.114.011432 1 Endocarditis Pathogen Promotes Vegetation Formation by Inducing Intravascular Neutrophil Extracellular Traps Through Activated Platelets Running title: Jung et al.; NETs promote vegetation formation Chiau-Jing Jung, PhD 1 ; Chiou-Yueh Yeh, PhD 2 ; Ron-Bin Hsu, MD 3 ; Chii-Ming, Lee, MD 4 ; Chia-Tung Shun, MD 5 ; Jean-San Chia, PhD 1,2 1 Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan; 2 Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan; 3 Dept of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; 4 Dept of Cardiology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; 5 Dept of Forensic Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan Address for Correspondence: Jean-San Chia, PhD Graduate Institute of Immunology, College of Medicine National Taiwan University No. 1 Jen Ai Road Section 1 Taipei, 10051, Taiwan, ROC Tel: 886-2-23123456 ext 88222 Fax: 886-2-23926238 E-mail: [email protected] Journal Subject Codes: Basic science research:[130] Animal models of human disease, Platelet adhesion:[178] Aggregation, Thrombosis:[92] Platelets, Heart failure:[11] Other heart failure 1 Graduate Institute of Microbiology, College of Medicine, National Taiwan Un niv iv iver er e si si sity ty ty , , Ta Ta Taip ip ipe ei, Taiwan; 2 Graduate Institute of Immunology, College of Medicine, National Taiw i an U U i niversit ity, f Taipei, Taiwan; 3 Dept of Surgery, National Taiwan University Hospital, National Taiwan U Un Univ iver er ers si ity y C C Co ol ollege of Medicine, Taipei, Ta aiw iw iwa a an; 4 Dept of Ca Ca ard r io olo lo log gy , National Taiwan Un Un Univ i ersity H Hos os sp pi p t tal, l, l N Nat at atio io iona na al l l Ta Ta Taiw iw wan an n U U Uni iv v ver r r sity y y C C Coll le leg ge ge o o of f f Me Me Medi di i i ci cine ne ne, T T Taip ip pei ei ei, , Ta Ta Taiw iw iwan an an; 5 5 De De Dept pt pt of Fo Fo Fore e ens n ic Med ed dici in ne e e, N Nat ati iona na al l T Ta Taiw i an an an U U Un n niver r rsi i ity Ho Ho Hosp sp it it ital al al, , Na Na Nation na a al T ai i iw wa wan n U Un Univ iver r rsi i ity ty C Co olle ege e e of n Me Medi di dic ci cine ne ne, Ta Tai ip ip i ei ei, Ta Ta Tai iw iwan an

Upload: others

Post on 16-Mar-2020

9 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

1

Endocarditis Pathogen Promotes Vegetation Formation by Inducing

Intravascular Neutrophil Extracellular Traps Through Activated Platelets

Running title: Jung et al.; NETs promote vegetation formation

Chiau-Jing Jung, PhD1; Chiou-Yueh Yeh, PhD2; Ron-Bin Hsu, MD3; Chii-Ming, Lee, MD4;

Chia-Tung Shun, MD5; Jean-San Chia, PhD1,2

1Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei,

Taiwan; 2Graduate Institute of Immunology, College of Medicine, National Taiwan University,

Taipei, Taiwan; 3Dept of Surgery, National Taiwan University Hospital, National Taiwan

University College of Medicine, Taipei, Taiwan; 4Dept of Cardiology, National Taiwan

University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; 5Dept of

Forensic Medicine, National Taiwan University Hospital, National Taiwan University College of

Medicine, Taipei, Taiwan

Address for Correspondence:

Jean-San Chia, PhD

Graduate Institute of Immunology, College of Medicine

National Taiwan University

No. 1 Jen Ai Road Section 1

Taipei, 10051, Taiwan, ROC

Tel: 886-2-23123456 ext 88222

Fax: 886-2-23926238

E-mail: [email protected]

Journal Subject Codes: Basic science research:[130] Animal models of human disease, Platelet adhesion:[178] Aggregation, Thrombosis:[92] Platelets, Heart failure:[11] Other heart failure

1Graduate Institute of Microbiology, College of Medicine, National Taiwan Unniviviverere sisisitytyty, , TaTaTaipipipeei,

Taiwan; 2Graduate Institute of Immunology, College of Medicine, National Taiwi an UU iniversitity, f

Taipei, Taiwan; 3Dept of Surgery, National Taiwan University Hospital, National Taiwan

UUnUniviverererssiityy CCCoolollege of Medicine, Taipei, Taaiwiwiwaaan; 4Dept of CaCaardr ioololologgy, National Taiwan

UnUnUnivi ersity HHosossppip ttal,l,l NNatatatioioionanaal l l TaTaTaiwiwwanann UUUniivvverrrsityyy CCCollleleggege ooof f f MeMeMedidii icicinenene, TTTaipippeieiei,, TaTaTaiwiwiwananan; 55DeDeDeptptpt of

FoFoForererensn ic Mededdiciinneee, NNatatiionanaal l TTaTaiwi ananan UUUnnniverrrsiiity HoHoHospsppitititalalal, , NaNaNationnaaal Taiiiwwawan n UUnUniviverrrsiiityty CCoolleegeee ofn

MeMedididiccicinenene, TaTaiipip ieiei, , TaTaTaiiwiwanan

Page 2: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

2

Abstract

Background—Endocarditis-inducing streptococci form multi-layered biofilms in complex with

aggregated platelets on injured heart valves. But host factors that interconnect and entrap these

bacteria-platelet aggregates to promote vegetation formation were unclear.

Methods and Results—In a Streptococcus mutans endocarditis rat model, we identified layers of

neutrophil extracellular traps (NETs) interconnecting and entrapping bacteria-platelet aggregates

inside vegetation that could be reduced significantly in size along with diminished colonizing

bacteria by prophylaxis with intravascular DNase I alone. The combination of activated platelets

and specific IgG-adsorbed bacteria are required to induce the formation of NETs through

multiple activation pathways. Bacteria play key roles in coordinating the signaling through

spleen tyrosine kinase, Src family kinases, phosphatidylinositol-3-kinase, and p38 mitogen-

activated protein kinase pathways to up-regulate the expression of P-selectin in platelets, while

inducing reactive oxygen species-dependent citrullination in the arm of neutrophils. NETs in turn

serve as the scaffold to further enhance and entrap bacteria-platelet aggregate formation and

expansion.

Conclusions—NETs promote and expand vegetation formation through enhancing and

entrapping bacteria-platelet aggregates on the injured heart valves.

Key words: infective endocarditis, infectious disease, platelet, neutrophil extracellular traps

and specific IgG-adsorbed bacteria are required to induce the formation of NETTs s thththrorr ugugughh h

multiple activation pathways. Bacteria play key roles in coordinating the signaling through

ppleleeenenen tttyryrrosososininine kikikinnanase, Src family kinases, phospspphahahattidylinositol-3-3-kinnasasasee,e, and p38 mitogen-

acctiivav ted protteieiinnn kkkinanasse ppatatathwhwhwayaysss totoo uup-p-reguuulaaate ttthhee expxpxpreressssiioon n ofoff P-P seseeleeectctininn iiin n plplplatatteleleleteets,,, wwhhih llele

nndududucicicingngng rreaeaeactcctiviveee ooxoxygyygeenn sspeeeciciieseses-d-d-deepepeenendededentntnt cccitititruruullliininaaattiononon iiinn thththee ararrm m m oofof nnneueutrtrt opopophihiilslsls.. NENEETTsTs iinnn tutuurnr

erve as the sscacacafffff ololold d d totoo fffurrthththerere eeenhnhnhanananceee aandndnd eennntrtrtrapapap bbbacaca teteeririr a-a-a plplp atatatelele ettt aaaggggggrereregagagatete fffororormamamatitit ono and

Page 3: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

3

Introduction

The release of neutrophil extracellular traps (NETs), in addition to phagocytosis, constitutes an

important innate effector mechanism to restrain bacterial spreading.1 NETs also exert direct

bacteria-killing activity against various Gram-positive and -negative pathogens through multiple

antimicrobial components that adhere to the fibril structures of NETs, such as elastase,

myeloperoxidase (MPO), cathepsin, and histones.2 Coagulation-generated fibrin barriers may

also restrict microbial dissemination, and interestingly, such activity could be promoted by NETs

through tissue factor and factor XII-dependent activation processes that require the proteolytic

activity of neutrophil serine protease.3 NET-induced thrombosis prevents the dissemination of

Escherichia coli during systemic infection by fostering the compartmentalization of bacteria in

liver microvessels and also by restricting bacterial extravasation into surrounding tissue

compartments. 3, 4 This septic thrombosis, which is also called immunothromobosis, is induced

by NETs and other innate immune cells upon interaction with invading pathogens and is

considered to be an evolutionarily conserved strategy to support innate immune response against

pathogen invasion. 3, 4

Platelet activation and aggregation following thrombin-induced coagulation activation

constitute another key mechanism in thrombosis. Interestingly, platelets also exhibit innate

effector characteristics to sense and respond to pathogen-associated molecular patterns such as

lipopolysaccharide (LPS).5 Disseminating bacteria or viruses in the bloodstream could be

entrapped by intravascular NETs that are frequently encountered in the liver and lung

vasculature.6, 7 Intravascular NETs in the liver could be induced by LPS-activated platelets and

involve TLR4 signaling; however, this only accounts for only part of the activation of NETs

induced by septic blood from patients.4, 5 Thus, it is possible that other activation signals directed

Escherichia coli during systemic infection by fostering the compartmentalizationnn of ff babaactctcterereriaiaia iin n

iver microvessels and also by restricting bacterial extravasation into surrounding tissue

coompmpmpararartmtmtmenenentstts. 3,3, 44 TTThis septic thrombosis, whichhh iiis aalso called immmumm nonoothththrromobosis, is induced

bbyy NNNETs and otothehh rrr innnnnatetete iiimmmmmmununee e ccecellls uupoonn inteeraraactiooonn n wwiwithhh iinnvvaadadininggg ppapaththhogogenensss anannddd iss

coonnsnsidididerere ededd tttooo bebe ann n evevvolllututioionnanariririlylyly cccononseseservrvrvededed ssstttrarar ttteggygy ttoo sususuppppppororo t t innnnananatetee immmmumumunnene rresesesppoponnnsee e agaggaiainsnsn t

pathogen invvasasasioioi n.n.n. 333, 444

Page 4: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

4

through activators present in septic blood or on activated platelets are required for the induction

of intravascular NET formation.

Infective endocarditis (IE) affecting the cardiovascular system is a typical infectious

disease caused by disseminating microorganisms into the bloodstream. Blood-borne

staphylococci from contaminated medical devices or streptococci from gingival wounds during

dental surgery are the most common endocarditis-inducing pathogens that can easily gain access

into bloodstream and colonize the injured heart valves to induce thrombus formation, which is

called vegetation.8, 9 Endocarditis-inducing Staphylococcus aureus or viridans streptococci

acquire distinct strategies to avoid immunosurveillance encountered in the bloodstream,10, 11 and

can induce platelet activation and aggregation through distinct mechanisms in a species-specific

manner.12, 13 Electron microscopic analysis has also revealed that bacteria are in complex with

platelets inside septic vegetation.14 We demonstrated recently that endocarditis pathogens, such

as Streptococcus mutans or Streptococcus gordonii, can form biofilms with multilayer

architecture that are characterized by bacterial floes embedded with platelet aggregates in the

form of mats or nidi and are refractory to antibiotic prophylaxis.15, 16 In the plasma, S. mutans-

induced biofilm formation through platelet aggregation required anti-bacteria specific IgG,16 a

potent opsonin designated for enhancing phagocytosis by neutrophils in order to clear bacteria in

the circulation. But the reason why neutrophil or intravascular immunity fails to prevent bacterial

colonization and biofilm formation on injured heart valves was unclear.

In this study, we present experimental evidence showing that intravascular NETs,

designated to control bacterial spreading through the bloodstream during infection, could be

hijacked by the endocarditis-inducing pathogen to aid in vegetation formation on injured heart

valves. We also delineate the molecular mechanisms underlying bacteria-platelet aggregate-

can induce platelet activation and aggregation through distinct mechanisms in a sspspecece ieeess-s-spspspecececifific

manner.12, 13 Electron microscopic analysis has also revealed that bacteria are in complex with

pllatatteleleletetetss inininsisisiddde ssepepeptitic vegetation.14 We demonstrrraata eed recently thatatt endodoocacacarditis pathogens, such

as SStStrer ptococcucuss mmumutataansnss ooorrr StStStrrereptptocococococcucuus gooorrddonniii,, caan n n foformrmm bbioioofifilmlms s wwiithth mmmulultitilalalayyeyerr

arrchchchitititecece tutureree ttthahat t arrree chchhararracacteerirr zezeeddd bybyby bbbacaccteteterririalll ffflolooesss embmbmbedededdedeed d d wwwiththth pplalaateeelelet tt agagaggrgrg egegegaaatesess iinn n ththhe

form of matss ooor r r niniidididi aandndnd arerere rerer frfrfracacactotooryry tttoo ananantitiibibibiototticicc ppprorophphphylyly axaxaxisisis..151515, 161616 IIIn n n thththe e e plplasasasmamama, , , S.S.S mutans-

Page 5: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

5

induced NET formation.

Methods

Bacterial strains and plasmid

S. mutans GS5 wild-type strain was grown and maintained in brain-heart infusion broth (BHI,

Difco Laboratories Inc., Detroit, MI). To generate green fluorescent protein (GFP)-tagged

bacteria, S. mutans was transformed with the GFPuv sequence containing the shuttle plasmid,

pPDGFPuv, which was described in our previous study. 11

Reagents

Detailed information is available in SUPPLEMENTAL MATERIAL.

Experimental streptococcal endocarditis rat model

A modified rat model of experimental streptococcal endocarditis was performed as previously

described.16, 17 The detailed procedures are available in the SUPPLEMENTAL MATERIAL.

Preparation of platelets and neutrophils

This study was approved by the National Taiwan University Hospital Committee for Regulation

of Human Specimens and Volunteers, and all human volunteers provided informed consent.

Preparation of human platelet rich plasma (PRP), platelet poor plasma (PPP), washed platelet

suspension (PS) and PMNs was performed as previously described.11, 18 The detailed procedures

are available in the SUPPLEMENTAL MATERIAL.

In vitro bacteria-platelet aggregate formation assay

We demonstrated previously that S. mutans adopts anti-bacteria specific IgG to activate platelets

and form biofilm in the plasma.16 We defined such bacterial biofilm embedded in aggregated

platelets as bacteria-platelet aggregates in the present study. The in vitro bacteria-platelet

Detailed information is available in SUPPLEMENTAL MATERIAL.

Experimental streptococcal endocarditis rat model

A A mmomodididifififiededed raaat mmmoododel of experimental streptocococccacac ll endocarditis wwwas ppperererfof rmed as previously

ddedescccrir bed.16,6 1717 TTThehee dddetetaiaiileleleddd prprprococededduurureess aaare aaavvaailaabblblee innn ttthehe SSUPUPU PLPLPLEMEMMENENNTATAALL L MAMAMATETETERIRIIALALAL.

PrPrepepeparararatatioioion n n ofof ppplalaatetelllettsts aandndd nnneueueutrtrtropopphihiilslsls

This study wwasasas aaapppppproror veveved d bybyby ttthehee NNNatattioioi nananal l TaTaTaiwiwwananan UnUnUnivivererersisisitytyty HHHososospipiitatatall l CoCoCommmmmmitittteteteeee fofofor r r ReR gulationn

Page 6: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

6

aggregate formation assay was performed as previously described.16 Bacterial biofilm growth

was initiated by inoculating individual wells of a 24-well plate with a round glass coverslip with

approximately 107 CFU bacteria in 200 l PRP or PPP supplemented with 1% (w/v) glucose. For

the induction of NETs and investigation of the role of NETs in promoting and enhancing

bacteria-platelet aggregate formation, peripheral blood-isolated neutrophils were added to a final

concentration of 2 x 106 neutrophils per ml. After 16 h incubation, the bacteria-platelet

aggregates that had formed on the glass coverslip was gently washed with PBS three times and

then fixed with 2% paraformaldehyde. After fixation, the bacteria-platelet aggregates were

incubated in 0.5% Triton X-100 for 15 min and then stained with rhodamine-conjugated

phalloidin (1:200 dilution; Invitrogen, Carlsbad, CA) and Hoechst 33258 (Sigma-Aldrich, St.

Louis, MO). After washing with PBS three times, the NET formation was observed with a

confocal microscope (Leica TCS SP5).

Induction of NET formation

Peripheral blood-isolated neutrophils were placed on the plasma-coated coverslip (100 l of 107

cells/ml in RPMI) positioned in wells of a 24-well plate and then incubated with 5% CO2 at 37oC

for 1 h. After removing the non-adhered neutrophils, the neutrophils on the coverslip were

further stimulated with S. mutans (108 CFU/ml), IgG (1 mg/ml), PS (108 platelets/ml) or

recombinant-P-selectin (1 g/ml), alone or combined in 300 l RPMI medium. In the inhibition

assay, the cells were pre-incubated with neutralizing antibodies or inhibitors, including aspirin

(0.5 mg/ml), anti-P-selectin mAb (20 g/ml), anti-PSGL-1 mAb (20 g/ml), anti-CD32 mAb (20

g/ml), anti-Toll like receptor mAb (TLR2) (25 g/ml), piceatannol (100 M), PP2 (20 M),

LY294002 (100 M), LFM-A13 (100 M), or SB203580 (50 M), or an equal volume of DMSO

as a vehicle control. For the detection of histone citrullination, the mixtures were incubated at

phalloidin (1:200 dilution; Invitrogen, Carlsbad, CA) and Hoechst 33258 (Sigmaaa-A-A-Aldldriririchchch, ,, StStSt. .

Louis, MO). After washing with PBS three times, the NET formation was observed with a

coonfnfnfococcalalal mmmicicicrrroscsccopopopee (Leica TCS SP5).

nndduduction of NNETETET ffororrmamamatititiononon

PePeriririphphpherere alal bbblololoododd-issosolalaateted d neneututu rropopophihihillsls wwwererereee ppplacacacededd ooon n tththeee plplplasassmamama-c-coaoaoateteedd ccocoveveversrsslilip p p (1(1(10000 l l ofoff 10007

cells/ml in RPRPPMIMIM ) ) ) popoposisis tititionnnededed in nn wewew llllllss ofofof aaa 242424-w-wwelelell l plplplata e e e ananand d d ththhenenen incncncubububatatatededed wwititi h h h 5%5%5% CCO2 at 37oCCC

Page 7: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

7

37oC with 5% CO2 for 3 h. Following the incubations, the cell lysates were prepared in SDS lysis

buffer (2% SDS in 62.5 mM Tris, pH 6.8, supplemented with 5% 2-mercaptoethanol and 10%

glycerol) and sonicated. The histone citrullination was further analyzed by Western blotting

using rabbit antibodies against citrullinated histone H3 or histone H3 (1:2000;

Abcam, Cambridge, MA). For observations by confocal microscopy, the samples were stained

with rhodamine-conjugated phalloidin (1:200 dilution; Invitrogen, Carlsbad, CA) for platelets or

Hoechst 33258 for NETs. The NET formation was further observed with a confocal microscope

(Leica TCS SP5).

P-selectin expression of platelets

The PS was stimulated with S. mutans (108 CFU/ml) alone or in combination with IgG (1mg/ml)

in the Tyrode solution. In the inhibition assay, the platelets were pre-incubated with inhibitors,

including aspirin (0.5 mg/ml), piceatannol (100 M), PP2 (20 M), LY294002 (100 M), or

SB203580 (50 M), or an equal volume of DMSO as a vehicle control. After incubation with 5%

at 37oC CO2 for 3 h, the P-selectin expression on platelets was further detected by flow

cytometry (FACS; Becton Dickinson, Franklin Lakes, NJ, USA) using a rabbit antibody against

human P-selectin (1:100 dilution; eBioscience, San Diego, CA) and FITC-conjugated anti-rabbit

IgG antibody (1:500 dilution; Jackson ImmunoResearch labs, West Grove, PA). For the detection

of phospho-p38 MAPK, the platelet lysate in SDS lysis buffer was analyzed by Western blotting

using a rabbit antibody against human phospho-p38 MAPK (1:2000 dilution; Cell Signaling

Technology, Danvers, MA).

Statistical analysis

The statistical significance of the difference between two sets of data was analyzed by using an

unpaired, two-tailed Student’s t-test. Differences between more than two sets of data were

The PS was stimulated with S. mutans (108 CFU/ml) alone or in combination wititthhh IgIgI G G G (1(1(1mgmgmg/m/ml))

n the Tyrode solution. In the inhibition assay, the platelets were pre-incubated with inhibitors, u

nnclclludududininingg asasaspipp rinnn (0(0(0.5. mg/ml), piceatannol (100 MMM), PP2 (20 M)M)M , LYLYY2929294002 (100 M), or

SSBSB220203580 (500 MMM), ooror aaan n eqeqequauuall vovovolululummeme of DMDMDMSOOO aas aaa vvevehhhicclclee ccocontntrorol.ll AAAftfteerer iincncubububatata ioioion n wiwiththt 5%

att 33377ooCC C COCOO222 ffofor r 33 hh,h, tthhehe PP--selelelececctititinnn eexexprprpresesessisisioonon ooon n plplaateeleletetetsss wawawasss fufuurtttheheher r dededetetectctctededd bbyy y flflflowww

cytometry (FFACACACS;S;S; BBBececctototon DiDiDickckckinininsososon,nn FFFrararanknknkliliin n n LaLaLakekekes,s,s NNNJ,J,J, UUUSASAA))) usssinining g g a a a rararabbbbb ititit aaantntntibibibodoo y against

Page 8: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

8

assessed using one-way analysis of variance (ANOVA) followed by the Bonferroni multiple-

comparisons test. For nonparametrically distributed data, Mann-Whitney U test and Kruskal-

Wallis test with subsequent Dunn’s test were used. A P < 0.05 was considered statistically

significant.

Results

NETs contribute to vegetation formation in situ

Using GFP-tagged S. mutans GS5 and the rat endocarditis model,11, 17 we previously

demonstrated that bacterial biofilm embedded in aggregated platelets form a unique layered

structure with unidentified interlayers of amorphous but well-organized meshes interconnecting

the bacteria-platelet layers inside the vegetation.16 Confocal laser scanning microscopic analysis

revealed the presence of extracellular DNA meshes intermixing with the bacteria biofilm inside

the vegetation (Figure 1). Immunostaining revealed positive co-localization of elastase, which is

a protein from neutrophil primary granules,19 and released DNA meshes, indicating that NETs

are present in the DNA meshes (Figure 1).2 Prophylactic administration of DNase I

intravenously before infection reduced the DNA meshes, the size of the vegetation and the

number of colonized bacteria in the vegetation (Figure 2A, 2B, 2C and 2D), and also improve

the aortic insufficiency (Supplemental Figure 1). DNase I treatment afterwards (4 h post-

infection) conducted in parallel experiments also exhibited therapeutic effects on the reduction of

both vegetation size and colonizing bacteria (data not shown). DNase I did not affect the growth

of S. mutans GS5 in vitro but increased the bacteremia in the infected rats, suggesting that intra-

vascular DNase I did not exert a bactericidal effect on S. mutans (Figure 2E and Supplemental

Figure 2). Prophylaxis with antibiotics for the prevention of IE is recommended by the

tructure with unidentified interlayers of amorphous but well-organized meshes ininnteteercrcononnenenectctctiining gr

he bacteria-platelet layers inside the vegetation.16 Confocal laser scanning microscopic analysis

eeveveealalalededed ttthehehe pppreesesesenncnce of extracellular DNA messhhehess intermixing wwwiti h ththhee e bab cteria biofilm inside

hhhe vev getationn (((FiFF ggugureree 111))). IIImmmmmmununnososostataiininiing rrrevvveallleddd posososititivivve coco--l-looocalallizizizaattioonn n oofof eelalalaststtasasee,e wwwhihihichhh is

a prprprotototeieie n n frfrromomom nneueueutrtropopphihiill pprimimimarararyyy grggranannululleseses,,,1119 aaandndnd reeeleaeaaseseedd d DNDNDNA A A mememeshshs eees, , inindidid cacacatitingngng thahaat NENEN TsTsTs

are present inn tttheheh DDDNANANA mmmeseseshehehes (((FiFiF gugugureree 111).).) 222 PPPrororophphphylylylacaca titiic c c adadadmimiminininists rararatitiiononon ooof f f DNDNDNasasase e e I II

Page 9: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

9

American Heart Association,20 but the efficacy remains controversial.21-23 Our previous and

current data also showed that prophylaxis with penicillin alone could not completely block

vegetation formation due to the deep-seated bacteria-platelet aggregates entrapped in NETs

(Supplemental Figure 3).16 Prophylactic administration of DNase I together with penicillin

significantly reduced the size of the vegetation, the number of colonized bacteria in the

vegetation, and the severity of bacteremia (Supplemental Figure 3). Together, these results

suggested that NETs may contribute directly to the formation of vegetation.

NETs increase bacteria-platelet aggregate formation

Based on the in vivo observation, we hypothesized that the bacteria-platelet aggregates could

induce NET formation, which in return promotes expansion and thickening of these aggregates

on the injured heart valves. The role and mechanism of NET formation was further investigated

using in vitro bacteria-platelet aggregate formation assay as previously described.16 Interestingly,

NET release from purified neutrophils could only be induced by the bacteria-platelet aggregates,

but not by the bacteria or ADP-activated platelets alone (Figure 3A, Supplemental Figure 4,

5A, and 5B), suggesting that the bacteria-platelet aggregate play a critical role in NET induction.

Positive staining for neutrophil elastase further confirmed that the DNA fibers were released

from neutrophils (Supplemental Figure 6). Pharmacological inhibition of NADPH oxidase with

diphenylene iodonium (DPI) attenuated NET release, suggesting that the NET formation induced

by the bacteria-platelet aggregates was dependent on NADPH oxidase activity (Figure 3B).

It has been shown that NETs can activate platelets directly through histone H3 and H4.24

We found that NETs and histones could further enhance bacteria-platelet aggregate formation

when bacteria were co-cultured in the PRP based on the quantification of the biomass (Figure

3C, 3D, Supplemental Figure 7A and 7B). NETs or extracellular histones can promote

nduce NET formation, which in return promotes expansion and thickening of thheeese ee agagggrgrgregegegatata eses

on the injured heart valves. The role and mechanism of NET formation was further investigated

ussininngg g ininin vvitititroror bacaccteteterir a-platelet aggregate formatiiionono assay as previoioouslylyy dddeescribed.16 Interestingly,

NNETTT release frfromomm ppururififieieeddd neneneuututrorophphphililss ccooulddd ooonlyy bbe indndnducucededed bbyy thhhe e bababactterriaiaa--pplalateteteleleet t aagggrgrgregeggatttese ,

buuttt nonoott t bybyby ttthehehe bbaaactteteririaaa ooror AADDPDP-a-aactctctivivivatatededed ppplalalateeleleletstss aalolonnne (((FiFiFiguguurerer 33AAA,,, SSuupppppleleememementnttalalal Fiiiguurure e 4,4,

5A, and 5B),),) sssuguguggegegeststs inining g g thhhatatat thehehe bbbacacacteteririria-a-a plplplatatatelelletett aaaggggggregegegatatate e e plplplayayay a cccririritititicacacal l l roror leee iiin n n NENENET T inductionnn

Page 10: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

10

thrombin generation in the PRP.25, 26 We also found that NETs or purified histone H4 could

enhance thrombin generation, when bacteria were co-cultured in the PRP (Supplemental Figure

7C). Therefore, activation of both coagulation cascade and platelet through histone H4 may

partially account for the bacteria-platelet aggregate formation enhanced by NETs. Such

enhancement of bacteria-platelet aggregate formation by NETs could be abolished in the

presence of DNase I (Figure 3C and 3D). Taken together, these data support our hypothesis and

indicate that bacteria-activated platelets can induce NET formation, which can further enhance

bacteria-platelet aggregate formation in the plasma.

Bacteria stimulate neutrophil citrullination and NET formation requires the combination

of bacteria and activated platelets

To delineate the mechanism by which S. mutans-activated platelets induce NET formation, the

sequential interaction of neutrophils or platelets with bacteria or each other were examined in a

purified system. NET formation was only induced by the bacteria-platelet aggregates (Figure

4A, Supplemental Figure 5C, 5D, and 8A) that formed in response to anti-S. mutans specific

IgG (Supplemental Figure 9). NET formation was also observed using purified rat neutrophils,

platelets, and IgG, thus confirming that bacteria-platelet aggregates could induce NET formation

in the rat IE experimental model (Supplemental Figure 10). Our previous study showed that

platelet activation induced by S. mutans could be inhibited by aspirin.16 Thus, we assessed the

addition of aspirin in this study and found that it inhibited significantly the NET formation,

further confirming that NET formation requires bacteria-activated platelets (Figure 4B).

Histone hypercitrullination can mediate chromatin decondensation, which is essential for

NET formation.27 We found that S. mutans could directly induce neutrophil histone citrullination,

which was enhanced when IgG was bridged together with the bacteria (Figure 4C and

of bacteria and activated platelets

To delineate the mechanism by which S. mutans-activated platelets induce NET formation, the

eequququenenntititialalal iiintntnteracacctititiono of neutrophils or platelets wwwittth bacteria or eeeaca h h ototothheher were examined in a r

puririifif ed system.m. NNNETT fororormamam tititioonon wwwasasas oonlnlly innnduuuceddd bbby thththee bbaacctctereriiia---plalateteeleeet agagagggrgregegatata eeses (((Fiiiggugurrere

4AAA, , SuSuSuppppppleleememmentnttalall FFiigiguururee 5C5CC, 5D5D5D,, , anannd d 8A8A8A))) ththhatata ffoorormmmeddd ininin rresesespopoonsnsnseee totoo aaantnti-i-i-S.S.S mmuututaaanss sppepeciciifficcc

gG (Suppleememementnttalalal FFFigigigurrreee 999). NNNETETET fororormamamatititiononon wwwasasa aaalslso o o obobobseseservrvrveeed d usususinining g g pupupuririr fifiiededed rrratatat nnneutrophils,,

Page 11: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

11

Supplemental Figure 8B) and inhibited by neutralizing antibodies against Toll-like receptor 2

(TLR2) or Fc receptor, or an Fc receptor binding inhibitor (Figure 4D and 4E). The essential

role of TLR2 was further confirmed using TLR2-deficient neutrophils isolated from TLR2

knockout mice (Supplemental Figure 11). The major pathogen-associated molecular patterns

(PAMPs) of S. mutans (a gram positive pathogen) for TLR2 recognition are lipoteichoic acid

(LTA) and peptidoglycan (PGN), and the receptors for recognizing LTA and PGN are TLR2 and

its signal partners, TLR1 and TLR6.28 LTA and S. mutans PGN induced histone citrullination in

neutrophils, and neutralizing antibodies against TLR1, 2, and 6 inhibited S. mutans-induced

histone citrullination in neutrophils (Supplemental Figure 12). Stimulation of neutrophil

NADPH oxidase may further induce histone citrullination.29 Inhibition of NADPH oxidase by

DPI reduced histone citrullination, suggesting that S. mutans-induced histone citrullination in

neutrophils was dependent on NADPH oxidase activity (Figure 4F). Taken together, these data

indicate that S. mutans can induce neutrophil histone citrullination through TLR2 and the Fc

receptor in a NADPH oxidase-dependent manner, and activated platelets are required for

promoting NET formation.

Induced P-selectin expression on platelets involves p38 MAPK activation

During hemostasis or inflammation, P-selectin that is expressed on activated platelets mediates

the initial tethering of circulating neutrophils through PSGL-1.30, 31 Neutralizing antibodies

against P-selectin or PSGL-1 inhibited the binding of bacteria-platelet aggregates to neutrophils

and also blocked NET formation, suggesting that the P-selectin expressed on S. mutans-activated

platelets mediates NET formation (Figure 5A, 5B and 5C). Our previous study indicated that S.

mutans-mediated platelet activation requires IgG.18 Consistent with this observation, induced P-

selectin expression on S. mutans-stimulated platelets also required specific IgG (Figure 5D).

NADPH oxidase may further induce histone citrullination.29 Inhibition of NADPHPHH oooxxixidadadasesese bbby y

DPI reduced histone citrullination, suggesting that S. mutans-induced histone citrullination in

neeutututrororophphphilililsss wwwas dededeppendent on NADPH oxidase aaactctiivity (Figure 444FF). TTTakakakene together, these data

nndiiicac te that S.S mmmuutanansss cccanann iindndnducuceee nneneuuttrroophhiili hhhistooonnne cccititrrululllil nnanatitiononon tthrhroououghgh TTTLLRLR222 ananndd d ththt eee FccFcrr

eecececeptptptororo iinn n a a a NANANADPDPHH oxxxididassse-ee dededepepepenndndenenenttt mmamannnnnnerer,, aaandndd aactctctivivivatttedede ppplalaateteteleleetss aareree rreqeqequiuiuirerered ffforr r

promoting NENEET T T fofoormrmrmatata ioioi n.n..

Page 12: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

12

We next investigated downstream signaling pathways of the Fc receptor in platelets in

order to identify the mechanisms by which IgG induces P-selectin expression. Pharmacological

inhibition of platelets suggested that Src family kinases, Syk, PI3K, and p38 MAPK were

involved in IgG-mediated P-selectin expression (Figure 5D). Furthermore, S. mutans together

with IgG induced phosphorylation of p38 MAPK in platelets, which was dependent on Src

family kinases (Figure 5E). Consistent with these findings, CLMS data showed that

pharmacological inhibition of Src family kinases, Syk, PI3K, or p38 MAPK inhibited bacteria-

platelet aggregation and NET formation (Figure 5F). Only inhibitors of Src family kinases or

Syk, but not aspirin or a P-selectin neutralizing antibody, could inhibit histone citrullination

induced by S. mutans-activated platelets (Supplemental Figure 13). These results confirmed

that S. mutans can stimulate histone citrullination directly through the bridging of the Fc

receptor by specific IgG on neutrophils, but not indirectly through activated platelets. Taken

together, these data suggest that bacteria stimulate NET formation by up-regulating P-selectin on

platelets, which involves Src family kinases, Src, PI3K, and p38 MAPK signaling events.

Roles of P-selectin and IgG in S. mutans-induced NET formation

The role of P-selectin in stimulating NET formation was further examined in a purified system

containing the recombinant-P-selectin, bacteria, and IgG. NET release was induced by the

combination of bacteria and recombinant-P-selectin and was enhanced by the addition of IgG

(Figure 6A and 6B). The downstream signaling pathways of PSGL-1 were further investigated

for their roles in P-selectin-mediated NET formation.32 The data from pharmacological inhibition

of neutrophils showed that Src family kinases, Syk, Tec kinase Bruton tyrosine kinase, and p38

MAPK were involved in P-selectin-mediated NET formation (Figure 6C). Taken together, these

data suggested that multiple signaling pathways, including those mediated through Fc receptor

nduced by S. mutans-activated platelets (Supplemental Figure 13). These resululttts ccconnnfififirmrmrmededed

hat S. mutans can stimulate histone citrullination directly through the bridging of the Fc

eececeeptptptooror bbbyy y spsppeccifififiicic IgG on neutrophils, but not iiindndndiirectly througgh h h actitiivavavatet d platelets. Taken

oogeeetht er, these e dadad ttaa ssuuugggegeststst ttthahahat t babaacctctererriaaa stimmmuuulateee NNENETT T foformrmmatatioioonn byby uuupp--rrm egegguluulatatininng g P-P-P-seselelelectcttininin on

plplatatatelelletetets,s,s, wwwhihihichch iinvnvvololvvevess s SrS cc fafamimimilylyly kkiininasasaseeesss, SSSrcrcr , ,, PPII3K3KK, anananddd p3p3p388 MAMAMAPPKK ssigiggnananalilil ngngng eeeveennntss.s.

Roles of P-sselellececectitiinnn ananand dd IgIggG G G inin S.S.S. mmmututtananansss-i-iindndnducucucededd NNNETETET fffororormamamatit ononon

Page 13: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

13

and P-selectin, are involved in NET formation stimulated by S. mutans-activated platelets.

Discussion

NETs are recognized as an important link between inflammation and thrombosis in host immune

surveillance.3 However, using a S. mutans-induced experimental endocarditis rat model, we have

demonstrated a dark side of NETs in vivo. We also demonstrated that the underlying mechanism

of NETs formation is well coordinated and controlled by the endocarditis pathogen. Our previous

study indicated that upon entering the blood stream, IE-inducing pathogens, such as S. mutans,

can hijack or adopt host innate immune effectors such as IgG or platelets to form biofilms.16 In

the present study, we further demonstrated that NETs formation contributes directly to the

expansion of vegetation formation. It is well known that bacterial extracellular DNA contributes

to formation of bacterial biofilms.33, 34 Only NETs and its component, histones, but not bacterial

DNA, could trigger subsequent platelet aggregation and coagulation activation (Supplemental

Figure 7).24, 25 In addition to histones, NETs could trap platelets through von Willebrand factor

and activate coagulation system through other pathways, including the direct activation of factor

XII or indirect activation of factor XII through elastase and myeloperoxidase.3 Therefore, NETs

but not bacterial extracellular DNA contributes directly to vegetation expansion observed in our

IE model.

Based on the results presented in this study, a putative model for the roles of neutrophils

and platelets in vegetation maturation and expansion in IE is proposed as follows (Figure 7): IE-

inducing pathogens activate platelets through specific IgG, which contributes to bacteria-platelet

aggregate formation on the damaged valve.16 In addition, the bacteria also activate the infiltrated

neutrophils through specific IgG and TLR2 to produce reactive oxygen ROS, which could

he present study, we further demonstrated that NETs formation contributes direcctctlyyy ttoo thththee e

expansion of vegetation formation. It is well known that bacterial extracellular DNA contributes

oo ffforoormmamatititiononon oof bababacctcterial biofilms.33, 34 Only NETTsTs aaand its componenen nt,, hihihisststones, but not bacterial

DNDNDNAA, could tririggggeeer ssuubu ssesequqquenenentt plpllatatateele eeet aaggrrregggatiooonnn annndd cocoagaggululaaatioon n acacctiivavatititioonon ((SuSuSupppppllel mmemenntn aaal

FiFigugugurerere 77).).).24,24,4, 25252 InInn aaaddddiittiooon n too hhhisisistototonenenes,s, NNNETETETsss cocooululu ddd ttrtrapapp ppplalalatteteleleetstst thrhrhrououo gghgh vvononn WWWilillelelebbbranannd d fafacctctoorr

and activate ccoaoaoagugugulalalatitiononon sysysystetetem m m thththroroouguggh hh ototo heheherr r papapathththwawaw ysyss,, inininclclc udududinining g thththeee dididirererectctc aaactctctivivvatatatioioi n of factorrr

Page 14: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

14

further induce histone citrullination that is required for chromatin decondensation.27 On the other

hand, bacteria induce platelet activation and P-selectin expression, which involves Src family

kinases, Syk, PI3K, and p38 MAPK signaling. This activation also provides other signals to

neutrophils for NET production. The resulting NETs not only induce bacteria-platelet aggregate

formation but also entrap these aggregates to promote vegetation formation. Such phenomenon

may also be applied to the human endocarditis, because NETs have been identified

histophathologicaly in human specimens.35 Our in vivo study with nine rats conducted here

showed that digestion of NETs with DNase I significantly reduced the number of bacteria and

size of vegetation, which confirmed the role of NETs in the pathogenesis in IE. Therefore,

targeting NETs provides a new strategy for controlling IE. Although it has been reported that

NETs can kill bacteria, our data showed that digestion of NETs with DNase I decreased, but not

increased, the colonized bacterial number in vivo, suggesting that NETs have a limited effect on

bacterial growth. In fact, we found that NETs had a limited effect on S. mutans growth in vitro

(Supplemental Figure 14). Taken together, these data suggest that NETs may trap S. mutans in

platelet aggregates but may not sufficiently kill the bacterium, which is consistent with previous

results.36

It was recently proposed that the link between coagulation and the innate immune

response may constitute an innate effector mechanism, named ‘immunothrombosis’, which

mediates the recognition of pathogens and damaged cells and inhibits pathogen dissemination

and survival.3 Immunothrombosis is therefore considered to be a physiological process crucial

for intravascular immunity, while dysregulation of immunothrombosis may be one of the

underlying events that triggers thrombotic disorders, such as disseminated intravascular

coagulation. Neurophils constitute a key player in the development of immunothrombosis

argeting NETs provides a new strategy for controlling IE. Although it has been rerrepopoportttededed ttthahahat t

NETs can kill bacteria, our data showed that digestion of NETs with DNase I decreased, but not

nncrcrreaeaeassesedd,d, ttthehehe colololoononized bacterial number in vivvooo, ssuuggesting that t NEN TsTsTs hhhave a limited effect on

bbaactcttere ial growwththh.. IInn fffaaact,t,, wwee fofofounund dd ththt aatt NNNETTTs haddd aaa limimimiteteddd eefffefeccct oon n S.SS mmmututtanananss ggrrrowwtwth hh innn vvitititrooo

SuSuupppppplelel memeentntntalal FiFiFigugug rrere 11144).) TTTakakakenenen tttogoggetettheheher,r,r, thehehesesee dddatataa a sususuggggggesesest tt ththhatatat NNNETETETs s mmamay y y trtrapapap SS. mmuututananns iinn

platelet agggreregagagatetess s bubub tt t mamam y y y nonon t t sususuffffficici ieeentntntlylyly kkkilili l l l ththhee e babab ctctterere iuiuium,m,m, wwwhihiccch h h isisis ccconononsisistststenenent tt wiwiwitht previous

Page 15: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

15

through the ejection of a meshwork of NETs.1, 2 On the other hand, NETs may also provide a

scaffold to activate platelets and stimulate thrombus formation.24 Our results provide direct

evidence suggesting that NET-mediated immunothrombosis may play an important role in the

formation and maturation of vegetation induced by endocarditis pathogen on injured heart

valves. Such bacteria-mediated immunothrombosis may also be involved in thromboembolic

events, which are serious complications associated with IE in clinics.8, 9, 16

Research on NETs spans nearly a decade, but the mechanism of NET formation is still

unclear and controversial.22, 37 NETosis requires ROS production, histone citrullination, and

chromatin decondensation.37 ROS may trigger Ca2+-dependent PAD4 activity and lead to histone

citrullination, which is required for chromatin decondensation in NETosis.27, 29 However,

stimulation of neutrophils with hydrogen peroxide alone or formyl-methionyl-leucyl-

phenylalanine (fMLP), which is a potent inducer of NADPH oxidase activity, does not induce

NETosis,38, 39 suggesting that additional processes are required to promote NET formation.37 This

may explain why only some groups observed NET formation in response to LPS while others

found that NETosis occurred only in the present of platelets.2, 5 Our data showed that S. mutans

could induce histone citrullination in neutrophils in an NADPH oxidase activity-dependent

manner, but still required activated platelets to promote NET formation.

The findings of this study indicate that specific IgG plays a key role in S. mutans-

mediated platelet activation and NET formation. This result is consistent with previous studies

showing that platelet activation by IE pathogens requires specific IgG.18, 40 However, the

underlying downstream signaling pathway of the Fc IIA receptor on platelets is not clear. In the

presence of IgG, S. sanguinis causes tyrosine phosphorylation of Fc IIA, which leads to the

phosphorylation of Syk, LAT, and PLC 2 and were dependent on Src family kinases.41 Our data

citrullination, which is required for chromatin decondensation in NETosis.27, 29 HHHowowwevvvererer,, ,

timulation of neutrophils with hydrogen peroxide alone or formyl-methionyl-leucyl-

phhenenenylylylalalalanannininineee (ffMLMMLP), which is a potent induceerrr ooff f NADPH oxidddasa e acacacttitiviv ty, does not induce

NNETTTosis,38, 3939 ssugugggggeststinii ggg thththatatat aaadddditititioioonnaal procceessesss aaare rerereququiiri eeded ttoo pprpromommootte e NENENETT fofoformrmmatattiooonn.n.373737 TTThis

mamaayy y exexexplplp aiaiinn n whwhyyy oononlylyy ssomomo e grgrg ouououpspsps oobbsbserererveveved NENEN TT fformrmrmatatatioioion nn inini rresesespopop nnnseee totoo LLLPSPSP wwwhhhileee oooththererrs

found that NNETETETososo isisis ooccccc uruu rereed d d ononnlylyly ininn thehehe ppprerereseseentntn oooff f plplplatateleleletetets.s.s.222, 555 OOOurrr dddatatata aa shshshowowowededed ttthahahatt t S.S mutans

Page 16: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

16

also showed that S. mutans-stimulated P-selectin expression was dependent on Src family

kinases and Syk. Furthermore, we also showed that PI3K and p38 MAPK play a role in P-

selectin expression on platelets, which might be similar to the signaling pathway of Fc in

phagocytes.42 Surprisingly, phosphorylation of p38 MAPK was dependent on Src family kinases,

but not Syk or PI3K in platelets. These results suggest that the signaling pathways downstream

of the Fc receptor in phagocytes and platelets are different. It was previously reported that p38

MAP kinase in human platelets could be activated by several platelet agonists, including

thrombin, thromboxane A2 (TxA2), ADP, and von Willebrand factor, but the upstream and

downstream pathways are not quite similar.43, 44 Therefore, the role and the underlying

mechanism of p38 MAPK in IgG-mediated platelet activation will require further investigation.

The binding of circulating neutrophils in the bloodstream to activated and immobilized

platelets is mainly mediated by the interaction of P-selectin on the activated platelet and PSGL-1

on the neutrophil.30, 31 The downstream signaling of PSGL-1 could induce integrin activation,

including Mac-1 (CD11b/CD18) and LFA-1 (CD11a/CD18), which contribute to the firm

adhesion of neutrophils to immobilized platelets.45, 46 Studies on sepsis have shown that

inhibition of the adhesion of platelets and neutrophils by targeting LFA-1 reduced NET

production.47 However, LPS could not stimulate P-selectin expression on platelets,5 and therefore

the underlying mechanism by which LPS stimulates platelet binding to neutrophils will require

further investigation. It is possible that different mechanisms are involved in LPS- and gram-

positive pathogens-regulated NETosis. However, neutralizing antibodies against LFA-1 also

reduced S. mutans-activated platelet-mediated NETosis (data not shown). Therefore, the

contribution of LFA-1 in this process cannot be excluded.

During NETosis induced by bacteria-activated platelets, the same signaling events are

mechanism of p38 MAPK in IgG-mediated platelet activation will require furthererr iinvnn esesestititigagagatititionon.

The binding of circulating neutrophils in the bloodstream to activated and immobilized

pllatatteleleletetetss isisis mmmaainlnllyy y mem diated by the interaction ofoff PP--selectin on thheee actitiivavavatet d platelet and PSGL-1

on ttthhe neutropphihihil.l 330, 313131 TThehehe dddowowownsnstrtrt eeaeammm ssignnalling off PSGSGSGL-L--1 cocouuuldd d inindudud ccece iintntntegegririinn n aacactititivaaattitiononn,

nnclclcludududinini gg g MaMaMac-c-11 (CCCDD111b1bb/C/CDD1D18)8)8) aaandndnd LLLFAFAFA-1-1-1 ((CDCDC 1111aa/CCCD1D1D188)8), whwhwhicichhh cocoontntriribubuutetee tto o thththee fififirmmm

adhesion of neneeututu roroophphphilills s to iiimmmmm obobobililizizizeddd ppplalalateteteleleletstst .454545, 464646 SStututudididieseses ooon n n sesepspsp isisis hhhavavaveee shshhowowown n n thththat

Page 17: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

17

required for platelet and neutrophil activation, including Src family kinases, and Syk. This could

explain why the pharmacological inhibition of these kinases completely inhibited NETosis

induced by bacteria-activated platelets (Figure 5F). Nevertheless, the potential adverse effects of

these inhibitors may not warrant their routine usage in clinics. In addition to S. mutans, we also

investigated the role of NETs in vegetation expansion by using another IE-inducing streptococci,

S. gordonii DL1 (Supplemental Figure 15), whose surface proteins Hsa or GspB and SspA or

SspB could directly bind to platelets to trigger the platelet activation in an IgG-independent

manner.48 S. gordonii-platelet aggregates also induced NET formation in vitro and in vivo, and

prophylactic administration of DNase I combined with penicillin significantly reduced the size of

the vegetation as well as the colonized bacteria in the vegetation (Supplemental Figure 15). Our

previous study showed that prophylaxis with aspirin or penicillin could not reduce S. gordonii-

induced vegetation formation and bacterial colonization.16 Therefore, the inclusion of DNase I

administration in addition to routine antibiotic agents, such as penicillin, may be helpful in

prophylactic therapy for controlling IE.

In summary, we have provided experimental evidence to indicate that endocarditis

pathogen could hijack NETs to promote the spreading and thickening of bacterial biofilms

accompanied with aggregated platelets leading to the expansion of vegetation.

Acknowledgments: We would like to thank John T Kung, Academia Sinica, for providing TLR2

KO mice, Shu-Chun Teng, National Taiwan University, for helpful discussion, and Chi-Ling

Chen, National Taiwan University, for statistical analysis.

Funding Sources: This work was supported by the National Science Council (grants NSC 102-

2321-B-002 -010 and 102-2321-B-002-055).

he vegetation as well as the colonized bacteria in the vegetation (Supplementalal FFigigigururre e 151515).).). OOur

previous study showed that prophylaxis with aspirin or penicillin could not reduce S. gordonii-

nnduduuceceeddd vevevegegeg ttatiiononon fformation and bacterial coloonninizaation.16 Therefofofore,, thththee e inclusion of DNase I

addmmiministrationn iiinnn aadaddididitit oonon tttoo rororoututininineee anannttiibiottticcc ageeennnts, ssuucuchh h aass pepeenniicicilllllinii ,, mamaay yy bebe hhhelelelpfpffululu iiinn

prpropopophyhyhylalactcticicic ttheherrarappypy fffororr cconontrtrt ololollililingngng IIEE.E.

In sumummmamam ryryry, , wewew havavave e e prprrovovovididdedede eeexpxpxperererimimimenenenttalalal eeviviidededencncnce e tototo iindnddiciccatatate e e thththatat eeendndndocococarara ditis

Page 18: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

18

Conflict of Interest Disclosures: None.

References:

1. Wartha F, Beiter K, Normark S, Henriques-Normark B. Neutrophil extracellular traps: casting the NET over pathogenesis. Curr Opin Microbiol. 2007;10:52-56. 2. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532-1535. 3. Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity. NatRev Immunol. 2013;13:34-45. 4. Phillipson M, Kubes P. The neutrophil in vascular inflammation. Nat Med. 2011;17:1381-1390. 5. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD, Chakrabarti S, McAvoy E, Sinclair GD, Keys EM, Allen-Vercoe E, DeVinney R, Doig CJ, Green FHY, Kubes P. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. 2007;13:463-469. 6. Hickey MJ, Kubes P. Intravascular immunity: the host-pathogen encounter in blood vessels. Nat Rev Immunol. 2009;9:364-375. 7. Jenne CN, Wong CH, Zemp FJ, McDonald B, Rahman MM, Forsyth PA, McFadden G, Kubes P. Neutrophils recruited to sites of infection protect from virus challenge by releasing neutrophil extracellular traps. Cell Host Microbe. 2013;13:169-180. 8. Mylonakis E, Calderwood SB. Infective endocarditis in adults. N Engl J Med. 2001;345:1318-1330. 9. Moreillon P, Que YA. Infective endocarditis. Lancet. 2004;363:139-149. 10. Kim HK, Thammavongsa V, Schneewind O, Missiakas D. Recurrent infections and immune evasion strategies of Staphylococcus aureus. Curr Opin Microbiol. 2012;15:92-99. 11. Jung CJ, Zheng QH, Shieh YH, Lin CS, Chia JS. Streptococcus mutans autolysin AtlA is a fibronectin-binding protein and contributes to bacterial survival in the bloodstream and virulence for infective endocarditis. Mol Microbiol. 2009;74:888-902. 12. McNicol A, Israels SJ. Mechanisms of oral bacteria-induced platelet activation. Can J Physiol Pharmacol. 2010;88:510-524.

13. Fitzgerald JR, Foster TJ, Cox D. The interaction of bacterial pathogens with platelets. NatRev Microbiol. 2006;4:445-457.

5. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KDD, , , ChChC akakrarababarrti S, McAvoy E, Sinclair GD, Keys EM, Allen-Vercoe E, DeVinney R, Doig CJ, GGrrreenenen FFFHYHYHY, ,,Kubes P. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteriria a ininin ssepepeptititicccblood. Nat Med. 2007;13:463-469.

6.. HHHicicickekekey y MJMJMJ, KuKuKubbebes P. Intravascular immunity::: ththt ee host-pathogeen nn enncccououounter in blood vessels. NaNaNat t RReRev Immumumunonolll.. 202009099;9;99:3:3:36464-3-33757575..

7.7.. Jeenenne CN,N, WWoonongg CHH,, Zeempmpmp FJ,J MMMccDDoonallld B, RaRaRahmhmmananan MMMMMM, FFFooorsythhh PPPAA,, MMMcFc aadadddedennn GGG, KKububbesP.P. NNNeueueutrtrt opopophihihilslls rreeecrruruititeeded ttoo siitetet ss ofofof iiinnfnfeecectititiononon prrrototo eecct t frf omomom vvviirirususus cchahahallllllenene ggege bby y rererelelel asassininng g nneneututu roroophhhili extrtracacelellululalar trrapaps.. CeCell HHoso t t MiMicrcrobe. 20201313;1;13:3:16169-18180.0

88 MMylylononakakisis EE CaCaldldererwowoodod SSBB IInnfefectctivivee enendodocacardrdititisis iinn adadulultsts NN EnEnglgl JJ MMeded 20200101;3;34545:1:1313188--

Page 19: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

19

14. Durack DT. Experimental bacterial endocarditis. IV. Structure and evolution of very early lesions. J Pathol. 1975;115:81-89. 15. Donlan RM, Costerton JW. Biofilms: survival mechanisms of clinically relevant microorganisms. Clin Microbiol Rev. 2002;15:167-193.

16. Jung CJ, Yeh CY, Shun CT, Hsu RB, Cheng HW, Lin CS, Chia JS. Platelets enhance biofilm formation and resistance of endocarditis-inducing streptococci on the injured heart valve. J Infect Dis. 2012;205:1066-1075. 17. Shun CT, Lu SY, Yeh CY, Chiang CP, Chia JS, Chen JY. Glucosyltransferases of viridans streptococci are modulins of interleukin-6 induction in infective endocarditis. Infect Immun. 2005;73:3261-3270. 18. Chia JS, Lin YL, Lien HT, Chen JY. Platelet aggregation induced by serotype polysaccharides from Streptococcus mutans. Infect Immun. 2004;72:2605-2617. 19. Borregaard N, Cowland JB. Granules of the human neutrophilic polymorphonuclear leukocyte. Blood. 1997;89:3503-3521. 20. Wilson W, Taubert KA, Gewitz M, Lockhart PB, Baddour LM, Levison M, Bolger A, Cabell CH, Takahashi M, Baltimore RS, Newburger JW, Strom BL, Tani LY, Gerber M, Bonow RO, Pallasch T, Shulman ST, Rowley AH, Burns JC, Ferrieri P, Gardner T, Goff D, Durack DT. Prevention of infective endocarditis: guidelines from the American Heart Association: a guideline from the American Heart Association Rheumatic Fever, Endocarditis and Kawasaki Disease Committee, Council on Cardiovascular Disease in the Young, and the Council on Clinical Cardiology, Council on Cardiovascular Surgery and Anesthesia, and the Quality of Care and Outcomes Research Interdisciplinary Working Group. Circulation. 2007;116:1736-1754. 21. Hall G, Heimdahl A, Nord CE. Bacteremia after oral surgery and antibiotic prophylaxis for endocarditis. Clin Infect Dis. 1999;29:1-8; quiz 9-10. 22. Chung M. Prescription of antibiotics for prophylaxis to prevent bacterial endocarditis. J Am Dent Assoc. 2009;140:1025-1026. 23. Oliver R, Roberts GJ, Hooper L. Penicillins for the prophylaxis of bacterial endocarditis in dentistry. Cochrane Database Syst Rev. 2004:CD003813. 24. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD, Jr., Wrobleski SK, Wakefield TW, Hartwig JH, Wagner DD. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A. 2010;107:15880-15885. 25. Semeraro F, Ammollo CT, Morrissey JH, Dale GL, Friese P, Esmon NL, Esmon CT. Extracellular histones promote thrombin generation through platelet-dependent mechanisms: involvement of platelet TLR2 and TLR4. Blood. 2011;118:1952-1961.

19. Borregaard N, Cowland JB. Granules of the human neutrophilic polymorphonunuuclclcleaear rfeukocyte. Blood. 1997;89:3503-3521.

20. Wilson W, Taubert KA, Gewitz M, Lockhart PB, Baddour LM, Levison M, Bolger A, Cabell CH, Takahashi M,, Baltimore RS, Newburger JWW, , Strom BL, Tani LY, GGerber M, Bonow RO, WWPaalllllasasaschchch TTT,, ShShShullmamaman n ST, Rowley AH, Burns JC, FeFeF rrrieri P, Gardneneer T,T, GGGoofo f D, Durack DT.PrPrrevvvention oofff innfefefectctc ivive e e enenndodod cacarrrdididititit s:s: ggguiuidedelilineness frfrromm thehee AAAmemm rriciccananan HHeaeaartrtr Assssococociaiaiatitionon:: aa a gguidddele ine fromm tthehehe AAAmmmeririricacacann n HeHHeararrtt AsAsA sosoociatttiooon RRhRheeumamamatitic c FFeevever, EEndnddooccaardrdditititiisis aandndnd KKKawawawaasasakakaki iDDDiseeease Commmmittttetee, CCCouuuncciill ooon CaCardrdr iioovvvascuululaar DDDissseaeasesese iiinn thhhe YYYouuung, anndnd tthehee CCouuunnccilil oonn rClCllinininicicicalala CCararardiddiolologogogy,y,y CCCououuncncilill oon nn CaCaCarrdrdioioovavavascscscululararar SSSururrgegeeryryry aaandnnd AAAneneeststthhhesese iiia,, anand dd thththe e QuQuQualliiityyy ofof CCaarareand d OuOutcomomess RResseaearcr h InInteerdrdisisciciplinarry y WoWorkrkining GrGrououp.p. CiCircrcullatatioonn.. 2000707;111616:173736-6 17175454.

2121 HaHallll GG HeHeimimdadahlhl AA NoNordrd CCEE BBacactetereremimiaa afafteterr ororalal ssururgegeryry aandnd aantntibibioiotiticc prpropophyhylalaxixiss foforr

Page 20: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

20

26. Gould TJ, Vu TT, Swystun LL, Dwivedi DJ, Mai SH, Weitz JI, Liaw PC. Neutrophil extracellular traps promote thrombin generation through platelet-dependent and platelet-independent mechanisms. Arterioscler Thromb Vasc Biol. 2014;34:1977-1984. 27. Wang Y, Li M, Stadler S, Correll S, Li P, Wang D, Hayama R, Leonelli L, Han H, Grigoryev SA, Allis CD, Coonrod SA. Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J Cell Biol. 2009;184:205-213. 28. Oliveira-Nascimento L, Massari P, Wetzler LM. The role of TLR2 in infection and immunity. Front Immunol. 2012;3:79. 29. Neeli I, Khan SN, Radic M. Histone deimination as a response to inflammatory stimuli in neutrophils. J Immunol. 2008;180:1895-1902. 30. Lalor P, Nash GB. Adhesion of flowing leucocytes to immobilized platelets. Br J Haematol. 1995;89:725-732. 31. Yang J, Furie BC, Furie B. The biology of P-selectin glycoprotein ligand-1: its role as a selectin counterreceptor in leukocyte-endothelial and leukocyte-platelet interaction. Thromb Haemost. 1999;81:1-7. 32. Hogg N, Patzak I, Willenbrock F. The insider's guide to leukocyte integrin signalling and function. Nat Rev Immunol. 2011;11:416-426. 33. Thomas VC, Thurlow LR, Boyle D, Hancock LE. Regulation of autolysis-dependent extracellular DNA release by Enterococcus faecalis extracellular proteases influences biofilm development. J Bacteriol. 2008;190:5690-5698. 34. Huseby MJ, Kruse AC, Digre J, Kohler PL, Vocke JA, Mann EE, Bayles KW, Bohach GA, Schlievert PM, Ohlendorf DH, Earhart CA. Beta toxin catalyzes formation of nucleoprotein matrix in staphylococcal biofilms. Proc Natl Acad Sci U S A. 2010;107:14407-14412. 35. Al-Salih G, Al-Attar N, Delbosc S, Louedec L, Corvazier E, Loyau S, Michel JB, Pidard D, Duval X, Meilhac O. Role of vegetation-associated protease activity in valve destruction in human infective endocarditis. PLoS One. 2012;7:e45695. 36. Menegazzi R, Decleva E, Dri P. Killing by neutrophil extracellular traps: fact or folklore? Blood. 2012;119:1214-1216. 37. Remijsen Q, Kuijpers TW, Wirawan E, Lippens S, Vandenabeele P, Vanden Berghe T. Dying for a cause: NETosis, mechanisms behind an antimicrobial cell death modality. Cell Death Differ. 2011;18:581-588. 38. Remijsen Q, Berghe TV, Wirawan E, Asselbergh B, Parthoens E, De Rycke R, Noppen S, Delforge M, Willems J, Vandenabeele P. Neutrophil extracellular trap cell death requires both autophagy and superoxide generation. Cell Res. 2010;21:290-304.

31. Yang J, Furie BC, Furie B. The biology of P-selectin glycoprotein ligand-1: its s rororolele aass a a electin counterreceptor in leukocyte-endothelial and leukocyte-platelet interacttioioon... ThThThrororombmbmb

Haemost. 1999;81:1-7.

32. Hoggg N,, Patzak I, Willenbrock F. The insider's gug ide to leukocyte integrin signalling and fuuncncctititioonon. NaNaNat tt Reev vv IImImmunol. 2011;11:416-426.

3333. TThomas VVC,C,C TThuurlrlr owww LLLR,R,R, BBoyoyylelele DDD, Hanncncoock k LLE. ReReR guguulaaatitioonn oof f auauutooolyysisisiss-s-dedepeppendndndeeent t texexxtrrraca ellular r DNDNNAA A releeaasse bybyy EEEntet roroococooccccus fffaaecallisss eextxtxtrararaceceellluulaarr ppproteeasasseses innfnfluluennnceees s bbibioofilmmm dedevvevelololopmpmpmenenent.tt. J J BaBaBactctererrioooll. 2000080808;1;1;1909090:5:56969690-0-0-55656989898.

34. Huseby MMMJ,J,J, KKKrururusesee AAAC,C,, DDDigigrerere JJ,, KoKoKohlhlhlererer PPPL,L,L, VVVococo kekek JJJA,A,A, MMMananann n n EEEEE,, BaBaBaylylylesese KKKW,W,W, BBBohoho ach GA,ScSchlhlieievevertrt PPMM OOhlhlenendodorfrf DDHH EEararhahartrt CCAA BBetetaa totoxixinn cacatatalylyzezess foformrmatatioionn ofof nnucucleleopoproroteteinin

Page 21: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

21

39. Urban CF, Ermert D, Schmid M, Abu-Abed U, Goosmann C, Nacken W, Brinkmann V, Jungblut PR, Zychlinsky A. Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 2009;5:e1000639. 40. Sullam PM, Jarvis GA, Valone FH. Role of immunoglobulin G in platelet aggregation by viridans group streptococci. Infect Immun. 1988;56:2907-2911. 41. Pampolina C, McNicol A. Streptococcus sanguis-induced platelet activation involves two waves of tyrosine phosphorylation mediated by FcgammaRIIA and alphaIIbbeta3. Thromb Haemost. 2005;93:932-939. 42. Joshi T, Butchar JP, Tridandapani S. Fcgamma receptor signaling in phagocytes. Int J Hematol. 2006;84:210-216. 43. Canobbio I, Reineri S, Sinigaglia F, Balduini C, Torti M. A role for p38 MAP kinase in platelet activation by von Willebrand factor. Thromb Haemost. 2004;91:102-10. 44. Begonja AJ, Geiger J, Rukoyatkina N, Rauchfuss S, Gambaryan S, Walter U. Thrombin stimulation of p38 MAP kinase in human platelets is mediated by ADP and thromboxane A2 and inhibited by cGMP/cGMP-dependent protein kinase. Blood. 2007;109:616-618. 45. Weber C, Springer TA. Neutrophil accumulation on activated, surface-adherent platelets in flow is mediated by interaction of Mac-1 with fibrinogen bound to alphaIIbbeta3 and stimulated by platelet-activating factor. J Clin Invest. 1997;100:2085-2093. 46. Diacovo TG, deFougerolles AR, Bainton DF, Springer TA. A functional integrin ligand on the surface of platelets: intercellular adhesion molecule-2. J Clin Invest. 1994;94:1243-1251. 47. McDonald B, Urrutia R, Yipp Bryan G, Jenne Craig N, Kubes P. Intravascular neutrophil extracellular traps capture bacteria from the bloodstream during sepsis. Cell Host Microbe. 2012;12:324-333. 48. Kerrigan SW, Jakubovics NS, Keane C, Maguire P, Wynne K, Jenkinson HF, Cox D. Role of Streptococcus gordonii surface proteins SspA/SspB and Hsa in platelet function. Infect Immun. 2007;75:5740-5747.

44. Begonja AJ, Geiger J, Rukoyatkina N, Rauchfuss S, Gambaryan S, Walter U.U. TThrhrh omomombibibin n ntimulation of p38 MAP kinase in human platelets is mediated by ADP and thrommboboboxaxanenene AAA222 ananandddnhibited by cGMP/cGMP-dependent protein kinase. Blood. 2007;109:616-618.

455. . WeWeWebebeberrr CC,C, Sprprrininingger TA. Neutrophil accumulatatatioionn on activatedd,, , surfrffacacacee-adherent platelets in flflowowow iis medidiiaaateded bbbyy ininteteteraraactctc iionon oooff f MaMac-c-11 wiwithth ffibibririnnogegenn n bobobounund d d tototo aalplpphahahaIII bbbbetetta3a3a3 aandnd sstititimumuulalalateted bbyy ppplal telet-actitivavaatiiingg ffacacctootorr.r. J J J ClClininn IIInvnvvessst. 1199997;11000:20808085-5-2020209393..

4666. DiDiDiacaca ovovvoo o TTGTG, ,, dedeeFoFoouguggererolllelel s s ARARAR, BaBaainnntototonnn DFDFDF,, SpSpprir ngngngererer TTTA.A.A AAA ffunununctctiiionnanal l inini tetetegrgrgrininin ligiggannnd d ooon tthehurfrfacace e ofof pplatetelel tss:: inintercecellulularar aadhd esioon n momolelecucule-22. J J ClClinin IInvvesestt. 1919944;9; 4:4:1212434 -11252 1.1.

4747 McMcDoDonanaldld BB UrUrrurutitiaa RR YYipippp BrBryayann GG JJenennene CCraraigig NN KuKubebess PP IIntntraravavascsculularar nneueutrtropophihill

Page 22: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

22

Figure Legends:

Figure 1. In situ NET formation inside vegetation on the damaged valve in bacterial endocarditis

rats. Catheterized Wistar rats were intravenously infected with green fluorescent protein-tagged

S. mutans GS5 at an inoculum of 109 CFUs. The vegetations were then harvested and stained

with Hoechst 33258 and anti-elastase antibody (1:50 dilution) followed by a Texas red-

conjugated secondary antibody (1:200 dilution). The co-localization of elastase and extracellular

DNA indicate the in situ NET formation inside the vegetation as observed by confocal

microscopy (magnification 630X). The bottom panels represent projections of z-stacks. The

experiments were repeated three times and a representative experiment is shown.

Figure 2. Digestion of NETs by DNase I reduces the bacterial colonization and vegetation size in

vivo. (A) DNase I (10000 Kunitz units/kg) was injected into the experimental endocarditis rats

30 min before inoculation of the bacteria. The top images show vegetation formation on the

damaged valve in situ. The vegetation that formed 24 h after injection with DNase I was smaller

in size. The bottom images are the CLSM images inside the vegetation. Bars indicate 10 m. (B)

The NETs inside the vegetation was quantified with a Volocity program from three separate

experiments. Colocalization of elastase and extracellular DNA was defined as NETs. The data

were analyzed by the Student’s t-test. **P < 0.01. The mean ± SD was 33.9 ± 7.3 in no treatment

group versus 8.4 ± 1.3 in DNase I group; n=3. The effects of DNase I in the vegetation formation

(C), bacterial colonization (D), and number of circulating bacteria (E) in vivo were further

analyzed for statistical significance. Scatter plots show values from individual rat (n=9) and

medians (bars). ***P < 0.001; **P < 0.01; *P < 0.05 as determined by Mann-Whitney U test.

The mean ± SD and median were 4.2 ± 1.9 and 4.0 in no treatment group versus 1.4 ± 1.3 and

experiments were repeated three times and a representative experiment is shownn..

Figure 2. Digeg stion of NETs by DNase I reduces the bacterial colonization and vegetation size in

viviivovovo.. (A(A) DNDNNasasa ee III (1(100000000000 KKKununitittzzz ununu itits/s/kgkg) ) wawass ininjejecctedd iiintntntoo o the e e exexexpepep ririmemem nttalal eeendndndococarardididititit s s rararatsts

30300 mmmin befororee innococculaattiooon ooff ttthhe bacaccteteririaa.a Thehehe toppp iimmagagageseses sshohoow vevegetaattioonon foorrmam tiiionnn ooon the

damamageg dd vavalvee iin sisitutu. Thhe e vevegegetatatition thahat t foformrmeded 244 h h afafteter r ininjeectctioion n wiw thth DDNaNase I wwass ssmamallleer

nn ssizizee TThehe bbotottotomm imimagageses aarere tthehe CCLSLSMM imimagageses iinsnsididee ththee vevegegetatatitionon BaBarsrs iindndicicatatee 1010 mm ((B)B))

Page 23: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

23

0.9 in DNase I group in panel C. The mean log10 CFU ± SD and median were 7.5 ± 0.7 and 7.5

in no treatment group versus 5.4 ± 1.1 and 5.4 in DNase I group in panel D. The mean log10 CFU

± SD and median were 1.9 ± 0.3 and 1.8 in no treatment group versus 2.4 ± 0.6 and 2.5 in DNase

I group in panel E.

Figure 3. NETs contribute to bacteria-platelet aggregate formation in the PRP. (A) S. mutans was

grown in 300 l PRP (left panel) or PPP supplemented with 1% (w/v) glucose (middle panel).

Addition of 5 g/ml ADP in 300 l PRP was performed as a control (right panel). For each well,

peripheral blood-isolated neutrophils were added to a final concentration of 2 x 106 neutrophils

per ml. S. mutans were labeled with GFP (green) and the platelets were visualized by staining

with rhodamine-conjugated phalloidin (1:200 dilution). The NET formation stained with Hoechst

33258 was observed by CLSM (magnification 630X). NETs were only induced by bacteria-

platelet aggregates in the PRP. (B) The NADPH oxidase inhibitor DPI (10 M) was added when

neutrophils were added in the bacteria-platelet aggregates in the PRP, and visualization was

performed on a CLSM (magnification 630X). (C) 3D-CLSM image of S. mutans-platelet

aggregates in PRP, PRP containing isolated neutrophils, or PRP containing isolated neutrophils

and 2 Kunitz units/ml DNase I. (D) The biomass of the bacteria-platelet aggregates represented

in (C) was analyzed using the Volocity program. The mean ± SD was 0.66 ± 0.07 in control

group, 1.21 ± 0.15 in neutrophil group, and 0.49 ± 0.07 in neutrophil+DNase I group; n=4. The

data were analyzed by one-way ANOVA. ***P < 0.001. These experiments were repeated three

times and a representative experiment is shown.

Figure 4. Bacteria stimulate histone citrullination in neutrophils and activated platelets are

required for promoting NET formation. (A) Peripheral blood-isolated neutrophils were placed on

per ml. S. mutans were labeled with GFP (green) and the platelets were visualizeed d d bybyby sstatataininininininggg

with rhodamine-conjugated phalloidin (1:200 dilution). The NET formation stained with Hoechs

332525888 wawawass s obobbseervrvrvedede by CLSM (magnification 636330X0X0X). NETs were onlly y y inininduced by bacteria-

plpllattteelet aggreregagaattetesss ininn ttthehehe PPRPRPRP. (B(B(B) )) ThThTheee NANANADPDPDPH H ooxoxiidasasseee inininhihihibibitototor DPDPDPI I I (1(1(10 M)M)M) wwwasasas aaaddddddededed wwwheheh n

neneeutututroror phphils wewew rere aaddddeedd iin ththhe e babab ctcterereriaia-p-plalatetelelett agaggrgrgregegaatateses iinn ththe e PPRRP,, aaanndd vvvisisuauau liizzaatititionnn wwwasas

pepeperfrfrfororormememeddd ononon aaa CCCLSLSLSMMM (m(m(magagagnininififificacacatititiononon 666303030X)X)X). (C(C(C))) 3D3D3D C-C-CLSLSLSMMM imimimagagageee ofofof SSS. mmmutututananansss p-p-plalalatetetelelelettt

Page 24: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

24

the plasma-coated coverslip (100 l of 107 cells/ml in RPMI) in wells of a 24-well plate for 1 h.

After removing the non-adhered neutrophils, the neutrophils were stimulated by S. mutans GS5

(108/ml) alone or in combination with IgG (1 mg/ml) or washed platelets. S. mutans were labeled

with GFP (green) and the platelets were visualized by staining with rhodamine-conjugated

phalloidin (1:200 dilution). NET formation stained with Hoechst 33258 was observed by CLSM

(magnification 630X). A yellow appearance is observed where S. mutans formed aggregates with

platelets. NETs could only be induced by bacteria-platelet aggregates. (B) Addition of aspirin

(500 g/ml) inhibited the bacteria forming aggregates with platelets and also inhibited NET

formation. (C) Histone H3 citrullination in neutrophils stimulated by S. mutans GS5, IgG, or

washed platelets, alone or together, were analyzed by Western blotting. The role of IgG, TLR2, or

NADPH oxidase activity in histone H3 citrullination in neutrophils was further confirmed by

incubating the samples with a neutralizing antibody against FcR (20 g/ml) or Fc binding

inhibitor (D), neutralizing antibody against TLR2 (25 g/ml) (E), or NADPH oxidase inhibitor,

DPI (10 M) (F).

Figure 5. IgG-mediated P-selectin expression on platelets involves p38 MAPK activation.

Inhibition of the binding of bacteria-platelet aggregates to neutrophils and NET formation

without (A) or with the addition of 20 g/ml of a neutralizing antibody against P-selectin (B) or

PSGL-1 (C) in neutrophils stimulated with S. mutans, washed platelets, and IgG. (D) P-selectin

expression on platelets was further analyzed by flow cytometry. Washed platelet suspensions

were pretreated with aspirin (500 g/ml), the Src family kinase inhibitor PP2 (20 M), the Syk

inhibitor piceatannol (100 M), the PI3K inhibitor LY294002 (100 M), the p38 inhibitor

SB203580 (50 M), or a vehicle control (DMSO) for 30 min and then stimulated by S. mutans

GS5 (108/ml) and IgG (1 mg/ml). The data were analyzed by one-way ANOVA from a triplicate

washed platelets, alone or together, were analyzed by Western blotting. The rolee ooof f IgIgI G,G,G, TTTLRLRLR22, o

NADPH oxidase activity in histone H3 citrullination in neutrophils was further confirmed by

nncucuubababatititingngg tttheheh samamamplp es with a neutralizing antibboododyyy against FcR (2(2(20 g/g/g/mmml) or Fc binding

nnhiiibbitor (D), nneueuuttrralallizizizinnng g ananantititibbobodydydy aaggaaiinnst TTTLLLR22 ((225 gg/g/mmml)) (E(E)), oor r NNANADPDPDPH H H oxoxidididasassee ininhihihibiibitotoor,r

DPDPPII I (1(1(100 M)M)M) ((F)F)).

Page 25: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

25

experiment. ***P < 0.001. The mean ± SD from left to light lane was 8.3 ± 0.5, 5.7 ± 0.2, 162.0

± 19.9, 27.6 ± 5.9, 4.8 ± 0.1, 49.1 ± 6.0, 7.7 ± 0.4, 6.4 ± 0.2 and 146.0 ± 17.7, respectively; n=3

(E) Phosphorylation of p38 MAPK in platelets was further analyzed by Western blotting with an

anti-phospho-p38 MAPK antibody. (F) Peripheral blood-isolated neutrophils were stimulated

with S. mutans GS5 (108/ml), IgG (1 mg/ml), and washed platelets, which were pretreated with

the Src family kinase inhibitor PP2 (20 M), the Syk inhibitor piceatannol (100 M), the PI3K

inhibitor LY294002 (100 M), the p38 inhibitor SB203580 (50 M), or an equal volume of

DMSO as a vehicle control. S. mutans were labeled with GFP (green) and the platelets were

visualized by staining with rhodamine-conjugated phalloidin (1:200 dilution). NET formation

stained with Hoechst 33258 was observed by CLSM (magnification 630X). These experiments

were repeated three times and a representative experiment is shown.

Figure 6. The roles of P-selectin and IgG in S. mutans GS5-stimulated NET formation.

Peripheral blood-isolated neutrophils were stimulated by S. mutans GS5 (108/ml) combined with

recombinant P-selectin (1 g/ml) (A) or S. mutans GS5 (108/ml) combined with recombinant P-

selectin (1 g/ml) and IgG (1 mg/ml) (B). S. mutans were labeled with GFP (green) and NET

formation stained with Hoechst 33258 was observed by CLSM (magnification 630X). (C)

Peripheral blood-isolated neutrophils were pretreated with the Src family kinase inhibitor PP2

(20 M), the Syk inhibitor piceatannol (100 M), the Tec kinase inhibitor LFM-A13 (100 M),

the p38 inhibitor SB203580 (50 M), or an equal volume of DMSO as a vehicle control for 30

min, and then stimulated with S. mutans GS5 (108/ml) in combination with recombinant P-

selectin (1 g/ml) and IgG (1 mg/ml) for 3 h. NET formations were stained with Hoechst 33258.

The area of NET DNA fibers and total DNA was quantified from three random fields per sample

using the Volocity program. Total DNA was taken as 100%. The data were analyzed by one-way

tained with Hoechst 33258 was observed by CLSM (magnification 630X). Theesesse eeexpxppereerimimimenenentsts

were repeated three times and a representative experiment is shown.

FFiFiguguure 6. ThThe e rororoleleles ofofof PPP-s-s-sellelececctititinn n anannd d d IgIgIgG GG ininin S.S.S mmuututaansss GGGS5S5S5-ss-stitimmumulalalateteted d d NNNET T T fofoformrmrmatatatioioonn.n.

PePeeririphphp ereral bblololoodod--isosolalateted d neneututu rrorophphilililss wewewerere ssstimumullaatteed d bybyby S.S. mmmuttaananss GSSS5 ((101008/m/m/ l)l) cccoomombbbinnened d wwiwith

eeecococombmbmbinininanananttt PP-P-seseselelelectctctininin (((111 g/g/g/mlmlml))) (A(A(A))) ororor SSS. mmmutututananansss GGGS5S5S5 (((10101088/m/m/ml)l)l) cccomomombibibineneneddd wiwiwiththth rrrecececomomombibibinananantntnt PPP--

Page 26: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

DOI: 10.1161/CIRCULATIONAHA.114.011432

26

ANOVA from a triplicate experiment. *P < 0.05; ***P < 0.001. The mean ± SD from left to

light lane was 5.9 ± 1.8, 5.4 ± 1.6, 23.4 ± 5.8, 37.9 ± 6.1, 3.0 ± 1.0, 4.7 ± 2.5, 22.9 ± 3.7, 5.7 ±

2.5 and 36.9 ± 7.9, respectively; n=3. These experiments were repeated three times and a

representative experiment is shown.

Figure 7. A hypothetical model for NET-induced vegetation expansion on injured heart valves.

IE pathogens form aggregates with platelets on the heart valve by stimulating platelets through

IgG bearing the Fc receptor. After activation, P-selectin is expressed on the platelets, which

involves Src family kinases, Syk, PI3K, and p38 MAPK signaling pathways. In addition, the

bacteria stimulate ROS production and histone citrullination in neutrophils through the Fc

receptor and TLR2. Two separate signals from the bacteria and the activated platelets,

respectively, eventually induce NETosis. The resulting NETs not only promote coagulation

activation and platelet aggregation but also entrap bacteria-platelet aggregates in situ to promote

and consolidate vegetation expansion.

bacteria stimulate ROS production and histone citrullination in neutrophils throuughghgh ttthehehe FFFccc

eceptor and TLR2. Two separate signals from the bacteria and the activated platelets,

esppececctititivevevelylyly,,, eve enenntutut ally induce NETosis. The resessululultitit ng NETs not onlyy ppproror mote coagulation

acacctivvation aandndd ppplalalatteleleet tt agagggrgrg egegegatatatioioion n n bububut tt alala sosoo eeentntn rarappp bbbacttterereriaiaia-p-p-plalal tetetelelet tt agagagggrgreeegatateseses ininin ssititituu tttoo prprpromomomooto e

anannd d ccoc nsn olididdatata e e vevevegegettaatiiion eexpxpxpanansisiionono .

Page 27: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

Figure 1

Page 28: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

Figure 2

Page 29: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

Figure 3

Page 30: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

Figure 4

Page 31: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

Figure 5

Page 32: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

Figure 6

Page 33: Endocarditis Pathogen Promotes Vegetation Formation by ... · colonization and biofilm formation on injured heart valves was unclear. In this study, we present experimental evidence

Figure 7