bacterias en neo.pdf

Embed Size (px)

Citation preview

  • 8/10/2019 bacterias en neo.pdf

    1/13

    Hindawi Publishing CorporationClinical and Developmental ImmunologyVolume , Article ID ,pageshttp://dx.doi.org/.//

    Review ArticleRole of Pore-Forming Toxins in Neonatal Sepsis

    Andreas F.-P. Sonnen and Philipp Henneke

    Center of Chronic Immunodeciency, Medical Center, University of Freiburg, Breisacher Strae , Freiburg, Germany

    Correspondence should be addressed to Andreas F.-P. Sonnen; [email protected]

    Received February ; Accepted March

    Academic Editor: Robert Bortolussi

    Copyright A. F.-P. Sonnen and P. Henneke. Tis is an open access article distributed under the Creative CommonsAttribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work isproperly cited.

    Protein toxins are important virulence actors contributing to neonatal sepsis. Te major pathogens o neonatal sepsis, group BStreptococci,Escherichia coli, Listeria monocytogenes, and Staphylococcus aureus, secrete toxins o differentmolecular nature, whichare key or dening the disease. Amongst these toxins are pore-orming exotoxins that are expressed as soluble monomers prior toengagement o the target cell membrane with subsequent ormation o an aqueous membrane pore. Membrane pore ormation isnot only a means or immediate lysis o the targeted cell but also a general mechanism that contributes to penetration o epithelialbarriers and evasion o the immune system, thus creating survival niches or the pathogens. Pore-orming toxins, however, can alsocontribute to the induction o inammation and hence to the maniestation o sepsis. Clearly, pore-orming toxins are not the soleactors that drive sepsis progression, but they ofen act in concert with other bacterial effectors, especially in the initial stages oneonatal sepsis maniestation.

    1. Introduction

    Te birth canal, that is, the area between the ully dilateduterus andthe outside o the vagina, harbours a polymicrobialcommunity that may ull the denition o a biolm []. Nextto apathogenic species, such asLactobacillusspp., potentiallypathogenic bacteria including group B streptococci (GBS),Escherichia coli(E. coli),Listeria monocytogenes(L. monocy-togenes), andStaphylococcus aureus(S. aureus) are ound inthe vagina o up to % o women. During birth, the etus

    needs to pass rom the sterile uterus through these bacteria.Accordingly, aspiration o bacteria during birth is regardedas a major cause o neonatal sepsis in the rst three to sevendays o lie (early-onset sepsis). In line with this model,early-onset sepsis is predominantly caused by GBS,E. coli,L.monocytogenes, and S. aureus. Yet most inants successullycontrol the bacteria at the mucocutaneous suraces.

    Subsequent to aspiration, bacteria like GBS can prolierateto striking densities in the neonatal lung, as shown in

    newborn primates with neonatal GBS pneumonia (911

    colony-orming units (CFUs)/g lung tissue, []). Te antimi-crobial quality o the local pulmonary environment, orexample, the concentration o suractant, may be important

    or the metabolic activity in the bacterial community andthereore orthe expression o bacterial virulence actors suchas bacterial toxins [].

    Sepsis imposes a major threat to newborn inants. Itis estimated that sepsis causes over hal a million neona-tal deaths annually, thereby accounting or about % oall neonatal deaths worldwide []. Whereas sepsis causesapproximately .% o inant deathsin developed countries, itis responsible or up to % o neonatal deaths in developingcountries[, ]. Moreover, neonatal sepsis ofen occurs as

    meningoencephalitis, which leaves almost % o affectedpatients with lielong disabilities []. On the other hand,GBS, E. coli, and S. aureus are normal components o themucocutaneous microbiome, and it is impossible to predictthe risk to an individual baby.

    2. Bacterial Membrane-Damaging Toxins

    Te rst membrane-damaging bacterial toxin was describedby Paul Ehrlich in [], who ound that Clostridiumtetani extracts lyse erythrocytes. oday, three different mech-anisms o membrane damage by proteinaceous agents canbe delineated. First, toxins can solubilise target membranes

    http://dx.doi.org/10.1155/2013/608456http://dx.doi.org/10.1155/2013/608456
  • 8/10/2019 bacterias en neo.pdf

    2/13

    Clinical and Developmental Immunology

    Detergent-like action Pore formation Lipid hydrolysis

    F : Ways to damage a lipid membrane. Tere are various mechanisms o membrane damage by protein toxins. Amphiphilic toxinscan integrate into the membrane and essentially solubilise the lipid membrane like a detergent (structure:S. aureus-toxin, PDB ID KAM).Similarly, the membrane lipids can be hydrolysed by phospholipases also resulting in the destructiono the membrane (structure:Clostridium,perfringens -toxin, PDB ID KHO, []). By ar the largest class o membrane damaging toxins is that o the pore-orming toxins (structure:-toxin rom S. aureus, PDB ID AHL,[]). Tese toxins integrate as stable channels into the lipid bilayer, thus creating an aqueousconnection between the cytosol and the extracellular space o the target cell.

    1

    2 3

    F : Pore ormation is a dynamic process with structurally and unctionally distinct states. Distinct molecular states exist on the pathto membrane pore ormation by PFs. Te toxin is secreted by the bacterial pathogen into the extracellular medium in a water-soluble orm,usually as a monomer. Upon engagement o the membrane via binding to a receptor (step ), or example, a membrane lipid or protein, themonomers assemble to a prepore oligomer (step ). Te membrane beneath the prepore oligomer remains intact and is only punctured once

    the preporereoldsto the membrane-inserted pore oligomer(step ). Tis stepusually goesalong with considerablestructural rearrangements.

    acting essentially as amphiphilic suractants.-oxins romvarious staphylococcal species [,] and the cyclolipopep-tides romBacillus subtilisare prominent examples[] (seeFigure ). Second, toxins can act as phospholipases anddamage membranes by enzymatic hydrolysis o phospholipidester bonds. -Hemolysin rom S. aureus, or instance, isa sphingomyelin-specic phospholipase, which cleaves sph-ingomyelin to ceramide and phosphorylcholine. However,the large majority o membrane damaging proteins belong

    to the class o pore-orming proteins/toxins (PFs). PFs,which make up approximately % o all protein toxins inpathogenic bacteria [], have evolved in all domains o lie.Tey are secreted as water-soluble proteins and subsequentlyintegrate into oreign membranes.

    3. Mechanism of Membrane Pore Formationby Pore-Forming Toxins

    Common structural themes o protein/membrane associa-tion are insertion o transmembrane -helices or -sheetbarrel arrangements, anchoring by prosthetic glycolipids, or

    direct linkage to hydrophobic lipid tails, such as myristic orpalmitic acid. Most pores or channels allowing or commu-nication across biological membranes are ormed by integralmembrane proteins spanning the lipid bilayer. However, PFsorm pores by acting initially extraneously o the lipid bilayer.Tey start out as soluble molecules and then turn themselvesinto integral membrane proteins, with a membrane-spanningregion that denes the pore. Pore ormation is a dynamicprocess with structurally and unctionally distinct states (see

    Figure ). Initial binding to the membrane, or example, to amembrane lipid or protein receptor, is ollowed by homotypicoligomerisation to a prepore state on the membrane surace(Figure , arrows and ). In this state, the protomer congu-ration resembles that o the soluble monomer, and the wholeoligomer still stands prone to the membrane with an intactlipid bilayer beneath the assembled ring. InFigure (a), thisis depicted or pneumolysin romStreptococcus pneumoniae,a close homologue o listeriolysin rom L. monocytogenesand, interestingly, also o perorin secreted by cytotoxic cells [], and o the complement membrane attack complex[, ], which indicates that bacterial attack and immunedeence employ the same mechanisms. Tis prepore state

  • 8/10/2019 bacterias en neo.pdf

    3/13

    Clinical and Developmental Immunology

    (a1)

    (a2)

    (b1) (b2)

    (c1)

    (c2)

    (c3)

    F : Structures o PFs that are important or neonatal sepsis. Panel (a) shows available structures o cholesterol-dependent cytolysinsto illustrate listeriolysins mechanism o pore ormation. (a) displays the crystal structure o the soluble, monomeric orm o perringolysinrom Clostridium perfringens (lef, PDB ID PFO, []). Te cryo-electron microscopy (cryo-EM) reconstruction o the prepore (EMdatabank: ) o the listeriolysin homologue pneumolysin romStreptococcus pneumoniae displayed on the right revealed that the protomerconguration in the prepore resembles that o the soluble monomer []. Lipid membrane is coloured yellow. Molecular modeling othe protomer tted into the cryo-EM pore structure below (EM databank: ) revealed the considerable structural rearrangements thataccompany membrane pore ormation. Te-helices that reold into-sheets are coloured in red. Panel (b) shows the different structuresavailable or ClyA romE. coli, (b) the soluble state (PDDid QOY, []) monomer and (b) a protomer rom the dodecameric pore state,which is shown as side and top view on the right (PDB ID WCD, []). Pore-lining-helices are in red and the -tongue in yellow. Panel(c) shows the PFs romS. aureus. (c) shows rom top to bottom LukF (PDB ID LKF, []), LukF-PV (PDB ID PVL,[]), and LukS-PV(PDB ID R, []). (c) shows the octameric pore structure o-hemolysin (PDB ID B,[]), protomer on the lef, side and top viewson the right. (c) displays the heptameric pore structure o the AF pore (PDB ID AHL, [ ]), individual protomer, side and tops views.Te-stem that unolds into the membrane lining, extended-hairpin is shown in red.

    then undergoes drastic conormational rearrangements to beinserted as a stable pore into the membrane (see Figure ,arrow ). Tis rearrangement can even involve the reold-ing o -helices in the soluble state to -sheets in themembrane-inserted orm []. While this general mechanismo pore ormation can be proposed or nearly all PFs, thestructural changes, which the individual proteins undergo,remain largely elusive. Detailed mechanistic models on howhydrophilic proteins can suddenly change their solubility andintegrate into biological membranes are available only ora ew PFs involved in neonatal sepsis (see Figure ). Notsurprising when considering that one not only needs to be

    able to study the solution state in sufficient structural detail,orexample, via X-ray crystallography, butthe structureo themembrane state also needs to be resolved. PFs are classicallydivided into two main groups based on the structural motisthat orm the pore[]. Pores can be ormed by-helices,-PFs or by-sheets, -PFs. For certain pore-ormingproteins, it was recently proposed that lipids might play adirect role in pore ormation, but our understanding o howthis can be achieved is limited by the available structural datato date[]. Te structures, where known, o the solubleand membrane states o the PFs discussed in this review aredisplayed inFigure .However, not all membrane pores areequal. Te pore diameter, or instance, o the -hemolysin

    membrane heptamer is considerably smaller than that olisteriolysin with up to protomers (c. Figures (a) and(c)). Clearly, the size o the membrane pore has importantconsequences or the targeted cell, as a large pore diameteris not selective or what it can conduct across a membrane,potentially mediating diffusion o larger molecules, such asAP or even small proteins.

    Interestingly, clearing o toxin pores rom host mem-branes, a mechanism that is o marked importance whenconsidering the amounts o toxin that are produced dur-ing sepsis, seems also to be size dependent. Cholesterol-

    dependent cytolysins, such as listeriolysin, can induce Ca2+-

    dependent resealing o membrane pores by induction oendocytosis[] but-hemolysin romS. aureuscannot[]. Tis is counterintuitive, as van der Goot and coworkersnicely state[] that small pores are harder to repair thanlarger ones.

    4. Bacterial Pore-Forming Toxins inPathogens Causing Neonatal Sepsis

    In the major pathogens isolated rom newborn inantswith sepsis, PFs are key virulence actors. Tey initiate amultitude o events ranging rom direct necrotic cell deaths

  • 8/10/2019 bacterias en neo.pdf

    4/13

    Clinical and Developmental Immunology

    to the induction o signalling cascades, or instance, Ca2+-mediated signalling []. Prominent PFs in the context oneonatal sepsis are listeriolysin O romL. monocytogenes,-hemolysin/cytolysin rom GBS (Streptococcus agalactiae),-hemolysin and cytolysin A rom E. coli, and -hemolysin,-hemolysin, and the leukocidins rom S. aureus. It has

    long been appreciated that PFs are especially importantduring initiation o bacterial inections through induction onecrosis and apoptosis o host epithelial and endothelial cells,which promotes microbial invasion and subverts deencemechanisms. However, PFs may also contribute to sepsis byreceptor mediated or membrane damaging mechanisms inimmune cells, which respond with the ormation o inam-matory mediators, as was shown or-hemolysin/cytolysinrom GBS[] and listeriolysin O[,] and-hemolysinand Panton-Valentine leukocidins romS. aureus[,].

    5.-Hemolysin/Cytolysin from

    Group B StreptococciStreptococcus agalactiae(group B streptococci, GBS) are themajor cause o sepsis and meningitis in newborn inantswithout underlying disease in the western world. In addition,they are a signicant cause o invasive inections in pregnantwoman and immuneocompromised patients [, ]. Tepore-orming toxin-hemolysin/cytolysin is one o the main

    virulence actors o GBS. It has been implicated in thepathogenesis o early- [] and late-onset neonatal sepsis,although its role in both cases remains controversial. Rabbitsinected with wild-type GBS had signicantly higherbacterialblood counts than those inected with GBS mutants lack-ing the-hemolysin/cytolysin []; mortality also increaseddramatically. Similarly, in a neonatal rat model o meningitiswild-type GBS induced more neuronal damage in the cortexand the hippocampus than cytolysin-decient mutants [].Te clinical outcome score, assessed in this study by weightedchanges and motor activity, decreased prooundly uponpresence o the cytolysin. Te -hemolysin/cytolysin lyticprotein agent is thought to be encoded by the cylE geneo the cyl operon [], since expression o cylE induces -hemolytic activity in nonhemolytic E. coli. However, the exactmolecular nature o the protein component responsible orhemolysis and pore ormation remains obscure, as it hasevaded purication to homogeneity as o yet. Interestingly,-hemolysin/cytolysin is also necessary or the synthesis

    o an orange carotenoid pigment [], with which it alsoassociates []. Hemolytic activity o partially puried -hemolytic activity containing the carotenoid pigment couldbe inhibited by addition o the lipid dipalmitoylphosphatidyl-choline (DPPC), the major component o suractant in thelung. Accordingly, suractant deciency may explain in partthe particular susceptibility o preterm inants to GBS sepsisand meningitis [, ]. As outlined above, breaching oepithelial barriers by GBS is the rst step in sepsis patho-genesis. Accordingly, it appears to be important that -hemolysin/cytolysin mediates not only injury o lung epithe-lial [] and o lung microvascular endothelial cells [] andinvasion o brain endothelial cells [] but also injury o

    proessional phagocytes [] and neurons []. Moreover,GBS mutants lacking clyE were more readily cleared rommouse and human blood []. Interestingly, Rubens andcolleagues initially proposed that-hemolysin/cytolysin wasno longer needed or systemic disease maniestation once theepithelial barriers have been breached []. However, pro-

    and anti-inammatory activity o-hemolysin/cytolysin inmacrophages was demonstrated, indicating more prooundimmunomodulatory unctions o the cytolysin [, ].One direct or indirect molecular-hemolysin/cytolysin tar-get with important implications or mononuclear phago-cyte activation is the NLPRP inammasome. Activationo the NLRP inammasome requires GBS expressing -hemolysin/cytolysin. Tis pathway is essential in a mousemodel o GBS sepsis, where deciency in NLRP or its sig-nalling partners apoptosis-associated speck-like protein andcaspase- increases lethality and bacterial dissemination [].Yet direct evidence or binding, engagement, and activationo LRs by the-hemolysin/cytolysin is not available.

    A second pore-orming toxin o GBS is the CAMP actorthat has long been used or microbiological identicationo GBS, since it characteristically synergizes with secretedsphingomyelinase oS. aureusto lyse erythrocytes on bloodagar plates []. However, its role in neonatal sepsis is notclear, as it was not required or systemic inection in a mousemodel o GBS inection [].

    6. Listeriolysin O fromListeria monocytogenes

    Listeriamonocytogenes (L. monocytogenes) is a Gram-positivebacterium that causes early- and late-onset neonatal sep-sis and meningitis. L. monocytogenes has the capacity tobreach the intestinal barrier, thereby causing ood-bornelisteriosis, the blood-brain barrier, causing meningitis, andthe maternal-placental barrier, causing early-onset listerio-sis. Listeriolysin O (LLO), a member o the PF class ocholesterol-dependent cytolysins (CDCs), is a major viru-lence actor oL. monocytogenes with multivalent unctions[]. In the late s, Kathariou et al. and Portnoy etal. reported that L. monocytogenes mutants lacking unc-tional LLO were avirulent in mice [, ]. Furthermore,LLO mutants did not induce secretion o cytokines suchas NF-, IL-, or IFN-, when injected intravenouslyinto CBL/ mice []. Recently, a single-gene signature-

    tag-based approach was used to assess the contribution oindividual amino acids o LLOs to its virulence in mice [].

    Based on structural homology with other toxins such aspneumolysin rom Streptococcus pneumoniae [] and per-ringolysin rom Clostridium perfringens [], common pore-orming properties can be proposed [] (seeFigure (a)).LLO engages cholesterol as a native membrane receptorin dependence on the two amino acids threonine andleucine , oligomerises to a prepore complex o up to monomers, and orms a membrane pore in a concertedreolding step with each protomer contributing two beta-hairpins to the membrane-spanning -barrel, which origi-nates rom ve-helices in the soluble state[,]. Te

  • 8/10/2019 bacterias en neo.pdf

    5/13

    Clinical and Developmental Immunology

    allosteric monomer assembly and prepore reolding processwere recently shown to rely on an undecapeptide sequence(-ECGLAWEWWR-), which was originally thoughtto be soley responsible or cholesterol binding []. Teexact nature o the membrane pore remains controversial,as arciorm pores, that is, membrane pores with a seemingly

    incomplete protein ring lining the aqueous membrane hole,are ofen observed in electron microscopic and atomic orcemicroscopic imaging o various CDCs [, ]. Moreover,LLO does not lose its membrane targeting properties aferincubation with cholesterol[].

    Listeria are classical intracellular pathogens [] and LLOpore ormation was traditionally thought to only mediateescape oListeriarom the phagolysosome []. Tis conceptwas based on the nding that LLO was active only at acidicand not at neutral pH, which is ound in the maturingphagolysosome []. However, host actors also play animportant part in regulating the activity o LLO in thephagolysosome. LLO hijacks the reductive capacity o the-intereron-inducible lysosomal thiol reductase GIL tomaintain cysteine o LLO in its reduced thiol state [],thus greatly increasing bacterial escape rom the phagolyso-some. Intriguingly, CDCs were originally, that is, beorethe identication o cholesterol as a membrane receptor,termed sulydryl-activated[] or thiol-activated, oxygen-labile cytolysins [], as chemical reduction activated thetoxins towards hemolysis o red blood cells. Additionally,the cystic brosis transmembrane conductance regulator(CFR), which transports chloride not only across the apicalplasma membrane o epithelial cells in the lung but alsointo the phagolysosomes o macrophages, potentiates LLOoligomerisation on the phagosomal membrane and its lyticactivity and phagolysosomal escape oL. monocytogenes [].However, the role o LLO extends beyond mediating phago-somal escape. LLO reduces ormation o reactive oxygenspecies (ROS) by inhibiting the NADPH oxidase NOXin RAW . macrophages []. Tis activity seems torely on pore ormation in the phagosomal membrane andprevents degradation o bacteria inside the phagosome. Poreormation at the plasma membrane o target cells inducesthe dynamin-/F-actin-dependent but clathrin-independentuptake o L. monocytogenes into HepG cells []. Tisnding questions the traditional model o LLO pore-ormingactivity being strictly dependent on the low phagosomal pH[], whereby premature lysis o target cells by (secreted)LLO is prevented[]. Residual lytic activity and structural

    integrity o LLO at neutral pH [,,] are in line withLLO-mediated calcium inux into epithelial Hep- [] andHEK cells [] along with concomitant L. monocytogenesuptake. Indeed LLO seems to orm pores at neutral pHin the plasma membrane, which do not result in lysis othe target cell but rather in uptake o the pathogen [].As pneumolysin rom S. pneumoniae can replace LLO inthe uptake o L. monocytogenes into HepG cells, CDCsrom other bacterial pathogens may similarly induce cellularuptake. Te contribution o LR signalling in responseto CDC has been subject o several studies. As examples,the LLO homologues anthrolysin (Bacillus anthracis) andpneumolysin can signal via LR [, ]. On the other

    hand, LLO induces an inammatory cellular response in aLR-independent ashion []. Moreover, LLO activity atthe plasma membrane induces clustering o lipid rafs[],suppressiono antigen-induced -cell activation [], inam-masome activation, and histone H dephosphorylation [],all o which might contribute to sepsis progression either at

    the stage o heightened inammation or at later stages oimmune suppression.

    7.-Hemolysin,-Hemolysin, and LeukocidinsfromStaphylococcus aureus

    Staphylococcus aureus (S. aureus) is well recognised as asignicant cause o neonatal sepsis []. Around ten bacteriaare sufficient to colonise the umbilical cord. Afer birth, S.aureuscan colonise the upper respiratory tract in up to %o inants []. S. aureus produces a number o PFs withdistinct specicity or target cell membranes. Although mostclinical isolates produce the PFs -hemolysin, bicomponent

    -hemolysins, and bicomponent leukocidins, none o these

    toxins was ound to be a necessary and sufficient virulencedeterminant o neonatal sepsis. In contrast, other actorssuchas the antigenic, peptidoglycan-associated protein A [],superantigens [], and sphingomyelinase C (-hemolysin,[]) contribute to host invasion, subversion o the immunesystem, and sepsis maniestation. However, there is evidencethat pore ormation by -hemolysin (-toxin, AF) con-tributes to the pathogenesis o sepsis []. As an example,erythrocyte lysis by AF could be directly imaged [].Downregulation o AF expression in vivo clearly reduces

    virulence oS. aureus [, ]. In a model using CBL/Jmice, AF activates the NLRP inammasome, thereby pro-moting necrotising pneumonia []. Moreover, monoclonalantibodies to AF are protective against staphylococcal pneu-monia[]. Indeed, a nonhemolytic variant o AF was usedto vaccinate rabbits, and antisera could be used to passivelyimmunize mice against an otherwise lethal challenge withwild-type S. aureus []. In a mouse model o mastitis,coagulase and AF proved to be the primary virulence deter-minants[]. Heat-inactivatedS. aureusand an AF mutantgreatly reduced the bacterial burdens in a mouse brainabscess model and attenuated the expression o inammatorymediators [].Inan in vivo model o corneal virulence, AFalso proved to be a decisive virulence actor [].

    AF is one o the best-studied PFs to date. It was therst toxin o which the membrane structure was solved by

    X-ray crystallography (seeFigure (c),[]). AF consistso amino acids and oligomerises on the plasma mem-brane o target cells to a heptamer (potentially a hexamer)prior to membrane insertion and pore ormation. It wasshown to have an important role in bacterial pathogenesis,especially by its ability to induce necrotic cell death [ ],by which it can cause vascular leakage when perused intothe lung[]. Recently the metalloprotease ADAM wasidentied as the membrane receptor o AF []. At lowtoxin concentrations, ADAM is required to mediate thecytotoxic effects o AF. Interestingly, binding o AF toADAM resultedin the upregulation o ADAM in alveolarepithelial cells and concomitant cleavage o E-cadherin. Tis

  • 8/10/2019 bacterias en neo.pdf

    6/13

    Clinical and Developmental Immunology

    leads to epithelial barrier disruption thereby aggravatingstaphylococcal pneumonia in mice []. Moreover, activa-tion o the NLRP-inammasome by AF might contributeto later stages o sepsis, although the molecular mechanismunderlying inammasome activation remains elusive at thisstage []. In this respect, it is interesting to note that, whereas

    direct LR activation has not been demonstrated or AF,NOD-dependent sensing oS. aureus was dependent onAF [].

    Next to AF,S. aureusexpresses the bicomponent cyto-toxins leukocidins (Luk) and the-hemolysins (Hlg). Bicom-ponent implies that class S toxins (LukS-PV, LukE, HlgA,HlgC, and LukS) have to associate with class F toxins (LukF,LukD, LukF-PV, and HlgB) in a : stoichiometric ratio toorm a unctional oligomer beore insertion into the mem-brane (see Figure (c)). Pathogenic S. aureus can produceseveral different bicomponent toxin pores, among the mostprominent are LukE/LukD [], Panton-Valentine leuko-cidin LukS-PV/LukF-PV, -hemolysins LukF/HlgA [],HlgA/HlgB, HlgB/HlgC, and the M/F-PV-like leukocidins,all o which might be expressed at different stages duringsepsis. Te Panton-Valentine leukocidin (PVL), which wasrst isolated rom uruncles in [], is probably themostwidely studied member. In a mouse model, secreted PVL pro-motes tissue invasion and causes necrotizing pneumonia, viamechanisms including upregulation o protein A and otheradhesins[]. PVL is an important actor in the early stageso skin inection, as shown in a rabbit skin inection model[]. Association opvl and spa (protein A) genes seemsto be an important virulence determinant in methicillin-resistant S. aureus (MRSA). Moreover, PVL was reported todirectly bind to LR and induce inammation in the mouselung []. LukE/LukD promotes systemic bacterial growth invivo by specically targeting neutrophils []. Interestingly,

    via engagement o their native receptor CCR (CC-moti-chemokine-receptor type ), LukE/LukD toxin pores clearantigen-presenting cells (macrophages and dendritic cells)and S. aureus-specic CCR-positive T/T cells [],thus greatly contributing to the spread oS. aureus in the host.

    AF, Hlg, and the leukocidins belong to the class o-pore-orming toxins []. Despite their moderate sequenceidentity (around % or the pairwise alignment with AF),they share a common structural old (see Figure (c)). Testructures o the soluble, monomeric LukF (Figure (c),top, []), LukF-PV (Figure (c), middle, []), LukS-PV(Figure (c), bottom, []), and the octameric, membrane

    complex o Hlg (LukF/HlgA, Figure (c), []) are available.A common molecular mechanism could be proposed inwhich a prestem, triple stranded -sheet in the solublemonomer, reolds to a double, stranded, membrane-inserted-sheet (Figures (c) and(c), lef), resulting in a hexadeca-or tetradecastranded-barrel that penetrates the membrane.

    Te target membrane specicities oS. aureusPFs hinttowards their role in neonatal sepsis. Whereas AF has abroad specicity and might thus be involved at the initialstages o sepsis maniestation, where the lung epitheliumneeds to be breached, bicomponent leukocidins and Hlgsmainly attack polymorphonuclear neutrophils, macrophagesand lymphocytes [,]. Bearing in mind that PVL does

    not attack lymphocytes and Hlg can be hemolytic, at leastin vitro, their restricted but highly directed mode o attackpredisposes the bicomponent leukocidins to be importantactors or the subversion o the immune system once theinitial barrier has been breached.

    8.-Hemolysin and Cytolysin A fromEscherichia coli

    Pathogenic Escherichia coli (E. coli) cause around % oinvasive neonatal sepsis [], and antibiotic resistance isan emerging threat in this context []. Generally, E. colican persist in the intestine as a normal constituent o theintestinal microbiota. However, extraintestinal pathogenicE.coli(ExPECs) are the most common gram-negative bacterialspecies isolated rom neonates with bacterial inections, andneonatal mortality rom gram-negative sepsis remains high[]. Urinary tract inections o pregnant women can leadto aspiration o ExPECs during partition with subsequentuncontrolledgrowth in the lung o the newborn andpotentialprogression to a systemic inection. Pathogenic E. coliofensecrete the pore-orming toxin-hemolysin (HlyA, CylA), a kDa member o the RX class o toxins [], which isusually associated with strains causing uropathogenic inec-tions []. Deletiono HlyA in E. coli greatly reduced mortal-ity and cytokine production as compared to the isogenic wildtype bacteria in an intravenous inection model[]. HlyAurthermore induced hemorrhagic bleeding o bladder tissueand exoliation o urothelium when pathogenic bacteria wereadministered into the urethra[]. HlyA is encoded by atleast % o all ExPEC clinical isolates []. Around %o meningitis- and sepsis-associatedE. colibelong to the Kserotype [, ]. Te E. coli K strain RS expressed

    HlyA in a zebrash model o systemic inection [] andthe hlyA gene was present in more than % oE. coliromthe genital tracts o pregnant women []. As a member othe repeats in toxin (RX) amily o hemolysins, the hlyAgene is part o the chromosomal hlyCABD operon, whichalso encodes a type ABC transporter or secretion o thePF. Te amino acid toxin repeats,which are locatedin the C-terminal portion o the protein,are composed o the sequenceGGXGCDXUX (with U being a large hydrophobic residue).

    Tese repeats are responsible or Ca2+ binding, which is aprerequisite or membrane association and pore ormationby the N-terminal, hydrophobic, and acylated domain oHlyA []. Interestingly, other pore-orming proteins, such

    as the human cytotoxic lymphocyte encoded perorin, alsorequire binding o Ca2+ or membrane association and poreormation []. HlyA oligomerises on the plasma membraneo target cells, where it accumulates in cholesterol-richmicrodomains [, ]. However, rather than cholesterolbeing a direct, lipid membrane receptor as in the case othe CDCs listeriolysin or pneumolysin, cholesterol seems tocontribute to the physicochemical environment necessaryor HlyA membrane engagement. Te exact nature o thepore ormation mechanism is under considerable debate. Itseems now accepted that HlyA orms membrane pores as anoligomer, at least in articial membrane mimics[],albeit possibly heterogeneous in size []. In this respect,

  • 8/10/2019 bacterias en neo.pdf

    7/13

    Clinical and Developmental Immunology

    it is intriguing that the PX receptor and pannexin wereound to mediate HlyA-dependent pore ormation [,].Sublytic doses o HlyA initiate degradation o paxillin andproteolytic cascades inside epithelial cells and macrophages,thus attenuating the inammatory host response and pro-moting epithelial exoliation []. Similarly, HlyA inhibits

    epithelial cytokine production potentially promoting epithe-lial invasion oE. coli[].Another PF oE. coli is ClyA (also termed hemolysin-

    E or SheA), which is expressed by various pathogenicand nonpathogenic E. coli including K strains, that arealso ound in clinical isolates o neonatal meningitis [],bacteremia [, ], and neonatal sepsis []. However,three neonatal meningitis K strains isolated by Ludwigand colleagues harboured deletion mutations at the clyAgene locus []. Nevertheless, a synergistic enhancemento extraintestinal inection was recently reported betweennonpathogenicE. coliK and pathogenic ExPEC strains ina mouse model o septicaemia [], which hints towardsa potential involvement o ClyA. ClyA is a kDa proteinbelonging to the class o-PFs. Te mechanism o poreormation is well understood, as crystal structures or thesoluble [] and membrane state [] are available (seeFigure (b)). Upon association with the membrane, insertiono the so-called -tongue induces a series o substantialstructural rearrangements in the now membrane-anchoredmonomer, resulting in a perpendicularposition to membranewith the amphiphatic helix a now lying along its surace.Afer oligomerisation to a dodecamer, helices a becomeinserted into the membrane orming a A hollow cylinderwith a A aperture protruding through the membrane.

    9. Synopsis and Medical OutlookNeonatal sepsis is a syndrome caused by systemic inam-mation and dened by clinical criteria such as tachycardia,respiratory distress, temperature instability, and unusuallyhigh amount o immature immune cells in the blood.Pathogenic bacteria that are aspirated by the etus in thebirth canal during parturition can cause neonatal sepsis itheir inection is not controlled locally by the innate deencemechanisms o the respiratory and alveolar epithelia. oxins,either proteinaceous or o other molecular nature, are impor-tantactors contributing to neonatal sepsis. Whileendotoxinssuch as the lipopolysaccharide (LPS) rom Gram-negativebacteria contribute to the sepsis phenotype by activating

    monocytes and macrophages via oll-like receptor binding,pore-orming proteinaceous exotoxins act by permeabilisingtarget membranes o host cells. Whereas the membranetargeting effects o PFs, that is, engagement o membrane,oligomerisation, and pore ormation, are well dened, theirsecondary downstream effects are maniold, owing to the actthat dened ionic and molecular gradients across cellularmembranes modulate a diverse set o signalling cascades.Unregulated cell death and its consequences are importantin neonatal sepsis []. Te PFs described above can causedirect necrotic cell death, which in the case oE. coli, GBS,and S. aureus contributes to overcoming the epithelial andendothelial barriers in the lung. LLO oL. monocytogenesis

    critical or cell invasion and cell-to-cell spread o this intra-cellular pathogen and thus also contributes to breaching theepithelial barriers o the host. Necrotic cell death can releaseproinammatory cytokines rom leukocytes thus contribut-ing to the inammatory storm during sepsis. Interestingly,several PFs discussed above can induce the inammasome:

    AF[], -hemolysin/cytolysin [], LLO [,], HlyA[], and leukocidins [, ]. Inammasome activationultimately may contribute to hyperinammation in newborninants similar to what has been shown in GBS and E. colisepsisin mice [, ]. PFs canalso elicitand alter apoptosiso host immune cells, that is, AF via caspase- [,],leukocidins via activation o caspases and [], LLO

    via release o cytochrome C rom mitochondria [], -hemolysin/cytolysin independently o caspase activation [,],HlyA[],and ClyA [], thus potentially contributingto theapparent immunodeciencyo the patient that is char-acteristic during stages o sepsis []. Despite the ormationo hydrophilic channels by all PFs, the ways in which theyinduce the inammasome are varied, hinting towards theact that unctions o PFs at cellular membranes are moresubtle than might be expected rom their general modeo action. In this respect, effects o sublytic concentrationso PFs have recently been explored. For instance, sublyticdoses o LLO induce mitochondrial network disorganisationwith transient alteration o the metabolic state o the targetcell, thus weakening the cell or L. monocytogenes entrywithout destroying it []. Moreover, targeting o organs byPFs might also contribute to the septic phenotype. GBS-hemolysin/cytolysin, or example, had marked effects oncardiomyocyte contractility and viability [].

    Due to their role in neonatal sepsis andbacterial inectionin general, PFs present attractive therapeutic targets. Incases where membrane receptors have been dened, specicinhibitors, akin to viral entry inhibitors, might preventmembrane binding and pore ormation. Monoclonal anti-bodies against PFs that prevent membrane binding and/orreolding to the pore state could be a way o neutralising thetoxin, at least in the blood stream. Moreover, vaccines basedon PFs may be used or immunizing women and therebyprotecting newborn inants through placental transer ospecic immunoglobulins, given the act that pneumolysinromS. pneumoniaeis considered a vaccine candidate [].Indeed, novel vaccines based on AF are currently developed[]. As PFs elicit specic cellular responses, it might,however, also be promising to designing therapeutics based

    on the pathways that the toxins induce in the target cell, ashas been proposed or pMAPK and-hemolysin/cytolysinrom GBS []. In any case, it is exactly these cellularresponses towards PFs that need to be investigated in thecontext o neonatal sepsisin theuture to improve therapeuticstrategies.

    Acknowledgment

    Tis work was supported by a Grant rom the GermanBundesministerium ur Bildung und Forschung (BMBF EO ).

  • 8/10/2019 bacterias en neo.pdf

    8/13

    Clinical and Developmental Immunology

    References

    [] H. Verstraelen and A. Swidsinski, Te biolm in bacterial vagi-nosis: implications or epidemiology, diagnosis and treatment,Current Opinion in Infectious Diseases, vol. , no. , pp. ,.

    [] C. E. Rubens, H. V. Raff, J. C. Jackson, E. Y. Chi, J. . Bielitzki,

    and S. L. Hillier, Pathophysiology and histopathology o groupB streptococcal sepsis in Macaca nemestrina primates inducedafer intraamniotic inoculation: evidence or bacterial cellularinvasion,Te Journal of Infectious Diseases, vol. , no. , pp., .

    [] E. Herting, X. Gan, P. Rauprich, C. Jarstrand, and B.Robertson, Combined treatment with suractant and specicimmunoglobulin reduces bacterial prolieration in experimen-tal neonatal group B streptococcal pneumonia, AmericanJournal of Respiratory and Critical Care Medicine, vol. , no., pp. , .

    [] R. E. Black, S. Cousens, H. L. Johnson et al., Global, regional,and national causes o child mortality in : a systematicanalysis,Te Lancet, vol. , no. , pp. , .

    [] K. D. Kochanek, J. Xu, S. L. Murphy, A. M. Minin o, and H.-C. Kung, Deaths: nal data or ,National Vital StatisticsReports, vol. , no. , pp. , .

    [] B. J. Stoll, Te global impact o neonatal inection,Clinics inPerinatology, vol. , no. , pp. , .

    [] R. Libster, K. M. Edwards,F.Levent et al., Long-term outcomeso group B streptococcal meningitis,Pediatrics, vol. , no. ,pp. , .

    [] P. Ehrlich, Diskussion waehrend der Gesellschaf der ChariteAerzte,Klinische Medizin, vol. , article , .

    [] A. Pokorny, . H. Birkbeck, and P. F. F. Almeida, Mechanismand kinetics o-lysin interaction with phospholipid vesicles,Biochemistry, vol. , no. , pp. , .

    [] J. H. Freer and J. P. Arbuthnott, oxins o Staphylococcusaureus,Pharmacology and Terapeutics, vol. , no. , pp. , .

    [] A. W. Bernheimer and B. Rudy, Interactions between mem-branes and cytolytic peptides, Biochimica et Biophysica Acta,vol. , no. , pp. , .

    [] J. E. Alou, Bacterial protein toxins. An overview,Methods inMolecular Biology, vol. , pp. , .

    [] R. H. P. Law, N. Lukoyanova, I. Voskoboinik et al., Testructural basis or membrane binding and pore ormation bylymphocyte perorin,Nature, vol. , no. , pp. ,.

    [] M. A. Hadders, D. X. Beringer, and P. Gros, Structure o C-MACPF reveals mechanism o membrane attack in comple-

    ment immune deense, Science, vol., no., pp. ,.

    [] C. J. Rosado, A. M. Buckle, R. H. P. Law et al., A common oldmediates vertebrate deense and bacterial attack,Science, vol., no. , pp. , .

    [] S. J. illey, E. V. Orlova, R. J. C. Gilbert, P. W. Andrew, and H. R.Saibil, Structural basis o pore ormation by the bacterial toxinpneumolysin,Cell, vol. , no. , pp. , .

    [] E. Gouaux, Channel-orming toxins: tales o transormation,Current Opinion in Structural Biology, vol. , no. , pp. ,.

    [] G. Anderluh and J. H. Lakey, Disparate proteins use similararchitectures to damage membranes, rends in BiochemicalSciences, vol. , no. , pp. , .

    [] M. W. Parker and S. C. Feil, Pore-orming protein toxins:romstructure to unction, Progress in Biophysics and MolecularBiology, vol. , no. , pp. , .

    [] R. J. C. Gilbert, Pore-orming toxins,Cellular and MolecularLife Sciences, vol. , no. , pp. , .

    [] G. Anderluh, M. Dalla Serra, G. Viero, G. Guella, P. Macek,

    and G. Menestrina, Pore ormation by equinatoxin II, aeukaryotic protein toxin, occurs by induction o nonlamellarlipid structures,Te Journal of Biological Chemistry, vol. ,no. , pp. , .

    [] R. J. C. Gilbert, Inactivation and activity o cholesterol-dependent cytolysins: what structural studies tell us,Structure,vol. , no. , pp. , .

    [] V. Idone, C. am, J. W. Goss, D. oomre, M. Pypaert, and N. W.Andrews, Repair o injured plasma membrane by rapid Ca2+

    dependent endocytosis,Journal of Cell Biology, vol. , no. ,pp. , .

    [] M. Corrotte, M. C. Fernandes, C. am, and N. W. Andrews,oxin pores endocytosed during plasma membrane repairtraffic into the lumen o MVBs or degradation,raffic, vol. ,

    no. , pp. , .

    [] C. am, V. Idone, C. Devlin et al., Exocytosis o acid sph-ingomyelinase by wounded cells promotes endocytosis andplasma membrane repair,Journal of Cell Biology, vol. , no., pp. , .

    [] M. Husmann, K. Dersch, W. Bobkiewicz, E. Beckmann, G.Veerachato, and S. Bhakdi, Differential role o p mitogenactivated protein kinase or cellular recovery rom attack bypore-orming S. aureus-toxin or streptolysin O,Biochemicaland Biophysical Research Communications, vol. , no. , pp., .

    [] M. Bischoerger, I. Iacovache, and F. G. van der Goot,Pathogenic pore-orming proteins: unction and host

    response, Cell Host & Microbe, vol. , no. , pp. , .[] A. Costa, R. Gupta, G. Signorino et al., Activation o the

    NLRP inammasome by group B streptococci, Journal ofImmunology, vol. , no. , pp. , .

    [] S. Mariathasan, D. S. Weiss, K. Newton et al., Cryopyrinactivates the inammasome in response to toxins and AP,Nature, vol. , no. , pp. , .

    [] M. A. Hamon and P. Cossart, K+ efflux is required or histoneH dephosphorylation byListeria monocytogeneslisteriolysin oand other pore-orming toxins, Infection and Immunity, vol. ,no. , pp. , .

    [] R. R. Craven, X. Gao,I. C. Allen et al., Staphylococcus aureus-hemolysin activates the NLRP-inammasome in human andmouse monocytic cells,PLoS ONE, vol. , no. , Article IDe, .

    [] D. Holzinger, L. Gieldon, V. Mysore et al., Staphylococcusaureus Panton-Valentine leukocidin induces an inammatoryresponse in human phagocytes via the NLRP inammasome,Journal of Leukocyte Biology, vol. , no. , pp. , .

    [] K. Fluegge, S. Supper, A. Siedler, and R. Berner, Serotypedistribution o invasive group B streptococcalisolates in inants:results rom a nationwide active laboratory surveillance studyover years in Germany, Clinical Infectious Diseases, vol. ,no. , pp. , .

    [] P. Henneke and R. Berner, Interaction o neonatal phagocyteswith group B streptococcus: recognition and response, Infec-tion and Immunity, vol. , no. , pp. , .

  • 8/10/2019 bacterias en neo.pdf

    9/13

    Clinical and Developmental Immunology

    [] M. E. Hensler, G. Y. Liu, S. Sobczak, K. Benirschke, V. Nizet,and G. P. Heldt, Virulence role o group B Streptococcus -hemolysin/cytolysin in a neonatal rabbit model o early-onsetpulmonary inection, Te Journal of Infectious Diseases, vol. ,no. , pp. , .

    [] A. Reiss, J. S. Braun, K. Jager et al., Bacterial pore-ormingcytolysins induce neuronal damage in a rat model o neonatal

    meningitis, Te Journal of Infectious Diseases, vol. ,no., pp., .

    [] C. A. Pritzlaff, J. C. W. Chang, S. P. Kuo, G. S. amura, C. E.Rubens, and V. Nizet, Genetic basis or the -haemolytic/cytolytic activity o group B streptococcus, Molecular Micro-biology, vol. , no. , pp. , .

    [] J. W. apsall, Pigmentproduction by Lanceeld-group-B strep-tococci (Streptococcus agalactiae),Journal of Medical Microbi-ology, vol. , no. , pp. , .

    [] G. Y. Liu, K. S. Doran, . Lawrence et al., Sword andshield: linked group B streptococcal -hemolysin/cytolysinand carotenoid pigment unction to subvert host phagocytedeense,Proceedings of the National Academy of Sciences of theUnited States of America, vol. , no. , pp. , .

    [] V. Nizet, R. L. Gibson, E. Y. Chi, P. E. Framson, M. Hulse, and C.E. Rubens, Group B streptococcal beta-hemolysin expressionis associated with injury o lung epithelial cells, Infection andImmunity, vol. , no. , pp. , .

    [] V. Nizet, R. L. Gibson, and C. E. Rubens, Te role o group Bstreptococci-hemolysin expression in newborn lung injury,Advances in Experimental Medicine and Biology, vol. , pp., .

    [] R. L. Gibson, V. Nizet, and C. E. Rubens, Group B streptococcal-hemolysinpromotes injury o lungmicrovascular endothelialcells, Pediatric Research, vol. , no. , part , pp. , .

    [] V. Nizet, K. S. Kim, M. Stins et al., Invasion o brain microvas-cular endothelial cells by group B streptococci, Infection and

    Immunity, vol. , no. , pp. , .[] C. E. Rubens, M. R. Wessels, J. M. Kuypers, D. L. Kasper, andJ. N. Weiser, Molecular analysis o two group B streptococcalvirulence actors, Seminars in Perinatology, vol. , no. ,supplement , pp. , .

    [] M. Bebien, M. E. Hensler, S. Davanture et al., Tepore-ormingtoxin hemolysin/cytolysin triggers p MAPK-dependent IL- production in macrophages and inhibits innate immunity,PLoS Pathogens, vol. , no. , Article ID , .

    [] A. Ring, C. Depnering, J. Pohl, V. Nizet, J. L. Shenep, andW. Stremmel, Synergistic action o nitric oxide release rommurine macrophages caused by group B streptococcal cell walland-hemolysin/cytolysin, Te Journal of Infectious Diseases,vol. , no. , pp. , .

    [] S. Lang and M. Palmer, Characterization o StreptococcusagalactiaeCAMP actor as a pore-orming toxin, Te Journalof Biological Chemistry, vol. , no. , pp. , .

    [] S. Lang, J. Xue, Z. Guo, and M. Palmer, Streptococcus agalac-tiae CAMP actor binds to GPI-anchored proteins, MedicalMicrobiology and Immunology, vol. , no. , pp. , .

    [] R. Christie, N. E. Atkins, and E. Munch-Petersen, A noteon a lytic phenomenon shown by group B streptococci,TeAustralian Journal of Experimental Biology and Medical Science,vol. , pp. , .

    [] M. E. Hensler, D. Quach, C. J. Hsieh, K. S. Doran, and V. Nizet,CAMP actor is not essential or systemic virulence o Group BStreptococcus,Microbial Pathogenesis, vol. , no. , pp. ,.

    [] M. A. Hamon, D. Ribet, F. Stavru, and P. Cossart, ListeriolysinO: the Swiss army knie o Listeria,rends in Microbiology, vol., no. , pp. , .

    [] D. A. Portnoy, P. S. Jacks, and D. J. Hinrichs, Role o hemolysinor the intracellular growth oListeria monocytogenes,Journalof Experimental Medicine, vol. , no. , pp. , .

    [] S. Kathariou, P. Metz, H. Ho, and W. Goebel, n-inducedmutations in the hemolysin determinant affecting virulence oListeria monocytogenes,Journal of Bacteriology, vol. , no. ,pp. , .

    [] R. M. Poston and R. J. Kurlander, Cytokine expression in vivoduring murine listeriosis: inection with live, virulent bacteriais required or monokine and lymphokine messenger RNAaccumulation in the spleen,Journal of Immunology, vol. ,no. , pp. , .

    [] J. A. Melton-Witt, S. L. McKay, and D. A. Portnoy, Develop-ment o a single-gene, signature-tag-based approach in com-bination with alanine mutagenesis to identiy listeriolysin Oresidues critical or the in vivo survival oListeria monocyto-genes, Infection and Immunity, vol. , no. , pp. ,

    .[] J. Rossjohn, R. J. C. Gilbert, D. Crane et al., Te molecular

    mechanism o pneumolysin, a virulence actor rom Streptococ-cus pneumoniae,Journal of Molecular Biology, vol. , no. ,pp. , .

    [] J. Rossjohn, S. C. Feil, W. J. McKinstry, R. K. weten, and M.W. Parker, Structure o a cholesterol-binding, thiol-activatedcytolysin and a model o its membrane orm,Cell, vol. , no., pp. , .

    [] D. W. Schuerch, E. M. Wilson-Kubalek, and R. K. weten,Molecular basis o listeriolysin O pH dependence, Proceedingsof the National Academy of Sciences of the United States ofAmerica, vol. , no. , pp. , .

    [] R. J. C. Gilbert, J. L. Jimenez, S. Chen et al., wo structural

    transitions in membrane pore ormation by pneumolysin, thepore-orming toxin o Streptococcus pneumoniae,Cell, vol. ,no. , pp. , .

    [] O. Shatursky, A. P. Heuck, L. A. Shepard et al., Te mechanismo membrane insertion or a cholesterol-dependent cytolysin: anovel paradigm or pore-orming toxins, Cell, vol., no. ,pp., .

    [] L. A. Shepard, A. P. Heuck, B. D. Hamman et al., Identicationo a membrane-spanning domain o the thiol-activated pore-orming toxinClostridium perfringensperringolysin O: an-helical to -sheet transition identied by uorescence spec-troscopy,Biochemistry, vol. , no. , pp. , .

    [] K. J. Dowd and R. K. weten, Te cholesterol-dependent

    cytolysin signature moti: a critical element in the allostericpathway that couples membrane binding to pore assembly,PLoS Pathogens, vol. , no. , Article ID , .

    [] S. Bhakdi, J. ranum-Jensen, and A. Sziegoleit, Mechanism omembrane damage by streptolysin-O,Infection and Immunity,vol. , no. , pp. , .

    [] M. Palmer, I. Vulicevic, P. Saweljew, A. Valeva, M. Kehoe,and S. Bhakdi, Streptolysin O: a proposed model o allostericinteraction between a pore-orming protein and its target lipidbilayer,Biochemistry, vol. , no. , pp. , .

    [] D.M. Czajkowsky, E. M. Hotze, Z. Shao,and R. K. weten, Ver-tical collapse o a cytolysin prepore moves its transmembrane-hairpins to the membrane, EMBO Journal, vol. , no. , pp., .

  • 8/10/2019 bacterias en neo.pdf

    10/13

    Clinical and Developmental Immunology

    [] . Jacobs, A. Darji, N. Frahm et al., Listeriolysin O: cholesterolinhibits cytolysis but not binding to cellular membranes,Molecular Microbiology, vol. , no. , pp. , .

    [] L. G. ilney and D. A. Portnoy, Actin laments andthe growth,movement, and spread o the intracellular bacterial parasite,Listeria monocytogenes,Journal of Cell Biology, vol. , no. ,part , pp. , .

    [] M. M. Gedde, D. E. Higgins, L. G. ilney, and D. A. Portnoy,Role o listeriolysin O in cell-to-cell spread oListeria mono-cytogenes,Infection and Immunity, vol. , no. , pp. ,.

    [] R. Singh, A. Jamieson, and P. Cresswell, GIL is a critical hostactor or Listeria monocytogenes inection, Nature, vol., no., pp. , .

    [] H. P. Bernheimerand J.G. iraby, Inhibitiono phageinectionby pneumococcus capsule,Virology, vol. , no. , pp. ,.

    [] C. J. Smyth and J. A. Duncan, Tiol-activated cytolysins, inBacterial oxIns and Cell Membranes, J. Jeljaszewicz and .Wadstrom, Eds., pp. , Academic Press, New York, NY,

    USA, .[] A. L. Radtke, K. L. Anderson, M. J. Davis, M. J. DiMagno,J. A. Swanson, and M. X. ORiordan, Listeria monocytogenesexploits cystic brosis transmembrane conductance regulator(CFR) to escape the phagosome, Proceedings of the NationalAcademy of Sciences of the United States of America, vol. , no., pp. , .

    [] G. Y. Lam, R. Fattouh, A. M. Muise, S. Grinstein, D. E. Higgins,and J. H. Brumell, Listeriolysin O suppresses phospholipaseC-mediated activation o the microbicidal NADPH oxidase topromote Listeria monocytogenes inection, Cell Host & Microbe,vol. , no. , pp. , .

    [] S. Vadia, E. Arnett, A. C. Haghighat, E. M. Wilson-Kubalek, R.K. weten, and S. Seveau, Te pore-orming toxin listeriolysin

    O mediates a novel entry pathway o L. monocytogenes intohuman hepatocytes,PLoS Pathogens, vol. , no. , Article IDe, .

    [] I. J. Glomski, M. M. Gedde, A. W. sang, J. A. Swanson, andD. A. Portnoy, Te Listeria monocytogenes hemolysin has anacidic pH optimum to compartmentalize activity and preventdamage to inected host cells, Journal of Cell Biology, vol. ,no. , pp. , .

    [] A. Bavdek, N. O. Gekara, D. Priselac et al., Sterol and pHinterdependence in the binding, oligomerization, and poreormation o listeriolysin O,Biochemistry, vol. , no. , pp., .

    [] A. Bavdek, R. Kostanjsek, V. Antonini et al., pH dependence olisteriolysin O aggregation and pore-orming ability,Te FEBS

    Journal, vol. , no. , pp. , .[] S. Dramsi and P. Cossart, Listeriolysin O-mediated calcium

    inux potentiates entry o Listeria monocytogenes into thehuman Hep- epithelial cell line,Infection and Immunity, vol., no. , pp. , .

    [] H. Repp, Z. Pamukci, A. Koschinski et al., Listeriolysin oListeria monocytogenesorms Ca2+-permeable pores leading tointracellular Ca2+ oscillations, Cellular Microbiology, vol. ,no., pp. , .

    [] R. Malley, P. Henneke, S. C. Morse et al., Recognition opneumolysin by oll-like receptor coners resistance topneumococcal inection,Proceedings of the National Academyof Sciences of the United States of America, vol. , no. , pp., .

    [] M. P. Jin, V. H. Ng, S. Maeda, R. F. Rest, and M. Karin,Anthrolysin O and other gram-positive cytolysins are toll-likereceptor agonists,Journal of Experimental Medicine, vol. ,no. , pp. , .

    [] N. O. Gekara, N. Dietrich, M. Lyszkiewicz, S. Lienenklaus, andS. Weiss, Signals triggered by a bacterial pore-orming toxin

    contribute to toll-like receptor redundancy in gram-positivebacterial recognition, Te Journal of Infectious Diseases, vol., no. , pp. , .

    [] N. O. Gekara, . Jacobs, . Chakraborty, and S. Weiss, Techolesterol-dependent cytolysin listeriolysin O aggregates rafsvia oligomerization, Cellular Microbiology, vol. , no. , pp., .

    [] N. O. Gekara, N. Zietara, R. Geffers, and S. Weiss, Liste-ria monocytogenes induces cell receptor unresponsivenessthrough pore-orming toxin listeriolysin O, Te Journal ofInfectious Diseases, vol. , no. , pp. , .

    [] G. H. McCracken Jr. and H. R. Shineeld, Changes in the pat-tern o neonatal septicemia and meningitis,American Journalof Diseases of Children, vol. , no. , pp. , .

    [] L. M. Harrison, J. A. Morris, D. R. elord, S. M. Brown, and K.Jones, Te nasopharyngeal bacterial ora in inancy: effects oage, gender, season, viral upper respiratory tract inection andsleeping position,FEMS Immunology and Medical Microbiol-ogy, vol. , no. -, pp. , .

    [] A. J. Brosnahan and P. M. Schlievert, Gram-positive bac-terial superantigen outside-in signaling causes toxic shocksyndrome,Te FEBS Journal, vol. , no. , pp. ,.

    [] G. Arad, R. Levy, I. Nasie et al., Binding o superantigen toxinsinto the CD homodimer interaceis essential or inductionocytokine genes that mediate lethal shock,PLoS Biology, vol. ,no. , Article ID e, .

    [] Y. Ramu, Y. Xu, and Z. Lu, Inhibition o CFR Cl- channelunction caused by enzymatic hydrolysis o sphingomyelin,Proceedings of the National Academy of Sciences of the UnitedStates of America, vol. , no. , pp. , .

    [] S. Bhakdi, F. Grimminger, N. Suttorp, D. Walmrath, and R.Seeger, Proteinaceous bacterial toxins and pathogenesis osepsis syndrome and septic shock: the unknown connection,Medical Microbiology and Immunology, vol. , no. , pp. , .

    [] R. Plummer, J. Bodkin, . W. Yau et al., Modelling Staphy-lococcus aureus-induced septicemia using NMR, MagneticResonance in Medicine, vol. , no. , pp. , .

    [] Y. Ji, A. Marra, M. Rosenberg, and G. Woodnutt, Regulatedantisense RNA eliminates alpha-toxin virulence in Staphylococ-cus aureus inection,Journal of Bacteriology, vol. , no. , pp., .

    [] D. S. Kernodle, R. K. R. Voladri, B. E. Menzies, C. C. Hager,and K. M. Edwards, Expression o an antisense hla ragment inStaphylococcus aureusreduces alpha-toxin production in vitroand attenuates lethal activity in a murine model,Infection andImmunity, vol. , no. , pp. , .

    [] C.Kebaier, R. R. Chamberland,I. C. Allen etal.,Staphylococcusaureusalpha-hemolysin mediates virulence in a murine modelo severe pneumonia through activation o the NLRP inam-masome,Te Journal of Infectious Diseases, vol. , no. , pp., .

  • 8/10/2019 bacterias en neo.pdf

    11/13

    Clinical and Developmental Immunology

    [] B. E. Ragle and J. Bubeck Wardenburg, Anti-alpha-hemolysinmonoclonal antibodies mediate protection againstStaphylococ-cus aureuspneumonia, Infection and Immunity, vol. , no. ,pp. , .

    [] B. E. Menzies and D. S. Kernodle, Passive immunization withantiserum to a nontoxic alpha-toxin mutant rom Staphylo-coccus aureus is protective in a murine model, Infection and

    Immunity, vol. , no. , pp. , .

    [] P. Jonsson, M. Lindberg, I. Haraldsson, and . Wadstrom,Virulence oStaphylococcus aureusin a mouse mastitis model:studies o alpha hemolysin, coagulase, and protein A as pos-sible virulence determinants with protoplast usion and genecloning,Infection and Immunity, vol. , no. , pp. ,.

    [] . Kielian, A. Cheung, and W. F. Hickey, Diminished virulenceo an alpha-toxin mutant oStaphylococcus aureus in experi-mental brain abscesses,Infection and Immunity, vol. , no. ,pp. , .

    [] M. C. Callegan, L. S. Engel, J. M. Hill, and R. J. OCallaghan,Corneal virulence oStaphylococcus aureus: roles o alpha-toxin and protein A in pathogenesis, Infection and Immunity,vol. , no. , pp. , .

    [] L. Song, M. R. Hobaugh, C. Shustak, S. Cheley, H. Bayley,and J. E. Gouaux, Structure o staphylococcal -hemolysin, aheptameric transmembrane pore,Science, vol. , no. ,pp. , .

    [] M. C. McElroy, H. R. Harty, G. E. Hosord, G. M. Boylan, J.F. Pittet, and . J. Foster, Alpha-toxin damages the air-bloodbarrier o the lung in a rat model oStaphylococcus aureus-induced pneumonia, Infection and Immunity, vol. , no. , pp., .

    [] G. A. Wilke and J. Bubeck Wardenburg, Role o a disintegrinand metalloprotease in Staphylococcus aureus-hemolysinmediated cellular injury,Proceedings of the National Academy

    of Sciences of the United States of America , vol. , no. , pp., .

    [] I. Inoshima, N. Inoshima, G. A. Wilke et al., AStaphylococcusaureuspore-orming toxin subverts the activity o ADAM tocause lethal inection in mice,Nature Medicine, vol. , no. ,pp. , .

    [] P. Hruz,A. S. Zinkernagel, G. Jenikovaet al., NOD contributesto cutaneous deense againstStaphylococcus aureusthrough-toxin-dependent innate immune activation,Proceedings of theNational Academy of Sciences of the United States of America ,vol. , no. , pp. , .

    [] A. Gravet, D. A. Colin, D. Keller, R. Giradot, H. Monteil, and G.Prevost, Characterization o a novel structural member, LukE-LukD, o the bi-component staphylococcal leucotoxins amily,

    FEBS Letters, vol. , no. , pp. , .[] J. Cooney, Z. Kienle, . J. Foster, and P. W. Ooole, Te

    gamma-hemolysin locus oStaphylococcus aureus comprisesthree linked genes, two o which are identical to the genes orthe F and S components o leukocidin, Infection and Immunity,vol. , no. , pp. , .

    [] J. Wright, Staphylococcal leucocidin (Neisser-Wechsberg type)and antileucociddin,Te Lancet, vol. , no. , pp. , .

    [] M. Labandeira-Rey, F. Couzon, S. Boisset et al., Staphylococcusaureus Panton-Valentine leukocidin causes necrotizing pneu-monia,Science, vol. , no. , pp. , .

    [] U. Lipinska, K. Hermans, L. Meulemans et al., Panton-Valentine leukocidin does play a role in the early stage o

    Staphylococcus aureus skin inections: a rabbit model, PLoSONE, vol. , no. , Article ID e, .

    [] A. Zivkovic, O. Shari, K. Stich et al., LR and CD mediateinnate immunity and lung inammation to staphylococcalpanton-valentine leukocidin in vivo, Journal of Immunology,vol. , no. , pp. , .

    [] F. Alonzo, M. A. Benson, J. Chen, R. P. Novick, B. Shopsin, andV. J. orres, Staphylococcus aureusleucocidin ED contributesto systemic inection by targeting neutrophils and promotingbacterial growth in vivo,Molecular Microbiology, vol. , no. ,pp. , .

    [] F. Alonzo, L. Kozhaya, S. A. Rawlings et al., CCR is a receptororStaphylococcus aureusleukotoxin ED,Nature, vol. , no., pp. , .

    [] R. Olson, H. Nariya, K. Yokota, Y. Kamio, and E. Gouaux,Crystal structure o staphylococcal lukF delineates conorma-tional changes accompanying ormation o a transmembranechannel, Nature Structural Biology, vol. , no. , pp. ,.

    [] J. D. Pedelacq, L. Maveyraud, G. Prevost et al., Te structure

    o a Staphylococcus aureus leucocidin component (LukF-PV)reveals the old o the water-soluble species o a amily otransmembranepore-ormingtoxins, Structure, vol., no. ,pp., .

    [] V. Guillet, P. Roblin, S. Werner et al., Crystal structure oleucotoxin S component: new insight into the staphylococcal-barrel pore-ormingtoxins, Te Journal of Biological Chemistry,vol. , no. , pp. , .

    [] K. Yamashita, Y. Kawai, Y. anaka et al., Crystal structureo the octameric pore o staphylococcal -hemolysin revealsthe-barrel pore ormation mechanism by two components,Proceedings of the National Academy of Sciences of the UnitedStates of America, vol. , no. , pp. , .

    [] F. Meyer, R. Girardot, Y. Piemont, G. Prevost, and D. A. Colin,Analysis o the specicity o panton-valentine leucocidin andgamma-hemolysin F component binding, Infection and Immu-nity, vol. , no. , pp. , .

    [] S. Szmigielski, E. Sobiczewska, G. Prevost, H. Monteil, D. A.Colin, and J. Jeljaszewicz, Effect o puried staphylococcalleukocidal toxins on isolated blood polymorphonuclear leuko-cytes and peritoneal macrophages in vitro, Zentralblatt furBakteriologie, vol. , no. , pp. , .

    [] E. J. Weston, . Pondo, M. M. Lewis et al., Te burden oinvasive early-onset neonatal sepsis in the United States, , Te Pediatric Infectious Disease Journal, vol. , no. , pp., .

    [] A. L. Shane and B. J. Stoll, Recent developments and currentissues in the epidemiology, diagnosis, and management obacterial and ungal neonatal sepsis, American Journal ofPerinatology, vol. , no. , pp. , .

    [] B. J. Stoll, N. Hansen, A. A. Fanaroff et al., Changes inpathogens causing early-onset sepsis in very-low-birth-weightinants,Te New England Journal of Medicine, vol. , no. ,pp. , .

    [] . J. Wiles and M. A. Mulvey, Te RX pore-orming toxin-hemolysin o uropathogenic Escherichia coli: progress andperspectives,Future Microbiology, vol. , pp. , .

    [] J. B. Kaper, J. P. Nataro, and H. L. . Mobley, PathogenicEscherichia coli,Nature Reviews Microbiology, vol. , no. , pp., .

  • 8/10/2019 bacterias en neo.pdf

    12/13

    Clinical and Developmental Immunology

    [] A. K. May, R. G. Sawyer, . Gleason, A. Whitworth, and .L. Pruett, In vivo cytokine response to Escherichia colialpha-hemolysin determined with genetically engineered hemolyticand nonhemolyticE. colivariants,Infection and Immunity, vol., no. , pp. , .

    [] Y. C. Smith, S. B. Rasmussen, K. K. Grande, R. M. Conran,and A. D. OBrien, Hemolysin o uropathogenic Escherichiacolievokes extensive shedding o the uroepithelium and hem-orrhage in bladder tissue within the rst hours afer intrau-rethral inoculation o mice, Infection and Immunity, vol. , no., pp. , .

    [] . J. Wiles, J. M. Bower, M. J. Redd, and M. A. Mulvey, Useo zebrash to probe the divergent virulence potentials andtoxin requirements o extraintestinal pathogenicEscherichiacoli,PLoS Pathogens, vol. , no. , Article ID e, .

    [] E. Bingen, B. Picard, N. Brahimi et al., Phylogenetic analysisoEscherichia colistrains causing neonatal meningitis suggestshorizontal gene transer rom a predominant pool o highlyvirulent B group strains, Te Journal of Infectious Diseases, vol., no. , pp. , .

    [] G. H. McCracken Jr., L. D. Sarff, and M. P. Glode, Relationbetween Escherichiacoli K capsular polysaccharideantigen andclinical outcome in neonatal meningitis,Te Lancet, vol. , no., pp. , .

    [] E. Guiral, J. Bosch, J. Vila, and S. M. Soto, Prevalence oEscherichia coli among samples collected rom the genitaltract in pregnant and nonpregnant women: relationship withvirulence,FEMS Microbiology Letters, vol. , no. , pp. , .

    [] R. A. Welch, RX toxin structure and unction: a storyo numerous anomalies and ew analogies in toxin biology,Current opics in Microbiology and Immunology, vol. , pp., .

    [] K. Baran, M. Dunstone, J. Chia et al., Te molecular basis or

    perorin oligomerization and transmembrane pore assembly,Immunity, vol. , no. , pp. , .

    [] L. Bakas, M. P. Veiga, A. Soloaga, H. Ostolaza, and F. M. Goni,Calcium-dependent conormation o E. coli -haemolysin.Implications or the mechanism o membrane insertion andlysis, Biochimica et Biophysica Acta, vol. , no. , pp. , .

    [] V. Herlax, S. Mate, O. Rimoldi, and L. Bakas, Relevance oatty acid covalently bound to Escherichia coli-hemolysin andmembrane microdomains in the oligomerization process,TeJournalof Biological Chemistry, vol. , no. , pp. ,.

    [] R. Benz, A. Dobereiner, A. Ludwig, and W. Goebel, Haemo-

    lysin oEscherichia coli: comparison o pore-orming proper-ties between chromosome and plasmid-encoded haemolysins,FEMS Microbiology Immunology, vol. , no. , pp. ,.

    [] R. Benz, A. Schmid, W. Wagner, and W. Goebel, Pore ormationby theEscherichia colihemolysin: evidence or an association-dissociation equilibrium o the pore-orming aggregates, Infec-tion and Immunity, vol. , no. , pp. , .

    [] A. Ludwig, R. Benz, and W. Goebel, Oligomerization oEscherichia colihaemolysin (HlyA) is involved in pore orma-tion,Molecular and General Genetics, vol. , no. -, pp. , .

    [] L. Bakas, A. Chanturiya, V. Herlax, and J. Zimmerberg, Para-doxical lipid dependence o pores ormed by the Escherichia

    coli -hemolysin in planar phospholipid bilayer membranes,Biophysical Journal, vol. , no. , pp. , .

    [] M. Skals, N. R. Jorgensen, J. Leipziger, and H. A. Praetorius, -hemolysin romEscherichia coliuses endogenous amplicationthrough PX receptor activation to induce hemolysis,Proceed-ings of the National Academy of Sciences of the United States ofAmerica, vol. , no. , pp. , .

    [] M. Skals, J. Leipziger, and H. A. Praetorius, Haemolysisinduced by-toxin rom Staphylococcus aureus requires PXreceptor activation,Pugers Archiv: European Journal of Physi-ology, vol. , no. , pp. , .

    [] B. K. Dhakaland M. A. Mulvey, Te UPEC pore-orming toxin-hemolysin triggers proteolysis o host proteins to disrupt celladhesion, inammatory, and survival pathways, Cell Host &Microbe, vol. , no. , pp. , .

    [] D. W. Hilbert et al., Clinical Escherichia coli isolates utilizealpha-hemolysin to inhibit in vitro epithelial cytokine produc-tion,Microbes and Infection / Institut Pasteur, vol. , no. -,pp. , .

    [] A. Cravioto, R. J.Gross, S. M. Scotland, and B.Rowe, Mannose-

    resistant haemagglutinationo human erythrocytesby strains oEscherichia colirom extraintestinal sources: lack o correlationwith colonisation actor antigen (CFA/I), FEMS MicrobiologyLetters, vol. , no. , pp. , .

    [] Z. Bian, A. Brauner, Y. Li, and S. Normark, Expression o andcytokine activation byEscherichia coli curli bers in humansepsis, Te Journal of Infectious Diseases, vol. , no. ,pp., .

    [] D. J. Evans Jr., D. G. Evans, and C. Hohne, Hemolysin and Kantigens in relation to serotype and hemagglutination type oM isolated rom extraintestinal inections,Journal of ClinicalMicrobiology, vol. , no. , pp. , .

    [] W. R. McCabe, B. Kaijser, and S. Olling, Escherichia coli inbacteremia: K and O antigens and serum sensitivity o strains

    rom adults and neonates, Te Journal of Infectious Diseases,vol. , no. , pp. , .

    [] A. Ludwig, C. Von Rhein, S. Bauer, C. Huttinger, and W.Goebel, Molecular analysis o cytolysinA (ClyA) in pathogenicEscherichia colistrains,Journal of Bacteriology, vol. , no. ,pp. , .

    [] J. ourret, M. Aloulou, L. Garry et al., Te interactionbetween a non-pathogenic and a pathogenic strain synergis-tically enhances extra-intestinal virulence in Escherichia coli,Microbiology, vol. , part , pp. , .

    [] A. J. Wallace, . J. Stillman, A. Atkins et al., E. colihemolysinE (Hlye, ClyA, SheA): X-ray crystal structure o the toxin andobservation o membrane pores by electron microscopy,Cell,vol. , no. , pp. , .

    [] M. Mueller, U. Grauschop, . Maier, R. Glockshuber, and N.Ban, Te structure o a cytolytic -helical toxin pore reveals itsassembly mechanism,Nature, vol. , no. , pp. ,.

    [] F. Pinheiro Da Silva and V. Nizet, Cell death during sepsis:integration o disintegration in the inammatory response tooverwhelming inection,Apoptosis, vol. , no. , pp. ,.

    [] J. Eitel, N. Suttorp, and B. Opitz, Innate immune recognitionand inammasome activation in listeria monocytogenes inec-tion,Frontiers in Microbiology, vol. , article , .

    [] M. Perret, C. Badiou, G. Lina et al., Cross-talk betweenStaphylococcus aureus leukocidins-intoxicated macrophages

  • 8/10/2019 bacterias en neo.pdf

    13/13

    Clinical and Developmental Immunology

    and lung epithelial cells triggers chemokine secretion in aninammasome-dependent manner,Cellular Microbiology, vol., no. , pp. , .

    [] J. S. Ayres, N. J. rinidad, and R. E. Vance, Lethal inam-masome activation by a multidrug-resistant pathobiont uponantibiotic disruption o the microbiota,Nature Medicine, vol., no. , pp. , .

    [] G. Imre and K. Rajalingam, Role or caspase- during pore-orming toxin-mediated apoptosis, Cell Cycle, vol. , no. , pp., .

    [] G. Imre, J. Heering, A. N. akeda et al., Caspase- is aninitiator caspase responsible or pore-orming toxin-mediatedapoptosis,EMBO Journal, vol. , no. , pp. , .

    [] A. L. Genestier, M. C. Michallet, G. Prevost et al., Staphylococ-cus aureus Panton-Valentine leukocidin directly targets mito-chondria and induces Bax-independent apoptosis o humanneutrophils,Te Journal of Clinical Investigation, vol. , no., pp. , .

    [] J. A. Carrero and E. R. Unanue, Mechanisms and immuno-logical effects o apoptosis caused by Listeria monocytogenes,

    Advances in Immunology, vol. , pp. , .[] C. Fernandez-Prada, B. D. all, S. E. Elliott, D. L. Hoover,

    J. P. Nataro, and M. M. Venkatesan, Hemolysin-positiveenteroaggregative and cell-detaching Escherichia coli strainscause oncosis o human monocyte-derived macrophages andapoptosis o murine J cells,Infection and Immunity, vol. ,no. , pp. , .

    [] X. H. Lai, I. Arencibia, A. Johansson et al., Cytocidal andapoptotic effects o the ClyA protein rom Escherichia colionprimary and cultured monocytes and macrophages,Infectionand Immunity, vol. , no. , pp. , .

    [] D. Rittirsch, M. A. Flierl, and P. A. Ward, Harmul molecularmechanisms in sepsis,Nature Reviews Immunology, vol. , no., pp. , .

    [] F. Stavru, F. Bouillaud, A. Sartori, D. Ricquier, and P. Cos-sart, Listeria monocytogenes transiently alters mitochon-drial dynamics during inection, Proceedings of the NationalAcademy of Sciences of the United States of America, vol. , no., pp. , .

    [] M. E. Hensler, S. Miyamoto, and V. Nizet, Group B strepto-coccal-Hemolysin/cytolysin directly impairs cardiomyocyteviability and unction, PLoS ONE, vol. , no. , Article IDe, .

    [] R. Malley and P. W. Anderson, Serotype-independent pneu-mococcal experimental vaccines that induce cellular as well ashumoral immunity, Proceedings of the National Academy ofSciences of the United States, vol. , no. , pp. , .

    [] R. P. Adhikari, H. Karauzum, J.Sarwaret al., Novel structurallydesigned vaccine or S. aureus alpha-hemolysin: protectionagainst bacteremia and pneumonia, PloS One, vol. , no. ,Article ID e, .

    [] N. Justin, N. Walker, H. L. Bullient et al., Te rst strain oClostridium perfringens isolated rom an avian source has analpha-toxin with divergent structural and kinetic properties,Biochemistry, vol. , no. , pp. , .