Lister Ios Is

download Lister Ios Is

of 6

Transcript of Lister Ios Is

  • 8/10/2019 Lister Ios Is

    1/6

    Listeriosis

    Klara M. Posfay-Barbe a,*, Ellen R. Wald b

    a Department of Pediatrics, Childrens Hospital of Geneva, University Hospitals of Geneva, 6 rue Willy-Donze, 1211 Geneva 14, Switzerlandb Department of Pediatrics, University of WisconsinMadison, School of Medicine and Public Health, 600 Highland Avenue, Box 4108, Madison, WI 53792, USA

    Keywords:

    Early-onset sepsis

    Late-onset sepsis

    Listeriosis

    Neonates

    Perinatal infection

    s u m m a r y

    Listeria monocytogenes, a small, facultative anaerobic, Gram-positive motile bacillus, is an important

    cause of foodborne illness which disproportionately affects pregnant women and their newborns. Lis-teria infects many types of animals and contaminates numerous foods including vegetables, milk,

    chicken and beef. This organism has a unique proclivity to infect the fetoplacental unit with the ability to

    invade cells, multiply intracellularly and be transmitted cell-to-cell. The organism possesses several

    virulence factors, including internalin A and internalin B, which facilitate the direct invasion of cells. Cell-

    to-cell transmission is promoted by the bacterial surface protein ActA which is regulated by a tran-

    scriptional activator known as positive regulatory factor A. Both innate and adaptive immune responses

    enable the host to eliminate this pathogen. Clinical manifestations of infection in the newborn fall into

    the traditional categories of early- and late-onset sepsis. Therapeutic recommendations include ampi-

    cillin and gentamicin for 1421 days.

    2009 Elsevier Ltd. All rights reserved.

    1. Microbiology

    Listeria monocytogenesis a small, facultatively anaerobic, Gram-

    positive, motile bacillus. It grows well in broth and on blood agar;

    some species produce a narrow zone of beta-hemolysis.1 In clinical

    specimens, the organism may be Gram-variable and look like cocci,

    diplococci, or diphtheroids, thereby misleading the laboratory

    technician. The organism tolerates low temperatures as well as

    high pH and high salt concentrations, which allow it to replicate in

    soil, water, sewage, manure, animal feed, and contaminated

    refrigerated foods. It can survive many months in soil and 2030

    days in tap water. Although persisting on environmental surfaces,

    pasteurization and most disinfecting agents eliminateListeria.

    Only four of the seven species ofListeria infect humans. Most

    diseases are due to three primary serotypes: 1a, 1b and 4b. The last

    is responsible for almost all outbreaks of listeriosis.2

    2. Epidemiology

    Listeria spp. are distributed worldwide, but human illness is

    reported most frequently in developed countries. Listeria spp. are

    an important cause of zoonoses, infecting many types of animals

    (domestic pets, livestock, other mammals, rodents, amphibians,

    fish, and arthropods) and more than 17 avian species. In

    mammals,L. monocytogenes can cause spontaneous abortions and

    is the cause of circling disease, a manifestation of basilar

    meningitis in which animals move incessantly in a circle. Fecal

    oral transmission is the probable means by which organisms are

    spread in animals. The pathogen can be transmitted directly from

    animals to humans and has been documented in veterinarians,

    farmers, and abattoir workers. Vertical transmission from mother

    to neonate occurs transplacentally or through an infected birth

    canal. Cross-infection in a neonatal unit through contact with

    contaminated mineral oil used to bathe infants led to one noso-

    comial outbreak.3

    Most cases of listeriosis appear to be foodborne, including those

    acquired during pregnancy. Many foods can be contaminated by L.

    monocytogenes, including raw vegetables, raw milk, fish, poultry,

    processed chicken, and beef. Approximately 1570% of hot dogs are

    reported to be contaminated withListeriaspp.Listeriaspp. also are

    found in the stools ofw5% of healthy adults.4 The infectious dose isestimated to be 104106 organisms per gram of ingested product

    but may be lower in immunocompromised hosts and patients who

    have diminished gastric acidity or have undergone ulcer surgery.5

    The incubation period has not been well-established, but is esti-

    mated to be three weeks.

    The first clearly documented foodborne (coleslaw) outbreak

    was in Nova Scotia in 1981.5 It was associated with a case fatality

    of 27%. In other sporadic outbreaks, 11% of all food samples

    retrieved from the refrigerator were contaminated, and 64% of the

    refrigerators of patients contained at least one contaminated food

    item.2* Corresponding author. Tel.: 41 22 382 5462; fax: 41 22 382 5490.

    E-mail address: [email protected](K.M. Posfay-Barbe).

    Contents lists available atScienceDirect

    Seminars in Fetal & Neonatal Medicine

    j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / s i n y

    1744-165X/$ see front matter 2009 Elsevier Ltd. All rights reserved.

    doi:10.1016/j.siny.2009.01.006

    Seminars in Fetal & Neonatal Medicine 14 (2009) 228233

    mailto:[email protected]://www.sciencedirect.com/science/journal/1744165Xhttp://www.elsevier.com/locate/sinyhttp://www.elsevier.com/locate/sinyhttp://www.sciencedirect.com/science/journal/1744165Xmailto:[email protected]
  • 8/10/2019 Lister Ios Is

    2/6

  • 8/10/2019 Lister Ios Is

    3/6

    was the first identified virulence factor ofL. monocytogenesand is

    required for bacterial escape from endocytic vacuoles. LLO-defec-

    tive strains are five to 10 times less invasive than wild-type

    strains.22 LLO is required in vivo for bacterial growth in tropho-

    blastic cells, and for subsequent fetal invasion.8 It is also known as

    a modulator of the innate immune response by inducing new

    cytokine synthesis in different immune cells and activating

    different immune-response-associated pathways, such as the NF-

    kappaB pathway.23 The mechanisms behind this activation process

    are, however, still unclear.

    3.3. Cell-to-cell transmission

    Once in the cytoplasm, listeria multiplies rapidly, migrates to the

    host cell periphery and into cell-wall protrusions, which will be

    taken up and ingested by neighboring cells where a new cycle will

    start again (Fig. 1). This migration to the surface of the hosts cell

    depends on ActA, a bacterial surface protein.

    3.3.1. ActA

    ActA is a virulence factor required for cell-to-cell spread of lis-

    teria. It uses components of the host cells actin cytoskeleton to

    generate a comet-tail which is rich in actin, enabling bacterialpropulsion in the cytosol leading to the infection of uninfected

    neighbor cells.24 Strains lacking ActA retain the ability to grow in

    the placenta although at a lower rate than wild-type strains but

    arestronglycurtailed in their ability to spread from cell to cell.8 This

    was demonstrated in an animal model, in which neonatal mice

    injected with an isolate ofL. monocytogenes lacking ActA induced

    a strong primary and secondary Th1 CD4 and CD8 T-cell response,

    but were protected against death from infection with listeria.

    Interestingly, motility can be conferred upon other non-motile

    micro-organisms (such asL. innocua) by adding ActA.25

    3.3.2. Positive regulatory factor A

    The expression of nearly all bacterial gene products that

    contribute to the survival and virulence of L. monocytogenes,including LLO and ActA, are regulated by a transcriptional activator

    known as positive regulatory factor A (PrfA). Strains lacking func-

    tional PrfA are not virulent. The effect of PrfA seems to be reduced

    at low temperature, but increased at low pH, probably explaining

    the increased virulence of strains in a warm, acidic environment

    after oral ingestion.26,27 Post-transcriptional mechanisms control-

    ling PrfA expression have also been described and contribute to the

    synthesis of Int1A, Int1B, and LLO.11

    In summary, it is currently believed that: In1A (especially) and

    In1B play a key role in the entry of listeria into host cells; LLO

    punches holes to allow the organism to multiply inside the cyto-

    plasm of the host cell; ActA produces a rocket-like mechanism to

    penetrate into neighboring cells; and that all these mechanisms are

    co-ordinated by PrfA. New virulence factors forL. monocytogenesaswell as genes involved in virulence or stress-response are described

    regularly, increasing the complexity of the bacteriums pathoge-

    nicity and its regulatory mechanisms.28

    4. Immunology

    4.1. Innate and adaptive immune response

    L. monocytogenes has been used for decades as a model

    organism to study innate and adaptive immune responses against

    intracellular pathogens. The innate immune response is immediate

    and involves multiple cellular types, cytokines, and bactericidal

    effector mechanisms. Monocytes and resident macrophages,

    Kupffer cells for example, are known to ingest and destroy listeria.

    Production of cytokines, such as interleukin (IL)-1, IL-6, and tumor

    necrosis factor (TNF)-a are central in decreasing susceptibility to

    disease by recruiting neutrophils. On the other hand, the adaptive

    immune response against infection induced by L. monocytogenes

    peaks about one week after infection, and is mainly a CD8 T-cell

    response. This response has twomain functions: the specific lysis of

    infected cells and rapidproduction of IFN-g in response to IL-12 and

    IL-18.29 It is now hypothesized that early IFN-gproduction leads to

    an accelerated formation of granulomas at sites of infection,

    thereby walling off the infection. In mouse strains of listeria that

    do not elicit an early IFN-gresponse, acute inflammation continues

    and listeria spreads between cells. Clearance ofL. monocytogenesis

    believed to be mediated through a Th1 immune response. IFN-gis

    a crucial contributor to the Th1 response by activating macro-

    phages, increasing antigen presentation via the MHC class I and II

    pathways, and by inhibiting the expansion of Th2 cells.30

    A number of research groups have been focused on describing

    the cells which produce IFN-g in infections caused by L. mono-

    cytogenes. It seems likely that several cells, including natural killer

    (NK) cells, dendritic cells during the early phase of innate immu-

    nity, antigen-specific CD4 TH1 cells,and effector and memory CD8

    T-cells during the later phase of adaptive immunity, produce IFN-g

    during infection.29,31 Tocomplicate ourunderstanding of the role ofIFN-gin listerial infection, it appears that IFN-gboth promotes the

    control of bacterial replication and also induces (paradoxically), the

    erosion of CD8 T-cell memory during infection, thereby impairing

    subsequent protective mechanisms.32 In summary, IFN-g plays

    a critical role in both innate and adaptive immune responses to L.

    monocytogenes.

    4.2. Toll-like receptors and autophagy

    Toll-like receptor (TLR) 2 and TLR5 have been reported to

    recognize L. monocytogenes.33 TLR recognition of listerias lip-

    oteichoic acid, lipoproteins, peptidoglycans and flagellin leads to

    the production of cytokines which inturn directly activate innate

    effectors and recruit other cells.34,35 Interestingly, however, ina murine model in which animals lack myeloid differentiation

    factor 88 (MyD88), an adaptor molecule downstream from TLRs, it

    was shown that the animals are highly susceptible to infectionwith

    listeria and that a MyD88-independent innate and adaptive

    immune response exists.36

    Macro-autophagy, also referred to as autophagy, is a recently

    described mechanism involved in the immune response of

    mammals to micro-organisms.37 Briefly, through autophagy, an

    infected cell can target an invading micro-organism and restrict its

    growth by destroying it in the lysosome. It then induces an adaptive

    immune response via presenting peptides on MHC Class I and II

    molecules. Recent studies have shown that autophagy targets L.

    monocytogenes before bacterial escape into the cytosol. However,

    listeria is capable of using alternate strategies, including ActA-dependent polymerization mentioned earlier, to avoid

    destruction.38

    4.3. Specific relation to immunology of pregnant woman and child

    The maternal immune system faces an important challenge

    during pregnancy, i.e. to prevent rejection of the semi-allogenic

    fetus and to protect itself and also the fetus from infection. Many

    observations suggest that pregnancy is associated with a shift from

    Th1 to Th2 cellular response and that the maternal immune system

    is biased toward humoral immunity and away from cell-mediated

    immunity (that could be harmful to the fetus). However, these

    changes appear mostly to occur locally, as there is no strong

    evidence suggesting that the maternal immune system is

    K.M. Posfay-Barbe, E.R. Wald / Seminars in Fetal & Neonatal Medicine 14 (2009) 228233230

  • 8/10/2019 Lister Ios Is

    4/6

    compromised overall during pregnancy. New immunomodulating

    mechanisms, not specific to L. monocytogenes, have been studied

    extensively and are reviewed elsewhere.39,40 L. monocytogenes, on

    the other hand, has to develop different strategies against the

    defence mechanisms of the host to survive. While some of these

    defences are not specific to pregnancy, such as gastric acidity, lis-

    teria has mastered manipulating the differences related to the

    immaturity of the immune system of the fetus. Neonatal immunity

    is reported to be Th2-biased. This avoids maternal rejection of fetal

    antigens induced by Th1-type inflammation that may result in

    spontaneous abortion or premature delivery.41 Furthermore, it

    appears that (murine) neonates have a lower expression of

    mannose-binding lectins and certain TLRs than adults.42 These

    mediators are necessary to develop Th1 immune responses and

    might explainwhy neonates are highly susceptible to infectionwith

    listeria. This fact has been questioned by others who believe that it

    is not a decreased expression of TLRs, but instead a TLR-induced

    TNF-a production which is significantly decreased in neonates.43

    Interestingly, the same authors acknowledge that neonates are

    capable of a robust IL-6 production, indicating that the intrinsic TLR

    pathway is functional at birth, and suggesting an abnormal, still

    unclear, TLR defect related to Th2.

    L. monocytogenes can also gain access to the neonate via oralexposure during the passage through the birth canal. Enteric anti-

    microbial peptides derived from Paneth cells provide protection

    from intestinal infection. However, these cells are not present

    during the first weeks of life. Recently, Menard et al. described an

    antimicrobially active form of peptide, called CRAMP (cathelin-

    related antimicrobial peptide), in the epithelium of neonatal mice,

    which is expressed constitutively during the first weeks of life

    only.44 CRAMP provides significant protection against bacterial

    growth in the gut. This developmental switch in innate immune

    effector expression (i.e. CRAMP being functional only while Paneth

    cells are lacking) is a newly described mechanism in the early

    protection against infection with listeria. Enhanced understanding

    of the specificity of the immune system in neonates may lead to

    novel treatment strategies in years to come.

    5. Neonatal disease

    Worldwide,L. monocytogenesis one of the three major causes of

    meningitis in neonates. Clinical manifestations may be very similar

    to those seen with group B streptococcal disease, and there is a high

    fatality rate (350%). There are two forms of neonatal listeriosis.

    5.1. Early onset

    The mothers of affected children often have a flu-like illness

    a few days before delivery, which may be preterm. Listeria

    bacteremia presents with an acute febrile illness, which is often

    accompanied by myalgias, arthralgias, backache and headache.Maternal illness is observed most often in the third trimester.

    Neonatal death and stillbirth occur in approximately one-fifth of

    maternal infections. Two-thirds of the infants who survive delivery

    to a woman who experiences listeriosis in pregnancy will develop

    neonatal infection. The infants are believed to be infected in utero

    because of the bacteremic phase of their mothers, but ascending

    infections have been described. The highest concentrations of L.

    monocytogenes are found in the lung and gut, suggesting that

    infection also can be acquired in utero via inhalation and ingestion

    of infected amniotic fluid as well as via the hematogenous route.

    During labor, brown-stained amniotic fluid is seen and maternal

    fever may be present. Serotypes 1a and 1b are most common.

    The mean onset of symptomsis 1.5 days after birth. A sepsis-like

    picture predominates, but other common manifestations are acute

    respiratory distress, pneumonia, and more rarely, meningitis or

    myocarditis. Strategies implemented to prevent group B strepto-

    coccal sepsis in the newborn may have secondarily decreased the

    rate of early-onset listerial infection.

    Granulomatosis infantisepticum is a widely disseminated

    granuloma characteristic of severe listerial disease. The lesions are

    more common in the liver, skin and placenta, but also appear in the

    brain, adrenal glands, spleen, kidney, lungs and gastrointestinal

    tract. Aspiration of infected fluid can contribute to acute respiratory

    failure and hemodynamic compromise in the neonate.

    5.2. Late onset

    This form of neonatal listeriosis is less common than the early-

    onset form, but it occurs more frequently in term infants who are

    the products of uncomplicated pregnancies. The babies are healthy

    at birth, and the maternal history is usually normal. The first

    manifestations of infection appear several days to weeks after birth,

    with a mean onset of illness at 14.3 days after birth. The clinical

    manifestation in this group is more likely to be meningitis than

    sepsis, but it can be subtle, with fever, irritability, anorexia, diar-

    rhea, and lethargy. Postpartum transmission is assumed to occur

    either during delivery or nosocomially. Serotype 4b is mostfrequent in late-onset disease.

    6. Diagnosis

    Listeriosis usually presents with leukocytosis, but unlike its

    name, rarely with monocytosis. Isolation of Listeria spp. from

    a normally sterile site defines the disease. Thirty-six hours of

    incubation usually are necessary for sufficient growth for identifi-

    cation. The pathogen often is observed on the Gram stain of the

    meconium of infected newborns. Carriage in the gastrointestinal

    tracts of older children may be common but difficult to demon-

    strate because the organism is fastidious and normal flora are

    plentiful. Screening of rectal or vaginal cultures is not clinically

    useful.Cold enrichment is not as good as selective media to isolate the

    organism from specimens containing multiple species (e.g. in food

    or stools). Rapid detection tests are based on the use of either

    monoclonal antibodies or nucleic acid hybridizations, but they only

    identify the genus Listeria.45

    When the central nervous system is infected, cerebrospinal fluid

    (CSF) is usually purulent, with leukocyte counts of 10010 000/mL

    (0.110 109/L). Polymorphonuclear leukocytes predominate in

    70% of cases, but the severity of the inflammatory response does

    notcorrelate with the prognosis. The Gram stain of CSFis positive in

  • 8/10/2019 Lister Ios Is

    5/6

    L. monocytogenes can multiply within macrophages, several

    prerequisites must be fulfilled to inhibit these hidden bacteria.

    Antibiotics must penetrate into and distribute within host cells and

    remain stable within this environment. Recommmendations are

    based on data obtained from in-vitro susceptibility testing, animal

    models, and clinical experience with small numbers of patients

    who were compared with historic controls.

    No controlled trials have established a drug of choice or duration

    of therapy for listeriosis. L. monocytogenes is sensitive in vitro to

    penicillin G, ampicillin, erythromycin, sulfamethoxazole, trimeth-

    oprim, chloramphenicol, rifampin, tetracyclines, and aminoglyco-

    sides. Bactericidal antibiotics include sulfamethoxazole,

    trimethoprim, and aminoglycosides. Gentamicin and tobramycin

    have been reported to have greater in-vitro activity than strepto-

    mycin, kanamycin, and amikacin. Listeria always are resistant to

    cephalosporins.48 Chloramphenicol should not be used because of

    unacceptable failure and relapse rates, and quinolones do not havegood in-vitro activity. Ampicillin, which is superior to penicillin,

    and gentamicin have shown synergistic effects in some studies.48

    Accordingly, gentamicin with ampicillin is recommended for

    treatment of listerial meningitis.

    If the patient is allergic to ampicillin or gentamicin, sulfame-

    thoxazole-trimethoprim is recommended because it is bactericidal

    and reaches adequate levels in the serum and CSF.48 No systematic

    study has examined the duration of therapy, but the current

    recommendations are 1421 days of treatment for meningitis due

    toL. monocytogenes.

    8. Prevention

    There is no vaccine for listeria infection. Dietary recommenda-

    tions for preventing foodborne listeriosis were established by the

    Centers for Disease Control and Prevention (CDC) in 1992. They are

    similar to those for other foodborne illnesses and include thorough

    cooking of raw food from animal sources; washing of raw vegeta-

    bles; avoidance of unpasteurized dairy products; keeping uncooked

    meats separate from vegetables; washing hands, knives, and

    cutting boards after exposure to uncooked food; and regular

    cleaning and disinfection of the insides of refrigerators. Persons at

    high risk for listeriosis should avoid soft cheeses, reheat (until

    steaming hot) leftover and ready-to-eat foods, and avoid cold cuts if

    unable to reheat them thoroughly.

    TheUS Department of Agriculture began surveillance in 1989 for

    L. monocytogenes in ready-to-eat processed meats and enforced

    regulations prohibiting the sale of contaminated meats. Since then,

    the number of cases of listerial infection has dropped substantially.

    The CDCs Division of Foodborne, Bacterial and Mycotic Diseases

    tracks the annual incidence of certain foodborne illnesses,

    including listeria, in 10 states through Food Net Surveillance.49

    Between 1996 and 2006, the incidence of listeria declined by 36%,

    although an outbreak in 2002, related to contaminated turkey

    meat, resulted in 54 illnesses, eight deaths and three fetal deaths in

    nine states (Fig. 2).49

    References

    1. Swaminathan B, Hayes PS, Przybyszewski VA, Plikaytis BD. Evaluation ofenrichment and plating media for isolating Listeria monocytogenes. J Assoc Off

    Anal Chem 1988;71:6648.2. Pinner RW, Schuchat A, Swaminathan B, et al. Role of foods in sporadic liste-

    riosis. II. Microbiologic and epidemiologic investigation. The Listeria StudyGroup. J Am Med Assoc1992;267:204650.

    3. Schuchat A, Lizano C, Broome CV, Swaminathan B, Kim C, Winn I. Outbreak ofneonatal listeriosis associated with mineral oil. Pediatr Infect Dis J 1991;10:1839.

    4. Schlech III WF. Foodborne listeriosis. Clin Infect Dis2000;31:7705.5. Schlech III WF. Listeria gastroenteritis old syndrome, new pathogen.N Engl J

    Med1997;336:1302.

    6. Kaur S, Malik SV, Vaidya VM, Barbuddhe SB. Listeria monocytogenes in spon-taneous abortions in humans and its detection by multiplex PCR. J ApplMicrobiol2007;103:188996.

    7. Redline RW, Lu CY. Specific defects in the anti-listerial immune response indiscrete regions of the murine uterus and placenta account for susceptibility toinfection. J Immunol 1988;140:394755.

    8. Le Monnier A, Autret N, Join-Lambert OF, et al. ActA is required for crossing ofthe fetoplacental barrier by Listeria monocytogenes. Infect Immun 2007;75:9507.

    9. DOrazio SE, Troese MJ, Starnbach MN. Cytosolic localization of Listeria mono-cytogenestriggers an early IFN-gamma response by CD8 T cells that correlateswith innate resistance to infection. J Immunol 2006;177:714654.

    10. Seveau S, Pizarro-Cerda J, Cossart P. Molecular mechanisms exploited by Lis-teria monocytogenes during host cell invasion. Microbes Infect2007;9:116775.

    11. Gray MJ, Freitag NE, Boor KJ. How the bacterial pathogenListeria monocytogenesmediates the switch from environmental Dr. Jekyll to pathogenic Mr. Hyde.Infect Immun 2006;74:250512.

    12. Hamon M, Bierne H, Cossart P. Listeria monocytogenes: a multifaceted model.Nat Rev Microbiol 2006;4:42334.

    13. Lecuit M, Dramsi S, Gottardi C, Fedor-Chaiken M, Gumbiner B, Cossart P.A single amino acid in E-cadherin responsible for host specificitytowar ds the h uman p athogen Listeria monocytogenes. EMBO J 1999;18:395663.

    14. Pentecost M, Otto G, Theriot JA, Amieva MR.Listeria monocytogenesinvades theepithelial junctions at sites of cell extrusion. PLoS Pathog2006;2:e3.

    15. Lecuit M, Nelson DM, Smith SD, et al. Targeting and crossing of the humanmaternofetal barrier by Listeria monocytogenes: role of internalin interactionwith trophoblast E-cadherin. Proc Natl Acad Sci USA 2004;101:61527.

    16. Bonazzi M, Veiga E, Cerda JP, Cossart P. Successive post-translational modifi-cations of E-cadherin are required for InlA-mediated internalisation ofListeriamonocytogenes. Cell Microbiol 2008; 10: 220822.

    17. Jacquet C, Doumith M, Gordon JI, Martin PM, Cossart P, Lecuit M. A molecularmarker for evaluating the pathogenic potential of foodborne Listeria mono-cytogenes. J Infect Dis 2004;189:2094100.

    18. Bakardjiev AI, Stacy BA, Fisher SJ, Portnoy DA. Listeriosis in the pregnant guineapig: a model of vertical transmission. Infect Immun 2004;72:48997.

    19. Shen Y, Naujokas M, Park M, Ireton K. InIB-dependent internalization of Listeriais mediated by the Met receptor tyrosine kinase. Cell 2000;103:50110.

    2.0

    1.0

    0.5

    0.4

    1996-1998

    1999 2000 2001 2002 2003

    Year

    Relative

    rate(logscale)

    2004 2005 2006 2007

    0.9

    0.8

    0.7

    0.6

    Fig. 2. Relative rates of laboratory-confirmed infections with listeria compared with

    19961998 rates, by year (Foodborne Diseases Active Surveillance Network, USA,

    19962007).

    Research directions

    To understand better how this quiet saprophyte

    becomes a deadly agent.

    To determine the infectious dose of listeria and to define

    the extent of human illness by searching for listeria inoutbreaks of febrile gastroenteritis where no other

    pathogens are identified.

    To identify new bacterial and host effectors.

    To examine interaction and activation processes

    between bacteria and host, especially in vulnerable

    populations, such as neonates.

    K.M. Posfay-Barbe, E.R. Wald / Seminars in Fetal & Neonatal Medicine 14 (2009) 228233232

  • 8/10/2019 Lister Ios Is

    6/6

    20. Greiffenberg L, Goebel W, Kim KS, et al. Interaction of Listeria monocytogeneswith human brain microvascular endothelial cells: InlB-dependent invasion,long-term intracellular growth, and spread from macrophages to endothelialcells.Infect Immun 1998;66:52607.

    21. Sabet C, Lecuit M, Cabanes D, Cossart P, Bierne H. LPXTG protein InlJ, a newlyidentified internalin involved inListeria monocytogenesvirulence.Infect Immun2005;73:691222.

    22. Dramsi S, Cossart P. Listeriolysin O-mediated calcium influx potentiates entry ofListeria monocytogenes into the human Hep-2 epithelial cell line. Infect Immun2003;71:36148.

    23. Park JM,Ng VH,Maeda S,RestRF,Karin M.AnthrolysinO andother gram-positivecytolysins are toll-like receptor 4 agonists. J Exp Med2004;200:164755.

    24. Portnoy DA, Auerbuch V, Glomski IJ. The cell biology ofListeria monocytogenesinfection: the intersection of bacterial pathogenesis and cell-mediated immu-nity. J Cell Biol 2002;158:40914.

    25. Kocks C, Marchand JB, Gouin E, et al. The unrelated surface proteins ActA ofListeria monocytogenesand IcsA ofShigella flexneriare sufficient to confer actin-based motility on Listeria innocua and Escherichia coli respectively. Mol Micro-biol 1995;18:41323.

    26. Johansson J, Mandin P, Renzoni A, Chiaruttini C, Springer M, Cossart P. An RNAthermosensor controls expression of virulence genes in Listeria mono-cytogenes.Cell 2002;110:55161.

    27. Ferreira A, Sue D, OByrne CP, Boor KJ. Role ofListeria monocytogenes sigma(B) insurvival of lethal acidic conditions and in the acquired acid tolerance response.

    Appl Environ Microbiol2003;69:26928.28. Chaturongakul S, Raengpradub S, Wiedmann M, Boor KJ. Modulation of stress

    and virulence in Listeria monocytogenes. Trends Microbiol2008;16:38896.29. Berg RE, Crossley E, Murray S, Forman J. Memory CD8T cells provide innate

    immune protection against Listeria monocytogenes in the absence of cognateantigen.J Exp Med 2003;198:158393.

    30. Pamer EG. Immune responses to Listeria monocytogenes. Nat Rev Immunol2004;4:81223.

    31. Plitas G, Chaudhry UI, Kingham TP, Raab JR, DeMatteo RP. NK dendritic cells areinnate immune responders to Listeria monocytogenes infection. J Immunol2007;178:44116.

    32. Dudani R, Murali-Krishna K, Krishnan L, Sad S. IFN-gamma induces the erosionof preexisting CD8 T cell memory during infection with a heterologous intra-cellularbacterium. J Immunol 2008;181:17009.

    33. Torres D, Barrier M, Bihl F, et al. Toll-like receptor 2 is required for optimalcontrol ofListeria monocytogenes infection. Infect Immun 2004;72:21319.

    34. Hayashi F, Smith KD, Ozinsky A, et al. The innate immune response to bacterialflagellin is mediated by Toll-like receptor 5. Nature 2001;410:1099103.

    35. Machata S, Tchatalbachev S, Mohamed W, Jansch L, Hain T, Chakraborty T.Lipoproteins of Listeria monocytogenes are critical for virulence and TLR2-mediated immune activation. J Immunol 2008;181:202835.

    36. Way SS, Kollmann TR, Hajjar AM, Wilson CB. Cutting edge: protective cell-mediated immunity to Listeria monocytogenes in the absence of myeloiddifferentiation factor 88. J Immunol 2003;171:5337.

    37. Kirkegaard K, Taylor MP, Jackson WT. Cellular autophagy: surrender, avoidanceand subversion by microorganisms. Nat Rev Microbiol 2004;2:30114.

    38. Birmingham CL,Canadien V, Gouin E, et al. Listeria monocytogenes evades killingby autophagy during colonization of host cells. Autophagy2007;3:44251.

    39. Poole JA, Claman HN. Immunology of pregnancy. Implications for the mother.Clin Rev Allergy Immunol 2004;26:16170.

    40. Seavey MM, Mosmann TR. Immunoregulation of fetal and anti-paternalimmune responses. Immunol Res2008;40:97113.

    41. Adkins B, Leclerc C, Marshall-Clarke S. Neonatal adaptive immunity comes ofage. Nat Rev Immunol 2004;4:55364.

    42. Byun HJ, Jung WW, Lee JB, et al. An evaluation of the neonatal immune systemusing a listeria infection model. Neonatology2007;92:8390.

    43. Levy O, Coughlin M, Cronstein BN, Ry RM, Desai A, Wessels M. The adenosinesystem selectively inhibits TLR-mediated TNF-a production in the humannewborn. J Immunol 2006;144:195666.

    44. Menard S, Forster V, Lotz M, et al. Developmental switch of intestinal antimi-crobial peptide expression. J Exp Med 2008;205:18393.

    45. Sontakke S, Farber JM. The use of PCR ribotyping for typing strains ofListeriaspp. Eur J Epidemiol 1995;11:66573.

    46. Dalton CB, Austin CC, Sobel J, et al. An outbreak of gastroenteritis and fever dueto Listeria monocytogenes in milk. N Engl J Med 1997;336:1005.

    47. Marget W, Seeliger HP. Listeria monocytogenes infections therapeutic possi-bilities and problems. Infection 1988;16:S1757.

    48. Espaze EP, Reynaud AE. Antibiotic susceptibilities ofListeria: in vitro studies.Infection1988;16:S1604.

    49. Anonymous. Preliminary foodnet data on the incidence of infection withpathogens transmitted commonly through food 10 states, 2007.MMWR MorbMortal Wkly Rep2008;57:36670.

    K.M. Posfay-Barbe, E.R. Wald / Seminars in Fetal & Neonatal Medicine 14 (2009) 228233 233