perinatal group b streptococcal infections: virulence factors, … · 2020-03-13 · are...

13
Review Perinatal Group B Streptococcal Infections: Virulence Factors, Immunity, and Prevention Strategies Jay Vornhagen, 1,2 Kristina M. Adams Waldorf, 3,4 and Lakshmi Rajagopal 1,2,5, * Group B streptococcus (GBS) or Streptococcus agalactiae is a b-hemolytic, Gram-positive bacterium that is a leading cause of neonatal infections. GBS commonly colonizes the lower gastrointestinal and genital tracts and, during pregnancy, neonates are at risk of infection. Although intrapartum antibiotic prophylaxis during labor and delivery has decreased the incidence of early- onset neonatal infection, these measures do not prevent ascending infection that can occur earlier in pregnancy leading to preterm births, stillbirths, or late- onset neonatal infections. Prevention of GBS infection in pregnancy is complex and is likely inuenced by multiple factors, including pathogenicity, host fac- tors, vaginal microbiome, false-negative screening, and/or changes in antibi- otic resistance. A deeper understanding of the mechanisms of GBS infections during pregnancy will facilitate the development of novel therapeutics and vaccines. Here, we summarize and discuss important advancements in our understanding of GBS vaginal colonization, ascending infection, and preterm birth. Infections by GBS during Pregnancy GBS is a leading cause of infection during pregnancy, preterm birth, and neonatal infection [13]. GBS was rst identied in 1887 as a cause of bovine mastitis [4], and later was isolated from the human vagina [5] and associated with cases of human disease [6]. Subsequently, GBS vaginal colonization was identied as a risk factor for the development of neonatal GBS disease [7,8] and preterm birth [2,3]. Women who are vaginally colonized during pregnancy are at risk for ascending infection or transmission of GBS to the newborn during delivery. Ascending infection is a widely accepted route by which vaginal bacteria move from the vagina, through the cervix, and into the uterus and penetrate gestational tissues (Figure 1). Once GBS has invaded the amniotic cavity, or come into contact with the placenta, there is the potential for chorioamnionitis or inammation of the placental membranes that is frequently associated with preterm births and stillbirths [9]. Globally, preterm birth is a signicant contributor to neonatal death. Every year, approximately 6 000 000 births are preterm, and more than 500 000 neonates die due to prematurity, accounting for 44% of all deaths under the age of 5 years [10,11]. The majority of early preterm births are due to microbial infection [9], and approximately 10% are attributable to GBS [1214]. The bacterial and host determinants that promote GBS vaginal colonization, ascending infection, and adverse perinatal outcomes are poorly understood. Trends Group B streptococcus (GBS) has to evade genitourinary immune responses for successful colonization and infection. A number of virulence factors promote GBS vaginal colonization and subse- quent ascension into the pregnant uterus. The GBS hemolytic pigment and hya- luronidase enable the pathogen to resist immune responses; however, dysregulation of the hemolytic pigment can also lead to bacterial clearance from the vagina. GBS invasion of the amniotic uid is not absolutely necessary for induction of inammatory responses and pre- term birth. Development of a GBS vaccine is cri- tical to reduce or prevent the risk of infection during pregnancy, thereby reducing GBS disease burden. 1 Department of Global Health, University of Washington, Seattle, WA, USA 2 Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, USA 3 Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA 4 Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden 5 Department of Pediatrics, University of Washington, Seattle, WA, USA *Correspondence: [email protected] (L. Rajagopal). Trends in Microbiology, November 2017, Vol. 25, No. 11 http://dx.doi.org/10.1016/j.tim.2017.05.013 919 © 2017 Elsevier Ltd. All rights reserved. Downloaded for Anonymous User (n/a) at University of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017. For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Upload: others

Post on 28-Jul-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

TrendsGroup B streptococcus (GBS) has toevade genitourinary immune responsesfor successful colonization and infection.

A number of virulence factors promoteGBS vaginal colonization and subse-quent ascension into the pregnantuterus.

The GBS hemolytic pigment and hya-luronidase enable the pathogen toresist immune responses; however,dysregulation of the hemolytic pigmentcan also lead to bacterial clearancefrom the vagina.

GBS invasion of the amniotic fluid isnot absolutely necessary for induction

ReviewPerinatal Group BStreptococcal Infections:Virulence Factors, Immunity,and Prevention StrategiesJay Vornhagen,1,2 Kristina M. Adams Waldorf,3,4 andLakshmi Rajagopal1,2,5,*

Group B streptococcus (GBS) or Streptococcus agalactiae is a b-hemolytic,Gram-positive bacterium that is a leading cause of neonatal infections. GBScommonly colonizes the lower gastrointestinal and genital tracts and, duringpregnancy, neonates are at risk of infection. Although intrapartum antibioticprophylaxis during labor and delivery has decreased the incidence of early-onset neonatal infection, these measures do not prevent ascending infectionthat can occur earlier in pregnancy leading to preterm births, stillbirths, or late-onset neonatal infections. Prevention of GBS infection in pregnancy is complexand is likely influenced by multiple factors, including pathogenicity, host fac-tors, vaginal microbiome, false-negative screening, and/or changes in antibi-otic resistance. A deeper understanding of the mechanisms of GBS infectionsduring pregnancy will facilitate the development of novel therapeutics andvaccines. Here, we summarize and discuss important advancements in ourunderstanding of GBS vaginal colonization, ascending infection, and pretermbirth.

of inflammatory responses and pre-term birth.

Development of a GBS vaccine is cri-tical to reduce or prevent the risk ofinfection during pregnancy, therebyreducing GBS disease burden.

1Department of Global Health,University of Washington, Seattle, WA,USA2Center for Global Infectious DiseaseResearch, Seattle Children’s ResearchInstitute, Seattle, WA, USA3Department of Obstetrics andGynecology, University ofWashington, Seattle, WA, USA4Sahlgrenska Academy, GothenburgUniversity, Gothenburg, Sweden5Department of Pediatrics, Universityof Washington, Seattle, WA, USA

*Correspondence:[email protected](L. Rajagopal).

Infections by GBS during PregnancyGBS is a leading cause of infection during pregnancy, preterm birth, and neonatal infection [1–3]. GBS was first identified in 1887 as a cause of bovine mastitis [4], and later was isolated fromthe human vagina [5] and associated with cases of human disease [6]. Subsequently, GBSvaginal colonization was identified as a risk factor for the development of neonatal GBS disease[7,8] and preterm birth [2,3]. Women who are vaginally colonized during pregnancy are at riskfor ascending infection or transmission of GBS to the newborn during delivery. Ascendinginfection is a widely accepted route by which vaginal bacteria move from the vagina, throughthe cervix, and into the uterus and penetrate gestational tissues (Figure 1). Once GBS hasinvaded the amniotic cavity, or come into contact with the placenta, there is the potential forchorioamnionitis or inflammation of the placental membranes that is frequently associated withpreterm births and stillbirths [9]. Globally, preterm birth is a significant contributor to neonataldeath. Every year, approximately 6 000 000 births are preterm, and more than 500 000neonates die due to prematurity, accounting for 44% of all deaths under the age of 5 years[10,11]. The majority of early preterm births are due to microbial infection [9], and approximately10% are attributable to GBS [12–14]. The bacterial and host determinants that promote GBSvaginal colonization, ascending infection, and adverse perinatal outcomes are poorlyunderstood.

Trends in Microbiology, November 2017, Vol. 25, No. 11 http://dx.doi.org/10.1016/j.tim.2017.05.013 919© 2017 Elsevier Ltd. All rights reserved.

Downloaded for Anonymous User (n/a) at University of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 2: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

Choriodecidual space

Uterus

Cervix

Vagina

Group B streptococcus

Endometrium

Decidua

Chorion

Amnion

Amnio�c fluid

Amnio�c epithelium

Figure 1. Ascending Group B Streptococcus (GBS) Infection. GBS vaginal colonization increases the risk ofascending infection during pregnancy. Ascending GBS infection during pregnancy involves bacterial trafficking from thevagina, ultimately leading to bacterial invasion of placental membranes (chorion and amnion), the amniotic cavity, and thefetus. GBS expresses a number of virulence factors that promote vaginal colonization, adhesion and invasion of host cells,and for either activation or suppression of inflammatory responses (Table 1 and Figure 2). These factors increase the risk ofascending infection, fetal injury, or preterm birth.

Recently, significant effort has been dedicated to measuring the global rates of GBS coloniza-tion (Box 1) [15,16], invasive disease [8,17,18], and related risk factors [19,20]. These studiesshow that increased GBS colonization in many low-income countries correlates with increasedneonatal infection and preterm birth [11]. Additionally, women of African descent have a higherincidence of GBS vaginal colonization [15,21,22] and neonatal disease [23–25]. In the USA andmany other countries, women are routinely screened in the late third trimester (between 35 and37 weeks' gestation) for GBS colonization by rectovaginal swab and subsequent culture [26]. Ifthe rectovaginal swab is culture-positive, or if the patient has GBS in the urine, or has a priorhistory of GBS perinatal infection, intrapartum prophylactic antibiotics are administered toprevent vertical transmission of GBS to the neonate during labor and delivery. Unlike the USA,some countries have not adopted the GBS screening program but instead administer anti-biotics upon the development of a risk factor for GBS neonatal disease (e.g., prolonged ruptureof membranes) [26]. However, these approaches have not fully eliminated neonatal GBSinfections. This is because these prevention strategies do not address the risk of ascendinginfection, which can potentially occur anytime during pregnancy, leading to preterm birth orstillbirth. Also, these approaches do not prevent late-onset GBS infections (observed in neo-nates who are older than 1 week of age) where vertical transmission is not the only mode ofacquisition [27]. Overall, prevention of GBS infection in pregnancy is still a complex question,with risk likely imparted by several factors, including: pathogenicity of the GBS strain, hostfactors, influence of the vaginal/rectal microbiome, false-negative screening results, and/orchanges in GBS antibiotic resistance. As current interventions targeting GBS infections arelimited to antibiotic therapy, and given that antibiotic resistance is on the rise [28], a deeperunderstanding of how GBS is able to colonize the vagina and cause neonatal disease is criticalfor the development of new therapeutics. Recently, a number of studies have described hostand bacterial factors important for GBS infections during pregnancy. In this review, we discussrecent advancements in GBS pregnancy-associated infections and therapeutic strategies.

920 Trends in Microbiology, November 2017, Vol. 25, No. 11

Downloaded for Anonymous User (n/a) at University of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 3: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

Box 1. Limited Knowledge of Colonization Prevalence and Risk

While regional Group B streptococcus (GBS)G vaginal colonization rates have been estimated [1], colonization rates inmany individual countries are unknown. Estimates of GBS vaginal colonization in different countries can be highlyvariable, with interstudy rates differing by as much as 20% [1]. This variability may be due to differences betweensubregions of large countries (e.g., India) or the means of diagnosing colonization (culture-based methods vs. PCR-based methods vs. serology-based methods). More comprehensive country-based and subregional-based studies arenecessary to fully understand the burden of GBS colonization. New studies should focus on areas where preterm birthrates and neonatal mortality rates are especially high, such as sub-Saharan Africa or south Asia. Additionally, fewstudies provide information about GBS serotype prevalence, colonization load, antibiotic-resistance profiles, or valuablegenetic information, such as virulence gene prevalence. While diagnostic technologies exist to evaluate these indicators,they can be time-consuming, expensive, technically challenging, and overall impractical. Diagnostic methodology forGBS colonization has not advanced as rapidly as our understanding of the disease itself, and new technologies need tobe developed to garner more information from future studies to further refine our knowledge of GBS colonization. Finally,more studies need to be designed to conclusively identify risk factors for GBS colonization, ascending infection, andGBS-associated preterm birth. It is clear that previous GBS colonization is a risk factor for colonization during asubsequent pregnancy [2], but few risk factors for initial GBS colonization have been identified. Recent studies haveidentified obesity [3,4] and black ethnicity as possible risk factors for colonization [9]. Future studies should be designedto identify population characteristics beyond ethnic demography and age of GBS-colonized women in an effort toimprove our ability to identify at-risk individuals. Ultimately, universal screening programs are needed in more countriesto measure the burden of GBS colonization and successfully prevent disease.

Mechanisms for GBS Infection during PregnancyIt is long known that GBS vaginal colonization during pregnancy is associated with increasedrates of neonatal infection [29], recurrent maternal colonization [30], early-term birth (weeks37–38, and 6 days of gestation) [31], preterm birth (weeks 14–36, and 6 days of gestation) [3],and stillbirth [32]. GBS is thought to be transmitted from person to person via multiple routes,including fecal–oral, sexual, and vertical transmission [33]. In the same woman, the closeproximity of the vagina and rectum likely enables GBS trafficking from intestinal flora into thevagina. Once GBS enters the vagina, colonization requires the bacteria to overcome a numberof challenges, including: physical barriers created by the mucus and epithelial layers, lowenvironmental pH, antimicrobial peptides, antibodies, microbicidal immune cells, and a vaginalmicrobiome dominated by lactobacilli.

How a nonmotile bacterium, such as GBS, manages to ascend into the uterus while evadinghost responses is not completely understood. The process of ascending infection is challeng-ing to study, as in vitro models are unsuitable, and in vivo models are limited in their ability torecapitulate human pregnancy [34,35]. Despite these limitations, a number of animal models,including pregnant mice and nonhuman primates, have been developed to study the mecha-nisms of ascending GBS infection [36–42], shedding new light on these complicated pro-cesses. While studies using these models have revealed a novel insight into the role of virulencefactors that contribute to ascending infection, more research is needed to fully understand theprocess of ascending GBS infection and adverse neonatal outcomes.

The host immune response evoked in the placenta in response to GBS infection is a keydeterminant of perinatal outcome, microbial invasion of the amniotic cavity (MIAC), and fetalinjury. A variety of fetal and maternal cells within the placental membranes are capable ofpathogen recognition for initiating and sustaining an inflammatory response; these includeamniotic epithelial cells, fetal macrophages, decidual macrophages, decidual NK cells, andneutrophils [9,39,43–46]. While a severe infection leading to early preterm birth is typicallyassociated with MIAC, an inflammatory response confined to the placenta even in the absenceof MIAC is also sufficient to induce preterm labor in some cases [38]. Interestingly, intra-amniotic administration of cytokines, such as tumor necrosis factor-alpha (TNF-a) and inter-leukin (IL)-1b alone (i.e., without any bacteria), can induce preterm labor in pregnant nonhumanprimates [47], and IL-1a, IL-1b, IL-6, and IL-8 drive infection-associated preterm birth inhumans (reviewed in [9,48]). Thus, placental inflammation induced by bacterial infection is

Trends in Microbiology, November 2017, Vol. 25, No. 11 921

Downloaded for Anonymous User (n/a) at University of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 4: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

likely a critical component of infection-associated preterm birth. Also, bacterial suppression ofplacental immune responses could contribute to MIAC, leading to stillbirths. A better under-standing of the mechanisms by which in utero GBS infections drive preterm births or stillbirthsmay lead to the development of new interventions to reduce the burden of disease. Below, wedescribe key bacterial and host factors that have been identified to influence GBS colonizationand perinatal infection.

Bacterial Factors That Promote GBS Vaginal Colonization, AscendingInfection, and Preterm BirthAdherence and Invasion FactorsGBS encodes a number of virulence factors that allow it to persist in the harsh vaginalenvironment and avoid clearance (Table 1). Many of these factors are involved in adherenceto, and invasion of, host epithelial cells that enable persistent colonization [49]. Adherence andinvasion appear to be mediated by GBS interactions with host extracellular matrix components(ECMs); these interactions may also promote GBS resistance to mechanical clearance,avoidance of immune surveillance, and paracellular transmigration [50]. A few examples ofGBS interaction with host ECMs are discussed below. The GBS extracellular protein BsaB(bacterial surface adhesin of GBS, also known as FbsC) interacts with host laminin [51] andfibrinogen [52], leading to increased adherence to cervicovaginal epithelial cells and biofilmformation [51,52]. The GBS Srr (serine-rich repeat) family of glycoproteins binds to epithelialcells [53] and interacts with host fibrinogen through a unique ‘dock, lock, and latch’ mechanism[54]. Fibrinogen binding leads to an ordered series of conformational changes in Srr1 and Srr2

Table 1. GBS Factors Involved in Vaginal Colonization, Ascending Infection, or Preterm Birth

Virulence factor Host target Function Phenotype/models Refs

HylB Hyaluronic acid Blocks TLR2/4 signaling Mouse vaginal colonization and ascendingInfection

[37,42]

BsaB/FbsC Fibrinogen and laminin Adherence to vaginal epithelial cells Immortalized human cell line [51,52]

Hemolytic pigment Amnion epithelial cellsNeutrophilsMast cellsMacrophages

CytolysisResistance to neutrophilsMast cell degranulationPyroptosis

Mouse vaginal colonization, humanchoriomamnion/placenta, nonhuman primatemodelPrimary human cellsImmortalized human cell line/s

[36,39,46,62,67,69,103]

Srr1/Srr2 Fibrinogen Adherence to vaginal epithelial cells,cervical epithelial cells

Mouse vaginal colonization, immortalized humancell line

[53,55,56,134]

Pili Collagen I Adherence to vaginal epithelial cells Mouse vaginal colonization, immortalized humancell line

[55]

Capsule Siglecsa Adherence to and invasionof cervical epithelial cellsBlunts siglec signaling inamnion epithelial cellsand neutrophils

Immortalized human cell linePrimary human cells

[50][98]

Alpha C protein Host cell surfaceglycosaminoglycan

Invasion of cervical epithelial cells Immortalized human cell line [59]

Two-component system External signal Function Model Refs

CovR/S pH Regulates hemolytic pigmentexpression (apart from other factors),adherence to vaginal epithelial cellsand cervical epithelial cells

Mouse vaginal colonization, immortalized humancell line

[68,69,72]

FspS/R Fructose 6-phosphate Vaginal persistence Mouse vaginal colonization [135]

aSiglecs, Sialic acid-binding immunoglobulin-type lectins.

922 Trends in Microbiology, November 2017, Vol. 25, No. 11

Downloaded for Anonymous User (n/a) at University of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 5: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

that result in enhanced adherence [54]. Deletion of the entire Srr1 glycoprotein, or only the latchdomain of Srr1, decreases vaginal colonization [55,56]. The GBS pili also mediate adherenceduring vaginal colonization via the binding of the PilA adhesin to host cell molecules. However,there is a discrepancy in our understanding of the nature of the host cell molecules whereinsome studies indicated that the PilA adhesion binds collagen type 1 [55,57] but others indicatedthat GBS clinical isolates do not bind collagen type 1 but rather bind to fibrinogen [58]. Furtherstudies will provide more insight into these factors during in vivo GBS colonization and infection.Finally, the GBS Alpha C protein, which contains a glycosaminoglycan-binding domain, isthought to mediate GBS invasion of cervical epithelial cells [59,60]; however, the specificglycosaminoglycan that binds Alpha C is not known. GBS invasion in endometrial cells ismediated via lipid rafts and signaling through phosphoinositide 3-kinase [61]. While clinical GBSisolates have been shown to be able to interact with ECM molecules [56,58] (Table 1),interactions between GBS and host ECM components during human vaginal colonizationare unknown. A better understanding of factors that regulate GBS vaginal colonization isnecessary for the development of preventive therapies. Given that there is no known benefit forhumans to be vaginally colonized by GBS, elimination of colonization during pregnancy is idealfor the prevention of GBS disease.

Hemolytic PigmentGBS is a b-hemolytic bacterium, and the hemolytic property of GBS is important for infectionand immune evasion. Hemolytic activity of GBS is due to the ornithine rhamnolipid pigment(hereafter referred to as‘hemolytic pigment’ or ‘pigment’) [62], which is produced by the genesof the cyl operon [63]. Transcription of cyl genes, and therefore production of the hemolyticpigment, is negatively regulated by the CovR/S two-component system (also known as CsrR/S) [62,64,65]. Consequently, deletion of covR/S renders GBS hyperhemolytic and hyperpig-mented [62,64,65]. Conversely, deletion of the cylE gene, which encodes an N-acyltransferasenecessary for pigment production [62], renders GBS nonpigmented and nonhemolytic [62,63].Identification of hemolytic, hyperhemolytic and nonhemolytic GBS strains in human casesallowed for a greater understanding of the role of hemolysin in GBS infection.

Whidbey et al. reported that the hemolytic pigment promotes GBS penetration of humanplacenta (chorioamniotic membranes) and induces loss of barrier function in human amnioticepithelial cells [62]. Furthermore, hyperpigmented GBS strains were isolated from either theamniotic fluid or chorioamniotic membranes of women in preterm labor [62]. Randis et al. alsonoted decreased bacterial dissemination, fetal injury, and preterm birth in mice that werevaginally inoculated with nonhemolytic GBS (i.e., GBS lacking cylE) [36]. Recently, Boldenowet al. showed that the increased hemolytic pigment expression accelerated GBS invasion of theamniotic cavity with significant uterine contractions and inflammatory responses indicative ofpreterm labor in a nonhuman primate model [39]. Although infection with hyperpigmented GBSinduced the formation of neutrophil extracellular traps (NETs) in chorioamniotic membranes ofnonhuman primates [39], these GBS strains were resistant to the antimicrobial activity of NETs,likely through increased pigment-mediated antioxidant activity [66]. Formation of NETs inresponse to GBS infection was also observed in murine models of colonization [67] andascending infection [41]. Collectively, these studies indicate a role for the hemolytic pigmentin promoting GBS dissemination in uterine, placental, and fetal tissues during pregnancy.

The GBS hemolytic pigment also affects vaginal colonization. Absence of the hemolyticpigment reduced the ability of GBS to successfully colonize the vagina, possibly due toincreased susceptibility to neutrophil clearance [36,67]. Surprisingly, hyperpigmented GBSalso exhibited decreased vaginal colonization in mice ([68,69]; see [70] for a visual of the murinevaginal model), likely due to increased pigment-mediated stimulation of neutrophil [68] andmast cell [69] inflammatory pathways. Consistent with these observations, hyperpigmented

Trends in Microbiology, November 2017, Vol. 25, No. 11 923

Downloaded for Anonymous User (n/a) at University of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 6: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

strains of GBS were rarely isolated from rectovaginal swabs of asymptomatic pregnant women[69]. These results emphasize the role of vaginal immune responses in pathogen colonization.

Apart from immune cells, pH regulates GBS gene expression and therefore influences vaginalcolonization. For instance, the GBS CovR/S system responds to pH wherein increased CovR/Sregulation was observed under low (acidic) pH [65,71–73]. Changes in pH also influence GBSadhesion [72,74], survival [75], and biofilm formation [76,77]. Of note, high vaginal pH and anon-lactobacilli-dominated vaginal microbiome [78] were associated with a higher incidence ofGBS vaginal colonization [15,21,22] and neonatal disease [23–25] in women of African descent. These studies indicate that GBS responds to environmental cues, such as pH, and evenutilizes regulatory systems such as CovR/S to temporally control virulence factor expression(e.g., hemolytic pigment) during pregnancy-associated infections. As such, this makes the GBSpigment an intriguing target for vaccine development.

HyaluronidaseThe GBS hyaluronidase, known as HylB, promotes vaginal colonization [42]. HylB is secretedby GBS and specifically targets and degrades host hyaluronic acid [79,80]. Hyaluronic acid isan extracellular matrix glycosaminoglycan composed of repeating disaccharide units (N-acetyl-D-glucosamine-D-glucuronic acid) and is important for cell migration, cell signaling, regulation ofinflammation [81], and the prevention of ascending infection [82,83]. GBS HylB degrades hosthyaluronic acid into its disaccharide components, which are immunosuppressive as they bindto TLR2/TLR4 receptors and block signaling [42]. Deletion of hylB led to increased clearance ofGBS from the mouse vagina [42]. Similarly, GBS lacking HylB was less able to ascend from thevagina to the uterus and was diminished for its ability to invade fetal tissues and cause pretermbirth [37]. By contrast, uterine tissue infected with HylB-proficient GBS showed decreasedlevels of inflammatory cytokines such as TNF-a, IL-6, and IL-8, leading to bacterial ascension[37]. Thus, suppression of key inflammatory responses also plays an important role in GBSinfection-associated fetal injury.

Other Virulence FactorsRecent studies have described a role for extracellular membrane vesicles (MVs) in the weak-ening of placental membranes [84]. GBS MVs contained multiple virulence factors, including: (i)HylB; (ii) CAMP factor (Christine, Atkins, Munch-Peterson factor [85]), a secreted pore-formingprotein [86] that may amplify [87], but is not essential for, GBS virulence [88]; (iii) IgA-bindingprotein, with the ability to bind human IgA [89] for host immune evasion [90], and (iv) multipleenzymes that may regulate ECM degradation [84]. Intra-amniotic administration of GBS MVs inpregnant mice caused significant damage to choriodecidual tissues and stimulated leukocyticinfiltration and inflammation, leading to membrane weakening [84]. The specific role played byeach virulence factor in the context of MVs remains unknown. MV weakening of choriodecidualmembranes represents a novel mechanism of GBS fetal injury.

Host Determinants of GBS Vaginal Colonization, Ascending Infection, andPreterm BirthVaginal ColonizationEvasion of the host immune response is essential for successful vaginal colonization. GBSvaginal immunity is mediated by its ability to resist many physical and cellular barriers, includingthe luminal mucus layer, vaginal epithelia, and immune cells in the vagina. Recent work, usinganimal models, has provided new information regarding the host immune response to GBSvaginal colonization [49,67–69]. Vaginal immune responses to GBS are largely mediated byneutrophils [49,67,68], mast cells [69], and macrophages [67] (Figure 2). The role of NK cellsand dendritic cells in GBS colonization is not known. Multiple soluble inflammatory cytokinesand chemokines have been identified as important for reducing GBS vaginal colonization and

924 Trends in Microbiology, November 2017, Vol. 25, No. 11

Downloaded for Anonymous User (n/a) at University of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 7: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

Epithelium

Lumen

Neutrophils

Macrophages

Mast cellsGroup Bstreptococcus

Degranula�on IL-5IL-8

TNFαHistamine

IL-1βIL-6IL-8

TNFα

Blocked TLR2/4

NLRP3 ac�va�on

NETforma�on

KEY:

HyIB

Hyaluronic acid

TLR2/4

Figure 2. Interaction of Group B Streptococcus (GBS) with Innate Immune Cells during Genital Infection.Upon primary vaginal colonization, vagina-resident mast cells are activated and degranulated through the hemolyticactivity of the GBS pigment. Degranulation leads to the release of inflammatory mediators, including IL-6, IL-8, TNF-a, andhistamine, which, in turn, recruit other immune cells, such as neutrophils and macrophages, to aid in bacterial clearance[69]. Neutrophils clear GBS through phagocytosis and extrusion of neutrophil extracellular traps (NETs) [67]. Additionally,GBS activates the NLRP3 inflammasome in neutrophils, leading to secretion of IL-1b, as well as other inflammatorycytokines. GBS is able to use the pigment to prevent phagocytic death through sequestration of reactive oxygen species[66], and to avoid being killed by NETs [39]. Macrophages also aid in GBS clearance through phagocytosis. GBS, in turn,activates the NLRP3 inflammasome in macrophages through membrane permeabilization by pigment, leading topyroptosis and fetal damage during pregnancy [46]. Abbreviations: IL, interleukin; TNF-a, tumor necrosis factor-alpha.

include IL-1b, IL-6, IL-8, IL-17, IL-23, and histamine [49,67,68]. Currently, the mucosal T cellresponse to GBS colonization is ill-defined. Studies have shown that IL-17 and IL-17+ cells playan important role in clearance of a hyperadherent and invasive GBS strain from the vagina [49],suggesting that the Th17 differentiation pathway is important for controlling persistent GBScolonization. Similarly, another study found cytokines involved in Th1, Th2, and Th17 differentia-tion pathways as important for decreased colonization [67]; however, T cells were not directlyidentified as being important in either study.

Trends in Microbiology, November 2017, Vol. 25, No. 11 925

Downloaded for Anonymous User (n/a) at University of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 8: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

Infection of Placental MembranesMany studies have focused on GBS infection of the placental membranes (chorioamnion) inorder to understand how GBS penetrates these barriers and induces chorioamnionitis. GBS isable to adhere to and invade both chorionic and amniotic epithelial cells [62,91]. Adherence andinvasion to these cells are mediated by several factors: (i) IagA, a glycosyltransferase that helpsanchor lipoteichoic acid to the cell surface [92]; (ii) the hemolytic pigment and its regulatorCovR/S [62]; and (iii) quorum sensing mediated by genes in the rgf operon [93]. GBS has alsobeen shown to induce secretion of multiple cytokines and defensins from placental membranesex vivo, including TNF-a, IL-1a, IL-1b, IL-6, and IL-8 [44,62,94–96]. Inflammation is stimulatedeither through pattern-recognition receptor (PRR) sensing of GBS antigens [44,94] or bypigment-mediated activation of nuclear factor-kB (NF-kB) [62]. An important family of PRRsthat mediate placental membrane immunity are the Siglecs [97,98], a family of cell-surface sialicacid-binding lectins that regulate innate and adaptive immune function [99]. GBS is able to bindSiglecs through the sialic acid capsule or b-protein to suppress immune cell activation [98,100–102] and placental membrane inflammation [98], potentially leading to increased rates ofGBS-associated preterm birth and stillbirth.

Fetal InjuryGBS invasion of the fetus in utero leads to a variety of adverse outcomes, including tissuedamage, inflammation, lung and brain injury, pneumonia, meningitis, sepsis, and fetal death.GBS can invade multiple fetal organs, including the lung, blood, liver, spleen, and gastrovas-cular cavity. Fetal tissue damage has been observed in the presence and absence of bacterialinvasion, which may be due to inflammation in the gestational tissues and amniotic fluid. GBSinvasion of fetal tissues induces inflammation and fetal death [32,36,37,46], and in the case ofthe hemolytic pigment, this involved induction of the NLRP3 inflammasome [46]. GBS stim-ulates the NLRP3 inflammasome in a number of immune cells, such as dendritic cells [103],macrophages [46], and neutrophils [104], which contribute to in vivo inflammation.

Interestingly, fetal injury is not entirely dependent on bacterial invasion of fetal tissues. Fetal lunginjury can also be caused by GBS-induced chorioamnionitis without bacterial invasion [38,105].Also, increases in amniotic fluid cytokines contribute to fetal lung injury [38] and dysregulation offetal lung development [105,106]. Moreover, intra-amniotic administration of MVs leads tosignificantly increased rates of fetal damage and preterm birth [84]. These studies suggest thatfetal injury can occur during transient or limited infection, and further emphasizes the impor-tance of developing therapeutics that prevent vaginal colonization and ascending infection.

Limitations and Next StepsWhile significant research has been committed to understanding the host and bacterial factorsinvolved in GBS vaginal colonization and ascending infection, further research is needed to fullygrasp these complex processes. Additionally, the research that has been completed is notwithout its limitations. These limitations and the next steps for research are discussed below.

Vaginal ColonizationDespite recent advances, there are many limitations in our understanding of GBS vaginalcolonization. Importantly, the complete repertoire of GBS virulence factors, host factors, andenvironmental factors that determine the extent and duration of vaginal colonization remainunknown. This point is key, given that the dynamics of colonization during pregnancy are highlyvariable [107]. In addition, experiments performed in laboratory settings are limited by the use ofanimal models that do not perfectly model the human vaginal environment. To fully understandGBS vaginal colonization, experimental models and outputs that more closely represent humandisease are needed. Finally, it is often assumed that neonates contract GBS from contaminatedvaginal fluids during the birthing process; however, given the kinetics of neonatal disease

926 Trends in Microbiology, November 2017, Vol. 25, No. 11

Downloaded for Anonymous User (n/a) at University of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 9: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

(frequently displaying clinical symptoms within 0–24 h of birth [108]), many cases are thought tobe a result of in utero infections caused by ascended GBS. This idea is also supported by thefrequent recovery of GBS from the amniotic fluid and chorioamniotic membranes in cases ofinfection-associated preterm birth [1,109,110]. Given its important implication for understand-ing GBS disease, colonization is an area of active research.

Ascension from the Vagina to the UterusWhile studies have provided some insight into virulence factors that enable GBS to gain accessto the uterine space from the lower genital tract, and host factors involved in this process,significant research is required to fully understand ascending GBS infection. Currently, little isknown about host factors that prevent ascending infection, such as the role of the cervicalbarrier, endocrine signaling, and cellular immunity. Further, host-specific factors and geneticsthat may influence ascending GBS infection are unknown. Studies aimed at filling theseknowledge gaps could lead to the development of therapeutics for preventing ascendinginfection. The choice of animal model is key in answering some of these questions due toimportant differences in pregnancy physiology, mechanism of labor, and placental structurebetween humans and mice [111]. Nonhuman primates are the closest animal model to fullyrecapitulate important aspects of human pregnancy [111] but are limited in their use by ethicalconstraints, availability, and cost. Ideally, a combination of lower animal and nonhuman primatemodels should be used in order to delineate relevant aspects of disease. Finally, clinical studiesshould be designed to identify biomarkers for ascending infection. Viral infection of the cervix[112] and diminished cervical hyaluronic acid levels [83] have been associated with increasedascending infection; however, little is known about the clinical relevance of these or otherfactors in the context of ascending GBS infection.

GBS Prevention and TherapeuticsCurrently, strategies focus on the prevention of GBS transmission during labor and deliverythrough the use of antibiotics. This strategy does not fully capture the biology of GBS infection,nor does it completely address the full burden of GBS disease. Moreover, antibiotic resistanceis increasing [28], and the use of antibiotics during pregnancy has consequential effects forneonatal health that are only now being appreciated [113,114]. To successfully eradicate theburden of disease, interventions need to be specifically targeted, have minimal detrimentaleffects on the microbiome, and target processes upstream of vertical transmission, such ascolonization and ascending infection.

Multiple studies have focused on a probiotic approach to reducing vaginal GBS colonization.Recent studies using probiotic Lactobacillus species have shown that pretreatment of thevagina can block GBS adherence to vaginal epithelial cells [115], and reduce colonization[116,117]. Probiotic administration of the oral colonizer Streptococcus salivarius has also beenshown to have the ability to reduce vaginal GBS burden through a yet unidentified antimicrobialactivity [118]. It is important to note that frequent doses (from three to seven) of probioticbacteria were required for a protective effect in vivo [116,117], raising the question as to thefeasibility of probiotic intervention in humans, and highlighting the need to continue to explorethis field to identify an efficient and feasible probiotic therapy to prevent GBS colonization.Moreover, little is known about the interactions between GBS and the vaginal microbiome, andwhether those interactions have any positive benefits to human health. Further studies that aimto understand these interactions would shed much needed light on this topic.

A vaccine to prevent GBS colonization would be the most effective intervention; however,development of a vaccine has proven to be challenging. Recent work has shown that vaccinationof mice with killed bacteria reduces preterm birth rates [119] and that mucosal vaccine delivery ismore effective than intramuscular delivery [120]. Multiple studies have identified maternal antibody

Trends in Microbiology, November 2017, Vol. 25, No. 11 927

Downloaded for Anonymous User (n/a) at University of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 10: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

Outstanding QuestionsWhy are humans intermittently colo-nized with GBS? What determineswhether a GBS strain will colonizethe vagina? How can we eliminateGBS without adverse consequenceson beneficial microbiota?

What factors determine the ability of aGBS strain to ascend into the uterusand invade the placenta and amnioticcavity? Is it environmental factors in thelower genital tract during pregnancy?

How do geographic differences in hostgenetics, environment, and diet con-tribute to differences in the vaginalmicrobiome and GBS colonization?

Can we determine which GBS strainsare likely to invade the amniotic cavityor cause late-onset neonatal sepsis tobetter target antibiotic therapy to ahigher-risk group of pregnant women?

levels to the GBS capsule as being protective against GBS infection [121–124], and it has longbeen known that anticapsular antibodies can confer protection to infection [5]. Unfortunately,vaccines targeting the GBS capsule alone are ineffective due to their poor immunogenicity andthus, conjugate–capsule vaccines, and a vaccine that targets the alpha C/Rib protein family, arenow being tested in clinical trials (reviewed in [125–127]). Despite the promise of these vaccinecandidates,challenges to the eradicationofGBSdisease will existevenafter the implementation ofa vaccine. While ten GBS capsular serotypes have been identified, serotypes Ia, Ib, II, III, and V arepredominantly responsible for GBS disease [127]. Consequently, current vaccine candidatestarget serotypes Ia, Ib, and III, [125], and a pentavalent vaccine targeting serotypes Ia, Ib, II, III, andV is in preclinical development [126]. The development of vaccines targeting various antigens,such as Alpha C/Rib, may prove to be vital, as nontypeable GBS strains can cause disease [128].Additionally, GBS strains can switch capsular serotypes [129], thereby evading host immunityconferred by vaccination.

It is possible that if these vaccines are developed, serotypes or strains that are not typically asignificant cause of GBS disease may emerge in vaccinated populations. Indeed, this phenome-non has been observed with implementation of conjugate vaccines against Streptococcuspneumoniae [130,131]. Thus, GBS surveillance programs would need to remain vigilant evenafter a vaccine becomes widely used. There are also significant challenges in resource-limitedsettingstoconsiderprior to implementation (reviewed in [132]).Regardless, it isclear thata vaccinewould be the most effective means of reducing the global burden of GBS disease [133].

Concluding RemarksGBS has long been recognized as a significant human pathogen [6], yet we are only nowbeginning to fully understand its pathogenesis almost a century later. It is clear that under-standing the interplay between the host and bacteria is vital for understanding how to effectivelyprevent GBS disease while having minimal adverse effects on the human host. Research in thisfield has revealed novel insights into the bacterial virulence factors necessary for successfullyestablishing disease (Table 1) and an appropriate host response to prevent ascending infectionand preterm birth (Figures 1 and 2). Any disturbance in this balance can lead to serious andlasting outcomes for the host, or disadvantageous pressures for the bacteria. Several examplesof how this balance can be perturbed to benefit human health have been described above, butmore research is needed to fully understand what triggers ascending GBS infection, the hostimmune response to colonization and ascending infection, and the physiological drivers of fetaldamage and preterm birth (see Outstanding Questions). Additionally, more epidemiologicaldata are required to describe the global burden of GBS colonization and disease (Box 1). AsGBS colonization during pregnancy is intermittent and variable [107], the current screeningmethodology likely misses a significant portion of colonization during pregnancy. This issueresults in a measurable risk for ascending GBS infection during pregnancy that could lead tostillbirth, preterm birth, and early-term birth. An evaluation of earlier and more frequent GBSscreening during pregnancy may shed light on these issues. With these data, effective andfeasible interventions to prevent GBS disease can be developed. Ultimately, vaccination willprove to be the most effect intervention. A combination of rational vaccine design, intelligentimplementation and monitoring strategies, and strong advocacy [26] may lead to the eradica-tion of GBS as a human pathogen.

AcknowledgmentsWe thank Jan Hamanishi for assistance with graphical design. We apologize to GBS researchers whose work was not

reviewed here due to constraints in space and/or focus of the topic. We acknowledge funding from the National Institutes

of Health, Grant R01AI100989 to L.R and K. A. W., and R21AI125907 and R01AI112619 to L.R. The NIH training grant

T32 AI07509 (PI: Lee Ann Campbell) supported J.V. The content in this manuscript is the responsibility of the authors and

does not necessarily represent the views of the National Institutes of Health.

928 Trends in Microbiology, November 2017, Vol. 25, No. 11

Downloaded for Anonymous User (n/a) at University of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 11: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

References

1. Hillier, S.L. et al. (1991) Microbiologic causes and neonatal

outcomes associated with chorioamnion infection. Am. J.Obstet. Gynecol. 165, 955–961

2. Allen, U. et al. (1999) Relationship between antenatal group Bstreptococcal vaginal colonization and premature labour.Paediatr. Child Health 4, 465–469

3. Lawn, J.E. et al. (2010) Global report on preterm birth andstillbirth (1 of 7): definitions, description of the burden andopportunities to improve data. BMC Pregnancy Childbirth 10(Suppl. 1), S1

4. Norcard, N. and Mollereau, R. (1887) Sur une mammite conta-gieuse des vaches laitieres. Ann. Inst. Pasteur 1, 109–126 (inFrench)

5. Lancefield, R.C. and Hare, R. (1935) The serological differentia-tion of pathogenic and non-pathogenic strains of hemolyticstreptococci from parturient women. J. Exp. Med. 61, 335–349

6. Fry, R.M. (1938) Fatal infections by hemolytic Streptococcusgroup B. Lancet 1, 199–201

7. Le Doare, K. and Heath, P.T. (2013) An overview of global GBSepidemiology. Vaccine 31 (Suppl. 4), D7–D12

8. Stoll, B.J. et al. (2011) Early onset neonatal sepsis: the burden ofgroup B streptococcal and E. coli disease continues. Pediatrics127, 817–826

9. Romero, R. et al. (2014) Preterm labor: one syndrome, manycauses. Science 345, 760–765

10. Liu, L. et al. (2015) Global, regional, and national causes of childmortality in 2000–13, with projections to inform post-2015 pri-orities: an updated systematic analysis. Lancet 385, 430–440

11. Mokdad, A.H. et al. (2016) Global burden of diseases, injuries,and risk factors for young people’s health during 1990-2013: asystematic analysis for the Global Burden of Disease Study2013. Lancet 387, 2383–2401

12. Hitti, J. et al. (2001) Amniotic fluid infection, cytokines, andadverse outcome among infants at 34 weeks’ gestation or less.Obstet. Gynecol. 98, 1080–1088

13. DiGiulio, D.B. et al. (2008) Microbial prevalence, diversity andabundance in amniotic fluid during preterm labor: a molecularand culture-based investigation. PLoS One 3, e3056

14. Han, Y.W. et al. (2009) Uncultivated bacteria as etiologic agentsof intra-amniotic inflammation leading to preterm birth. J. Clin.Microbiol. 47, 38–47

15. Kwatra, G. et al. (2016) Prevalence of maternal colonisation withGroup B Streptococcus: a systematic review and meta-analysis.Lancet Infect. Dis. 16, 1076–1084

16. Barcaite, E. et al. (2008) Prevalence of maternal group B strep-tococcal colonisation in European countries. Acta Obstet. Gyne-col. Scand. 87, 260–271

17. Nan, C. et al. (2015) Maternal Group B Streptococcus-relatedstillbirth: a systematic review. BJOG 122, 1437–1445

18. Petersen, K.B. et al. (2014) Increasing prevalence of group Bstreptococcal infection among pregnant women. Dan. Med. J.61, A4908

19. Kleweis, S.M. et al. (2015) Maternal obesity and rectovaginalGroup B Streptococcus colonization at term. Infect. Dis. Obstet.Gynecol. 2015, 586767

20. Colicchia, L.C. et al. (2015) Recurrence of Group B Streptococ-cus colonization in successive pregnancies. J. Perinatol. 35,173–176

21. Hickman, M.E. et al. (1999) Changing epidemiology of group Bstreptococcal colonization. Pediatrics 104 (2 Pt 1), 203–209

22. Taylor, J.K. et al. (2002) The incidence of Group B Streptococ-cus in the vaginal tracts of pregnant women in central Alabama.Clin. Lab. Sci. 15, 16–17

23. Lin, F.Y. et al. (2003) Prematurity is the major risk factor for late-onset Group B Streptococcus disease. J. Infect. Dis. 188, 267–271

24. Peltier, M.R. et al. (2012) Amniotic fluid and maternal raceinfluence responsiveness of fetal membranes to bacteria.J. Reprod. Immunol. 96, 68–78

Downloaded for Anonymous User (n/a) at UniversiFor personal use only. No other

25. Weston, E.J. et al. (2011) The burden of invasive early-onsetneonatal sepsis in the United States, 2005–2008. Pediatr. Infect.Dis. J. 30, 937–941

26. Burns, G. and Plumb, J. (2013) GBS public awareness,advocacy, and prevention – what’s working, what’s not and whywe need a maternal GBS vaccine. Vaccine 31 (Suppl 4), D58–D65

27. Tazi, A. et al. (2010) The surface protein HvgA mediates Group BStreptococcus hypervirulence and meningeal tropism in neo-nates. J. Exp. Med. 207, 2313–2322

28. Castor, M.L. et al. (2008) Antibiotic resistance patterns in inva-sive group B streptococcal isolates. Infect. Dis. Obstet. Gynecol.2008, 727505

29. Hoogkamp-Korstanje, J.A. et al. (1982) Maternal carriage andneonatal acquisition of group B streptococci. J. Infect. Dis. 145,800–803

30. Cheng, P.J. et al. (2008) Risk factors for recurrence of Group BStreptococcus colonization in a subsequent pregnancy. Obstet.Gynecol. 111, 704–709

31. Mitchell, K. et al. (2013) Group B Streptococcus colonizationand higher maternal IL-1beta concentrations are associatedwith early term births. J. Matern. Fetal Neonatal Med. 26, 56–61

32. Monari, F. et al. (2013) Fetal bacterial infections in antepartumstillbirth: a case series. Early Hum. Dev. 89, 1049–1054

33. Manning, S.D. et al. (2004) Prevalence of Group B Streptococ-cus colonization and potential for transmission by casual con-tact in healthy young men and women. Clin. Infect. Dis. 39, 380–388

34. De Clercq, E. et al. (2013) Animal models for studying femalegenital tract infection with Chlamydia trachomatis. Infect.Immun. 81, 3060–3067

35. McDuffie, R.S. and Gibbs, R.S. (1994) Animal models ofascending genital-tract infection in pregnancy. Infect. Dis.Obstet. Gynecol. 2, 60–70

36. Randis, T.M. et al. (2014) Group B Streptococcus beta-hemo-lysin/cytolysin breaches maternal-fetal barriers to cause pretermbirth and intrauterine fetal demise in vivo. J. Infect. Dis. 210,265–273

37. Vornhagen, J. et al. (2016) Bacterial hyaluronidase promotesascending GBS infection and preterm birth. mBio 7, e00781-16

38. Adams Waldorf, K.M. et al. (2011) Choriodecidual group Bstreptococcal inoculation induces fetal lung injury withoutintra-amniotic infection and preterm labor in Macaca nemes-trina. PLoS One 6, e28972

39. Boldenow, E. et al. (2016) Group B Streptococcus circumventsneutrophils and neutrophil extracellular traps during amnioticcavity invasion and preterm labor. Sci. Immunol. 1, eaah4576

40. Ancona, R.J. and Ferrieri, P. (1979) Experimental vaginal colo-nization and mother–infant transmission of group B streptococciin rats. Infect. Immun. 26, 599–603

41. Kothary, V. et al. (2017) Group B Streptococcus inducesneutrophil recruitment to gestational tissues and elaboration ofextracellular traps and nutritional immunity. Front. Cell. Infect.Microbiol. 7, 19

42. Kolar, S.L. et al. (2015) Group B Streptococcus evades hostimmunity by degrading hyaluronan. Cell Host Microbe 18,694–704

43. Singh, U. et al. (2005) Immunological properties of humandecidual macrophages – a possible role in intrauterine immunity.Reproduction 129, 631–637

44. Boldenow, E. et al. (2015) Role of cytokine signaling in Group BStreptococcus-stimulated expression of human beta defensin-2in human extraplacental membranes. Am. J. Reprod. Immunol.73, 263–272

45. Duriez, M. et al. (2014) Human decidual macrophages and NKcells differentially express Toll-like receptors and display distinctcytokine profiles upon TLR stimulation. Front. Microbiol. 5, 316

46. Whidbey, C. et al. (2015) A streptococcal lipid toxin inducesmembrane permeabilization and pyroptosis leading to fetalinjury. EMBO Mol. Med. 7, 488–505

Trends in Microbiology, November 2017, Vol. 25, No. 11 929

ty of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017. uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 12: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

47. Sadowsky, D.W. et al. (2006) Preterm labor is induced by intra-amniotic infusions of interleukin-1beta and tumor necrosis fac-tor-alpha but not by interleukin-6 or interleukin-8 in a nonhumanprimate model. Am. J. Obstet. Gynecol. 195, 1578–1589

48. Cappelletti, M. et al. (2016) Inflammation and preterm birth.J. Leukoc. Biol. 99, 67–78

49. Patras, K.A. et al. (2015) Characterization of host immunityduring persistent vaginal colonization by Group B Streptococ-cus. Mucosal Immunol. 8, 1339–1348

50. Soriani, M. et al. (2006) Group B Streptococcus crosses humanepithelial cells by a paracellular route. J. Infect. Dis. 193, 241–250

51. Jiang, S. and Wessels, M.R. (2014) BsaB, a novel adherencefactor of Group B Streptococcus. Infect. Immun. 82, 1007–1016

52. Buscetta, M. et al. (2014) FbsC, a novel fibrinogen-bindingprotein, promotes Streptococcus agalactiae–host cell interac-tions. J. Biol. Chem. 289, 21003–21015

53. Mistou, M.Y. et al. (2009) Molecular dissection of the secA2locus of Group B Streptococcus reveals that glycosylation of theSrr1 LPXTG protein is required for full virulence. J. Bacteriol.191, 4195–4206

54. Seo, H.S. et al. (2013) Characterization of fibrinogen binding byglycoproteins Srr1 and Srr2 of Streptococcus agalactiae. J. Biol.Chem. 288, 35982–35996

55. Sheen, T.R. et al. (2011) Serine-rich repeat proteins and pilipromote Streptococcus agalactiae colonization of the vaginaltract. J. Bacteriol. 193, 6834–6842

56. Wang, N.Y. et al. (2014) Group B streptococcal serine-richrepeat proteins promote interaction with fibrinogen and vaginalcolonization. J. Infect. Dis. 210, 982–991

57. Banerjee, A. et al. (2011) Bacterial pili exploit integrin machineryto promote immune activation and efficient blood-brain barrierpenetration. Nat. Commun. 2, 462

58. Dramsi, S. et al. (2012) Epidemiologically and clinically relevantGroup B Streptococcus isolates do not bind collagen but displayenhanced binding to human fibrinogen. Microbes Infect. 14,1044–1048

59. Bolduc, G.R. et al. (2002) The alpha C protein mediates inter-nalization of Group B Streptococcus within human cervicalepithelial cells. Cell. Microbiol. 4, 751–758

60. Baron, M.J. et al. (2007) Identification of a glycosaminoglycanbinding region of the alpha C protein that mediates entry ofgroup B streptococci into host cells. J. Biol. Chem. 282, 10526–10536

61. Goluszko, P. et al. (2008) Group B Streptococcus exploits lipidrafts and phosphoinositide 3-kinase/Akt signaling pathway toinvade human endometrial cells. Am. J. Obstet. Gynecol. 199,e1–e9

62. Whidbey, C. et al. (2013) A hemolytic pigment of Group BStreptococcus allows bacterial penetration of human placenta.J. Exp. Med. 210, 1265–1281

63. Pritzlaff, C.A. et al. (2001) Genetic basis for the beta-haemolytic/cytolytic activity of Group B Streptococcus. Mol. Microbiol. 39,236–247

64. Lamy, M.C. et al. (2004) CovS/CovR of Group B Streptococcus:a two-component global regulatory system involved in virulence.Mol. Microbiol. 54, 1250–1268

65. Jiang, S.M. et al. (2005) Regulation of virulence by a two-com-ponent system in Group B Streptococcus. J. Bacteriol. 187,1105–1113

66. Liu, G.Y. et al. (2004) Sword and shield: linked group B strepto-coccal beta-hemolysin/cytolysin and carotenoid pigment func-tion to subvert host phagocyte defense. Proc. Natl. Acad. Sci. U.S. A. 101, 14491–14496

67. Carey, A.J. et al. (2014) Infection and cellular defense dynamicsin a novel 17beta-estradiol murine model of chronic humanGroup B Streptococcus genital tract colonization reveal a rolefor hemolysin in persistence and neutrophil accumulation. J.Immunol. 192, 1718–1731

68. Patras, K.A. et al. (2013) Group B Streptococcus CovR regula-tion modulates host immune signalling pathways to promotevaginal colonization. Cell. Microbiol. 15, 1154–1167

930 Trends in Microbiology, November 2017, Vol. 25, No. 11

Downloaded for Anonymous User (n/a) at UniversiFor personal use only. No other

69. Gendrin, C. et al. (2015) Mast cell degranulation by a hemolyticlipid toxin decreases GBS colonization and infection. Sci. Adv. 1,e1400225

70. Patras, K.A. and Doran, K.S. (2016) A murine model of Group BStreptococcus vaginal colonization. J. Vis. Exp. Published onlineNovember 16, 2016. http://dx.doi.org/10.3791/54708

71. Cumley, N.J. et al. (2012) The CovS/CovR acid response regu-lator is required for intracellular survival of Group B Streptococ-cus in macrophages. Infect. Immun. 80, 1650–1661

72. Park, S.E. et al. (2012) CsrRS and environmental pH regulateGroup B Streptococcus adherence to human epithelial cells andextracellular matrix. Infect. Immun. 80, 3975–3984

73. Santi, I. et al. (2009) CsrRS regulates Group B Streptococcusvirulence gene expression in response to environmental pH: anew perspective on vaccine development. J. Bacteriol. 191,5387–5397

74. Tamura, G.S. et al. (1994) Adherence of group B streptococci tocultured epithelial cells: roles of environmental factors and bac-terial surface components. Infect. Immun. 62, 2450–2458

75. Borges, S. et al. (2012) Survival and biofilm formation by GroupB streptococci in simulated vaginal fluid at different pHs. AntonieVan Leeuwenhoek 101, 677–682

76. D’Urzo, N. et al. (2014) Acidic pH strongly enhances in vitrobiofilm formation by a subset of hypervirulent ST-17 Strepto-coccus agalactiae strains. Appl. Environ. Microbiol. 80, 2176–2185

77. Ho, Y.R. et al. (2013) The enhancement of biofilm formation inGroup B streptococcal isolates at vaginal pH. Med. Microbiol.Immunol. 202, 105–115

78. Ravel, J. et al. (2011) Vaginal microbiome of reproductive-agewomen. Proc. Natl. Acad. Sci. U. S. A. 108 (Suppl 1), 4680–4687

79. Gochnauer, T.A. and Wilson, J.B. (1951) The production ofhyaluronidase by Lancefield’s Group B streptococci. J. Bacter-iol. 62, 405–414

80. Baker, J.R. and Pritchard, D.G. (2000) Action pattern and sub-strate specificity of the hyaluronan lyase from group B strepto-cocci. Biochem. J. 348 (Pt 2), 465–471

81. Stern, R. et al. (2006) Hyaluronan fragments: an information-richsystem. Eur. J. Cell Biol. 85, 699–715

82. Mahendroo, M. (2012) Cervical remodeling in term and pretermbirth: insights from an animal model. Reproduction 143, 429–438

83. Akgul, Y. et al. (2014) Hyaluronan in cervical epithelia protectsagainst infection-mediated preterm birth. J. Clin. Invest. 124,5481–5489

84. Surve, M.V. et al. (2016) Membrane vesicles of Group B Strep-tococcus disrupt feto-maternal barrier leading to preterm birth.PLoS Pathog. 12, e1005816

85. Christie, R. et al. (1944) A note on a lytic phenomenon shown bygroup B streptococci. Aust. J. Exp. Biol. Med. Sci. 22, 197–200

86. Lang, S. and Palmer, M. (2003) Characterization of Streptococ-cus agalactiae CAMP factor as a pore-forming toxin. J. Biol.Chem. 278, 38167–38173

87. Jurgens, D. et al. (1987) Unspecific binding of group B strepto-coccal cocytolysin (CAMP factor) to immunoglobulins and itspossible role in pathogenicity. J. Exp. Med. 165, 720–732

88. Hensler, M.E. et al. (2008) CAMP factor is not essential forsystemic virulence of Group B Streptococcus. Microb. Pathog.44, 84–88

89. Kvam, A.I. et al. (1992) Binding of human IgA to HCl-extracted cprotein from group B streptococci (GBS). APMIS 100, 1129–1132

90. Nordstrom, T. et al. (2011) Human Siglec-5 inhibitory receptorand immunoglobulin A (IgA) have separate binding sites instreptococcal beta protein. J. Biol. Chem. 286, 33981–33991

91. Winram, S.B. et al. (1998) Characterization of group B strepto-coccal invasion of human chorion and amnion epithelial cells invitro. Infect. Immun. 66, 4932–4941

92. Doran, K.S. et al. (2005) Blood–brain barrier invasion by Group BStreptococcus depends upon proper cell-surface anchoring oflipoteichoic acid. J. Clin. Invest. 115, 2499–2507

ty of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017. uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 13: Perinatal Group B Streptococcal Infections: Virulence Factors, … · 2020-03-13 · are attributable to GBS [12–14]. The bacterial and host determinants that promote GBS vaginal

93. Parker, R.E. et al. (2017) Contribution of the RgfD quorumsensing peptide to rgf regulation and host cell association inGroup B Streptococcus. Genes (Basel) 8, 23

94. Boldenow, E. et al. (2015) The trichloroethylene metabolite S-(1,2-dichlorovinyl)-L-cysteine but not trichloroacetate inhibitspathogen-stimulated TNF-alpha in human extraplacental mem-branes in vitro. Reprod. Toxicol. 52, 1–6

95. Boldenow, E. et al. (2013) Antimicrobial peptide response toGroup B Streptococcus in human extraplacental membranes inculture. Placenta 34, 480–485

96. Flores-Herrera, H. et al. (2012) An experimental mixed bacterialinfection induced differential secretion of proinflammatory cyto-kines (IL-1beta, TNFalpha) and proMMP-9 in human fetal mem-branes. Placenta 33, 271–277

97. Brinkman-Van der Linden, E.C. et al. (2007) Human-specificexpression of Siglec-6 in the placenta. Glycobiology 17, 922–931

98. Ali, S.R. et al. (2014) Siglec-5 and Siglec-14 are polymorphicpaired receptors that modulate neutrophil and amnion signalingresponses to Group B Streptococcus. J. Exp. Med. 211, 1231–1242

99. Crocker, P.R. et al. (2007) Siglecs and their roles in the immunesystem. Nat. Rev. Immunol. 7, 255–266

100. Carlin, A.F. et al. (2009) Group B Streptococcus suppression ofphagocyte functions by protein-mediated engagement ofhuman Siglec-5. J. Exp. Med. 206, 1691–1699

101. Carlin, A.F. et al. (2007) Group B streptococcal capsular sialicacids interact with siglecs (immunoglobulin-like lectins) onhuman leukocytes. J. Bacteriol. 189, 1231–1237

102. Carlin, A.F. et al. (2009) Molecular mimicry of host sialylatedglycans allows a bacterial pathogen to engage neutrophil Siglec-9 and dampen the innate immune response. Blood 113, 3333–3336

103. Costa, A. et al. (2012) Activation of the NLRP3 inflammasome bygroup B streptococci. J. Immunol. 188, 1953–1960

104. Mohammadi, N. et al. (2016) Neutrophils directly recognizegroup b streptococci and contribute to Interleukin-1beta pro-duction during infection. PLoS One 11, e0160249

105. McAdams, R.M. et al. (2012) Choriodecidual infection down-regulates angiogenesis and morphogenesis pathways in fetallungs from Macaca nemestrina. PLoS One 7, e46863

106. McAdams, R.M. et al. (2015) Choriodecidual Group B strepto-coccal infection induces miR-155-5p in the fetal lung in Macacanemestrina. Infect. Immun. 83, 3909–3917

107. Hansen, S.M. et al. (2004) Dynamics of Streptococcus agalac-tiae colonization in women during and after pregnancy and intheir infants. J. Clin. Microbiol. 42, 83–89

108. Schrag, S.J. et al. (2016) Epidemiology of invasive early-onsetneonatal sepsis, 2005 to 2014. Pediatrics 138, e20162013

109. Romero, R. et al. (2001) The role of infection in preterm labourand delivery. Paediatr. Perinat. Epidemiol. 15 (Suppl 2), 41–56

110. Romero, R. et al. (1992) Infection and labor. VII. Microbial inva-sion of the amniotic cavity in spontaneous rupture of mem-branes at term. Am. J. Obstet. Gynecol. 166 (1 Pt 1), 129–133

111. Adams Waldorf, K.M. et al. (2011) Use of nonhuman primatemodels to investigate mechanisms of infection-associated pre-term birth. Br. J. Obstet. Gynaecol. 118, 136–144

112. Racicot, K. et al. (2013) Viral infection of the pregnant cervixpredisposes to ascending bacterial infection. J. Immunol. 191,934–941

113. Langdon, A. et al. (2016) The effects of antibiotics on the micro-biome throughout development and alternative approaches fortherapeutic modulation. Genome Med. 8, 39

114. Bokulich, N.A. et al. (2016) Antibiotics, birth mode, and dietshape microbiome maturation during early life. Sci. Transl. Med.8, 343ra82

115. Ortiz, L. et al. (2014) Effect of two probiotic strains of Lactoba-cillus on in vitro adherence of Listeria monocytogenes, Strepto-coccus agalactiae, and Staphylococcus aureus to vaginalepithelial cells. Curr. Microbiol. 68, 679–684

Downloaded for Anonymous User (n/a) at UniversiFor personal use only. No other

116. De Gregorio, P.R. et al. (2015) Preventive effect of Lactobacillusreuteri CRL1324 on Group B Streptococcus vaginal coloniza-tion in an experimental mouse model. J. Appl. Microbiol. 118,1034–1047

117. De Gregorio, P.R. et al. (2016) Immunomodulation of Lactoba-cillus reuteri CRL1324 on Group B Streptococcus vaginal colo-nization in a murine experimental model. Am. J. Reprod.Immunol. 75, 23–35

118. Patras, K.A. et al. (2015) Streptococcus salivarius K12 limitsGroup B Streptococcus vaginal colonization. Infect. Immun. 83,3438–3444

119. Bernardini, R. et al. (2016) Neonatal protection and preterm birthreduction following maternal Group B Streptococcus vaccina-tion in a mouse model. J. Matern. Fetal Neonatal Med. Pub-lished online December 14, 2016. http://dx.doi.org/10.1080/14767058.2016.1265932

120. Baker, J.A. et al. (2017) Mucosal vaccination promotes clear-ance of Streptococcus agalactiae vaginal colonization. Vaccine35, 1273–1280

121. Fabbrini, M. et al. (2016) The protective value of maternal GroupB Streptococcus antibodies: quantitative and functional analysisof naturally acquired responses to capsular polysaccharides andpilus proteins in European maternal sera. Clin. Infect. Dis. 63,746–753

122. Dangor, Z. et al. (2015) Correlates of protection of serotype-specific capsular antibody and invasive Group B Streptococcusdisease in South African infants. Vaccine 33, 6793–6799

123. Dangor, Z. et al. (2015) Association between maternal Group BStreptococcus surface-protein antibody concentrations andinvasive disease in their infants. Expert Rev. Vaccines 14,1651–1660

124. Kwatra, G. et al. (2015) Natural acquired humoral immunityagainst serotype-specific Group B Streptococcus rectovaginalcolonization acquisition in pregnant women. Clin. Microbiol.Infect. 21 (568), e13–21

125. Heath, P.T. (2016) Status of vaccine research and developmentof vaccines for GBS. Vaccine 34, 2876–2879

126. Kobayashi, M. et al. (2016) WHO consultation on Group BStreptococcus vaccine development: Report from a meetingheld on 27–28 April 2016. Vaccine Published online December22, 2016. http://dx.doi.org/10.1016/j.vaccine.2016.12.029

127. Kobayashi, M. et al. (2016) Group B Streptococcus vaccinedevelopment: present status and future considerations, withemphasis on perspectives for low and middle income countries.F1000Res. 5, 2355

128. Ramaswamy, S.V. et al. (2006) Molecular characterization ofnontypeable Group B Streptococcus. J. Clin. Microbiol. 44,2398–2403

129. Bellais, S. et al. (2012) Capsular switching in Group B Strepto-coccus CC17 hypervirulent clone: a future challenge for poly-saccharide vaccine development. J. Infect. Dis. 206, 1745–1752

130. Weinberger, D.M. et al. (2011) Serotype replacement in diseaseafter pneumococcal vaccination. Lancet 378, 1962–1973

131. Hanage, W.P. (2007) Serotype replacement in invasive pneu-mococcal disease: where do we go from here? J. Infect. Dis.196, 1282–1284

132. Nishihara, Y. et al. (2017) Challenges in reducing Group BStreptococcus disease in African settings. Arch. Dis. Child102, 72–77

133. Kim, S.Y. et al. (2014) Cost-effectiveness of a potential group Bstreptococcal vaccine program for pregnant women in SouthAfrica. Vaccine 32, 1954–1963

134. Six, A. et al. (2015) Srr2, a multifaceted adhesin expressed by ST-17 hypervirulent Group B Streptococcus involved in binding toboth fibrinogen and plasminogen. Mol. Microbiol. 97, 1209–1222

135. Faralla, C. et al. (2014) Analysis of two-component systems inGroup B Streptococcus shows that RgfAC and the novel FspSRmodulate virulence and bacterial fitness. mBio 5, e00870-14

Trends in Microbiology, November 2017, Vol. 25, No. 11 931

ty of Manitoba - Canada Consortium from ClinicalKey.com by Elsevier on November 29, 2017. uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.