Skip to main content
Clinical Microbiology Reviews logoLink to Clinical Microbiology Reviews
. 2017 Aug 16;30(4):973–989. doi: 10.1128/CMR.00019-17

Intrinsic Maturational Neonatal Immune Deficiencies and Susceptibility to Group B Streptococcus Infection

Michelle L Korir a, Shannon D Manning a, H Dele Davies b,
PMCID: PMC5608877  PMID: 28814408

SUMMARY

Although a normal member of the gastrointestinal and vaginal microbiota, group B Streptococcus (GBS) can also occasionally be the cause of highly invasive neonatal disease and is an emerging pathogen in both elderly and immunocompromised adults. Neonatal GBS infections are typically transmitted from mother to baby either in utero or during passage through the birth canal and can lead to pneumonia, sepsis, and meningitis within the first few months of life. Compared to the adult immune system, the neonatal immune system has a number of deficiencies, making neonates more susceptible to infection. Recognition of GBS by the host immune system triggers an inflammatory response to clear the pathogen. However, GBS has developed several mechanisms to evade the host immune response. A comprehensive understanding of this interplay between GBS and the host immune system will aid in the development of new preventative measures and therapeutics.

KEYWORDS: group B streptococcus, immunology, inflammation, prevention

INTRODUCTION

Group B Streptococcus (GBS) (Streptococcus agalactiae) commonly colonizes the human gastrointestinal and/or genitourinary tracts in approximately 30% of healthy adults (15). GBS can be found primarily in the outer mucus layer of the colon as well as the small intestine (6). In addition to being a commensal, GBS also causes severe disease in neonates and in elderly and immunocompromised individuals. The Active Bacterial Core Surveillance report estimates that there are 28,550 cases of invasive GBS disease resulting in approximately 1,770 deaths annually in the United States (7).

GBS is a highly diverse species and can be classified by using serotyping and multilocus sequence typing (MLST). Serotyping is based on the capsular polysaccharide (CPS) and categorizes GBS into 10 types: types Ia, Ib, and II through IX (8). These 10 antigenically distinct CPS types play a major role in GBS virulence, with types Ia, Ib, II, III, and V most often resulting in disease. Structural and sequence comparisons of the 10 types indicate that the differences across CPS types are more likely due to horizontal gene transfer rather than gradual mutagenesis (9). MLST uses the allelic profile of seven conserved genes in order to group GBS strains into sequence types (STs), which can be clustered into clonal complexes (CCs) (10). Several studies have shown that ST-17, a serotype III lineage, causes severe neonatal disease more often, indicating that ST-17 may be more virulent than other GBS STs (1015). Moreover, this lineage has a number of ST-17-specific genes that may contribute to its ability to cause meningitis, a topic that has been reviewed in detail elsewhere (16).

There are two different types of neonatal disease, early-onset disease (EOD) and late-onset disease (LOD), which differ based on the age of the baby at the time of clinical presentation as well as the possible mechanism of transmission. EOD typically presents as pneumonia and sepsis, which occur within hours after birth and up to 1 week of age. Vertical transmission of GBS occurs when the baby inhales infected vaginal fluid during birth or may occur due to ascending GBS infection from the vaginal canal crossing the extraplacental membranes to infect the amniotic fluid (17, 18). LOD typically presents as bloodstream infections leading to meningitis and occurs after 7 days of age but before the first 3 months of life (19). The transmission and pathogenesis of LOD are not well understood, although premature birth has been shown to be a major risk factor (20). In the United States, current rates of EOD are 0.23 cases per 1,000 live births, and LOD rates are 0.34 cases per 1,000 live births (7). Preventative measures against neonatal GBS disease involve intrapartum antibiotic prophylaxis (IAP) given to women who test positive for GBS colonization or those who are in preterm labor in order to reduce the likelihood of transmission to the baby during birth. These practices have successfully reduced the number of cases of EOD; however, the incidence of LOD has remained the same, and overall case rates have plateaued over the years, indicating a need for alternative therapies (21). Because women can remain persistently colonized by GBS even after IAP, they are still able to transmit the bacterium even after birth (22).

The first step in neonatal GBS disease progression is asymptomatic colonization of vaginal epithelial cells in the pregnant mother. Heavy maternal colonization is a primary risk factor for EOD (23). Vertical transmission results in infection via the lungs, where GBS then adheres to and invades lung epithelial cells. From the lungs, GBS can gain access to the bloodstream, causing sepsis. In the most severe cases, GBS is able to breach the blood-brain barrier, resulting in meningitis (17). Although the pathogenesis of LOD is not fully understood, it is possible that the baby acquires the bacterium from the mother (24). A number of case studies, for instance, have identified infected breast milk as a possible source (2527). However, a number of LOD cases have occurred after the baby was fed formula or in the absence of GBS-infected breast milk (24, 28), suggesting nosocomial, community, or other environmental sources. Since babies can become asymptomatically colonized by GBS in their intestines following birth (5), it is also possible that GBS invades across the intestinal epithelium, resulting in LOD.

The severity of disease can be attributed to the susceptibility of the newborn and the ability of GBS to avoid immunological clearance and adapt to changing environments throughout disease progression. Infants generally become infected by GBS during the first 3 months of life, suggesting that the immature state of the immune system contributes to susceptibility to infection. Moreover, GBS infections in nonpregnant adults typically present when the host is in an immunocompromised or relatively compromised state, such as diabetes, cancer, HIV, and others, with diabetes being the predominating underlying condition (2932). The common theme of GBS infection appears to be that optimal conditions for the pathogenesis of GBS invasion occur when a part of the immune defense system is compromised. A greater understanding of the capacity of GBS to interact with the deficient immune system will aid in the development of novel therapies or preventative measures for invasive disease. Examining which immune cells are deficient in these cases will provide clues about the predominating cell types that keep GBS under control in colonized individuals. The process by which GBS transitions from a colonizing state to an invasive pathogen and its interactions with innate immune cells were recently reviewed by Landwehr-Kenzel and Henneke (33). Here, we focus on innate immune deficiencies in the newborn that enhance susceptibility to disease, host immune responses to GBS infection, and mechanisms that GBS uses to evade immune responses.

DEFICIENCIES IN NEONATAL IMMUNITY

The relatively underdeveloped newborn immune system includes a reduced number of available immune cells, resulting in heightened susceptibility to infectious diseases. Moreover, neonatal immune cells can be present in different proportions in different sites relative to adult immune cell populations (34). The general characteristics of neonatal immune cells compared to adult immune cells are listed in Table 1. The neonatal immune system is also relatively naive, resulting in a lack of preexisting memory immune cells, which leads to a dependency on the maternal transfer of antibodies. Furthermore, the newborn immune system produces higher levels of anti-inflammatory cytokines than proinflammatory cytokines. A thorough understanding of these deficiencies and their implications is an important step toward helping to protect neonates from invading pathogens such as GBS. The neonatal immune system was recently reviewed in detail elsewhere (34, 35), and we only briefly discuss neonatal immune deficiencies here.

TABLE 1.

Deficiencies in neonatal immune cells compared to adult cells

Cell type Characteristic of neonatal cells relative to adult cells Reference(s)
Neutrophils Reduced no. of stored cells 36
Reduced recruitment to sites of infection 37
Initially reduced phagocytic ability 38
Delayed NET response 39, 40
Monocytes Higher no. of cells 41, 42
Reduced recruitment to sites of infection 43
Similar phagocytic ability 38
Reduced cytokine production in response to stimuli 44
Diminished antigen presentation capacity 47
Macrophages Low no. of alveolar macrophages immediately after birth 49
Reduced antigen processing and presentation 50
Delayed response to recruit monocytes and neutrophils to site of infection 49
Similar migration and ROS production 41
Dendritic cells Reduced capacity to stimulate other immune cells 54
Reduced IFN-α/β production 56
Similar level of proinflammatory cytokine production 57
T cells Higher no. of Th2 cells 63
Diminished no. of Th1 cells 63
Diminished no./lack of Th17 cells 64
B cells Immature development of surface Ig 65
Deficient signaling through the BCR 66

Innate Immunity Deficiencies

Since the adaptive immune system has limited exposures to antigens in utero, resulting in a deficient adaptive immune response, neonates rely mainly on the innate immune response to pathogens. Neutrophils are one of the main phagocyte types found in the blood and are the first cells recruited to the site of infection. The neutrophil storage pool, however, is much smaller than that in adults; also, neonatal rats challenged with GBS developed neutropenia, and neutrophil storage pools rapidly became depleted (36). In addition to the small pool of stored neutrophils, neonatal neutrophils show impaired rolling adhesion, transmigration, and chemotaxis, resulting in poor recruitment to infection sites (37). Neutrophils from both preterm and term neonates also show reduced levels of phagocytosis compared to adult neutrophils but become comparable to adult neutrophils by 3 days after birth (38). Neonatal neutrophils are capable of producing functional neutrophil extracellular traps (NETs), but the response is delayed and requires extended stimulation, making them less able to aid in clearing pathogens (39, 40). These findings show not only that there are fewer neutrophils being recruited to the infection site but also that the neutrophils that make it there are deficient in their ability to clear infection, making neonates particularly susceptible to infection within the first few days after birth.

In contrast to neutrophils, the numbers of monocytes are much higher in neonates than in adults, while preterm neonates have even higher numbers of monocytes than do term neonates (41, 42). Although the phagocytic ability of neonatal monocytes is the same as that of adult monocytes (38), monocyte chemotaxis and recruitment to the site of infection are attenuated (43), and cytokine concentrations are lower, resulting in a reduced in inflammatory response (44). Additionally, a study that examined differences between adult peripheral and cord blood monocytes in their interactions with GBS found no difference in phagocytic uptake, bacterial degradation, and reactive oxygen species (ROS) production. However, there was a reduced level of cell death following GBS infection in cord blood monocytes compared to adult monocytes (45). Since it is possible that a higher level of apoptosis early during sepsis leads to improved outcomes in patients, this reduced level of apoptosis in cord blood monocytes contributes to poorer outcomes among neonates with sepsis (46). Neonatal monocytes also have reduced levels of major histocompatibility complex (MHC) class II expression on their surface, resulting in a diminished capacity for antigen presentation (47). Toll-like receptor (TLR)-mediated signal transduction pathways are also impaired in neonatal monocytes, resulting in the reduced activation of NF-κB, which is an important transcription factor involved in immune response regulation (48).

Once monocytes travel to tissues, they differentiate into macrophages. Numbers of alveolar macrophages are much lower in newborns than in adults; however, the number rises to adult levels 24 to 48 h after birth (49). As inhalation of GBS during birth is the main predisposing mechanism for pneumonia, the initial reduction of pulmonary macrophages predisposes newborns to an inability to rapidly clear the infection, resulting in EOD. Relative to adult murine macrophages, neonatal murine macrophages had reduced gene expression levels of MHC class II, CD11b, CD14, CD80, CD86, TLR2, TLR4, and TLR9, all of which are involved in processing and presenting antigens, with a corresponding reduction in the ability to induce T-cell proliferation (50). While neonatal macrophages have a delayed response in recruiting neutrophils and monocytes to the site of infection (49), migration and production of ROS are normal relative to adult macrophages (51). Upon stimulation through TLRs 1, 2, and 4, neonatal macrophages have an enhanced production of interleukin-6 (IL-6), demonstrating the ability to secrete proinflammatory cytokines despite having other deficits (52).

Many different subtypes of dendritic cells (DCs) can be found, which vary in tissue localization as well as surface receptor expression and function. Although DCs are highly specialized, potent antigen-presenting cells (53), cord blood DCs are very immature. Indeed, cord blood DCs are unable to stimulate either adult or cord blood mononuclear or T cells, in contrast to adult DCs, suggesting a deficit in cord blood DCs (54). In addition, cord blood DCs have reduced levels of expression of MHC classes I and II, ICAM-1/CD54, CD40, CD80, CD83, and CD86 relative to adult DCs, which is indicative of immaturity (54, 55). DCs stimulated by TLR7/9 have a reduced ability to produce alpha/beta interferons (IFN-α/β), which are important immune regulators. This deficiency is due to the reduced translocation of the transcription factor interferon regulatory factor 7 (IRF7) into the nucleus (56). However, stimulated cord blood monocyte-derived DCs have similar levels of NF-κB signaling as well as secretion of the proinflammatory cytokines tumor necrosis factor alpha (TNF-α), IL-6, and IL-8 compared to those of adult DCs (57).

In addition to the reduced numbers and function of neonatal innate immune cells, the complement system is also underdeveloped. Depending on the stimuli, the complement system can be activated through either the classical, alternative, or lectin pathway through a cascade of enzymatic reactions. Regardless of the activation pathway used, the result is the formation of the membrane attack complex (MAC), which creates a channel in cell membranes that results in cell lysis. Additionally, throughout the cascade, a number of enzymatic intermediates and cleavage products are formed, which play a role in immune responses, such as immune cell activation or bacterial cell opsonization (58). Complement proteins cannot be transplacentally transferred from mother to fetus, and the numbers of neonatal complement proteins are only 10 to 80% of those found in adults (59). More specifically, the classical pathway components C1q, C3, and C4 as well as the alternative pathway components properdin and factor B are deficient in neonates (5961). These deficiencies in the neonatal complement system result in a reduced ability to activate the complement cascade, thereby leading to reduced phagocytosis, a reduced ability to lyse pathogens, and reduced recruitment of immune cells to sites of infection (59).

Adaptive Immunity Deficiencies

Deficiencies in the innate immune system can lead to reduced adaptive immune responses, and there are a number of deficiencies and differences in neonatal adaptive immune cells relative to those of adults. The neonatal adaptive immune response can range from no response to a strong response similar to that of adults (62). Although neonates mainly rely on their innate immune response to pathogens within the timeline of GBS transmission, understanding how neonates differ in their adaptive immune response compared to that of adults may greatly influence vaccine development efforts.

T cells can be classified into different subclasses that play specific roles in the immune response. CD4+ T cells, also known as T helper (Th) cells, play an important role in activating or stimulating the maturation of other immune cells and can be further differentiated into other subtypes, with the two major subtypes being Th1 and Th2. Th1 cells aid in the production of inflammatory responses to microbial pathogens, whereas Th2 cells secrete cytokines in response to parasites and allergens. Interestingly, the neonatal immune system has a much larger population of Th2 cells and diminished numbers of Th1 cells (63). In addition to the reduced number of Th1 cells, neonates also have reduced numbers or even a complete lack of Th17 cells, which aid in developing immunity to both bacterial and fungal infections at mucosal surfaces (64).

Neonates have been shown to have defective B-cell responses resulting in deficient humoral immunity as well. This defective response could be due to the immature development of surface immunoglobulins (Igs) and a reduced level of antigen exposure. Additionally, follicular Th (TFH) cells play an important role in developing an antibody response by eliciting the proliferation and maturation of B cells. Neonates have a reduced frequency of TFH cells, which are regulated by IL-4 production by Th2 cells (65). Moreover, B-cell signaling through the B-cell receptor (BCR) is deficient in neonatal B cells, a phenotype that could be caused by higher expression levels of CD22, a negative regulator of BCR signaling in neonatal B cells (66).

Despite these deficiencies in adaptive immunity, neonates have protective antibodies that are passed on from mother to neonate either transplacentally or through breast milk. These maternal antibodies help protect the neonate from infection but can also impact the neonatal immune response to infection and vaccination (67). A study that examined specific antibody concentrations at birth and after immunization found an inverse correlation between birth concentrations and increases in antibody concentrations after immunization. These data suggest that these higher concentrations of antibodies at birth could inhibit the neonatal immune response to vaccines. Nonetheless, most of the neonates in that study developed antibodies, suggesting that there is not a complete inhibition of neonatal antibody development by higher concentrations of maternal antibodies (68).

IMMUNE RESPONSE TO GBS AND MECHANISMS OF IMMUNE EVASION

Recognition of and Cytokine Response to GBS by the Innate Immune System

GBS is able to elicit a strong host inflammatory response. Upon ex vivo GBS infection, neonatal monocytes produce proinflammatory cytokines, including TNF and IL-6, but at reduced levels compared to those produced by adult monocytes (69). GBS strains belonging to different sequence types also elicited different cytokine responses in primary human monocyte cells. More specifically, infection by strains belonging to CC17 and -19, both of which are more frequently associated with infection (10), resulted in significantly higher levels of production of TNF-α, IL-6, and IL-8 than those induced by strains of other lineages (70).

GBS activates phagocytes via interactions with TLR2 and TLR6, and this activation is dependent on the TLR adaptor protein myeloid differentiation factor 88 (MyD88) (71, 72). Additionally, GBS-induced activation of inflammatory cytokines requires the c-Jun kinase pathway (73), while phagosomal GBS induces interferon in DCs via TLR7, MyD88, and the transcription factor IRF1 (74). Furthermore, GBS single-stranded RNA (ssRNA) is recognized by monocytes and macrophages via a complex comprising MyD88 and UNC-93B (75). The recognition of GBS ssRNA results in the increased production of nitric oxide (NO) by host cells, which activates macrophages and aids in phagosome acidification (76). The presence of GBS DNA also induces the release of IL-6, IL-12, and TNF-α via TLR9 but does not upregulate IFN-β or NO secretion (77). In contrast, IFN-β production was shown to be induced by GBS DNA in murine bone marrow-derived macrophages as well as THP-1 human monocytes in a TLR-independent manner. Rather, cytoplasmic GBS DNA is sensed by cyclic GMP-AMP synthase (cGAS), which activates stimulator of interferon genes (STING) that leads to IFN-β production (78, 79). GBS also releases cyclic di-AMP (c-di-AMP) into its environment, which can directly activate STING without cGAS; however, a GBS-expressed ectonucleotidase (CdnP) degrades c-di-AMP in order to reduce STING activation (79). Elevated levels of TNF-α occur during GBS sepsis, which is believed to play a role in clinical outcomes and is released from both monocytes and macrophages in response to GBS. The deposition of complement on GBS, more specifically C3 activation via the alternative pathway, triggers TNF-α production by monocytes (80). Monocytes are the most abundant innate immune cells in neonates, which could contribute to the abundance of monocyte-derived TNF-α production (49).

GBS produces a surface-associated beta-hemolysin/cytolysin toxin that is encoded by the cyl operon and is a major virulence factor (81). This ornithine rhamnolipid also generates pigmentation in GBS and has been shown to aid in crossing human extraplacental membranes (82). Not only does GBS beta-hemolysin/cytolysin contribute to pathogenicity through its cytolytic properties and by promoting invasion across host cell barriers, it also stimulates a potent proinflammatory cytokine response via the release of IL-1 and IL-6 and NO production in macrophages (83). Moreover, purified beta-hemolysin/cytolysin increased membrane permeabilization, resulting in the osmotic lysis of red blood cells and pyroptosis induction in macrophages (84). Both purified beta-hemolysin/cytolysin and hyperpigmented GBS were also cytotoxic to adult neutrophils but not through apoptosis or pyroptosis (85). Activation of the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome by GBS is dependent on the expression of beta-hemolysin/cytolysin. Inflammasomes are multiprotein complexes located inside innate immune cells that activate the immune system in response to pathogens through the activation of caspase-1, which leads to an inflammatory response (86). In macrophages, GBS beta-hemolysin/cytolysin can cause leakage of the lysosome containing GBS, which allows the escape of bacterial RNA. This RNA then activates the NLRP3 inflammasome, inducing the production of IL-1β (87).

GBS Immune System Evasion

GBS employs several mechanisms to resist immune detection and phagocytosis, thereby increasing the chance of survival in the host. These mechanisms are summarized in Fig. 1. One example is the expression of the polysaccharide capsule (CPS), which is considered a major virulence factor, as unencapsulated GBS strains are less virulent in animal models (88). The GBS capsule contains a terminal sialic acid (Sia). Since Sia is also present on the surface of vertebrate cells, the Sia on the surface of GBS allows it to mimic host cells and avoid immune detection (89).

FIG 1.

FIG 1

Mechanisms used by GBS to evade the immune system. GBS expresses many factors that help it evade the immune system and increase its survival in the host. The sialic acid capsule and fibrin fragments cleaved by CspA that coat the surface help GBS present as “self” to the immune system. The capsule also blocks C3 deposition and recognition by phagocytes. Sialic acid in the capsule, β-protein, ScpB, CIP, and BibA inhibit the complement system by binding or cleaving complement components. The GBS β-protein also binds the Fc region of IgA1 to inhibit immune activation. HylB and CspA inhibit or cleave cytokines, while PilB, PBP1a, and proteins encoded by the dlt operon assist in resisting antimicrobial peptides. NucA degrades the DNA matrix of neutrophil extracellular traps. Glutathione, carotenoid pigment, and SodA all aid in defense against reactive oxygen species, and both β-hemolysin/cytolysin (β-h/c) and GAPDH aid in inducing apoptosis in phagocytes.

Sia-binding immunoglobulin-like lectins (Siglecs) are located primarily on the surface of leukocytes and are responsible for distinguishing between “self” and “nonself” to determine if an immune response should be activated. The Sia in the GBS capsule binds to Siglecs in order to reduce the activation of NF-κB and mitogen-activated protein kinase (MAPK) signaling, thus inhibiting the immune response. Siglec-9 expressed by human neutrophils recognizes Sia on the surface of GBS and dampens the immune response (90). Additionally, the surface-expressed β-protein of GBS binds to both Siglec-5 and Siglec-14 on the surface of neutrophils (91, 92). Interestingly, ligand binding to Siglec-5 elicits an inhibitory response in phagocytes, whereas Siglec-14 binding elicits an activating response. Since both Siglecs have similar ligand-binding motifs, it has been suggested that they are paired receptors that play a role in balancing the immune response to invading bacteria. Moreover, Siglec-5/14 expression was found on the surface of the amniotic membrane in human extraplacental membranes (92). This unusual location for Siglec expression is of particular interest, since GBS is capable of crossing extraplacental membranes (18). GBS binding to Siglecs results in the impairment of phagocytosis, reduced ROS generation, and poor extracellular trap formation in leukocytes (90, 91). Macrophages lacking Siglecs show enhanced production of proinflammatory cytokines, phagocytosis, and bacterial killing of GBS (93). Macrophages also express sialoadhesin on their surface, which is a unique type of Siglec that contains an elongated extracellular portion capable of recognizing Sia on the surface of pathogens and mounting an inflammatory response. Sialoadhesin aids in clearing GBS infection and blocking dissemination to organs in mice (94).

Another mechanism of host cell mimicry employed by GBS is coating itself with the highly adhesive fibrin breakdown product of fibrinogen. GBS uses the cell surface protein CspA to cleave fibrinogen similarly to thrombin, which results in the exposure of the regions responsible for fibrinogen polymerization, leading to the aggregation of GBS and coating of the bacterial surface with fibrin. This fibrin coating allows GBS to appear as “self” to host immune cells and reduces the access of opsonins to the surface, thereby inhibiting opsonophagocytosis (95).

The CPS can also inhibit opsonophagocytosis by blocking the deposition of C3b on the surface of GBS. Both unencapsulated and encapsulated strains lacking sialic acid, for instance, bound more C3 molecules than did a wild-type (WT) strain (96). GBS also expresses other surface components that prevent opsonophagocytosis as well as the activation of the complement cascade. BibA, for example, resists opsonophagocytic killing by neutrophils via the specific binding of the C4-binding protein, which is a regulator of the complement pathway (97). The secreted complement-interfering protein (CIP) binds to C4b, inhibiting its interaction with C2 to reduce complement activation through the classical and lectin pathways but not the alternative pathway (98). Similarly, the GBS β-protein binds the soluble complement inhibitor factor H to the bacterial surface in a way that inhibits C3b deposition and opsonophagocytosis (99); the Sia residues in the CPS can also bind factor H (100). Another important factor is a serine protease, ScpB, which is a C5a peptidase that proteolytically cleaves complement-activated C5a, a powerful chemoattractant involved in the recruitment of inflammatory cells (101). In addition to its ability to cleave fibrinogen, CspA is also capable of cleaving and, therefore, inactivating CXC chemokines that recruit neutrophils to different infection sites (102).

In a pregnant mouse model, GBS was shown to ascend the vaginal tract to infect the decidua, placenta, and fetus. This invasion was marked by a large recruitment of neutrophils to the infection site in the decidua and placenta, which is similar to what is seen in chorioamnionitis in human patients. Neutrophils isolated from mice also produced NETs in response to GBS (103); similar results were observed in a nonhuman primate model of amniotic cavity infection by GBS (85). NETs are produced by neutrophils in response to invading bacteria and consist of DNA and antimicrobial peptides (AMPs). These NETs ensnare bacteria and eliminate them to help clear infections (104). High expression levels of beta-hemolysin/cytolysin, as well as purified beta-hemolysin/cytolysin, induce NET formation in adult neutrophils, although beta-hemolysin/cytolysin also conferred resistance to killing by these NETs (85). GBS-induced NETs contain lactoferrin, which sequesters iron, preventing invading pathogens from using it as a nutrient source. Lactoferrin is capable of repressing GBS growth and could be one way in which these NETs prevent some GBS strains from invading (103). Nonetheless, GBS also produces nuclease A (NucA), which degrades the DNA in the NETs to allow GBS to escape. In a previous study, NucA was needed for GBS persistence in lung tissue, and a nucA mutant was less virulent than the WT in a mouse model, suggesting that NucA is important for both initial infection as well as dissemination (105).

In response to tissue injury following pathogen invasion, hyaluronan (HA), a component of the extracellular matrix, is quickly degraded by host hyaluronidases and ROS (106). The small cleavage products are recognized by TLR2 and/or TLR4 to stimulate an inflammatory response to clear the pathogen as well as initiate wound healing (107, 108). GBS secretes hyaluronidase, encoded by hylB, to degrade HA to assist in dissemination. Interestingly, HylB plays roles in enhancing survival inside macrophages, inhibiting proinflammatory cytokine expression, and utilizing HA as a carbon source in the host (109). The GBS hyaluronidase degrades HA into disaccharides instead of 4- to 16-mer fragments that produce a proinflammatory response. These HA disaccharides are capable of blocking TLR2/4 signaling, resulting in reduced proinflammatory cytokine production (110).

Phagocytic Uptake of GBS

Despite the above-described mechanisms employed by GBS to avoid immune detection and phagocytosis, GBS is easily phagocytosed and killed by phagocytic cells in the presence of serotype-specific antibodies via Fc receptors (111). Internalization of GBS can also occur through complement receptor 3 (CR3) in the presence of other opsonins like lectins and L-ficolin (112). Since GBS elicits a poor antibody response and neonates have low levels of complement, opsonin-independent pathways of phagocytosis would be the more likely mechanism of uptake of GBS. Additionally, GBS is rapidly taken up by macrophages in the absence of opsonins (111). Because CR3 is important for opsonin-independent phagocytosis by macrophages, GBS was suggested to interact with CR3 in a C3-independent manner (113). Furthermore, the uptake of GBS requires actin (111) and varies by strain type (114). Besides the complement-binding domain, CR3 also contains a lectin domain that is able to bind the type III CPS to initiate phagocytosis in neutrophils (115).

GBS Induction of Apoptosis in Macrophages

One strategy used to avoid immune activation after a pathogen is taken up by a phagocyte and to persist at the site of infection is to induce the apoptosis of immune cells before they become activated (116). Apoptosis is a process of programmed cell death that is less likely to elicit a strong inflammatory response, such as that seen with necrosis or pyroptosis. However, there are certain cases in which apoptosis can be inflammatory (117). Since apoptosis plays a role in the maintenance of cell populations in tissues as well as during development and aging, it is a tightly regulated process. This process involves protein kinase C (PKC) activity and modulation of cytoplasmic calcium levels and is regulated by the caspase family of cysteine-directed proteases (caspase-dependent pathway) or calpains (caspase-independent pathway) as well as Bcl-2 family regulators (118).

GBS is capable of inducing apoptosis in macrophages, which requires internalization and is bacterial dose dependent (119). During induction, GBS stimulates the sustained activation of c-Jun NH2-terminal kinase (JNK) and p38 but inhibits extracellular signal-regulated kinase (ERK), all of which are members of the MAPK family (120). Moreover, GBS infection of macrophages also induces the expression of TNF-α, IL-1, and inducible nitric oxide synthase (iNOS), leading to apoptosis. Inhibition of iNOS expression inhibited GBS-induced apoptosis, but inhibition of TNF-α and IL-1 did not. Also, the addition of NO alone without infection induced apoptosis, indicating a direct effect of GBS-induced NO production on apoptosis (121).

The role of caspases in GBS-induced apoptosis is not clear. One study showed that GBS-induced apoptosis was independent of caspase-1 and -3 (119), whereas another study showed that caspase-3 and -9 were important for this process (121). These contradictory results could be due to the use of different GBS strains in those studies: both studies used serotype III strains, but different strains of the same serotype have been shown to have various host-pathogen interactions (122). Therefore, it is possible that diverse strains of GBS are capable of using different mechanisms for inducing apoptosis. Moreover, those studies were done by using cell culture, making it difficult to fully understand the mechanism of GBS-induced apoptosis in vivo. Interestingly, one study used an ex vivo fetal rat lung model to show that caspase-3 activation results in apoptosis in macrophages and erythroblasts in the lung interstitium following GBS infection (123).

Through beta-hemolysin/cytolysin-induced plasma membrane permeability, GBS is able to cause a massive increase in calcium levels inside macrophages leading to the activation of the calcium-sensitive calpains, which leads to the degradation of structural and regulatory cytoskeletal proteins as well as the induction of apoptosis (124, 125). GBS-induced calcium influx also results in PKC activation (119) as well as the activation of gelsolin, an important regulator of the actin cytoskeleton and apoptosis (126). Glyceraldehyde-3-phosphate dehydrogenases (GAPDHs) are surface-localized enzymes that are capable of binding to host cell components and have immunomodulatory effects. Interestingly, GAPDHs from GBS and other pathogens, including Streptococcus pyogenes and Staphylococcus aureus, can induce apoptosis in macrophages, indicating yet another role of bacterial GAPDH in pathogenesis (127).

GBS Survival inside Phagocytes

Once a bacterium is taken up by a phagocytic cell, it gets trapped within a vacuole that goes through phagosomal maturation, in which the vacuole fuses with various compartments in the endocytic pathway. The end product is a fully mature phagolysosome, which consists of a harsh, highly acidic, and nutrient-limiting environment where AMPs, ROS, and reactive nitrogen species (RNS) are generated to kill the bacterium (128). Although most bacteria are efficiently killed by this process, many pathogens have developed ways to overcome these defense mechanisms. For instance, some pathogens can disrupt cellular signaling to prevent or slow down the phagosome maturation process and live inside the phagosome. Other pathogens can escape from the phagosome by lysing the membrane to replicate in the cytosol, while others can remain inside the phagolysosome, defending against the many stressors (129).

GBS is capable of persisting within macrophages and remains inside the phagosome, which recruits late endosomal markers. This recruitment indicates that GBS does not inhibit phagosome maturation as a survival strategy and likely uses a phagosomal stress defense mechanism (111, 130). This ability to survive inside innate immune cells allows GBS to avoid immune detection, protect against antibiotics, and facilitate dissemination to other sites of the body, making it a particularly important topic of study (131, 132). Indeed, opsonization of GBS significantly reduces the ability of GBS to survive intracellularly (111). Although the CPS helps GBS avoid phagocytosis, it does not aid in intracellular survival, as unencapsulated mutants were internalized at a higher rate in a previous study; the time of survival intracellularly, however, was no different than that for the encapsulated WT strain (133).

GBS has several strategies to help it survive under the antimicrobial conditions of the phagosome. Upon infection, macrophages undergo a number of changes in protein expression that result in the decreased expression of enzymes that impact ROS production and NO synthesis, both of which are important for antimicrobial responses. Since these changes were not observed in macrophages infected with heat-inactivated GBS, it is likely that GBS actively induces these changes (134). In addition to its ability to inhibit ROS production, GBS also has the ability to inactivate ROS through the use of superoxide dismutase (SodA), which functions to convert superoxide into oxygen and hydrogen peroxide (135). Although GBS is catalase negative, sequencing shows that the GBS genome contains NADH peroxidase, a thiol peroxidase, and an alkylhydroperoxide reductase, all of which could possibly be used to detoxify hydrogen peroxide (136). Moreover, GBS has been shown to produce glutathione (137), which protects the bacterial cell from oxidative stress, low pH, as well as other stresses (138). In addition to its immunomodulatory effects and cytolytic properties, beta-hemolysin/cytolysin also produces an orange carotenoid pigment, which has also been shown to protect GBS from oxidative damage (139).

In addition to ROS and RNS production, a number of AMPs and hydrolases are present in the phagosome to kill bacteria (140). Penicillin-binding protein 1a (PBP1a), for example, is important for resisting host AMPs (141). One mechanism used to avoid the effect of cationic AMPs used by GBS is to increase the number of d-alanine residues in lipoteichoic acids, which is regulated by the dlt operon (142). Initially, it was thought that d-alanylation would reduce the electronegativity of the cell wall and therefore decrease the affinity of cationic AMPs. However, a previous study showed that d-alanylation altered the rigidity and permeability of the cell wall, which blocked certain cationic AMPs from crossing it (143).

Additionally, GBS pili have been shown to mediate resistance to AMPs in addition to aiding in host cell attachment. There are three distinct pilus islands (PIs), PI-1, PI-2a, and PI-2b, that encode structurally different pili in GBS (144). PilB, a pilus protein subunit, was shown to play a role in intracellular survival in murine macrophages and human neutrophils by conferring resistance to cathelicidin and defensin families of AMPs and facilitates bloodstream survival in a mouse model. Moreover, the expression of GBS PilB in Lactococcus lactis, which is susceptible to AMPs, conferred resistance to AMPs (145). Contradictory to these results, one study found no significant difference in survival inside murine macrophages between WT and ΔpilB strains (146). One possible explanation for this difference could be that different strains were used, which may vary in the mechanism used to survive inside the phagosome (114). The pilus backbone protein specific for ST-17 lineages, Spb1, was also shown to enhance both phagocytosis and intracellular survival of GBS. Additionally, the presence of spb1 in GBS strains did not alter NO or TNF-α responses in macrophages (147). Another ST-17-specific gene, srr2, plays a role in binding both fibrinogen and plasminogen but has also been shown to increase phagocytic uptake and intracellular survival in macrophages and neutrophils (148). Although having a protein that would enhance the phagocytic uptake of the pathogen seems counterintuitive, that same protein can also be used to enhance survival inside macrophages while promoting dissemination. These proteins, along with several other ST-17-specific virulence factors, may partly explain the enhanced ability of ST-17 strains to survive inside macrophages as well as their increased virulence and association with neonatal infections (16).

As a lactic acid-producing bacterium, GBS has mechanisms to withstand low pH and should be expected to withstand the low pH of the phagosome. Indeed, a previous study demonstrated that ∼18% of the genes in the GBS genome were differentially expressed at pH 5.5 relative to pH 7.0, and most of these genes are regulated by the CovR/S (also known as CsrRS) two-component regulatory system (149). In addition to regulating many virulence factors, this CovR/S acid response regulator was found to be required for GBS to survive inside macrophages. Some of the genes upregulated at low pH encode transporters, which may allow GBS to increase its scavenging ability to facilitate survival under the nutrient-limiting conditions of the phagosome (130). Moreover, inhibition of the acidification of the phagosome significantly reduced the ability of GBS to survive in macrophages, suggesting that acidic pH is needed for GBS to survive phagosomal stress. This reduced survival, however, was not observed in all of the strains examined, suggesting that diverse strains of GBS are using alternative mechanisms to withstand phagosomal stress (114).

Antibody Response to GBS and Vaccine Development

Because of the large number of deficits in the neonatal innate immune system, maternal antibody transfer is very important in passive immune protection of the newborn. A deficiency in maternal antibody responses targeting GBS has been considered to be important for neonatal infections (150). Moreover, CPS type III strains induce a lower antibody response than those induced by strains of other CPS types (151). This finding is consistent with data from our previous study showing that CPS type III strains representing multiple STs survived better in a multiple-stress medium comprising key phagosomal stressors than did strains of other genotypes with various CPS types. Indeed, enhanced survival in macrophages could result in decreased bacterial killing and presentation of CPS antigens to the adaptive immune system (114).

Since human colostrum and milk contain high concentrations of secretory IgA, it is probable that IgA plays an important role in neonatal protective immunity. Known roles of IgA include recognizing pathogens and triggering a response to eliminate them. Once IgA recognizes a pathogen, it interacts with CD89 on the surface of phagocytes to induce phagocytosis, ROS production, and the production of inflammatory mediators (152). The GBS surface-expressed β-protein also plays a role in binding to the Fc region of IgA, which inhibits IgA binding to CD89 and blocks proactive immunity from maternal IgA (153).

Due to the high level of diversity across GBS strains, vaccine development efforts have been difficult. Since CPS types are both antigenically and structurally unique, CPS-based vaccines do not offer protection against other CPS types (154). Studies have switched toward examining conserved antigenic proteins as vaccine candidates. Interestingly, one study found that both mothers and their newborns naturally produced antibodies against the GBS surface protein Sip. This finding suggests not only that mothers can produce Sip antibodies but also that these antibodies are transferred transplacentally and can persist in the infant (155). Another study found that GBS-colonized mothers who delivered healthy babies had higher levels of naturally occurring antibodies against both CPS and pilus proteins than did mothers whose babies developed GBS infection or noncolonized mothers (156). This finding further supports the role of maternal antibodies in protecting neonates from GBS infections and suggests that a vaccine strategy targeting pregnant women has potential merit and warrants further investigation. The possibilities of such a vaccine as well as the status of vaccine development have been reviewed elsewhere (157, 158). Current efforts have also focused on developing both CPS-protein conjugate vaccines and protein-based vaccines that target conserved GBS proteins (159).

CONCLUDING REMARKS AND FUTURE DIRECTIONS

The neonatal immune system has several deficiencies and limitations that render neonates more susceptible to infection. Furthermore, GBS has an arsenal of immune evasion strategies and virulence factors that make it an extremely successful pathogen in neonates. Although previous studies examined the interaction between GBS and the immune system, many of those studies were conducted in vitro by using cell lines or primary cells, and hence, it is difficult to know how these findings correlate with those of in vivo studies. Additionally, many studies have used immune cells derived from adults, which have properties and functions distinct from those of neonatal cells. It would therefore be interesting and informative to explore more of these interactions using neonatal or deficient immune cells. Similarly, most in vivo studies utilize murine models, which have important differences from humans (160) and also limit our ability to correlate findings to natural human infections. The development of humanized strains of mice has helped overcome a number of these differences and has become a popular method for studying specific aspects of the immune system (161). Interestingly, humanized mice have deficiencies in several immune cells and the complement system, which are similar to those found in neonates, making humanized mice a promising model to study neonatal responses to infections. The use of specific-pathogen-free or germfree murine models will also be useful to mimic the naive nature of the neonatal immune system. Indeed, Ernst et al. recently introduced a neonatal humanized model of GBS sepsis, which represents an intriguing system to further explore neonatal infections (162).

Despite the large number of advancements in our understanding of neonatal GBS infections, there are still many areas left to be explored. Although a number of studies have begun to explore variation across GBS strains, it is important to further examine these differences to determine why certain strains/lineages have a greater capacity to cause disease than do others. Some aspects of the phagocytic uptake of GBS in the absence of opsonins have been explored; however, more details of the precise mechanisms still need to be elucidated. Moreover, the mechanism by which GBS induces apoptosis in vivo is another interesting area to be explored, as most previous studies were performed in vitro. Although GBS survives inside a mature phagolysosome and likely uses a stress defense mechanism, only a few bacterial factors have been identified to be important for this process to date. Future studies should therefore focus on identifying additional mechanisms that are important for resisting phagosomal stress, particularly in those genotypes that more commonly cause neonatal infections.

GBS is a highly versatile organism that causes invasive disease in neonates in addition to elderly and immunocompromised adults. Since GBS is a leading cause of neonatal sepsis and meningitis, many studies have focused on these infections. The steady rate of EOD in neonates despite current preventative measures, as well as high frequencies of antibiotic resistance, emphasizes the need to find additional or alternative therapeutics and preventatives. Additionally, the current preventative practice of IAP has not had an effect on the incidence of LOD. In order to better tailor efforts in developing new therapeutic and preventive measures, a more thorough understanding of the how GBS interacts with the immune system is required.

Biographies

graphic file with name zcm0041726010002.jpg

Michelle L. Korir recently defended her Ph.D. thesis in Microbiology and Molecular Genetics at Michigan State University in December 2016. Her research focused on the ability of diverse strains of group B Streptococcus to interact with host cells as well as mechanisms of survival in human macrophages. She will be starting her postdoctoral research in the Department of Microbiology and Immunology at the University of Minnesota. Her research interests are in bacterial pathogenesis and host-pathogen interactions.

graphic file with name zcm0041726010003.jpg

Shannon D. Manning is a Michigan State University (MSU) Foundation Associate Professor in the Department of Microbiology and Molecular Genetics (MMG). She earned her M.P.H. and Ph.D. from the University of Michigan (UM) in 1998 and 2001, respectively. For her postdoctoral training, she served as an emerging infectious diseases research fellow through the Centers for Disease Control and Prevention and held research faculty positions at UM and MSU. She joined the MMG faculty in 2010 and is interested in the pathogenesis and molecular epidemiology of bacterial pathogens, namely diarrheagenic Escherichia coli and group B Streptococcus.

graphic file with name zcm0041726010004.jpg

H. Dele Davies is Professor of Pediatrics, Infectious Diseases, and Public Health at the University of Nebraska Medical Center. Dr. Davies received his M.D. from the University of Toronto in 1985, followed by pediatric residency and infectious diseases fellowships at the Hospital for Sick Children in Toronto, Canada (1992). Dr. Davies holds dual master's degrees in Epidemiology (Toronto) and in Health Care Management (Harvard School of Public Health). Dr. Davies has a long-standing research area of interest in clinical and molecular epidemiology, virulence factors, and host responses related to group B streptococcus infections. Dr. Davies was previously Director of the Child Health Research Unit and Chair of the Child Health Research Group at the University of Calgary and Professor and Chair of Pediatrics at Michigan State University. He has served or currently serves on different U.S. and Canadian national advisory boards related to prevention and management of Infectious Diseases.

REFERENCES

  • 1.Hickman ME, Rench MA, Ferrieri P, Baker CJ. 1999. Changing epidemiology of group B streptococcal colonization. Pediatrics 104:203–209. [DOI] [PubMed] [Google Scholar]
  • 2.Bliss SJ, Manning SD, Tallman P, Baker CJ, Pearlman MD, Marrs CF, Foxman B. 2002. Group B Streptococcus colonization in male and nonpregnant female university students: a cross-sectional prevalence study. Clin Infect Dis 34:184–190. doi: 10.1086/338258. [DOI] [PubMed] [Google Scholar]
  • 3.Manning SD, Neighbors K, Tallman PA, Gillespie B, Marrs CF, Borchardt SM, Baker CJ, Pearlman MD, Foxman B. 2004. Prevalence of group B Streptococcus colonization and potential for transmission by casual contact in healthy young men and women. Clin Infect Dis 39:380–388. doi: 10.1086/422321. [DOI] [PubMed] [Google Scholar]
  • 4.Hansen SM, Uldbjerg N, Kilian M, Sørensen BS. 2004. Dynamics of Streptococcus agalactiae colonization in women during and after pregnancy and in their infants. J Clin Microbiol 42:83–89. doi: 10.1128/JCM.42.1.83-89.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Weindling AM, Hawkins JM, Coombes MA, Stringer J. 1981. Colonisation of babies and their families by group B streptococci. Br Med J (Clin Res Ed) 283:1503–1505. doi: 10.1136/bmj.283.6305.1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Anthony BF, Carter JA, Eisenstadt R, Rimer DG. 1983. Isolation of group B streptococci from the proximal small intestine of adults. J Infect Dis 147:776. doi: 10.1093/infdis/147.4.776. [DOI] [PubMed] [Google Scholar]
  • 7.Centers for Disease Control and Prevention. 2015. Active bacterial core surveillance report, Emerging Infections Program Network, group B Streptococcus. Centers for Disease Control and Prevention, Atlanta, GA. [Google Scholar]
  • 8.Slotved H-C, Kong F, Lambertsen L, Sauer S, Gilbert GL. 2007. Serotype IX, a proposed new Streptococcus agalactiae serotype. J Clin Microbiol 45:2929–2936. doi: 10.1128/JCM.00117-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Cieslewicz MJ, Chaffin D, Glusman G, Kasper D, Madan A, Rodrigues S, Fahey J, Wessels MR, Rubens CE. 2005. Structural and genetic diversity of group B Streptococcus capsular polysaccharides. Infect Immun 75:3096–3103. doi: 10.1128/IAI.73.5.3096-3103.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Jones N, Bohnsack JF, Takahashi S, Karen A, Chan M, Kunst F, Glaser P, Rusniok C, Crook DWM, Rosalind M, Bisharat N, Spratt BG, Oliver KA, Harding RM. 2003. Multilocus sequence typing system for group B Streptococcus. J Clin Microbiol 41:2530–2536. doi: 10.1128/JCM.41.6.2530-2536.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bohnsack JF, Whiting A, Gottschalk M, Dunn DM, Weiss R, Azimi PH, Philips JB III, Weisman LE, Rhoads GG, Lin F-YC. 2008. Population structure of invasive and colonizing strains of Streptococcus agalactiae from neonates of six U.S. academic centers from 1995 to 1999. J Clin Microbiol 46:1285–1291. doi: 10.1128/JCM.02105-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lin FC, Whiting A, Adderson E, Takahashi S, Dunn DM, Weiss R, Azimi PH, Philips JB, Weisman LE, Regan J, Clark P, Rhoads GG, Frasch CE, Troendle J, Moyer P, Bohnsack JF. 2006. Phylogenetic lineages of invasive and colonizing strains of serotype III group B streptococci from neonates: a multicenter prospective study. J Clin Microbiol 44:1257–1261. doi: 10.1128/JCM.44.4.1257-1261.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Luan S, Granlund M, Sellin M, Lagergård T, Spratt BG, Norgren M. 2005. Multilocus sequence typing of Swedish invasive group B Streptococcus isolates indicates a neonatally associated genetic lineage and capsule switching. J Clin Microbiol 43:3727–3733. doi: 10.1128/JCM.43.8.3727-3733.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Manning SD, Springman AC, Lehotzky E, Lewis MA, Whittam TS, Davies HD. 2009. Multilocus sequence types associated with neonatal group B streptococcal sepsis and meningitis in Canada. J Clin Microbiol 47:1143–1148. doi: 10.1128/JCM.01424-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tazi A, Disson O, Bellais S, Bouaboud A, Dmytruk N, Dramsi S, Mistou M-Y, Khun H, Mechler C, Tardieux I, Trieu-Cuot P, Lecuit M, Poyart C. 2010. The surface protein HvgA mediates group B Streptococcus hypervirulence and meningeal tropism in neonates. J Exp Med 207:2313–2322. doi: 10.1084/jem.20092594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tazi A, Bellais S, Tardieux I, Dramsi S, Trieu-Cuot P, Poyart C. 2012. Group B Streptococcus surface proteins as major determinants for meningeal tropism. Curr Opin Microbiol 15:44–49. doi: 10.1016/j.mib.2011.12.002. [DOI] [PubMed] [Google Scholar]
  • 17.Doran KS, Nizet V. 2004. Molecular pathogenesis of neonatal group B streptococcal infection: no longer in its infancy. Mol Microbiol 54:23–31. doi: 10.1111/j.1365-2958.2004.04266.x. [DOI] [PubMed] [Google Scholar]
  • 18.Katz V, Bowes WA. 1988. Perinatal group B streptococcal infections across intact amniotic membranes. J Reprod Med 33:445–449. [PubMed] [Google Scholar]
  • 19.Schuchat A. 1998. Epidemiology of group B streptococcal disease in the United States: shifting paradigms. Clin Microbiol Rev 11:497–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lin F-YC, Weisman LE, Troendle J, Adams K. 2003. Prematurity is the major risk factor for late-onset group B streptococcus disease. J Infect Dis 188:267–271. doi: 10.1086/376457. [DOI] [PubMed] [Google Scholar]
  • 21.Schrag SJ, Phil D, Zywicki S, Farley MM, Reingold AL, Harrison LH, Lefkowitz LB, Hadler JL, Danila R, Cieslak PR, Schuchat A. 2000. Group B streptococcal disease in the era of intrapartum antibiotic prophylaxis. N Engl J Med 342:15–20. doi: 10.1056/NEJM200001063420103. [DOI] [PubMed] [Google Scholar]
  • 22.Manning SD, Lewis MA, Springman AC, Lehotzky E, Whittam TS, Davies HD. 2008. Genotypic diversity and serotype distribution of group B Streptococcus isolated from women before and after delivery. Clin Infect Dis 46:1829–1837. doi: 10.1086/588296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Benitz WE, Gould JB, Druzin ML. 1999. Risk factors for early-onset group B streptococcal sepsis: estimation of odds ratios by critical literature review. Pediatrics 103:e77 http://pediatrics.aappublications.org/content/103/6/e77.long. [DOI] [PubMed] [Google Scholar]
  • 24.Berardi A, Rossi C, Lugli L, Creti R, Bacchi Reggiani ML, Lanari M, Memo L, Pedna MF, Venturelli C, Perrone E, Ciccia M, Tridapalli E, Piepoli M, Contiero R, Ferrari F. 2013. Group B Streptococcus late-onset disease: 2003–2010. Pediatrics 131:e361–e368. doi: 10.1542/peds.2012-1231. [DOI] [PubMed] [Google Scholar]
  • 25.Kotiw M, Zhang GW, Daggard G, Reiss-Levy E, Tapsall JW, Numa A. 2003. Late-onset and recurrent neonatal group B streptococcal disease associated with breast-milk transmission. Pediatr Dev Pathol 6:251–256. doi: 10.1007/s10024-001-0276-y. [DOI] [PubMed] [Google Scholar]
  • 26.Lanari M, Serra L, Cavrini F, Liguori G, Sambri V. 2007. Late-onset group B streptococcal disease by infected mother's milk detected by polymerase chain reaction. New Microbiol 30:253–254. [PubMed] [Google Scholar]
  • 27.Gagneur A, Héry-Arnaud G, Croly-Labourdette S, Gremmo-Feger G, Vallet S, Sizun J, Quentin R, Tandé D. 2009. Infected breast milk associated with late-onset and recurrent group B streptococcal infection in neonatal twins: a genetic analysis. Eur J Pediatr 168:1155–1158. doi: 10.1007/s00431-008-0903-y. [DOI] [PubMed] [Google Scholar]
  • 28.Doran KS, Benoit VM, Gertz RE, Beall B, Nizet V. 2002. Late-onset group B streptococcal infection in identical twins: insight to disease pathogenesis. J Perinatol 22:326–330. doi: 10.1038/sj.jp.7210675. [DOI] [PubMed] [Google Scholar]
  • 29.Sendi P, Johansson L, Norrby-Teglund A. 2008. Invasive group B streptococcal disease in non-pregnant adults: a review with emphasis on skin and soft-tissue infections. Infection 36:100–111. doi: 10.1007/s15010-007-7251-0. [DOI] [PubMed] [Google Scholar]
  • 30.Kernéis S, Plainvert C, Barnier J-P, Tazi A, Dmytruk N, Gislain B, Loubinoux J, El Sayed F, Cattoir V, Desplaces N, Vernet V, Morand P, Poyart C. 26 April 2017. Clinical and microbiological features associated with group B Streptococcus bone and joint infections, France 2004–2014. Eur J Clin Microbiol Infect Dis doi: 10.1007/s10096-017-2983-y. [DOI] [PubMed] [Google Scholar]
  • 31.Skoff TH, Farley MM, Petit S, Craig AS, Schaffner W, Gershman K, Harrison LH, Lynfield R, Mohle-Boetani J, Zansky S, Albanese BA, Stefonek K, Zell ER, Jackson D, Thompson T, Schrag SJ. 2009. Increasing burden of invasive group B streptococcal disease in nonpregnant adults, 1990–2007. Clin Infect Dis 49:85–92. doi: 10.1086/599369. [DOI] [PubMed] [Google Scholar]
  • 32.Smith EM, Khan MA, Reingold A, Watt JP. 2015. Group B Streptococcus infections of soft tissue and bone in California adults, 1995–2012. Epidemiol Infect 143:3343–3350. doi: 10.1017/S0950268815000606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Landwehr-Kenzel S, Henneke P. 2014. Interaction of Streptococcus agalactiae and cellular innate immunity in colonization and disease. Front Immunol 5:519. doi: 10.3389/fimmu.2014.00519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kumar SKM, Bhat BV. 2016. Distinct mechanisms of the newborn innate immunity. Immunol Lett 173:42–54. doi: 10.1016/j.imlet.2016.03.009. [DOI] [PubMed] [Google Scholar]
  • 35.Basha S, Surendran N, Pichichero M. 2014. Immune responses in neonates. Expert Rev Clin Immunol 10:1171–1184. doi: 10.1586/1744666X.2014.942288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Christensen RD, Macfarlane JL, Taylor NL, Hill HR. 1982. Blood and marrow neutrophils during experimental group B streptococcal infection: quantification of the stem cell, proliferative, storage and circulating pools. Pediatr Res 16:549–553. doi: 10.1203/00006450-198207000-00011. [DOI] [PubMed] [Google Scholar]
  • 37.Urlichs F, Speer CP. 2004. Neutrophil function in preterm and term infants. Neoreviews 5:e417–e430. doi: 10.1542/neo.5-10-e417. [DOI] [Google Scholar]
  • 38.Filias A, Theodorou GL, Mouzopoulou S, Varvarigou AA, Mantagos S, Karakantza M. 2011. Phagocytic ability of neutrophils and monocytes in neonates. BMC Pediatr 11:29. doi: 10.1186/1471-2431-11-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yost CC, Cody MJ, Harris ES, Thornton NL, McInturff AM, Martinez ML, Chandler NB, Rodesch CK, Albertine KH, Petti CA, Weyrich AS, Zimmerman GA. 2009. Impaired neutrophil extracellular trap (NET) formation: a novel innate immune deficiency of human neonates. Blood 113:6419–6427. doi: 10.1182/blood-2008-07-171629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Tagariello G, Iorio A, Mannucci PM. 2009. Delayed but functional neutrophil extracellular trap formation in neonates. Blood 114:4908–4912. doi: 10.1182/blood-2009-09-242388. [DOI] [PubMed] [Google Scholar]
  • 41.Christensen RD, Jensen J, Maheshwari A, Henry E. 2010. Reference ranges for blood concentrations of eosinophils and monocytes during the neonatal period defined from over 63000 records in a multihospital health-care system. J Perinatol 30:540–545. doi: 10.1038/jp.2009.196. [DOI] [PubMed] [Google Scholar]
  • 42.Marchant EA, Kan B, Sharma AA, van Zanten A, Kollmann TR, Brant R, Lavoie PM. 2015. Attenuated innate immune defenses in very premature neonates during the neonatal period. Pediatr Res 78:492–497. doi: 10.1038/pr.2015.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Marodi L, Scorba S, Nagy B. 1980. Chemotactic and random movement of human newborn monocytes. Eur J Pediatr 135:73–75. doi: 10.1007/BF00445897. [DOI] [PubMed] [Google Scholar]
  • 44.Valero N, Mosquera J, Levy A, Añez G, Marcucci R, Alvarez-Mon M. 2014. Differential induction of cytokines by human neonatal, adult, and elderly monocyte/macrophages infected with dengue virus. Viral Immunol 27:151–159. doi: 10.1089/vim.2013.0123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gille C, Leiber A, Mundle I, Spring B, Abele H, Spellerberg B, Hartmann H, Poets CF, Orlikowsky TW. 2009. Phagocytosis and postphagocytic reaction of cord blood and adult blood monocyte after infection with green fluorescent protein-labeled Escherichia coli and group B streptococci. Cytometry B Clin Cytom 76:271–284. doi: 10.1002/cyto.b.20474. [DOI] [PubMed] [Google Scholar]
  • 46.Moraes TJ, Downey GP. 2006. Death of the septic monocyte: is more better? Crit Care 10:146. doi: 10.1186/cc4950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jones CA, Holloway JA, Warner JO. 2002. Phenotype of fetal monocytes and B lymphocytes during the third trimester of pregnancy. J Reprod Immunol 56:45–60. doi: 10.1016/S0165-0378(02)00022-0. [DOI] [PubMed] [Google Scholar]
  • 48.Li YP, Yu SL, Huang ZJ, Huang J, Pan J, Feng X, Zhang XG, Wang JH, Wang J. 2015. An impaired inflammatory cytokine response to gram-negative LPS in human neonates is associated with the defective TLR-mediated signaling pathway. J Clin Immunol 35:218–226. doi: 10.1007/s10875-015-0128-6. [DOI] [PubMed] [Google Scholar]
  • 49.Remington JS, Klein JO, Wilson CB, Nizet V, Maldonando YA (ed). 2010. Infectious diseases of the fetus and newborn infant, 7th ed Elsevier Health Sciences, Philadelphia, PA. [Google Scholar]
  • 50.Winterberg T, Vieten G, Meier T, Yu Y, Busse M, Hennig C, Hansen G, Jacobs R, Ure BM, Kuebler JF. 2015. Distinct phenotypic features of neonatal murine macrophages. Eur J Immunol 45:214–224. doi: 10.1002/eji.201444468. [DOI] [PubMed] [Google Scholar]
  • 51.Speer CP, Gahr M, Wieland M, Eber S. 1988. Phagocytosis-associated functions in neonatal monocyte-derived macrophages. Pediatr Res 24:213–216. doi: 10.1203/00006450-198808000-00015. [DOI] [PubMed] [Google Scholar]
  • 52.Liao S-L, Yeh K-W, Lai S-H, Lee W-I, Huang J-L. 2013. Maturation of Toll-like receptor 1-4 responsiveness during early life. Early Hum Dev 89:473–478. doi: 10.1016/j.earlhumdev.2013.03.013. [DOI] [PubMed] [Google Scholar]
  • 53.Hochrein H, O'Keeffe M. 2008. Dendritic cell subsets and Toll-like receptors. Handb Exp Pharmacol 183:153–179. doi: 10.1007/978-3-540-72167-3_8. [DOI] [PubMed] [Google Scholar]
  • 54.Hunt DW, Huppertz HI, Jiang HJ, Petty RE. 1994. Studies of human cord blood dendritic cells: evidence for functional immaturity. Blood 84:4333–4343. [PubMed] [Google Scholar]
  • 55.De Wit D, Olislagers V, Goriely S, Vermeulen F, Wagner H, Goldman M, Willems F. 2004. Blood plasmacytoid dendritic cell responses to CpG oligodeoxynucleotides are impaired in human newborns. Blood 103:1030–1032. doi: 10.1182/blood-2003-04-1216. [DOI] [PubMed] [Google Scholar]
  • 56.Danis B, George TC, Goriely S, Dutta B, Renneson J, Gatto L, Fitzgerald-Bocarsly P, Marchant A, Goldman M, Willems F, De Wit D. 2008. Interferon regulatory factor 7-mediated responses are defective in cord blood plasmacytoid dendritic cells. Eur J Immunol 38:507–517. doi: 10.1002/eji.200737760. [DOI] [PubMed] [Google Scholar]
  • 57.Willems F, Vollstedt S, Suter M. 2009. Phenotype and function of neonatal DC. Eur J Immunol 39:26–35. doi: 10.1002/eji.200838391. [DOI] [PubMed] [Google Scholar]
  • 58.Sarma JV, Ward PA. 2011. The complement system. Cell Tissue Res 343:227–235. doi: 10.1007/s00441-010-1034-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.McGreal EP, Hearne K, Spiller OB. 2012. Off to a slow start: underdevelopment of the complement system in term newborns is more substantial following premature birth. Immunobiology 217:176–186. doi: 10.1016/j.imbio.2011.07.027. [DOI] [PubMed] [Google Scholar]
  • 60.Belderbos ME, Levy O, Meyaard L, Bont L. 2013. Plasma-mediated immune suppression: a neonatal perspective. Pediatr Allergy Immunol 24:102–113. doi: 10.1111/pai.12023. [DOI] [PubMed] [Google Scholar]
  • 61.Wolach B, Dolfin T, Regev R, Gilboa S, Schlesinger M. 1997. The development of the complement system after 28 weeks' gestation. Acta Paediatr 86:523–527. doi: 10.1111/j.1651-2227.1997.tb08924.x. [DOI] [PubMed] [Google Scholar]
  • 62.Adkins B, Leclerc C, Marshall-Clarke S. 2004. Neonatal adaptive immunity comes of age. Nat Rev Immunol 4:553–564. doi: 10.1038/nri1394. [DOI] [PubMed] [Google Scholar]
  • 63.Zaghouani H, Hoeman CM, Adkins B. 2009. Neonatal immunity: faulty T-helpers and the shortcomings of dendritic cells. Trends Immunol 30:585–591. doi: 10.1016/j.it.2009.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.De Roock S, Stoppelenburg AJ, Scholman R, Hoeks SBEA, Meerding J, Prakken BJ, Boes M. 2013. Defective Th17 development in human neonatal T cells involves reduced RORC2 mRNA content. J Allergy Clin Immunol 132:754.e3–756.e3. doi: 10.1016/j.jaci.2013.04.014. [DOI] [PubMed] [Google Scholar]
  • 65.Debock I, Jaworski K, Chadlaoui H, Delbauve S, Passon N, Twyffels L, Leo O, Flamand V. 2013. Neonatal follicular Th cell responses are impaired and modulated by IL-4. J Immunol 191:1231–1239. doi: 10.4049/jimmunol.1203288. [DOI] [PubMed] [Google Scholar]
  • 66.Viemann D, Schlenke P, Hammers H-J, Kirchner H, Kruse A. 2000. Differential expression of the B cell-restricted molecule CD22 on neonatal B lymphocytes depending upon antigen stimulation. Eur J Immunol 30:550–559. doi:. [DOI] [PubMed] [Google Scholar]
  • 67.Glezen WP. 2003. Effect of maternal antibodies on the infant immune response. Vaccine 21:3389–3392. doi: 10.1016/S0264-410X(03)00339-6. [DOI] [PubMed] [Google Scholar]
  • 68.Jones C, Pollock L, Barnett SM, Battersby A, Kampmann B. 2014. The relationship between concentration of specific antibody at birth and subsequent response to primary immunization. Vaccine 32:996–1002. doi: 10.1016/j.vaccine.2013.11.104. [DOI] [PubMed] [Google Scholar]
  • 69.Currie AJ, Curtis S, Strunk T, Riley K, Liyanage K, Prescott S, Doherty D, Simmer K, Richmond P, Burgner D. 2011. Preterm infants have deficient monocyte and lymphocyte cytokine responses to group B Streptococcus. Infect Immun 79:1588–1596. doi: 10.1128/IAI.00535-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.De Francesco MA, Gargiulo F, Negrini R, Gelmi M, Manca N. 2008. Different sequence strains of Streptococcus agalactiae elicit various levels of cytokine production. Immunol Invest 37:741–751. doi: 10.1080/08820130802403283. [DOI] [PubMed] [Google Scholar]
  • 71.Mancuso G, Midiri A, Beninati C, Biondo C, Galbo R, Akira S, Henneke P, Golenbock D, Teti G. 2004. Dual role of TLR2 and myeloid differentiation factor 88 in a mouse model of invasive group B streptococcal disease. J Immunol 172:6324–6329. doi: 10.4049/jimmunol.172.10.6324. [DOI] [PubMed] [Google Scholar]
  • 72.Henneke P, Takeuchi O, van Strijp JA, Guttormsen HK, Smith JA, Schromm AB, Espevik TA, Akira S, Nizet V, Kasper DL, Golenbock DT. 2001. Novel engagement of CD14 and multiple Toll-like receptors by group B streptococci. J Immunol 167:7069–7076. doi: 10.4049/jimmunol.167.12.7069. [DOI] [PubMed] [Google Scholar]
  • 73.Kenzel S, Mancuso G, Malley R, Teti G, Golenbock DT, Henneke P. 2006. c-Jun kinase is a critical signaling molecule in a neonatal model of group B streptococcal sepsis. J Immunol 176:3181–3188. doi: 10.4049/jimmunol.176.5.3181. [DOI] [PubMed] [Google Scholar]
  • 74.Mancuso G, Gambuzza M, Midiri A, Biondo C, Papasergi S, Akira S, Teti G, Beninati C. 2009. Bacterial recognition by TLR7 in the lysosomes of conventional dendritic cells. Nat Immunol 10:587–594. doi: 10.1038/ni.1733. [DOI] [PubMed] [Google Scholar]
  • 75.Deshmukh SD, Kremer B, Freudenberg M, Bauer S, Golenbock DT, Henneke P. 2011. Macrophages recognize streptococci through bacterial single-stranded RNA. EMBO Rep 12:71–76. doi: 10.1038/embor.2010.189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Deshmukh SD, Müller S, Hese K, Rauch KS, Wennekamp J, Takeuchi O, Akira S, Golenbock DT, Henneke P. 2012. NO is a macrophage autonomous modifier of the cytokine response to streptococcal single-stranded RNA. J Immunol 188:774–780. doi: 10.4049/jimmunol.1101383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Talati AJ, Kim HJ, Kim YI, Yi AK, English BK. 2008. Role of bacterial DNA in macrophage activation by group B streptococci. Microbes Infect 10:1106–1113. doi: 10.1016/j.micinf.2008.06.001. [DOI] [PubMed] [Google Scholar]
  • 78.Charrel-Dennis M, Latz E, Halmen KA, Trieu-Cuot P, Fitzgerald KA, Kasper DL, Golenbock DT. 2008. TLR-independent type I interferon induction in response to an extracellular bacterial pathogen via intracellular recognition of its DNA. Cell Host Microbe 4:543–554. doi: 10.1016/j.chom.2008.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Andrade WA, Firon A, Schmidt T, Hornung V, Fitzgerald KA, Kurt-Jones EA, Trieu-Cuot P, Golenbock DT, Kaminski PA. 2016. Group B Streptococcus degrades cyclic-di-AMP to modulate STING-dependent type I interferon production. Cell Host Microbe 20:49–59. doi: 10.1016/j.chom.2016.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Levy O, Jean-Jacques RM, Cywes C, Sisson RB, Zarember KA, Godowski PJ, Christianson JL, Guttormsen H, Carroll MC, Nicholson-Weller A, Wessels MR. 2003. Critical role of the complement system in group B Streptococcus-induced tumor necrosis factor alpha release. Infect Immun 71:6344–6353. doi: 10.1128/IAI.71.11.6344-6353.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Spellerberg B, Pohl B, Haase G, Martin S, Weber-Heynemann J, Lütticken R. 1999. Identification of genetic determinants for the hemolytic activity of Streptococcus agalactiae by ISS1 transposition. J Bacteriol 181:3212–3219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Whidbey C, Harrell MI, Burnside K, Ngo L, Becraft AK, Iyer LM, Aravind L, Hitti J, Waldorf KMA, Rajagopal L. 2013. A hemolytic pigment of group B Streptococcus allows bacterial penetration of human placenta. J Exp Med 210:1265–1281. doi: 10.1084/jem.20122753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ring A, Depnering C, Pohl J, Nizet V, Shenep JL, Stremmel W. 2002. Synergistic action of nitric oxide release from murine macrophages caused by group B streptococcal cell wall and beta-hemolysin/cytolysin. J Infect Dis 186:1518–1521. doi: 10.1086/344895. [DOI] [PubMed] [Google Scholar]
  • 84.Whidbey C, Vornhagen J, Gendrin C, Boldenow E, Samson JM, Doering K, Ngo L, Ezekwe EAD, Gundlach JH, Elovitz MA, Liggitt D, Duncan JA, Adams Waldorf KM, Rajagopal L. 2015. A streptococcal lipid toxin induces membrane permeabilization and pyroptosis leading to fetal injury. EMBO Mol Med 7:488–505. doi: 10.15252/emmm.201404883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Boldenow E, Gendrin C, Ngo L, Bierle C, Vornhagen J, Coleman M, Merillat S, Armistead B, Whidbey C, Alishetti V, Santana-Ufret V, Ogle J, Gough M, Srinouanprachanh S, MacDonald JW, Bammler TK, Bansal A, Liggitt HD, Rajagopal L, Waldorf KMA. 2017. Group B Streptococcus circumvents neutrophils and neutrophil extracellular traps during amniotic cavity invasion and preterm labor. Sci Immunol 1:eaah4576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Costa A, Gupta R, Signorino G, Malara A, Cardile F, Biondo C, Midiri A, Galbo R, Trieu-Cuot P, Papasergi S, Teti G, Henneke P, Mancuso G, Golenbock DT, Beninati C. 2012. Activation of the NLRP3 inflammasome by group B streptococci. J Immunol 188:1953–1960. doi: 10.4049/jimmunol.1102543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Gupta R, Ghosh S, Monks B, DeOliveira RB, Tzeng TC, Kalantari P, Nandy A, Bhattacharjee B, Chan J, Ferreira F, Rathinam V, Sharma S, Lien E, Silverman N, Fitzgerald K, Firon A, Trieu-Cuot P, Henneke P, Golenbock DT. 2014. RNA and B-hemolysin of group B Streptococcus induce interleukin-1B (IL-1B) by activating NLRP3 inflammasomes in mouse macrophages. J Biol Chem 289:13701–13705. doi: 10.1074/jbc.C114.548982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Rubens CE, Wessels MR, Heggen LM, Kasper DL. 1987. Transposon mutagenesis of type III group B Streptococcus: correlation of capsule expression with virulence. Proc Natl Acad Sci U S A 84:7208–7212. doi: 10.1073/pnas.84.20.7208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wessels MR, Rubens CE, Benedí VJ, Kasper DL. 1989. Definition of a bacterial virulence factor: sialylation of the group B streptococcal capsule. Proc Natl Acad Sci U S A 86:8983–8987. doi: 10.1073/pnas.86.22.8983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Carlin AF, Uchiyama S, Chang YC, Lewis AL, Nizet V, Varki A. 2009. Molecular mimicry of host sialylated glycans allows a bacterial pathogen to engage neutrophil Siglec-9 and dampen the innate immune response. Blood 113:3333–3336. doi: 10.1182/blood-2008-11-187302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Carlin AF, Chang Y-C, Areschoug T, Lindahl G, Hurtado-Ziola N, King CC, Varki A, Nizet V. 2009. Group B Streptococcus suppression of phagocyte functions by protein-mediated engagement of human Siglec-5. J Exp Med 206:1691–1699. doi: 10.1084/jem.20090691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ali SR, Fong JJ, Carlin AF, Busch TD, Linden R, Angata T, Areschoug T, Parast M, Varki N, Murray J, Nizet V, Varki A. 2014. Siglec-5 and Siglec-14 are polymorphic paired receptors that modulate neutrophil and amnion signaling responses to group B Streptococcus. J Exp Med 211:1231–1242. doi: 10.1084/jem.20131853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Chang Y-C, Olson J, Beasley FC, Tung C, Zhang J, Crocker PR, Varki A, Nizet V. 2014. Group B Streptococcus engages an inhibitory siglec through sialic acid mimicry to blunt innate immune and inflammatory responses in vivo. PLoS Pathog 10:e1003846. doi: 10.1371/journal.ppat.1003846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Chang Y-C, Olson J, Louie A, Crocker PR, Varki A, Nizet V. 2014. Role of macrophage sialoadhesin in host defense against the sialylated pathogen group B Streptococcus. J Mol Med 92:951–959. doi: 10.1007/s00109-014-1157-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Harris TO, Shelver DW, Bohnsack JF, Rubens CE. 2003. A novel streptococcal surface protease promotes virulence, resistance to opsonophagocytosis, and cleavage of human fibrinogen. J Clin Invest 111:61–70. doi: 10.1172/JCI200316270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Marques MB, Kasper DL, Pangburn MK, Wessels MR. 1992. Prevention of C3 deposition by capsular polysaccharide is a virulence mechanism of type III group B streptococci. Infect Immun 60:3986–3993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Santi I, Scarselli M, Mariani M, Pezzicoli A, Masignani V, Taddei A, Grandi G, Telford JL, Soriani M. 2007. BibA: a novel immunogenic bacterial adhesin contributing to group B Streptococcus survival in human blood. Mol Microbiol 63:754–767. doi: 10.1111/j.1365-2958.2006.05555.x. [DOI] [PubMed] [Google Scholar]
  • 98.Pietrocola G, Rindi S, Rosini R, Buccato S, Speziale P, Margarit I. 2016. The group B Streptococcus-secreted protein CIP interacts with C4, preventing C3b deposition via the lectin and classical complement pathways. J Immunol 196:385–394. doi: 10.4049/jimmunol.1501954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Jarva H, Hellwage J, Jokiranta TS, Lehtinen MJ, Zipfel PF, Meri S. 2004. The group B streptococcal beta and pneumococcal Hic proteins are structurally related immune evasion molecules that bind the complement inhibitor factor H in an analogous fashion. J Immunol 172:3111–3118. doi: 10.4049/jimmunol.172.5.3111. [DOI] [PubMed] [Google Scholar]
  • 100.Maruvada R, Blom AM, Prasadarao NV. 2008. Effects of complement regulators bound to Escherichia coli K1 and group B Streptococcus on the interaction with host cells. Immunology 124:265–276. doi: 10.1111/j.1365-2567.2007.02764.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Cheng Q, Stafslien D, Purushothaman SS, Cleary P. 2002. The group B streptococcal C5a peptidase is both a specific protease and an invasin. Infect Immun 70:2408–2413. doi: 10.1128/IAI.70.5.2408-2413.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Bryan JD, Shelver DW. 2009. Streptococcus agalactiae CspA is a serine protease that inactivates chemokines. J Bacteriol 191:1847–1854. doi: 10.1128/JB.01124-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Kothary V, Doster RS, Rogers LM, Kirk LA, Boyd KL, Romano-Keeler J, Haley KP, Manning SD, Aronoff DM, Gaddy JA. 2017. Group B Streptococcus induces neutrophil recruitment to gestational tissues and elaboration of extracellular traps and nutritional immunity. Front Cell Infect Microbiol 7:19. doi: 10.3389/fcimb.2017.00019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Amulic B, Hayes G. 2011. Neutrophil extracellular traps. Curr Biol 21:R297–R298. doi: 10.1016/j.cub.2011.03.021. [DOI] [PubMed] [Google Scholar]
  • 105.Derré-Bobillot A, Cortes-Perez NG, Yamamoto Y, Kharrat P, Couvé E, Da Cunha V, Decker P, Boissier MC, Escartin F, Cesselin B, Langella P, Bermúdez-Humarán LG, Gaudu P. 2013. Nuclease A (Gbs0661), an extracellular nuclease of Streptococcus agalactiae, attacks the neutrophil extracellular traps and is needed for full virulence. Mol Microbiol 89:518–531. doi: 10.1111/mmi.12295. [DOI] [PubMed] [Google Scholar]
  • 106.Jiang D, Liang J, Noble PW. 2007. Hyaluronan in tissue injury and repair. Annu Rev Cell Dev Biol 23:435–461. doi: 10.1146/annurev.cellbio.23.090506.123337. [DOI] [PubMed] [Google Scholar]
  • 107.Taylor KR, Trowbridge JM, Rudisill JA, Termeer CC, Simon JC, Gallo RL. 2004. Hyaluronan fragments stimulate endothelial recognition of injury through TLR4. J Biol Chem 279:17079–17084. doi: 10.1074/jbc.M310859200. [DOI] [PubMed] [Google Scholar]
  • 108.Scheibner KA, Lutz MA, Boodoo S, Fenton MJ, Powell JD, Horton MR. 2006. Hyaluronan fragments act as an endogenous danger signal by engaging TLR2. J Immunol 177:1272–1281. doi: 10.4049/jimmunol.177.2.1272. [DOI] [PubMed] [Google Scholar]
  • 109.Wang Z, Guo C, Xu Y, Liu G, Lu C, Liu Y. 2014. Two novel functions of hyaluronidase from Streptococcus agalactiae are enhanced intracellular survival and inhibition of proinflammatory cytokine expression. Infect Immun 82:2615–2625. doi: 10.1128/IAI.00022-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kolar SL, Kyme P, Tseng CW, Soliman A, Kaplan A, Liang J, Nizet V, Jiang D, Murali R, Arditi M, Underhill DM, Liu GY. 2015. Group B Streptococcus evades host immunity by degrading hyaluronan. Cell Host Microbe 18:694–704. doi: 10.1016/j.chom.2015.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Valentin-Weigand P, Benkel P, Rohde M, Chhatwal GS. 1996. Entry and intracellular survival of group B streptococci in J774 macrophages. Infect Immun 64:2467–2473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Fujieda M, Aoyagi Y, Mastubara K, Takeuchi Y, Fujimaki W, Matsuchita M, Bohnsack J, Takahashi S. 2012. L-ficolin and capsular polysaccharide-specific IgG in cord serum contribute synergistically to opsonophagocytic killing of serotype III and V group B streptococci. Infect Immun 80:2053–2060. doi: 10.1128/IAI.06232-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Antal JM, Cunningham JV, Goodrum KJ. 1992. Opsonin-independent phagocytosis of group B streptococci: role of complement receptor type three. Infect Immun 60:1114–1121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Korir ML, Laut C, Rogers LM, Plemmons JA, Aronoff DM, Manning SD. 28 October 2016. Differing mechanisms of surviving phagosomal stress among group B Streptococcus strains of varying genotypes. Virulence doi: 10.1080/21505594.2016.1252016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Albanyan EA, Edwards MS. 2000. Lectin site interaction with capsular polysaccharide mediates nonimmune phagocytosis of type III group B streptococci. Infect Immun 68:5794–5802. doi: 10.1128/IAI.68.10.5794-5802.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.DeLeo FR. 2004. Modulation of phagocyte apoptosis by bacterial pathogens. Apoptosis 9:399–413. doi: 10.1023/B:APPT.0000031448.64969.fa. [DOI] [PubMed] [Google Scholar]
  • 117.Rock KL, Kono H. 2008. The inflammatory response to cell death. Annu Rev Pathol 3:99–126. doi: 10.1146/annurev.pathmechdis.3.121806.151456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Elmore S. 2007. Apoptosis: a review of programmed cell death. Toxicol Pathol 35:495–516. doi: 10.1080/01926230701320337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Fettucciari K, Rosati E, Scaringi L, Cornacchione P, Migliorati G, Sabatini R, Fetriconi I, Rossi R, Marconi P. 2000. Group B Streptococcus induces apoptosis in macrophages. J Immunol 165:3923–3933. doi: 10.4049/jimmunol.165.7.3923. [DOI] [PubMed] [Google Scholar]
  • 120.Fettucciari K, Fetriconi I, Bartoli A, Rossi R, Marconi P. 2003. Involvement of mitogen-activated protein kinases in group B Streptococcus-induced macrophage apoptosis. Pharmacol Res 47:355–362. doi: 10.1016/S1043-6618(03)00004-5. [DOI] [PubMed] [Google Scholar]
  • 121.Ulett GC, Adderson EE. 2005. Nitric oxide is a key determinant of group B Streptococcus-induced murine macrophage apoptosis. J Infect Dis 191:1761–1770. doi: 10.1086/429693. [DOI] [PubMed] [Google Scholar]
  • 122.Korir ML, Knupp D, LeMerise K, Boldenow E, Loch-Caruso R, Aronoff DM, Manning SD. 2014. Association and virulence gene expression vary among serotype III group B Streptococcus isolates following exposure to decidual and lung epithelial cells. Infect Immun 82:4587–4595. doi: 10.1128/IAI.02181-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Kling DE, Tsvang I, Murphy MP, Newburg DS. 2013. Group B Streptococcus induces a caspase-dependent apoptosis in fetal rat lung interstitium. Microb Pathog 61–62:1–10. doi: 10.1016/j.micpath.2013.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Fettucciari K, Fetriconi I, Mannucci R, Nicoletti I, Bartoli A, Coaccioli S, Marconi P. 2006. Group B Streptococcus induces macrophage apoptosis by calpain activation. J Immunol 176:7542–7556. doi: 10.4049/jimmunol.176.12.7542. [DOI] [PubMed] [Google Scholar]
  • 125.Fettucciari K, Quotadamo F, Noce R, Palumbo C, Modesti A, Rosati E, Mannucci R, Bartoli A, Marconi P. 2011. Group B Streptococcus (GBS) disrupts by calpain activation the actin and microtubule cytoskeleton of macrophages. Cell Microbiol 13:859–884. doi: 10.1111/j.1462-5822.2011.01584.x. [DOI] [PubMed] [Google Scholar]
  • 126.Fettucciari K, Ponsini P, Palumbo C, Rosati E, Mannucci R, Bianchini R, Modesti A, Marconi P. 2015. Macrophage induced gelsolin in response to group B Streptococcus (GBS) infection. Cell Microbiol 17:79–104. doi: 10.1111/cmi.12338. [DOI] [PubMed] [Google Scholar]
  • 127.Oliveira L, Madureira P, Andrade EB, Bouaboud A, Morello E, Ferreira P, Poyart C, Trieu-Cuot P, Dramsi S. 2012. Group B Streptococcus GAPDH is released upon cell lysis, associates with bacterial surface, and induces apoptosis in murine macrophages. PLoS One 7:e29963. doi: 10.1371/journal.pone.0029963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Flannagan RS, Cosío G, Grinstein S. 2009. Antimicrobial mechanisms of phagocytes and bacterial evasion strategies. Nat Rev Microbiol 7:355–366. doi: 10.1038/nrmicro2128. [DOI] [PubMed] [Google Scholar]
  • 129.Thi EP, Lambertz U, Reiner NE. 2012. Sleeping with the enemy: how intracellular pathogens cope with a macrophage lifestyle. PLoS Pathog 8:e1002551. doi: 10.1371/journal.ppat.1002551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Cumley NJ, Smith LM, Anthony M, May RC. 2012. The CovS/CovR acid response regulator is required for intracellular survival of group B Streptococcus in macrophages. Infect Immun 80:1650–1661. doi: 10.1128/IAI.05443-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Kaplan EL, Chhatwal GS, Rohde M. 2006. Reduced ability of penicillin to eradicate ingested group A streptococci from epithelial cells: clinical and pathogenetic implications. Clin Infect Dis 43:1398–1406. doi: 10.1086/508773. [DOI] [PubMed] [Google Scholar]
  • 132.Thwaites GE, Gant V. 2011. Are bloodstream leukocytes Trojan horses for the metastasis of Staphylococcus aureus? Nat Rev Microbiol 9:215–222. doi: 10.1038/nrmicro2508. [DOI] [PubMed] [Google Scholar]
  • 133.Cornacchione P, Scaringi L, Fettucciari K, Rosati E, Sabatini R, Orefici G, von Hunolstein C, Modesti A, Modica A, Minelli F, Marconi P. 1998. Group B streptococci persist inside macrophages. Immunology 93:86–95. doi: 10.1046/j.1365-2567.1998.00402.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Susta F, Chiasserini D, Fettucciari K, Orvietani PL, Quotadamo F, Noce R, Bartoli A, Marconi P, Corazzi L, Binaglia L. 2010. Protein expression changes induced in murine peritoneal macrophages by group B Streptococcus. Proteomics 10:2099–2112. doi: 10.1002/pmic.200900642. [DOI] [PubMed] [Google Scholar]
  • 135.Poyart C, Pellegrini E, Gaillot O, Boumaila C, Baptista M, Trieu-Cuot P. 2001. Contribution of Mn-cofactored superoxide dismutase (SodA) to the virulence of Streptococcus agalactiae. Infect Immun 68:5098–5106. doi: 10.1128/IAI.69.8.5098-5106.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Glaser P, Rusniok C, Buchrieser C, Chevalier F, Frangeul L, Msadek T, Zouine M, Couvé E, Lalioui L, Poyart C, Trieu-Cuot P, Kunst F. 2002. Genome sequence of Streptococcus agalactiae, a pathogen causing invasive neonatal disease. Mol Microbiol 45:1499–1513. doi: 10.1046/j.1365-2958.2002.03126.x. [DOI] [PubMed] [Google Scholar]
  • 137.Janowiak BE, Griffith OW. 2005. Glutathione synthesis in Streptococcus agalactiae: one protein accounts for gamma-glutamylcysteine synthetase and glutathione synthetase activities. J Biol Chem 280:11829–11839. doi: 10.1074/jbc.M414326200. [DOI] [PubMed] [Google Scholar]
  • 138.Masip L, Veeravalli K, Georgiou G. 2006. The many faces of glutathione in bacteria. Antioxid Redox Signal 8:753–762. doi: 10.1089/ars.2006.8.753. [DOI] [PubMed] [Google Scholar]
  • 139.Liu GY, Doran KS, Lawrence T, Turkson N, Puliti M, Tissi L, Nizet V. 2004. Sword and shield: linked group B streptococcal beta-hemolysin/cytolysin and carotenoid pigment function to subvert host phagocyte defense. Proc Natl Acad Sci U S A 101:14491–14496. doi: 10.1073/pnas.0406143101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Maisey HC, Doran KS, Nizet V. 2008. Recent advances in understanding the molecular basis of group B Streptococcus virulence. Expert Rev Mol Med 10:e27. doi: 10.1017/S1462399408000811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Hamilton A, Popham DL, Carl DJ, Lauth X, Nizet V, Jones AL. 2006. Penicillin-binding protein 1a promotes resistance of group B Streptococcus to antimicrobial peptides. Infect Immun 74:6179–6187. doi: 10.1128/IAI.00895-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Poyart C, Lamy M-C, Boumaila C, Fiedler F, Trieu-Cuot P. 2001. Regulation of d-alanyl-lipoteichoic acid biosynthesis in Streptococcus agalactiae involves a novel two-component regulatory system. J Bacteriol 183:6324–6334. doi: 10.1128/JB.183.21.6324-6334.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Saar-Dover R, Bitler A, Nezer R, Shmuel-Galia L, Firon A, Shimoni E, Trieu-Cuot P, Shai Y. 2012. d-Alanylation of lipoteichoic acids confers resistance to cationic peptides in group B Streptococcus by increasing the cell wall density. PLoS Pathog 8:e1002891. doi: 10.1371/journal.ppat.1002891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Rosini R, Rinaudo CD, Soriani M, Lauer P, Mora M, Maione D, Taddei A, Santi I, Ghezzo C, Brettoni C, Buccato S, Margarit I, Grandi G, Telford JL. 2006. Identification of novel genomic islands coding for antigenic pilus-like structures in Streptococcus agalactiae. Mol Microbiol 61:126–141. doi: 10.1111/j.1365-2958.2006.05225.x. [DOI] [PubMed] [Google Scholar]
  • 145.Maisey HC, Quach D, Hensler ME, Liu GY, Gallo RL, Nizet V, Doran KS. 2008. A group B streptococcal pilus protein promotes phagocyte resistance and systemic virulence. FASEB J 22:1715–1724. doi: 10.1096/fj.07-093963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Papasergi S, Brega S, Mistou MY, Firon A, Oxaran V, Dover R, Teti G, Shai Y, Trieu-Cuot P, Dramsi S. 2011. The GBS PI-2a pilus is required for virulence in mice neonates. PLoS One 6:e18747. doi: 10.1371/journal.pone.0018747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Chattopadhyay D, Carey AJ, Caliot E, Webb RI, Layton JR, Wang Y, Bohnsack JF, Adderson EE, Ulett GC. 2011. Phylogenetic lineage and pilus protein Spb1/SAN1518 affect opsonin-independent phagocytosis and intracellular survival of group B Streptococcus. Microbes Infect 13:369–382. doi: 10.1016/j.micinf.2010.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Six A, Bellais S, Bouaboud A, Fouet A, Gabriel C, Tazi A, Dramsi S, Trieu-Cuot P, Poyart C. 2015. Srr2, a multifaceted adhesin expressed by ST-17 hypervirulent group B Streptococcus involved in binding to both fibrinogen and plasminogen. Mol Microbiol 97:1209–1222. doi: 10.1111/mmi.13097. [DOI] [PubMed] [Google Scholar]
  • 149.Santi I, Grifantini R, Jiang S-M, Brettoni C, Grandi G, Wessels MR, Soriani M. 2009. CsrRS regulates group B Streptococcus virulence gene expression in response to environmental pH: a new perspective on vaccine development. J Bacteriol 191:5387–5397. doi: 10.1128/JB.00370-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Baker CJ, Kasper DL. 1976. Correlation of maternal antibody deficiency with susceptibility to neonatal group B streptococcal infection. N Engl J Med 294:753–756. doi: 10.1056/NEJM197604012941404. [DOI] [PubMed] [Google Scholar]
  • 151.Davies HD, Adair C, McGeer A, Ma D, Robertson S, Mucenski M, Kowalsky L, Tyrell G, Baker CJ. 2001. Antibodies to capsular polysaccharides of group B Streptococcus in pregnant Canadian women: relationship to colonization status and infection in the neonate. J Infect Dis 184:285–291. doi: 10.1086/322029. [DOI] [PubMed] [Google Scholar]
  • 152.Woof JM, Kerr MA. 2006. The function of immunoglobulin A in immunity. J Pathol 208:270–282. doi: 10.1002/path.1877. [DOI] [PubMed] [Google Scholar]
  • 153.Pleass RJ, Areschoug T, Lindahl G, Woof JM. 2001. Streptococcal IgA-binding proteins bind in the Cα2-Cα3 interdomain region and inhibit binding of IgA to human CD89. J Biol Chem 276:8197–8204. doi: 10.1074/jbc.M009396200. [DOI] [PubMed] [Google Scholar]
  • 154.Johri AK, Paoletti LC, Glaser P, Dua M, Sharma PK, Grandi G, Rappuoli R. 2006. Group B Streptococcus: global incidence and vaccine development. Nat Rev Microbiol 4:932–942. doi: 10.1038/nrmicro1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Manning SD, Wood S, Kasha K, Martin D, Rioux S, Brodeur B, Davies HD. 2006. Naturally occurring antibodies for the group B streptococcal surface immunogenic protein (Sip) in pregnant women and newborn babies. Vaccine 24:6905–6912. doi: 10.1016/j.vaccine.2006.06.020. [DOI] [PubMed] [Google Scholar]
  • 156.Fabbrini M, Rigat F, Rinaudo CD, Passalaqua I, Khacheh S, Creti R, Baldassarri L, Carboni F, Anderloni G, Rosini R, Maione D, Grandi G, Telford JL, Margarit I. 2016. The protective value of maternal group B Streptococcus antibodies: quantitative and functional analysis of naturally acquired responses to capsular polysaccharides and pilus proteins in European maternal sera. Clin Infect Dis 63:746–753. doi: 10.1093/cid/ciw377. [DOI] [PubMed] [Google Scholar]
  • 157.Chen VL, Avci FY, Kasper DL. 2013. A maternal vaccine against group B Streptococcus: past, present, and future. Vaccine 31:D13–D19. doi: 10.1016/j.vaccine.2012.12.080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Nuccitelli A, Rinaudo CD, Maione D. 2015. Group B Streptococcus vaccine: state of the art. Ther Adv Vaccines 3:76–90. doi: 10.1177/2051013615579869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Heath PT. 2016. Status of vaccine research and development of vaccines for GBS. Vaccine 34:2876–2879. doi: 10.1016/j.vaccine.2015.12.072. [DOI] [PubMed] [Google Scholar]
  • 160.Mestas J, Hughes CCW. 2004. Of mice and not men: differences between mouse and human immunology. J Immunol 172:2731–2738. doi: 10.4049/jimmunol.172.5.2731. [DOI] [PubMed] [Google Scholar]
  • 161.Shultz LD, Brehm MA, Garcia-Martinez JV, Greiner DL. 2012. Humanized mice for immune system investigation: progress, promise and challenges. Nat Rev Immunol 12:786–798. doi: 10.1038/nri3311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Ernst W, Zimara N, Hanses F, Männel DN, Seelbach-Göbel B, Wege AK. 2013. Humanized mice, a new model to study the influence of drug treatment on neonatal sepsis. Infect Immun 81:1520–1531. doi: 10.1128/IAI.01235-12. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical Microbiology Reviews are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES