You are on page 1of 8

INFECTION AND IMMUNITY, Apr. 2006, p. 1999–2006 Vol. 74, No.

4
0019-9567/06/$08.00⫹0 doi:10.1128/IAI.74.4.1999–2006.2006
Copyright © 2006, American Society for Microbiology. All Rights Reserved.

MINIREVIEW
Neonatal Innate Immunity to Infectious Agents
László Maródi*
Department of Infectious and Pediatric Immunology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary

Host defenses to microbial invasion include the phylogeneti- responses for innate immune recognition are encoded in the
cally older but rapidly developing antigen-independent or in- germ line DNA and, in contrast to adaptive immune responses,
nate immunity and the much more slowly developing specific do not require gene rearrangement (35).
or adaptive immunity (2, 35, 82, 91). Innate immune responses
are triggered by bacteria, viruses, protozoa, and fungi, as non-
NEONATAL INNATE IMMUNITY
self, and involve nonspecific activation of neutrophils, mono-
cytes and macrophages, dendritic cells (DCs), natural killer Human neonates and young infants are more vulnerable
(NK) cells, and complement. The importance of innate immu- to infectious agents than older children and adults and are
nity in defense against mycobacteria is illustrated by the ob- especially susceptible to infections with intracellular patho-

Downloaded from iai.asm.org by on October 29, 2008


servation that patients with T-cell-negative, B-cell-negative, gens. Some of the pathogens causing infections in utero, intra-
and NK cell-positive severe combined immunodeficiency partum, and postpartum evoke fetal and neonatal innate im-
(SCID) may survive inadvertent vaccination with bacillus mune responses. These agents include group B streptococci
Calmette-Guérin vaccine (64). (GBS), Escherichia coli, Listeria monocytogenes, herpes simplex
Phagocytosis as a mechanism of innate immune defense has virus (HSV), cytomegalovirus (CMV), Epstein-Barr virus
served as the classical model for studying host-parasite inter- (EBV), varicella-zoster virus (VZV), respiratory syncytial virus
actions, and significant progress has been made toward under- (RSV), Toxoplasma gondii, and Candida species. Innate immu-
standing the molecular mechanisms of phagocytic uptake and nity against these pathogens represents the critical first-line
microbial killing (19, 25, 54, 57, 59). Recently, Toll-like recep- barrier of host defenses, as newborns have a naı̈ve adaptive
tors (TLRs) have emerged as central points of innate immunity immune system. The past decade has brought great strides in
(82, 91). TLRs represent a conserved family of immune recep- our understanding of innate immune mechanisms in humans.
tor sensing molecules on a wide variety of pathogens. These An increasing body of evidence suggests that neonatal innate
receptors recognize pathogen-associated molecular patterns, responses may not be fully developed, allowing early dissemi-
which results in activation of NF-␬B and other transcription nation of infections. This review describes recent advances and
factors including interferon (IFN) regulatory factors. TLRs are current understanding of innate neonatal immunity to infec-
expressed on the surface of monocytes, macrophages, DCs, tious agents that are thought to be responsible for significant
and epithelial cells or in the cytoplasm of cells from different morbidity and mortality in newborns. Neonatal infection by
tissues. Other immune receptors involved in innate immune sexually transmitted disease pathogens (Treponema pallidum,
responses are the macrophage mannose receptor (MR) and Neisseria gonorrheae, and Chlamydia trachomatis), human im-
dectin-1 (25, 93). Ligand binding to innate receptors generates munodeficiency virus (HIV), and hepatitis viruses will not be
intracellular signals, initiates gene activation, and enhances the discussed here. A better understanding of molecular mecha-
release of cytokines and chemokines at the site of immune nisms that underlie neonatal immune functions may improve
activation. Chemokines recruit innate immune effector cells our ability to prevent and treat neonatal infections.
such as granulocytes, monocytes, macrophages, and NK cells
(32, 63, 65). An important humoral component of innate im-
munity is the complement system, which can be activated INNATE RESPONSES TO NEONATAL PATHOGENS
through the alternative and lectin pathways, in addition to the GBS. GBS is the foremost cause of neonatal bacterial infec-
classical pathway, leading to antibody-independent opsoniza- tions, and mortality of invasive GBS disease in newborns re-
tion and opsonophagocytosis (55, 75). mains high despite advances in intensive care and susceptibility
Innate immunity is ontogenetically older than adaptive im- of the pathogen to penicillin and gentamicin (16, 78). The lack
munity, but innate recognition of pathogens is the first step in of pathogen-specific maternal antibodies to the capsular poly-
inducing adaptive immunity (35). In vertebrates, innate and saccharide antigen was supposed to contribute to susceptibility
adaptive immunity are overlapping and intervening. One major and the severe course of disease in newborns. Accordingly, it
difference in the biology of the two systems is that several was believed that effective vaccination would be a way to re-
duce the incidence of GBS disease over the long term. In a
study including 321 healthy term newborns, immunoglobulin G
* Corresponding author. Mailing address: Department of Infectious
and Pediatric Immunology, Medical and Health Science Center, Univer-
(IgG) antibodies against capsular polysaccharides of GBS se-
sity of Debrecen, Nagyerdei Krt. 98, H-4012 Debrecen, Hungary. Phone: rotypes Ia, II, and III were present in 98 to 100% of cord sera
36 52 416 841. Fax: 36 52 430 323. E-mail: lmarodi@jaguar.dote.hu. (10). However, the concentrations of IgG antibodies were of-

1999
2000 MINIREVIEW INFECT. IMMUN.

ten low and might not have been sufficient for protection. The capsule of GBS is well characterized as one of the
Naturally occurring IgG antibodies with the capacity to opson- virulence factors of streptococci. The capsule protects GBS
ize GBS type III in a complement-dependent manner may also from opsonization by C3 through inhibition of the alternative
play a role in host defense against these pathogens (22). complement pathway in the absence of type-specific antibodies
Neutrophils are the predominant mobile phagocytes of cir- (76). In addition, streptococcal proteins localized to the sur-
culating blood and may contribute to killing of GBS even more face of bacteria may bind complement factor H, retaining its
than mononuclear phagocytes do. Importantly, exposure to ability to down-regulate complement activation (6). Recently,
recombinant human IFN-␥ was found to activate cord blood a surface-localized protease, CspA, that may play an important
neutrophils and to result in enhanced chemotaxis and in- role in GBS pathogenesis as an antiphagocytic surface factor
creased concentrations of free intracellular calcium (31). was described (28). CspA was found to be required for GBS
These data suggest that IFN-␥ may enhance the newborns’ cleavage of human fibrinogen. GBS mutants that failed to
own host defense by activating neutrophils. express cspA, the gene coding for CspA, displayed a signifi-
Monocytes and macrophages have a rich diversity of cell cantly decreased virulence in a neonatal rat model of GBS
surface receptors complementing the diversity of microbial infection and an increased sensitivity to opsonophagocytic kill-
molecules that they are likely to encounter, often in the context ing. Further characterization of the expression and function of
of soluble opsonins such as complement and antibodies. Ear- surface-localized GBS proteins and enzymes will help us to
lier studies showed that the capacity of cord blood monocytes understand better how GBS that evade the host innate im-
to kill serum-opsonized GBS type III was decreased compared mune response cause severe infections in newborns.
to the capacity of adult blood monocytes (56). Interactions E. coli. E. coli is one of the leading gram-negative bacteria
between serum-opsonized GBS and monocyte-derived macro- that cause neonatal meningitis and sepsis (84). The mortality
phages isolated from cord blood were also studied by using rate and the neurological squeal remain high despite advances

Downloaded from iai.asm.org by on October 29, 2008


resident and cytokine-activated cells (53). These results in antimicrobial therapy. Intracellular survival of E. coli rep-
showed that resident cord and adult macrophages efficiently resents one important pathogenicity mechanism. E. coli K1,
which causes meningitis in neonates, is able to enter and sur-
phagocytosed serum-opsonized GBS, but the ingested bacteria
vive in human macrophages and peripheral blood monocytes
survived inside the cells. Bacterial killing by cord macrophages
(85). Outer membrane protein A (OmpA) expression on the
was augmented by granulocyte-macrophage colony-stimulating
surface of bacteria plays an important role in binding to and
factor but not by IFN-␥, suggesting differential modulation of
phagocytosis by macrophages in the absence of opsonization.
bacterial killing by these cytokines. Survival of GBS in neonatal
E. coli expressing OmpA is able to bind the classical comple-
macrophages provides an additional explanation to the severity
ment fluid-phase regulator C4b-binding protein to avoid depo-
of GBS disease in early life.
sition of C3 and C5, subsequent phagocytosis by granulocytes,
GBS types Ia and III may impair microbicidal systems in
and activation of the membrane attack complex (69). In addi-
murine macrophages by inhibiting protein kinase C-dependent
tion, IgG does not bind efficiently to the surface of E. coli K1,
signal transduction pathways (17). Alternatively, macrophages
allowing the bacteria to avoid recognition via the Fc␥ receptors
may fail to kill GBS unless they are activated. After phago-
of granulocytes. Deficiency of the alternative complement
cytosis, these cells may become permissive for bacterial repli- pathway in cord blood contributes further to the opsonic defect
cation. Therefore, ingestion by macrophages of opsonized in neonates against E. coli (55). Under such conditions, entry
GBS may not only enhance but also interfere with elimination and survival within macrophages could play an important role
of these bacteria at the site of tissue infection. in the development of bacteremia and the course of meningeal
GBS vigorously activates inflammatory cytokine responses infection by E. coli (85).
by innate immune cells (36). Impaired interleukin-12 (IL-12) Neonatal innate immune cells are characterized by de-
production by GBS-exposed mononuclear phagocytes has re- creased responses to pathogen-derived or physiologic stimuli
cently been proposed to be linked to IFN-␥ deficiency in new- like lipopolysaccharide (LPS) and IFN-␥, respectively (44,
borns. GBS-stimulated mRNA accumulation and protein se- 106). LPS, the primary constituent of E. coli and other gram-
cretion of both IFN-␥ and IL-12 in mononuclear cells from negative bacteria, induces inflammation by binding to the
cord and adult blood were studied (36). By using reverse trans- TLR4/MD2/CD14 complex on macrophages (35). Recent re-
criptase PCR and quantitative densitometry assays, the kinet- search has shown normal expression of both TLR-4 and CD14
ics of GBS-stimulated accumulation of IFN-␥ mRNA and molecules on cord blood mononuclear cells but decreased
IL-12 mRNA were compared in cord and adult cells. After 12 TLR-4-mediated signaling and ligand-induced tumor necrosis
to 18 h of incubation, significantly decreased mRNA accumu- factor ␣ (TNF-␣) release by these cells exposed to LPS (44,
lation for both IFN-␥ and IL-12 was detected in cord cells 106). These data suggest that the risk of overwhelming infec-
compared to adults. The concentrations of IFN-␥ and IL-12 in tion by E. coli in human neonates may be related to impaired
suspensions of GBS-exposed cord mononuclear cells were also TLR-4-mediated responses by macrophages, in addition to
significantly lower than in adults at 12 and 18 h. These data decreased opsonophagocytosis. Other authors found that neo-
suggest that, in addition to lymphocyte immaturity, IFN-␥ de- natal mononuclear cells produce an enhanced amount of
ficiency in neonates may be linked to decreased production of TNF-␣ in response to LPS or GBS (102). These findings are in
IL-12 by cord mononuclear phagocytes, at least when these concert with the enhanced sensitivity of neonates to TNF-
cells are stimulated with GBS. This observation also suggests induced shock.
that strategies to enhance neonatal host defense against GBS Listeria. Listeria infection in humans occurs most commonly
may include administration of IL-12. in newborns and in immunosuppressed children and adults
VOL. 74, 2006 MINIREVIEW 2001

(45). Perinatal infections caused principally by L. monocyto- pared with older children and adults, which may be responsible
genes are usually secondary to maternal infection or coloniza- for rapid progression of the disease (14, 86). Recent studies
tion. Macrophage activation is critically important for an effi- have shown that both HSV-1 and HSV-2 induced secretion of
cient killing of Listeria, and macrophage activation in vivo by IL-6 and IL-8 from adult peripheral blood mononuclear cells
IFN-␥ is a sine qua non for protection (9, 46). The effectiveness (PBMCs) in a dose-dependent manner (41). In addition,
of innate immunity in host defense against Listeria has been HSV-1 and HSV-2 activated NF-␬B in TLR-2-transfected
exemplified by studies using SCID mice that lack both T-cell HEK 293 cells but not control HEK cells or TLR-4-transfected
and B-cell immunity. These mice were remarkably resistant to HEK cells (40). Analysis of IL-6 and IL-8 responses revealed
infection with L. monocytogenes due to a rapid neutrophil that cord blood cells produced significantly higher amounts of
response followed by activation of macrophages and were able these cytokines in response to stimulation with HSV-1 than did
to control infection for several days (18). However, listeriosis adult blood cells (40). These findings are in concert with pre-
in mice with an SCID mutation results in a chronic infection viously published data indicating that term and preterm infants
characterized by abundant granulomas, microabscesses, and produce enhanced IL-6 and IL-8 and that clinical manifesta-
neutrophil infiltrates occurring mostly in the liver (11). There- tion of HSV infection is associated with increased production
fore, even though the innate immunity is effective to provide of inflammatory cytokines (79). Nevertheless, the link between
protection, an adequate immune response, i.e., clearance of in vitro and in vivo data is only indirect, and further research is
bacteria, granuloma formation with lymphocytes, and disap- needed to determine whether ongoing overproduction of in-
pearance of microabscesses, requires specific immunity. Adop- flammatory cytokines is a consistent component of HSV pa-
tive transfer studies showed a decisive role of CD4⫹ and CD8⫹ thology in newborns.
T cells in augmenting innate antibacterial host defenses and The effect of FL on neonatal innate immunity to HSV
ensuring long-term survival of Listeria-infected adult mice (11). infection has recently been studied in mice (95). After FL

Downloaded from iai.asm.org by on October 29, 2008


The capacity of CpG oligodeoxynucleotides (ODN) to stim- treatment, the nature and quality of resistance were ana-
ulate protective immune responses to Listeria was recently lyzed for short-term innate effect and for survival of neona-
studied in newborn mice (34). These studies showed that DCs, tal mice. Data showed that FL induced an IFN-␣/␤-associ-
macrophages, and B cells from 3-day-old mice responded to ated immune response in newborn animals by expanding
CpG stimulation by secreting IFN-␥, IL-12, and TNF-␣. In cells of the DC linage. A significant number of mice lacking
addition, spleen cells from CpG-treated newborn mice pro- mature T and B cells died after challenge with HSV-1,
duced large amounts of cytokines and nitric oxide when ex- whereas 30 to 40% of FL-treated mice survived HSV-1
posed to Listeria in vitro. In concert with these findings, new- infection for more than 21 days (95). This observation indi-
borns treated with CpG ODN were protected from lethal cated that innate immunity was decisive in defense against
Listeria challenge (34). These data suggest that cellular ele- HSV and that manipulation of the innate immune system by
ments of the neonatal immune system, similar to those of adult cytokine treatment may provide a tool to improve clinical
mice, may respond to stimulation by CpG ODN, thereby re- outcomes of neonatal HSV infection.
ducing host susceptibility to infectious pathogens. CMV. CMV is the most common cause of intrauterine in-
The hematopoietic growth factor Flt3 ligand (FL) was found to fection, affecting 0.3 to 2.2% of live-born infants (4). Congen-
induce a ⬎100-fold increase in the innate resistance to Listeria ital CMV infection is a leading cause of sensorineural hearing
infection in neonatal mice (95). In particular, FL induced in- loss, cognitive and visual impairment, and cerebral palsy. The
creases in DC numbers as well as IL-12 production by these cells virus can be transmitted to the fetus during primary maternal
(96). The increased IL-12 production may be crucial in defense infection in pregnancy, but it can also be transmitted even
against Listeria in vivo through stimulating IFN-␥ release by T when maternal infection occurred years prior to conception
cells and NK cells and most likely explains the increased survival (24, 81). Earlier studies suggested that the increased suscepti-
of FL-treated neonatal mice. Although these studies did not bility of the fetus to CMV infection could be related to defec-
clearly define differential responses to FL by adult versus neonatal tive cell-mediated immunity (26, 67). Recently, the presence of
mice, they indicate that newborn mice treated with this hemato- functionally mature cytolytic CD8⫹ T lymphocytes in newborns
poietic growth factor have a distinct advantage over untreated with congenital CMV infection was reported (47). This finding
littermates to control Listeria infections. suggests that intrauterine antigen stimulation has the potential
TLRs (TLR-2, TLR-4, and TLR-5) have been implicated in to elicit protective immunity in the fetus and that, in contrast
mice and humans as signaling receptors for L. monocytogenes to CD4⫹ T cells, the expression of efficient CD8⫹ effector
(23, 80, 94, 98). Studies in mice showed that TLR-2 plays a function in newborns may be preserved. Pertinent to this find-
critical role in controlling Listeria infection (94). In particular, ing, functionally mature CD8⫹ cytotoxic responses were doc-
TLR-2-deficient mice were more susceptible to systemic infec- umented in infants during primary infection with RSV (61).
tion by Listeria than were wild-type mice, with a reduced sur- The overall efficiency of CD8-dependent T-cell function in
vival rate and an increased bacterial burden in the liver. fetal or neonatal life, however, remains unclear. Evidence sug-
HSV. HSV is a formidable pathogen causing disseminated or gests that neonatal CD4⫹ T cells are deficient in activation-
central nervous system disease with a high mortality rate in the associated intracellular signaling and require high levels of
first weeks of life (101). Infection is acquired during the birth costimulation to achieve maximal activation (2, 27, 96). In this
process as the neonate comes in contact with the virus during regard it is noteworthy that CD4⫹ T cells play an essential
passage through an infected birth canal or through contact role in promoting the long-term activation and terminal
with individuals with active HSV lesions. Cellular immune differentiation of CD8⫹ T cells and in reactivation of CD8⫹
responses mediated by T cells are impaired in newborns com- memory cells.
2002 MINIREVIEW INFECT. IMMUN.

CMV, as a cofactor, may be involved in the pathogenesis of organ involvement and a high mortality rate are characteristic
HIV infection and AIDS (72). A cohort-based prospective features of perinatal varicella (13). Innate immunity in the
study was performed to examine the possible association of “antibody-free” window period is therefore critical to control
CMV infection with the progression of HIV disease in infants infection. PBMCs from adults were found to produce a large
who were born to HIV-1-infected women and whose CMV amount of IFN-␥ in response to VZV antigen, suggesting that
status was known (38). At birth, the frequency of CMV infec- a Th1 response with IFN-␥ production may be important in
tion in HIV-1-infected infants and in infants not infected with early host defense against VZV (7, 30). Remarkably, VZV did
HIV-1 was 4.3% and 4.5%, respectively, which was higher than not drive cord blood mononuclear cells (MC) to release sig-
the rates of 0.3 to 2.2% in the general population. However, at nificant IFN-␥ production (107). A real-time reverse-transcrip-
6 months of age, CMV infection was diagnosed in 39.9% of tion PCR analysis of IFN-␥ mRNA expression showed that
HIV-1-infected infants and in only 15.3% of noninfected in- VZV induced a significantly higher IFN-␥ mRNA in PBMCs
fants. The cumulative rates of CMV infection over a period of than in cord blood MC. IFN-␥ production is regulated by T-bet
48 months remained significantly higher among HIV-1-in- expression mediated by STAT-1 (signal transducer and activa-
fected children, and the rate of CMV transmission from moth- tor of transcription 1) (3, 73). Recent data suggested that VZV
ers to offspring was especially high during the first 12 months did not upregulate T-bet mRNA significantly in cord blood
(38). These data suggest that HIV-1-infected children have a MC in contrast to its effect in adult PBMCs. These data indi-
higher rate of CMV infection acquired postnatally and that cate a poor Th1 response and an impaired innate immune
CMV infection is associated with an increased risk of HIV-1 response to VZV in neonates.
disease progression. It is likely that CMV and HIV-1, two RSV. RSV infection is one of the most common human viral
immunosuppressive viruses, may act synergistically to acceler- diseases worldwide, and virtually every child is infected by the
ate disease progression. Whatever the mechanism, these ob- third birthday (66). The virus does not normally replicate out-

Downloaded from iai.asm.org by on October 29, 2008


servations suggest that strategies to prevent vertical and hori- side of the bronchopulmonary tree, and the infection is exquis-
zontal CMV infection in HIV-1-infected infants and children itely restricted to the respiratory mucosa. RSV proteins such as
should be applied in order to decrease and prolong disease the major surface glycoprotein (G) and the fusion (F) protein,
progression and central nervous system disease. which is a large envelope glycoprotein, are essential for viral
EBV. EBV infection occurring in early childhood is usually attachment and penetration, respectively, and are important in
not associated with any defined clinical disease (87). However, initiating immune responses (66, 99). Both G and F glycopro-
if primary infection is delayed until adolescence or adulthood, teins are able to induce neutralizing antibody responses and
a high proportion of affected individuals develop infectious long-term immunity. However, in young infants antibody-me-
mononucleosis (IM), characterized by increased numbers of diated immunity might contribute to lung pathology as well.
EBV-infected B cells in the peripheral blood and massive Despite the presence through the first few months of life of
oligoclonal expansion of EBV-specific CD8⫹ T cells (88). IM maternal antiviral antibodies passively transferred to the fetus,
can be expected to occur when primary EBV infection is not prevalence of more severe forms of RSV disease is greatest in
adequately controlled, leading to a subsequent overstimulation young infants. In a cohort of infants, not only did maternal
of CD8⫹ T cells by EBV-infected B cells. This concept is in neutralizing antibodies fail to prevent infection with RSV, but
agreement with fulminant IM occurring in patients with X- also the severity of pneumonia was inversely related to the
linked lymphoproliferative disease, an inherited immune defi- level of neutralizing antibodies, an intriguing observation as far
ciency characterized by defective immune responses to EBV as passive neonatal immunity is concerned (39, 42). In contrast,
infection. However, this would also imply that EBV infection administration of RSV-specific immunoglobulin or monoclo-
may be controlled better in newborns and infants than in ad- nal antibody preparations to high-risk infants may prevent
olescents or adults. Recently, there have been a number of bronchiolitis and hospitalization (33, 71). These data clearly
studies of CD4⫹ T cells, which are able to inhibit EBV-trans- indicate that further research is needed to define the role of
formed lymphoblastoid B cell line growth (89, 103). These specific antibodies in antiviral immunity in RSV disease in
transformed B cells can activate CD4⫹ T cells and NK cells early life.
from both adult and fetal blood. Differences in the activities of Several reports suggested detectable innate cytokine re-
CD4⫹ T cells and NK cells may not explain the immunological sponses to RSV at birth. In vitro, both cord and adult mono-
and clinical phenotypes of EBV infection in different age cyte-derived macrophages exhibited production of high levels
groups. However, CD8⫹ T-cell responses to EBV-infected B of IL-6 and TNF-␣ within 24 h after viral exposure (60). In
cells may be weaker in newborns and infants, explaining the contrast to adult cells, little or no production by cord macro-
lack of clinical manifestation of infection in early life. phages of these cytokines was observed 24 h after exposure to
VZV. VZV may cause significant morbidity and mortality in live RSV. These data indicated that neonatal macrophages
fetuses and newborns, and vaccinating VZV-susceptible may be less efficient in a sustained induction of inflammatory
women prior to pregnancy can prevent both vertical and hor- cytokine production. Others found that cord mononuclear cells
izontal transmission of varicella, suggesting a role for antibody- showed no proliferation response to exposure to inactivated
mediated immunity (68). Fetal varicella syndrome arises in RSV and, when exposed to live virus, produced fewer innate
about 2% of cases of maternal varicella, occurring during the and no adaptive cytokines (39). The major difference in cyto-
first 20 weeks of gestation (68, 70). VZV infection in newborns kine responses of cord and adult mononuclear cells to RSV
may result from either vertical or horizontal transmission. Peri- exposure appears to be that cord cells produce almost entirely
natally acquired varicella occurs mostly after the onset of ma- innate cytokines, whereas both innate and adaptive cytokines
ternal viremia but before maternal antibody develops. Visceral are produced by adult cells. Consequently, adaptive cytokine
VOL. 74, 2006 MINIREVIEW 2003

responses may be required for an efficient innate immune motility that is used by T. gondii organisms to actively invade
response to RSV. Humans are born with the capacity to mount their vertebral host cells (100). Calcium-mediated protein se-
innate cytokine responses to RSV, but due to the lack of in cretion and MIC2, a thrombospondin-related protein that
utero sensitization, infants may remain highly susceptible to serves as adhesion for T. gondii, have been implicated in the
the virus until adaptive cellular immunity develops. process of gliding motility. By using this invasion strategy,
In mice sensitized with recombinant vaccinia virus vector, the G Toxoplasma escapes phagocytic uptake, and the host cell plays
and F glycoproteins differentially regulated cytokine responses little role in controlling the entry of the parasite. During pen-
(5). Whereas G protein induced a Th2-type response character- etration of host cells, T. gondii restricts access of host cell
ized by secretion of IL-4 and IL-5, F protein induced the secretion proteins to the vacuole, thus creating a fusion-incompetent
of IL-2 and IFN-␥. In addition to inflammatory and immunoregu- vacuole that lies segregated from the endocytic network (100).
latory cytokines, chemokines are also induced in the respiratory This unique intracellular lifestyle provides protection from
tract after natural RSV infection. Studies of children with RSV host surveillance.
bronchiolitis have shown an increased production of chemokines Defense against T. gondii infection is mediated primarily by
including CXCL8, CXCL5, CXCL3, and CXCL2 in the upper cellular responses involving killing by macrophages and cyto-
respiratory tract (32, 65). Intriguing recent findings on chemokine toxic T cells and release of inflammatory cytokines that help
production in the lower respiratory tract in infants with RSV infected cells to kill the parasite or to maintain it in a quiescent
bronchiolitis were reported (63). CXC chemokines (CXCL10 and stage. Cellular immunity is mainly targeted to infected cells
CXCL8) were found to be the most abundant, but CC chemo- that express peptides from the parasite (21). Killing of Toxo-
kines (CCL2 and CCL3) were also present. Remarkably, plasma by and the survival and replication of this parasite in
CXCL10, one of the few chemokines capable of binding receptors resident mononuclear phagocytes from newborns and adults,
from different classes (both CXCR3 and CCR3), was present in respectively, are comparable (104, 105). In the immune mech-

Downloaded from iai.asm.org by on October 29, 2008


very large quantity in the RSV-infected lung. Whether chemokine anism through which acute T. gondii infection is controlled,
responses are protective or contribute to the pathogenesis of RSV IFN-␥ plays a central role as a strong activator of resident
disease needs to be determined. Further clinical studies are re- macrophages to limit intracellular growth of tachyzoites. In
quired to discover whether chemokine responses induced by RSV
vitro studies showed that GTPases are required for IFN-␥-
occur in other viral or respiratory tract infections in children.
induced suppression of T. gondii growth in macrophages (15).
Precise elucidation of the role of chemokines in the pathogenesis
In particular, a 47-kDa protein that possesses inherent GTPase
of RSV bronchiolitis has potentially therapeutic implications be-
activity and binds to the endoplasmic reticulum and Golgi was
cause a number of chemokine receptor antagonists are in devel-
found to regulate host resistance to T. gondii through its ability
opment.
to inhibit parasite growth within the macrophage. In human
The clinical spectrum of RSV disease is extremely variable,
newborns, both generation of IFN-␥ and response to IFN-␥ by
ranging from mild upper-respiratory tract disease to severe
mononuclear phagocytes are impaired (49, 104). This age-
respiratory distress (66). It is likely that genetic heterogeneity
related deficiency is likely to be one critical factor responsible
contributes to disease severity in addition to known risk factors
for the increased susceptibility of newborns to T. gondii infec-
including prematurity, congenital heart anomaly, and chronic
lung disease. Efficient host immune responses to viral patho- tion.
gens are mediated by Th1 cytokines. As the production of Th1 Candida. Infections by Candida are the most common of the
cytokines can be inhibited by cytokines secreted by Th2 lym- fungal infections in newborns (8, 83). Body surfaces are colo-
phocytes, an adequate balance of Th1 and Th2 cytokines is nized at birth by Candida species residing in the birth canal.
essential for the efficient eradication of RSV. Several studies Overgrowth of colonizing Candida may lead to mucosal or
have shown a correlation between predominant Th2 responses mucocutaneous candidiasis. The role of passively acquired hu-
in infants with RSV disease severity (1, 12, 74). TLR-4 and moral antibody in defense against invasive candidal disease
CD14 have been shown to sense RSV, and TLR-4-deficient appears to be negligible in newborns (48). The unique suscep-
mice developed delayed clearance of RSV as well as a pre- tibility to oropharyngeal candidiasis during the first weeks re-
dominant Th2 response which correlated with disease progres- sults most likely from the down-regulation of Th1 responses
sion (29, 92). An association between TLR-4 mutations (43, 48, 49).
(Asp299Gly and Thr394Ile) and severe RSV disease has re- Invasive candidal disease in neonates is a life-threatening
cently been reported, whereas no association between CD14 condition which may be explained by developmental deficien-
polymorphisms and RSV bronchiolitis was found (92). cies in the newborn’s innate immune system (52). Candida
Toxoplasma. T. gondii, an obligate intracellular pathogen, albicans is part of the common commensal of the gastrointes-
causes subclinical chronic infections in humans, where it is also tinal tract and invasive candidal disease can arise from entero-
an important opportunistic pathogen (62). The human fetus circular translocation of the gut flora (58, 77). B cell knockout
and newborn are especially susceptible to infection by T. gon- mice, which lack functional antibodies, are as resistant to mu-
dii. The placenta may act as a barrier to transplacental trans- cosal or invasive candidiasis of endogen origin as are immuno-
mission of parasites from mother to fetus, which occurs mostly competent controls (97). In addition, patients with X-linked
in the third trimester. In congenitally infected newborns, in- agammaglobulinemia or severe hypogammaglobulinemia do
fections with T. gondii may result in fibrous or calcified cerebral not exhibit an increased susceptibility to either mucocutaneous
lesions or ocular lesions that threaten vision (62). However, or invasive candidal infections (52). In such patients T cells and
disseminated infection is rare in congenitally infected infants innate immune cells ensure defense against Candida, and macro-
(20). This may be explained by the unique process of gliding phages can prevent candidal invasion by phagocytosis and kill-
2004 MINIREVIEW INFECT. IMMUN.

ing of nonopsonized yeasts through innate immune receptors several innate immune mechanisms are impaired in neonates.
(19, 25, 54, 90). It is also clear, however, that neonates are immunocompetent
Recognition and uptake of Candida yeasts involve the macro- to mount mature innate as well as adaptive immune responses
phage MR, which is a type I membrane protein with three types like nonopsonic uptake of fungi or, under certain circum-
of domains in the extracellular region (19, 54, 59, 93). Well- stances, adult-level T-cell responses. Thus, the challenge of
characterized lectin activities of the MR are mediated by the future research will include the discovery of mechanisms that
cysteine-rich domain which can recognize sulfated sugars, underlie differential immune responses in newborns so that
whereas mannose recognition takes place through the C-type prevention and treatment of neonatal infections can more
lectin-like domains. The extent of phagocytosis and killing of safely be targeted.
nonopsonized Candida organisms by resident monocyte-derived
macrophages were comparable in newborns and adults, and both ACKNOWLEDGMENTS
mannan and mannose-bovin serum albumin complex inhibited This work was supported by grants from the Hungarian Research
ingestion in a concentration-dependent manner, suggesting a role Fund (OTKA T038095 and OTKA T049017).
for the MR (50, 52). Exposure of adult macrophages to IFN-␥ (up
REFERENCES
to a concentration 100 U/ml) resulted in increased phagocytosis
1. Aberle, J. H., S. W. Aberle, M. N. Dworzak, C. W. Mandl, W. Rebhandl, G.
and killing. In contrast, no enhancement with cord macrophages Vollnhofer, M. Kundi, and T. Popow-Kraupp. 1999. Reduced interferon-
could be detected under the same experimental condition, and at gamma expression in peripheral blood mononuclear cells of infants with
a concentration of 500 U/ml IFN-␥ there was still significantly severe respiratory syncytial virus disease. Am. J. Respir. Crit. Care Med.
160:1263–1268.
lower killing and superoxide release by cord macrophages com- 2. Adkins, B., C. Leclerc, and S. Marshall-Clarke. 2004. Neonatal adaptive
pared to adult cells (57). These data suggested that neonatal immunity comes of age. Nat. Rev. Immunol. 4:553–564.
macrophages have a normal capacity to ingest and kill Candida 3. Afkarian, M., J. R. Sedy, J. Yang, N. G. Jacobson, N. Cereb, S. Y. Yang,

Downloaded from iai.asm.org by on October 29, 2008


T. L. Murphy, and K. M. Murphy. 2002. T-bet is a STAT1-induced regu-
through the MR but cannot be fully activated by IFN-␥, a finding lator of IL-12R expression in naı̈ve CD4⫹ T cells Nat. Immunol. 3:549–557.
that could not be attributed to lower expression or binding to its 4. Alford, C. A., S. Stagno, R. F. Pass, and W. J. Britt. 1990. Congenital and
perinatal cytomegalovirus infections. Rev. Infect. Dis. 7:S745–S753.
ligand of IFN-␥ receptor on neonatal cells. Remarkably, in re- 5. Alwan, W. H., W. J. Kozlowska, and P. J. Openshaw. 1994. Distinct types of
sponse to IFN-␥, a significantly decreased STAT-1 phosphoryla- lung disease caused by functional subsets of antiviral T cells. J. Exp. Med.
tion was detected in neonatal cells, suggesting the possibility of 179:81–89.
6. Areschoug, T., M. Stalhammar-Carlemalm, I. Karlsson, and G. Lindahl.
negative regulation of IFN-␥ receptor signaling in newborns (51). 2002. Streptococcal beta protein has separate binding sites for human factor
The precise mechanism by which signaling through innate im- H and IgA-Fc. J. Biol. Chem. 277:12642–12648.
mune receptors may be down-modulated in neonates remains 7. Arvin, A. M., C. M. Koropchak, B. R. Williams, F. C. Grumet, and S. K.
Foung. 1986. Early immune response in healthy and immunocompromised
unclear. subjects with primary varicella-zoster virus infection. J. Infect. Dis. 154:422–
Dectin-1, the receptor for binding fungal-derived ␤-glucan 429.
8. Baley, J. E., R. M. Kliegman, and A. A. Fanaroff. 1984. Disseminated fungal
by macrophages and neutrophils, is a small type II transmem- infections in very low-birth-weight infants: clinical manifestations and epi-
brane protein containing one lectin-like carbohydrate recogni- demiology. Pediatrics 73:144–152.
tion domain (25). This receptor can recognize live Saccharo- 9. Bancroft, G. J., R. D. Schreiber, G. C. Bosma, M. J. Bosma, and E. R.
Unanue. 1987. A T cell-independent mechanism of macrophage activation
myces cerevisiae and, to a lesser extent, C. albicans. It was by interferon gamma. J. Immunol. 139:1104–1107.
previously reported that ligand binding to the MR is not cou- 10. Berg, S., S. Kasvi, B. Trollfors, J. Pilichowska-Paszkiet, A. Fattom, I.
pled to the activation of the respiratory burst oxydase and Tessin, and T. Lagergard. 1998. Antibodies to group B streptococci in
neonates and infants. Eur. J. Pediatr. 157:221–224.
superoxide release in macrophages, in contrast to binding an- 11. Bhardwaj, V., O. Kanagawa, P. E. Swanson, and E. R. Unanue. 1998.
tibody-opsonized Candida to Fc receptors on these cells (54). Chronic Listeria infection in SCID mice: requirements for the carrier state
and the dual role of T cells in transferring protection or suppression.
The effect of ligand binding to dectin-1 on the respiratory burst J. Immunol. 160:376–384.
activity has not been studied. However, a TNF-␣ response was 12. Bont, L., and J. L. Kimpen. 2002. Immunological mechanisms of severe
generated by macrophages upon exposure to ␤-glucan. Re- respiratory syncytial virus bronchiolitis. Intensive Care Med. 28:616–621.
13. Brunell, P. A. 1983. Fetal and neonatal varicella-zoster infections. Semin.
markably, the production of TNF-␣ was significantly greater Perinatol. 7:47–56.
when macrophages were exposed to S. cerevisiae than to C. 14. Burchett, S. K., L. Corey, K. M. Mohan, J. Westall, R. Ashley, and C. B.
albicans. This observation is in concert with published data that Wilson. 1992. Diminished interferon-gamma and lymphocyte proliferation
in neonatal and postpartum primary herpes simplex virus infection. J. In-
␤-glucan is buried within the cell wall of C. albicans and that fect. Dis. 165:813–818.
Candida uptake by macrophages and keratinocytes can be in- 15. Butcher, B. A., R. I. Greene, S. C. Henry, K. L. Annecharico, J. B. Weinberg,
E. Y. Denkers, A. Sher, and G. A. Taylor. 2005. p47 GTPases regulate
hibited by mannan and, to a lower extent, by glucan (37, 54, Toxoplasma gondii survival in activated macrophages. Infect. Immun. 73:
90). It is also possible that S. cerevisiae may have a higher 3278–3286.
density of ␤-glucan exposed on its surface compared to that of 16. Cordero, L., M. Sananes, and L. W. Ayers. 1999. Bloodstream infections in
a neonatal intensive-care unit: 12 years’ experience with an antibiotic con-
C. albicans. trol program. Infect. Control Hosp. Epidemiol. 20:242–246.
17. Cornacchione, P., L. Scaringi, and K. Fettucciari. 1998. Group B strepto-
cocci persist inside macrophages. Immunology 93:86–95.
SUMMARY AND CONCLUSION 18. Deschryver-Kecskemeti, K., G. J. Bancroft, G. C. Bosma, M. J. Bosma, and
E. R. Unanue. 1988. Pathology of Listeria infection in murine severe com-
Human neonates are highly susceptible to infection by a bined immunodeficiency. A study by immunohistochemistry and electron
microscopy. Lab. Investig. 58:698–705.
wide range of bacteria, viruses, protozoa, and fungi. The 19. Ezekowitz, R. A., K. Sastry, P. Bailly, and A. Warner. 1990. Molecular
heightened susceptibility and the severe course of infections in characterization of the human macrophage mannose receptor: demonstra-
early life can be attributed, at least in part, to the lack of tion of multiple carbohydrate recognition-like domains and phagocytosis of
yeasts in Cos-1 cells. J. Exp. Med. 172:1785–1794.
preexisting immunological memory and competent adaptive 20. Fatoohi, A. F., G. J. Cozon, M. Wallon, S. Kahi, F. Gay-Andrieu, T.
immunity. In addition, a large body of evidence suggests that Greenland, and F. Peyron. 2003. Cellular immunity to Toxoplasma gondii in
VOL. 74, 2006 MINIREVIEW 2005

congenitally infected newborns and immunocompetent infected hosts. Eur. 45. Lorber, B. 1997. Listeriosis. Clin. Infect. Dis. 24:1–9.
J. Clin. Microbiol. Infect. Dis. 22:181–184. 46. Mackaness, G. B. 1962. Cellular resistance to infection. J. Exp. Med. 116:
21. Fatoohi, A. F., G. J. Cozon, P. Gonzalo, M. Mayencon, T. Greenland, S. 381–406.
Picot, and F. Peyron. 2004. Heterogeneity in cellular and humoral immune 47. Marchant, A., V. Appay, M. Van Der Sande, N. Dulphy, C. Liesnard, M. Kidd,
responses against Toxoplasma gondii antigen in humans. Clin. Exp. Immu- S. Kaye, O. Ojuola, G. M. Gillespie, A. L. Vargas Cuero, V. Cerundolo, M.
nol. 136:535–541. Callan, K. P. McAdam, S. L. Rowland-Jones, C. Donner, A. J. McMichael, and
22. Feldman, R. G., M. A. Breukels, S. David, and G. T. Rijkers. 1998. Prop- H. Whittle. 2003. Mature CD8(⫹) T lymphocyte response to viral infection
erties of human anti-group B streptococcal type III capsular IgG antibody. during fetal life. J. Clin. Investig. 111:1747–1755.
Clin. Immunol. Immunopathol. 86:161–169. 48. Maródi, L. 1997. Local and systemic host defense mechanisms against
23. Flo, T. H., O. Halaas, E. Lien, L. Ryan, G. Teti, D. T. Golenbock, A. Sundan, Candida: immunopathology of candidal infections. Pediatr. Infect. Dis. J.
and Espevik, T. 2000. Human Toll-like receptor 2 mediates monocyte 16:795–801.
activation by Listeria monocytogenes, but not by group B streptococci or 49. Maródi, L. 2002. Down-regulation of Th1 responses in human neonates.
lipopolysaccharide. J. Immunol. 164:2064–2069. Clin. Exp. Immunol. 128:1–2.
24. Fowler, K. B., S. Stagno, and R. F. Pass. 2003. Maternal immunity and 50. Maródi, L., D. E. Campbell, R. Káposzta, R. A. Polin, J. Csongor, and R. B.
prevention of congenital cytomegalovirus infection. JAMA 289:1008–1011. Johnston, Jr. 1994. Candidacidal mechanisms in the human neonate: im-
25. Gantner, B. N., R. M. Simmons, and D. M. Underhill. 2005. Dectin-1 paired IFN-gamma activation of macrophages in newborn infants. J. Im-
mediates macrophage recognition of Candida albicans yeast but not fila- munol. 153:5643–6549.
ments. EMBO J. 24:1277–1286. 51. Maródi, L., K. Goda, A. Palicz, and G. Szabó. 2001. Cytokine receptor
26. Gehrz, R. C., S. C. Marker, S. O. Knorr, J. M. Kalis, and H. H. Balfour. signaling in neonatal macrophages: defective STAT-1 phosphorylation in
1977. Specific cell-mediated immune defect in active cytomegalovirus in- response to stimulation with IFN-␥. Clin. Exp. Immunol. 126:456–460.
fection of young children and their mothers. Lancet 2:844–847. 52. Maródi, L., and R. B. Johnston, Jr. 2003. Mechanisms of resistance to
27. Han, P., T. McDonald, and G. Hodge. 2004. Potential immaturity of the fungal infections, p. 1487–1489. In R. A. Polin, W. W. Fox, and S. Abman
T-cell and antigen-presenting cell interaction in cord blood with particular (ed.), Fetal and neonatal physiology. Elsevier, Philadelphia, Pa.
emphasis on the CD40-CD40 ligand costimulatory pathway. Immunology 53. Maródi, L., R. Káposzta, and É. Nemes. 2000. Survival of group B Strep-
113:26–34. tococcus type III in mononuclear phagocytes: differential regulation of
28. Harris, T. O., D. W. Shelver, J. F. Bohnsack, and C. E. Rubens. 2003. A bacterial killing in cord macrophages by human recombinant gamma inter-
novel streptococcal surface protease promotes virulence, resistance to op- feron and granulocyte-macrophage colony-stimulating factor. Infect. Im-
sonophagocytosis, and cleavage of human fibrinogen. J. Clin. Investig. 111: mun. 68:2167–2170.
61–70.

Downloaded from iai.asm.org by on October 29, 2008


54. Maródi, L., H. M. Korchak, and R. B. Johnston, Jr. 1991. Mechanisms of
29. Haynes, L. M., D. D. Moore, E. A. Kurt-Jones, R. W. Finberg, L. J. Anderson, host defense against Candida species. I. Phagocytosis by monocytes and
and R. A. Tripp. 2001. Involvement of Toll-like receptor 4 in innate immunity monocyte-derived macrophages. J. Immunol. 146:2783–2789.
to respiratory syncytial virus. J. Virol. 75:10730–10737. 55. Maródi, L., P. C. Leijh, A. Braat, M. R. Daha, and R. van Furth. 1985.
30. Hayward, A. R., M. Cosyns, M. Jones, M. J. Levin, E. Villanueba, A. Opsonic activity of cord blood sera against various species of microorgan-
Weinberg, and C. Y. Chan. 1998. Cytokine production in varicella-zoster ism. Pediatr. Res. 19:433–436.
virus-stimulated cultures of human blood lymphocytes. J. Infect. Dis. 178:
56. Maródi, L., P. C. J. Leijh, and R. van Furth. 1984. Characteristics and
S95–98.
functional capacities of human cord blood granulocytes and monocytes.
31. Hill, H. R., N. H. Augustine, and H. S. Jaffe. 1991. Human recombinant
Pediat. Res. 18:1127–1131.
interferon gamma enhances neonatal polymorphonuclear leukocyte activa-
57. Maródi, L., S. Schreiber, D. Anderson, R. P. MacDermott, H. M. Korchak,
tion and movement, and increases free intracellular calcium. J. Exp. Med.
and R. B. Johnston, Jr. 1993. Enhancement of macrophage candidacidal
173:767–770.
activity by IFN-gamma: increased phagocytosis, killing and calcium signal
32. Hornsleth, A., L. Loland, and L. B. Larsen. 2001. Cytokines and chemo-
mediated by a decreased number of mannose receptors. J. Clin. Investig.
kines in respiratory secretion and severity of disease in infants with respi-
91:2596–2601.
ratory syncytial virus (RSV) infection. J. Clin. Virol. 21:163–170.
58. Maródi, L., C. Tournay, R. Káposzta, R. B. Johnston, Jr., and N. Mogui-
33. The Impact-RSV Study Group. 1998. Palivizumab, a humanized respiratory
levsky. 1998. Augmentation of human macrophage candidacidal capacity by
syncytial virus infection in high-risk infants. Pediatrics 102:531–537.
recombinant human myeloperoxidase and granulocyte-macrophage colony-
34. Ito, S., K. J. Ishii, M. Gursel, H. Shirotra, A. Ihata, and D. M. Klinman.
stimulating factor. Infect. Immun. 66:2750–2754.
2005. CpG oligodeoxynucleotides enhance neonatal resistance to Listeria
infection. J. Immunol. 174:777–782. 59. Martinez-Pomares, L., L. G. Hanitsch, R. Stillion, S. Keshav, and S. Gordon.
35. Janeway, C. A., Jr., and R. Medzhitov. 2002. Innate immune recognition. 2005. Expression of mannose receptor and ligands for its cysteine-rich domain
Annu. Rev. Immunol. 20:197–216. in venous sinuses of human spleen. Lab. Investig. 85:1238–1249.
36. Joyner, J. L., N. H. Augustine, K. A. Taylor, T. R. La Pine, and H. R. Hill. 60. Matsuda, K., H. Tsutsumi, S. Sone, Y. Yoto, K. Oya, Y. Okamoto, P. L.
2000. Effects of group B streptococci on cord and adult mononuclear cell Ogra, and S. Chiba. 1996. Characteristics of IL-6 and TNF-alpha produc-
interleukin-12 and interferon-␥ mRNA accumulation and protein secre- tion by respiratory syncytial virus-infected macrophages in the neonate.
tion. J. Infect. Dis. 182:974–977. J. Med. Virol. 48:199–203.
37. Klis, F. M., P. de Groot, and K. Hellingwerf. 2001. Molecular organization 61. Mbawuike, I. N., J. Wells, R. Byrd, S. G. Cron, W. P. Glezen, and P. A.
of the cell wall of Candida albicans. Med. Mycol. 39:S1–8. Piedra. 2001. HLA-restricted CD8⫹ cytotoxic T lymphocyte, interferon-
38. Kovacs, A., M. Schluchter, K. Easley, G. Demmler, W. Shearer, P. La gamma, and interleukin-4 responses to respiratory syncytial virus infection
Russa, J. Pitt, E. Cooper, J. Goldfarb, D. Hodes, M. Kattan, and K. in infants and children. J. Infect. Dis. 183:687–696.
McIntosh. 1999. Cytomegalovirus infection and HIV-1 disease progression 62. McLeod, R., J. Wisner, and K. Boyer. 1992. Toxoplasmosis, p. 518–550. In
in infants born to HIV-1-infected women. N. Engl. J. Med. 341:77–84. S. Krugman, S. L. Katz, A. A. Gershon, and C. M. Wilfert (ed.), Infectious
39. Krishnan, S., M. Craven, R. C. Welliver, N. Ahmad, and M. Halonen. 2003. diseases of children, 9th ed. Mosby Year Book, St. Louis, Mo.
Differences in participation of innate and adaptive immunity to respiratory 63. McNamara, P. S., B. F. Flanagan, C. A. Hart, and R. L. Smyth. 2005.
syncytial virus in adults and neonates. J. Infect. Dis. 188:433–439. Production of chemokines in the lungs of infants with severe respiratory
40. Kurt-Jones, E. A., J. Belko, C. Yu, P. E. Newburger, J. Wang, M. Chan, syncytial virus bronchiolitis. J. Infect. Dis. 191:1225–1232.
D. M. Knipe, and R. W. Finberg. 2005. The role of Toll-like receptors in 64. Mosdósi, B., T. Decsi, K. Nagy, Gy. Soltész, and L. Maródi. 2002. Early
herpes simplex infection in neonates. J. Infect. Dis. 191:746–748. recognition and successful treatment of an infant with severe combined
41. Kurt-Jones, E. A., M. Chan, S. Zhou, J. Wang, G. Reed, R. Bronson, M. M. immune deficiency. Orv. Hetil. 143:2081–2083.
Arnold, D. M. Knipe, and R. W. Finberg. 2004. Herpes simplex virus 1 65. Noah, T. L., S. S. Ivins, P. Murphy, I. Kazachkova, B. Moats-Staats, and
interaction with Toll-like receptor 2 contributes to lethal encephalitis. Proc. F. W. Henderson. 2002. Chemokines and inflammation in the nasal pas-
Natl. Acad. Sci. USA 101:1315–1320. sages of infants with respiratory syncytial virus bronchiolitis. Clin. Immunol.
42. Lamprecht, C. L., H. E. Krause, and M. A. Mufson. 1976. Role of maternal 104:86–95.
antibody in pneumonia and bronchiolitis due to respiratory syncytial virus. 66. Ogra, P. L. 2004. Respiratory syncytial virus: the virus, the disease and the
J. Infect. Dis. 134:211–217. immune response. Paediatr. Respir. Rev. 5:S119–126.
43. La Pine, T. R., J. L. Joyner, N. H. Augustine, S. D. Kwak, and H. R. Hill. 67. Pass, R. F., S. Stagno, W. J. Britt, and C. A. Alford. 1983. Specific cell-
2003. Defective production of IL-18 and IL-12 by cord blood mononuclear mediated immunity and the natural history of congenital infection with
cells influences the T helper-1 interferon gamma response to group B cytomegalovirus. J. Infect. Dis. 148:953–961.
streptococci. Pediatr. Res. 54:276–281. 68. Pastuszak, A. L., M. Levy, B. Schick, C. Zuber, M. Feldkamp, J. Gladstone,
44. Levy, O., K. A. Zarember, R. M. Roy, C. Cywes, P. J. Godowski, and M. R. F. Bar-Levy, E. Jackson, A. Donnenfeld, and W. Meschino. 1994. Outcome
Wessels. 2004. Selective impairment of TLR-mediated innate immunity in after maternal varicella infection in the first 20 weeks of pregnancy. N. Engl.
human newborns: neonatal blood plasma reduces monocyte TNF-alpha J. Med. 330:901–905.
induction by bacterial lipopeptides, lipopolysaccharide, and imiquimod, but 69. Prasadarao, N. V., A. M. Blom, B. O. Villoutreix, and L. C. Linsangan.
preserves the response to R-848. J. Immunol. 173:4627–4634. 2002. A novel interaction of outer membrane protein A with C4b binding
2006 MINIREVIEW INFECT. IMMUN.

protein mediates serum resistance of Escherichia coli K1. J. Immunol. 89. Sun, Q., R. L. Burton, K. E. Pollok, D. J. Emanuel, and K. G. Lucas. 1999.
169:6352–6360. CD4(⫹) Epstein-Barr virus-specific cytotoxic T-lymphocytes from human
70. Preblud, S. R., D. J. Bregman, and L. L. Vernon. 1985. Deaths from umbilical cord blood. Cell. Immunol. 195:81–88.
varicella in infants. Pediatr. Infect. Dis. J. 4:503–507. 90. Szolnoky, G., Z. Bata-Csörgó, A. S. Kenderessy, M. Kiss, A. Pivarcsi, Z.
71. The Prevent Study Group. 1997. Reduction of respiratory syncytial virus Novák, K. Nagy Newman, G. Michel, T. Ruzicka, L. Maródi, A. Dobozy, and
hospitalization among premature infants and infants with bronchopulmo- L. Kemény. 2001. A mannose-binding receptor is expressed on human
nary dysplasia using respiratory syncytial virus immune globulin prophy- keratinocytes and mediates killing of Candida albicans. J. Investig. Derma-
laxis. Pediatrics 99:93–99. tol. 117:205–213.
72. Rabkin, C. S., A. Hatzakis, P. D. Griffiths, D. Pillay, M. V. Ragni, M. W. 91. Takeda, K., and S. Akira. 2005. Toll-like receptors in innate immunity. Int.
Hilgartner, J. J. Goedert, et al. 1993. Cytomegalovirus infection and risk of Immunol. 17:1–14.
AIDS in human immunodeficiency virus-infected hemophilia patients. 92. Tal, G., A. Mandelberg, I. Dalal, K. Cesar, E. Somekh, A. Tal, A. Oron, S.
J. Infect. Dis. 168:1260–1263. Itskovich, A. Ballin, S. Houri, A. Beigelman, O. Lider, G. Rechavi, and N.
73. Robinson, D. S., and A. O’Garra. 2002. Further checkpoints in Th1 devel- Amariglio. 2004. Association between common Toll-like receptor 4 muta-
tions and severe respiratory syncytial virus disease. J. Infect. Dis. 189:2057–
opment. Immunity 16:755–758.
2063.
74. Roman, M., W. J. Calhoun, K. L. Hinton, L. F. Avendano, V. Simon, A. M.
93. Taylor, M. E., J. T. Conary, M. R. Lennartz, P. D. Stahl, and K. Drickamer.
Escobar, A. Gaggero, and P. V. Diaz. 1997. Respiratory syncytial virus
1990. Primary structure of the mannose receptor contains multiple motifs
infection in infants is associated with predominant Th-2-like response.
resembling carbohydrate-recognition domains. J. Biol. Chem. 265:12156–
Am. J. Respir. Crit. Care Med. 156:190–195.
12162.
75. Rooijakkers, S. H., M. Ruyken, A. Roos, M. R. Daha, J. S. Presanis, R. B. 94. Torres D., M. Barrier, F. Bihl, V. J. Quesniaux, I. Maillet, S. Akira, B.
Sim, W. J. van Wamel, K. P. van Kessel, and J. A. van Strijp. 2005. Immune Ryffel, and F. Erard. 2004. Toll-like receptor 2 is required for optimal
evasion by a staphylococcal complement inhibitor that acts on C3 conver- control of Listeria monocytogenes infection. Infect. Immun. 72:2131–2139.
tases. Nat. Immunol. 6:920–927. 95. Vollstedt, S., M. Franchini, H. P. Hefti, B. Odermatt, M. O’Keeffe, G. Alber,
76. Rubens, C. E., M. R. Wessels, L. M. Heggen, and D. L. Kasper. 1987. B. Glanzmann, M. Riesen, M. Ackermann, and M. Suter. 2003. Flt3 ligand-
Transposon mutagenesis of type III group B Streptococcus: correlation of treated neonatal mice have increased innate immunity against intracellular
capsule expression with virulence. Proc. Natl. Acad. Sci. USA 84:7208– pathogens and efficiently control virus infections. J. Exp. Med. 197:575–584.
7212. 96. Vollstedt, S., M. O’Keeffe, B. Odermatt, R. Beat, B. Glanzmann, M. Riesen,
77. Scherer, S., and D. A. Stevens. 1988. Candida albicans dispersed, repeated K. Shortman, and M. Suter. 2004. Treatment of neonatal mice with Flt3
gene family and its epidemiological applications. Proc. Natl. Acad. Sci. ligand leads to changes in dendritic cell subpopulations associated with

Downloaded from iai.asm.org by on October 29, 2008


USA 85:1452–1456. enhanced IL-12 and IFN-alpha production. Eur. J. Immunol. 34:1849–1860.
78. Schuchat, A. 1999. Group B Streptococcus. Lancet 353:51–56. 97. Wagner, R. D., A. Vazquez-Torres, J. Jones-Carson, T. Warner, and E.
79. Schultz, C., C. Rott, P. Temming, P. Schlenke, J. C. Moller, and P. Bucsky. Balish. 1996. B cell knockout mice are resistant to mucosal and systemic
2002. Enhanced interleukin-6 and interleukin-8 synthesis in term and pre- candidiasis of endogenous origin but susceptible to experimental systemic
term infants. Pediatr. Res. 51:317–322. candidiasis. J. Infect. Dis. 174:589–597.
80. Seki, E., H. Tsutsui, N. M. Tsuji, N. Hayashi, K. Adachi, H. Nakano, S. 98. Way S. S., L. J. Thompson, J. E. Lopes, A. M. Hajjar, T. R. Kollmann, N. E.
Futatsugi-Yumikura, O. Takeuchi, K. Hoshino, S. Akira, J. Fujimoto, and Freitag, and C. B. Wilson. 2004. Characterization of flagellin expression
K. Nakanishi. 2002. Critical roles of myeloid differentiation factor 88- and its role in Listeria monocytogenes infection and immunity. Cell Micro-
dependent proinflammatory cytokine release in early phase clearance of biol. 6:235–242.
Listeria monocytogenes in mice. J. Immunol. 169:3863–3868. 99. Welliver, R. C. 2003. Review of epidemiology and clinical risk factors for
81. Stagno, S., D. W. Reynolds, E. S. Huang, S. D. Thames, R. J. Smith, and severe Respiratory syncytial virus infection. J. Pediatr. 143:S112–117.
C. A. Alford. 1977. Congenital cytomegalovirus infection. N. Engl. J. Med. 100. Wetzel, D. M., L. A. Chen, F. A. Ruiz, S. N. Moreno, and L. D. Sibley. 2004.
296:1254–1258. Calcium-mediated protein secretion potentiates motility in Toxoplasma
82. Staros, E. B. 2005. Innate immunity: new approaches to understanding its gondii. J. Cell Sci. 117:5739–5748.
clinical significance. Am. J. Clin. Pathol. 123:305–312. 101. Whitley, R. 2004. Neonatal herpes simplex virus infection. Curr. Opin.
Infect. Dis. 17:243–246.
83. Stephenson, J. 2001. Can a common medical practice transform Candida
102. Williams, P. A., J. F. Bohnsack, N. H. Augustine, W. K. Drummond, C. E.
infections from benign to deadly? JAMA 286:2531–2532.
Rubens, and H. R. Hill. 1993. Production of tumor necrosis factor by
84. Stoll, B. J., N. I. Hansen, R. D. Higgins, A. A. Fanaroff, S. Duara, R.
human cells in vitro and in vivo, induced by group B streptococci. J. Pediatr.
Goldberg, A. Laptook, M. Walsh, W. Oh, and E. Hale. 2005. Very low birth
123:292–300.
weight preterm infants with early onset neonatal sepsis: the predominance
103. Wilson, A. D., and A. J. Morgan. 2002. Primary immune responses by cord
of Gram-negative infections continues in the National Institute of Child blood CD4⫹ T cells and NK cells inhibit Epstein-Barr virus B-cell trans-
Health and Human Development Neonatal Research Network, 2002–2003. formation in vitro. J. Virol. 76:5071–5081.
Pediatr. Infect. Dis. J. 24:635–639. 104. Wilson, C. B., and D. B. Lewis. 1990. Basis and implications of selectively
85. Sukumaran, S. K., H. Shimada, and N. V. Prasadarao. 2003. Entry and diminished cytokine production in neonatal susceptibility to infection. Rev.
intracellular replication of Escherichia coli K1 in macrophages require ex- Infect. Dis. 4:S410–420.
pression of outer membrane protein A. Infect. Immun. 71:5951–5961. 105. Wilson, C. B., and J. E. Haas. 1984. Cellular defenses against Toxoplasma
86. Sullender, W. M., J. L. Miller, L. L. Yasukawa, J. S. Bradley, S. B. Black, gondii in newborns. J. Clin. Investig. 73:1606–1616.
A. S. Yeager, and A. M. Arvin. 1987. Humoral and cell-mediated immunity 106. Yan, S. R., G. Qing, D. M. Byers, A. W. Sdadnyk, W. Al-Hertani, and R.
in neonates with herpes simplex virus infection. J. Infect. Dis. 155:28–37. Bortolussi. 2004. Role of MyD88 in diminished tumor necrosis factor alpha
87. Sumaya, C. V., and Y. Ench. 1985. Epstein-Barr virus infectious mono- production by newborn mononuclear cells in response to lipopolysaccha-
nucleosis in children. I. Clinical and general laboratory findings. Pediatrics ride. Infect. Immun. 72:1223–1229.
75:1003–1010. 107. Yu, H. R., J. C. Chang, R. F. Chen, H. Chuang, K. C. Hong, L. Wang, and
88. Sumaya, C. V., and Y. Ench. 1985. Epstein-Barr virus infectious mono- K. D. Yang. 2003. Different antigens trigger different Th1/Th2 reactions in
nucleosis in children. II. Heterophil antibody and viral-specific responses. neonatal mononuclear cells (MNCs) relating to T-bet/GATA-3 expression.
Pediatrics 75:1011–1019. J. Leukoc. Biol. 74:952–958.

Editor: J. B. Kaper

You might also like