Developmental Immunology and Role of Host Defenses in Fetal and Neonatal Susceptibility to Infection


The human fetus and neonate are unduly susceptible to infection with a wide variety of microbes, many of which are not pathogenic in more mature individuals. This susceptibility results from limitations of both innate and adaptive (antigen-specific) immunity and their interactions. This chapter focuses on the ontogeny of the immune system in the fetus, neonate, and young infant and the relationship between limitations in immune function and susceptibility to specific types of infection.

The immune system includes innate protective mechanisms against pathogens provided by the skin, respiratory and gastrointestinal epithelia, and other mucosa; humoral factors, such as cytokines ( Tables 4-1 and 4-2 ) and complement components ( Fig. 4-1 ); and innate and adaptive immune mechanisms mediated by hematopoietic cells, including mononuclear phagocytes (circulating monocytes and tissue macrophages), granulocytes, dendritic cells (DCs), and lymphocytes. Certain nonhematopoietic cells, such as follicular dendritic cells and thymic epithelial cells, also play important roles in adaptive immunity.

Table 4-1
Major Human Cytokines and Tumor Necrosis Factor Family Ligands: Structure, Cognate Receptors, and Receptor-Mediated Signal Transduction Pathways
Cytokine Family Members Structure Cognate Receptor Family Proximal Signal Transduction Pathways
IL-1 β-Trefoil, monomers; processed and secreted IL-1 receptor (IL-1R) superfamily (three immunoglobulin-like ectodomains) MyD88, IRAKs, NF-κB, MAPK, JNK
IL-1 subfamily IL-1α, IL-1β, IL-33, IL-1 receptor antagonist (RA)
IL-18 subfamily IL-18, IL-37
IL-36 subfamily IL-36α, IL-36β, IL-36γ, IL-36RA, IL-38
Hematopoietin IL-2–IL-7, IL-9–IL-13, IL-15, IL-17, IL-19 to IL-32, IL-34, IL-35, CSFs, oncostatin-M, IFNs (α, β, γ, and others); class II subfamily consists of IL-10, IL-19, IL-20, IL-22, IL-24, IL-26, IFNs (including IFN-αs, IFN-β, IL-27/28) Four α-helical; monomers except for IL-5 and IFNs (homodimers) and IL-12, IL-23, IL-27, IL-30, IL-35 (heterodimers); secreted Hematopoietin receptors JAK tyrosine kinases/STAT, SRC kinases, PI3K, PKB
TNF ligand TNF-α, lymphotoxin-α, lymphotoxin-β, CD27L, CD30L, CD40L, OX40L, TRAIL, others β-Jellyroll, homotrimers; type II membrane proteins and secreted TNF receptor family TRAFs and proteins mediating apoptosis
TGF-β TGF-β1, TGF-β2, TGF-β3, bone morphogenetic proteins Cysteine knot; processed and secreted TGF-β receptors type 1 and type 2 heterodimers (intrinsic serine threonine kinases) SMAD proteins
Chemokines Three-stranded β-sheet; all but fractalkine are secreted Seven membrane-spanning domains G protein–mediated
CXC ligand subfamily CXCL1-14, CXCL16 CXCR1 to CXCR6
CC ligand subfamily CCL1-5, CCL7, CCL8, CCL11, CCL13 to CCL28 CCR1 to CCR10
C ligand subfamily XCL1 (lymphotactin), XCL2 (SCM-1β) XCR1
CX3C ligand subfamily CX3CL (fractalkine) CX3CR1
CSF, Colony-stimulating factor; IFN, interferon; IL, interleukin; IRAK, IL-1 receptor–associated serine/threonine kinase; JAK, Janus tyrosine kinase; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor-kappa B; PI3K, phosphoinositide-3-kinase; PKB, protein kinase B; RA, receptor antagonist; SMAD, SMA- and MAD-related; SRC, sarcoma (family); STAT, signal transducer and activator of transcription; TGF, transforming growth factor; TNF, tumor necrosis factor; TRAFs, TNF-α receptor–associated factors; TRAIL, TNF-related apoptosis–inducing ligand.

Table 4-2
Immunoregulatory Effects of Select Cytokines, Chemokines, and Tumor Necrosis Factor (TNF) Ligand Family Proteins
Cytokine Principal Cell Source Major Biologic Effects
IFN-γ T cells and NK cells Pleiotropic, including enhancement of MΦ activation and antigen processing and MHC expression; promotes Th1 differentiation of activated CD4 T cells; regulates B-cell isotype switching
IL-1α, IL-1β Many cell types; MΦ are a major source Fever, inflammatory response, cofactor in T- and B-cell growth
IL-2 T cells T-cell > B-cell growth, increased cytotoxicity by T and NK cells, increased cytokine production and sensitivity to apoptosis by T cells, growth and survival of regulatory T cells
IL-3 T cells Growth of early hematopoietic precursors (also known as multi-CSF)
IL-4 T cells, mast cells, basophils, eosinophils Required for IgE synthesis; enhances B-cell growth and MHC class II expression; promotes T-cell growth and Th2 differentiation, mast-cell growth factor; enhances endothelial VCAM-1 expression
IL-5 T cells, NK cells, mast cells, basophils, eosinophils Eosinophil growth, differentiation, and survival
IL-6 MΦ, fibroblasts, T cells Hepatic acute-phase protein synthesis, fever, T-cell and B-cell growth and differentiation
IL-7 Stromal cells of bone marrow and thymus Essential thymocyte growth factor, promotes survival and homeostatic proliferation of peripheral T cells
IL-8 (CXCL8) MΦ, endothelial cells, fibroblasts, epithelial cells, T cells Chemotaxis and activation of neutrophils
IL-10 MΦ, T, cells, B cells, NK cells, keratinocytes, eosinophils Inhibits cytokine production by T cells and mononuclear cell inflammatory function and MHC expression; promotes B-cell growth and isotype switching; inhibits NK-cell cytotoxicity
IL-12 DCs, MΦ Induces Th1 and TFH differentiation of CD4 T cells
IL-13 T cells (Th2) and mast cells Promotes B-cell isotype switching to IgE, mucous production by epithelial cells
IL-15 DCs, MΦ Pleiotropic, including enhancement of NK-cell differentiation and survival and function of CD8 T cells
IL-21 T cells Pleiotropic, including promotion of B-cell growth, plasma-cell differentiation, NK-cell cytotoxicity
IL-23 DCs, MΦ Promotes Th17 differentiation of activated CD4 T cells
TNF-α MΦ, T cells, NK cells Fever and inflammatory response effects similar to IL-1, shock, hemorrhagic necrosis of tumors, and increased VCAM-1 expression on endothelium; induces catabolic state
CD40 ligand (CD154) T cells, lower amounts by B cells and DCs B-cell growth factor; promotes isotype switching, promotes IL-12 production by DCs; activates MΦ
Fas ligand Activated T cells, NK cells retina, testicular epithelium Induces apoptosis of cells expressing Fas, including effector B and T cells
Flt-3 ligand Bone marrow stromal cells Potent DC growth factor; promotes growth of myeloid and lymphoid progenitor cells in conjunction with other cytokines
G-CSF MΦ, fibroblasts, epithelial cells Growth of granulocyte precursors
GM-CSF MΦ, endothelial cells, T cells Growth of granulocyte-MΦ precursors and DCs; enhances granulocyte-MΦ function and B-cell antibody production
CCL3 (MIP-1α) MΦ, T cells MΦ chemoattractant; T-cell activator
CCL5 (RANTES) MΦ, T cells, fibroblasts, epithelial cells MΦ and memory T-cell chemoattractant; enhances T-cell activation; blocks HIV coreceptor
TGF-β MΦ, T cells, fibroblasts, epithelial cells, others Inhibits MΦ activation; inhibits Th1 T-cell responses
CSF, Colony-stimulating factor; DC, dendritic cell; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-macrophage colony-stimulating factor; HIV, human immunodeficiency virus; IL, interleukin; MΦ, mononuclear phagocytes; MHC, major histocompatibility complex; MIP, macrophage inhibitory protein; NK, natural killer; RANTES, regulated on activation, normal T-cell expressed and secreted; TFH, T-follicular helper; TGF, transforming growth factor; Th1, T helper 1; Th17, T helper 17; VCAM-1, vascular cell adhesion molecule-1.

Figure 4-1
Complement activation. Classical and mannose-binding lectin (MBL) pathways of activation intersect with the alternative pathway at C3. The MBL pathway of activation is identical to the classical pathway, starting with the cleavage of C4. When C3 is activated, this is followed by activation of the terminal components, which generate the membrane attack complex (C5b6789). Enzymatically active proteases, which serve to cleave and activate subsequent components, are shown with an overbar.

Innate immunity, unlike adaptive immunity to be discussed later, does not require prior exposure to be immediately effective and is equally efficient on primary and subsequent encounter with a microbe but does not provide long-lasting protection against reinfection. Innate defenses consist of fixed epithelial barriers and resident tissue macrophages, which act immediately or within the first minutes to hours of encounter with a microbe. These “frontline” defenses are sufficient for protection from the vast majority of microbes in the environment, which do not produce disease in healthy individuals. If the microbial insult is too great or the organism is able to evade these initial defenses, these cells release mediators that incite an inflammatory response, through which soluble and cellular defenses are recruited and help to limit or eradicate the infection over the next hours to days and to initiate the antigen-specific immune response that follows.

Epithelial Barriers

Epithelia form a crucial physical and chemical barrier against infection. Tight junctions between epithelial cells prevent direct entry of microbes into deeper tissues, and physical injury that disrupts epithelial integrity can greatly increase the risk for infection. In addition to providing a physical barrier, mechanical and chemical factors and colonization by commensal microbes contribute to the protective functions of the skin and of the mucosal epithelia of the gastrointestinal and respiratory tracts.

Antimicrobial Peptides and Proteins

A general feature of epithelial defenses is the production of one or more antimicrobial peptides, which include the α-defensins and β-defensins and the cathelicidin LL-37. Defensins and cathelicidin have direct antimicrobial activity against gram-positive and gram-negative bacteria and some fungi; viruses, such as, influenza and respiratory syncytial virus (RSV); and protozoa. Some of these antimicrobial peptides also exhibit proinflammatory and immunomodulatory activities.

There are six known human α-defensins: human neutrophil proteins (HNP) 1 to 4 and human defensins (HD) 5 and 6. HNPs 1 to 4 are expressed in leukocytes (white blood cells). HD-5 and HD-6 are produced and secreted by Paneth cells, located at the base of crypts in the small intestine. There are at least six human β-defensins, but only four (hBD-1 to -4) have been well characterized. hBD-1 is constitutively expressed by skin keratinocytes, whereas exposure to bacteria or proinflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) (see Tables 4-1 and 4-2 ), induces expression of hBD-2 and hBD-3 in keratinocytes and hBD-4 in lung epithelial cells, respectively. hBD-2 appears to be a normal component of human amniotic fluid, with levels increasing in the setting of chorioamnionitis and preterm labor. hBD-1 and hBD-2 are active against gram-negative bacteria and streptococci but are less active against Staphylococcus aureus, whereas hBD-3 is broadly active against gram-positive and gram-negative bacteria and Candida species. The cathelicidin LL-37 is expressed in leukocytes, for instance, neutrophils, mononuclear phagocytes, and mast cells, and its expression is induced by microbes and proinflammatory cytokines in epithelial cells of the skin, gut, and respiratory tract; LL-37 is active against gram-positive and gram-negative bacteria but less active against S . aureus than hBD-3 ; it also has activity in vitro against RSV and in vivo against influenza A virus.

In addition to the defensins, other antimicrobial proteins also contribute to epithelial defenses. These include the C-type lectin hepatocarcinoma-intestine-pancreas/pancreatic-associated protein (HIP/PAP), which is expressed by Paneth cells and enterocytes and which binds to gram-positive bacterial peptidoglycan ; phospholipase A2 (PLA2), which hydrolyses bacterial membrane phospholipids and is produced by Paneth cells and mononuclear phagocytes ; psoriasin (S100A7), which is produced by keratinocytes and inhibits gram-negative bacteria; and RNase7, which is expressed by the skin and the gastrointestinal and genitourinary epithelia, and inhibits gram-positive bacteria, including S. aureus skin colonization.

Skin

The barrier function of the skin is mediated primarily by its outermost layer, the stratum corneum , which consists of keratinocytes and the lipid rich matrix that surrounds them. These lipids, particularly ceramides, inhibit microbial growth, as does the low pH environment they help to create. The lipid content and acidic pH of the skin are established postnatally, reaching maturity by 2 to 4 weeks in term neonates but at a later age in premature neonates. Epithelial integrity and the antimicrobial barrier this provides are easily disrupted at this age. The skin of neonates is also coated by a water, protein, and lipid-rich material, the vernix caseosa. The skin is rapidly colonized by environmental bacteria after birth, creating a normal flora of commensal bacteria that help to prevent colonization by pathogens. This flora normally consists of coagulase-negative staphylococci (e.g., Staphylococcus epidermidis ), micrococci, and other species. Commensal bacteria, such as S. epidermidis, may play a protective role in the skin by secreting lipopeptides that bind to Toll-like receptor 2 (TLR2) on neonatal keratinocytes and stimulating them to produce hBD-2 and hBD-3 ; the role of TLRs in innate immunity is discussed in detail later. The organisms that make up the initial microbiota of the skin in newborns are heavily influenced by the mode of delivery, with vaginally delivered infants acquiring bacterial communities similar to the vaginal microbiome.

Antimicrobial peptides are expressed by neonatal keratinocytes and are present in the vernix caseosa . As in adults, the stratum corneum of skin from normal term neonates contains hBD-1. By the end of the second trimester of gestation, fetal skin constitutively expresses hBD-3, psoriasin, and RNase7, and neonatal skin expresses hBD-2 and LL-37, which are absent or present only in very low amounts in the skin of normal adults. The mechanisms underlying the apparent constitutive production of hBD-2 and LL-37 by neonatal keratinocytes are not known, but their presence may help to provide an immediate barrier against bacterial invasion during the initial exposure of the neonate to environmental microbes. Vernix caseosa may augment skin defenses and contains HNPs 1 to 3 and additional antibacterial proteins, including lysozyme and psoriasin (S100A7).

The Gastrointestinal Tract

The proximal gastrointestinal epithelium of the mouth and esophagus consists of a squamous epithelium, whereas the stomach, small intestine, and colon have a columnar epithelium with microvilli, which, along with intestinal peristalsis, help to maintain the longitudinal movement of fluid. The acidic pH of the stomach acts as a chemical barrier in adults. Gastric acidification is not yet fully developed in neonates, but digestion of milk lipids by gastric lipases may compensate in part by generating free fatty acids. The gastrointestinal tract is coated with a mucin-rich glycocalyx, which forms a viscous coating that helps to protect the epithelium and to which commensal intestinal bacteria bind. The composition of the intestinal glycocalyx in neonates differs from adults and may contribute to differences in commensal flora.

The application of high-throughput, comprehensive, culture-independent molecular approaches to assess microbial diversity has shown that the commensal intestinal flora of humans is a highly diverse ecologic system consisting of approximately 10 14 microorganisms, representing the most abundant and diverse microbial community in our body and exceeding the numbers (by a factor of 10) and genetic content of human cells in an individual. In adults, colonic and stool flora are dominated by gram-negative anaerobic bacteria (Bacteroides) and one phylum of gram-positive bacteria (Firmicutes), whereas Actinobacteria, Proteobacteria, and Verrucomicrobia, although found in many people, are typically minor constituents, and aerobic gram-negative bacteria (e.g., Escherichia coli ) are present in much lower abundance. The composition of stool flora in the first year of life is highly dynamic. Based on a longitudinal study of 14 term infants from whom serial samples were collected from birth to 1 year of age, the flora of individual infants differs substantially from one infant to another in the first months of life, initially most closely resembling the maternal fecal, vaginal, or breast-milk flora. In addition, the bacterial composition in infants is highly influenced by environmental factors, such as mode of delivery, geographic location, and life events, such as illness, antibiotic use, and changes in diet. This interindividual variability typically diminishes over time and by 1 year of age converges on a pattern similar to that found in adults. As more data become available linking microbiota composition to disease states, there will be a better understanding of how such factors affect the intestinal flora and, perhaps modulate the risk for necrotizing enterocolitis (NEC) and other inflammatory, infectious or allergic diseases in neonates and infants.

The dynamic interaction between host and microbe in the gut has an important impact on nutrition, intestinal homeostasis and development of innate and adaptive immunity. Such immunity, in turn, restricts these microbes to the gut and primes the immune system to respond properly to dangerous microbes. For example, recent murine studies have shown that commensal bacteria are important for systemically maintaining a relatively low threshold for activation in mononuclear phagocytes, which allows these cells to efficiently respond to systemic viral infection. Commensal bacteria appear to act by helping to maintain the appropriate epigenetic structure of promoters of genes involved in the antiviral inflammatory response so that these genes can be efficiently expressed in response to innate immune receptor engagement. Other murine studies have found a similar requirement of commensal bacteria for maintaining an appropriate threshold for DC function—in this case, the response to necrotic cells. If this priming of innate immune function by commensals occurs in humans, which appears likely, then limitations in neonatal innate immune cellular function would be expected to be most prominent immediately after birth, with increases in function as the commensal flora is established postnatally.

At the same time, commensal bacteria dampen the response to the normal flora, harmless environmental antigens, and self-antigens to prevent self-injury. Certain intestinal epithelial cells play special roles in intestinal immunity: goblet cells produce mucus, Paneth cells (located at the base of small intestinal crypts) secrete antimicrobial factors, and M cells deliver, by transcytosis, a sample of the distal small intestinal microbiota to antigen-presenting DCs located beneath the epithelium; some DCs also directly sample the intestinal lumen of the distal small intestine.

The intestinal epithelium can directly recognize and respond to microbes using a limited set of invariant cell surface, endosomal, and cytosolic innate immune pattern recognition receptors, including TLRs and others described later. How commensals prime innate and adaptive immunity in the gut without inducing deleterious inflammation and how potentially dangerous pathogens are discriminated from harmless commensals is an area of active investigation. This is achieved in part by innate immune receptor location. For example, intestinal epithelial cells normally express few or no TLRs on their luminal surface, where they are in contact with commensals. Conversely, pathogens that invade through or between epithelial cells can be recognized by endosomal TLRs, cytosolic innate immune recognition receptors, and TLRs located on the basolateral surface of epithelial cells. Moreover, certain commensal bacteria inhibit signaling and inflammatory mediator production downstream of these receptors or induce antiinflammatory cytokine production, thereby actively suppressing gut inflammation. The importance of one of these antiinflammatory cytokines, IL-10, in maintaining intestinal immune homeostasis after birth has been dramatically demonstrated by patients with genetic immunodeficiencies of IL-10 or its receptor components, all of whom present with severe inflammatory bowel disease in early infancy.

Adaptation of the intestinal epithelium to avoid unwarranted inflammatory responses to the normal flora is developmentally and/or environmentally regulated. Human 20- to 24-week fetal small intestinal organ cultures produced much more of the proinflammatory cytokine IL-8 (see Tables 4-1 and 4-2 ) when exposed to bacterial lipopolysaccharide (LPS) or IL-1 than similar cultures from infants or adults. Developmental differences in inflammatory signaling may also contribute to aberrant intestinal inflammation in preterm neonates with NEC. Recently, neonatal CD71 + erythroid cells in the mouse have been shown to have immunosuppressive properties to dampen immune activation in response to rapid colonization of microorganisms in the gut, and it will be of interest to determine if these cells play a similar role in the human intestine.

Intestinal epithelial cells produce and secrete defensins and other antimicrobial factors. Epithelial cells of the esophagus, stomach, and colon constitutively produce hBD-1 and LL-37, and produce hBD-2, hBD-3, and hBD-4 in response to infection and inflammatory stimuli. Intrinsic host defense of the small intestine is provided by Paneth cells, which constitutively produce HD-5 and lysozyme. The abundance of HD-5 in the neonatal small intestine correlates with the abundance of Paneth cells, which are present but much less abundant in the midgestation fetus than at term and which, in turn, are much less abundant than in adults. These data suggest that intrinsic small intestinal defenses may be compromised in human neonates, particularly when preterm. However, a study in the mouse suggests another possibility. The intestinal epithelium of neonatal mice expresses abundant amounts of cathelicidin, which is lost by 14 days of postnatal age, by which time Paneth cells expressing murine defensins reach adult numbers. Whether a similar “switch” in intestinal antimicrobial defenses occurs in humans is not known.

The Respiratory Tract

The respiratory tract is second only to the gut in epithelial surface area. The upper airways and larger airways of the lung are lined by pseudostratified ciliated epithelial cells, with smaller numbers of mucin-producing goblet cells, whereas the alveoli are lined by nonciliated type I pneumocytes and by smaller numbers of surfactant-producing type II pneumocytes. Airway surface liquid and mucociliary clearance mechanisms provide an important first line of defense. Airway surface liquid contains a number of antimicrobial factors, including lysozyme, secretory leukoprotease inhibitor (SLPI), defensins and LL-37, and surfactant apoproteins A and D (SP-A, SP-D). Collectively, these factors likely account for the lack of microbes in the lower respiratory tract of normal individuals.

Lung parenchymal cells express a diverse set of TLRs and other innate immune receptors. Lower airway epithelial cells express and respond to ligands for TLR2, TLR4, and TLR5, whereas airway epithelial cells express TLR3, TLR7, and TLR9 primarily on the apical surface in the human trachea. However, to our knowledge, there are no data carefully comparing TLR function in the airways of the human fetus and neonate versus human adults; data from neonatal sheep (and rodents) indicate developmental differences. TLR2 and TLR4 are expressed in the lungs of fetal sheep in the latter part of gestation: messenger RNA (mRNA) abundance increases from 20% of adult values at the beginning of the third trimester to 50% at term. TLR4 mRNA was present in the airway epithelium and parenchyma, whereas TLR2 expression was found primarily in inflammatory cells after intraamniotic administration of LPS, which resulted in increased expression of TLR2 and TLR4. TLR3 expression was approximately 50% of adult values and unchanged in response to LPS. Similar developmental differences have been observed in mice. In the human fetal lung, transcripts for TLR2, TLR3, TLR5, TLR6, TLR7, TLR8, and TLR10 increase between 3 months and 4 to 5 months of gestation, whereas TLR4 expression remains unchanged. However, the relationship of these TLR mRNA levels to protein expression, the cellular sources of these transcripts, and the biologic activity of these TLRs during fetal development, which requires the presence of intracellular signaling adapters, for instance, MyD88 (myeloid differentiation primary response protein 88), and in some cases, accessory surface molecules, for instance, CD14 and MD2 for TLR4 signaling, remains unclear.

Airway epithelial cells express hBD-1 constitutively and hBD-2, hBD-3, hBD-4, and LL-37 in response to a variety of microbial stimuli and inflammatory cytokines, including IL-1. Lung explants from term but not preterm fetuses expressed hBD-2 and smaller amounts of hBD-1 (although the amount even at term appeared to be less than at older ages) but did not contain hBD-3 mRNA. By contrast, LL-37 mRNA was present and appeared not to vary at these ages. Consistent with these findings, tracheal aspirates from mechanically ventilated term, but not preterm neonates, contained hBD-2, whereas lower amounts of LL-37 were found, but these amounts were similar in aspirates from term and preterm neonates. However, another study found no difference in the abundance of hBD-1, hBD-2, and LL-37 in aspirates from ventilated neonates, ranging in ages from 22 to 40 weeks,

Surfactant proteins SP-A and SP-D are produced by type II pneumocytes and by Clara cells, which are progenitors of ciliated epithelial cells located at the bronchoalveolar junction. SP-A and SP-D are members of the collectin family. Collectins are involved in both antimicrobial host defense as well as the clearance of apoptotic cells by human alveolar macrophages and other mononuclear phagocytes. Collectins bind to carbohydrates, including mannose, glucose, and fucose, found on the surface of gram-positive and gram-negative bacteria, yeasts, and some viruses, including RSV. Once bound, collectins can result in aggregation of microbes, which may inhibit their growth or facilitate their mechanical removal, or they can opsonize microbes, that is, facilitate their ingestion by phagocytic cells. Mice lacking SP-A or SP-D have impaired lung clearance of group B streptococci (GBS), Haemophilus influenzae, Pseudomonas aeruginosa, and RSV. SP-A and SP-D are detectable in human fetal lungs by 20 weeks of gestation, and amounts appear to increase with increasing fetal maturity and in response to antenatal steroid administration. Low levels of SP-A or SP-D are correlated with a higher risk of infection and the development of chronic lung disease in premature infants.

Summary

The skin of neonates, particularly preterm neonates, is more readily disrupted and lacks the protection provided by an acidic pH until approximately 1 month of postnatal age. Counterbalancing these factors is the constitutive production in neonates of a broader array of antimicrobial peptides by the skin epithelium and the presence of such peptides in the vernix caseosa . The lack of an acidic pH in the stomach may facilitate the establishment of the protective commensal flora, which at birth varies substantially from infant to infant, converging by 1 year of age to resemble adult flora. The lack of gastric acidity and diminished numbers of antimicrobial peptide-producing Paneth cells in the small intestine of preterm and, to a lesser degree, term neonates may increase their risk for enterocolitis and invasion by pathogens; these deficits may be counterbalanced by more robust production of antimicrobial peptides by other intestinal epithelial cells, but as yet, this has only been demonstrated in animal models. Innate defenses of the respiratory epithelium—TLRs, antimicrobial peptides, SP-A, and SP-D—mature in the last trimester. Consequently, these defenses may be compromised in preterm infants. Reduced numbers of resident alveolar macrophages may further impair lung innate defenses in preterm infants (see “Mononuclear Phagocytes” ).

Complement and Other Humoral Mediators of Innate Immunity

Collectins, Pentraxins, and Ficolins

C-reactive protein (CRP) and mannose-binding lectin (MBL) are soluble proteins that can bind to structures found on the surface of microbes and infected or damaged host cells and facilitate their clearance by phagocytes. Both are produced by the liver. Their concentrations in the blood increase in response to infection and tissue injury as part of the acute-phase response, allowing them to contribute to early host defense to infection and the clearance of damaged cells.

CRP is a member of the pentraxin family of proteins, which binds to phosophocholine and other lipids and carbohydrates on the surface of certain gram-positive bacteria, particularly Streptococcus pneumoniae, fungi, and apoptotic host cells. It does not cross the placenta. Term and preterm neonates can produce CRP as well as adults. Values of CRP in cord blood from term infants are low, rising to concentrations found in adult blood in the first days of life, paralleling a postnatal increase in serum IL-6 and microbial colonization.

MBL (like SP-A and SP-D described earlier) is a member of the collectin family and binds to carbohydrates, including mannose, glucose, and fucose, on the surface of bacteria, yeasts, and some viruses. When bound, MBL activates complement and enhances phagocytosis by neutrophils and macrophages. Engagement of MBL is impeded by capsular polysaccharides of most virulent bacterial pathogens. MBL abundance in neonates is affected by three interacting variables: MBL genotype, gestational age, and postnatal age. In neonates with a MBL genotype lacking mutations, MBL concentrations are 50% to 75% of those in adults and reach adult values by 7 to 10 days and 20 weeks of age in term and preterm neonates, respectively. Concentrations are more than fivefold lower, and these increases are less evident in those with variant MBL genotypes. Preterm neonates with low concentrations of MBL found in those with variant genotypes appear to be at greater risk for sepsis or pneumonia.

Ficolins are multimeric lectins and come in three forms: L-ficolin (ficolin-2), which is made by the liver; H-ficolin (ficolin-3), which is produced by the liver and lung; and M-ficolin (ficolin-1), which is produced by mononuclear phagocytes. All three ficolins are detectable in serum and can activate the lectin-dependent complement pathway, which is described later. Ficolins recognize acetylated compounds, including N -acetyl-glucosamine, N -acetyl-galactosamine, N -acetylneuraminic acid, and sialic acid and bind bacteria, such as S. aureus, GBS, Salmonella, and E. coli. Although serum levels of ficolins are highly variable, in one study, all three ficolins were in lower concentration in prematurely born infants compared with term neonates and older children and adults; only M-ficolin in children older than 1 year and H-ficolin in term neonates and children were comparable to the values found in adults. However, despite these differences, there were still substantial levels of all three ficolins in premature infants, and it is unclear to what extent these modest decreases limit lectin-dependent complement activation and predispose to neonatal bacteremia and sepsis.

Complement

The complement system is composed of serum proteins that can be activated sequentially through one of three pathways—the classical, MBL, and alternative pathways, each of which leads to the generation of activated C3, C3, and C5 convertases and the membrane attack complex (see Fig. 4-1 ).

Classical and Mannose-Binding Lectin Pathways

Activation of the classical pathway is initiated when antibodies capable of engaging C1q to their Fc portion (immunoglobulin M [IgM], IgG1, IgG2, and IgG3 in humans) complex with microbial (or other) antigens. After C1q binds to the immune complex, there is sequential binding of C1r and C1s to C1q. C1s can then cleave C4, followed by C2, and the larger fragments of these bind covalently to the surface of the microbe or particle, forming the classical pathway C3 convertase (C2aC4b). C3 convertase cleaves C3, thereby liberating C3b, which binds to the microbe or particle, and C3a, which is released into the fluid phase. This pathway can also be activated directly by CRP. When CRP binds to the surface of a microbe, its conformation is altered such that it can bind C1q and activate the classical pathway. Similarly, when MBL or ficolins engage the surface of a microbe, their confirmation is altered, creating a binding site for mannose-binding lectin–associated serine protease (MASP)1 and MASP2. MASP2, in turn, cleaves C4 and C2, leading to the formation of the C3 convertase. Independently of their activation of downstream complement components, Clq and MBL can also bind apoptotic cells and initiate their uptake into mononuclear phagocytes by macropinocytosis.

Alternative Pathway

The alternative pathway is activated constitutively by the continuous low-level hydrolysis of C3 in solution, creating a binding site for factor B. This complex is in turn cleaved by factor D, generating C3b and Bb. If C3b and Bb bind to a microorganism, they form a more efficient system, which binds and activates additional C3 molecules, depositing C3b on the microbe and liberating C3a into the fluid phase. This interaction is facilitated by factor P (properdin) and inhibited by alternative pathway factors H and I. The classical pathway, by creating particle-bound C3b, also can activate the alternative pathway, thereby amplifying complement activation. This amplification step may be particularly important in the presence of small amounts of antibody. Bacteria vary in their capacity to activate the alternative pathway, which is determined by their ability to bind C3b and to protect the complex of C3b and Bb from the inhibitory effects of factors H and I. Sialic acid, a component of many bacterial polysaccharide capsules, including those of GBS and E. coli K1, favors factor H binding. Thus many bacterial pathogens are protected from the alternative pathway by their capsules and/or by proteins that bind to factor H. Antibody is needed for efficient opsonization of such organisms.

Terminal Components, Membrane Attack Complex, and Biologic Consequences of Complement Activation

Binding of C3b on the microbial surface facilitates microbial killing or removal through the interaction of C3b with CR1 receptors on phagocytes. C3b also is cleaved to C3bi, which binds to the CR3 receptor (Mac-1, Cd11b-CD18) and CR4 receptor (CD11c-CD18). C3bi receptors are β 2 integrins, which are present on neutrophils, mononuclear phagocytes, and certain other cell types and also play a role in leukocyte adhesion. Along with IgG antibody, which binds to Fcγ receptors on phagocytes, C3b and C3bi promote phagocytosis and killing of bacteria and fungi. Bound C3b, together with C4b and C2a or together with Bb, form C5 convertases, which cleave C5. The smaller fragment, C5a, is released into solution. The larger fragment, C5b, triggers the recruitment of the terminal components, C6 to C9, which together form the membrane attack complex. This complex is assembled in lipid-containing cell membranes, which include the outer membrane of gram-negative bacteria and the plasma membrane of infected host cells. Once assembled in the membrane, this complex can lyse the cell. Such lysis appears to be a central defense mechanism against meningococcal and systemic gonococcal infection. Certain gram-negative organisms have mechanisms to impede complement-mediated lysis, and gram-positive bacteria are intrinsically resistant to complement-mediated lysis because they do not have an outer membrane.

The soluble fragment of C5, C5a, and, to a more limited degree, C3a and C4a cause vasodilatation and increase vascular permeability. C5a also is a potent chemotactic factor for phagocytes. In addition to these roles for complement in innate immunity, complement facilitates B-cell responses to T-cell–dependent antigens as discussed in the section B-Cell Activation and Immune Selection .

Complement in the Fetus and Neonate

Complement components are synthesized by hepatocytes and, for some components, also by macrophages. Little, if any, maternal complement is transferred to the fetus. Fetal synthesis of complement components can be detected in tissues as early as 6 to 14 weeks of gestation, depending on the specific complement component and tissue examined.

Table 4-3 summarizes published reports on classical pathway complement activity (CH 50 ) and alternative pathway complement activity (AP 50 ) and individual complement components in neonates. Substantial individual variability is seen, and in many term neonates, values of individual complement components or of CH 50 or AP 50 are within the adult range. Alternative pathway activity and components are more consistently decreased than are classical pathway activity and components. The most marked deficiency is in the terminal complement component C9, which correlates with poor killing of gram-negative bacteria by serum from neonates. The C9 deficiency in neonatal serum appears to be a more important factor in the inefficient killing of E. coli K1 than the deficiency in antigen-specific IgG antibodies. Preterm infants demonstrate a greater and more consistent decrease in both classical and alternative pathway complement activity and components. Mature but small-for-gestational-age infants have values similar to those for healthy term infants. The concentration of most complement proteins increases postnatally and reaches adult values by 6 to 18 months of age.

Table 4-3
Summary of Published Complement Levels in Neonates
Data from Johnston RB, Stroud RM: Complement and host defense against infection, J Pediatr 90:169-179, 1977; Notarangelo LD, Chirico G, Chiara A, et al: Activity of classical and alternative pathways of complement in preterm and small for gestational age infants, Pediatr Res 18:281-285, 1984; Davis CA, Vallota EH, Forristal J: Serum complement levels in infancy: age related changes, Pediatr Res 13:1043-1046, 1979; Lassiter HA, Watson SW, Seifring ML, et al: Complement factor 9 deficiency in serum of human neonates, J Infect Dis 166:53-57, 1992; Wolach B, Dolfin T, Regev R, et al: The development of the complement system after 28 weeks’ gestation, Acta Paediatr 86:523-527, 1997; and Zilow G, Brussau J, Hauck W, et al: Quantitation of complement component C9 deficiency in term and preterm neonates, Clin Exp Immunol 97:52-59, 1994.
Mean % of Adult Levels
Complement Component Term Neonate Preterm Neonate
CH 50 56-90 (5) 45-71 (4)
AP 50 49-65 (4) 40-55 (3)
CIq 61-90 (4) 27-58 (3)
C4 60-100 (5) 42-91 (4)
C2 76-100 (3) 67-96 (2)
C3 60-100 (5) 39-78 (4)
C5 73-75 (2) 67 (1)
C6 47-56 (2) 36 (1)
C7 67-92 (2) 72 (1)
C8 20-36 (2) 29 (1)
C9 <20-52 (3) <20-41 (2)
B 35-64 (4) 36-50 (4)
P 33-71 (6) 16-65 (3)
H 61 (1)
C3bi 55 (1)

Number of studies.

Opsonization is the process whereby soluble factors present in serum or other body fluids bind to the surface of microbes (or other particles) and thereby enhance their phagocytosis and killing. Some organisms are effective activators of the alternative pathway, whereas others require antibody to activate complement. Thus, depending on the organism, opsonic activity reflects antibody, MBL, ficolin, CRP, classical or alternative complement pathway activity, or combinations of these. Accordingly, it is not surprising that the efficiency with which neonatal sera opsonize organisms is quite variable. For example, although opsonization of S. aureus was normal in neonatal sera in all studies, opsonization of GBS, S. pneumoniae, E. coli, and other gram-negative rods was decreased against some strains and in some studies but not in others.

Neonatal sera generally are less able to opsonize organisms in the absence of antibody. This difference is compatible with deficits in the function of the alternative and MBL pathways and with the moderate reduction in alternative pathway components. This difference is not due to a reduced ability of neonatal sera to initiate complement activation through the alternative pathway. Neonatal sera also are less able to opsonize some strains of GBS in a classical pathway–dependent but antibody-independent manner. The deficit in antibody-independent opsonization is accentuated in sera from premature neonates, in whom there is significantly reduced levels of circulating MBL, MASP, and all three ficolins and may be further impaired by the depletion of complement components in septic neonates.

Sera from term neonates generate less chemotactic activity than adult sera. This diminished activity reflects a defect in complement activation rather than lack of antibody. These observations notwithstanding, preterm and term neonates do generate substantial amounts of activated complement products in response to infection in vivo.

Summary

Compared with adults, neonates have moderately diminished alternative complement pathway activity, slightly diminished classical complement pathway activity, and decreased abundance of some terminal complement components. Neonates with much reduced concentrations of MBL resulting from genetic variation and prematurity appear to be at greater risk for sepsis or pneumonia. Consistent with these findings, neonatal sera are less effective than adult sera in opsonization when concentrations of specific antibody are limiting and in the generation of complement-derived chemotactic activity; these differences are greater in preterm than in term neonates. These deficiencies, in concert with phagocyte deficits described later, may contribute to delayed inflammatory responses and impaired bacterial clearance in neonates.

Phagocytes

Hematopoiesis

Phagocytes as well as all leukocytes of the immune system are derived from self-renewing, pluripotent hematopoietic stem cells (HSCs), which have the capacity for indefinite self-renewal ( Fig. 4-2 ). Most circulating HSCs in cord blood and adult bone marrow are identified by their CD34 + CD45 + CD133 + CD143 + surface phenotype, combined with a lack of expression of CD38 and markers found on specific lineages of mature leukocytes; for instance, they lack CD3, a T-cell marker, and are thus CD34 positive and lineage-marker negative (CD34 + Lin ). HSCs are generated during ontogeny from embryonic para-aortic tissue, fetal liver, and bone marrow. The yolk sac, which is extraembryonic, is a major site of production of primitive erythrocytes and some primitive mononuclear phagocytes, starting at about the third week of embryonic development. HSCs that give rise to both erythrocyte and all nonerythroid hematopoietic cell lineages appear in the fetal liver after 4 weeks of gestation and in the bone marrow by 11 weeks of gestation. Liver-mediated hematopoiesis ceases by 20 weeks of gestation, with the bone marrow becoming the sole site of hematopoiesis thereafter. All major lineages of hematopoietic cells that are part of the immune system are present in the human by the beginning of the second trimester.

Figure 4-2, Myeloid and lymphoid differentiation and the tissue compartments in which they occur. CDP, Committed dendritic cell progenitor; CFU-GM, colony-forming unit–granulocyte-macrophage; CMP, common myeloid progenitor; NK, natural killer.

HSCs can subsequently differentiate into common lymphoid progenitors or common myeloid-erythroid progenitors (see Fig. 4-2 ). Common lymphoid progenitors give rise to T, B, and natural killer (NK) lymphocytes, which will be discussed in later sections of this chapter. Common myeloid-erythroid progenitors give rise to the megakaryocyte, erythroid, and myeloid lineages. Myeloid and lymphoid cells represent the two largely distinct but functionally interrelated immune cell lineages, with DCs, which are myeloid derived, providing a key functional bridge between these lineages (see “Dendritic Cells” later).

As was first described in mice and birds, fetal HSCs and adult HSCs in humans appear to give rise to T-cell populations that have distinct properties. For example, HSCs of the second trimester of pregnancy give rise to naïve CD4 T cells that have many features of regulatory T cells (see “Regulatory T Cells” in the “T Cells and Antigen Presentation” section), whereas those of the adult give rise to naïve CD4 T cells that tend to become effector cells rather than regulatory cells. This appears to be a cell-intrinsic property of fetal versus adult HSCs, and it is possible that other HSC derivatives, such as monocytes and NK cells, may also differ between the fetus and adult because of these developmentally related cell-intrinsic properties.

Phagocyte Production by the Bone Marrow

Phagocytes are derived from a common precursor myeloid stem cell, which often is referred to as the colony-forming unit–granulocyte-monocyte (CFU-GM) (see Fig. 4-2 ). The formation of myeloid stem cells from pluripotent HSCs and further differentiation of the myeloid precursor into mature granulocytes and monocytes are governed by bone marrow stromal cells and soluble colony-stimulating factors (CSFs) and other cytokines (see Table 4-2 ). In response to an infectious or inflammatory stimulus, the production of granulocyte-specific colony-stimulating factor (G-CSF), granulocyte-macrophage–specific colony-stimulating factor (GM-CSF), and certain other cytokines is increased, resulting in increased production and release of granulocytes and monocytes. Similarly, when given exogenously, these factors enhance production and function of the indicated cell lineages.

Innate Immune Pattern Recognition Receptors Used by Leukocytes

Monocytes, macrophages, DCs, neutrophils, and other cells of the innate immune system use invariant innate immune pattern receptors to discriminate between microbes and self, or things that are “dangerous” or are not. These receptors recognize microbial structures (commonly referred to as pathogen-associated molecular patterns [PAMPs] and danger-associated molecular patterns [DAMPS]), produced by infected or injured host cells—molecules such as uric acid, adenosine triphosphate (ATP), heat shock proteins (HSPs), and high-mobility group box 1 (HMGB1) protein. Recognition is followed by signals that activate the innate immune response.

Toll-like Receptors

Toll-like receptors are a family of structurally related proteins and are the most extensively characterized set of innate immune pattern recognition receptors. Ten different TLRs have been defined in humans. Their distinct ligand specificities, subcellular localization, and patterns of expression by specific cell types are shown in Table 4-4 .

Table 4-4
Human Toll-like Receptors (TLRs)
TLR Microbial Ligands Site of Interaction with Ligand Signal Transduction/Effector Molecules Expression by Antigen-Presenting Cells
TLR1 See TLR2 Cell surface MyD88-dependent induction of cytokines Monocytes and B cells > cDCs
TLR2 Bacterial peptidoglycan, lipoteichoic acid and lipopeptides; mycobacterial lipoarabinomannan, recognition of some ligands is mediated by TLR2/TLR1 or TLR2/TLR6 heterodimers Cell surface MyD88-dependent induction of cytokines Monocytes and cDCs > B cells
TLR3 Double-stranded RNA Endosome TRIF-dependent induction of type I IFNs and cytokines cDCs
TLR4 LPS, RSV Cell surface MyD88-dependent and TRIF-dependent induction of cytokines; TRIF-dependent induction of type I IFNs Monocytes > cDCs
TLR5 Flagellin Cell surface MyD88-induction of cytokines Monocytes > cDCs
TLR6 See TLR2 Cell surface MyD88-dependent induction of cytokines Monocytes and cDCs, B cells > pDCs
TLR7 Single-stranded RNA, imidazoquinoline drugs Endosome MyD88-dependent induction of type I IFNs and cytokines pDCs and B cells >> cDCs > monocytes
TLR8 Single-stranded RNA, imidazoquinoline drugs Endosome MyD88-dependent induction of type I IFNs and cytokines Monocytes and cDCs >> B cells
TLR9 Unmethylated CpG DNA Endosome MyD88-dependent induction of cytokines and type I IFNs pDCs and B cells
TLR10 Viral RNA-protein complexes Unknown Unknown Monocytes (postviral infection), B cells > cDCs and pDCs
cDCs, Conventional dendritic cells; CpG, cytosine-guanine dinucleotide; IFN, interferon; LPS, lipopolysaccharide; MyD88, myeloid differentiation primary response protein 88; pDCs, plasmacytoid dendritic cells; RSV, respiratory syncytial virus; TLR, Toll-like receptor; TRIF, TIR-domain-containing adaptor-inducing interferon-β.

TLR4 forms a functional LPS receptor with myeloid differentiation (MD-2) protein, a soluble protein required for surface expression of the TLR4/MD-2 receptor complex. CD14, which is expressed abundantly on the surface of monocytes and also exists in a soluble form in the plasma, facilitates recognition by the TLR4/MD-2 complex and is essential for recognition of smooth LPS present on pathogenic gram-negative bacteria. TLR2, which forms a heterodimer with TLR1 or TLR6, recognizes bacterial lipopeptides, lipoteichoic acid, and peptidoglycan, and this recognition is facilitated by CD14. TLR2 has a central role in the recognition of gram-positive bacteria and also contributes to recognition of fungi, including Candida species. TLR5 recognizes bacterial flagellin. Consistent with their role in recognition of microbial cell surface structures, these TLRs are displayed on the cell surface.

By contrast, TLRs 3, 7, 8, and 9 recognize nucleic acids: TLR3 binds double-stranded RNA, TLR7 and TLR8 bind single-stranded RNA, and TLR9 binds nonmethylated cytosine-guanine dinucleotide (CpG)-containing DNA. These TLRs appear primarily to function in antiviral recognition and defense, and TLR9 also contributes to defense against bacteria and fungi. These TLRs preferentially recognize features of nucleic acids that are more common in microbes than mammals, but their specificity may be as much based on location as nucleic sequence: TLRs 3, 7, 8, and 9 detect nucleic acids in a location where they should not be found—acidified late endolysosomes. Individuals lacking TLR3; TRIF (TIR-domain–containing adaptor-inducing interferon-β), which is required for intracellular signaling by TLR3; or a protein UNC93B, required for proper localization of TLRs 3, 7, 8, and 9 to endosomes, are unduly susceptible to infection with herpesviruses, particularly primary herpes simplex encephalitis.

A conserved cytoplasmic Toll/IL-1 receptor (TIR) domain links TLRs to downstream signaling pathways by interacting with adaptor proteins, including myeloid differentiation primary response protein 88 (MyD88) and TRIF. MyD88 is involved in signaling downstream of all TLRs with the exception of TLR3, which signals solely through TRIF. TLR signaling via MyD88 leads to the activation and translocation of the transcription factor nuclear factor kappa B (NF-κB) to the nucleus and to the induction or activation by extracellular signal-regulated kinase (ERK)/p38/c-Jun N -terminal kinase (JNK)/mitogen-activated protein kinases (MAPK) of other transcription factors, resulting in the production of the proinflammatory cytokines, including TNF-α, IL-1, and IL-6. Production of the antiinflammatory and immunomodulatory cytokine IL-10 is dependent on signal transducer and activator of transcription 3 (STAT3) activation in addition to NF-κB and MAPK activation.

In addition to these transcription factors, activation of interferon regulatory factor 3 (IRF3) and/or IRF7 is required for the induction of type I interferons (IFNs), which are key mediators of antiviral innate immunity. Activation of IRF3 and the production of type I IFNs downstream of TLR4 are dependent on TRIF. TRIF is also essential for the activation of IRF3 and for the production of type I IFNs and other cytokines via TLR3. Conversely, TLRs 7, 8, and 9 use the adaptor MyD88 to activate IRF7 and to induce the production of type I IFNs and other cytokines. Consistent with their role in detection of bacterial but not viral structures, TLR2 and TLR5 do not induce type I IFNs.

The production of IL-12 and IL-27 (but not of the structurally related cytokine IL-23) is, like the production of type I IFNs, dependent on IRF3 and IRF7. Consequently, signals via TLR3, TLR4, and TLR7/8, but not via TLR2 and TLR5, can induce the production of these two cytokines; by contrast, each of these TLRs, except TLR3, which signals exclusively via TRIF, can induce the production of IL-23. IL-23 promotes the production of IL-17 and IL-22, which contribute to host defenses to extracellular bacterial and fungal pathogens, whereas IL-12 and IL-27 stimulate interferon-gamma (IFN-γ) production by NK cells and thereby facilitate defense against viruses and other intracellular pathogens. Thus, through the concerted regulation of IL-12, IL-27, and type I IFNs, IRF3 and IRF7 link TLR recognition to host defenses against intracellular pathogens.

Recent studies have also found that engagement of TLR7 and/or TLR8 can induce the production of high levels of IL-1β in a caspase-1–dependent manner in monocytes and DCs. As discussed immediately below, IL-1β secretion in response to many non-TLR stimuli involves the activation of caspase-1 by the inflammasome and the caspase-1–dependent cleavage of pro–IL-1β, but it is unclear if caspase-1 activation by the TLR7/8 pathway involves the inflammasome or a novel mechanism.

Nucleotide-Binding Domain– and Leucine-Rich Repeat–Containing Receptors (NLRs)

NLRs are a family of 22 proteins in humans, most of which play a role in innate immunity activation in response to microbial products or endogenously derived danger signals. These include nucleotide oligomerization domain 1 (NOD1), NOD2, and NLRP3 (also known as NALP3), which recognize components of bacterial peptidoglycan; NLRP3 is also involved in responses to components of gram-positive bacteria, including bacterial RNA and DNA, products of injured host cells such as uric acid, and noninfectious foreign substances, including asbestos and aluminum salts, which are widely used as the adjuvant alum ; NOD-like receptor C4 (NLRC4) (also known as IL-1-converting enzyme protease activating factor, or IPAF) is involved in responses to Salmonella and, like TLR5, the PAMP recognized is flagellin. NOD1 and NOD2 can activate MAP kinase and NF-κB pathways and proinflammatory cytokines in synergy with TLRs. By contrast, NLRP3 interacts with the mitochondrial antiviral signaling (MAVS) adaptor protein to activate the inflammasome, a protein platform that activates caspase-1, which, in turn, cleaves pro–IL-1 and pro–IL-18, allowing the secretion of the mature forms of these inflammatory cytokines. NLRC4 directly binds to caspase-1 and induces its activation. In addition to leading to IL-1 and IL-18 secretion, caspase-1 activation results in pores in the cell membrane and, as a consequence, a form of cell death known as pyroptosis.

Three NLR members that are not associated with inflammasomes—CTIIA, NLRC5, and NLRP10—have distinct roles in augmenting antigen presentation rather than in regulating innate immunity responses: CTIIA (class II activator) is a transcriptional coactivator that is essential for the upregulation of major histocompatibility complex (MHC) class II expression by IFN-γ ; NLRC5, also known as CITA, plays an analogous role for the IFN-γ–dependent enhancement of the MHC class I antigen presentation pathway by augmenting transcription of the MHC class I heavy and light chain genes and transporter associated with antigen processing (TAP)-1; NLRP10 appears to play an essential role in the migration of DCs from inflamed tissues via afferent lymphatics to the TD areas of draining lymph nodes.

Retinoic Acid–Inducible Gene-I–Like Receptors

There are three members of the retinoic acid–inducible gene-I (RIG)-I–like receptor (RLR) family: RIG-I, melanoma-differentiation–associated protein (MDA-5), and Laboratory of Genetics and Physiology 2 (LGP2). RLRs are present in the cytoplasm of nearly all mammalian cells, where they provide rapid, cell-intrinsic, antiviral surveillance. RIG-I is important for host resistance to a wide variety of RNA viruses, including influenza, parainfluenza, and hepatitis C virus, whereas MDA-5 is important for resistance to picornaviruses. RIG-I and MDA-5 interact with a common signaling adaptor (MAVS or IFN-β–promoter stimulator 1 [IPS-1]), which, like TRIF in the TLR3/4 pathway, induces the phosphorylation of IRF3 to stimulate type I IFN production.

C-Type Lectin Receptors

C-type lectin receptors (CLRs) are a family of surface proteins, which include DC-SIGN (dendritic cell–specific ICAM-3–grabbing nonintegrin), a receptor on DCs that is involved in their interaction with human immunodeficiency virus (HIV); the macrophage mannose receptor; dectin-1 and dectin-2, which are expressed by DCs and macrophages; and CLEC9A, which facilitates the uptake of necrotic material by DCs. Dectin-1 and dectin-2 bind to fungi, such as Candida, and activate cytokine production by a signaling pathway that involves the spleen tyrosine kinase (Syk) and caspase activation and recruitment domain 9 (CARD9), an adaptor protein that links Syk activity to the induction of NF-κB and activator protein-1 (AP-1), resulting in the production of cytokines. The recognition by dectins of the hyphal forms of fungi, which is indicative of invasion rather than colonization of a tissue, results in the preferential induction of cytokines, for instance, IL-1β, IL-6, and IL-23, that favor naïve CD4 T cells recognizing fungal antigens differentiating into Th17 cells. The secretion of IL-1β after dectin-1 engagement involves the processing of pro–IL-1β via a novel caspase-8 inflammasome rather than a canonical caspase-1–containing inflammasome. The importance of this pathway for fungal host defense is illustrated by the patients prone to opportunistic fungal infection who have genetic deficiencies of dectin, CARD9, or IL-17.

Cytoplasmic DNA Receptors

Cytoplasmic receptors for double-stranded DNA are found in leukocytes and other cell types that, upon binding of DNA, lead to the production of type I IFNs. The best characterized of these receptors involves cyclic guanosine monophosphate–adenosine monophosphate (GMP-AMP) synthase (cGAS) coupled with the STING (stimulator of interferon genes) adaptor protein. Upon DNA binding, cGAS synthesizes a novel cyclic dinucleotide, cyclic guanosine monophosphate–adenosine monophosphate (cGAMP), that binds to STING, which, in turn, activates kinase pathways that result in IRF3 phosphorylation and its translocation to the nucleus where type I IFN transcription is induced. The source of the DNA can be viral or bacterial and, unlike TLR9 DNA recognition, there is no preference for a particular nucleotide sequence, and the cytosine methylation status of CpGs also does not influence receptor activity. STING may also be coupled with other mammalian cytoplasmic DNA receptors, such as DDX41, but their role in host defense versus that of cGAS remains unclear. STING and innate immune responses can also be directly triggered by cyclic dinucleotides that are generated by bacteria, for instance, during intracellular infection with Listeria monocytogenes, but are biochemically distinct from the cyclic dinucleotides generated by cGAS.

Decoding the Nature of the Threat Through Combinatorial Receptor Engagement

The differing molecular components of specific microbes result in the engagement of different combinations of innate immune recognition receptors. The innate immune system uses combinatorial receptor recognition patterns to decode the nature of the microbe and then tailors the ensuing early innate response and the subsequent antigen-specific response to combat that specific type of infection. Extracellular bacteria engage TLRs 2, 4, and/or 5 on the cell surface and also activate NLRs, providing a molecular signature of this type of pathogen. This leads to the production of proinflammatory cytokines and IL-23 to recruit neutrophils and support the development of a T-helper (Th)17-type T-cell response (see “Differentiation of Activated Naïve T Cells into Effector and Memory Cells” ). Fungal products engage TLR2 and dectins, leading to a similar response. Conversely, virus recognition via TLRs 3, 7, 8, 9, and RLRs (for RNA viruses) and cGAS/STING (for DNA viruses) stimulates the production of type I IFNs and IFN-induced chemokines (e.g., CXCL10), which, in turn, induce and recruit CD8 and Th1 CD4 T cells. Nonviral intracellular bacterial pathogens also induce type I IFNs, such as via the TLR9 and cGAS/STING pathways, which collaborate with signals from cell surface TLRs and NLRs to induce the production of IL-12, resulting in Th1-type responses. The importance of these innate sensing mechanisms is underscored by strategies that pathogenic microbes have evolved to evade them and the mediators they induce.

Neutrophils

Production

Polymorphonuclear leukocytes or granulocytes, including neutrophils, eosinophils, and basophils, are derived from CFU-GM. Neutrophils are the principal cells of interest in relation to defense against pyogenic pathogens. The first identifiable committed neutrophil precursor is the myeloblast, which sequentially matures into myelocytes, metamyelocytes, bands, and mature neutrophils. Myelocytes and more mature neutrophilic granulocytes cannot replicate and constitute the postmitotic neutrophil storage pool. The postmitotic neutrophil storage pool is an important reserve because these cells can be rapidly released into the circulation in response to inflammation. Mature neutrophils enter the circulation, where they remain for approximately 8 to 10 hours and are distributed equally and dynamically between circulating cells and those cells adherent to the vascular endothelium. After leaving the circulation, neutrophils do not recirculate and die after approximately 24 hours. Release of neutrophils from the marrow may be enhanced in part by cytokines, including IL-1, IL-17, and TNF-α, in response to infection or inflammation.

Neutrophil precursors are detected at the end of the first trimester, appearing somewhat later than macrophage precursors. Mature neutrophils are first detected by 14 to 16 weeks of gestation, but at midgestation, the numbers of postmitotic neutrophils in the fetal liver and bone marrow remain markedly lower than in term newborns and adults. By term, the numbers of circulating neutrophil precursors are 10- to 20-fold higher in the fetus and neonate than in the adult, and neonatal bone marrow also contains an abundance of neutrophil precursors. However, the rate of proliferation of neutrophil precursors in the human neonate appears to be near maximal, suggesting that the capacity to increase numbers in response to infection may be limited.

At birth, neutrophil counts are lower in preterm than term neonates and in neonates born by cesarean section without labor. Within hours of birth, the numbers of circulating neutrophils increase sharply. The number of neutrophils normally peaks shortly thereafter, whereas the fraction of neutrophils that are immature (bands and less mature forms) remains constant at about 15%. Peak counts occur at approximately 8 hours in infants born at greater than 28 weeks gestation and at approximately 24 hours in those born at less than 28 weeks gestation, then decline to a stable level by approximately 72 hours in those born without complications. Thereafter, the lower limit of normal for term and preterm neonates is approximately 2500 and 1000 per μL, respectively, and the upper limit of normal is approximately 7000 per μL for both term and preterm neonates.

Values may be influenced by a number of additional factors. Most important is the response to sepsis. Septic infants may have normal or increased neutrophil counts. Sepsis and other perinatal complications, including maternal hypertension, periventricular hemorrhage, and severe asphyxia, can cause neutropenia, however, and severe or fatal sepsis often is associated with persistent neutropenia, particularly in preterm neonates. Neutropenia may be associated with increased margination of circulating neutrophils, which occurs early in response to infection. However, neutropenia that is sustained often reflects depletion of the neonate’s limited postmitotic neutrophil storage pool. Septic neutropenic neonates in whom the neutrophil storage pool is depleted are more likely to die than are those with normal neutrophil storage pools. Leukemoid reactions also are observed at a frequency of approximately 1% in term neonates in the absence of an identifiable cause. Such reactions appear to reflect increased neutrophil production.

Circulating G-CSF levels in healthy infants are highest in the first hours after birth, and levels in premature neonates are generally higher than in term neonates. Levels decline rapidly in the neonatal period and more slowly thereafter. Plasma G-CSF levels tend to be elevated in infected neonates. Mononuclear cells and monocytes from midgestation fetuses and premature neonates generally produce less G-CSF and GM-CSF after stimulation in vitro than comparable adult cell types, whereas cells from term neonates produce amounts that are similar to or modestly less than those of adults.

Migration to Sites of Infection or Injury

After release from the bone marrow into the blood, neutrophils circulate until they are called upon to enter infected or injured tissues. Neutrophils adhere selectively to endothelium in such tissues but not in normal tissues. The adhesion and subsequent migration of neutrophils through blood vessels into tissues and to the site of infection result from a multistep process, which is governed by the pattern of expression on their surface of adhesion molecules and receptors for chemotactic factors and by the local patterns and gradients of adhesion molecule and chemotactic factors in the tissues.

The adhesion molecules involved in neutrophil migration from the blood into tissues include selectins, integrins, and the molecules to which they adhere. The selectins are named by the cell types in which they are primarily expressed: L-selectin by leukocytes, E-selectin by endothelial cells, and P-selectin by platelets and endothelial cells. L-selectin is constitutively expressed on leukocytes and appears to bind to tissue- or inflammation-specific carbohydrate-containing ligands on endothelial cells. E-selectin and P-selectin are expressed on activated not resting endothelial cells or platelets. E- and P-selectin bind to sialylated glycoproteins on the surface of leukocytes, including P-selectin glycoprotein ligand-1. L-selectin binds to glycoproteins and glycolipids, which are expressed on vascular endothelial cells in specific tissues. The integrins are a large family of heterodimeric proteins composed of an α and a β chain. The β 2 integrins LFA-1 (CD11a-CD18) and Mac-1 (CD11b-CD18) play a critical role in neutrophil function because neutrophils do not express other integrins in substantial amounts. The β 2 integrins are constitutively expressed on neutrophils, but their abundance and avidity for their endothelial ligands are increased after activation of neutrophils in response to chemotactic factors. Their endothelial ligands include intercellular adhesion molecule 1 (ICAM-1) and ICAM-2. Both are constitutively expressed on endothelium, but ICAM-1 expression is increased markedly by exposure to inflammatory mediators, including IL-1, TNF-α, and LPS.

Chemotactic factors may be derived directly from bacterial components, such as n -formylated-Met-Leu-Phe (fMLP) peptide; from activated complement, including C5a; and from host cell lipids, including leukotriene B 4 (LTB 4 ). In addition, a large family of chemotactic cytokines (chemokines) is synthesized by macrophages and many other cell types (see Tables 4-1 and 4-2 ). Chemokines constitute a cytokine superfamily, with more than 50 members known at present, most of which are secreted and of relatively low molecular weight. Chemokines attract various leukocyte populations, which bear the appropriate G protein–linked chemokine receptors. They can be divided into four families according to their pattern of amino-terminal cysteine residues: CC, CXC, C, and CX3C (X represents a noncysteine amino acid between the cysteines). A nomenclature for the chemokines and their receptors has been adopted, in which the family is first denoted (e.g., CC), followed by L for ligand (the chemokine itself) and a number, or followed by R (for receptor) and a number. Functionally, chemokines also can be defined by their principal function—homeostatic or inflammatory cell migration—and by the subsets of cells on which they act. Neutrophils are attracted by the subset of CXC chemokines that contain a glutamine-leucine-arginine motif, including the prototypical neutrophil chemokine CXLC8, also known as IL-8.

These adhesion molecules and chemotactic factors act in a coordinated fashion to allow neutrophil recruitment. In response to injury or inflammatory cytokines, E-selectin and P-selectin are expressed on the endothelium of capillaries or postcapillary venules. Neutrophils in the blood adhere to these selectins in a low-avidity fashion, allowing them to roll along the vessel walls. This step is transient and reversible unless a second, high-avidity interaction is triggered. If, at the time of the low-avidity binding, neutrophils also encounter chemotactic factors released from the tissues or from the endothelium itself, they rapidly upregulate the avidity and abundance of LFA-1 and Mac-1 on the neutrophil cell surface. This process results in high-avidity binding of neutrophils to endothelial cells, which, in the presence of a gradient of chemotactic factors from the tissue to the blood vessel, induces neutrophils to migrate across the endothelium and into the tissues by diapedesis.

The profound importance of integrin- and selectin-mediated leukocyte adhesion is illustrated by the genetic leukocyte adhesion deficiency (LAD) syndromes. In LAD 1, deficiency of the common β 2 integrin chain (CD18) results in inability of leukocytes to exit the bloodstream and reach sites of infection and injury in the tissues. Affected patients are profoundly susceptible to infections with pathogenic and nonpathogenic bacteria and may present in early infancy with delayed separation of the umbilical cord, omphalitis, and severe bacterial infection without pus formation. Two related syndromes, LAD 2 and LAD 3, are due, respectively, to a defect in synthesis (fucosylation) of the carbohydrate selectin ligands and mutations in the gene encoding kindlin-3, a protein required for integrin activation.

Migration of Neonatal Neutrophils

The ability of neonatal neutrophils to migrate from the blood into sites of infection and inflammation is reduced or delayed, and the transition from a neutrophilic to mononuclear cell inflammatory response is delayed. This diminished delivery of neutrophils may result in part from defects in adhesion and chemotaxis.

Adhesion of neonatal neutrophils under resting conditions is normal or at most modestly impaired, whereas adhesion of activated cells is deficient. Adhesion and rolling of neonatal neutrophils to activated endothelium under conditions of flow similar to those found in capillaries or postcapillary venules is variable but on average approximately 50% of that observed with adult neutrophils. This decreased adhesion appears to reflect, at least in part, decreased abundance and shedding of L-selectin and decreased binding of neonatal neutrophils to P-selectin. Resting neonatal and adult neutrophils have similar amounts of Mac-1 and LFA-1 on their plasma membrane, but neonatal neutrophils have a reduced ability to upregulate expression of these integrins after exposure to chemotactic agents. Reduced integrin upregulation is associated with a parallel decrease in adhesion to activated endothelium or ICAM-1 suggesting that a deficit in adhesion underlies the diminished ability of neonatal neutrophils to migrate through endothelium into tissues, particularly in preterm neonates.

In nearly all studies in which neutrophil migration has been examined in vitro, chemotaxis of neonatal neutrophils was less than that of adult neutrophils. The response of neonatal neutrophils to a variety of chemotactic factors, including fMLP, LTB 4 , and neutrophil-specific chemokines, including IL-8, is reduced. Chemotactic factor binding and dose response patterns of neonatal neutrophils appear similar to adult neutrophils, whereas downstream processes, including expression of Ras-related C3 botulinum toxin substrate 2 (Rac2), the increases in the free (nonprotein bound) intracellular calcium concentration [Ca 2+ ] i and inositol phospholipid generation and the change in cell membrane potential are impaired. An additional factor may be the reduced deformability of neonatal neutrophils, which may limit their ability to enter the tissues after binding to the vascular endothelium. Decreased generation of chemotactic factors in neonatal serum may compound the intrinsic chemotactic deficits of neonatal neutrophils. However, the generation of other chemotactic agents, such as LTB 4 , by neonatal neutrophils appears to be normal.

Phagocytosis

Having reached the site of infection, neutrophils must bind, phagocytose, and kill the pathogen. Opsonization greatly facilitates this process. Neutrophils express on their surface receptors for multiple opsonins, including receptors for the Fc portion of the IgG molecule (Fcγ receptors): FcγRI (CD64), FcγRIIA (CD32), and FcγRIIIB (CD16). Neutrophils also express receptors for activated complement components C3b and C3bi, which are bound by CR1, CR3 (CD11b-CD18), and CR4 (CD11c-CD18), respectively. Opsonized bacteria bind and cross link Fcγ and C3b-C3bi receptors. This cross-linkage transmits a signal for ingestion and for the activation of the cell’s microbicidal mechanisms.

Under optimal in vitro conditions, neutrophils from healthy neonates bind and ingest gram-positive and gram-negative bacteria as well as or only slightly less efficiently than an adult’s neutrophils. However, the concentrations of opsonins are reduced in serum from neonates, in particular, preterm neonates, and when concentrations of opsonins are limited, neutrophils from neonates ingest bacteria less efficiently than those from adults. Consistent with this finding, phagocytosis of bacteria by neutrophils from preterm, but not term, neonates is reduced compared with adult neutrophils when assayed in whole blood. Why neonatal neutrophils have impaired phagocytosis when concentrations of opsonins are limiting is incompletely understood. Basal expression of receptors for opsonized bacteria is not greatly different. Neutrophils from neonates, particularly preterm neonates, express greater amounts of the high-affinity FcγRI, lesser amounts of FcγRIII, and similar or slightly reduced amounts of FcγRIIA at birth, compared with adults; values in preterm neonates approach those of term neonates by 1 month of age. Expression of complement receptors on neutrophils from term neonates and adults is similar, but reduced expression of CR3 on neutrophils from preterm neonates has been reported in some studies. Neutrophils from preterm neonates also are less able to upregulate CR3 in response to LPS and chemotactic factors. Lower expression of proteins involved in the engulfment process, including Rac2 as noted above, may also contribute to impaired phagocytosis when concentrations of opsonins are limited.

Killing

After ingestion, neutrophils kill ingested microbes through oxygen-dependent and oxygen-independent mechanisms. Oxygen-dependent microbicidal mechanisms are of central importance, as illustrated by the severe compromise in defenses against a wide range of pyogenic pathogens (with the exception of catalase-negative bacteria) observed in individuals with a genetic defect in this system. Children with this disorder have a defect in one of several proteins that constitute the phagocyte oxidase, which is activated during receptor-mediated phagocytosis. The assembly of the oxidase in the plasma membrane results in the generation and delivery of reactive oxygen metabolites, including superoxide anion, hydrogen peroxide, and hydroxyl radicals. These oxygen radicals, along with the granule protein myeloperoxidase, are discharged into the phagocytic vacuole, where they collaborate in killing ingested microbes. In addition to this oxygen-dependent pathway, neutrophils contain other granule proteins with potent microbicidal activity, including the defensins HNPs 1 to 4, LL-37, elastase, cathepsin G, and a protein that binds selectively to and helps to kill gram-negative bacteria, bactericidal permeability-increasing (BPI) protein.

Oxygen-dependent and oxygen-independent microbicidal mechanisms of neutrophils from neonates and adults do not differ greatly. Generation of superoxide anion and hydrogen peroxide by neutrophils from term neonates is generally similar to or greater than by cells from adults in response to soluble stimuli. Although a modest reduction in the generation of reactive oxygen metabolites by neutrophils from preterm compared with term neonates was seen in response to some strains of coagulase-negative staphylococci, this was not observed with other strains nor with a strain of GBS and is of uncertain significance. By contrast, LPS primes adult neutrophils for increased production of reactive oxygen metabolites, but priming is much reduced with neonatal neutrophils, which could limit their efficacy in response to infection in vivo. The few studies of oxygen-independent microbicidal mechanisms suggest that neonatal neutrophils contain and release reduced amounts of BPI (approximately two to threefold) and lactoferrin (approximately twofold) compared with adult neutrophils but contain or release comparable amounts of myeloperoxidase, defensins, and lysozyme.

Consistent with these findings, killing of ingested gram-positive and gram-negative bacteria and Candida organisms by neutrophils from neonates and adults is generally similar. However, variable and usually mildly decreased bactericidal activity has been noted against Pseudomonas aeruginosa, S. aureus, and certain strains of GBS. Deficits in killing of engulfed microbes by neonatal neutrophils are more apparent at high ratios of bacteria to neutrophils, as is killing by neutrophils from sick or stressed neonates (i.e., those born prematurely or who have sepsis, respiratory impairment, hyperbilirubinemia, premature rupture of membranes, or hypoglycemia).

Neutrophils also function in trapping and killing extracellular bacteria in neutrophil extracellular traps (NETs), which consist of a complex of DNA, histones, granule enzymes, and antimicrobial proteins that are extruded from the cell. Neonatal neutrophils from both term and preterm infants are unable to form NETs and therefore have impaired extracellular bacterial killing in vitro.

Activation by Innate Immune Receptors

Although neutrophils were formally viewed by many as cells highly specialized for the uptake and killing of bacteria and fungi, human neutrophils also express a variety of innate immune receptors that are functional, including all TLRs except TLR3 and TLR7, NLRs (NOD2 and NLRP3), and RLRs (RIG-I and MDA-5). Moreover, neutrophils respond to TLR agonists or NOD2 ligands with production of IL-8 and other chemokines, superoxide generation, and shedding of L-selectin, and they also produce IL-1β after NLRP3 activation. In mice, neutrophil-derived IL-1β is sufficient for the formation of abscesses in S. aureus infection, with cytokine production involving both TLRs and NLRs. This suggests that neutrophils may be an important in vivo source of proinflammatory cytokines, at least in certain contexts. There is little information on neonatal neutrophil activation by innate immune receptors. One study of infants with severe RSV found that neutrophils of the blood and lung had reduced expression of TLR4. As TLR4 is a receptor for RSV G protein, it is plausible that this decreased expression could contribute to the disease severity.

Neutrophil Clearance and Resolution of Neutrophilic Inflammation

One to 2 days after egress from the bone marrow, neutrophils undergo apoptosis and are efficiently cleared by tissue macrophages without producing inflammation or injury. In the context of infection or sterile inflammation, their survival is prolonged by CSFs and other inflammatory mediators, allowing them to aid in microbial clearance, while at the same time augmenting or perpetuating tissue injury. Studies from several groups have shown that spontaneous and anti-Fas–induced apoptosis of isolated neonatal neutrophils is reduced when these cells are cultured in vitro. The greater survival of neonatal than adult neutrophils was associated with reduced expression of the apoptosis-inducing Fas receptor and pro-apoptotic members of the Bcl-2 family. The increased survival of neonatal neutrophils has led some to speculate that this may help to compensate for the neonate’s limited neutrophil storage pool in protection against infection, but this might also contribute to persistent untoward inflammation and tissue injury.

Effects of Immunomodulators

After systemic treatment with G-CSF and GM-CSF, the numbers of neutrophils increase in neonates, as does expression of CR3 on these cells. The increased numbers likely reflect increased production and survival. GM-CSF and IFN-γ enhance the chemotactic response of neonatal neutrophils, although, at high concentrations, GM-CSF inhibits chemotaxis while augmenting oxygen radical production. Of potential concern, indomethacin, which is used clinically to facilitate ductal closure in premature neonates, impairs chemotaxis of cells from term and preterm neonates.

Eosinophils

In adults and older children, eosinophils represent a small percentage of the circulating granulocytes. In the healthy fetus and neonate, eosinophils commonly represent a larger fraction (10%-20%) of total granulocytes than in adults. Numbers of eosinophils increase postnatally, peaking at the third to fourth week of postnatal life. A relative increase in the abundance of eosinophils in inflammatory exudates of various causes is also seen in neonates, paralleling their greater numbers in the circulation. Thus eosinophil-rich inflammatory exudates do not so strongly suggest the presence of allergic disease or helminthic infection, as they do in older individuals, and are not associated with increased amounts of circulating IgE. The degree of eosinophilia is greater yet in preterm neonates and in those with Rh disease, total parenteral nutrition, and transfusions. The basis for the eosinophilic tendency of the neonate is not known. By contrast to the diminished migration of neonatal neutrophils, neonatal eosinophils exhibit greater spontaneous and chemotactic factor–induced migration than adult eosinophils. Their greater numbers and ability to migrate may contribute to the relatively greater abundance of eosinophils in neonatal inflammatory infiltrates, including those seen in physiologic conditions such as erythema toxicum.

Mononuclear Phagocytes

Production and Differentiation of Monocytes and Resident Tissue Macrophages

Together, monocytes and tissue macrophages are referred to as mononuclear phagocytes. Under steady-state conditions, monocytes are released from the bone marrow within 24 hours and circulate in the blood for 1 to 3 days before moving to the tissues, where they differentiate into tissue macrophages. All monocytes express CD14, which serves as a co-receptor for recognition of LPS by TLR4/MD-2. CD14 is commonly used as a lineage marker for these cells because it is the only cell type that expresses it in high amounts. Additional commonly used mononuclear phagocyte markers include CD68, a glycoprotein that binds to low density lipoprotein, and CD163, a scavenger receptor for hemoglobin-haptoglobin complexes. Monocytes also express histocompatibility leukocyte antigen (HLA)-DR (MHC class II) and can present antigens to CD4 T cells, which is discussed in detail in the section “Antigen Presentation by Classical Major Histocompatibility Complex Molecules,” although the amounts expressed and efficiency of antigen presentation are less than by DCs. Monocytes are heterogeneous. In the healthy adult circulation, CD14 + monocytes can be divided into three subsets based on their levels of expression of CD14 and CD16 (a Fc receptor for IgG)—CD14 ++ CD16 , CD14 + CD16 + , and CD14 dim CD16 ++ , which comprise approximately 85%, 6%, and 9% of total blood monocytes, respectively. CD14 ++ CD16 monocytes have relatively low levels of surface expression of CD11c, CD80/86, CD163, and HLA-DR compared with CD14 + CD16 + monocytes. Both CD11c, a β 2 integrin molecule, and L-selectin (CD62-L), are involved in leukocyte adhesion, and CD80 and CD86 are important for co-stimulation to T cells. The CD14 dim CD16 ++ cell subset, which was only recently described, is highly responsive to viral nucleic acids but not bacterial products, such as LPS, although this point is controversial. Studies in humanized mice suggest that CD14 dim CD16 ++ monocytes “patrol” the endothelium in an L-selectin–dependent manner.

Macrophages are resident in tissues throughout the body and include distinct populations of alveolar macrophages (lung), histiocytes (interstitial connective tissue), Kupffer cells (liver), microglia (brain), and osteoclasts (bone). Macrophages have multiple functions, including the clearance of dead host cells (efferocytosis), phagocytosis and killing of microbes, secretion of inflammatory mediators, and presentation of antigen to T cells. They can also act to limit or suppress inflammation in certain contexts and promote tissue repair. With the exception of inflammatory macrophages, which are similar to neutrophils in being short-lived, most macrophage populations are relatively long-lived cells, but their life span may vary from days to possibly years, depending on the particular macrophage population.

Macrophages are detectable as early as 4 weeks of fetal life in the yolk sac and are found shortly thereafter in the liver and then in the bone marrow. Recent studies in mice indicate that the major tissue subsets, including liver Kupffer cells and lung alveolar, splenic, and peritoneal macrophages, are established before birth (21 days of gestation) and are subsequently maintained by proliferation in situ rather than by replenishment from circulating monocytes. These findings, along with others documenting tissue macrophage proliferation in the mouse in diverse inflammatory responses, for instance, tissue nematode infection and atherosclerosis, suggest that tissue macrophage proliferation could be an important part of human mononuclear cell homeostasis and regulation by inflammation.

The capacity of the fetus and the neonate to produce monocytes is at least as great as that of adults. The numbers of monocytes per volume of blood in neonates are equal to or greater than those in adults. Cord blood has a similar fraction of monocytes that are CD14 ++ CD16 and CD14 + CD16 + as adult peripheral blood. Cord blood monocytes express approximately 50% as much HLA-DR as adult monocytes, and a larger fraction of neonatal monocytes lack detectable HLA-DR ; this reduced expression in cord blood applies to both the CD14 ++ CD16 and CD14 + CD16 + subsets. In contrast, other surface markers are expressed similarly, with the CD14 + CD16 + subset of both cord blood and adult peripheral blood having significantly higher levels of CD11c, CD80/86, and CD163 than CD14 ++ CD16 monocytes. To the best of our knowledge, there have been no studies reporting the frequency and phenotype of the recently described CD14 dim CD16 ++ monocyte subset in human neonates.

The numbers of tissue macrophages in human neonates are not well characterized. Limited data in humans, which are consistent with data in nonhuman primates and other mammals, suggest that the lung contains few macrophages until shortly before term. Postnatally, the numbers of lung macrophages increase to adult levels by 24 to 48 hours in healthy monkeys. A similar increase occurs in humans, although the data are less complete and by necessity derived from individuals with clinical problems necessitating tracheobronchial lavage. The blood of premature neonates contains increased numbers of pitted erythrocytes or erythrocytes containing Howell-Jolly bodies, suggesting that the ability of splenic and liver macrophages to clear these effete cells, and perhaps microbial cells, may be reduced in the fetus and premature infant.

Migration to Sites of Infection and Delayed Hypersensitivity Responses

Like neutrophils, mononuclear phagocytes express the adhesion molecules L-selectin and β 2 integrins. These cells also express substantial amounts of the α 4 β 1 integrin very late antigen-4 (VLA-4), allowing them, unlike neutrophils, to adhere efficiently to endothelium expressing vascular cell adhesion molecule 1 (VCAM-1), the ligand for VLA-4. Interaction of VLA-4 with VCAM-1 allows monocytes to enter tissues in states in which there is little or no neutrophilic inflammation. Chemokines that are chemotactic for neutrophils are not generally chemotactic for monocytes, and vice versa. Monocytes respond to a range of CC chemokines, such as CCL2 (monocyte chemotactic protein-1 [MCP-1]).

The acute inflammatory response is characterized by an initial infiltration of neutrophils that is followed within 6 to 12 hours by influx of mononuclear phagocytes. Some inflammatory responses, including delayed-type hypersensitivity (DTH) reactions induced by the injection of antigens, for instance, purified protein derivative (PPD), to which the individual is immune (i.e., has developed an antigen-specific T-cell response) are characterized by the influx of mononuclear phagocytes and lymphocytes with very minimal or no initial neutrophilic phase.

The influx of monocytes into sites of inflammation, including DTH responses, is delayed and attenuated in neonates compared with adults. This is true even when antigen-specific T-cell responses are evident in vitro, suggesting that decreased migration of monocytes and lymphocytes into the tissues is predominantly responsible for the poor response in neonates. Whether this delay results from impaired chemotaxis of neonatal monocytes or impaired generation of chemotactic factors or both is unresolved

Antimicrobial Properties of Monocytes and Macrophages

Although neutrophils ingest and kill pyogenic bacteria more efficiently, resident macrophages are the initial line of phagocyte defense against microbial invasion in the tissues. When the microbial insult is modest, these cells are sufficient. If not, they produce cytokines and other inflammatory mediators to direct the recruitment of circulating neutrophils and monocytes from the blood. Monocytes and macrophages express receptors that allow them to bind to microbes. These receptors include FcγRI, II, and III, which bind IgG-coated microbes ; FcαR, which binds IgA-coated microbes ; and the CR1 and CR3 receptors, which bind microbes coated with C3b and C3bi, respectively. Microbes bound through these receptors are efficiently engulfed by macrophages and once ingested can be killed by microbicidal mechanisms using many methods similar to those used by neutrophils. Mononuclear phagocytes generate reactive oxygen metabolites but in lesser amounts than neutrophils. Circulating monocytes, but not tissue macrophages, contain myeloperoxidase, which facilitates the microbicidal activity of hydrogen peroxide. The expression of microbicidal granule proteins differs somewhat in mononuclear phagocytes and neutrophils; for instance, human mononuclear phagocytes express β-defensins but not α-defensins, whereas neutrophils express both.

The microbicidal activity of resident tissue macrophages is relatively modest, especially compared with the robust activity of neutrophils. This limited activity may be important in allowing macrophages to remove dead or damaged host cells and small numbers of microbes without excessively damaging host tissues. However, in response to infection, macrophage microbicidal and proinflammatory functions are enhanced in a process referred to as classical (M1) macrophage activation. Macrophage activation results from the integration of signals from TLRs and other innate immune pattern recognition receptors and receptors for activated complement components, immune complexes, cytokines, and ligands produced by other immune cells, including IFN-γ, CD40 ligand, TNF-α, and GM-CSF.

The increased antimicrobial activity of M1-activated macrophages results in part from increased expression of FcγRI, enhanced phagocytic activity, and increased production of reactive oxygen metabolites. Other antimicrobial mechanisms induced by activation of these cells include the catabolism of tryptophan by indoleamine 2,3-dioxygenase, scavenging of iron, and production of nitric oxide and its metabolites by the inducible nitric oxide synthase (iNOS). The last is a major mechanism by which activated murine macrophages inhibit or kill a variety of intracellular pathogens. However, the role of nitric oxide in the antimicrobial activity of human macrophages is controversial, although polymorphisms of the iNOS gene locus in humans have been associated with the host susceptibility to Mycobacteria tuberculosis. Activated mononuclear phagocytes also secrete a number of noncytokine products that are potentially important in host defense mechanisms. These include complement components, fibronectin, and lysozyme.

Classical (M1) activation of macrophages plays a critical role in defense against infection with intracellular bacterial and protozoan pathogens that replicate within phagocytic vacuoles. Support for this notion comes from studies in humans and mice with genetic deficiencies that impair the activation of macrophages by IFN-γ. Humans with genetic defects involving IL-12, which induces IFN-γ production by NK and T cells, the IL-12 receptor, the IFN-γ receptor or the transcription factor STAT1, which is activated via this receptor, suffer unduly from infections with mycobacteria and Salmonella. Treatment of humans with antagonists of TNF-α also impair antimycobacterial defenses. Patients with the hyper-IgM syndrome, which is due to a defect in CD40 ligand, or with autosomal recessive hyper-IgM due to a mutation in CD40, the receptor for CD40 ligand, are predisposed to disease caused by Pneumocystis jirovecii and Cryptosporidium parvum, in addition to the problems they experience from defects in antibody production (see section CD4 T-Cell Help for Antibody Production ). These findings are consistent with the notion that IFN-γ, TNF-α, and CD40 ligand–mediated M1 macrophage activation is important in host defense against these pathogens and that these molecules activate macrophages, at least in part, in a nonredundant manner.

By contrast to this canonical pathway of macrophage activation, macrophages exposed to cytokines produced by Th2 cells, such as IL-4, which are induced by infection with parasitic helminths and as part of allergic responses, are activated in an alternative manner. These alternatively activated macrophages, also termed M2 cells, dampen acute inflammation, impede the generation of reactive nitrogen products, limit proinflammatory T-cell responses, and foster fibrosis through the production of arginase and other mediators. Although best characterized in mice, this alternative pathway appears to be relevant in humans as well, and a recent study suggests that there is substantial additional diversity in the function of human macrophage subsets beyond the M1 and M2 paradigms.

Antimicrobial Activity and Activation of Neonatal Monocytes and Macrophages

Monocytes from human neonates and adults ingest and kill S. aureus, E. coli, and GBS with similar efficiency. Consistent with these findings, the production of microbicidal oxygen metabolites by neonatal and adult monocytes is similar. Neonatal and adult monocytes, monocyte-derived macrophages, and fetal macrophages are comparable in their ability to prevent herpes simplex virus (HSV) from replicating within them. And although neonatal monocytes may be slightly less capable of killing HSV-infected cells than adult monocytes in the absence of antibody, they are equivalent in the presence of antibody.

The ability of neonatal and adult monocyte-derived macrophages (monocytes cultured in vitro) to phagocytose GBS, other bacteria, and Candida through receptors for mannose-fucose, IgG, and complement components is similar. Despite comparable phagocytosis, neonatal monocyte-derived macrophages kill Candida and GBS less efficiently. Moreover, GM-CSF, but not IFN-γ, activates neonatal monocyte-derived macrophages to produce superoxide anion and to kill these organisms, whereas both of these cytokines activate adult macrophages. The lack of response to IFN-γ by neonatal macrophages was associated with normal binding to its receptor but decreased activation of STAT1. Studies with alveolar macrophages from newborn and, particularly, premature newborn monkeys and other mammals also have shown reduced phagocytic and/or microbicidal activity, suggesting this might apply to humans. By contrast to these reports of decreased antimicrobial activity and failure of macrophage activation by IFN-γ, blood monocytes and IFN-γ–treated monocyte-derived and placental macrophages from neonates kill and restrict the growth of Toxoplasma as effectively as cells from adults.

Mononuclear Phagocytes Produce Cytokines and Other Mediators That Regulate Inflammation and Immunity

Monocytes and macrophages produce cytokines, chemokines, colony-stimulating factors and other mediators in response to ligand binding by TLRs and other pattern recognition receptors expressed by these cells (described in the next section), cytokines produced by other cell types, activated complement components and other mediators, and engagement of CD40 on their surface by CD40 ligand expressed on activated CD4 T cells. These include the cytokines IL-1α, IL-1β, TNF-α, and IL-6, which induce the production of prostaglandin E2, which, in turn, induces fever, accounting for the antipyretic effect of drugs that inhibit prostaglandin synthesis. Fever may have a beneficial role in host resistance to infection by inhibiting the growth of certain microorganisms and by enhancing host immune responses. TNF-α, IL-1, and IL-6 also act on the liver to induce the acute-phase response, which is associated with decreased albumin synthesis and increased synthesis of certain complement components, fibrinogen, CRP, and MBL. G-CSF, GM-CSF, and macrophage-specific colony-stimulating factor (M-CSF) enhance the production of their respective target cell populations, increasing the numbers of phagocytes available. At the sites of infection or injury, TNF-α and IL-1 increase endothelial cell expression of adhesion molecules, including E-selectin and P-selectin, ICAM-1, and VCAM-1, increase endothelial cell procoagulant activity and enhance neutrophil adhesiveness by upregulating β 2 integrin expression. IL-6 may help to terminate neutrophil recruitment into tissues and to facilitate a switch from an inflammatory infiltrate rich in neutrophils to one dominated by monocytes and lymphocytes. IL-8 and other Glu-Leu-Arg (ELR)-containing CXC chemokines enhance the avidity of neutrophil β 2 integrins for ICAM-1 and attract neutrophils into the inflammatory-infectious focus; CC chemokines play a similar role in attracting mononuclear phagocytes and lymphocytes. These and additional factors contribute to edema, redness, and leukocyte infiltration, which characterize inflammation.

In addition to secreting cytokines that regulate the acute inflammatory response and play a crucial role in host defense to extracellular bacterial and fungal pathogens, monocytes and macrophages (and DCs; see later) produce cytokines that mediate and regulate defense against intracellular viral, bacterial, and protozoan pathogens. Type I IFNs directly inhibit viral replication in host cells, as do IFN-γ and TNF-α. IL-12, IL-23, and IL-27 are members of a family of heterodimeric cytokines that help to regulate T-cell and NK-cell differentiation and function. IL-12 is composed of IL-12/23 p40 and p35; IL-23 is composed of IL-12/23 p40 and p19; and IL-27 is composed of Epstein-Barr virus–induced gene 3 protein (EBI-3) and p28. IL-12, in concert with IL-15 and IL-18, enhance NK-cell lytic function and production of IFN-γ and facilitate the development of type 1 CD4 T helper (Th1) and CD8 effector T cells, which are discussed more fully in the section “Differentiation of Activated Naïve T Cells into Effector and Memory Cells” and which play a critical role in control of infection with intracellular bacterial, protozoal, and viral pathogens. IFN-γ activates macrophages, allowing them to control infection with intracellular pathogens, and enhances their capacity to produce IL-12 and TNF-α, which, in turn, amplifies IFN-γ production by NK cells and causes T cells to differentiate into IFN-γ–producing Th1 T cells. IL-27 also facilitates IFN-γ production, while at the same time inducing the expression of IL-10, which dampens inflammatory and Th1 T-cell responses to limit tissue injury. By contrast, IL-23 favors IL-17–producing Th17 T-cell responses, in which IL-17 promotes neutrophil production, acute inflammation, and defense to extracellular pathogens.

The production of cytokines by mononuclear phagocytes normally is restricted temporally and anatomically to cells in contact with microbial products, antigen-stimulated T cells, or other agonists. When excess production of proinflammatory cytokines occurs systemically, septic shock and disseminated intravascular coagulation may ensue, underscoring the importance of closely regulated and anatomically restricted production of proinflammatory mediators. Tight control of inflammation normally is achieved by a combination of positive and negative feedback regulation. For example, TNF-α, IL-1, and microbial products that induce their production also cause macrophages to produce cytokines that attenuate inflammation and dampen immunity, including IL-10 and the IL-1 receptor antagonist. Inflammation is also attenuated by the production of antiinflammatory lipid mediators (resolvins), including the lipoxins, protectins, and maresins.

Cytokine Production, Toll-like Receptors, and Regulation of Innate Immunity and Inflammation by Neonatal Monocytes and Macrophages

Much of the older literature suggested that blood mononuclear cells (BMCs) obtained from cord blood or neonatal peripheral blood were less efficient in general than adult BMC in the production of cytokines in response to LPS, other TLR ligands, or whole bacteria. BMCs consist of monocytes, DCs, and B, T, and NK lymphocytes. Because of the greater than 10-fold abundance of monocytes compared with DCs in blood and BMCs, and the relatively limited production by lymphocytes of cytokines in response to TLR agonists, monocytes were likely to be the predominant source for most of these cytokines. The recent development of routine flow cytometric analysis of 6- to 12-color monoclonal antibody staining for intracellular cytokines and surface markers applied to BMC now permits an assessment of the levels of production of multiple cytokines per cell for each cell type. Also, because of simplicity and a desire to minimize manipulations that might activate or alter blood leukocyte function, many recent studies have been done with whole blood to which TLR ligands are added directly ex vivo, with cytokine production evaluated by enzyme-linked immunosorbent assay (ELISA) or bead-based fluorescent assays or, in some cases, by multiparameter flow cytometry. The findings for recent flow cytometric studies using whole blood or BMC will be emphasized here, although secreted cytokine levels will also be included where the likely cell source can be inferred. As TLR3 and TLR9 are expressed by certain DC population but not monocytes, and monocytes, in general, produce type I IFN poorly, the results for type I IFN production and other cytokines in response to TLR3 or TLR9 ligands in whole-blood assays will be discussed separately in the section “Dendritic Cells: The Link Between Innate and Adaptive Immunity.”

The preponderance of the currently available data does not support the notion of a general inability of neonatal monocytes (and, as discussed later, DCs) to produce cytokines, but rather suggests a difference in the nature of their response.

There is a clear, substantial, and with rare exception, consistent deficit in the production of IL-12p70 and IFN-γ in response to TLR agonists that act on monocytes (TLR2, TLR4, TLR7/8). In assays using whole blood or BMC, the IFN-γ produced is probably mainly derived from NK cells responding to IL-12p70 rather than from T cells. These deficits of cord blood leukocytes are likely cell intrinsic because they are consistently observed with either whole-blood assays or assays of BMC cultured with heterologous adult serum or with fetal calf serum. As discussed in the section “Dendritic Cells: The Link Between Innate and Adaptive Immunity,” there are also marked reductions in type I IFN production by DCs in cord blood. Because type I IFNs synergize with IL-12p70 and IFN-γ in promoting Th1 development from naïve CD4 T cells, this cytokine profile combined with T-cell intrinsic mechanisms, which are discussed later, would be expected to limit the generation of Th1 effectors in neonates, as has been observed in neonatal HSV infection. As LPS-induced IL-12p70 production by monocytes is itself markedly increased by IFN-γ, it is noteworthy that IL-12p70 secretion and IL-12p35 mRNA expression in cord blood after combined LPS and IFN-γ stimulation is decreased, and this decrease persisted until at least 1 month of age. Whole-blood assays indicate that adult levels of production of IL-12p70 in response to LPS are achieved by 6 months of age. A large body of evidence, including from human genetic immunodeficiencies, has clearly demonstrated that IL-12p70 and IFN-γ are essential for Th1-type immune control of pathogens, especially mycobacteria. Thus it is plausible that these antigen-presenting cell (APC) limitations in Th1 cytokine production are an important mechanism contributing to the well-described vulnerability of the neonate and young infant to Mycobacterium tuberculosis infection.

A similar deficit in TNF-α production is evident in response to stimulation with TLR2, TLR3, TLR4, and TLR5 agonists, particularly when whole blood is used or BMC or monocytes are cultured in high (≥50%) concentrations of neonatal serum. In contrast, TNF-α production in response to TLR8 agonists or whole gram-positive or gram-negative bacteria is similar. Further, in flow cytometric analysis of BMC cultured in adult serum, production of TNF-α by cord blood and adult peripheral blood monocytes was similar. As discussed later, these findings are explained at least in part by the presence of an inhibitory factor in cord blood plasma and suggest that any cell-intrinsic limitations in TNF-α production by cord blood monocytes may be relatively subtle.

In any case, with the exception of TNF-α, production by neonatal cells of cytokines central to host defense against extracellular bacterial and fungal pathogens, acute inflammation, and Th17-type responses appear not to be greatly decreased and, in some cases, is more robust. IL-1 production by cells from term neonates and adults is similar or, at most, marginally reduced. In response to TLR agonists, neonatal monocytes produced equal or greater amounts of IL-6, IL-23, and IL-10, compared with adult cells, but were less able to produce multiple cytokines simultaneously. In one recent longitudinal study of the ontogeny of TLR-mediated cytokine responses in South African infants, this high level of production of IL-6 and IL-23, which was particularly robust in response to TLR2 agonists, gradually declined in the first year of life to adult peripheral blood levels. As IL-1, IL-6, and IL-23 production by APCs instructs the differentiation of naïve CD4 T cells into Th17-type effector cells, these findings are in agreement with apparently intact Th17 immunity in the neonate. Although some early studies suggested that the production of the immunoregulatory and antiinflammatory cytokine IL-10 by neonatal cells was reduced, most studies have found greater production of IL-10 by whole blood and equal or greater production by BMCs. These high levels of IL-10 in whole-blood assays, which are particularly prominent after TLR2 stimulation, gradually decline to adult levels in the first year of life, indicating that they are not merely a feature of cord blood leukocytes.

The basis for lower production of certain cytokines by neonatal monocytes and macrophages in response to microbial products that signal through TLRs is incompletely understood. Cell surface expression of TLR2, TLR4, and TLR8 by adult and neonatal monocytes is similar in most studies, and expression of CD14, which facilitates responses to LPS and TLR2 agonists, is similar or, at most, slightly reduced on neonatal monocytes. Monocytes from premature infants express less TLR2 and TLR4 at birth but increase their expression to amounts comparable to term infants by 2 weeks of age. By reverse-transcriptase polymerase chain reaction (RT-PCR) analysis, neonatal and adult monocytes contain similar amounts of TLRs 1 to 9, MD-2, CD14, MyD88, TIR domain-containing adaptor protein (TIRAP), and interleukin-1 receptor-associated kinase 4 (IRAK4) mRNA. One group reported that neonatal monocytes have reduced amounts of MyD88 protein but found no difference between adult and neonatal monocytes in LPS-induced activation of ERK1/2 and p38 kinases and phosphorylation and degradation of inhibitor of kappa B (IκB), events that are downstream of MyD88. Moreover, decreased expression of MyD88 is not sufficient to explain the diminished induction of HLA-DR and CD40 on neonatal monocytes in response to LPS, or of CD40 and CD80 on neonatal dendritic cells in response to LPS and polyinosinic:polycytidylic (poly [I:C]) acid, because these TLR ligands induce co-stimulatory molecules by TRIF- and type I IFN–dependent but MyD88-independent pathways.

Humoral Influences on Mononuclear Phagocyte Cytokine Production

Levy and colleagues identified adenosine as a factor in cord blood plasma that inhibited TNF-α production but preserved IL-6 production by monocytes in response to TLR2, TLR4, and TLR7 ligands. Compared with adult monocytes, cord blood monocytes also had a greater sensitivity to adenosine-induced (most likely A3 receptor–mediated) accumulation of cyclic adenosine monophosphate (cAMP). Adenosine was modestly elevated in cord blood compared with adult peripheral blood plasma, and the elevated cAMP had an inhibitory effect on TLR2- or TLR4-induced TNF-α but not on IL-6 production, and it was associated with decreased p38 kinase activation. In contrast, with TLR8 stimulation, cord blood plasma did not reduce the levels of p38 activation by monocytes or diminish the amounts of TNF-α, IL-1β, and multiple other cytokines produced. The increased amounts of adenosine may be related, in part, to hypoxia occurring during the birth process, to the relatively low amounts of adenosine deaminase (which converts adenosine to inosine) in cord blood, and to increased adenosine generation by ectoenzymes that convert ATP/adenosine diphosphate (ADP) to adenosine. In addition to adenosine, estrogen and progesterone are also at high levels in the fetus during pregnancy and in cord blood. These hormones may contribute to the suppressive effect of cord blood on TLR-induced monocyte proinflammatory cytokine production by inhibiting TLR-dependent activation of NF-κB activity, and, as in the case of adenosine, cord blood monocytes may have greater sensitivity to the inhibitory effects of these hormones than adult cells.

It is plausible that immunosuppressive effect of humoral factors, such as adenosine, may be a transient phenomenon alleviated shortly after birth. This possibility is supported by studies that found that cord blood plasma, but not plasma from 1-month-old infants, suppressed IL-12p70 production and increased IL-10 production by adult peripheral BMCs treated with LPS and IFN-γ. Moreover, a study of rhesus macaques evaluating TLR-induced cytokine production using whole-blood assays found that, similar to humans, TNF-α and IL-1β production by cord blood cells in response to TLR2/6 agonist stimulation was markedly reduced compared with the amounts produced by adult peripheral blood cells, but cytokine production from blood at 1 week of age was similar to (TNF-α) or exceeded that of adult blood (IL-1β); whether this normalization by 1 week after birth reflects decreases in adenosine levels or monocyte sensitivity to adenosine or some other mechanism remains unclear.

Compared with blood from healthy adults, neonatal cord blood contains lower amounts of soluble CD14 and similar or modestly reduced amounts of soluble LPS-binding protein (LBP); concentrations of CD14 and LBP rise to adult levels in the first week of life and rise further in response to infection, as they do in adults. The reduced amounts of these two proteins may account for the earlier observation that neonatal cord blood contains lower amounts of a soluble protein(s) that facilitates the response of monocytes to LPS. However, the addition of soluble CD14 to neonatal plasma did not restore TNF-α production by neonatal monocytes.

Resolution of Mononuclear Phagocytic Inflammation

The resolution of mononuclear phagocytic inflammation involves the secretion of antiinflammatory cytokines, such as IL-10, IL-1 receptor antagonist, and resolvin lipid mediators. Phagocytosis-induced cell death (PICD) after ingestion of bacteria may also potentially be beneficial or harmful to the host, depending on whether cell death results in the death of the ingested microbe or its release into the extracellular space. As was observed for neonatal neutrophils, cord blood monocytes had reduced PICD compared with adult peripheral blood monocytes, which could contribute to persistent inflammation and sequelae, such as bronchopulmonary dysplasia and periventricular leukomalacia, in septic neonates.

Summary

The most critical deficiency in phagocyte defenses in the term neonate and particularly in the preterm neonate is the limited ability to accelerate neutrophil production in response to infection. This age-specific limitation appears to result in large part from a limited neutrophil storage pool and perhaps a more limited ability to increase neutrophil production in response to infection. Impaired migration of neutrophils into tissues is likely to also be a factor, whereas phagocytosis and intracellular killing do not appear to be greatly impaired. However, the inability of neonatal neutrophils to form extracellular traps and the subsequent defect in extracellular bacterial killing may contribute to the increased susceptibility of neonates to bacterial infection. Persistent inflammation and tissue injury may result from impaired clearance of infection and protracted neutrophilic inflammation once the infection is cleared.

Monocytes and macrophages are detected in early fetal life and are present in blood and tissues by late gestation in numbers similar to adults. An exception is lung alveolar macrophages, which are few in number before birth, increase rapidly after birth in term neonates, but may be delayed in preterm neonates. Recruitment of monocytes to sites of infection and inflammation is slower than in adults. Ingestion and killing of pathogens by neonatal monocytes is as competent as in adults, but neonatal macrophages may be less efficient and be activated less efficiently by IFN-γ. Whereas expression by neonatal and adult monocytes of TLRs and other innate immune receptors appears not to differ greatly, their responses to stimulation via these receptors differ. In response to most but not all microbial stimuli, neonatal blood mononuclear cells produce (1) substantially lower amounts of IL-12 and type I IFNs, which are cytokines produced primarily by DCs and important for defense against intracellular pathogens; (2) moderately less TNF-α; (3) similar or somewhat greater amounts of other proinflammatory cytokines and IL-23, which are cytokines that are important in defense against extracellular bacterial and fungal pathogens; and (4) similar or greater amounts of the antiinflammatory and immunoregulatory cytokine IL-10.

Dendritic Cells: the Link between Innate and Adaptive Immunity

Overview

Dendritic cells, which have aptly been referred to as sentinels of the immune system, derive their name from the characteristic cytoplasmic protrusions or “dendrites” found on their mature form. DCs are bone marrow–derived myeloid cells that are found in all tissues and also circulate in the blood, where they represent approximately 0.5% to 1% of circulating leukocytes. Some DCs that are newly produced by the bone marrow enter into the blood and exit directly into lymphoid tissues, where they permanently reside as lymphoid tissue (LT) DCs. Other newly produced circulating DCs exit into the nonlymphoid tissues and are known as migratory DCs, which later migrate via afferent lymphatics into lymph nodes, particularly after exposure to inflammatory stimuli.

Human DCs can be divided into two major groups of cells that express high levels of the CD11c/CD18 β 2 integrin protein, hereafter referred to as conventional DCs (cDCs) and plasmacytoid cells (pDCs), which are CD11c and express other distinct markers, such as CD123 (a component of the IL-3 receptor). Langerhans cells, which are also CD11c + , are a migratory DC of squamous epithelium that has a distinct developmental origin from other DCs and will be discussed separately, as will inflammatory DCs, which are derived from mature monocytes rather than immature bone marrow precursors.

The DC cell surface lacks molecules characteristic of other bone marrow–derived cell lineages (a feature that is termed Lin ), including molecules that are typically expressed on T cells (e.g., CD3-ε), monocytes or neutrophils (e.g., CD14), B cells (e.g., CD19 or CD20), and natural killer (NK) cells (e.g., CD16 or CD56). Resting DCs express MHC class II, and, upon activation/maturation, express greater amounts than any other cell type in the body. Relatively high amounts of MHC class I are also expressed. The role of MHC molecules in the presentation of antigenic peptide to T cells is discussed later in the section “Antigen Presentation by Classic Major Histocompatibility Complex Molecules.”

cDC and pDC development in the bone marrow requires the expression by DC precursors of Flt3, a cytokine receptor, and its binding to the Flt3 ligand, which is mainly produced by nonhematopoietic stromal cells. Murine studies suggest that the DC and monocyte lineages are derived from a common bone marrow cell precursor, the monocyte and DC progenitor (MDP), which can differentiate into either monocytes or committed DC progenitors (CDPs). The CDP gives rise to pre-cDCs, which enter the blood and then are presumed to rapidly enter into lymphoid or nonlymphoid organs, where, respectively, they differentiate in situ into immature lymphoid tissue cDCs or migratory cDCs. In the mouse, this final differentiation step includes the acquisition of their final DC subset surface phenotype, the characteristic cytoplasmic protrusions, and probing behavior. pDCs leaving the bone marrow appear to be immature functionally but otherwise fully differentiated. Unlike cDCs, pDCs only acquire cytoplasmic protrusions and high levels of MHC class II after they undergo terminal maturation by exposure to pathogen-derived products or viral infection.

Conventional Dendritic Cells

Conventional dendritic cells (cDCs) play a unique and essential role in the initiation and modulation of the adaptive immune response as well as the maintenance of tolerance to self-proteins. They integrate signals from receptors that recognize microbial-derived PAMPs or DAMPs; the latter are of host origin and include molecules that are induced by inflammation, danger, or cellular stress, which are frequently induced in the setting of infection. In the absence of these warning signs of infection, there is a constant low-level turnover of both migratory and lymphoid tissue cDCs. By mechanisms that are poorly understood, some migratory cDCs in uninflamed tissues undergo a maturation process and travel via the afferent lymphatics to lymph nodes, where they interact with T cells in the T-cell–rich extrafollicular areas. These migratory cDCs play a central role in maintaining a state of T-cell tolerance to self-peptides derived from tissue proteins by presenting them to T cells in the absence of co-stimulatory signals required for T-cell activation. This results in self-antigen–specific T cells that either die by apoptosis or that become anergic or regulatory cells, which are suppressive rather than immunostimulatory. Resident lymphoid tissue DCs may also participate in tolerance induction for self-peptides of proteins expressed within lymphoid tissues in steady-state conditions.

Alternatively, in cases of infection within the tissues, immature migratory cDCs take up microbes and microbial antigens by micropinocytosis and macropinocytosis of extracellular fluid and particulate debris from perturbed tissues and process proteins into peptides, which are loaded onto MHC class I and class II molecules. At the same time, cDCs start to express on their surface the CCR7 chemokine receptor and to lose expression of receptors for chemokines that target them to nonlymphoid tissues. This change in chemokine receptor expression enhances cDC migration via lymphatics to T-cell–rich areas of the draining lymph nodes, which constitutively express chemokines that bind to CCR7 (CCL19 and CCL21). Concomitant with their migration to the draining lymph nodes, cDCs mature and cease their pinocytotic uptake and display on their cell surface high amounts of MHC molecules enriched for antigenic peptides derived from previously internalized microbes and antigens. These peptide-MHC complexes are present on the surface of mature DCs in great abundance, as are the co-stimulatory molecules CD40, CD80 (B7-1), and CD86 (B7-2), which together allow these cells to present antigens to T cells in a highly effective manner (see Antigen Presentation by Classic Major Histocompatibility Complex Molecules ). A similar maturation process most likely occurs in situ with lymphoid tissue cDCs in response to lymphoid tissue inflammation. Because activated DCs display very high levels of peptide/MHC complexes and co-stimulatory ligands, they are the most efficient APCs for initiating the T-cell immune response to neoantigens, that is, antigens that have not been previously encountered by the host. However, cDCs are also important for maximizing the memory T-cell response to bacterial and viral pathogens.

cDCs not only play a critical role in T-cell activation, they influence the quality of the T-cell response that ensues through the production of cytokines. Naïve CD4 T cells may become Th1, Th2, Th9, Th17, Th22, or T-follicular helper (TFH) effector cells, each with a distinct cytokine-secretion profile and role in host defense. The secretion by DCs of particular cytokines instructs CD4 T-cell effector differentiation. For example, IL-12, IL-27, and type I IFNs instruct naïve CD4 T cells to produce IFN-γ and to differentiate into Th1 cells, which help to protect against intracellular bacteria, by multiple effects on mononuclear phagocytes, as well as certain viruses. IL-1, IL-6, TGF-β, and IL-23 induce differentiation to Th17 cells, which secrete IL-17A and IL-17F and help to protect against extracellular bacteria and fungi by increasing neutrophil production and activity and the expression in mucosal tissues of antimicrobial peptides and proteins. Th2 cell development is instructed by IL-33, thymic stromal lymphopoietin (TSLP), and IL-4 (from a non–T-cell source, such as basophils or innate lymphoid cells), and Th2 cells produce IL-4, IL-5, and IL-13 to protect against infections with multicellular pathogens, such as worms, but also are important in the pathogenesis of allergic disorders.

cDC migration and maturation can be triggered by a variety of stimuli, including pathogen-derived products that are recognized directly by innate immune receptors, by cytokines, including IL-1, TNF-α, and type I IFNs (see Tables 4-1 and 4-2 ), and by engagement of CD40 on the DC surface by CD40 ligand (CD154) on the surface of activated CD4 T cells ( Table 4-5 ). Given their role as early detectors of infection or perturbations that are often associated with infection, it is not surprising that cDCs express a great diversity of receptors for PAMPs, including TLRs, NLRs, CLRs, and RLRs, Human cDCs express multiple TLRs but do not express TLR9 (see Table 4-4 ) and consequently are not activated by unmethylated CpG DNA, a potent inducer of IFN-α production by pDCs, which do express TLR9. Unlike pDCs (and monocytes), cDCs express TLR3 (see Table 4-4 ), which, along with RIG-I, allows them to produce type I IFNs and other cytokines in response to double-stranded RNAs, including poly I:C. cDCs also express receptors for DAMPs, such as extracellular ATP and heat shock proteins, as well as receptors that facilitate the uptake of necrotic cell debris.

Table 4-5
Selected Pairs of Surface Molecules Involved in T-Cell–Antigen-Presenting–Cell (APC) Interactions
T Cell Surface Molecule T Cell Distribution Corresponding Ligands on APCs APC Distribution
CD2 Most T cells; higher on memory cells, lower on adult naïve and neonatal T cells LFA-3 (CD58), CD59 Leukocytes
CD4 Subset of αβ T cells with predominantly helper activity MHC class II β chain DCs MΦ, B cells, others (see text)
CD5 All T cells CD72 B cells, MΦ
CD8 Subset of αβ T cells with predominantly cytotoxic activity MHC class I heavy chain Ubiquitous
LFA-1 (CD11a/CD18) All T cells; higher on memory cells, lower on adult naïve and neonatal T cells ICAM-1 (CD54) Leukocytes (ICAM-3 > ICAM-1, ICAM-2) and endothelium (ICAM-1, ICAM-2); most ICAM-1 expression requires activation
ICAM-2 (CD102)
ICAM-3 (CD50)
CD28 Most CD4 T cells, subset of CD8 T cells CD80 (B7-1) DCs, MΦ, activated B cells
CD86 (B7-2)
ICOS Effector and memory T cells, particularly CD4 + T follicular helper cells; not on resting naïve cells B7RP-1 (ICOS-L, B7h) B cells, MΦ, DCs, endothelial cells
VLA-4 (CD49d/CD29) All T cells; higher on memory cells, lower on adult naïve and neonatal T cells VCAM-1 (CD106) Activated or inflamed endothelium (increased by TNF, IL-1, IL-4)
ICAM-1 (CD54) All T cells; higher on memory cells, lower on adult virgin and neonatal T cells LFA-1 (CD11a/CD18) Leukocytes
CTLA-4 (CD152) Activated T cells CD80 DCs, MΦ, activated B cells, activated T cells
CD86
CD40 ligand (CD154) Activated CD4 T cells; lower on neonatal CD4 T cells CD40 DCs, MΦ, B cells, thymic epithelial cells
PD-1 Activated CD4 and CD8 T cells PD-L1, PD-L2 DCs, MΦ, B cells, regulatory T cells
CTLA-4, Cytotoxic T-lymphocyte antigen-4; DCs, dendritic cells; ICAM, intercellular adhesion molecule; ICOS, inducible co-stimulator; IL, interleukin; LFA, leukocyte function antigen; MΦ, mononuclear phagocytes; MHC, major histocompatibility complex; PD-1, programmed death molecule-1; TNF, tumor necrosis factor; VCAM, vascular cell adhesion molecule; VLA-4, very late antigen-4.

Given that the role of cDCs in regulating T-cell immunity is highly nuanced and potentially involves the recognition of diverse types of pathogens in different tissues, it is perhaps not surprising that cDCs are heterogeneous in their ontogeny, location, migration, phenotype, and function. In the human, there is specialization of cDC function that is carried out by two major subsets of human cDCs: CD1c + and CD141 + cDCs.

CD1c + cDCs, which outnumber CD141 + cDCs by about 10-fold, are the major population of cDCs in the blood, tissues, and lymphoid organs and appear to be the human equivalent of the murine CD11b + cDC subset. CD1c + cDCs are equipped with a wide range of pattern recognition receptors and respond well to ligands for TLRs 1 to 8 and express high levels of the dectin-1 and dectin-3 CLRs, which are involved in fungal recognition. They produce TNF-α, IL-8, IL-10, IL-12, and IL-23. Like murine CD11b cDCs, they are particularly good stimulators of CD4 T cells, and, based on their cytokine profile, are likely important for the induction of both Th1 and Th17 immunity. They have a relatively limited ability to cross present antigens to and stimulate CD8 T cells. The importance of CD1c + cDCs in mycobacterial immunity is supported by the report of an infant with disseminated bacillus Calmette-Guérin (BCG) infection with an IRF8 missense mutation that results in selective depletion of CD1c + cDCs and, as a consequence, impaired TLR7/8 ligand–induced IL-12p70 production by peripheral blood mononuclear cells (PBMCs). Because CD1c is involved in nonclassical antigen presentation of mycobacterial products (mycoketides and lipopeptides) to T cells, limitations in mycobacterial antigen presentation might also have played a role in this patient.

CD141 + cDCs, which appear to be the human equivalent of the murine CD8α + CD103 + cDC subset, comprise about 10% of human blood cDCs (i.e., only about 0.1% of PBMCs). CD141 + cDCs are also found among resident cDCs of lymphoid tissue, including lymph node, tonsil, and spleen, and nonlymphoid tissues, including the skin, liver, and lung. Like murine CD8α + CD103 + cDCs, human CD141 + cDCs express the CLEC9A CLR, which facilitates the uptake of necrotic cellular material for antigen processing, and are highly efficient at cross-presentation, in which externally derived proteins that are taken up by endocytosis, pinocytosis, or by CLEC9A are diverted from loading onto MHC class II molecules and, instead, are loaded onto MHC class I molecules for presentation to CD8 T cells (see Antigen Presentation by Classic Major Histocompatibility Complex Molecules ). These DCs are capable of efficiently phagocytosing dead cells by using CLEC9. In contrast to mice, in which CD8α + DCs are highly specialized for cross-presentation, most other human DC populations also have some capacity to cross present antigens to CD8 T cells. CD141 + cDCs produce high levels of IFN-λ (IL-28/29), TNF-α, and the chemokine CXCL10 after engagement of TLR3 by poly I:C, a mimic of viral double-stranded RNA.

Neonatal Conventional Dendritic Cells

Conventional dendritic cells overall constitute a similar fraction (0.5%-1.0%) of blood mononuclear cells in neonates, children, and adults, but cDCs only constitute about 25% of the total of circulating DCs in neonates, whereas cDCs constitute about 75% of the total in adults. The absolute numbers of cDCs remain constant from the neonatal period into adulthood, whereas the fraction and absolute numbers of pDCs decline with increasing postnatal age, reaching numbers similar to adults at greater than or equal to 5 years of age. To the best of our knowledge, the relative proportion of circulating cDCs in the newborn that are CD1c + and CD141 + has not yet been reported. It is likely that the CD1c + cDC subset predominates in the neonatal circulation as in the adult circulation (10:1 ratio with respect to CD141 + cells), so that the results of cDC (Lin CD11c + ) studies using cord blood or infant peripheral blood mainly reflect the properties of the CD1c + subset.

Older flow cytometric studies reported that expression of MHC class II (HLA-DR), CD40, CD80, and CD86 on cord blood and adult peripheral blood cDCs were similar, although a more recent study found that the basal surface expression of HLA-DR and CD80 on cord blood cDCs was substantially decreased compared with circulating adult cDCs. Although cord blood cDCs can stimulate allogeneic cord blood T cells in vitro, it is unclear whether neonatal cDCs are as proficient as adult cDCs in processing and presenting foreign antigens to T cells. Limitations in cord blood cDC antigen presentation are plausible because cord blood cDCs do not upregulate CD40 and CD80 to the same degree as adult cDCs in response to agonists for TLR2/6 (macrophage-activated lipopeptide [MALP]), TLR3 (poly I:C), TLR4 (LPS), or TLR7, whereas TLR8 agonists induce equivalent increases in expression. Decreased maturation of cord blood cDCs was also observed in response to pertussis toxin.

Flow cytometric studies of cytokine expression by cord blood cDCs indicate that they were approximately 50% as efficient as adult cDCs at producing TNF-α in response to LPS, whereas TNF-α production in response to TLR8 agonists was similar. In contrast to TNF-α, IL-1α and IL-6 production by adult cDCs and cDCs from cord blood of term infants in response to LPS was similar, as was production of IL-12/IL-23p40 in response to most TLR agonists. However, cDCs in the cord blood of premature infants have markedly lower responses to these stimuli, which is paralleled by reduced secretion of IL-23 by cord blood mononuclear cell cultures. Given the importance of IL-23 production by cDCs in instructing naïve CD4 T cells for differentiation into Th17 effector cells, it is plausible that premature infants may have compromised Th17 immunity, which may increase their risk of extracellular bacterial and fungal infection.

However, these limitations in cDC function present at birth may be quite transient. A recent longitudinal study of a cohort of South African infants that evaluated cDC cytokine accumulation of TNF-α, IL-6 and IL-12/23p40 by flow cytometry after whole-blood stimulation with TLR agonists found that the cytokine level per cell was higher at 2 weeks of age compared with adults. This enhanced cytokine accumulation was consistent in that it was observed for multiple cytokines (TNF-α, IL-6, and IL-12/23p40) and after stimulation with TLR2/1, TLR4, or TLR7/8 agonists. Of interest, in most cases, the cytokine level per cell gradually declined to adult levels by 1 year of age. This apparently rapid postnatal maturation cDC function may not be limited to cytokine production capacity because there are also marked increases in the cDC expression of HLA-DR and CD80 by 3 months of age compared with at birth.

Earlier studies using stimulation of whole blood or unfractionated blood mononuclear cells also indicated that cord blood cDC responses to TLR3 stimulation were substantially reduced compared with those of adults. TLR3-induced cDC function can be inferred after poly I:C stimulation of whole blood or blood mononuclear cells because poly I:C activates blood cells primarily via TLR3, and cDCs are the only cell type in blood that expresses substantial amounts of TLR3. Therefore the observation that poly I:C–stimulated cord blood and cord blood mononuclear cells had modestly diminished production of type I IFNs and markedly decreased production of IL-12p70 suggested that cord blood cDCs had decreased TLR3-dependent function. The production of IFN-γ by poly I:C–stimulated cord blood and cord blood mononuclear cells also likely reflected diminished TLR3-mediated IL-12p70 and type I IFN production by cDCs, which, in turn, resulted in diminished production of IFN-γ by cord blood NK cells (see Fetal and Neonatal Natural Killer Cell-Mediated Cytotoxicity and Cytokine Production ).

Several studies suggest that the reduced TLR3-mediated cDC function increases during the first month of life. For example, a study in The Gambia found that poly I:C stimulation of whole blood induced significantly higher levels of TNF-α and IFN-γ at 1 month of age compared with at birth (cord blood). Similarly, a study of Belgian infants showed a significant increase in IL-12p70 secretion after poly I:C whole blood stimulation at 1 month of age compared with at birth. In a longitudinal study of South African infants peripheral blood samples from 2-week-old infants had substantially higher levels of IL-12p70, IL-23, IFN-γ, and chemokines that are upregulated by IFN-γ (MCP-1 and IFN-γ–induced protein-10[IP-10]) than did adult blood samples, and these high levels declined to those of adults by approximately 1 year of age. In contrast to these studies, which indicated rapid postnatal acquisition of adult levels of competency for TLR3-induced cDC function, one study of Canadian infants found that poly I:C treatment of cord blood mononuclear cells induced levels of IL-12/23p40, IL-12p70, IFN-γ, and type I IFN that were substantially lower than those of adult peripheral blood mononuclear cells, and they remained so at 1 and 2 years of age. These different results, at least those in the first month of life, could reflect humoral factors, such as adenosine or maternally derived hormones, acting to reduce the cDC-derived production of cytokines in whole blood. However, the precise basis for these findings is unclear, and there is precedent for substantially different levels of TLR-induced cytokine production by cDCs when they are assayed as purified cells versus in whole-blood assays.

A recent study used transcriptional profiling to compare the response of DCs (a mixture of cDCs and pDCs) from cord blood and adult peripheral blood to incubation with RSV. Of interest, cord blood DCs had a transcriptional profile indicating increased transforming growth factor (TGF)-β– and TGF-β–dependent gene expression, and secreted increased levels of TGF-β compared with adult DCs. When RSV-infected DCs were co-cultured with autologous circulating T cells, the adult co-cultured cells secreted relatively high levels of IL-12p70, TNF-α, IL-2, IL-10, IL-13, and IFN-γ, whereas the cord blood co-cultured cells secreted low levels of these cytokines and, compared to the adult cell co-cultures, high levels of IL-1β, IL-4, IL-6, and IL-17. Blockade of TGF-β signaling using a chemical inhibitor in the cord blood co-cultures markedly increased IL-12p70 secretion and more modestly enhanced the levels of other cytokines, such as IL-1β and IL-6. This approach did not address to what extent the results are due to differences between adult and neonatal T-cell populations, that is, a substantial fraction of effector memory T-cells were already committed to the Th1 lineage in adult preparations, whereas the cord blood T-cells were predominantly uncommitted antigenically-naïve T-cells. The importance of cDCs versus pDCs for TGF-β production is also unclear. Despite these limitations, the findings suggest that cord blood cDCs in response to RSV secrete a cytokine milieu (high levels of IL-1β, IL-6, and TGF-β and low levels IL-12p70) that favor Th17 differentiation rather than Th1 differentiation, which could contribute to the immunopathogenesis of RSV infection in young infants.

Plasmacytoid Dendritic Cells

Plasmacytoid dendritic cell have a Lin− CD11c HLA-DR + CD123(IL-3 receptor) + CD303(BDCA-2) + CD304(BDCA4; neuropilin) + surface phenotype. They also express CD4 and CD45RA. Immature pDCs are found in the blood and secondary lymphoid organs. In contrast to migratory cDCs, pDCs appear to migrate to secondary lymphoid organs from the blood via entry into high endothelial venules. They also accumulate at particularly high levels in inflamed lymph nodes. Their characteristic function is the capacity to produce high levels of type I IFNs and certain other cytokines, which achieve systemic levels and directly help to protect the host from viral infection in particular. Although pDCs in the blood and uninflamed tissues have a very limited capacity for antigen uptake and presentation, stimulation of these cells via TLR7 or TLR9 results in their upregulation of CCR7 and migration to T-cell–rich areas of lymph nodes, upregulation of HLA-DR and co-stimulatory molecules, and increased capacity to present antigen to T cells, including cross-presentation of antigen to the CD8 T-cell subset. Consistent with their specialized function, pDCs express only high levels of two TLRs—TLR7, which allows them to respond to single-stranded RNA from RNA viruses such as influenza, parainfluenza, RSV, and HIV-1, and TLR9, which allows them to respond to unmethylated CpG DNA from bacteria, such as S. aureus, and DNA viruses, such as HSV. Because pDCs are the only circulating leukocyte that expresses TLR9, the production of type I IFN by CpG stimulation of whole blood or BMC is often used to infer pDC function. pDCs also express CLRs that are mainly involved in antigen uptake, for instance, BDCA-2 (CLEC4C), DEC-205, dectin-1, DCIR (dendritic cell immunoreceptor), and Fc receptor CD32, rather than innate immune receptors that are directly linked to proinflammatory intracellular signaling pathways.

Neonatal Plasmacytoid Dendritic Cells

Cells with the histologic features of immature pDCs are found in fetal lymph nodes between 19 to 21 weeks of gestation. Using flow cytometry, pDC-lineage cells (Lin CD11c CD4 + CD45RA + CD123 + ) have been identified in fetal liver and bone marrow as early as 16 weeks of gestation; these can be further divided into CD34 + and CD34 subsets that are likely to be pro-pDCs and immature pDCs, respectively. Of interest, both fetal liver pro-pDCs and immature pDCs were capable of high levels of type I IFN production in response to stimulation with irradiated HSV-1, indicating this function of pDCs is established early in ontogeny.

The absolute concentration of pDCs in the term neonate is approximately twofold higher than in the adult and gradually declines after birth, reaching numbers similar to adults at greater than or equal to 5 years of age. The biologic significance of the predominance of pDCs in the neonatal circulation is uncertain. In the prematurely born neonate, there is a trend for a greater proportion of cord blood pDCs with a pro-pDC (CD34 + ) surface phenotype than is observed in adult blood.

Compared with adult pDCs, multiple studies have found that cord blood pDCs produce less type I IFNs in response to unmethylated CpG oligonucleotide, a potent TLR9 ligand. or to TLR7 agonists. Because pDCs appear to be the main sources of type I IFN in response to CpG oligonucleotide stimulation of whole blood or blood mononuclear cells, type I IFN production from these cells can be attributed to pDCs. Thus diminished type I IFN expression in 4-day-old neonates suggests that decreased pDC function persists until at least this age, These reductions in type I IFN production in response to TLR7 and TLR9 agonists are more pronounced in the cord blood of prematurely born infants and are associated with greater immaturity of premature infant pDCs, based on their decreased expression of CD304 (BDCA-4) and “immature” morphology by electron microscopy. The defect in neonatal pDC type I IFN production in response to CpG stimulation appears to result in part from impaired activation and translocation of IRF7 to the nucleus. Consistent with these findings, TNF-α is more modestly reduced, and production of IL-6 is comparable to adult pDCs.

In addition to diminished type I IFN responses, cord blood pDCs also express lower amounts of HLA-DR, CD40, CD80, CD86, and CCR7 after stimulation with TLR7 or TLR9 agonists than adult pDCs. Cord blood pDCs may also have a tendency for reduced survival during culture with CpG oligonucleotides, which could contribute to their reduced responses to these stimuli.

In older studies, type I IFN production by blood cells from neonates in response to direct viral stimulation was significantly diminished, for instance, to HSV, cytomegalovirus (CMV), and parainfluenza virus. As for stimulation by TLR7 or TLR9 ligands, this reduced type I IFN response to herpesviruses was associated with decreased IRF7 cytoplasmic to nuclear translocation. However, a recent report found that type I IFN production in response to influenza A virus (live or heat-inactivated), HIV, or HSV was similar for cord blood and adult peripheral blood pDCs, regardless of whether whole blood, blood mononuclear cells, or purified pDCs were used. The production of TNF-α and the chemokines CCL3 and CCL4 in this study were also similar for cord blood and adult peripheral blood pDCs by using these assay conditions. The reasons for these different findings remain unclear, but they raise the possibility that production of such cytokines by neonatal pDCs may be as competent as those of adults in response to strong viral stimulation. Therefore additional studies to determine pDC function ex vivo in the setting of neonatal viral infection will be of interest.

In limited studies, the age at which responses to TLR ligands become comparable to adult cells has been assessed. CpG-induced upregulation of HLA-DR and CD80 on pDCs did not reach adult levels until 6 to 9 months of age. pDC-derived chemokine production (interferon gamma induced protein 10 [IP-10] and monokine induced by gamma interferon [MIG]) in whole blood was lower during the first year of life compared with adult pDCs. Of interest, pDC-derived IL-6, IL-8, IL-10, and IL-1β were significantly higher than in adult cells from 3 months of age onward, suggesting that neonatal pDCs have a unique cytokine profile that may inhibit Th1 responses (i.e., IL-10) and promote Th17 responses (IL-6 and IL-1β). Consistent with this finding, which suggested a maturation of pDC function by 9 months of age, one study found that pDC production of TNF-α and IL-6 in response to either TLR7/8 or TLR9 agonists at 1 year of age was similar to those of adult pDCs.

Langerhans Cells

Langerhans cells are a unique type of DC found only in the epidermis, in the interstices of keratinocytes, where they can be differentiated from dermal cDCs by their expression of CD1a and Birbeck granules and lack of expression of the factor XIIIa coagulation factor. Langerhans cells also express high levels of Langerin (CD207), although this is not specific for this cell type because subsets of cDCs also express Langerin but do so at lower levels. Langerhans cells are distinct from other DC populations in their ability to undergo local self-renewal in the epidermis (for up to 10 years after limb transplant) and their dependence on M-CSF for their development rather than the Flt3 ligand or GM-CSF. Langerhans cells do not express TLRs 2, 4, and 5, which may account for their limited responsiveness to bacterial stimulation. In uninflamed human skin Langerhans cells seem to mainly be involved in the maintenance of tolerance by selectively promoting the activation of skin-resident regulatory T cells, but in response to infection, for instance, the injection into skin of Candida albicans, can present antigen to skin-resident pathogen-specific memory T cells. However, cell lineage tracing studies suggest that Langerhans cells may play a relatively minor role compared with migratory dermal cDCs in the activation of T cells in the draining lymph nodes, even though they may cross present protein antigens as efficiently as cDCs.

In the embryonic mouse, Langerhans cells appear to arise largely from fetal liver monocytes that seed the skin before bone marrow hematopoiesis initiates. This may also apply to humans because Langerhans cells can be detected in human embryonic skin as early as 6 to 7 weeks of gestation. However, there is little information as to the function of Langerhans cells in the human fetus and neonate.

Inflammatory and Monocyte-Derived Dendritic Cells

Murine inflammatory DCs, in contrast to cDCs, are derived from mature monocytes that are exposed to strong inflammatory stimuli, such as LPS or gram-negative bacteria. Murine inflammatory DCs, like cDCs, have cytoplasmic protrusions, upregulate CCR7, home to T-cell areas of peripheral lymphoid tissue, and can efficiently present antigen to and activate CD4 and CD8 T cells; however, inflammatory DCs are distinct in expressing DC-SIGN (CD209a). Human inflammatory DCs with many of these features, and that are likely to have been derived from monocytes, have been described in psoriatic skin, inflammatory tumor ascites fluid, and synovial fluid of patients with rheumatoid arthritis. Cells similar, although not identical to, inflammatory DCs can be generated by culturing monocytes in GM-CSF plus IL-4 to produce monocyte-derived DCs (moDCs), which can then be matured by treatment with TNF-α.

Whether inflammatory DCs occur in the neonate in settings of strong inflammatory stimuli, such as bacterial sepsis, is not known. Studies comparing neonatal and adult moDCs suggest that the capacity of neonatal monocytes to differentiate into inflammatory DCs that can instruct Th1 immunity is limited. Most studies have found that neonatal moDCs produce much less IL-12p70 in response to LPS, poly I:C, and TLR8 ligands or engagement of CD40 than adult moDCs. Reduced production of IL-12p70 is due to reduced amounts of the IL-12p35 component, whereas the p40 component that is common to IL-12p70 and IL-23 (IL-12/23p40) is produced in similar amounts by neonatal and adult cells. Diminished IL-12p35 production appears to result from a defect in IRF3 binding to and remodeling of the IL-12p35 promoter, whereas more proximal aspects of signaling resulting in IRF3 translocation from the cytoplasm to the nucleus appear to be intact. Although neonatal moDCs stimulated with live Mycobacterium bovis BCG or with a TLR8 agonist plus LPS or poly I:C secreted much less IL-12 than adult moDCs, when moDCs were stimulated with these combinations of TLR ligands in cultures also containing autologous naïve CD4 T cells, comparable IL-12–dependent IFN-γ production was observed. Together, these observations suggest that defective production of IFN-γ–inducing cytokines, including IL-12, by neonatal DCs can be overcome by combined signaling from TLRs, NLRs, and direct physical interactions between T cells and DCs.

Summary

DCs are detectable by 16 weeks of gestation. At birth, the concentration of cDCs is similar and the concentration of pDCs greater than in adult blood. Although adult and neonatal blood cDCs and moDCs express on their surface the MHC class II molecule HLA-DR and co-stimulatory molecules in similar abundance, expression by neonatal DCs increases less in response to stimulation via TLRs. TLR-stimulated neonatal cDCs and moDCs generally produce substantially less IL-12 and type I IFNs, cytokines that contribute to early innate defenses and subsequent T-cell–mediated defenses against intracellular pathogens. By contrast, these neonatal DCs produce proinflammatory cytokines and IL-23, which are important in defense against extracellular bacterial and fungal pathogens, more efficiently than their adult counterparts. These differences may limit the ability of neonatal DCs to activate naïve pathogen-specific T cells and, in particular, to induce IFN-γ–producing Th1 T-cell responses rather than Th17 or Th2 T-cell responses. However, neonatal DCs may be able to produce IL-12 and support IFN-γ production by neonatal T cells in response to combinatorial activation of innate immune receptors and when in contact with T cells. Whether neonatal pDCs are deficient in the production of type I IFNs, IFN-dependent chemokines, and other functions during in vivo viral infections remains unclear; these cells are markedly less responsive to stimulation with chemical TLR7 and TLR9 agonists.

Natural Killer Cells and Innate Lymphoid Cells

Natural Killer Cells

Overview and Development

NK cells are large granular lymphocytes with cytotoxic function, which, unlike T and B lymphocytes, lack antigen-specific T-cell receptor (TCR) or B-cell receptor (BCR) receptors characteristic of the adaptive immune system and instead express a diverse array of activating and inhibitory receptors. Although NK cells are generally considered to be a component of the innate rather than adaptive immune system, in mouse models they can be primed by infection, such as with herpesviruses, and retain certain features of memory/effector T cells, such as enhanced cytotoxicity and cytokine secretion for at least several months postinfection. In clinical practice, NK cells are usually defined as lymphoid cells that express CD16 and CD56 but not CD3, that is, have a CD16 + CD56 + CD3 surface phenotype. Virtually all circulating NK cells from adults also express the NK-cell–specific NKp30 and NKp46 receptors, along with CD2 and CD161, and approximately 50% express CD57, but these molecules are found on other lymphocyte types as well.

The fetal liver produces NK cells as early as 6 weeks of gestation, but the bone marrow is the major site for NK-cell production from late gestation onward. NK cells are derived from bone marrow cells that lack surface molecules specific for other cell lineages (i.e., CD34 + Lin cells) but express CD7 or CD38. NK-cell development is dependent on the IL-15/IL-15 receptor (which consists of the IL-15 receptor α chain, the IL-2 receptor β chain, and the common γ chain)/Janus tyrosine kinase 3 (JAK3) pathway. Based on the lack of NK-cell development in genetic immunodeficiencies, human NK-cell development also requires adenylate kinase 2 and the GATA binding protein 2 transcription factor. In vitro studies suggest a NK lineage cell developmental sequence in which CD161 is acquired early in NK development. At the next developmental stage, NKp30, NKp46, 2B4, and NKG2D are expressed on the cell surface, followed by members of the killer cell inhibitor receptor (KIR) family, CD94-NKG2A, CD2, and CD56; the function of these molecules is discussed in the sections that follow.

NK cells are functionally defined by their natural ability to lyse virally infected or tumor target cells in a non–HLA-restricted manner that does not require prior sensitization. NK cells preferentially recognize and kill cells expressing ligands for activating receptors that are not antigen-specific in conjunction with reduced or absent expression of self-HLA class I molecules, a property referred to as natural cytotoxicity. These ligands for activating receptors are characteristically increased in response to stresses, such as infection or malignant transformation. This is in contrast with cytotoxic CD8 T cells, which are triggered to lyse targets after the recognition of foreign antigenic peptides bound to self-HLA class I molecules or self-peptides bound to foreign HLA class I molecules. NK cells also have the ability to kill target cells that are coated with IgG antibodies, a process known as antibody-dependent cellular cytotoxicity (ADCC). ADCC requires the recognition of IgG bound to the target cell by the NK-cell FcγRIIIB receptor (CD16).

Mature NK cells can be subdivided into CD56 hi CD16 lo and CD56 lo CD16 hi populations. CD56 hi CD16 lo cells usually are only a minority of mature NK cells in the circulation but express CCR7 and L-selectin and predominate in lymph node tissue. CD56 hi CD16 lo cells have limited cytotoxic capacity but produce cytokines and chemokines efficiently, whereas the inverse is true for CD56 lo CD16 hi NK cells. These features suggest that the CD56 hi CD16 lo subset could regulate lymph node T cells and DCs through cytokine secretion. Developmental studies suggest that CD56 hi CD16 lo NK cells are less mature than CD56 lo CD16 hi NK cells, but the precise precursor-product relationship of these subsets under various conditions in vivo has not been firmly established.

NK cells are particularly important in the early containment of viral infections, especially with pathogens that may initially avoid control by adaptive immune mechanisms. Infection of host cells by the herpesvirus group, including HSV, CMV, and varicella-zoster virus (VZV), and some adenoviruses leads to decreased surface expression of HLA class I molecules. Viral protein–mediated decreases in expression of HLA class I may limit the ability of CD8 T cells to lyse virally infected cells and to clonally expand from naïve precursors. These virus-mediated effects may be particularly important during early infection, when CD8 T cells with appropriate antigen specificity are present at a low frequency. By contrast, decreased HLA class I expression in conjunction with the expression of ligands for activating receptors facilitates recognition and lysis by NK cells. The importance of NK cells in the initial control of human herpesvirus infections is suggested by the observation that persons with selective deficiency of NK-cell numbers or function are prone to severe infection with HSV, CMV, and VZV.

Natural Killer Cell Receptors

NK-cell cytotoxicity is regulated by a complex array of inhibitory and activating receptor–ligand interactions with target cells ( Fig. 4-3 ). The expression of multiple combinations of inhibitory and activating receptors leads to a large amount of NK-cell diversity. By some estimates, there are 6000 to 30,000 different NK-cell populations within an individual. Individual NK cells in the bone marrow appear to undergo a tuning process so that the threshold for their activation is appropriate for their particular repertoire of receptors. NK-cell activation is inhibited by recognition of HLA class I molecules expressed on nontransformed, uninfected cells; this recognition is presumed to provide a net inhibitory signal that predominates over activating signals. Infection or other perturbations of the host target cell, such as malignant transformation, can reduce the amount of HLA class I on the cell surface, thereby reducing inhibitory signaling, and upregulate other molecules that promote NK-cell activation, such as MHC class I–related chains A and B (MICA and MICB).

Figure 4-3, Positive and negative regulation of NK-cell cytotoxicity by receptor–ligand interactions. NK-cell cytotoxicity is inhibited by engagement of KIR by MHC class I molecules, such as HLA-B and HLA-C. In addition, NK cells are inhibited when CD94-NKG2A complex, a member of the C-type lectin family, on the NK cell is engaged by HLA-E. HLA-E binds hydrophobic leader peptides derived from HLA-A, HLA-B, and HLA-C molecules and requires these for its surface expression. Thus HLA-E surface expression on a potential target cell indicates the overall production of conventional MHC class I molecules. These inhibitory influences on NK-cell cytotoxicity are overcome if viral infection of the target cell results in decreased MHC class I and HLA-E levels. NK-cell cytotoxicity is positively regulated by the engagement of NKG2D, which interacts with MICA, MICB, and ULBPs; 2B4, which interacts with CD48; and natural cytotoxicity receptors, such as NKp30 and, not shown, NKp46, for which the ligands on the target cell induced by infection are unknown. CD16 is an Fc receptor for IgG and mediates antibody-dependent cellular cytotoxicity against cells coated with antibody, for instance, against viral proteins found on the cell surface. Positive receptors mediate their intracellular signals via associated CD3-ζ, DAP10 or DAP12 proteins. DAP, DNAX-activating protein; HLA, human leukocyte antigen; IgG, immunoglobulin G; KIR, killer inhibitory receptors; MHC, major histocompatibility complex; MICA and MICB, MHC class I–related chains A and B; NK, natural killer; SAP, SLAM-associated protein; ULBPs, UL16-binding proteins (UL16 is a cytomegalovirus protein).

There are two major families of inhibitory NK-cell receptors that recognize HLA class I molecules in humans: KIR and the CD94-containing C-type lectin families. KIRs with a long cytoplasmic domain transmit signals that inhibit NK-cell activation; most, although not all, NK cells express one or more inhibitory KIRs on their surface. Most NK cells, including all those not expressing any inhibitory KIRs, also express inhibitory CD94-NKG2A receptors. KIRs bind to polymorphic HLA-B, HLA-C, or HLA-A molecules, whereas CD94-NKG2A binds to HLA-E, which is monomorphic (HLA molecules are discussed later in the sections “Antigen Presentation by Classic Major Histocompatibility Complex Molecules” and “Nonclassic Antigen-Presentation Molecules”). Because HLA-E reaches the cell surface only when its peptide binding groove is occupied by hydrophobic peptides derived from the leader sequences of HLA-A, HLA-B, and HLA-C molecules, the amount of HLA-E on the cell surface reflects the overall levels of HLA-A, HLA-B, and HLA-C molecules on that cell.

In addition to CD94-NKG2A, a third group of inhibitory receptors that broadly recognize HLA class I molecules are the leukocyte immunoglobulin-like receptors B1 and B2 (LILRB1 and LILRB2). LILRB1 and LILRB2, also referred to as LIR1/CD85J and LIR2/CD85d, bind to HLA-A, HLA-B, and HLA-C molecules, as well as the nonconventional class I molecules HLA-E, HLA-F, and HLA-G. HLA-G is the only HLA class I molecule constitutively expressed on the surface of fetal trophoblasts. Thus the interaction of LILRB1 and LILRB2 with HLA-G is thought to protect the placenta from injury by maternal NK cells.

Countering the effects of these inhibitory receptors are multiple types of activating receptors. NKG2D is found on NK cells as well as in certain T-cell populations. NKG2D recognizes MICA, MICB, and UL16-binding proteins (ULBPs 1-4). MICA and MICB are nonclassical HLA class I molecules that are expressed on stressed or infected cells. ULBPs are a group of HLA class I–like molecules expressed on many cell types that were first identified and named based on their ability to bind to the human CMV UL16 viral protein. In human CMV infection, UL16 probably limits NK-cell– and T-cell–mediated activation by binding to ULBPs internally and preventing their surface expression on the infected cell.

NK cells also express NKp30, NKp44, and NKp46. NKp44 and NKp46 can trigger NK-cell cytotoxicity through their recognition of influenza virus hemagglutinin and Sendai (parainfluenza family) virus hemagglutinin-neuraminidase. These receptors also recognize ligands on tumor cells and cells infected with herpesviruses, but with the exception of NKp30, which recognizes the tumor ligand B7-H6, the definitive nature of the ligands is not known.

The proteins 2B4 (CD244) and NTBA are members of the signaling lymphocytic-activation molecule (SLAM) protein family and are expressed on most NK cells. 2B4 binds to CD48, whereas the ligand for NTBA remains unclear. Both 2B4 and NTBA engagement triggers NK-cell activation through SLAM-associated protein (SAP), an intracellular adapter protein that is mutated in the X-linked lymphoproliferative syndrome.

Finally, NK cells may express KIRs with short cytoplasmic tails, which unlike their counterparts with long cytoplasmic tails, activate NK cells by a DNAX-activating protein 12 (DAP12) signaling mechanism. Also, unlike CD94-NKG2A, CD94-NKG2C is an activating receptor complex that also activates via DAP12 signaling. These activating KIRs and CD94-NKG2C and their respective inhibitory forms have identical or very similar ligand specificities. How NK cells integrate the effects on natural cytotoxicity of these multiple inhibitory and activating receptors, particularly those that recognize the same or similar ligands, remains unclear.

Natural Killer Cell Cytotoxicity

After binding via adhesion molecule interactions and activation, NK cells release perforin and granzymes from preformed cytotoxic granules into a synapse formed between the NK cell and its target, leading to death by apoptosis of the target cell. NK cell–mediated cytotoxicity also may be mediated by Fas ligand or TRAIL (TNF-related apoptosis-inducing ligand) expressed on the activated NK cell surface. Fas–Fas ligand interactions appear not to be essential for human NK cell control of viral infections because persons with dominant-negative mutations of the Fas or Fas ligand genes develop autoimmunity but do not experience an increased severity of virus infections. In contrast with natural cytotoxicity, in which perforin/granzyme-dependent mechanisms appear to be predominant, ADCC appears to use both perforin/granzyme- and Fas ligand–dependent cytotoxic mechanisms.

Natural Killer Cell Cytokine Responsiveness and Dependence

NK cell proliferation and cytotoxicity are enhanced in vitro by cytokines produced by T cells (IL-2, IFN-γ), APCs (IL-1β, IL-12p70, IL-18, and type I IFNs), and nonhematopoietic cells (IL-15, stem cell factor, Flt3 ligand, IFN-β). IL-15, which appears critical for the development of NK cells, also promotes the survival of mature NK cells and, like IL-12, increases the expression of perforin and granzymes. A subset of NK cells found in mucosal-associated lymphoid tissues responds to IL-23 by producing cytokines, including IL-22, that help to protect the gut from bacterial pathogens.

Natural Killer Cell Cytokine and Chemokine Production

NK cells are also important producers of IFN-γ and TNF-α in the early phase of the immune response to viruses, and IFN-γ may promote the development of CD4 T cells into Th1 effector cells (see “Differentiation of Activated Naïve T Cells into Effector and Memory Cells” ). NK-cell–mediated IFN-γ production may be induced by the ligation of surface β1 integrins on the NK-cell surface, as well as by the cytokines IL-1, IL-12, IL-15, and IL-18, which are produced by DCs and mononuclear phagocytes. The combination of IL-12 and IL-15 also potently induces NK cells to produce the CC chemokine macrophage inhibitory protein-1α (MIP-1α; CCL-3), which may help to attract other types of mononuclear cells to sites of infection, where NK-cell–mediated lysis takes place. NK cells from HIV-infected persons also are able to produce a variety of CC chemokines, including MIP-1α, MIP-1β (CCL-4), and RANTES (regulated on activation, normal T-cell expressed and secreted; CCL-5) in response to treatment with IL-2 alone; these chemokines may help prevent HIV infection of T cells and mononuclear phagocytes by acting as antagonists of the HIV co-receptor CCR5. NK cells also can be triggered to produce a similar array of cytokines during ADCC in vitro, but the role of such ADCC-derived cytokines in regulating immune responses in vivo is poorly defined. Some of the cytokine-dependent mechanisms by which NK cells, T cells, and APCs may influence each other’s function, such as in response to infection with viruses and other intracellular pathogens, are summarized in Figure 4-4 .

Figure 4-4, Cytokines link innate and antigen-specific Th1 immune mechanisms against intracellular pathogens. Activation of T cells by antigen-presenting cells, such as dendritic cells and mononuclear phagocytes, results in the expression of CD40 ligand (CD40L) and the secretion of cytokines, such as interleukin-2 (IL-2) and interferon- γ (IFN-γ). Mononuclear phagocytes are activated by IFN-γ and the engagement of CD40 with increased microbicidal activity. Mononuclear phagocytes produce tumor necrosis factor-α (TNF-α), which enhances their microbicidal activity in a paracrine or autocrine manner. Mononuclear phagocytes and dendritic cells also secrete cytokines IFN-α/β, IL-12p70, IL-18, and IL-27. These cytokines promote Th1 effector cell differentiation, and most also promote activation of natural killer (NK) cells. Mononuclear phagocytes and dendritic cells, as well as nonhematopoietic cells, also secrete IL-15, which is particularly important for NK-cell maturation and the generation and homeostasis of effector and memory CD8 T cells. NK-cell activation is augmented further by other cytokines, such as IL-2 and IL-21, which are produced by CD4 T cells and other cell types. Activated NK cells secrete IFN-γ, which enhances mononuclear phagocyte activation and Th1 effector-cell differentiation futher.

Natural Killer Cells of the Maternal Decidua and Human Leukocyte Antigen G

The maternal decidua contains a prominent population of NK cells, which may help contribute to the maintenance of pregnancy. NK cells belonging to the CD56 hi CD16 lo subset, which have a high capacity for cytokine production but low capacity for cytotoxicity, predominate. Murine studies suggest that maternal NK-cell–derived cytokines, such as IFN-γ, may help to remodel the spiral arteries of the placenta. Although the NK-cell populations of the decidua have a low capacity for cytotoxicity, their presence in a tissue lacking expression of HLA-A, HLA-B, and HLA-C molecules could potentially contribute to placental damage and fetal rejection. As noted earlier, the expression by human fetal trophoblast of HLA-G is thought to protect this tissue from attack by maternal NK cells through binding to the inhibitory receptors LILRB1 and LILRB2.

Natural Killer–Cell Numbers and Surface Phenotype in the Fetus and Neonate

Circulating NK cells become increasingly abundant during the second trimester, and at term, their numbers in the neonatal circulation (approximately 15% of total lymphocytes) are typically the same as or greater than in adults. Relatively high frequencies of NK cells are found in the fetal liver, lung, and spleen, whereas frequencies in the bone marrow and mesenteric lymph nodes are relatively low. The fraction of blood neonatal and adult NK cells that are CD56 hi CD16 −/lo (approximately 10%) and CD56 lo CD16 + NK cells (approximately 90%) is similar. Earlier studies of cell surface molecule expression on neonatal NK cells varied in their conclusions regarding the expression of molecules involved with adhesion, activation, inhibition, and cytotoxicity by neonatal NK cells. More recent studies using newer and more reliable methods suggest that neonatal NK cells have decreased expression of ICAM-1 but similar expression of other adhesion molecules, similar or greater expression of the inhibitory CD94-NKG2A complex but reduced expression of the inhibitory LILRB1 (LIR1) receptor, similar or greater expression of the activating NKp30 and NKp46 receptors, similar or slightly reduced expression of the activating NKG2D receptor, and similar to reduced surface expression of CD57. The abundance of the cytotoxic molecules perforin, granzyme B, Fas ligand, and TRAIL is as great or greater in neonatal NK cells as in adult NK cells. Thus these findings suggest that neonatal NK cells differ phenotypically from but are not simply immature versions of adult NK cells.

Congenital viral or Toxoplasma infection during the second trimester can increase the number of circulating NK cells, which have phenotypic features of activated cells.

Fetal and Neonatal Natural Killer Cell–Mediated Cytotoxicity and Cytokine Production

Recent studies of NK cells of the fetal lung and other tissues have found that KIR-expressing cells are hyporesponsive in terms of cytotoxicity compared with those of adult peripheral blood or lung tissue. Fetal tissue NK-cell function also appears to be substantially more sensitive to the suppressive effects of TGF-β than adult NK cells. The cytotoxic function of circulating NK cells increases progressively during fetal life to reach values approximately 50% (a range of 15%-60% in various studies) of those in adult cells at term, as determined in assays using tumor cell targets and either unpurified or NK cell–enriched preparations. Reduced cytotoxic activity by neonatal NK cells has been observed in studies using cord blood from vaginal or cesarean section deliveries or peripheral blood obtained 2 to 4 days after birth ; full function is not achieved until at least 9 to 12 months of age. Decreased cytotoxic activity by neonatal NK cells compared with adult cells also is consistently observed with HSV- and CMV-infected target cells. By contrast, both neonatal and adult NK cells had equivalent cytotoxic activity against HIV-1–infected cells. These results suggest that ligands on the target cell or the target cell’s intrinsic sensitivity to induction of apoptosis may influence fetal and neonatal NK-cell function. The mechanisms of these pathogen-related differences remain unclear but may contribute to the severity of neonatal HSV infection. Paralleling the reduction in natural cytotoxic activity of neonatal cells, ADCC of neonatal mononuclear cells is approximately 50% of that of adult mononuclear cells, including against HSV-infected targets.

The reduced cytotoxic activity of neonatal NK cells appears not to reflect decreased expression of cytotoxic molecules but, instead, may result from diminished adhesion to target cells, perhaps as a result of decreased expression of ICAM-1 or diminished recycling of cells to kill multiple targets. However, the mechanisms responsible for diminished neonatal NK-cell cytotoxicity have not been conclusively defined. Cytokines, including IL-2, IL-12, IL-15, IFN-α, IFN-β, and IFN-γ, can augment the cytotoxic activity of neonatal NK cells, as they do for adult NK cells, and with fetal tissue NK cells, the augmentation of cytotoxicity may exceed that for adult NK cells. Consistent with the ability of IL-2 and IFN-γ to augment their cytolytic activity, neonatal NK cells express on their surface receptors for IL-2/IL-15 and IFN-γ in numbers that are equal to or greater than those of adult NK cells. Treatment of neonatal NK cells with ionomycin and phorbol myristate acetate (PMA) also enhances natural cytotoxicity to levels present in adult NK cells. This increase is blocked by inhibitors of granule exocytosis, indicating that decreased release of granules containing perforin and granzyme may contribute to reduced neonatal NK cytotoxicity. Finally, decreased neonatal NK cytotoxicity is not determined at the level of the precursor cells of the NK-cell lineage: Donor-derived NK cells appear early after cord blood transplantation, with good cytotoxicity effected through the perforin/granzyme and Fas–Fas ligand cytotoxic pathways.

Neonatal NK cells produce IFN-γ as effectively as adult NK cells in response to exogenous IL-2, IL-12, IL-18, HSV, and polyclonal stimulation with ionomycin and PMA, but fewer neonatal NK cells express TNF-α than do adult NK cells after ionomycin and PMA stimulation. Neonatal NK cells produce chemokines that suppress the growth of HIV strains that use CCR5 as a co-receptor to infect CD4 T cells but not those strains that use CXCR4 as a receptor.

Innate Lymphoid Cells

Innate lymphoid cells (ILCs) are a family of lymphocytes derived from a common developmental pathway that is closely related to NK cells. ILCs are involved in tissue development and immunity. Like NK cells, they are distinguished from other cell types by their absence of antigen-specific T-cell receptors or B-cell receptors, a lack of DC or myeloid cell markers, and a lymphoid morphology. ILCs lack most markers and receptors that are characteristic of NK cells and typically have a relatively limited ability to mediate cell-mediated cytotoxicity. Their distinct functional feature is their ability to secrete cytokines in patterns that in many cases follow the patterns of CD4 T-cell helper (Th) effector subsets (see “Regulation of CD4 Effector T-Cell Subset Differentiation” ). Subsets of ILCs have now been described that have the cytokine production profiles of several of the T-helper effector cell subsets, that is, they secrete cytokines in a mutually exclusive pattern that is characteristic of the major Th effector subsets, including IFN-γ (Th1), IL-4 and/or IL-13 (Th2), and IL-17 and/or IL-22 (Th17). Human ILCs were first described in the lymph nodes and spleen of the second trimester fetus and have a Lin CD45 mid CD127 hi surface phenotype. These cells can be found in the mesentery of the human fetal intestine as early 8 to 9 weeks of gestation before the appearance of distinct lymph nodes in the second trimester of pregnancy. They appear to be analogous to murine lymphoid tissue inducer (LTi) cells in playing a role in lymph node organogenesis by expressing lymphotoxin-α, lymphotoxin-β, and IL-17 and inducing mesenchymal cells to increase expression of adhesion molecules important in lymphoid tissue organogenesis.

More recent studies reveal that IL-17/IL-22– and IL-4/IL-13–producing ILCs are found in mucosal tissues, including within the epithelium and in lymphoid-associated tissues such as Peyer patches and the lamina propria. They are intimately involved in interactions between epithelial cells, immune cells, and commensal microbes and help to maintain mucosal homeostasis. Based on murine studies, ILCs of the intestines appear to present antigens on MHC class II molecules to CD4 T cells, but rather than inducing activation, they limit CD4 T-cell responses to intestinal commensal bacteria. Also, based on murine studies, their IL-22 production likely helps maintain high levels of defensins and other antimicrobial proteins and mucosal integrity, and their IL-17 and IL-22 production is likely an important firs-tline defense against bacterial and fungal pathogens. However, like Th17 cells, dysregulated expression of these cytokines by ILCs has been associated with inflammatory bowel disease. Human IFN-γ–secreting ILCs have also been implicated in the pathogenesis of inflammatory bowel disease. IL-4/IL-13–secreting ILCs are potentially first responders to mucosal infection with multicellular parasites, such as worms, but, like Th2 cells, may also play an important role in the pathogenesis of allergic diseases.

Little is known of ILCs in the intestines of the neonate and young infant, but given the importance of these cells in mucosal homeostasis and regulation of commensal bacteria, such knowledge may be important in understanding normal mucosal immune development after birth as well as the pathogenesis of certain diseases such as NEC.

Summary

NK cells appear early during gestation and are present in normal numbers by mid to late gestation. However, certain phenotypic features of NK cells differ from those of adult NK cells. Neonatal NK cells appear to be as capable as adult cells of producing IFN-γ and chemokines that inhibit the ability of CCR5-trophic HIV strains to infect CD4 T cells but may produce less TNF-α and chemokines that inhibit infection by CXCR4-trophic strains of HIV. Compared with adult NK cells, neonatal NK cells have decreased cytotoxicity to many types of target cells, including HSV- and CMV-infected but not HIV-infected cells. This decreased cytotoxicity is particularly evident in the tissues. Neonatal NK cell cytotoxicity can be augmented by incubation with cytokines, such as IL-15 in vitro, suggesting a potential immunotherapeutic strategy, and can also be expanded into large numbers for cellular immunotherapy.

T Cells and Antigen Presentation

Overview

T cells are so named because the vast majority of these cells originate in the thymus. They, along with B cells, which in mammals develop in the bone marrow, comprise the adaptive or antigen-specific immune system. T cells play a central role in antigen-specific immunity because they directly mediate and regulate cellular immune responses and play a critical role in facilitating antigen-specific humoral immune responses by B cells. Most T cells recognize antigen in the form of peptides bound to MHC molecules on APCs. Antigen-specific TCRs are heterodimeric molecules composed of either α and β chains (αβ-TCRs) ( Fig. 4-5 ) or γ and δ chains (γδ-TCRs), with the amino-terminal portion of each of these chains variable and involved in antigen recognition. This variability is generated, in large part, as a result of TCR gene rearrangement of variable (V), diversity (D), and joining (J) segments. The TCR on the cell surface is invariably associated with the nonpolymorphic complex of CD3 proteins, which include CD3-γ, -δ, -ε, and -ζ (see Fig. 4-5 ). The cytoplasmic domains of proteins of the CD3 complex include 10 immunoreceptor tyrosine-based activation motifs (ITAMs), which serve as docking sites for the lck and ZAP-70 (CD3 zeta-associated protein of 70 kilodaltons) intracellular tyrosine kinases that transduce proximal activation signals to the interior of the cell after the TCR has been engaged by antigen.

Figure 4-5, T-cell recognition of antigen and activation. αβ T-cell receptor (αβ-TCR) recognizes antigen presented by the antigen-presenting cell (APC) in the form of antigenic peptides bound to major histocompatibility complex (MHC) molecules on the APC surface. Most CD4 T cells recognize peptides bound to MHC class II, whereas most CD8 T cells recognize peptides bound to MHC class I. This MHC restriction is the result of a thymic selection process and is due in part to an intrinsic affinity of the CD4 and CD8 molecules for the MHC class II and class I molecules, respectively. Once antigen is recognized, the CD3 protein complex, which is invariably associated with the TCR, acts as docking site for tyrosine kinases that transmit activating intracellular signals. Interaction of the T-cell CD28 molecule with either CD80 (B7-1) or CD86 (B7-2) provides an important co-stimulatory signal to the T cell, leading to complete activation, rather than partial activation or functional inactivation (anergy).

Most T cells that bear an αβ-TCR (or αβ T cells), also express on their surface the CD4 or CD8 co-receptors in a mutually exclusive manner and are commonly referred to as CD4 or CD8 T cells, respectively. Nearly all CD8 T cells recognize protein antigens in the form of 7- to 9-mer peptide fragments bound to MHC class I molecules of the classic type (HLA-A, HLA-B, and HLA-C in humans). CD4 T cells recognize antigen presented by MHC class II molecules (HLA-DR, HLA-DP, and HLA-DQ in humans); most of these antigens are in the form of peptide fragments typically ranging between 12 to 22 amino acids in length. MHC class II presentation of certain zwitterionic bacterial polysaccharides, such as those derived from Bacteroides fragilis, can also occur.

APCs, which include DCs, mononuclear phagocytes, and B cells, constitutively express both MHC class I and class II molecules, which allows them to present antigenic peptides to CD8 and CD4 T cells, respectively. DCs are particularly important for presentation to T cells that are antigenically naïve and that have not been previously activated by foreign antigen. γδ T cells, which mainly recognize stress-induced molecules rather than peptide/MHC complexes, have distinct immune functions from αβ T cells and are discussed later in a separate section.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here