Abstract
Recent research has pointed to the developing immune system as a remarkably sensitive toxicologic target for environmental chemicals and drugs. In fact, the perinatal period before and just after birth is replete with dynamic immune changes, many of which do not occur in adults. These include not only the basic maturation and distribution of immune cell types and selection against autoreactive lymphocytes but also changes designed specifically to protect the pregnancy against immune-mediated miscarriage. The newborn is then faced with critical immune maturational adjustments to achieve an immune balance necessary to combat myriad childhood and later-life diseases. All these processes set the fetus and neonate completely apart from the adult regarding immunotoxicologic risk. Yet for decades, safety evaluation has relied almost exclusively upon exposure of the adult immune system to predict perinatal immune risk. Recent workshops and forums have suggested a benefit in employing alternative exposures that include exposure throughout early life stages. However, issues remain concerning when and where such applications might be required. In this review we discuss the reasons why immunotoxic assessment is important for current childhood diseases and why adult exposure assessment cannot predict the effect of xenobiotics on the developing immune system. It also provides examples of developmental immunotoxicants where age-based risk appears to differ. Finally, it stresses the need to replace adult exposure assessment for immune evaluation with protocols that can protect the developing immune system.
Keywords: allergy, atopy, autoimmunity, children’s health, developmental immunotoxicology, immune balance, immunoglobulin E, perinatal risk, safety testing
The premise of this review is that the developing immune system represents a particularly sensitive xenobiotic target that is not effectively modeled through routine screening for immunotoxicity using adult exposure. Hence, adult exposure testing for immunotoxicity is limited in application, cannot address the most significant immune vulnerabilities, and should be replaced with a more predictive assessment protocol. This conclusion is drawn from recent developmental immunotoxicity findings, including those from our own laboratory, as well as from the conclusions of numerous conferences and workshops. These sources point to the special vulnerabilities of the perinatal immune system compared with the fully matured and dispersed immune system of the adult.
Individuals in early-life stages have been recognized as a special subset of the population that is likely to be at greater toxicologic risk than adults (Daston et al. 2004; Kimmel 2005; Landrigan et al. 2004; Selevan et al. 2000). Within this broader framework, a significant number of recent review and consensus workshop reports have stressed that early-life exposure to xenobiotics poses the greatest environmental risk for the immune system and would be expected to exert the greatest effect on subsequent human health (Dietert 2005; Dietert et al. 2000, 2002; Holladay 1999; Holladay and Smialowicz 2000; Holsapple et al. 2005; Kimmel et al. 2005; Luebke et al. 2006; Luster et al. 2005; Van Loveren and Piersma 2004). Yet, immune-associated safety from problematic exposure to environmental chemicals as well as drugs has hinged on adult exposure assessment [Hinton et al. 2000; Luster et al. 1992 U.S. Food and Drug Administration 1999]. Developmental immunotoxicity screening was not included in one recent immunotoxicity draft guidance covering human pharmaceuticals (U.S. Food and Drug Administration 2004), although its potential application within safety screening is under ongoing consideration (Holsapple et al. 2005; Ladics et al. 2005).
In this review we highlight some novel processes of perinatal immune development that both contribute to the immunotoxic vulnerability of the developing immune system and cannot be effectively examined via current adult-exposure assessment. Additionally, specific examples of the problems associated with reliance on adult-induced immunotoxicity assessment are shown for a variety of immunotoxicants.
Perinatal Immune Development versus the Adult Immune System
Immune development has been characterized from a toxicologic perspective through a series of discrete functional changes representing critical windows of differential vulnerability to toxicants (Dietert et al. 2000; Holsapple et al. 2003; Landreth 2002; Landreth and Dodson 2005; Leibnitz 2005). These reviews have emphasized that each “window” of development likely has different immunologic risks associated with immunotoxicant exposure, and indeed, examples of differential immunotoxic outcomes among these windows do exist (Bunn et al. 2001b; Lee et al. 2001). Although it is not practical or necessary to directly evaluate the differential risk of limited exposures over different periods within perinatal development, it is important to accurately estimate immunologic risk across the entire period of immune development because of the important role of effective immune function in children’s health (Daston et al. 2004; Kimmel et al. 2005).
Table 1 draws upon the broader critical windows of immune development (Dietert et al. 2000) to illustrate a set of seven discrete events that are either unique to perinatal immune development or critical to the post-natal immune integrity while serving a different role in the adult. These immune events include those that are restricted solely to the immune system as well as some involving the role of the immune system in host organ/tissue homeostasis. In most cases, clear associations exist between exposure to specific toxicants and disruption of the perinatal event. It is not simply by chance that this set of early immune events seems to impinge primarily on the risk of atopy, autoimmune disease, and later-life immune balance (which also influences risk of cancer, etc.). In fact, the perinatal period of immune development is precisely the period in which immune balance must shift from that of an allogeneic yet full-term fetus to that of an offspring ready to meet the complete spectrum of disease challenges. At the heart of the issue is the fact that impact of a xenobiotic on that shift cannot be tested with adult exposure assessment.
Table 1.
Key perinatal immune events | Timing in humans | Benefit to host | Examples of concern | Health ramifications | Key references |
---|---|---|---|---|---|
Differentiation and seeding of macrophages to tissues | 6–24 WG | Self-renewing populations of microglia, Kupffer cells, and alveolar macrophages; resident macrophage functioning in tissues (e.g., testis) | Lead, LPS, ozone, cyclophosphamide | Inflammation of lung, brain, or liver tissue dysfunction (e.g., male infertility) | Cao et al. 2004; Hao et al. 2001; Janossy et al. 1986; Pace et al. 2005 |
Seeding of thymus by pro–T cells and thymopoiesis to expand populations | Seeding 8–12 WG, massive expansion of populations 14–26 WG | Production of T-cell clones necessary to establish peripheral T-lymphocyte populations | PAHs, T-2 toxin, tributyltins, TCDD | Thymic atrophy, decreased postnatal T cells and T-dependent function, increased risk of cancer and infectious diseases | Gehrs and Smialowicz 1997; Holladay and Smith 1994, 1995; Holladay et al. 1993b, 1995; Smialowicz et al. 1989, 1994; Vos et al. 1990; Walker et al. 2004 |
Negative selection and apoptosis of autoreactive thymocytes | 15–26 WG | Elimination of most peripheral T-lymphocyte clones | TCDD promotes unnecessary negative thymocyte selection increasing apoptotic cell death | If promoted, then deceased numbers of thymocytes. If impaired, then, increased risk of later-life self- reactivity | Camacho et al. 2004; Fisher et al. 2004 |
Treg cell (CD4+CD25+ high) population generation in thymus, seeding and activation in periphery | Thymus appearance 12–13 WG; periphery 14–16 WG | Active suppression of postnatal autoreactive T-cell clones | Possible low-dose cyclophosphamide, selected doses of cyclosporin A | If excessively promoted, then possible immune suppression. If impaired, then increased risk of later autoimmunity or allergy (e.g. breaking tolerance to nickel) | Cavani 2005; Cupedo et al. 2005; Darrasse-Jeze et al. 2005; Kawai 2005; Lutsiak et al. 2005; Robinson et al. 2004; Valmori et al. 2005 |
Perinatal dendritic cell maturation to support TH1 responses | Birth–juvenile | Increase in dendritic cell maturation and TH1-promoting capacity after birth to achieve necessary TH1 balance | Dexamethasone, nicotine | Increased risk of allergy and some forms of autoimmunity (e.g., type 1 diabetes) | Andersson et al. 2004; De Wit et al. 2003; Krumbiegel et al. 2005, Mainali and Tew 2004; Nouri-Shirazi and Guinet 2003; Renkl et al. 2005; Skarsvik et al. 2004 |
Increase in TH1 response capacity among peripheral T lymphocytes after birth | Birth–juvenile | Needed to avoid life-long TH2 skewing | Lead, mercury, kynurenines selectively impair TH1 cells, 1-methyl-tryptophan may promote TH1 | With depressed TH1, increased risk of TH2 associated diseases such as atopy and asthma | Bunn et al. 2001b, 2001c; Fallarino et al. 2003; Mellor et al. 2002; Miller et al. 1998; Silva et al. 2005; Snyder et al. 2000 |
Maturation and regulation of fetal macrophages via interactions with surfactants A and D and glutathione sources | 16 WG neonatal period SP-D; 19 WG neonatal period SP-A | Needed to avoid oxidative damage to lung and increased risk of respiratory disease; needed to facilitate parturition, needed to regulate macrophages | Ethanol | Increased risk of childhood respiratory disease; potential problems with labor, increased risk of autoimmune disease | Gauthier et al. 2005; Kaneko et al. 2001; Palaniyar et al. 2005; Pryhuber et al. 1991; Seppanen et al. 2005; Zimmermann et al. 2005 |
Abbreviations: LPS, lipopolysaccharide; PAHs, polycyclic aromatic hydrocarbons; SP-A, surfactant protein A; SP-D, surfactant protein D; T, thymic derived; TCDD, 2,3,7,8-tetra-chlorodibenzo-p-dioxin; TH1, T helper 1; TH2, T helper 2; Treg, T regulatory; WG, weeks of gestation.
Establishing and Renewing Macrophage-Derived Cells in Critical Tissues
One of the early events connecting the immune system to virtually all organs is the differentiation and seeding of myelomonocytic lineage macrophages and macrophage-derived cells to various sites, including the bronchial (e.g., alveolar macrophages), hepatic (Kupffer cells), neurologic (microglia), and reproductive systems (testicular macrophages). These cells provide regulatory and host defense roles in these tissues. Specific examples describe the vulnerability of these tissues during the perinatal period when exposure to toxicants impairs macrophages, including the possibility that the heavy metal lead can impair both the function and the self-renewal of testicular macrophages, which contributes to male sterility problems (Pace et al. 2005). Similarly, pulmonary and alveolar macrophages play a key role in lung development (Beyea et al. 2005), and sensitivity of the perinatal lung to some environmental agents is directly related to alterations in early-life macrophage populations (Cao et al. 2004; Li et al. 2001). In the brain, inappropriate cytokine production from microglial cells and/or astrocytes is now recognized as an early component of many postnatal neurologic diseases (Bell and Hallenbeck 2002; Cacci et al. 2005; Mesples et al. 2005; Ravizza et al. 2005). With Kupffer cells in the liver (Naito et al. 1997), researchers recently found that their capacity to develop a tolerance for lipopolysacharride (LPS) (Uhrig et al. 2005) is critical for the ability of the liver to control inflammation.
Lymphoid seeding of the thymus and thymopoiesis.
Another early immune process critical for subsequent host defense is the migration of pro–T lymphocytes to the thymus and their expansion during thymopoiesis. During the perinatal period, the thymus is central to the production of T lymphocytes. Even in children, the thymus continues to play the major role in T-lymphocyte generation (Mackall et al. 1995; Schonland et al. 2003). In contrast, the thymus has a much different role in the adult. Although the adult thymus retains some capacity for the production of cells, particularly with severe immune depletion, its role remains minor in the repopulation of T lymphocytes (Hakim et al. 2005; Petrie 2002). Instead, most T-lymphocyte production comes from the periphery in adults (Hakim et al. 2005). The ramifications of this are that the targeting of pro–T lymphocytes by chemicals or drugs and/or induction of thymus atrophy would be expected to have different consequences depending upon age. As shown in Table 1, several environmental agents appear to target either pro–T lymphocytes or the thymopoiesis process.
Negative selection in the thymus of auto-reactive T-cell clones.
A third early immune process critical for host integrity is the negative selection and removal of autoreactive T-lymphocyte clones in the thymus. This occurs during the process sometimes designated as “T-cell education.” Meylan et al. (2005) provided a clear demonstration of this process as it occurs in humans. Partially mature thymocytes undergo negative selection at the corticomedullary boundary and in the medulla of thymus (Sprent and Kishimoto 2002). This process is essential if self-reactive clones are to be eliminated before birth. Conversely, excessive loss of thymocytes during negative selection leads to T-cell depletion. Although explicit examples of chemicals or drugs blocking negative selection have yet to be determined, this perinatally unique stage of immune development would appear to be a significant factor in later-life autoimmune disease. Metals represent one category of immunotoxicants warranting examination based on the capacity of mercury and other metals to either trigger or accelerate the progression of autoimmune manifestations (Fournie et al. 2002; Lawrence and McCabe 2002; Rowley and Monestier 2005). Conversely, some chemicals are known to disrupt the negative selection process by overpromoting negative selection and inappropriate thymocyte apoptosis. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is an example of such a toxicant (Camacho et al. 2004; Fisher et al. 2004).
Thymic Generation of Regulatory T-Cells (CD4+CD25+ High Expression) and Acquisition of Tolerance
A parallel protection against self-reactivity resides in the gestational thymic production and subsequent activation of a specialized population of regulatory T lymphocytes. The specialized regulatory T-cell (Treg) population carries the phenotype FoxP3+CD4+CD25+ and develops during gestation (~ 12–13 weeks) in the human fetus (Cupedo et al. 2005; Darrasse-Jeze et al. 2005). Tregs are critical in the suppression of autoreactive lymphocytes that have escaped elimination through negative selection in the thymus (Cupedo et al. 2005; Kronenberg and Rudensky 2005; Sakaguchi and Sakaguchi 2005). It appears that Tregs acquire their regulatory and suppressive phenotype while within the fetal thymus and are further activated in peripheral lymphoid organs (Cupedo et al. 2005). The process through which these cells emerge and acquire the capacity to identify and suppress self-reactive lymphoid clones occurs embryonically. As with negative selection discussed previously, the active perinatal process of producing and activating Tregs is a logical stage where toxicant-induced risk of later-life autoimmune disease would be great. Evidence suggests that some doses of cyclophosphamide (Lutsiak et al. 2005) and cyclosporin A (Kawai et al. 2005) cause inhibition of Treg populations. Treg suppression is thought to be one route to increase allergic disease (Robinson et al. 2004), and the breaking of tolerance to nickel has been associated with Treg suppression (Cavani 2005).
Maturation of dendritic cells from the fetal (immature) phenotype.
Dendritic cells are known to be important in antigen presentation and in determining the nature of subsequent immune responses. A fifth perinatal process involves the maturation of immature dendritic cells to produce interleukin (IL-12) (counterbalancing IL-10 production) and acquire the capability of promoting T-helper 1 (TH1) or type 1 responses. This does not happen in humans until parturition under normal circumstances (Holt and Jones 2000; Holt and Sly 2002).
In fact, the TH1 response must be suppressed until after birth to protect the pregnancy from TH1-mediated immunologic rejection (Lim et al. 2000). One of the processes for accomplishing this is the metabolism of the amino acid tryptophan by the enzyme indoleamine-2,3-dioxygenase (IDO) to produce tryptophan metabolites such as kynurenines (Fallarino et al. 2003; Gutierrez et al. 2003; Meisel et al. 2004; Mellor et al. 2002). These metabolites selectively suppress TH1 function by inducing apoptosis in TH1 but not TH2 cells (Fallarino et al. 2003), thereby skewing responses toward TH2. In the fetus, this is required to avoid allogeneically induced miscarriage (Mellor et al. 2002). But in the newborn this must be corrected to provide adequate immune balance. Not surprisingly, imbalances in IDO activity have been associated with diseases such as colitis (Gurtner et al. 2003) and inflammatory bowel disease (Wolf et al. 2004).
The perinatal system is exquisitely sensitive to these time/life-stage–dependent shifts in immune balance. For example, Malamitsi-Puchner et al. (2005) demonstrated that even the mode of birth delivery can influence the acquisition of TH1 cytokine production capacity in humans. Newborns delivered by cesarean section remained more TH2 skewed compared with vaginally delivered newborns. This emphasizes the potential problems in using an adult exposure assessment protocol for immunotoxicity to model the perinatal immune changes surrounding birth.
In keeping with this idea, a recent study demonstrated that human cord blood–derived dendritic cells respond completely differently than their adult counterparts when exposed to dexamethasone (Mainali and Tew 2004). Dexamethasone exposure of these immature cells prevents their maturation to promote TH1 responses and locks in the TH2 IgE-promoting phenotype. The TH2 skewing effect appears to be long-lasting (Mainali et al. 2005). This type of early-life-stage–restricted immunotoxicity appears to contribute to an increased risk of atopy and asthma. Andersson et al. (2004) showed that maturation of newborn immature dendritic cells with LPS reduced the development of a TH2-associated birch allergen response. In contrast, the lack of dendritic cell maturation from the fetal immature stage was associated with children at risk for type 1 diabetes (Skarsvik et al. 2004). In addition to dexamethasone (Mainali and Tew 2004), nicotine (Nouri-Shirazi and Guinet 2003) has been reported to block dendritic cell maturation. Again, such toxicant-induced perinatal alterations cannot be examined with adult-only exposure because extensive dendritic cell maturation would have occurred after birth and before adult exposure to the test xenobiotic.
Shifting TH balance for later life.
Beyond dendritic cells, some xenobiotics such as the heavy metals and tryptophan metabolites may directly affect TH cells and contribute to skewed immune responses in later life. Because mammals are born with a TH2-skewed functional capacity (Protonotariou et al. 2003), perinatal versus adult exposure assessment actually measures two different alterations. In the perinatal case, the issue is whether a xenobiotic locks in the already existing TH2 bias among T lymphocytes, thereby preventing the genetically influenced adult balance to be achieved postnatally. This would allow as the default an increased risk of neonatal TH2-associated diseases (Holt and Sly 2002). In contrast, under the best circumstances adult exposure assessment could measure only whether a xenobiotic selectively impaired TH1 cells in an already balanced system. At physiologic levels of exposure, many more environmental factors may be capable of delaying or reducing the efficiency of perinatal TH1 maturation (thereby perpetuating the fetal imbalance) than can clinically alter adult TH balance.
Surfactant modulation of macrophages.
Beyond the gestational seeding of macrophages to different tissues and initial maturation in situ, there is a special perinatal maturation of macrophages (particularly alveolar) that enables them to acquire increasing host defense capabilities (phagocytosis, chemotaxis, tumor necrosis factor-α production, antibody-dependent cellular cytotoxicity) with increased postnatal age (Goldman et al. 2004). Hence, perinatal exposure to chemicals and drugs would target functionally immature cells in a manner unlike the fully functional population in exposed adults. Among the critical factors in this perinatal macrophage maturation is exposure to various factors known as “collectins” (surfactant proteins).
Although collectins can be immunomodulatory for adult alveolar macrophages (Hickling et al. 2004), they seem to provide important maturational signals for perinatal macrophages that go well beyond defense of the lung (Mendelson and Condon 2005). Palaniyar et al. (2005) discussed the role of surfactant protein D in enhancing macrophage clearance of DNA and in minimizing anti-DNA antibody production. Additionally, Brinker et al. (2001) demonstrated that surfactant interactions with macrophages and dendritic cells help to shift responses from purely innate to acquired immune responses. Surfactant protein A signals amniotic fluid macrophages to migrate to the uterus and initiate the parturition process (Condon et al. 2004; Mendelson and Condon 2005). Complicating the age issue is the fact that surfactant content varies with age (Egberts et al. 1992). Obviously, such perinatal alterations in macrophage activities are difficult to evaluate using adult-only exposure to potential immunotoxicants.
Although these developmental immune events illustrate the biological problem with modeling immunotoxicologic risk using adult exposures, the resulting underestimation of perinatal sensitivity can take several forms. These are described in the following section.
Nature of Increased Perinatal Immunotoxic Sensitivity
Dose sensitivity.
The increased sensitivity of the developing versus the adult immune system to immunotoxic alteration can take several forms. First, early-life stages have increased dose sensitivity to most toxicants. There are several examples suggesting that the developing immune system is altered by significantly lower doses of toxicants than those required to produce effects in the adult. Such comparisons were recently reviewed in Luebke et al. (2006). Lead (Chen et al. 2004; Heo et al. 1996; McCabe et al. 1999; Miller et al. 1998; Snyder et al. 2000) appears to differ across ages for immunotoxic end points ranging from 3- to 12-fold in lowest observed adverse effect levels (LOAELS). Similarly, mercury appears to have age-based differences (Havarinasab et al. 2004; Hultman and Hansson-Georgiadis 1999; Silva et al. 2005). With TCDD the age-based range in LOAELS appears to be even greater (Gehrs and Smialowicz 1997, 1999; Gehrs et al. 1997; Smialowicz et al. 1994; Walker et al. 2004; see also Table 2).
Table 2.
Chemical/drug | Nature of age-based difference | Reference(s) |
---|---|---|
Benzo[a]pyrene | Severity of effects (e.g., impact of fetalthymic atrophy) | Holladay and Smith 1994; Lummus and Henningsen 1995; Rodriguez et al. 1999; Wolisi et al. 2001 |
Chlordane | Dose sensitivity, spectrum of effects | Barnett et al. 1985; Blaylock et al. 1990; Spyker-Cranmer et al. 1982; Theus et al. 1992 |
Cyclosporin A | Persistence of effects | Hussain et al. 2005b |
Dexamethasone | Dose sensitivity
Spectrum of effects |
Dietert et al. 2003; Mainali and Tew 2004 |
Diazepam | Dose sensitivity
Spectrum/severity of effects |
Descotes et al. 1982; Schlumpf et al. 1989 |
DES | Persistence of effects
Latency |
Fenaux et al. 2004; Kalland and Forsberg 1978; Luster et al. 1980 |
7,12-Dimethybenz[a]anthracene | Severity of effects (e.g., impact of fetal thymic atrophy) | Holladay and Smith 1995 |
Ethanol | Latency, different developmental window effects | Giberson and Weinberg 1995, 1997; Giberson et al. 1997 |
Genistein | Different spectrum of effects | Guo et al. 2002 |
Lead | Dose sensitivity
Differences in spectrum of effects Latency |
Bunn et al. 2001a, 2001b, 2001c, Chen et al. 2004; Faith et al. 1979; Heo et al. 1996; Lee et al. 2001, 2002; McCabe et al. 1999; Miller et al. 1998; Snyder et al. 2000 |
Methoxychlor | Spectrum/severity of effects | White et al. 2005 |
Mercury | Dose sensitivity | Havarinasab et al. 2004; Hultman and Hansson-Georgiadis 1999; Silva et al. 2005 |
Nonylphenol | Spectrum/severity of effects | Karrow et al. 2004 |
Paracetamol | Dose sensitivity | Shaheen et al. 2005 |
T-2 toxin | Severity of effects (e.g., impact of fetal thymic atrophy) | Cooray and Jonsson 1990; Holladay et al. 1993b; Holladay et al. 1995 |
TCDD | Dose sensitivity | Gehrs and Smialowicz 1997, 1999; Gehrs et al. 1997; Smialowicz et al. 1994; Walker et al. 2004 |
Tributyltins | Dose sensitivity
Spectrum/severity of effects |
Tryphonas et al. 2004; Vos et al. 1990;
Smialowicz et al. 1989 |
Range and severity of effects.
A second measure of differential age-based sensitivity to immunotoxicants concerns the spectrum and severity of effects. Not surprisingly, immunotoxicants frequently produce a different and unpredictable array of alterations when the exposure occurs in utero or in the early neonate versus the adult. Among those immunotoxicants that produce different ranges or severities of outcomes depending upon age of exposure are lead (Bunn et al. 2001b; Lee et al. 2001), methoxychlor (White et al. 2005), T-2 toxin (Holladay et al. 1993b), benzo[a]pyrene (Holladay and Smith 1994; Rodriguez et al. 1999), chlordane (Barnett et al. 1985), 7,12-dimethylbenz[a]anthracene (Cooray and Jonsson 1990; Holladay and Smith 1994; Holladay et al. 1995), ethanol (Giberson and Weinberg 1995, 1997; Giberson et al. 1997), nonylphenol (Karrow et al. 2004), tributyltins (Smialowicz et al. 1989; Tryphonas et al. 2004; Vos et al. 1990), and genistein (Guo et al. 2002).
For example, with methoxychlor exposure of rats, F1 males had significantly elevated levels of splenic antibody-forming cells, unlike their exposed mothers, whereas F1 females had a significantly reduced percentage of CD8+ T cells (at all doses examined) with no corresponding effect in the exposed dams (White et al. 2005). Likewise with genistein exposure, exposed F0 rat dams displayed altered natural killer (NK) activity, whereas their daughters exposed in utero had altered antibody-forming cell activity but no change in NK activity (Guo et al. 2002). With TCDD exposure in rats to assess persistent effects, exposed offspring had a significant reduction in contact hypersensitivity with no effect in the exposed dams (Walker et al. 2004). These examples illustrate that adult exposure assessment is inherently ineffective in predicting the range of likely immunotoxic effects after in utero exposure.
Persistence of effects.
Another feature of developmental immunotoxicity is that alterations after early exposure are frequently persistent and last long after exposure, frequently into adulthood of the exposed offspring. Examples where early xenobiotic exposure results in a greater persistence of effects than would be predicted from adult exposure assessment are found with diethylstilbestrol (DES) (Fenaux et al. 2004; Holladay et al. 1993a; Kalland and Forsberg 1978; Luster et al. 1979, 1980) and cyclosporin A (Hussain et al. 2005b).
Latency.
Finally, one of the anomalies of early exposure is that a sublethal exposure to a toxicant may produce an unrecognizable immunotoxic alteration until the postnatal immune system is placed under subsequent stress. This hidden or cryptic state is referred to as “latency.” A classic example exists for early exposure to DES (Fenaux et al. 2004). In this case, an apparently innocuous early exposure to DES alters the immune system in such a manner that it responds to a second adult estrogenic exposure (which of itself has no effect) with a completely aberrant cytokine production profile. The in utero exposure to DES primes the immune system for postnatal unpredictable responses. A similar example has been seen after low-level exposure to lead where postnatal viral infection resulted in unpredictable alterations in leukocyte mobilization (Lee et al. 2002). Obviously, adult exposure assessment affords no opportunity to examine this phenomenon of embryonic-induced immune latency.
Sex Differences in Sensitivity Outcome
Differential immunotoxic effects between sexes are neither universal after early exposure (Voderstrasse et al. 2004) nor unique to early-life stages. However, a surprising number of examples exist in which males and females have different immune outcomes after perinatal xenobiotic exposure. Xenobiotics can have different effects on the developing immune system based on the hormonal environment (Hussain et al. 2005a). Among the chemicals listed in Table 2, gender differences in developmental immunotoxicity have been reported for lead (Bunn et al. 2001a, 2001b, 2001c; Miller et al. 1998), mercury (Silva et al. 2005), genistein (Guo et al. 2002), nonylphenol (Karrow et al. 2004), TCDD (Gehrs and Smialowicz 1999), and methoxychlor (White et al. 2005).
Need for Nonadult Exposure Assessment
Table 2 lists examples of immunotoxicants where age-based comparisons exist and adult exposure assessment is not predictive of perinatal sensitivity to the xenobiotic. Although basic hazard identification could be performed on most of the toxicants listed using only adult exposure data, there would be little guidance for protecting early life stages from problematic exposure of the developing immune system. This is one reason that several recent reviews have suggested the benefit of exposure regimes that include all nonadult (conception, gestation, lactation, juvenile) stages of development (Holsapple et al. 2003; Kimmel et al. 2005; Luster et al. 2005). Recent findings of key maturation events surrounding birth and of chemical- and drug-induced disruption of those immune-associated events (Mainali and Tew 2004; Mainali et al. 2005; Shaheen et al. 2005) are further indications that adult-only exposure protocols are unlikely to accurately predict the risk of perinatal immunotoxicity. Exposure over the nonadult stages of immune development is more likely to include those age-based populations at greatest risk.
Conclusions
Many critical processes occurring during peri-natal immune development are either nonexistent or comparatively unimportant in the adult (e.g., Table 1). Therefore, when safety limits are established on the basis of adult immune exposure data, they likely have limited use for predicting developmental immunotoxicity and protection of the nonadult. For the chemicals and drugs compared across age groups to date, the developing immune system has a greater sensitivity than that of the fully matured adult. Because this increased sensitivity can take different forms (e.g., Table 2), use of magnitude safety factors is of limited benefit in the absence of relevant exposure data. Where adult exposure protocols are used as the only yardstick of immunotoxic safety, consideration should be given to replacing these protocols with exposure regimes extending throughout the nonadult period of development. Given the specific pattern of perinatal immune alterations, it is likely that they explain, in part, the increased incidence of such human diseases as atopy, asthma, and certain autoimmune manifestations.
Footnotes
Support of the authors for research on this topic, including partial salary recovery, was provided by grants or contracts from the National Institute of Environmental Health Sciences [Superfund Basic Research and Education Program via the U.S. Environmental Protection Agency (EPA)], the American Chemistry Council (ACC), the U.S. EPA, and the U.S. Department of Agriculture (USDA).
The ACC and the USDA provided salary support for M.S.P. Research support for R.R.D. during the past 5 years has been from the ACC and the USDA (Developmental Immunotoxicology) and from the USDA and New York State (Environment and Breast Cancer).
References
- Andersson AC, Seppala U, Rudin A. Activation of human neonatal monocyte-derived dendritic cells by lipopolysaccharide down-regulate birch allergen-induced Th2 differentiation. Eur J Immunol. 2004;34:3516–3524. doi: 10.1002/eji.200425544. [DOI] [PubMed] [Google Scholar]
- Barnett JB, Soderberg LSF, Menna JH. The effect of pre-natal chlordane exposure on the delayed hypersensitivity response of Balb/c mice. Toxicol Lett. 1985;25:173–183. doi: 10.1016/0378-4274(85)90079-7. [DOI] [PubMed] [Google Scholar]
- Bell MJ, Hallenbeck JM. Effects of intrauterine inflammation on developing rat brain. J Neurosci Res. 2002;70:570–579. doi: 10.1002/jnr.10423. [DOI] [PubMed] [Google Scholar]
- Beyea JA, Olson DM, Harvey S. Growth hormone expression in the perinatal and postnatal rat lung. Dev Dyn. 2005;232:1037–1046. doi: 10.1002/dvdy.20255. [DOI] [PubMed] [Google Scholar]
- Blaylock BL, Soderberg LSF, Gandy J, Menna JH, Denton R, Barnett JB. Cytotoxic T-lymphocyte and NK responses in mice treated prenatally with chlordane. Toxicol Lett. 1990;51:41–49. doi: 10.1016/0378-4274(90)90223-9. [DOI] [PubMed] [Google Scholar]
- Brinker KG, Martin E, Borron P, Mostaghel E, Doyle C, Harding CV, et al. Surfactant protein d enhances bacterial antigen presentation by bone marrow derived dendritic cells. Am J Physiol Lung Cell Mol Physiol. 2001;281:1253–1563. doi: 10.1152/ajplung.2001.281.6.L1453. [DOI] [PubMed] [Google Scholar]
- Bunn TL, Ladics GS, Holsapple MP, Dietert RR. Developmental immunotoxicology assessment in the rat: age, gender and strain comparisons after exposure to Pb. Toxicol Methods. 2001a;11:41–58. [Google Scholar]
- Bunn TL, Parsons PJ, Kao E, Dietert RR. Exposure to lead during critical windows of embryonic development: differential immunotoxic outcome based on stage of exposure and gender. Toxicol Sci. 2001b;64:57–66. doi: 10.1093/toxsci/64.1.57. [DOI] [PubMed] [Google Scholar]
- Bunn TL, Parsons PJ, Kao E, Dietert RR. Gender-based profiles of developmental immunotoxicity to lead in the rat: assessment in juveniles and adults. J Toxicol Environ Health A. 2001c;64:101–118. doi: 10.1080/15287390152543708. [DOI] [PubMed] [Google Scholar]
- Cacci E, Claasen JH, Kokaia Z. Microglia-derived tumor necrosis factor-alpha exaggerates death of newborn hippocampal progenitor cells in vitro. J Neurosci Res. 2005;80:789–797. doi: 10.1002/jnr.20531. [DOI] [PubMed] [Google Scholar]
- Camacho IA, Nagarkatti M, Nagarkatti PS. Evidence for the induction of apoptotsis in T cells from murine fetal thymus following perinatal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) Toxicol Sci. 2004;78:96–106. doi: 10.1093/toxsci/kfh048. [DOI] [PubMed] [Google Scholar]
- Cao L, Liu C, Cai B, Jia X, Kang L, Speer CP, et al. Nuclear factor-kappa B expression in aveolar macrophage of mechanically ventilated neonate with respiratory distress syndrome. Biol Neonate. 2004;86:116–123. doi: 10.1159/000078940. [DOI] [PubMed] [Google Scholar]
- Cavani A. Breaking tolerance to nickel. Toxicology. 2005;209:119–121. doi: 10.1016/j.tox.2004.12.021. [DOI] [PubMed] [Google Scholar]
- Chen S, Golemboski KA, Piepenbrink M, Dietert RR. Developmental immunotoxicity of lead in the rat: influence of maternal diet. J Toxicol Environ Health A. 2004;67:495–511. doi: 10.1080/15287390490276520. [DOI] [PubMed] [Google Scholar]
- Condon JC, Jeyasuria P, Faust JM, Mendelson CR. Surfactant protein secreted by the maturing mouse fetal lung acts as a hormone that signals the initiation of parturition. Proc Natl Acad Sci USA. 2004;101:4978–4983. doi: 10.1073/pnas.0401124101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cooray R, Jonsson P. Modulation of resistance to mastitis pathogens by pretreatment of mice with T-2 toxin. Food Chem Toxicol. 1990;28:687–692. doi: 10.1016/0278-6915(90)90144-c. [DOI] [PubMed] [Google Scholar]
- Cupedo T, Nagasawa M, Weijer K, Blom B, Spits H. Development and activation of regulatory T cells in the human fetus. Eur J Immunol. 2005;35:383–390. doi: 10.1002/eji.200425763. [DOI] [PubMed] [Google Scholar]
- Darrasse-Jeze G, Marodon G, Salomon BL, Catala M, Klatzmann D. Ontogeny of CD4+CD25+ regulatory/suppressor T cells in human fetuses. Blood. 2005;105:4715–4721. doi: 10.1182/blood-2004-10-4051. [DOI] [PubMed] [Google Scholar]
- Daston G, Faustman E, Ginsberg G, Fenner-Crisp P, Olin S, Sonawane B, et al. A framework for assessing risks to children from exposure to environmental agents. Environ Health Perspect. 2004;112:238–256. doi: 10.1289/ehp.6182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Descotes J, Tedone R, Evreux JC. Suppression of humoral and cellular immunity in normal mice by diazepam. Immunol Lett. 1982;5:41–43. doi: 10.1016/0165-2478(82)90089-x. [DOI] [PubMed] [Google Scholar]
- De Wit D, Tonon S, Olislagers V, Goriely S, Boutriaux M, Goldman M, et al. Impaired responses to toll-like receptor and toll-like receptor 3 ligands in human cord blood. J Autoimmunity. 2003;21:277–281. doi: 10.1016/j.jaut.2003.08.003. [DOI] [PubMed] [Google Scholar]
- Dietert RR. New developments in the assessment of developmental immunotoxicology. J Immunotoxicol. 2005;2:185–190. doi: 10.1080/15476910500362788. [DOI] [PubMed] [Google Scholar]
- Dietert RR, Etzel RA, Chen D, Halonen M, Holladay SD, Jarabek AM, et al. Workshop to identify critical windows of exposure for children’s health: immune and respiratory systems work groups summary. Environ Health Perspect. 2000;108(suppl 3):483–490. doi: 10.1289/ehp.00108s3483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dietert RR, Lee J-E, Bunn TL. Developmental immunotoxicology: emerging issues. Hum Exp Toxicol. 2002;21:479–485. doi: 10.1191/0960327102ht285oa. [DOI] [PubMed] [Google Scholar]
- Dietert RR, Lee J-E, Olsen J, Fitch K, Marsh JA. Developmental immunotoxicity of dexamethasone: comparison of fetal versus adult exposures. Toxicology. 2003;196:163–176. doi: 10.1016/j.tox.2003.07.001. [DOI] [PubMed] [Google Scholar]
- Egberts J, Sprengers BM, Sietaram MA. Comparison of pumonary surfactant content in alveolar macrophages of newborn, young and adult rats. Exp Lung Res. 1992;18:275–285. doi: 10.3109/01902149209031685. [DOI] [PubMed] [Google Scholar]
- Faith RE, Luster MI, Kimmel CA. Effect of chronic developmental lead exposure on cell-mediated immune functions. Clin Exp Immunol. 1979;35:413–420. [PMC free article] [PubMed] [Google Scholar]
- Fallarino F, Grohmann U, Vacca C, Orabona C, Spreca A, Fioretti MC, et al. T cell apoptosis by kynurenines. Adv Exp Med Biol. 2003;527:183–190. doi: 10.1007/978-1-4615-0135-0_21. [DOI] [PubMed] [Google Scholar]
- Fenaux JB, Gogal RM, Jr, Ahmed SA. Diethylstilbesterol exposure during fetal development affects thymus: studies in fourteen-month-old mice. J Reprod Immunol. 2004;64:75–90. doi: 10.1016/j.jri.2004.08.003. [DOI] [PubMed] [Google Scholar]
- Fisher MT, Nagarkatti M, Nagarkatti PS. Combined screening of thymocytes using apoptosis-specific cDNA array and promoter analysis yields novel gene targets mediating TCDD-induced toxicity. Toxicol Sci. 2004;78:116–124. doi: 10.1093/toxsci/kfh058. [DOI] [PubMed] [Google Scholar]
- Fournie GJ, Saoudi A, Druet P, Pelletier L. Th-2 type immunopathological manifestations induced by mecuric chloride or gold salts in the rat: signal transduction pathways, cellular mechanisms and genetic control. Autoimmun Rev. 2002;1:205–212. doi: 10.1016/s1568-9972(02)00052-6. [DOI] [PubMed] [Google Scholar]
- Gauthier TW, Ping XD, Harris FL, El Bahesh H, Brown LA. Fetal alcohol exposure impairs alveolar macrophage function via decreased glutathione availability. Pediatr Res. 2005;57:76–81. doi: 10.1203/01.PDR.0000149108.44152.D3. [DOI] [PubMed] [Google Scholar]
- Gehrs BC, Riddle MM, Williams WC, Smialowicz RJ. Alterations in the developing immune system of the F344 rat after perinatal exposure to 2,3,7,8-tetraclorobenzo-p-dioxin. II. Effects on the pup and the adult. Toxicology. 1997;122:229–240. doi: 10.1016/s0300-483x(97)00099-1. [DOI] [PubMed] [Google Scholar]
- Gehrs BC, Smialowicz RJ. Alteration in the developing immune system of the F344 rat after perinatal exposure to 2,3,7,8-tetrachlorodibenzo-p-doxin. I. Effects on the fetus and the neonate. Toxicology. 1997;122:219–228. doi: 10.1016/s0300-483x(97)00098-x. [DOI] [PubMed] [Google Scholar]
- Gehrs BC, Smialowicz RJ. Persistent suppression of delayed-type hypersensitivity in adult F344 rats after perinatal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicology. 1999;134:70–88. doi: 10.1016/s0300-483x(99)00024-4. [DOI] [PubMed] [Google Scholar]
- Giberson PK, Kim CK, Hutchison S, Yu W, Junker A, Weinberg J. The effect of cold stress on lymphocyte proliferation in fetal ethanol-exposed rats. Alcohol Clin Exp Res. 1997;21:1440–1447. [PubMed] [Google Scholar]
- Giberson PK, Weinberg J. Effects of prenatal ethanol exposure and stress in adulthood on lymphocyte populations in rats. Alcohol Clin Exp Res. 1995;19:1286–1294. doi: 10.1111/j.1530-0277.1995.tb01613.x. [DOI] [PubMed] [Google Scholar]
- Giberson PK, Weinberg J. Effect of surrogate fostering on splenic lymphocytes in fetal ethanol exposed rats. Alcohol Clin Exp Res. 1997;21:44–55. [PubMed] [Google Scholar]
- Goldman A, Rubin C, Gomez S, Palermo MS, Tasat DR. Functional age-dependent changes in bronchoalveolar lavage rat cells. Cell Mol Biol. 2004;50:649–655. [PubMed] [Google Scholar]
- Guo TL, White KL, Jr, Brown RD, Declos KB, Newbold RR, Weis C, et al. Genistein modulates splenic natural killer cell activity, antibody forming cell response, and phenotypic marker expression in F0 and F1 generations of Sprague-Dawley rats. Toxicol Appl Pharmacol. 2002;181:219–227. doi: 10.1006/taap.2002.9418. [DOI] [PubMed] [Google Scholar]
- Gurtner GJ, Newberry RD, Schloemann SR, McDonald KG, Stenson WF. Inhibition of indoleamine 2,3-dioxygenase augments trinitrobenzene sulfonic acid colitis in mice. Gastroenterology. 2003;125:1762–1773. doi: 10.1053/j.gastro.2003.08.031. [DOI] [PubMed] [Google Scholar]
- Gutierrez G, Fitzgerald JS, Pohlmann T, Hoppe I, Markert UR. Comparative effects of l-tryptophan and 1-methyl-trytophan on immunoregulation induced by sperm, human pre-implantation embryo and trophoblast supernatants. Am J Reprod Immunol. 2003;50:309–315. doi: 10.1034/j.1600-0897.2003.00093.x. [DOI] [PubMed] [Google Scholar]
- Hakim FT, Memon SA, Cepeda R, Jones EC, Chow CK, Kasten-Sportes C, et al. Age-dependent incidence, time course, and consequences of thymic renewal in adults. J Clin Invest. 2005;115:930–039. doi: 10.1172/JCI22492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hao AJ, Dheen ST, Ling EA. Response of amoeboid microglia/brain macrophages in fetal rat brain exposed to a teratogen. J Neurosci Res. 2001;64:79–93. doi: 10.1002/jnr.1056. [DOI] [PubMed] [Google Scholar]
- Havarinasab S, Lambertsson L, Qvarnstrom J, Hultman P. Dose-reponse study of thimerosal-induced murein systemic autoimmunity. Toxicol Appl Pharmacol. 2004;194:169–179. doi: 10.1016/j.taap.2003.09.006. [DOI] [PubMed] [Google Scholar]
- Heo Y, Parsons PJ, Lawrence DA. Lead differentially modifies cytokine production in vitro and in vivo. Toxicol Appl Pharmacol. 1996;138:149–157. doi: 10.1006/taap.1996.0108. [DOI] [PubMed] [Google Scholar]
- Hickling TP, Clark H, Mahotra R, Sim RB. Collectins and their role in lung immunity. J Leuk Biol. 2004;75:27–33. doi: 10.1189/jlb.0703304. [DOI] [PubMed] [Google Scholar]
- Hinton DM. US FDA “Redbook II” immunotoxicity testing guidelines and research in immunotoxicity evaluations of food chemicals and new food proteins. Toxicol Pathol. 2000;28:467–478. doi: 10.1177/019262330002800318. [DOI] [PubMed] [Google Scholar]
- Holladay SD. Prenatal immunotoxicant exposure and postnatal autoimmune disease. Environ Health Perspect. 1999;107:687–691. doi: 10.1289/ehp.99107s5687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holladay SD, Blaylock BL, Comment CE, Heindel JJ, Fox WM, Korach KS, et al. Selective prothymocyte targeting by prenatal diethylstilbesterol exposure. Cell Immunol. 1993a;152:131–142. doi: 10.1006/cimm.1993.1273. [DOI] [PubMed] [Google Scholar]
- Holladay SD, Blaylock BL, Comment CE, Heindel JJ, Luster MI. Fetal thymic atrophy after exposure to T-2 toxin: selectivity for lymphoid progenitor cells. Toxiol Appl Pharmacol. 1993b;121:8–14. doi: 10.1006/taap.1993.1122. [DOI] [PubMed] [Google Scholar]
- Holladay SD, Smialowicz RJ. Development of the murine and human immune system: differential effects of immunotoxicants depend on time of exposure. Environ Health Perspect. 2000;108:463–473. doi: 10.1289/ehp.00108s3463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holladay SD, Smith BJ. Fetal hematopoietic alterations after maternal exposure to benzo[a]pyrene: a cytometric evaluation. J Toxicol Environ Health. 1994;42:259–273. doi: 10.1080/15287399409531878. [DOI] [PubMed] [Google Scholar]
- Holladay SD, Smith BJ. Alterations in murine fetal thymus and liver hematopoietic cell populations following developmental exposure to 7,12-dimethylbenz[a]anthracene. Environ Res. 1995;68:106–113. doi: 10.1006/enrs.1995.1014. [DOI] [PubMed] [Google Scholar]
- Holladay SD, Smith BJ, Luster MI. B lymphocyte precursor cells represent sensitive targets of T-2 mycotoxin exposure. Toxicol Appl Pharmacol. 1995;131:309–315. doi: 10.1006/taap.1995.1073. [DOI] [PubMed] [Google Scholar]
- Holsapple MP, Burn-Naas LA, Hastings KL, Ladics GS, Lavin GS, Makris SL, et al. A proposed testing framework for developmental immunotoxicology (DIT) Toxicol Sci. 2005;83:18–24. doi: 10.1093/toxsci/kfh299. [DOI] [PubMed] [Google Scholar]
- Holsapple MP, West LJ, Landreth KS. Species comparison of anatomical and functional immune system development. Birth Defects Res B. 2003;68:321–334. doi: 10.1002/bdrb.10035. [DOI] [PubMed] [Google Scholar]
- Holt PG, Jones CA. The development of the immune system during pregnancy and early life. Allergy. 2000;55:688–697. doi: 10.1034/j.1398-9995.2000.00118.x. [DOI] [PubMed] [Google Scholar]
- Holt PG, Sly PD. Interactions between RSV infections, asthma, and atopy: unravelling the complexities. J Exp Med. 2002;196:1271–1275. doi: 10.1084/jem.20021572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hultman P, Hansson-Georgiadis H. Methyl mercury-induced autoimmunity in mice. Toxicol Appl Pharmacol. 1999;154:203–211. doi: 10.1006/taap.1998.8576. [DOI] [PubMed] [Google Scholar]
- Hussain I, Piepenbrink MS, Dietert RR. Impact of in ovo-administered lead and testosterone on developing female thymocytes. J Toxicol Environ Health A. 2005a;68:1309–1319. doi: 10.1080/15287390590953554. [DOI] [PubMed] [Google Scholar]
- Hussain I, Piepenbrink MS, Fitch KJ, Marsh JA, Dietert RR. Developmental immunotoxicity of cyclosporin-A in rats: age-associated differential effects. Toxicology. 2005b;206:273–284. doi: 10.1016/j.tox.2004.08.019. [DOI] [PubMed] [Google Scholar]
- Janossy G, Bofill M, Poulter LW, Rawlings E, Burford GD, Navarrete C, et al. Separate ontogeny of two macrophage-like accessory cell populations in the human fetus. J Immunol. 1986;136:4354–4361. [PubMed] [Google Scholar]
- Kalland T, Forsberg JG. Delayed hypersensitivity response to oxazolone in neonatally estrogenized mice. Cancer Lett. 1978;4:141–146. doi: 10.1016/s0304-3835(78)94017-x. [DOI] [PubMed] [Google Scholar]
- Kaneko K, Shimizu H, Arakawa H, Ogawa Y. Pulmonary surfactant protein A in sera for assessing neonatal lung maturation. Early Hum Dev. 2001;62:11–21. doi: 10.1016/s0378-3782(00)00133-x. [DOI] [PubMed] [Google Scholar]
- Karrow NA, Guo TL, Declos KB, Newbold RR, Weis C, Germolec DR, et al. Nonylphenol alters the activity of splenic NK cells and the number of leukocyte subpopulations in Sprague-Dawley rats: a two generation feeding study. Toxicology. 2004;196:237–245. doi: 10.1016/j.tox.2003.11.009. [DOI] [PubMed] [Google Scholar]
- Kawai M, Kitade H, Mathieu C, Waer M, Pirenne J. Inhibitory and stimulatory effects of cyclosporine A on the development of regulatory T cells in vivo. Transplantation. 2005;79:1073–1077. doi: 10.1097/01.tp.0000153505.73700.32. [DOI] [PubMed] [Google Scholar]
- Kimmel CA, King MD, Makris SL. 2005. Risk assessment perspectives for developmental immunotoxicology. In: Developmental Immunotoxicology (Holladay SD, ed). Boca Raton, FL:CRC Press, 45–63.
- Kimmel GL. An overview of children as a special population-relevance to predictive biomarkers. Toxicol Appl Pharmacol. 2005;206:215–218. doi: 10.1016/j.taap.2004.06.032. [DOI] [PubMed] [Google Scholar]
- Kronenberg M, Rudensky A. Regulation of immunity by self reactive T cells. Nature. 2005;435:598–604. doi: 10.1038/nature03725. [DOI] [PubMed] [Google Scholar]
- Krumbiegel D, Rohr J, Schmidtke P, Knuf M, Zepp F, Meyer CU. Efficient maturation and cytokine production of neonatal DCs requires combined proinflammatory cytokine signals. Clin Dev Immunol. 2005;12:99–105. doi: 10.1080/17402520500116772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ladics GS, Chapin RE, Hastings KL, Holsapple MP, Makris SL, Sheet LP, et al. Developmental toxicology evaluations—issues including neurotoxicology and immunotoxicology assessments in reproductive toxicology studies. Toxicol Sci. 2005;88:24–29. doi: 10.1093/toxsci/kfi299. [DOI] [PubMed] [Google Scholar]
- Landreth KS. Critical windows in development of the rodent immune system. Hum Exp Toxicol. 2002;21:493–498. doi: 10.1191/0960327102ht287oa. [DOI] [PubMed] [Google Scholar]
- Landreth KS, Dodson SVM. 2005. Development of the rodent immune system. In: Developmental Immunotoxicology (Hollday SD, ed). Boca Raton, FL:CRC Press, 3–19.
- Landrigan PL, Kimmel CA, Correa A, Eskenazi B. Children’s health and the environment: public health issues and challenges for risk assessment. Environ Health Perspect. 2004;112:257–265. doi: 10.1289/ehp.6115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lawrence DA, McCabe MJ., Jr Immunomodulation by metals. Int Immunopharmacol. 2002;2:293–302. doi: 10.1016/s1567-5769(01)00180-1. [DOI] [PubMed] [Google Scholar]
- Lee J-E, Chen S, Golemboski KA, Parsons PJ, Dietert RR. Developmental windows of differential lead-induced immunotoxicity in chickens. Toxicology. 2001;156:161–170. doi: 10.1016/s0300-483x(00)00350-4. [DOI] [PubMed] [Google Scholar]
- Lee J-E, Naqi SA, Kao E, Dietert RR. Embryonic exposure to lead: comparison of immune and cellular responses in unchallenged and virally stressed chickens. Arch Toxicol. 2002;75:717–724. doi: 10.1007/s00204-001-0283-9. [DOI] [PubMed] [Google Scholar]
- Leibnitz R. 2005. Development of the human immune system. In: Developmental Immunotoxicology (Hollday SD, ed). Boca Raton, FL:CRC Press, 21–42.
- Li YH, Yan ZQ, Brauner A, Tullus K. Meconium undces expression of inducible no synthase and activation of NF-kappaB in rat alveolar macrophages. Pediatr Res. 2001;49:820–825. doi: 10.1203/00006450-200106000-00018. [DOI] [PubMed] [Google Scholar]
- Lim KJ, Odukoya OA, Ajjan RA, Li TC, Weetman AP, Cooke ID. The role of T-helper cytokines in human reproduction. Fertil Steril. 2000;73:136–142. doi: 10.1016/s0015-0282(99)00457-4. [DOI] [PubMed] [Google Scholar]
- Luebke RW, Chen DH, Dietert RR, Yang Y, King M. The comparative immunotoxicity of five selected compounds following developmental or adult exposure. J Toxicol Environ Health B Crit Rev. 2006;9:1–26. doi: 10.1080/15287390500194326. [DOI] [PubMed] [Google Scholar]
- Lummus ZL, Henningsen G. Modulation of T-cell ontogeny by transplacental benzo(a)pyrene. Int J Immunopharmacol. 1995;17:339–350. doi: 10.1016/0192-0561(94)00098-9. [DOI] [PubMed] [Google Scholar]
- Luster MI, Boorman GA, Dean JH, Luebke RW, Lawson LD. The effect of adult exposure to diethylstilbesterol in the mouse: alterations in imunological functions. J Reticuloendothel Soc. 1980;28:561–569. [PubMed] [Google Scholar]
- Luster MI, Faith RE, McLauchlan JA, Clark GC. Effect of in utero exposure to diethylstilbesterol on the immune response in mice. Toxicol Appl Pharmacol. 1979;47:279–285. doi: 10.1016/0041-008x(79)90322-3. [DOI] [PubMed] [Google Scholar]
- Luster MI, Johnson VJ, Yucesoy B, Simeonova P. Biomarkers to assess potential developmental immunotoxicity in children. Toxicol Appl Pharmacol. 2005;206:229–236. doi: 10.1016/j.taap.2005.02.010. [DOI] [PubMed] [Google Scholar]
- Luster MI, Portier C, Pait DG, White KLJ, Gennings C, Munson AE, et al. Risk assessment in immunotoxicology. I. Sensitivity and predictability of immune tests. Fundam Appl Toxicol. 1992;18:200–210. doi: 10.1016/0272-0590(92)90047-l. [DOI] [PubMed] [Google Scholar]
- Lutsiak ME, Semnani RT, De Pascalis R, Kashmiri SV, Schlom J, Sabzevari H. Inhibition of CD4+CD25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide. Blood. 2005;105:2862–2868. doi: 10.1182/blood-2004-06-2410. [DOI] [PubMed] [Google Scholar]
- MacKall CL, Fleisher TA, Brown MR, Andrich MP, Chen CC, Feuerstein IM, et al. Age, thymopoiesis, and CD4+ T lymphocyte regeneration after intensive chemotherapy. N Engl J Med. 1995;332:143–149. doi: 10.1056/NEJM199501193320303. [DOI] [PubMed] [Google Scholar]
- Mainali ES, KiKuchi T, Tew JG. Dexamethasone inhibits maturation and alters function of monocyte-derived dendritic cells from cord blood. Pediatr Res. 2005;58:125–131. doi: 10.1203/01.PDR.0000157677.72136.09. [DOI] [PubMed] [Google Scholar]
- Mainali ES, Tew JG. Dexamethasone selectively inhibits differentiation of cord blood stem cell derived-dendritic cells (DC) precursors into mature DCs. Cell Immunol. 2004;232:127–136. doi: 10.1016/j.cellimm.2005.03.002. [DOI] [PubMed] [Google Scholar]
- Malamitsi-Puchner A, Protonotariou E, Boutsikou T, Makrakis E, Sarandakou A, Creatas G. The influence of the mode of delivery on circulating cytokine concentration in the perinatal period. Early Hum Dev. 2005;81:387–392. doi: 10.1016/j.earlhumdev.2004.10.017. [DOI] [PubMed] [Google Scholar]
- McCabe MJ, Jr, Singh KP, Reiners JJ., Jr Lead intoxication impairs the generation of a delayed type hypersensitivity response. Toxicology. 1999;139:255–264. doi: 10.1016/s0300-483x(99)00147-x. [DOI] [PubMed] [Google Scholar]
- Meisel R, Zibert A, Laryea M, Gobel U, Daubener W, Dilloo D. Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation. Blood. 2004;103:4619–4623. doi: 10.1182/blood-2003-11-3909. [DOI] [PubMed] [Google Scholar]
- Mellor AL, Chandler P, Lee GK, Johnson T, Keskin DB, Lee J, et al. Indoleamine 2,3,-dioxygenase, immunosuppression and pregnancy. J Reprod Immunol. 2002;57:143–150. doi: 10.1016/s0165-0378(02)00040-2. [DOI] [PubMed] [Google Scholar]
- Mendelson CR, Condon JC. New insights into the molecular endocrinology of parturition. J Steroid Biochem Mol Biol. 2005;93:113–119. doi: 10.1016/j.jsbmb.2004.12.027. [DOI] [PubMed] [Google Scholar]
- Mesples B, Plaisant F, Fontaine RH, Gressens P. Pathophysiology of neonatal brain lesions: lesson from animal models of excitotoxicity. Acta Paediatr. 2005;94:185–190. doi: 10.1111/j.1651-2227.2005.tb01888.x. [DOI] [PubMed] [Google Scholar]
- Meylan F, De Smedt M, LeClercq G, Plum J, Leupin O, Marguerat S, et al. Negative thymocyte selection to HERV-K18 superantigens in humans. Blood. 2005;105:4377–4382. doi: 10.1182/blood-2004-07-2596. [DOI] [PubMed] [Google Scholar]
- Miller TE, Golemboski KA, Ha RS, Bunn T, Sanders FS, Dietert RR. Developmental exposure to lead causes persistent immunotoxicity in Fischer 344 rats. Toxicol Sci. 1998;42:129–135. doi: 10.1006/toxs.1998.2424. [DOI] [PubMed] [Google Scholar]
- Naito M, Hasegawa G, Takahashi K. Development, differentiation, and maturation of Kupffer cells. Microsc Res Tech. 1997;39:350–364. doi: 10.1002/(SICI)1097-0029(19971115)39:4<350::AID-JEMT5>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
- Nouri-Shirazi M, Guinet E. Evidence for the immunosuppressive role nicotine has on human dendritic cell function. Immunology. 2003;109:365–273. doi: 10.1046/j.1365-2567.2003.01655.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pace BM, Lawrence DA, Behr MJ, Parsons PJ, Dias JA. Neonatal lead exposure changes quality of sperm and number of macrophages in testes of BALB/c mice. Toxicology. 2005;210:247–256. doi: 10.1016/j.tox.2005.02.004. [DOI] [PubMed] [Google Scholar]
- Palaniyar N, Clark H, Nadesalingam J, Shih MJ, Hawgood S, Reid KB. Innate immune collectin surfactant protein D enhances the clearance of DNA by macrophages and minimizes anti-DNA antibody generation. J Immunol. 2005;174:7352–7358. doi: 10.4049/jimmunol.174.11.7352. [DOI] [PubMed] [Google Scholar]
- Petrie HT. Role of thymic organ structure and stromal composition in steady-state postnatal T-cell production. Immunologic Rev. 2002;189:8–19. doi: 10.1034/j.1600-065x.2002.18902.x. [DOI] [PubMed] [Google Scholar]
- Protonotariou E, Malamitsi-Puchner A, Rizos D, Sarandakou A, Makrakis E, Salamolekis E. Alterations in TH1/TH2 cytokine concentrations in early neonatal life. J Matern Fetal Neonatal Med. 2003;14:407–410. doi: 10.1080/14767050412331312270. [DOI] [PubMed] [Google Scholar]
- Pryhuber GS, Hull WM, Fink I, McMahan MJ, Whitsett JA. Ontogeny of surfactant proteins A and B in human amniotic fluid as indices of fetal lung maturity. Pediatr Res. 1991;30:597–605. doi: 10.1203/00006450-199112000-00023. [DOI] [PubMed] [Google Scholar]
- Ravizza T, Rizzi M, Perego C, Richichi C, Veliskova J, Moshe SL, et al. Inflammatory response and glia activation in developing rat hippocampus after status epilepticus. Epilepsia. 2005;46:113–117. doi: 10.1111/j.1528-1167.2005.01006.x. [DOI] [PubMed] [Google Scholar]
- Renkl AC, Wussler J, Ahrens T, Thoma K, Kon S, Uede T, et al. Osteoponin functionally activates dendritic cells and induces their differentiation toward a Th1-polarizing pheno type. Blood. 2005;106:946–955. doi: 10.1182/blood-2004-08-3228. [DOI] [PubMed] [Google Scholar]
- Robinson DS, Larche M, Durham SR. Tregs and allergic disease. J Clin Invest. 2004;114:1389–1397. doi: 10.1172/JCI23595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodriguez JW, Kirlin WG, Wirsiy YG, Matheravidathu S, Hodge TW, Urso P. Maternal exposure to benzo[a]-pyrene alters development of T lymphocytes in offspring. Immunopharmacol Immunotoxicol. 1999;21:379–396. doi: 10.3109/08923979909052769. [DOI] [PubMed] [Google Scholar]
- Rowley B, Monestier M. Mechanisms of heavy metal-induced autoimmunity. Mol Immunol. 2005;42:833–838. doi: 10.1016/j.molimm.2004.07.050. [DOI] [PubMed] [Google Scholar]
- Sakaguchi S, Sakaguchi N. Regulatory T cells in immunologic self-tolerance and autoimmune disease. Int Rev Immunol. 2005;24:3–4. doi: 10.1080/08830180590934976. [DOI] [PubMed] [Google Scholar]
- Schlumpf M, Ramseier H, Lichtensteiger W. Prenatal diazepam induced persisting depression of cellular immune responses. Life Sci. 1989;44:493–501. doi: 10.1016/0024-3205(89)90465-7. [DOI] [PubMed] [Google Scholar]
- Schonland SO, Zimmer JK, Lopez-Benitez CM, Widmann T, Ramin KD, Goronzy JJ, et al. Homeostatic control of T cell generation in neonates. Blood. 2003;102:1428–1434. doi: 10.1182/blood-2002-11-3591. [DOI] [PubMed] [Google Scholar]
- Selevan SG, Kimmel CA, Mendola P. Identifying critical windows of exposure for children’s health. Environ Health Perspect. 2000;108:451–455. doi: 10.1289/ehp.00108s3451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seppanen O, Glumoff V, Paananen R, Rounioja S, Hallman M. Transcription factors NF-kappaC and C/EBPdelta and IL-1-induced expression of surfactant protein a in lung explants during perinatal period. Biol Neonate. 2005;87:152–159. doi: 10.1159/000082312. [DOI] [PubMed] [Google Scholar]
- Shaheen SO, Newson RB, Henderson AJ, Headley JE, Stratton FD, Jones RW, et al. Prenatal paracetamol exposure and risk of asthma and elevated immunoglobulin E in childhood. Clin Exp Allergy. 2005;35:18–25. doi: 10.1111/j.1365-2222.2005.02151.x. [DOI] [PubMed] [Google Scholar]
- Silva IA, El Nabawi M, Hoover D, Silbergeld EK. Prenatal HgCl2 exposure in BALB/c mice: gender specific effects on the ontogeny of the immune system. Dev Comp Immunol. 2005;29:171–183. doi: 10.1016/j.dci.2004.05.008. [DOI] [PubMed] [Google Scholar]
- Skarsvik S, Tittanen M, Lindstrom A, Casas R, Ludvigsson J, Vaarala O. Poor in vitro maturation and pro-inflammatory cytokine response of dendritic cells in children at genetic risk of type 1 diabetes. Scan J Immunol. 2004;60:647–652. doi: 10.1111/j.0300-9475.2004.01521.x. [DOI] [PubMed] [Google Scholar]
- Smialowicz RJ, Riddle MM, Rogers RR, Luebke RW, Copeland CB. Immunotoxicity of tributyltin oxide in rats exposed as adults or pre-weanlings. Toxicology. 1989;57:97–111. doi: 10.1016/0300-483x(89)90037-1. [DOI] [PubMed] [Google Scholar]
- Smialowicz RJ, Riddle MM, Williams WC, Dilberto JJ. Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on humoral immunity and lymphocyte subpopulations: difference between mice and rats. Toxicol Appl Pharmacol. 1994;124:248–256. doi: 10.1006/taap.1994.1029. [DOI] [PubMed] [Google Scholar]
- Snyder JE, Filipov NM, Parsons PJ, Lawrence DA. The efficiency of maternal transfer of lead and its influence on plasma IgE and splenic cellularity of mice. Toxicol Sci. 2000;57:87–94. doi: 10.1093/toxsci/57.1.87. [DOI] [PubMed] [Google Scholar]
- Sprent J, Kishimoto H. The thymus and negative selection. Immunol Rev. 2002;185:126–135. doi: 10.1034/j.1600-065x.2002.18512.x. [DOI] [PubMed] [Google Scholar]
- Spyker-Cranmer JM, Barnett JB, Avery DL, Cranmer MF. Immunoteratology of chlordane: cell-mediated and humoral immune responses in adult mice exposed in utero. Toxicol Appl Pharmacol. 1982;62:402–408. doi: 10.1016/0041-008x(82)90141-7. [DOI] [PubMed] [Google Scholar]
- Theus SA, Lau KA, Tabor DR, Soderberg LSF, Barnett JB. In vivo prenatal chlordane exposure induces development of endogenous inflammatory macrophages. . J Leukoc Biol. 1992;51:366–372. doi: 10.1002/jlb.51.4.366. [DOI] [PubMed] [Google Scholar]
- Tryphonas H, Cooke G, Caldwell D, Bondy G, Parenteau M, Hayward S, et al. Oral (gavage), in utero and postnatal exposure of Sprague-Dawley rats to low doses of tributyltin chloride. Part II: Effects on the immune system. Food Chem Toxicol. 2004;42:221–235. doi: 10.1016/j.fct.2003.08.019. [DOI] [PubMed] [Google Scholar]
- Uhrig A, Banafsche R, Kremer M, Hengenbarth S, Hamann A, Neurath M, et al. Development and functional consequences of LPS tolerance in sinusoidal endothelial cells of the liver. J Leukoc Biol. 2005;77:626–633. doi: 10.1189/jlb.0604332. [DOI] [PubMed] [Google Scholar]
- U.S. Food and Drug Administration 1999. Guidance for Industry and FDA Reviewers—Immunotoxicity Testing Guidance. Washington, DC:Center for Devices and Radiological Health, U.S. Food and Drug Administration.
- U.S. Food and Drug Administration 2004. S8 Immunotoxicity Studies for Human Pharmaceuticals. Draft Guidance. November 18 2004. Washington, DC:U.S. Food and Drug Administration.
- Valmori D, Merlo A, Souleimanian NE, Hesdorffer CS, Ayyoub M. A peripheral circulating compartment of naive CD4 Tregs. J Clin Invest. 2005;115:1953–1962. doi: 10.1172/JCI23963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Loveren H, Piersma A. Immunotoxicological consequences of perinatal chemical exposures. Toxicol Lett. 2004;149:141–145. doi: 10.1016/j.toxlet.2003.12.028. [DOI] [PubMed] [Google Scholar]
- Voderstrasse B, Cundiff J, Lawrence B. Developmental exposure to aryl hydrocarbon receptor agonist 2,3,7,8-tetra-chkorodibenzo-p-doxin impairs the cell-mediated immune response to infection with influenza A virus, but enhances elements of innate immunity. J Immunotoxicol. 2004;1:103–112. doi: 10.1080/15476910490509244. [DOI] [PubMed] [Google Scholar]
- Vos JG, De Klerk A, Krajnc EI, Van Loveren H, Rozing J. Immunotoxicity of bis (tri-n-butyltin) oxide in the rat: effect on thymus-dependent immunity and on nonspecific resistance following long-term exposure in young versus aged rats. Toxicol Appl Pharmacol. 1990;105:144–155. doi: 10.1016/0041-008x(90)90366-3. [DOI] [PubMed] [Google Scholar]
- Walker DB, Williams WC, Copeland CB, Smialowicz RJ. Persistent suppression of contact hypersensitivity, and altered T-cell parameters in F344 rats exposed perinatally to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) Toxicology. 2004;197:57–66. doi: 10.1016/j.tox.2003.12.012. [DOI] [PubMed] [Google Scholar]
- White KL, Jr, Germolec DR, Booker CD, Hernendez DM, McCay JA, Declos KB, et al. Dietary methoxychlor exposure modulates splenic natural killer cell activity, antibody-forming cell response and phenotypic marker expression in F0 and F1 generations of Sprague Dawley rats. Toxicology. 2005;207:271–281. doi: 10.1016/j.tox.2004.09.011. [DOI] [PubMed] [Google Scholar]
- Wolf AM, Wolf D, Rumpold H, Moschen AR, Kaser A, Obrist P, et al. Overexpression of indoleamine 2,3-dioxygenase in human inflammatory bowel disease. Clin Immunol. 2004;113:47–55. doi: 10.1016/j.clim.2004.05.004. [DOI] [PubMed] [Google Scholar]
- Wolisi GO, Majekodunmi J, Bailey GB, Urso P. Immunomodulation in progeny from thymectomized primiparous mice exposed to benzo[a]pyrene during mid-pregnancy. Immunopharmacol Immunotoxicol. 2001;23:267–280. doi: 10.1081/iph-100103865. [DOI] [PubMed] [Google Scholar]
- Zimmerman LJI, Janssen DJMT, Tibboel D, Hamvas A, Carnielli VP. Surfactant metabolism in the neonate. Biol Neonate. 2005;87:296–307. doi: 10.1159/000084877. [DOI] [PubMed] [Google Scholar]