Skip to main content
Clinical and Experimental Immunology logoLink to Clinical and Experimental Immunology
. 2005 Jul;141(1):10–18. doi: 10.1111/j.1365-2249.2005.02799.x

T cell-mediated immune responses in human newborns: ready to learn?

A Marchant 1, M Goldman 1
PMCID: PMC1809424  PMID: 15958064

Abstract

Infections with intracellular pathogens are often more severe or more prolonged in young infants suggesting that T cell-mediated immune responses are different in early life. Whereas neonatal immune responses have been quite extensively studied in murine models, studies of T cell-mediated immunity in human newborns and infants are scarce. Qualitative and quantitative differences when compared with adult immune responses have been observed but on the other hand mature responses to certain vaccines and infectious pathogens were demonstrated during the postnatal period and even during foetal life. Herein, we review the evidence suggesting that under appropriate conditions of stimulation, protective T cell-mediated immune responses could be induced by vaccines in early life.

Keywords: vaccines, human studies, paediatric, dendritic cells, T cells

Introduction

Young infants are exposed to a large number of infectious microorganisms, including viruses, bacteria and parasites. During the first months of life, maternal antibodies transferred to the foetus and infant attenuate the severity of infectious episodes caused by microorganisms against which the mother has developed immunological memory [1]. In contrast, maternal lymphocytes normally do not cross the placental barrier so that T cell-mediated immunity is not transferred to the offspring. Maternal T lymphocytes are present at relatively low concentrations in breastmilk but their role in the recipient infant is currently unknown [2]. Therefore, young infants are dependent on their own immune system to fight infections caused by intracellular pathogens for which immune defences merely depend on T lymphocytes. Viruses, including respiratory syncytial virus (RSV), herpes simplex virus (HSV), human immunodeficiency virus (HIV) and human cytomegalovirus (HCMV), Mycobacterium tuberculosis and Plasmodium falciparum are common causes of severe infections in early life, particularly in developing countries [37]. Prolonged replication of hepatitis B virus (HBV) and HCMV is also observed when infection occurs in early life [6,8]. These clinical observations suggest that T cell-mediated immune responses are different in young infants [3,4,9]. This would limit the efficacy of vaccines against intracellular pathogens. Whereas neonatal immune responses have been quite extensively studied in murine models (reviewed in 4,9), studies comparing T cell-mediated immunity in human infants and adults are scarce. Both qualitative and quantitative differences have been observed but on the other hand adult-like immune responses to certain infectious pathogens and vaccines were demonstrated during the postnatal period and even during foetal life. These observations suggest that under appropriate conditions of stimulation neonatal T lymphocytes can be instructed to fight intracellular pathogens.

In vitro studies of human neonatal T cells

Circulating neonatal T lymphocytes are fundamentally different from naïve adult T cells and have characteristics of recent thymic emigrants (Table 1). They contain high concentrations of T-cell-receptor excision circles (TRECs), episomal DNA byproducts of TCR α-chain rearrangement that are not replicated but diluted during cell division [10,11]. Like adult naïve cells, most neonatal T lymphocytes express the CD45 RA+ isoform and the costimulatory molecules CD27 and CD28. In contrast to adult naïve lymphocytes, neonatal lymphocytes express the CD38 molecule. In addition, a high proportion of circulating neonatal T cells are in cycle and display an increased susceptibility to apoptosis indicating high cell turn-over [1014]. Proliferation of naïve T lymphocytes can also be detected during fetal life (our unpublished observation) and could last up to five years of age [15]. The high cell turn-over observed in early life probably plays a central role in the establishment of the T cell repertoire. Despite their high turn-over, T cells preserve long telomeric sequences through a high constitutive telomerase activity [10]. In vitro apoptosis of neonatal T lymphocytes can be prevented by cytokines signalling through the γ-chain of the IL-2 receptor, namely IL-2, IL-4, IL-7 and IL-15 [11,14,16]. Among these cytokines, IL-7 and IL-15 also induce the proliferation of neonatal T lymphocytes in the absence of other stimuli [10,11,13,14,16,17]. IL-7 is involved in thymocyte development at a stage preceding the T cell receptor rearrangement [18]. Circulating neonatal T cells express higher levels of the IL-7 receptor α-chain (CD127) than adult naïve T cells [10,16]. Interestingly, IL-7-induced proliferation of newborn T lymphocytes is dependent on caspases, molecules that are classically involved in activation-induced cell death [19]. Proliferation induced by IL-7 is not associated with T cell differentiation [10,13]. IL-15 preferentially stimulates the proliferation of CD8 rather than CD4 T cells [10]. In contrast to IL-7, IL-15 induces the differentiation of CD8 T lymphocytes in vitro[10,20]. Although IL-7 is likely to play a role in the high turn-over of neonatal T lymphocytes, other factors are also probably involved. In neonatal mice, homeostatic proliferation of CD4 T lymphocytes is critically dependent on class II MHC—TCR interaction and only partly involves IL-7 [21].

Table 1.

In vitro studies of human neonatal T cells

References
Contain high concentrations of TRECs [10,11]
High cell turn-over [1014]
Have high constitutive telomerase activity and long telomeric sequences [10]
Have increased susceptibility to apoptosis, prevented by cytokines signalling through the IL-2 receptor γ-chain [11,14,16]
Proliferation induced by IL-7 and IL-15 [10,11,13,14,16,17,19,20]
Low production of IFN-γ by CD4 T lymphocytes associated with hypermethylation of IFN-γ promoter [25,26,28]
Expression of CD40 ligand by CD4 T lymphocytes is controversial [29,30,31,32,33,34,35]
Expression of NFAT by CD4 T lymphocytes is controversial [3638]
CD25+ regulatory T lymphocytes occur spontaneously during foetal life [4144]

Several mechanisms limit T helper 1 (Th1) type responses in early life. In utero, Th1 responses are toxic to the placenta and are inhibited by trophoblast-derived IL-10 and progesterone [2224]. At birth, Th1 responses are still of lower magnitude than later in life. In vitro, newborn CD4 T cells produce lower levels of IFN-γ than adult naïve T cells and are hypermethylated at CpG and non-CpG sites within the IFN-γ promoter [25]. In the presence of suboptimal CD28 costimulation, IL-12 stimulates the production of both IL-4 and IFN-γ by neonatal CD4 T lymphocytes whereas adult cells do not produce IL-4 under similar conditions [26]. In response to polyclonal or superantigen activation, postnatal thymocytes develop into Th2 cytokine producing CD4 cells whereas IL-12 is required to stimulate the production of IFN-γ[27]. In contrast, neonatal CD8 T cells produce similar levels of IFN-γ and have a pattern of IFN-γ promoter methylation comparable to that of naïve adult cells [25]. In addition, neonatal CD8 T lymphocytes are strictly dependent on the presence of IL-4 at the time of priming to differentiate into IL-4-producing cells [28].

The capacity of neonatal CD4 T lymphocytes to express CD40 ligand (CD154), a molecule playing a critical role in the help to B lymphocytes and CD8 T cells, remains controversial. Several authors reported a lower expression of CD154 by cord blood CD4 T cells activated by polyclonal stimuli as compared to adult cells [2933]. In contrast, Splawski et al. [34] reported high levels of CD154 expression by newborn CD4 T cells stimulated with anti-CD3 Ab but a low expression when cells were stimulated with PMA and ionomycin. More recently, Suarez et al. [35] demonstrated that cAMP-inducing agents combined to ionomycin induce the expression of high levels of CD154 by neonatal CD4 T cells.

Together, the available data indicate that naïve T lymphocytes are differently programmed in neonates and in adults. Recent studies have attempted to determine whether this different program involves specific signal transduction pathways. The group of Laughlin reported that neonatal CD4 T cells express lower levels of the nuclear factors of activated T cell (NFAT) than adult cells, including NFAT-1 a transcription factor involved in the activation of CD154 and IFN-γ genes [36,37]. In contrast, O'Neill et al. [38] reported similar levels of NFAT-1 in resting or activated neonatal and adult CD4 T cells. The basis of these conflicting results remains unclear. Importantly, most of these experiments involved the comparison of cord blood cells, including mostly naïve lymphocytes, with adults cells that include both naïve and memory lymphocytes. Direct comparison of cord cells with naïve adult cells is needed. In order to define the profiles of genes expressed during human T cell differentiation, Lee et al. [39] performed microarray gene analysis on resting thymocytes, cord blood CD4 T lymphocytes and adult naïve CD4 T lymphocytes. Interestingly, they observed that the greatest degree of expression change occurs at the transition between double positive and single positive CD4 thymocytes while the least amount of changes exists between cord blood and naïve adult CD4 lymphocytes. Yet, some markers of cord blood cells were identified, including CCR9, a chemokine receptor binding the thymus-expressed chemokine (TECK) and previously been proposed as a marker of recent thymic emigrants [40].

Essential regulatory mechanisms controlling T cell responses are in place in early life. CD25+ CD4 T lymphocytes naturally develop in the human foetus and newborn [4144]. This subset inhibits the proliferation of CD4 and CD8 T lymphocytes and thereby controls autoimmune reactions as well as allogeneic and antimicrobial immune responses. In contrast to adult cells, cord blood CD25+ CD4 lymphocytes have a naïve phenotype. Interestingly, CD25+ regulatory cells are detected at higher frequencies in preterm as compared to term newborns, suggesting that this subset could play an important immunoregulatory role during intrauterine life (43 and our unpublished observations). Regulatory properties can also be acquired in vitro by CD4 lymphocytes cultured in the presence of IL-10 and IFN-α[45].

Vaccines as models of controlled antigenic challenges in young infants

In order to determine whether differences exist between neonatal and adult T cell-mediated immune responses in vivo, vaccines currently administered to young infants have been used as models of controlled antigenic exposures (Table 2). Following World Health Organization recommendations, infants living in tropical countries are immunized at birth with the antituberculosis vaccine BCG, the oral poliovirus vaccine (OPV) and the anti-hepatitis B vaccine (HBVac) [3,4]. In countries were these diseases are not endemic, infants receive their first vaccines around 2 months of age.

Table 2.

Immune responses to vaccines and infectious pathogens in early life

Vaccine Immune responses References
Hepatitis B vaccine Defective early Th1 and increased memory Th2 responses in newborns as compared to naïve adults; higher antibody response than adults [46]
Oral poliomyelitis vaccine Defective Th1 response and similar antibody response in newborns as compared to immune adults [47]
M. bovis BCG vaccine Adult-like Th1 response in newborns; promotes antibody, Th1 and Th2 responses to unrelated vaccines [4850,58]
Whole cell pertussis vaccine Th1 response in 2 months-old infants [52]
Measles vaccine Lower Th1 response in 6–12 months-old infants as compared to immune adults [53]
Human cytomegalovirus Mature CD8 T cell response and defective CD4 T cell response in fetuses and infants as compared to adults [70,71,75,98,99]
Human immunodeficiency virus Defective CD4 and CD8 T cell responses in newborns and infants as compared to adults [76,77,78,79,80,81,82,100,101]
Trypanosoma cruzi Adult-like CD8 T cell response [88]
Herpes simplex virus Delayed IFN-γ response in infants as compared to adults [102]
Bordetella pertussis Th1 response in infants [52]

Infants immunized at birth with HBVac or OPV develop lower Th1 type responses than adults [46,47]. These lower Th1 responses are associated with higher antibody responses than in adults. During the early phase of the response, Th2 type cytokine production is similar in infants and adults but memory responses to HBVac is characterized by higher Th2 cytokine production in infants [46]. The low Th1 responses to HBVac and OPV contrast with the mature Th1 response induced by BCG in newborns [4850]. The response induced by BCG at birth is quantitatively and qualitatively similar to that observed in immune adults. In tuberculosis endemic countries, adults develop relatively weak immune responses to BCG, probably because of their chronic exposure to environmental mycobacteria [51]. In these countries, newborns who have not yet been chronically exposed to mycobacteria actually develop stronger Th1 responses to BCG than adults. Th1 responses were detected in 2 months-old infants immunized with the whole cell pertussis vaccine [52] but comparison with naïve adults could not be performed. Lower IFN-γ responses to measles vaccine were detected in 6–12 months-old infants as compared to immune adults [53].

The variable capacity of different vaccines to induce Th1 responses in young infants suggests that the quality of the signal delivered by antigen-presenting cells to T lymphocytes plays an important role. Mycobacteria as well as Bordetella pertussis toxin are potent activators of dendritic cells (DC), the antigen-presenting cells required for priming of naïve T lymphocytes [5457]. The Th1 response induced by BCG and whole cell pertussis vaccines in early life could therefore be related to their potent DC activating properties. As it will be discussed below, neonatal DCs have a lower capacity to produce cytokines promoting Th1 responses. The induction of Th1 responses in early life would therefore require vaccines that are particularly efficient at promoting this type of response. In order to determine whether the potent Th1 response induced by BCG could influence the response to the other vaccines, Ota et al. [58] conducted a randomized controlled trial measuring the response to HBVac and OPV in infants who received BCG at the time of priming or after the last dose of HBVac and OPV. This study showed that BCG increases the cytokine and antibody responses to unrelated vaccine antigens. Surprisingly, the production of both Th1 and Th2 cytokines was promoted suggesting that the magnitude but not the quality of neonatal responses to vaccines was influenced by BCG. Whether this phenomenon is related to the age of the study population and whether Th1 polarization of vaccine responses would be induced by BCG in adults has not yet been determined. Several epidemiological studies have suggested that BCG immunization in early life could influence the immune response to allergens and infectious pathogens. BCG vaccination of young infants was associated with a reduced risk of atopy in some populations but not in others [5964]. In Guinea Bissau, BCG immunization was associated with a reduced risk of acute respiratory tract infections and of all causes mortality [65,66]. The possibility that vaccines such as BCG may have nonspecific clinical effects remains a subject of intense controversy [6769]. Controlled epidemiological and immunological studies conducted in different populations are needed to answer this important question. The observation that BCG increases both Th1 and Th2 cytokine responses to unrelated vaccines in infants indicates that its potential nonspecific effects are unlikely to be related to a shift from Th2 to Th1 type immune responses [58].

T cell-mediated immune responses to infectious pathogens in early life

Only a restricted number of studies have described T cell responses to infectious pathogens in early life (Table 2). As observed with infant vaccines, the magnitude and the quality of cellular immune responses vary from one pathogen to another. HCMV is the most common cause of congenital infection. Acute HCMV infection is most often asymptomatic in immunocompetent adults. In contrast, about 10% of infected newborns develop severe symptoms [6]. If the pathogenesis of symptomatic infections remains poorly understood, the cellular immune response to HCMV has been recently studied in asymptomatic newborns and infants. Following congenital infection, newborns develop a mature CD8 T cell response to HCMV [70,71]. This response is similar to that detected in adults and is characterized by large oligoclonal expansions directed against a restricted number of immunodominant antigens. HCMV-specific CD8 T cells express a late differentiation phenotype and have antiviral effector functions, producing IFN-γ and having perforin-dependent cytolytic activity [7275]. CD8 T cell responses to HCMV have been measured in infected foetuses as early as after 28 weeks of gestation [71]. This mature CD8 T cell response to HCMV contrasts with the low CD8 T cell response to HIV. In HIV-infected infants, CD8 T cell responses are of lower magnitude and are less commonly detected than in infected adults [7681]. In coinfected infants, high frequencies of HCMV-specific CD8 T cells are detectable but anti-HIV-specific CD8 T cells are infrequently detected [82]. Similar results were obtained using IFN-γ Elispot and HLA class I-peptide multimers. The low CD8 T lymphocyte responses to HIV could play an important role in the rapid disease progression observed in young children. An estimated 600 000 new paediatric infections occur each year. Whereas children account for 4% cases of HIV infection, they represent 20% of AIDS deaths [5].

The mechanisms underlying the difference in the capacity of young infants to develop CD8 T cell responses to HIV and HCMV is unclear. They could be related to differences in the quality of T cell priming. Indeed, HCMV-specific CD8 T cells display a more advanced differentiation phenotype and express higher levels of perforin than HIV-specific cells [72,83,84]. Recent data indicate that the intensity of T cell activation plays a critical role in CD8 T cell differentiation [85]. The poor CD8 T cell response to HIV in infants could also be related to a low CD4 T cell help; interestingly, the potent CD8 T cell response to HCMV may not require CD4 help. Alternatively, the transmission of HIV mutants that have escaped maternal CD8 T cells could prevent the development of infant CD8 T cell responses to dominant epitopes [86]. Epidemiological studies have shown that HLA concordance between mother and child increases the risk of HIV transmission [87]. Finally, specific immune evasion mechanisms may be expressed by HIV that are particularly efficient at inhibiting the response of neonatal T cells. HCMV is not the only infectious pathogen stimulating potent CD8 T cell responses in early life. Newborns with congenital Trypanosoma cruzi infection also display large clonal expansions of CD8 T cells recognizing parasite antigens and producing IFN-γ[88]. CD8 T cell responses to RSV and measles can be detected in infants but whether these responses are of similar quality and magnitude than those of adults has not yet been established [8991].

Our knowledge of CD4 T lymphocyte responses to infectious pathogens in early life is even more restricted than that of CD8 T cells. Although foetuses and infants develop mature CD8 T cell responses to HCMV, they have lower CD4 T cell responses to this virus than adults. Adults develop strong CD4 T cell responses during the acute and persistent phases of HCMV infection [9296]. In contrast, children who acquired HCMV in utero or during infancy have very low proliferative and IFN-γ responses to HCMV antigens [97,98]. Intriguingly, these low responses can persist for several years post infection. In parallel, children infected in early life excrete the virus for prolonged periods of time. In the mouse model of CMV infection CD4 T lymphocytes are required to control viral replication in salivary glands [99]. The low CD4 T cell response to HCMV infection observed in infants could be involved in their persistent viral excretion and thereby allow virus spread in the population [6]. Low CD4 T cell responses are also detected in HIV-infected infants whereas CD4 T cell responses to this virus can be detected in older children and in adults [100,101]. The mechanisms involved in the low CD4 T cell responses to HCMV and HIV are not yet understood. They probably involve both intrinsic T cell characteristics previously mentioned as well as immune evasion mechanisms. Indeed, as discussed above, mature CD4 T cell responses can be triggered by vaccines in early life and Th1 type CD4 T cell responses develop following whooping cough in young infants [52]. Along the same line, IFN-γ-producing CD4 T cells were detected in HSV-infected infants, although their responses were established with a slower rate than in adults [102]. Also, following in utero exposure to helminth antigens newborn T lymphocytes produce similar cytokines as those produced by T lymphocytes obtained from infected adults [103,104].

Properties of neonatal dendritic cells

The variable T cell responses induced by vaccines and infectious pathogens in early life suggest that the function of antigen-presenting cells, in particular DCs, could be different in newborns and adults. Recent studies have compared the responses of neonatal and adult myeloid and plasmacytoid DCs to Toll-like receptor (TLR) ligands. TLRs are relatively conserved molecules recognizing microbial products that stimulate DCs and influence their ability to activate T lymphocytes [105]. A number of studies indicate that neonatal DCs have a lower capacity to produce cytokines promoting Th1 responses than adult DCs. Neonatal monocytes-derived DC (mDC) produce low concentrations of IL-12(p70) in response to lipopolysaccharide (LPS), poly I:C, Bordetella pertussis toxin or CD40 ligation [56,106]. This phenomenon is related to a reduced expression of the gene coding for the p35 subunit of IL-12 whereas IL-12 (p40) gene expression is comparable to that induced in adult cells. The low IL-12 (p35) synthesis depends on a reduced nucleosomal remodelling process that is required for IL-12(p35) gene transcription [107,108]. Importantly, addition of IFN-γ to LPS-activated DCs increases nucleosomal remodelling and IL-12 (p35) synthesis by neonatal mDC to adult-like levels [108]. Low IL-12 production by neonatal mDCs was also observed by Langrish et al. [109]. On the other hand, Upham et al. [110] observed that the production of IL-12 by neonatal monocytes is lower than in adults but that differentiation of monocytes into mDCs increases IL-12 production to the levels detected in adults. These discrepant results are probably related to differences in the conditions under which DCs were cultured and are compatible with the notion that neonatal mDCs are able to produce adult-like levels of IL-12 under some experimental conditions, including the addition of IFN-γ.

The function of neonatal plasmacytoid (p)DCs remains largely unexplored. In order to assess the ability of these cells to produce IFN-α, we used as stimulus CpG oligodeoxynucleotides which are considered as potential adjuvants to prime Th1 responses. In response to both stimuli, neonatal pDC were virtually incapable to synthesize IFNα although they expressed adult-like levels of TLR9 mRNA [111]. Together these results indicate that neonatal DCs have different characteristics than adult DCs. The mechanisms involved in their maturation have not been defined. Intriguingly, recent data indicate that adult-like levels of IL-12 are produced only after several years of life (110 and our own unpublished observation). This slow maturation of DC functions may be involved in the persistent defects in the CD4 T cell responses to HCMV and HIV [97,98,100,101]. Indeed, these viruses impair the function of DCs and reduce their capacity to produce IL-12 and activate T cells. In adults, the function of DCs may be sufficient to allow T cell priming even when infected with suppressive pathogens. In young infants, HCMV or HIV infection of immature DCs may suppress their costimulation capacity below the threshold required to activate CD4 T cells. The intensity and/or the nature of the costimulatory signals required to prime neonatal CD8 T lymphocytes may be lower or different than for CD4 T lymphocytes. Indeed, newborns with congenital HCMV infection have mature CD8 but lower CD4 T cell responses [70,71,97,98]. Also, in vitro neonatal mDCs are less efficient than adult mDCs at stimulating allogeneic CD4 T cells but are able to prime antigen-specific CD8 T lymphocytes at similar levels than adult mDCs [112].

Conclusion

In vitro investigations established that the capacity of neonatal CD4 T cells to produce IFN-γ and of neonatal DCs to promote Th1 responses is lower in infants as compared to adults. In vivo, Th1 responses to a number of vaccines and infectious pathogens are poor during early life. However, mature Th1 responses can develop in certain conditions such as neonatal BCG vaccination and Bordetella pertussis infection, probably in relation with a more efficient activation of DCs. In vitro studies as well as clinical investigations also suggest that the priming of neonatal CD8 T cell responses may require a lower threshold of DC activation than that required for Th1 responses. Other features of neonatal T lymphocytes include homeostatic proliferation and increased susceptibility to apoptosis, but the impact of this high cell turn-over on the establishment of memory responses remains to be clarified.

The classical paradigm that newborns have incompetent T lymphocytes developing only weak or even tolerogenic responses should clearly be reconsidered. The observation that mature cellular immune responses can be developed in early life suggests that under appropriate conditions of stimulation neonatal T lymphocytes can be instructed to fight intracellular pathogens (Fig. 1). We can therefore hope that the identification of molecular pathways leading to DC and T cell activation in human neonates will lead to the development of new vaccines eliciting efficient and safe protective responses against these agents early after birth.

Fig. 1.

Fig. 1

Youth denotes aptness to apprehend and learn. From the Iconologia or Moral Emblems by Cæsar Ripa, London, 1709; reproduced with the permission of the Special Collections Library, the Pennsylvania State University Libraries.

Acknowledgments

The authors thank Prof Sarah Rowland-Jones for critical reading of the manuscript.

References

  • 1.Zinkernagel RM. Maternal antibodies, childhood infections, and autoimmune diseases. N Engl J Med. 2001;345:1331–5. doi: 10.1056/NEJMra012493. [DOI] [PubMed] [Google Scholar]
  • 2.Rassin DK, Garofalo RP, Ogra PL. Human milk. In: Remington JS, Klein JO, editors. Infectious Diseases of the Fetus and the Newborn Infant. Philadelphia: W.B. Saunders Company; 2001. pp. 169–204. [Google Scholar]
  • 3.Marchant A, Newport M. Prevention of infectious diseases by neonatal and early infantile immunization: prospects for the new millennium. Curr Opin Infect Dis. 2000;13:241–6. doi: 10.1097/00001432-200006000-00007. [DOI] [PubMed] [Google Scholar]
  • 4.Siegrist CA. Neonatal and early life vaccinology. Vaccine. 2001;19:3331–46. doi: 10.1016/s0264-410x(01)00028-7. [DOI] [PubMed] [Google Scholar]
  • 5.Goulder PJ, Jeena P, Tudor-Williams G, Burchett S. Paediatric HIV infection: correlates of protective immunity and global perspectives in prevention and management. Br Med J. 2001;58:89–108. doi: 10.1093/bmb/58.1.89. [DOI] [PubMed] [Google Scholar]
  • 6.Stagno S. Cytomegalovirus. In: Remington JS, Klein JO, editors. Infectious Diseases of the Fetus and the Newborn Infant. Philadelphia: W.B. Saunders Company; 2001. pp. 389–424. [Google Scholar]
  • 7.Arvin A. Whitley. Herpes simplex virus infections. In: Remington JS, Klein JO, editors. Infectious Diseases of the Fetus and the Newborn Infant. Philadelphia: W.B. Saunders Company; 2001. pp. 425–46. [Google Scholar]
  • 8.Mahoney FJ. Kane. Hepatitis B vaccine. In: Plotkin SA, Orenstein WA, editors. Vaccines. Philadelphia: W.B. Saunders Company; 1999. pp. 158–82. [Google Scholar]
  • 9.Adkins B, Leclerc C, Marshall-Clarke S. Neonatal adaptive immunity comes of age. Nat Rev Immunol. 2004;4:553–64. doi: 10.1038/nri1394. [DOI] [PubMed] [Google Scholar]
  • 10.Schonland SO, Zimmer JK, Lopez-Benitez CM, Widmann T, Ramin KD, Goronzy JJ, Weyand CM. Homeostatic control of T-cell generation in neonates. Blood. 2003;102:1428–34. doi: 10.1182/blood-2002-11-3591. [DOI] [PubMed] [Google Scholar]
  • 11.Hassan J, Reen DJ. Human recent thymic emigrants – identification, expansion, and survival characteristics. J Immunol. 2001;167:1970–6. doi: 10.4049/jimmunol.167.4.1970. [DOI] [PubMed] [Google Scholar]
  • 12.Szabolcs P, Park KD, Reese M, Marti L, Broadwater G, Kurtzberg J. Coexistent naive phenotype and higher cycling rate of cord blood T cells as compared to adult peripheral blood. Exp Hematol. 2003;31:708–14. doi: 10.1016/s0301-472x(03)00160-7. [DOI] [PubMed] [Google Scholar]
  • 13.Hassan J, Reen DJ. IL-7 promotes the survival and maturation but not differentiation of human post-thymic CD4+ T cells. Eur J Immunol. 1998;28:3057–65. doi: 10.1002/(SICI)1521-4141(199810)28:10<3057::AID-IMMU3057>3.0.CO;2-Z. [DOI] [PubMed] [Google Scholar]
  • 14.Soares MV, Borthwick NJ, Maini MK, Janossy G, Salmon M, Akbar AN. IL-7-dependent extrathymic expansion of CD45RA+ T cells enables preservation of a naive repertoire. J Immunol. 1998;161:5909–17. [PubMed] [Google Scholar]
  • 15.Hazenberg MD, Otto SA, van Rossum AM, et al. Establishment of the CD4+ T-cell pool in healthy children and untreated children infected with HIV-1. Blood. 2004;104:3513–9. doi: 10.1182/blood-2004-03-0805. [DOI] [PubMed] [Google Scholar]
  • 16.Fukui T, Katamura K, Abe N, Kiyomasu T, Iio J, Ueno H, Mayumi M, Furusho K. IL-7 induces proliferation, variable cytokine-producing ability and IL-2 responsiveness in naive CD4+ T-cells from human cord blood. Immunol Lett. 1997;59:21–8. doi: 10.1016/s0165-2478(97)00093-x. [DOI] [PubMed] [Google Scholar]
  • 17.Webb LM, Foxwell BM, Feldmann M. Putative role for interleukin-7 in the maintenance of the recirculating naive CD4+ T-cell pool. Immunology. 1999;98:400–5. doi: 10.1046/j.1365-2567.1999.00906.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Freeden-Jeffry U, Solvason N, Howard M, Murray R. The earliest T lineage-committed cells depend on IL-7 for Bcl-2 expression and normal cell cycle progression. Immunity. 1997;7:147–54. doi: 10.1016/s1074-7613(00)80517-8. [DOI] [PubMed] [Google Scholar]
  • 19.O'Neill RM, Hassan J, Reen DJ. IL-7-regulated homeostatic maintenance of recent thymic emigrants in association with caspase-mediated cell proliferation and apoptotic cell death. J Immunol. 2003;170:4524–31. doi: 10.4049/jimmunol.170.9.4524. [DOI] [PubMed] [Google Scholar]
  • 20.Cookson S, Reen D. IL-15 drives neonatal T cells to acquire CD56 and become activated effector cells. Blood. 2003;102:2195–7. doi: 10.1182/blood-2003-01-0232. [DOI] [PubMed] [Google Scholar]
  • 21.Min B, McHugh R, Sempowski GD, Mackall C, Foucras G, Paul WE. Neonates support lymphopenia-induced proliferation. Immunity. 2003;18:131–40. doi: 10.1016/s1074-7613(02)00508-3. [DOI] [PubMed] [Google Scholar]
  • 22.Krishnan L, Guilbert LJ, Wegmann TG, Belosevic M, Mosmann TR. T helper 1 response against Leishmania major in pregnant C57BL/6 mice increases implantation failure and fetal resorptions. Correlation with increased IFN-gamma and TNF and reduced IL-10 production by placental cells. J Immunol. 1996;156:653–62. [PubMed] [Google Scholar]
  • 23.Roth I, Corry DB, Locksley RM, Abrams JS, Litton MJ, Fisher SJ. Human placental cytotrophoblasts produce the immunosuppressive cytokine interleukin 10. J Exp Med. 1996;184:539–48. doi: 10.1084/jem.184.2.539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Szekeres-Bartho J, Faust Z, Varga P, Szereday L, Kelemen K. The immunological pregnancy protective effect of progesterone is manifested via controlling cytokine production. Am J Reprod Immunol. 1996;35:348–51. doi: 10.1111/j.1600-0897.1996.tb00492.x. [DOI] [PubMed] [Google Scholar]
  • 25.White GP, Watt PM, Holt BJ, Holt PG. Differential patterns of methylation of the IFN-gamma promoter at CpG and non-CpG sites underlie differences in IFN-gamma gene expression between human neonatal and adult CD45RO- T cells. J Immunol. 2002;168:2820–7. doi: 10.4049/jimmunol.168.6.2820. [DOI] [PubMed] [Google Scholar]
  • 26.Ohshima Y, Delespesse G. T cell-derived IL-4 and dendritic cell-derived IL-12 regulate the lymphokine-producing phenotype of alloantigen-primed naive human CD4 T cells. J Immunol. 1997;158:629–36. [PubMed] [Google Scholar]
  • 27.Mingari MC, Maggi E, Cambiaggi A, Annunziato F, Schiavetti F, Manetti R, Moretta L, Romagnani S. Development in vitro of human CD4+ thymocytes into functionally mature Th2 cells. Exogenous interleukin-12 is required for priming thymocytes to produce both Th1 cytokines and interleukin-10. Eur J Immunol. 1996;26:1083–7. doi: 10.1002/eji.1830260519. [DOI] [PubMed] [Google Scholar]
  • 28.Delespesse G, Yang LP, Ohshima Y, Demeure C, Shu U, Byun DG, Sarfati M. Maturation of human neonatal CD4+ and CD8+ T lymphocytes into Th1/Th2 effectors. Vaccine. 1998;16:1415–9. doi: 10.1016/s0264-410x(98)00101-7. [DOI] [PubMed] [Google Scholar]
  • 29.Durandy A, De Saint BG, Lisowska-Grospierre B, Gauchat JF, Forveille M, Kroczek RA, Bonnefoy JY, Fischer A. Undetectable CD40 ligand expression on T cells and low B cell responses to CD40 binding agonists in human newborns. J Immunol. 1995;154:1560–8. [PubMed] [Google Scholar]
  • 30.Nonoyama S, Penix LA, Edwards CP, Lewis DB, Ito S, Aruffo A, Wilson CB, Ochs HD. Diminished expression of CD40 ligand by activated neonatal T cells. J Clin Invest. 1995;95:66–75. doi: 10.1172/JCI117677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fuleihan R, Ahern D, Geha RS. Decreased expression of the ligand for CD40 in newborn lymphocytes. Eur J Immunol. 1994;24:1925–8. doi: 10.1002/eji.1830240832. [DOI] [PubMed] [Google Scholar]
  • 32.Brugnoni DP, Airo D, Graf M, et al. Ugazio, and. Ineffective expression of CD40 ligand on cord blood T cells may contribute to poor immunoglobulin production in the newborn. Eur J Immunol. 1994;24:1919–24. doi: 10.1002/eji.1830240831. [DOI] [PubMed] [Google Scholar]
  • 33.Jullien P, Cron RQ, Dabbagh K, Cleary A, Chen L, Tran P, Stepick-Biek P, Lewis DB. Decreased CD154 expression by neonatal CD4+ T cells is due to limitations in both proximal and distal events of T cell activation. Int Immunol. 2003;15:1461–72. doi: 10.1093/intimm/dxg145. [DOI] [PubMed] [Google Scholar]
  • 34.Splawski JB, Nishioka J, Nishioka Y, Lipsky PE. CD40 ligand is expressed and functional on activated neonatal T cells. J Immunol. 1996;156:119–27. [PubMed] [Google Scholar]
  • 35.Suarez A, Mozo L, Gayo A, Simo A, Gutierrez C. Induction of functional CD154 (CD40 ligand) in neonatal T cells by cAMP-elevating agents. Immunology. 2000;100:432–40. doi: 10.1046/j.1365-2567.2000.00051.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kadereit S, Mohammad SF, Miller RE, et al. Reduced NFAT1 protein expression in human umbilical cord blood T lymphocytes. Blood. 1999;94:3101–7. [PubMed] [Google Scholar]
  • 37.Kaminski BA, Kadereit S, Miller RE, et al. Reduced expression of NFAT-associated genes in UCB versus adult CD4+ T lymphocytes during primary stimulation. Blood. 2003;102:4608–17. doi: 10.1182/blood-2003-05-1732. [DOI] [PubMed] [Google Scholar]
  • 38.O'Neill RM, Reen DJ. Equivalent functional nuclear factor of activated T cell 1 mRNA and protein expression in cord blood and adult T cells. Transplantation. 2003;76:1526–8. doi: 10.1097/01.TP.0000084308.19565.2B. [DOI] [PubMed] [Google Scholar]
  • 39.Lee M, Hanspers SK, Barker CS, Korn AP, McCune JM. Gene expression profiles during human CD4+ T cell differentiation. Int Immunol. 2004;16:1109–24. doi: 10.1093/intimm/dxh112. [DOI] [PubMed] [Google Scholar]
  • 40.Olaussen RW, Farstad IN, Brandtzaeg P, Rugtveit J. Age-related changes in CCR9+ circulating lymphocytes: are CCR9+ naive T cells recent thymic emigrants? Scand J Immunol. 2001;54:435–9. doi: 10.1046/j.1365-3083.2001.01008.x. [DOI] [PubMed] [Google Scholar]
  • 41.Ng WF, Duggan PJ, Ponchel F, Matarese G, Lombardi G, Edwards AD, Isaacs JD, Lechler RI. Human CD4(+)CD25(+) cells: a naturally occurring population of regulatory T cells. Blood. 2001;98:2736–44. doi: 10.1182/blood.v98.9.2736. [DOI] [PubMed] [Google Scholar]
  • 42.Thornton CA, Upham JW, Wikstrom ME, Holt BJ, White GP, Sharp MJ, Sly PD, Holt PG. Functional maturation of CD4+CD25+CTLA4+CD45RA+ T regulatory cells in human neonatal T cell responses to environmental antigens/allergens. J Immunol. 2004;173:3084–92. doi: 10.4049/jimmunol.173.5.3084. [DOI] [PubMed] [Google Scholar]
  • 43.Takahata Y, Nomura A, Takada H, et al. CD25+CD4+ T cells in human cord blood: an immunoregulatory subset with naive phenotype and specific expression of forkhead box p3 (Foxp3) gene. Exp Hematol. 2004;32:622–9. doi: 10.1016/j.exphem.2004.03.012. [DOI] [PubMed] [Google Scholar]
  • 44.Godfrey WR, Spoden DJ, Ge YG, et al. Cord blood CD4+CD25+ derived T regulatory cell lines express FoxP3 protein and manifest potent suppressor function. Blood. 2004;105:750–8. doi: 10.1182/blood-2004-06-2467. [DOI] [PubMed] [Google Scholar]
  • 45.Levings M, Sangregorio KR, Galbiati F, Squadrone S, de Waal MR, Roncarolo MG. IFN-alpha and IL-10 induce the differentiation of human type 1 T regulatory cells. J Immunol. 2001;166:5530–9. doi: 10.4049/jimmunol.166.9.5530. [DOI] [PubMed] [Google Scholar]
  • 46.Ota MO, Vekemans J, Schlegel-Haueter SE, et al. Hepatitis B immunisation induces higher antibody and memory Th2 responses in new-borns than in adults. Vaccine. 2004;22:511–9. doi: 10.1016/j.vaccine.2003.07.020. [DOI] [PubMed] [Google Scholar]
  • 47.Vekemans J, Ota MO, Wang EC, et al. T cell responses to vaccines in infants: defective IFNgamma production after oral polio vaccination. Clin Exp Immunol. 2002;127:495–8. doi: 10.1046/j.1365-2249.2002.01788.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hussey GD, Watkins ML, Goddard EA, Gottschalk S, Hughes EJ, Iloni K, Kibel MA, Ress SR. Neonatal mycobacterial specific cytotoxic T-lymphocyte and cytokine profiles in response to distinct BCG vaccination strategies. Immunology. 2002;105:314–24. doi: 10.1046/j.1365-2567.2002.01366.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Marchant A, Goetghebuer T, Ota MO, et al. Newborns develop a Th1-type immune response to Mycobacterium bovis bacillus Calmette-Guerin vaccination. J Immunol. 1999;163:2249–55. [PubMed] [Google Scholar]
  • 50.Vekemans JA, Amedei MO, Ota MM, D'Elios T, Goetghebuer J, Ismaili MJ, Newport G, Del Prete M, Goldman KP. McAdam, and A. Marchant. Neonatal bacillus Calmette-Guerin vaccination induces adult-like IFN-gamma production by CD4+ T lymphocytes. Eur J Immunol. 2001;31:1531–5. doi: 10.1002/1521-4141(200105)31:5<1531::AID-IMMU1531>3.0.CO;2-1. [DOI] [PubMed] [Google Scholar]
  • 51.Black GF, Weir RE, Floyd S, et al. BCG-induced increase in interferon-gamma response to mycobacterial antigens and efficacy of BCG vaccination in Malawi and the UK: two randomised controlled studies. Lancet. 2002;20:1393–401. doi: 10.1016/S0140-6736(02)08353-8. [DOI] [PubMed] [Google Scholar]
  • 52.Mascart FV, Verscheure A, Malfroot M, et al. Bordetella pertussis infection in 2-month-old infants promotes type 1 T cell responses. J Immunol. 2003;170:1504–9. doi: 10.4049/jimmunol.170.3.1504. [DOI] [PubMed] [Google Scholar]
  • 53.Gans H, Yasukawa L, Rinki M, et al. Immune responses to measles and mumps vaccination of infants at 6, 9, and 12 months. J Infect Dis. 2001;184:817–26. doi: 10.1086/323346. [DOI] [PubMed] [Google Scholar]
  • 54.Henderson RA, Watkins SC, Flynn JL. Activation of human dendritic cells following infection with Mycobacterium tuberculosis. J Immunol. 1997;159:635–43. [PubMed] [Google Scholar]
  • 55.Inaba K, Inaba M, Naito M, Steinman RM. Dendritic cell progenitors phagocytose particulates, including bacillus Calmette-Guerin organisms, and sensitize mice to mycobacterial antigens in vivo. J Exp Med. 1993;178:479–88. doi: 10.1084/jem.178.2.479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Tonon SS, Goriely E, Aksoy O, et al. Bordetella pertussis toxin induces the release of inflammatory cytokines and dendritic cell activation in whole blood: impaired responses in human newborns. Eur J Immunol. 2002;32:3118–25. doi: 10.1002/1521-4141(200211)32:11<3118::AID-IMMU3118>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
  • 57.Ausiello CM, Fedele G, Urbani F, Lande R, Di Carlo B, Cassone A. Native and genetically inactivated pertussis toxins induce human dendritic cell maturation and synergize with lipopolysaccharide in promoting T helper type 1 responses. J Infect Dis. 2002;186:351–60. doi: 10.1086/341510. [DOI] [PubMed] [Google Scholar]
  • 58.Ota MO, Vekemans J, Schlegel-Haueter SE, et al. Influence of Mycobacterium bovis bacillus Calmette-Guerin on antibody and cytokine responses to human neonatal vaccination. J Immunol. 2002;168:919–25. doi: 10.4049/jimmunol.168.2.919. [DOI] [PubMed] [Google Scholar]
  • 59.Aaby P, Shaheen SO, Heyes CB, Goudiaby A, Hall AJ, Shiell AW, Jensen H, Marchant A. Early BCG vaccination and reduction in atopy in Guinea-Bissau. Clin Exp Allergy. 2000;30:644–50. doi: 10.1046/j.1365-2222.2000.00803.x. [DOI] [PubMed] [Google Scholar]
  • 60.Annus T, Montgomery SM, Riikjarv MA, Bjorksten B. Atopic disorders among Estonian schoolchildren in relation to tuberculin reactivity and the age at BCG vaccination. Allergy. 2004;59:1068–73. doi: 10.1111/j.1398-9995.2004.00557.x. [DOI] [PubMed] [Google Scholar]
  • 61.Bager P, Rostgaard K, Nielsen NM, Melbye M, Westergaard T. Age at bacille Calmette-Guerin vaccination and risk of allergy and asthma. Clin Exp Allergy. 2003;33:1512–7. doi: 10.1046/j.1365-2222.2003.01796.x. [DOI] [PubMed] [Google Scholar]
  • 62.da Cunha SS, Cruz AA, Dourado I, Barreto ML, Ferreira LD, Rodrigues LC. Lower prevalence of reported asthma in adolescents with symptoms of rhinitis that received neonatal BCG. Allergy. 2004;59:857–62. doi: 10.1111/j.1398-9995.2004.00517.x. [DOI] [PubMed] [Google Scholar]
  • 63.Townley RG, Barlan IB, Patino C, et al. The effect of BCG vaccine at birth on the development of atopy or allergic disease in young children. Ann Allergy Asthma Immunol. 2004;92:350–5. doi: 10.1016/S1081-1206(10)61574-8. [DOI] [PubMed] [Google Scholar]
  • 64.von Hertzen LC, Haahtela T. Immunization and atopy: possible implications of ethnicity. J Allergy Clin Immunol. 2004;113:401–6. doi: 10.1016/j.jaci.2003.12.010. [DOI] [PubMed] [Google Scholar]
  • 65.Kristensen I, Aaby P, Jensen H. Routine vaccinations and child survival: follow up study in Guinea-Bissau, West Africa. Br Med J. 2000;321:1435–8. doi: 10.1136/bmj.321.7274.1435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Stensballe LG, Nante E, Jensen IP, Kofoed PE, Poulsen A, Jensen H, Newport M, Marchant A, Aaby P. Acute lower respiratory tract infections and respiratory syncytial virus in children in Guinea-Bissau: a beneficial effect of BCG vaccination for girls. Vaccine. 2005;23:1251–7. doi: 10.1016/j.vaccine.2004.09.006. [DOI] [PubMed] [Google Scholar]
  • 67.Aaby P, Jensen H. Routine vaccination and child survival in Guinea-Bissau. Author's reply to commentary. Br Med J. 2001;322:360. [PMC free article] [PubMed] [Google Scholar]
  • 68.Shann F. Non-specific effects of vaccines in developing countries. We need evidence about the effect of vaccines on mortality from all causes. Br Med J. 2000;321:1423–4. doi: 10.1136/bmj.321.7274.1423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Fine P. Commentary: an unexpected finding that needs confirmation or rejection. Br Med J. 2000;321:1439. [PubMed] [Google Scholar]
  • 70.Gibson L, Piccinini G, Lilleri D, Revello MG, Wang Z, Markel S, Diamond DJ, Luzuriaga K. Human cytomegalovirus proteins pp65 and immediate early protein 1 are common targets for CD8+ T cell responses in children with congenital or postnatal human cytomegalovirus infection. J Immunol. 2004;172:2256–64. doi: 10.4049/jimmunol.172.4.2256. [DOI] [PubMed] [Google Scholar]
  • 71.Marchant AV, Appay SM, Van Der N, et al. Mature CD8(+) T lymphocyte response to viral infection during fetal life. J Clin Invest. 2003;111:1747–55. doi: 10.1172/JCI17470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Appay VPR, Dunbar M, Callan P, et al. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nat Med. 2002;8:379–85. doi: 10.1038/nm0402-379. [DOI] [PubMed] [Google Scholar]
  • 73.Tomiyama H, Takata H, Matsuda T, Takiguchi M. Phenotypic classification of human CD8+ T cells reflecting their function: inverse correlation between quantitative expression of CD27 and cytotoxic effector function. Eur J Immunol. 2004;34:999–1010. doi: 10.1002/eji.200324478. [DOI] [PubMed] [Google Scholar]
  • 74.Wills MR, Okecha G, Weekes MP, Gandhi MK, Sissons PJ, Carmichael AJ. Identification of naive or antigen-experienced human CD8(+) T cells by expression of costimulation and chemokine receptors: analysis of the human cytomegalovirus-specific CD8(+) T cell response. J Immunol. 2002;168:5455–64. doi: 10.4049/jimmunol.168.11.5455. [DOI] [PubMed] [Google Scholar]
  • 75.Chen SF, Tu WW, Sharp MA, et al. Antiviral CD8 T cells in the control of primary human cytomegalovirus infection in early childhood. J Infect Dis. 2004;189:1619–27. doi: 10.1086/383249. [DOI] [PubMed] [Google Scholar]
  • 76.Chakraborty R, Gillespie GM, Reinis M, et al. HIV-1-specific CD8 T cell responses in a pediatric slow progressor infected as a premature neonate. AIDS. 2002;16:2085–7. doi: 10.1097/00002030-200210180-00017. [DOI] [PubMed] [Google Scholar]
  • 77.Sandberg JK, Fast NM, Jordan KA, et al. HIV-specific CD8+ T cell function in children with vertically acquired HIV-1 infection is critically influenced by age and the state of the CD4+ T cell compartment. J Immunol. 2003;170:4403–10. doi: 10.4049/jimmunol.170.8.4403. [DOI] [PubMed] [Google Scholar]
  • 78.Buseyne F, Burgard M, Teglas JP, Bui E, Rouzioux C, Mayaux MJ, Blanche S, Riviere Y. Early HIV-specific cytotoxic T lymphocytes and disease progression in children born to HIV-infected mothers. AIDS Res Hum Retroviruses. 1998;14:1435–44. doi: 10.1089/aid.1998.14.1435. [DOI] [PubMed] [Google Scholar]
  • 79.Pikora CA, Sullivan JL, Panicali D, Luzuriaga K. Early HIV-1 envelope-specific cytotoxic T lymphocyte responses in vertically infected infants. J Exp Med. 1997;185:1153–61. doi: 10.1084/jem.185.7.1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Luzuriaga K, Holmes D, Hereema A, Wong J, Panicali DL, Sullivan JL. HIV-1-specific cytotoxic T lymphocyte responses in the first year of life. J Immunol. 1995;154:433–43. [PubMed] [Google Scholar]
  • 81.Wasik TJ, Jagodzinski PP, Hyjek EM, Wustner J, Trinchieri G, Lischner HW, Kozbor D. Diminished HIV-specific CTL activity is associated with lower type 1 and enhanced type 2 responses to HIV-specific peptides during perinatal HIV infection. J Immunol. 1997;158:6029–36. [PubMed] [Google Scholar]
  • 82.Scott ZA, Chadwick EG, Gibson LL, et al. Infrequent detection of HIV-1-specific, but not cytomegalovirus-specific, CD8(+) T cell responses in young HIV-1-infected infants. J Immunol. 2001;167:7134–40. doi: 10.4049/jimmunol.167.12.7134. [DOI] [PubMed] [Google Scholar]
  • 83.Appay VDF, Nixon SM, Donahoe GM, et al. HIV-specific CD8(+) T cells produce antiviral cytokines but are impaired in cytolytic function. J Exp Med. 2000;192:63–75. doi: 10.1084/jem.192.1.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Champagne PGS, Ogg AS, King C, et al. Skewed maturation of memory HIV-specific CD8 T lymphocytes. Nature. 2001;410:106–11. doi: 10.1038/35065118. [DOI] [PubMed] [Google Scholar]
  • 85.Papagno L, Spina CA, Marchant A, et al. Immune Activation and CD8(+) T-Cell Differentiation towards Senescence in HIV-1 Infection. Plos Biol. 2004;2:E20. doi: 10.1371/journal.pbio.0020020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Goulder PJ, Brander C, Tang Y, et al. Evolution and transmission of stable CTL escape mutations in HIV infection. Nature. 2001;412:334–8. doi: 10.1038/35085576. [DOI] [PubMed] [Google Scholar]
  • 87.MacDonald KS, Embree J, Njenga S, et al. Mother-child class I HLA concordance increases perinatal human immunodeficiency virus type 1 transmission. J Infect Dis. 1998;177:551–6. doi: 10.1086/514243. [DOI] [PubMed] [Google Scholar]
  • 88.Hermann E, Truyens C, Alonso-Vega C, et al. Human fetuses are able to mount an adultlike CD8 T-cell response. Blood. 2002;100:2153–8. [PubMed] [Google Scholar]
  • 89.Isaacs D, Bangham CR, McMichael AJ. Cell-mediated cytotoxic response to respiratory syncytial virus in infants with bronchiolitis. Lancet. 1987;2:769–71. doi: 10.1016/s0140-6736(87)92502-5. [DOI] [PubMed] [Google Scholar]
  • 90.Mbawuike IN, Wells J, Byrd R, Cron SG, Glezen WP, Piedra PA. HLA-restricted CD8+ cytotoxic T lymphocyte, interferon-gamma, and interleukin-4 responses to respiratory syncytial virus infection in infants and children. J Infect Dis. 2001;183:687–96. doi: 10.1086/318815. [DOI] [PubMed] [Google Scholar]
  • 91.Jaye A, Magnusen AF, Sadiq AD, Corrah T, Whittle HC. Ex vivo analysis of cytotoxic T lymphocytes to measles antigens during infection and after vaccination in Gambian children. J Clin Invest. 1998;102:1969–77. doi: 10.1172/JCI3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Gamadia LE, Remmerswaal EB, Weel JF, Bemelman F, van Lier RA, Ten Berge IJ. Primary immune responses to human CMV. a critical role for IFN-gamma-producing CD4+ T cells in protection against CMV disease. Blood. 2003;101:2686–92. doi: 10.1182/blood-2002-08-2502. [DOI] [PubMed] [Google Scholar]
  • 93.Weekes MP, Wills MR, Sissons JG, Carmichael AJ. Long-term stable expanded human CD4+ T cell clones specific for human cytomegalovirus are distributed in both CD45RAhigh and CD45ROhigh populations. J Immunol. 2004;173:5843–51. doi: 10.4049/jimmunol.173.9.5843. [DOI] [PubMed] [Google Scholar]
  • 94.van Leeuwen EM, Remmerswaal EB, Vossen MT, Rowshani AT, Wertheim-van Dillen PM, van Lier RA, Ten Berge IJ. Emergence of a CD4+CD28— granzyme B+, cytomegalovirus-specific T cell subset after recovery of primary cytomegalovirus infection. J Immunol. 2004;173:1834–41. doi: 10.4049/jimmunol.173.3.1834. [DOI] [PubMed] [Google Scholar]
  • 95.Bitmansour AD, Douek DC, Maino VC, Picker LJ. Direct ex vivo analysis of human CD4(+) memory T cell activation requirements at the single clonotype level. J Immunol. 2002;169:1207–18. doi: 10.4049/jimmunol.169.3.1207. [DOI] [PubMed] [Google Scholar]
  • 96.Kern F, Bunde T, Faulhaber N, et al. Cytomegalovirus (CMV) phosphoprotein 65 makes a large contribution to shaping the T cell repertoire in CMV-exposed individuals. J Infect Dis. 2002;185:1709–16. doi: 10.1086/340637. [DOI] [PubMed] [Google Scholar]
  • 97.Tu W, Chen S, Sharp M, Dekker C, et al. Persistent and selective deficiency of CD4+ T cell immunity to cytomegalovirus in immunocompetent young children. J Immunol. 2004;172:3260–7. doi: 10.4049/jimmunol.172.5.3260. [DOI] [PubMed] [Google Scholar]
  • 98.Pass RF, Stagno S, Britt WJ, Alford CA. Specific cell-mediated immunity and the natural history of congenital infection with cytomegalovirus. J Infect Dis. 1983;148:953–61. doi: 10.1093/infdis/148.6.953. [DOI] [PubMed] [Google Scholar]
  • 99.Jonjic S, Mutter W, Weiland F, Reddehase MJ, Koszinowski UH. Site-restricted persistent cytomegalovirus infection after selective long-term depletion of CD4+ T lymphocytes. J Exp Med. 1989;169:1199–212. doi: 10.1084/jem.169.4.1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Feeney ME, Draenert R, Roosevelt KA, et al. Reconstitution of virus-specific CD4 proliferative responses in pediatric HIV-1 infection. J Immunol. 2003;171:6968–75. doi: 10.4049/jimmunol.171.12.6968. [DOI] [PubMed] [Google Scholar]
  • 101.Scott ZA, Beaumier CM, Sharkey M, Stevenson M, Luzuriaga K. HIV-1 replication increases HIV-specific CD4+ T cell frequencies but limits proliferative capacity in chronically infected children. J Immunol. 2003;170:5786–92. doi: 10.4049/jimmunol.170.11.5786. [DOI] [PubMed] [Google Scholar]
  • 102.Burchett SK, Corey L, Mohan KM, Westall J, Ashley R, Wilson CB. Diminished interferon-gamma and lymphocyte proliferation in neonatal and postpartum primary herpes simplex virus infection. J Infect Dis. 1992;165:813–8. doi: 10.1093/infdis/165.5.813. [DOI] [PubMed] [Google Scholar]
  • 103.Malhotra I, Mungai P, Wamachi A, Kioko J, Ouma JH, Kazura JW, King CL. Helminth- and Bacillus Calmette-Guerin-induced immunity in children sensitized in utero to filariasis and schistosomiasis. J Immunol. 1999;162:6843–8. [PubMed] [Google Scholar]
  • 104.Malhotra I, Ouma J, Wamachi A, et al. In utero exposure to helminth and mycobacterial antigens generates cytokine responses similar to that observed in adults. J Clin Invest. 1997;99:1759–66. doi: 10.1172/JCI119340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nat Immunol. 2004;5:987–95. doi: 10.1038/ni1112. [DOI] [PubMed] [Google Scholar]
  • 106.Goriely SB, Vincart P, Stordeur J, Vekemans F, Willems M, Goldman , De Wit D. Deficient IL-12 (p35) gene expression by dendritic cells derived from neonatal monocytes. J Immunol. 2001;166:2141–6. doi: 10.4049/jimmunol.166.3.2141. [DOI] [PubMed] [Google Scholar]
  • 107.Goriely S, Demonte D, Nizet S, De Wit D, Willems F, Goldman M, Van Lint C. Human IL-12 (p35) gene activation involves selective remodeling of a single nucleosome within a region of the promoter containing critical Sp1-binding sites. Blood. 2003;101:4894–902. doi: 10.1182/blood-2002-09-2851. [DOI] [PubMed] [Google Scholar]
  • 108.Goriely S, Van Lint C, Dadkhah R, Libin M, De Wit D, Demonte D, Willems F, Goldman M. a defect in nucleosome remodeling prevents IL-12 (p35) gene transcription in neonatal dendritic cells. J Exp Med. 2004;199:1011–6. doi: 10.1084/jem.20031272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Langrish CL, Buddle JC, Thrasher AJ, Goldblatt D. Neonatal dendritic cells are intrinsically biased against Th-1 immune responses. Clin Exp Immunol. 2002;128:118–23. doi: 10.1046/j.1365-2249.2002.01817.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Upham JW, Lee PT, Holt BJ, Heaton T, Prescott SL, Sharp MJ, Sly PD, Holt PG. Development of interleukin-12-producing capacity throughout childhood. Infect Immun. 2002;70:6583–8. doi: 10.1128/IAI.70.12.6583-6588.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.De Wit D, Olislagers V, Goriely S, Vermeulen F, Wagner H, Goldman M, Willems F. Blood plasmacytoid dendritic cell responses to CpG oligodeoxynucleotides are impaired in human newborns. Blood. 2004;103:1030–2. doi: 10.1182/blood-2003-04-1216. [DOI] [PubMed] [Google Scholar]
  • 112.Salio M, Dulphy N, Renneson J, Herbert M, McMichael A, Marchant A, Cerundolo V. Efficient priming of antigen-specific cytotoxic T lymphocytes by human cord blood dendritic cells. Int Immunol. 2003;15:1265–73. doi: 10.1093/intimm/dxg123. [DOI] [PubMed] [Google Scholar]

Articles from Clinical and Experimental Immunology are provided here courtesy of British Society for Immunology

RESOURCES