Skip to main content

REVIEW article

Front. Immunol., 20 November 2020
Sec. Alloimmunity and Transplantation
This article is part of the Research Topic Recent Developments in Haploidentical Stem Cell Transplantation: Therapy and Complications View all 10 articles

T cell Tolerance in Early Life

  • 1Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
  • 2Institute of Systems Biomedicine, Peking University Health Science Center, Beijing, China
  • 3Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China

T cell-mediated immune tolerance is a state of unresponsiveness of T cells towards specific self or non-self antigens. This is particularly essential during prenatal/neonatal period when T cells are exposed to dramatically changing environment and required to avoid rejection of maternal antigens, limit autoimmune responses, tolerate inert environmental and food antigens and antigens from non-harmful commensal microorganisms, promote maturation of mucosal barrier function, yet mount an appropriate response to pathogenic microorganisms. The cell-intrinsic and cell extrinsic mechanisms promote the generation of prenatal/neonatal T cells with distinct features to meet the complex and dynamic need of tolerance during this period. Reduced exposure or impaired tolerance in early life may have significant impact on allergic or autoimmune diseases in adult life. The uniqueness of conventional and regulatory T cells in human umbilical cord blood (UCB) may also provide certain advantages in UCB transplantation for hematological disorders.

Introduction

Immune tolerance is a state of unresponsiveness of the immune cells towards specific self or non-self antigens. It is an essential mechanism to prevent unwanted or self-reactive immune responses. In allogeneic hematopoietic stem cell transplantation (HSCT), failure to develop immune tolerance to autoantigens and alloantigens results in chronic graft-versus-host disease (GVHD), a leading cause of non-relapse morbidity and mortality (1).

Immune tolerance was first discovered in neonatal dizygotic cattle twins with cellular chimerism that was due to naturally occurring placental anastomoses and exchange of non-self antigens (2). Anderson et al. then showed that skin grafts between these calves were well accepted (3). Since then, the concepts of neonatal immune tolerance and transplant tolerance were first described (4, 5).

T cells play an essential role in neonatal immune tolerance. Thymectomy at day 3 (d3Tx) after birth quickly leads to the development of an autoimmune wasting disease in mice which could be rescued by a thymus transplant (6, 7). At the neonatal period (from birth through the first month of life in human or the first 1–2 weeks in mice), T cells are exposed to a rapidly and dramatically changing environment, not only from the thymus to peripheral tissues with variable maturity, but also from a relatively pathogen-free and stable environment in utero to the diverse microbial environment in the outside world. During this period, T cells need to avoid rejection of the maternal host, limit autoimmune responses, tolerate inert environmental and food antigens and antigens from non-harmful commensal microorganisms, promote maturation of mucosal barrier function, yet mount an appropriate response to pathogenic microorganisms (8). The clonal deletion of autoreactive T cells in the thymus (central tolerance) (9, 10) and the suppressive activity of regulatory T cells (Tregs) in the periphery (peripheral tolerance) (1115) are both crucial in immune tolerance. But the mechanisms underlying the uniqueness of neonatal T cell tolerance and its adaptation to the adult state are just beginning to be understood after decades of comparison between neonatal and adult T cells. In this review, we will summarize current knowledge on T cell tolerance in early life and subsequent advantages of umbilical cord blood (UCB) T cells in tolerance development in allogeneic HSCT.

T Cell Repertoire Before Thymic Selection in Early Life

The stepwise T cell development, selection, and the generation of a functional T cell repertoire occur in the thymus (16). Compared to adult T cells, both human and murine neonatal conventional T (Tconv) cells and Treg cells have shorter T cell receptor (TCR) or shorter complementarity determining region (CDR)3α stretches, fewer N-region additions (more germ line-encoded clonotypes), and are less clonally expanded (1727). Human UCB T cells also revealed higher percentage of nonfunctional TCRβ mRNAs, likely due to suppressed nonsense-mediated decay mechanism (26). The shorter TCRs in neonatal T cells do not limit TCR diversity. The results from deep sequencing and single cell sequencing demonstrate higher diversity of TCR repertoire in human neonatal Tconv and Tregs when compared to adult ones (28, 29). In addition, UCB Treg cells are also shown to have more clones with TCRs specific for autoantigens (28).

Terminal deoxynuceotidyl transferase (TdT) is responsible for template-independent nucleotide addition during the V(D)J rearrangement. It contributes to 90% of TCRαβ diversity. The activity of TdT is believed to be low in the fetal period of both humans and mice. In particular, TdT expression could be only detected until 4–5 days after birth in mice and beyond 20th week of gestation in human. Such delayed TdT expression not only makes a significant contribution to short CDR3 length and less N-addition in TCRs of human and murine neonatal T cells (26, 3032), but also leads to relatively high numbers of public clonotypes shared among human UCB samples (26).

In addition to different diversity, neonatal TCR repertoire is also biased toward TCRs with high affinity and high cross-reactivity. This is mainly based on the studies of Tdt-deficient mice but is confirmed later with other mouse models. T cells lacking Tdt showed increased affinity of TCR to the α helices of self-MHC (major histocompatibility complex) (33, 34). One of the surface markers that can report the TCR avidity for peptide/MHC complexes is CD5. Higher levels of CD5 (peaked at day 7 after birth) were found in wild type and several types of mutant murine neonatal Tconv and Tregs when compared to their adult counterparts (35). However, the high affinity between TCRs and self-peptide/MHC complexes did not increase the likelihood to generate autoreactive T cells during neonatal period or incidence of autoimmune pathologies (3638), at least in a rodent model with the transplantation of NOD thymi to NOD.scid mice (39). Instead, it promotes Tregs’ capability to undergo proliferation and likely, to modulate specific immune responses (40, 41). Tdt-deficient T cells also had an increased frequency for a given antigen, including self, commensal, and pathogenic ones (33, 34, 42). Such promiscuous peptide recognition is clearly an advantage to defend against a variety of environmental or infectious insults during neonatal period or during reconstitution after HSCT when the number of peripheral T cells is limited. Indeed, specific and competent CD8+ T cell responses against a range of viral infections (Vesicular Stomatitis Virus, Vaccinia Virus, and Lymphocytic Choriomeningitis Virus) in vivo have been observed in murine Tdt-deficient or neonatal T cells (34, 43, 44). In human samples, T cells in UCB had higher level of CD5 expression and higher precursor frequency for certain tumor-associated antigens or pathogens than T cells in adults (Table 1) (28, 42, 45). Together with delayed TdT expression and similar TCR sequencing feature between human fetal T cells and mouse neonatal T cells, it is believed, although more evidence is needed, that human TCR repertoire also has high cross-reactivity.

TABLE 1
www.frontiersin.org

Table 1 Unique features of human Tconv and Treg cells in umbilical cord blood.

Thymic Selection in Early Life

During thymocyte development, the stochastic V(D)J recombination of TCR α and β chains inevitably generates thymocyte clones with high potential for self-reactivity. These autoreactive clones will either be removed by negative selection or develop into self-reactive thymic Tregs (tTregs) by agonist selection (59, 60). Thymic epithelial cells in the medulla (mTECs) are essential in these thymic selections by displaying a broad spectrum of self-peptide called tissue-specific self antigens (TSAs) to developing T cells (61). The expression of these TSAs in mTECs is regulated, in a significant part, by the transcriptional modulator autoimmune regulator (AIRE). Other regulators include but not limited to the transcription factor forebrain embryonic zinc fingerlike protein 2 (Fezf2) and mTECs’ autophagy machinery (6264). Other cell types in the thymus, including cortical TECs, corneocyte-like mTECs (16), various types of dendritic cells (DC) (6567), and B cells (68, 69), also contribute to negative selection of conventional T (Tconv) cells and agonist selection of tTregs. These different types of antigen presenting cells (APCs), with their different ways to sample and process self antigens, likely have non-redundant roles in thymic selection and in the determination of negative selection versus agonist selection (70, 71).

The uniqueness of thymic selection during neonatal period is not fully understood yet. Most of the evidence so far comes from murine studies. For instance, the interaction of developing thymocytes with medullary APCs may be limited due to small “islands” of thymic medulla in newborn animals in comparison with large and organized structure in adult ones (39). The spectrum of peptide presented by various thymic APCs is also different between neonatal and adult mice. Perinatal mTECs had a much lower ratio of HLA-DO : HLA-DM (non-classical MHC-II molecules that regulate peptide loading of MHC-II) and lower level of CD74/CLIP (MHC-II-associated invariant chain peptide) expression when compared to adult mTECs, indicating that mTECs in young animals have higher efficiency in loading a diverse repertoires of TSA peptides in the antigen-binding grooves of MHC-II molecules (27). MHC-IIhiCD8α+ conventional DC (cDC) that can cross-present diverse TSAs to thymocytes, however, are less in perinatal than in adult thymi (27). The seeding of migratory DCs, including B220+ plasmacytoid DCs and Sirpα+CD11b+ cDCs, to induce negative selection against peripheral self- and non-self antigens in the thymus also takes time, in particular when the number of DCs and the expression levels of MHC-II, CD86, and IL-12p70 in DCs were low during neonatal period (7275).

The impact of the unique antigen presentation in neonatal thymus was demonstrated recently. Tconv cells specific for islet β cells can be observed within 1 week after birth, and the appearance of Tconv and tTreg specific for Peptidyl arginine deiminase, type IV (Padi4) and Adducin 2 (Add2) was restricted to 1–3-week-old mice (39, 76). Beyond the above indicated period, β cell-, Padi4- or Add2-reactive CD4 single positive T cells or tTreg cells were depleted in the thymus. The coincidence of bone marrow (BM)-derived cells accumulating in the thymus beyond weaning age indicates the likelihood of migratory DCs in inducing a late stage negative selection of these autoreactive T cells (76). The second piece of evidence comes from Aire-related studies. Mathis’s group found that the level of Aire expression and the repertories of Aire-dependent transcripts in mTECs were indistinguishable between <3-day-old and 5-week-old mice (27). However, thymectomy at day 3 after birth, turning off Aire expression before or shortly after birth, or tuning on Aire expression only after birth in the inducible Aire transgenic mice quickly led to the wasting disease and multiorgan autoimmune pathology (77), while turning off Aire expression beyond weaning age induced a different spectrum of pathologies (7780). In addition, the multiorgan pathology in Aire-deficient mice could be ameliorated by the adoptive transfer of perinatal Tregs, but not adult Tregs (27). Collectively, these murine studies clearly demonstrate the differences in the antigen presentation machineries and post-selected repertoires between neonatal and adult thymi. Whether different selection machineries also exist in human thymi over the course of a lifespan is not clear. But infants who receive fully allogeneic thymi from unrelated infants generate Treg cells with diverse repertoires and Tconv being tolerant to self as well as the thymic transplant (8183).

Treg Cells in Early Life

Treg cells are an essential mode of immune tolerance that can be transferred into naïve animals to prevent rejection of tissue/cell transplantation, development of autoimmune diseases and atopic disorders, such as allergies (1113, 8486). The importance of Treg cells specifically in fetal tolerance is realized by the onset of IPEX (immune dysregulation, polyendocrinopathy, entheropathy, X-linked)-related autoimmunity at second-trimester in humans that lack functional FOXP3 (87). Using a Foxp3-DTR transgenic mouse system, we and Yang et al. showed that Treg depletion during the day 0–10 or day 7–11 age-window quickly resulted in significant weight loss and autoimmune pathology (27, 41). When Treg cells were depleted beyond weaning age (35–45-day window), only scattered individual mouse developed mild autoimmune inflammation (27). Collectively, these data demonstrate an active and tight control of fetal/neonatal autoimmune responses by Treg cells

In addition to self antigens, Treg cell-mediated immune tolerance to commensal microbiota-derived antigens is also critical at barrier sites. Notably, the preferential barrier sites for neonatal Treg regulation are the intestine in humans but the skin in mouse. In humans, Treg cells with gut tropism (integrin β7 expression) and resting phenotype are found most abundant at birth and decreased with age, while the frequency of Treg cells with skin tropism (cutaneous lymphocyte antigen (CLA) expression) and activated phenotype is increased later in life (55) (Table 1). IL-2 and IL-7, but not retinoic acid, promote the expression of β7 in Treg cells after thymic egress (55). Reduced tTreg cells in UCB were found to be associated with higher susceptibility to food allergies in infants (88). Thus, human neonatal tTreg cells may preferentially migrate to the gut and promote the establishment of mucosal immune tolerance (oral tolerance), in preparing for progressive exposure of microbial, diet, and environmental antigens after birth (89, 90). The reason for the delayed acquisition of skin homing potential in human neonatal Treg cells is not clear. But with impaired barrier function, such as in atopic dermatitis, late coming Tregs may increase the susceptibility to allergen sensitization through the skin (55).

In mouse, however, a unique neonatal Treg population was recently found to migrate to hair follicles and get activated at 1–2 weeks after birth, coinciding with the initial colonization of microbes to the skin (91, 92). Such rapid recruitment of Treg cells in neonatal skin depends on Ccl20–Ccr6 pathway stimulated by commensal bacteria and their surface molecules. Blocking Treg cell entry into hair follicles during neonatal window or colonization of bacteria during adult period all leads to increased antigen-specific effector T cells in the draining lymph nodes, demonstrating the importance of murine neonatal Tregs in promoting immune tolerance to skin commensal microbiota. It further indicates that certain chronic tissue inflammation in adults may be closely associated with impaired tolerance to commensal microbiota established during the neonatal period. Whether murine Treg cells (9396) accumulate in other barrier sites, including lung and gut, during a defined early developmental period is not as clearly studied as the ones in the mouse skin.

A second difference between human and murine Treg cells is the timing of appearance, with the former emerging at gestational week 13 (97, 98) while the latter being detected in the thymus 2–3 days after birth (27, 99, 100). The frequency of human Treg cells in CD4+ T cells significantly increases during the second trimester then decreases during the third trimester. Within the first week after birth, Treg cell ratio rapidly increases again (56, 101, 102). Depletion of CD25+ Treg cells enhanced fetal T cell activation against self and maternal cells, but not against unrelated donor cells (103). Loss of FOXP3 leads to the occurrence of autoimmune inflammation specifically at second-trimester. Thus, the early appearance of human Treg cells in fetus plays a unique but critical role in maintaining self-tolerance as well as feto-maternal tolerance (8, 103, 104).

Murine neonatal Tregs and human fetal Tregs also have common features. They are more proliferative, have higher percentage of activated/effector cells, and perform better in the suppression assay in vitro when compared to adult Treg cells (27, 56). The transcriptome of human neonatal/fetal Tregs is also different from that of adult Treg cells, supporting the enhanced cell division and suppressive functions (57, 58).

Origin of T Cells in Early Life

Although having different dynamics in T cell emergence, the origin of human and murine prenatal/perinatal T cells with distinct intrinsic properties, including short TCR, promiscuous antigen recognition, and high CD5 expression, is the same, i.e. both are derived from hematopoietic stem cells (HSCs) from fetal liver (53, 58, 105108). High expression of Lin28b and high expression of let-7 microRNA mark the difference between fetal liver/thymus and adult BM/thymus, respectively. The detailed in vivo experiments in murine system further demonstrate that ectopic expression of Lin28b or loss of Ezh2 in adult BM hematopoietic stem/progenitor cells (HSPCs) induces activation of fetal-specific genes (including let-7 target genes) in HSPCs and fetal-like lymphopoiesis, including the development of B-1 cells, marginal zone B cells, and γδ T cells (106, 109).

Both human or mouse fetal/neonatal CD4+ T cells preferentially differentiate into induced Tregs (iTregs) when compared to adult CD4+ T cells (58, 103, 110, 111). Inhibiting Lin28b in human fetal CD4+ T cells leads to let-7 upregulation and reduced Treg cell differentiation (112). Human fetal naïve T cells also express higher level of Helios, and deletion of Helios results in impaired Treg differentiation and regulatory function (113). These results demonstrate that fetal liver-derived T cells have unique intrinsic properties to promote Treg cell differentiation.

Persistence of Neonatal T Cells in Adulthood

The uniqueness of neonatal T cells and their roles in immune tolerance are not restricted to early life. Using a fate-mapping model, Yang et al. found that the number and function of murine neonatal Tregs were stably maintained in adulthood (27). Thus, the adoptive transfer of the persisting neonate-derived Treg cells from adult mice suppressed the progression of multi-organ autoimmune pathology in Aire-deficient mice. Similarly, human fetal Treg cells specific for maternal antigens can be found more than a decade later, right into the teenage year (103). Therefore, Treg cells produced during a specific ontogenic window in early life are unique and essential in maintaining self-tolerance in adulthood.

Notably, the persistence of fetal T cells in young adults is not limited to Treg cells. The analysis of deep sequencing data of human TCR repertoire recently reveals that large numbers of naïve T cell clones without N-region addition (fetal origin) are public clones and also persist for decades (114). A better understanding of the impact of these persisting fetal/neonatal T cells on self-tolerance and immune responses against pathogen/tumor in adults will thus be important and may bring benefits in the development of vaccine and therapeutics.

Early-Life T Cell Tolerance and Umbilical Cord Blood Transplantation

Allogeneic HSCT from an HLA-matched related or unrelated donor has been more and more widely used to treat patients with malignant or non-malignant hematological disorders (115). The HSCs used in the transplantation can be derived from BM, peripheral blood, or UCB. Multiple comparisons between the transplantation of UCB and BM/peripheral blood HSCs have shown that UCB grafts are associated with lower incidence of GVHD, and in some cases such as patients with pre-transplant persistent minimal residual disease, better long-term outcomes (116). When CD34+ cells from a third-party HLA-haploidentical donor were transplanted together with unrelated UCB cells, an early haploidentical engraftment was frequently replaced by durable UCB engraftment (117, 118). The distinct features of fetal liver-derived HSCs and Tconv/Treg cells described above may build the basis for these advantages in UCB transplantation (UCBT). Whether T cells reconstituted from UCBT could provide further benefits, such as better self-tolerance and lower incidence of autoimmune diseases later in life, will be an interesting question to investigate.

Concluding Remarks

T cell-mediated immune tolerance is essential in preventing unwanted or self-reactive immune responses throughout life. The distinct features of prenatal/neonatal Tconv and Treg cells provide a unique layer of tolerance against maternal and self antigens, certain allergens, and commensal microbes-derived products. The in-depth investigation of these T cell populations in early life may shed light on a better understanding of the immune responses in infants, the early-life root of certain adult immune alterations, and the choice and prognosis of UCBT in treating hematological disorders.

Author Contributions

LY and QG wrote the manuscript. DL and RJ gave critical comments and revision. All authors contributed to the article and approved the submitted version.

Funding

This work was supported by grants from the National Key Research and Development Program of China (2017YFA0104500), the National Natural Science Foundation of China (32070897, 81471525, 31671244, 31872734), the Foundation for Innovative Research Groups of the National Natural Science Foundation of China (81621001), Beijing Natural Science Foundation (7202079), the Non-Profit Central Research Institute Fund of Chinese Academy of Medical Sciences, 2019PT320006.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Whangbo JS, Antin JH, Koreth J. The role of regulatory T cells in graft-versus-host disease management. Expert Rev Hematol (2020) 13(2):141–54. doi: 10.1080/17474086.2020.1709436

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Owen RD. Immunogenetic Consequences of Vascular Anastomoses between Bovine Twins. Science (1945) 102(2651):400–1. doi: 10.1126/science.102.2651.400

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Anderson D, Billingham RE, Lampkin GH, Medawar PB. The Use of Skin Grafting to Distinguish between Monozygotic and Dizygotic Twins in Cattle. Heredity (1951) 5(3):379–+. doi: 10.1038/hdy.1951.38

CrossRef Full Text | Google Scholar

4. Billingham RE, Brent L, Medawar PB. Actively acquired tolerance of foreign cells. Nature (1953) 172(4379):603–6. doi: 10.1038/172603a0

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Burnet FM, Fenner F. Genetics and Immunology. Heredity (1948) 2(3):289–324. doi: 10.1038/hdy.1948.19

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Miller JFAP. Effect of Neonatal Thymectomy on Immunological Responsiveness of Mouse. Proc R Soc Ser B-Biol Sci (1962) 156(964):415–+. doi: 10.1098/rspb.1962.0048

CrossRef Full Text | Google Scholar

7. Nishizuka Y, Sakakura T. Thymus and reproduction: sex-linked dysgenesia of the gonad after neonatal thymectomy in mice. Science (1969) 166(3906):753–5. doi: 10.1126/science.166.3906.753

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Burt TD. Fetal regulatory T cells and peripheral immune tolerance in utero: implications for development and disease. Am J Reprod Immunol (2013) 69(4):346–58. doi: 10.1111/aji.12083

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Kappler JW, Roehm N, Marrack P. T cell tolerance by clonal elimination in the thymus. Cell (1987) 49(2):273–80. doi: 10.1016/0092-8674(87)90568-x

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Kisielow P, Bluthmann H, Staerz UD, Steinmetz M, von Boehmer H. Tolerance in T-cell-receptor transgenic mice involves deletion of nonmature CD4+8+ thymocytes. Nature (1988) 333(6175):742–6. doi: 10.1038/333742a0

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Fowell D, Mason D. Evidence that the T cell repertoire of normal rats contains cells with the potential to cause diabetes. Characterization of the CD4+ T cell subset that inhibits this autoimmune potential. J Exp Med (1993) 177(3):627–36. doi: 10.1084/jem.177.3.627

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Powrie F, Leach MW, Mauze S, Caddle LB, Coffman RL. Phenotypically distinct subsets of CD4+ T cells induce or protect from chronic intestinal inflammation in C. B-17 scid mice. Int Immunol (1993) 5(11):1461–71. doi: 10.1093/intimm/5.11.1461

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol (1995) 155(3):1151–64.

PubMed Abstract | Google Scholar

14. Thornton AM, Korty PE, Tran DQ, Wohlfert EA, Murray PE, Belkaid Y, et al. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J Immunol (2010) 184(7):3433–41. doi: 10.4049/jimmunol.0904028

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Weiss JM, Bilate AM, Gobert M, Ding Y, Curotto de Lafaille MA, Parkhurst CN, et al. Neuropilin 1 is expressed on thymus-derived natural regulatory T cells, but not mucosa-generated induced Foxp3+ T reg cells. J Exp Med (2012) 209(10):1723–42, S1. doi: 10.1084/jem.20120914

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Kadouri N, Nevo S, Goldfarb Y, Abramson J. Thymic epithelial cell heterogeneity: TEC by TEC. Nat Rev Immunol (2019) 20(4):239–53. doi: 10.1038/s41577-019-0238-0

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Schelonka RL, Raaphorst FM, Infante D, Kraig E, Teale JM, Infante AJ. T cell receptor repertoire diversity and clonal expansion in human neonates. Pediatr Res (1998) 43(3):396–402. doi: 10.1203/00006450-199803000-00015

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Carey AJ, Hope JL, Mueller YM, Fike AJ, Kumova OK, van Zessen DBH, et al. Public Clonotypes and Convergent Recombination Characterize the Naive CD8(+) T-Cell Receptor Repertoire of Extremely Preterm Neonates. Front Immunol (2017) 8:1859. doi: 10.3389/fimmu.2017.01859

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Venturi V, Nzingha K, Amos TG, Charles WC, Dekhtiarenko I, Cicin-Sain L, et al. The Neonatal CD8+ T Cell Repertoire Rapidly Diversifies during Persistent Viral Infection. J Immunol (2016) 196(4):1604–16. doi: 10.4049/jimmunol.1501867

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Rudd BD, Venturi V, Smith NL, Nzingha K, Goldberg EL, Li G, et al. Acute neonatal infections ‘lock-in’ a suboptimal CD8+ T cell repertoire with impaired recall responses. PloS Pathog (2013) 9(9):e1003572. doi: 10.1371/journal.ppat.1003572

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Rudd BD, Venturi V, Davenport MP, Nikolich-Zugich J. Evolution of the antigen-specific CD8+ TCR repertoire across the life span: evidence for clonal homogenization of the old TCR repertoire. J Immunol (2011) 186(4):2056–64. doi: 10.4049/jimmunol.1003013

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Ruckwardt TJ, Malloy AM, Gostick E, Price DA, Dash P, McClaren JL, et al. Neonatal CD8 T-cell hierarchy is distinct from adults and is influenced by intrinsic T cell properties in respiratory syncytial virus infected mice. PloS Pathog (2011) 7(12):e1002377. doi: 10.1371/journal.ppat.1002377

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Feeney AJ. Junctional sequences of fetal T cell receptor beta chains have few N regions. J Exp Med (1991) 174(1):115–24. doi: 10.1084/jem.174.1.115

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Bogue M, Gilfillan S, Benoist C, Mathis D. Regulation of N-region diversity in antigen receptors through thymocyte differentiation and thymus ontogeny. Proc Natl Acad Sci U S A (1992) 89(22):11011–5. doi: 10.1073/pnas.89.22.11011

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Bogue M, Candeias S, Benoist C, Mathis D. A special repertoire of alpha:beta T cells in neonatal mice. EMBO J (1991) 10(12):3647–54. doi: 10.1002/j.1460-2075.1991.tb04931.x

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Britanova OV, Shugay M, Merzlyak EM, Staroverov DB, Putintseva EV, Turchaninova MA, et al. Dynamics of Individual T Cell Repertoires: From Cord Blood to Centenarians. J Immunol (2016) 196(12):5005–13. doi: 10.4049/jimmunol.1600005

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Yang S, Fujikado N, Kolodin D, Benoist C, Mathis D. Immune tolerance. Regulatory T cells generated early in life play a distinct role in maintaining self-tolerance. Science (2015) 348(6234):589–94. doi: 10.1126/science.aaa7017

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Motwani K, Peters LD, Vliegen WH, El-Sayed AG, Seay HR, Lopez MC, et al. Human Regulatory T Cells From Umbilical Cord Blood Display Increased Repertoire Diversity and Lineage Stability Relative to Adult Peripheral Blood. Front Immunol (2020) 11:611. doi: 10.3389/fimmu.2020.00611

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Gao K, Chen L, Zhang Y, Zhao Y, Wan Z, Wu J, et al. Germline-Encoded TCR-MHC Contacts Promote TCR V Gene Bias in Umbilical Cord Blood T Cell Repertoire. Front Immunol (2019) 10:2064. doi: 10.3389/fimmu.2019.02064

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Cabaniols JP, Fazilleau N, Casrouge A, Kourilsky P, Kanellopoulos JM. Most alpha/beta T cell receptor diversity is due to terminal deoxynucleotidyl transferase. J Exp Med (2001) 194(9):1385–90. doi: 10.1084/jem.194.9.1385

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Gilfillan S, Dierich A, Lemeur M, Benoist C, Mathis D. Mice lacking TdT: mature animals with an immature lymphocyte repertoire. Science (1993) 261(5125):1175–8. doi: 10.1126/science.8356452

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Bonati A, Zanelli P, Ferrari S, Plebani A, Starcich B, Savi M, et al. T-cell receptor beta-chain gene rearrangement and expression during human thymic ontogenesis. Blood (1992) 79(6):1472–83. doi: 10.1182/blood.V79.6.1472.bloodjournal7961472

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Rudd BD. Neonatal T Cells: A Reinterpretation. Annu Rev Immunol (2020) 38:229–47. doi: 10.1146/annurev-immunol-091319-083608

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Gavin MA, Bevan MJ. Increased peptide promiscuity provides a rationale for the lack of N regions in the neonatal T cell repertoire. Immunity (1995) 3(6):793–800. doi: 10.1016/1074-7613(95)90068-3

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Dong M, Artusa P, Kelly SA, Fournier M, Baldwin TA, Mandl JN, et al. Alterations in the Thymic Selection Threshold Skew the Self-Reactivity of the TCR Repertoire in Neonates. J Immunol (2017) 199(3):965–73. doi: 10.4049/jimmunol.1602137

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Conde C, Weller S, Gilfillan S, Marcellin L, Martin T, Pasquali JL. Terminal deoxynucleotidyl transferase deficiency reduces the incidence of autoimmune nephritis in (New Zealand Black x New Zealand White)F1 mice. J Immunol (1998) 161(12):7023–30.

PubMed Abstract | Google Scholar

37. Feeney AJ, Lawson BR, Kono DH, Theofilopoulos AN. Terminal deoxynucleotidyl transferase deficiency decreases autoimmune disease in MRL-Fas(lpr) mice. J Immunol (2001) 167(6):3486–93. doi: 10.4049/jimmunol.167.6.3486

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Robey IF, Peterson M, Horwitz MS, Kono DH, Stratmann T, Theofilopoulos AN, et al. Terminal deoxynucleotidyltransferase deficiency decreases autoimmune disease in diabetes-prone nonobese diabetic mice and lupus-prone MRL-Fas(lpr) mice. J Immunol (2004) 172(7):4624–9. doi: 10.4049/jimmunol.172.7.4624

PubMed Abstract | CrossRef Full Text | Google Scholar

39. He Q, Morillon YM,2, Spidale NA, Kroger CJ, Liu B, Sartor RB, et al. Thymic development of autoreactive T cells in NOD mice is regulated in an age-dependent manner. J Immunol (2013) 191(12):5858–66. doi: 10.4049/jimmunol.1302273

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Tuovinen H, Laurinolli TT, Rossi LH, Pekkarinen PT, Mattila I, Arstila TP. Thymic production of human FOXP3(+) regulatory T cells is stable but does not correlate with peripheral FOXP3 expression. Immunol Lett (2008) 117(2):146–53. doi: 10.1016/j.imlet.2008.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Li M, Zhao W, Wang Y, Jin L, Jin G, Sun X, et al. A wave of Foxp3(+) regulatory T cell accumulation in the neonatal liver plays unique roles in maintaining self-tolerance. Cell Mol Immunol (2019) 17(5):507–18. doi: 10.1038/s41423-019-0246-9

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Mandl JN, Monteiro JP, Vrisekoop N, Germain RN. T cell-positive selection uses self-ligand binding strength to optimize repertoire recognition of foreign antigens. Immunity (2013) 38(2):263–74. doi: 10.1016/j.immuni.2012.09.011

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Gilfillan S, Bachmann M, Trembleau S, Adorini L, Kalinke U, Zinkernagel R, et al. Efficient immune responses in mice lacking N-region diversity. Eur J Immunol (1995) 25(11):3115–22. doi: 10.1002/eji.1830251119

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Robins HS, Srivastava SK, Campregher PV, Turtle CJ, Andriesen J, Riddell SR, et al. Overlap and effective size of the human CD8+ T cell receptor repertoire. Sci Transl Med (2010) 2(47):47ra64. doi: 10.1126/scitranslmed.3001442

PubMed Abstract | CrossRef Full Text | Google Scholar

45. St John LS, Wan L, He H, Garber HR, Clise-Dwyer K, Alatrash G, et al. PR1-specific cytotoxic T lymphocytes are relatively frequent in umbilical cord blood and can be effectively expanded to target myeloid leukemia. Cytotherapy (2016) 18(8):995–1001. doi: 10.1016/j.jcyt.2016.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

46. D’Arena G, Musto P, Cascavilla N, Di Giorgio G, Fusilli S, Zendoli F, et al. Flow cytometric characterization of human umbilical cord blood lymphocytes: immunophenotypic features. Haematologica (1998) 83(3):197–203.

PubMed Abstract | Google Scholar

47. Grozdics E, Berta L, Gyarmati B, Veres G, Zadori D, Szalardy L, et al. B7 costimulation and intracellular indoleamine 2,3-dioxygenase expression in umbilical cord blood and adult peripheral blood. Biol Blood Marrow Transplant (2014) 20(10):1659–65. doi: 10.1016/j.bbmt.2014.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Walk J, Westerlaken GH, van Uden NO, Belderbos ME, Meyaard L, Bont LJ. Inhibitory receptor expression on neonatal immune cells. Clin Exp Immunol (2012) 169(2):164–71. doi: 10.1111/j.1365-2249.2012.04599.x

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Jacks RD, Keller TJ, Nelson A, Nishimura M II, White P, Iwashima M. Cell intrinsic characteristics of human cord blood naive CD4T cells. Immunol Lett (2018) 193:51–7. doi: 10.1016/j.imlet.2017.11.011

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Kwoczek J, Riese SB, Tischer S, Bak S, Lahrberg J, Oelke M, et al. Cord blood-derived T cells allow the generation of a more naive tumor-reactive cytotoxic T-cell phenotype. Transfusion (2018) 58(1):88–99. doi: 10.1111/trf.14365

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Fike AJ, Kumova OK, Carey AJ. Dissecting the defects in the neonatal CD8(+) T-cell response. J Leukoc Biol (2019) 106(5):1051–61. doi: 10.1002/JLB.5RU0319-105R

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Hiwarkar P, Qasim W, Ricciardelli I, Gilmour K, Quezada S, Saudemont A, et al. Cord blood T cells mediate enhanced antitumor effects compared with adult peripheral blood T cells. Blood (2015) 126(26):2882–91. doi: 10.1182/blood-2015-06-654780

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Smith NL, Patel RK, Reynaldi A, Grenier JK, Wang J, Watson NB, et al. Developmental Origin Governs CD8(+) T Cell Fate Decisions during Infection. Cell (2018) 174(1):117–130 e14. doi: 10.1016/j.cell.2018.05.029

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Galindo-Albarran AO, Lopez-Portales OH, Gutierrez-Reyna DY, Rodriguez-Jorge O, Sanchez-Villanueva JA, Ramirez-Pliego O, et al. CD8(+) T Cells from Human Neonates Are Biased toward an Innate Immune Response. Cell Rep (2016) 17(8):2151–60. doi: 10.1016/j.celrep.2016.10.056

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Hsu PS, Lai CL, Hu M, Santner-Nanan B, Dahlstrom JE, Lee CH, et al. IL-2 Enhances Gut Homing Potential of Human Naive Regulatory T Cells Early in Life. J Immunol (2018) 200(12):3970–80. doi: 10.4049/jimmunol.1701533

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Hayakawa S, Ohno N, Okada S, Kobayashi M. Significant augmentation of regulatory T cell numbers occurs during the early neonatal period. Clin Exp Immunol (2017) 190(2):268–79. doi: 10.1111/cei.13008

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Zahran AM, Saad K, Abdel-Raheem YF, Elsayh K II, El-Houfey AA, Aboul-Khair MD, et al. Characterization of Regulatory T Cells in Preterm and Term Infants. Arch Immunol Ther Exp (Warsz) (2019) 67(1):49–54. doi: 10.1007/s00005-018-0530-x

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Mold JE, Venkatasubrahmanyam S, Burt TD, Michaelsson J, Rivera JM, Galkina SA, et al. Fetal and adult hematopoietic stem cells give rise to distinct T cell lineages in humans. Science (2010) 330(6011):1695–9. doi: 10.1126/science.1196509

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Cheng M, Anderson MS. Thymic tolerance as a key brake on autoimmunity. Nat Immunol (2018) 19(7):659–64. doi: 10.1038/s41590-018-0128-9

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Owen DL, Sjaastad LE, Farrar MA. Regulatory T Cell Development in the Thymus. J Immunol (2019) 203(8):2031–41. doi: 10.4049/jimmunol.1900662

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Aschenbrenner K, D’Cruz LM, Vollmann EH, Hinterberger M, Emmerich J, Swee LK, et al. Selection of Foxp3+ regulatory T cells specific for self antigen expressed and presented by Aire+ medullary thymic epithelial cells. Nat Immunol (2007) 8(4):351–8. doi: 10.1038/ni1444

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Perniola R. Twenty Years of AIRE. Front Immunol (2018) 9:98. doi: 10.3389/fimmu.2018.00098

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Takaba H, Morishita Y, Tomofuji Y, Danks L, Nitta T, Komatsu N, et al. Fezf2 Orchestrates a Thymic Program of Self-Antigen Expression for Immune Tolerance. Cell (2015) 163(4):975–87. doi: 10.1016/j.cell.2015.10.013

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Liang Z, Zhang L, Su H, Luan R, Na N, Sun L, et al. MTOR signaling is essential for the development of thymic epithelial cells and the induction of central immune tolerance. Autophagy (2018) 14(3):505–17. doi: 10.1080/15548627.2017.1376161

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Guerri L, Peguillet I, Geraldo Y, Nabti S, Premel V, Lantz O. Analysis of APC types involved in CD4 tolerance and regulatory T cell generation using reaggregated thymic organ cultures. J Immunol (2013) 190(5):2102–10. doi: 10.4049/jimmunol.1202883

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Leventhal DS, Gilmore DC, Berger JM, Nishi S, Lee V, Malchow S, et al. Dendritic Cells Coordinate the Development and Homeostasis of Organ-Specific Regulatory T Cells. Immunity (2016) 44(4):847–59. doi: 10.1016/j.immuni.2016.01.025

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Hadeiba H, Lahl K, Edalati A, Oderup C, Habtezion A, Pachynski R, et al. Plasmacytoid dendritic cells transport peripheral antigens to the thymus to promote central tolerance. Immunity (2012) 36(3):438–50. doi: 10.1016/j.immuni.2012.01.017

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Frommer F, Waisman A. B cells participate in thymic negative selection of murine auto-reactive CD4+ T cells. PloS One (2010) 5(10):e15372. doi: 10.1371/journal.pone.0015372

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Fujihara C, Williams JA, Watanabe M, Jeon H, Sharrow SO, Hodes RJ. T cell-B cell thymic cross-talk: maintenance and function of thymic B cells requires cognate CD40-CD40 ligand interaction. J Immunol (2014) 193(11):5534–44. doi: 10.4049/jimmunol.1401655

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Klein L, Kyewski B, Allen PM, Hogquist KA. Positive and negative selection of the T cell repertoire: what thymocytes see (and don’t see). Nat Rev Immunol (2014) 14(6):377–91. doi: 10.1038/nri3667

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Klein L, Robey EA, Hsieh CS. Central CD4(+) T cell tolerance: deletion versus regulatory T cell differentiation. Nat Rev Immunol (2019) 19(1):7–18. doi: 10.1038/s41577-018-0083-6

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Li J, Park J, Foss D, Goldschneider I. Thymus-homing peripheral dendritic cells constitute two of the three major subsets of dendritic cells in the steady-state thymus. J Exp Med (2009) 206(3):607–22. doi: 10.1084/jem.20082232

PubMed Abstract | CrossRef Full Text | Google Scholar

73. De Wit D, Tonon S, Olislagers V, Goriely S, Boutriaux M, Goldman M, et al. Impaired responses to toll-like receptor 4 and toll-like receptor 3 ligands in human cord blood. J Autoimmun (2003) 21(3):277–81. doi: 10.1016/j.jaut.2003.08.003

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Langrish CL, Buddle JC, Thrasher AJ, Goldblatt D. Neonatal dendritic cells are intrinsically biased against Th-1 immune responses. Clin Exp Immunol (2002) 128(1):118–23. doi: 10.1046/j.1365-2249.2002.01817.x

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Willems F, Vollstedt S, Suter M. Phenotype and function of neonatal DC. Eur J Immunol (2009) 39(1):26–35. doi: 10.1002/eji.200838391

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Stadinski BD, Blevins SJ, Spidale NA, Duke BR, Huseby PG, Stern LJ, et al. A temporal thymic selection switch and ligand binding kinetics constrain neonatal Foxp3(+) Treg cell development. Nat Immunol (2019) 20(8):1046–58. doi: 10.1038/s41590-019-0414-1

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Guerau-de-Arellano M, Martinic M, Benoist C, Mathis D. Neonatal tolerance revisited: a perinatal window for Aire control of autoimmunity. J Exp Med (2009) 206(6):1245–52. doi: 10.1084/jem.20090300

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Fontenot JD, Rasmussen JP, Williams LM, Dooley JL, Farr AG, Rudensky AY. Regulatory T cell lineage specification by the forkhead transcription factor foxp3. Immunity (2005) 22(3):329–41. doi: 10.1016/j.immuni.2005.01.016

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Kim JM, Rasmussen JP, Rudensky AY. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol (2007) 8(2):191–7. doi: 10.1038/ni1428

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Chen Z, Benoist C, Mathis D. How defects in central tolerance impinge on a deficiency in regulatory T cells. Proc Natl Acad Sci U S A (2005) 102(41):14735–40. doi: 10.1073/pnas.0507014102

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Markert ML, Devlin BH, McCarthy EA. Thymus transplantation. Clin Immunol (2010) 135(2):236–46. doi: 10.1016/j.clim.2010.02.007

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Chinn IK, Milner JD, Scheinberg P, Douek DC, Markert ML. Thymus transplantation restores the repertoires of forkhead box protein 3 (FoxP3)+ and FoxP3- T cells in complete DiGeorge anomaly. Clin Exp Immunol (2013) 173(1):140–9. doi: 10.1111/cei.12088

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Li B, Li J, Devlin BH, Markert ML. Thymic microenvironment reconstitution after postnatal human thymus transplantation. Clin Immunol (2011) 140(3):244–59. doi: 10.1016/j.clim.2011.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Modigliani Y, Thomas-Vaslin V, Bandeira A, Coltey M, Le Douarin NM, Coutinho A, et al. Lymphocytes selected in allogeneic thymic epithelium mediate dominant tolerance toward tissue grafts of the thymic epithelium haplotype. Proc Natl Acad Sci U S A (1995) 92(16):7555–9. doi: 10.1073/pnas.92.16.7555

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Ohki H, Martin C, Corbel C, Coltey M, Le Douarin NM. Tolerance induced by thymic epithelial grafts in birds. Science (1987) 237(4818):1032–5. doi: 10.1126/science.3616623

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Salaun J, Bandeira A, Khazaal I, Calman F, Coltey M, Coutinho A, et al. Thymic epithelium tolerizes for histocompatibility antigens. Science (1990) 247(4949 Pt 1):1471–4. doi: 10.1126/science.2321009

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Bacchetta R, Barzaghi F, Roncarolo MG. From IPEX syndrome to FOXP3 mutation: a lesson on immune dysregulation. Ann N Y Acad Sci (2018) 1417(1):5–22. doi: 10.1111/nyas.13011

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Zhang Y, Collier F, Naselli G, Saffery R, Tang ML, Allen KJ, et al. Cord blood monocyte-derived inflammatory cytokines suppress IL-2 and induce nonclassic “T(H)2-type” immunity associated with development of food allergy. Sci Transl Med (2016) 8(321):321ra8. doi: 10.1126/scitranslmed.aad4322

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Fujimura KE, Sitarik AR, Havstad S, Lin DL, Levan S, Fadrosh D, et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat Med (2016) 22(10):1187–91. doi: 10.1038/nm.4176

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Riedler J, Braun-Fahrlander C, Eder W, Schreuer M, Waser M, Maisch S, et al. Exposure to farming in early life and development of asthma and allergy: a cross-sectional survey. Lancet (2001) 358(9288):1129–33. doi: 10.1016/S0140-6736(01)06252-3

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Scharschmidt TC, Vasquez KS, Truong HA, Gearty SV, Pauli ML, Nosbaum A, et al. A Wave of Regulatory T Cells into Neonatal Skin Mediates Tolerance to Commensal Microbes. Immunity (2015) 43(5):1011–21. doi: 10.1016/j.immuni.2015.10.016

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Scharschmidt TC, Vasquez KS, Pauli ML, Leitner EG, Chu K, Truong HA, et al. Commensal Microbes and Hair Follicle Morphogenesis Coordinately Drive Treg Migration into Neonatal Skin. Cell Host Microbe (2017) 21(4):467–477 e5. doi: 10.1016/j.chom.2017.03.001

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, Santacruz N, et al. Peripheral education of the immune system by colonic commensal microbiota. Nature (2011) 478(7368):250–4. doi: 10.1038/nature10434

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Cebula A, Seweryn M, Rempala GA, Pabla SS, McIndoe RA, Denning TL, et al. Thymus-derived regulatory T cells contribute to tolerance to commensal microbiota. Nature (2013) 497(7448):258–62. doi: 10.1038/nature12079

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Powell BR, Buist NR, Stenzel P. An X-linked syndrome of diarrhea, polyendocrinopathy, and fatal infection in infancy. J Pediatr (1982) 100(5):731–7. doi: 10.1016/s0022-3476(82)80573-8

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Gollwitzer ES, Saglani S, Trompette A, Yadava K, Sherburn R, McCoy KD, et al. Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat Med (2014) 20(6):642–7. doi: 10.1038/nm.3568

PubMed Abstract | CrossRef Full Text | Google Scholar

97. 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(2):383–90. doi: 10.1002/eji.200425763

PubMed Abstract | CrossRef Full Text | Google Scholar

98. 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(12):4715–21. doi: 10.1182/blood-2004-10-4051

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Fontenot JD, Dooley JL, Farr AG, Rudensky AY. Developmental regulation of Foxp3 expression during ontogeny. J Exp Med (2005) 202(7):901–6. doi: 10.1084/jem.20050784

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Konkel JE, Jin W, Abbatiello B, Grainger JR, Chen W. Thymocyte apoptosis drives the intrathymic generation of regulatory T cells. Proc Natl Acad Sci U S A (2014) 111(4):E465–73. doi: 10.1073/pnas.1320319111

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Takahata Y, Nomura A, Takada H, Ohga S, Furuno K, Hikino S, 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(7):622–9. doi: 10.1016/j.exphem.2004.03.012

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Grindebacke H, Stenstad H, Quiding-Jarbrink M, Waldenstrom J, Adlerberth I, Wold AE, et al. Dynamic development of homing receptor expression and memory cell differentiation of infant CD4+CD25high regulatory T cells. J Immunol (2009) 183(7):4360–70. doi: 10.4049/jimmunol.0901091

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Mold JE, Michaelsson J, Burt TD, Muench MO, Beckerman KP, Busch MP, et al. Maternal alloantigens promote the development of tolerogenic fetal regulatory T cells in utero. Science (2008) 322(5907):1562–5. doi: 10.1126/science.1164511

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Michaelsson J, Mold JE, McCune JM, Nixon DF. Regulation of T cell responses in the developing human fetus. J Immunol (2006) 176(10):5741–8. doi: 10.4049/jimmunol.176.10.5741

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Ikuta K, Kina T, MacNeil I, Uchida N, Peault B, Chien YH, et al. A developmental switch in thymic lymphocyte maturation potential occurs at the level of hematopoietic stem cells. Cell (1990) 62(5):863–74. doi: 10.1016/0092-8674(90)90262-d

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Yuan J, Nguyen CK, Liu X, Kanellopoulou C, Muljo SA. Lin28b reprograms adult bone marrow hematopoietic progenitors to mediate fetal-like lymphopoiesis. Science (2012) 335(6073):1195–200. doi: 10.1126/science.1216557

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Adkins B. Peripheral CD4+ lymphocytes derived from fetal versus adult thymic precursors differ phenotypically and functionally. J Immunol (2003) 171(10):5157–64. doi: 10.4049/jimmunol.171.10.5157

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Wang J, Wissink EM, Watson NB, Smith NL, Grimson A, Rudd BD. Fetal and adult progenitors give rise to unique populations of CD8+ T cells. Blood (2016) 128(26):3073–82. doi: 10.1182/blood-2016-06-725366

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Oshima M, Hasegawa N, Mochizuki-Kashio M, Muto T, Miyagi S, Koide S, et al. Ezh2 regulates the Lin28/let-7 pathway to restrict activation of fetal gene signature in adult hematopoietic stem cells. Exp Hematol (2016) 44(4):282–96 e3. doi: 10.1016/j.exphem.2015.12.009

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Wang G, Miyahara Y, Guo Z, Khattar M, Stepkowski SM, Chen W. “Default” generation of neonatal regulatory T cells. J Immunol (2010) 185(1):71–8. doi: 10.4049/jimmunol.0903806

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Do JS, Zhong F, Huang AY, Van’t Hof WJ, Finney M, Laughlin MJ. Foxp3 expression in induced T regulatory cells derived from human umbilical cord blood vs. adult peripheral blood. Bone Marrow Transplant (2018) 53(12):1568–77. doi: 10.1038/s41409-018-0205-6

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Bronevetsky Y, Burt TD, McCune JM. Lin28b Regulates Fetal Regulatory T Cell Differentiation through Modulation of TGF-beta Signaling. J Immunol (2016) 197(11):4344–50. doi: 10.4049/jimmunol.1601070

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Ng MSF, Roth TL, Mendoza VF, Marson A, Burt TD. Helios enhances the preferential differentiation of human fetal CD4(+) naive T cells into regulatory T cells. Sci Immunol (2019) 4(41):eaav5947. doi: 10.1126/sciimmunol.aav5947

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Pogorelyy MV, Elhanati Y, Marcou Q, Sycheva AL, Komech EA, Nazarov V II, et al. Persisting fetal clonotypes influence the structure and overlap of adult human T cell receptor repertoires. PloS Comput Biol (2017) 13(7):e1005572. doi: 10.1371/journal.pcbi.1005572

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Copelan EA. Hematopoietic stem-cell transplantation. N Engl J Med (2006) 354(17):1813–26. doi: 10.1056/NEJMra052638

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Algeri M, Gaspari S, Locatelli F. Cord blood transplantation for acute leukemia. Expert Opin Biol Ther (2020) 107(5):513–8. doi: 10.1007/s12185-018-2412-8

CrossRef Full Text | Google Scholar

117. Liu H, Rich ES, Godley L, Odenike O, Joseph L, Marino S, et al. Reduced-intensity conditioning with combined haploidentical and cord blood transplantation results in rapid engraftment, low GVHD, and durable remissions. Blood (2011) 118(24):6438–45. doi: 10.1182/blood-2011-08-372508

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Magro E, Regidor C, Cabrera R, Sanjuan I, Fores R, Garcia-Marco JA, et al. Early hematopoietic recovery after single unit unrelated cord blood transplantation in adults supported by co-infusion of mobilized stem cells from a third party donor. Haematologica (2006) 91(5):640–8.

PubMed Abstract | Google Scholar

Keywords: neonatal period, T cell tolerance, regulatory T cells, conventional T cells, allogeneic hematopoietic stem cell transplantation

Citation: Yang L, Jin R, Lu D and Ge Q (2020) T cell Tolerance in Early Life. Front. Immunol. 11:576261. doi: 10.3389/fimmu.2020.576261

Received: 25 June 2020; Accepted: 21 October 2020;
Published: 20 November 2020.

Edited by:

William Ying Khee Hwang, National Cancer Centre Singapore, Singapore

Reviewed by:

Simrit Parmar, University of Texas MD Anderson Cancer Center, United States
Nelson Chao, Duke University, United States

Copyright © 2020 Yang, Jin, Lu and Ge. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Qing Ge, qingge@bjmu.edu.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.