Open access peer-reviewed chapter

Lymphoid Hematopoiesis and Lymphocytes Differentiation and Maturation

Written By

Luciana Cavalheiro Marti, Nydia Strachman Bacal, Laiz Camerão Bento, Rodolfo Patussi Correia and Fernanda Agostini Rocha

Reviewed: 07 April 2017 Published: 12 July 2017

DOI: 10.5772/intechopen.69058

From the Edited Volume

Lymphocyte Updates - Cancer, Autoimmunity and Infection

Edited by Gheorghita Isvoranu

Chapter metrics overview

3,347 Chapter Downloads

View Full Metrics

Abstract

Lymphocytes belong to the lymphoid lineage and are considered as divergent from other blood cells lineages as those from the myeloid or erythroid lineage. Lymphoid hematopoiesis is not trivial, because although lymphocytes are found in the bloodstream and their precursor originates in the bone marrow, they mainly belong to the separate lymphatic system, which interacts with the blood circulation. We will discuss B cell differentiation in the bone marrow and the later stages of maturation in secondary lymphoid tissues, besides the B cell profiles in interfollicular, perifollicular, and follicular areas. In addition, we will also discuss T-cell precursor and natural killer cells derivation in the marrow. Furthermore, we will also discuss T-cell precursor migration to thymus, differentiation, rearrangement, thymic selection, involved transcription factors, and, finally, T-cell profiles and subsets in secondary lymphoid organs. We will provide flow cytometry plots showing strategies to identify and characterize NK, T and B lymphocytes and their subsets in circulation. Furthermore, we will provide illustrations to help the reader to understand and visualize the information provide over the chapter. Furthermore, the comprehension about lymphocytes and their contribution to the immune response will favor their application in developmental hematology and immunology. These topics are very important for the continuous development of knowledge.

Keywords

  • hematopoiesis
  • B cells
  • T cells
  • differentiation

1. Introduction

Lymphocytes belong to the lymphoid lineage and are considered as divergent from other blood cells lineages as those from the myeloid or erythroid lineage. Lymphoid hematopoiesis is not trivial, because although lymphocytes are found in the bloodstream and their precursor originates in the bone marrow, they mostly belong to the separate lymphatic system, which interacts with the blood circulation. Lymphoid and myeloid lineages are separated at the progenitor level; the common lymphoid progenitors (CLPs) can differentiate into all types of lymphocytes but lack the myeloid potential under physiological conditions, although some myeloid-related genes can be detected in CLPs depending on experimental conditions [1, 2].

After transplanting a single hematopoietic stem cell (HSC) into an irradiated mouse, long‐term reconstitution of both lymphoid and myeloid compartments was achieved demonstrating that the HSC is the common predecessor of all blood cells [35].

Hematopoiesis studies, mainly in the fetal liver indicated that the difference between the lymphoid and myeloid lineages may not be as simple as imagined. Nevertheless, a corresponding adult common myeloid progenitor (CMP) has been recognized in a fetal liver, but the presence of a lymphoid‐committed progenitor (CLPs) has not yet been demonstrated [6, 7].

The bi‐directional T and B cell derivation from a single cell was not found in the fetal liver progenitors at clonal levels, but T cell and granulocytes and macrophages (GM) or B cell and GM progenitor ability of differentiation were present [8]. Regarding these findings, it has been wondered whether these differences observed in fetal and adult hematopoiesis are due to different intrinsic mechanisms in fetal and adult progenitors or due to the liver and bone marrow environmental differences.

Several hematopoietic and lymphoid progenitors, such as multipotent progenitors (MPPs) and CLPs, are mobilized from bone marrow and initiate T‐cell development in the thymus [9]. MPPs CCR9+ are the major bone marrow population that transit to the thymus. Once the hematopoietic progenitor cell home to the thymus, B cell development potential is immediately turned off by stimulation through Notch, while B cell differentiation will be regulated by several transcription factors in the bone marrow.

Next, we will discuss B lymphocyte differentiation in the bone marrow and later their stages of maturation in secondary lymphoid tissues and profiles in interfollicular, perifollicular and follicular areas. In addition, we will also discuss T‐cell precursor and natural killer cell derivation in the marrow. Furthermore, we will also discuss T-cell precursor migration to thymus, differentiation, rearrangement, thymic selection, involved transcription factors, and, finally, T-cell profiles and subsets in secondary lymphoid organs.

1.1. B Lymphocytes

1.1.1. B Lymphocytes ontogeny

Lymphocytes are cells from the adaptive immune system and are derived from hematopoietic progenitor cells. These cells are first produced in the yolk sac, next, they are formed in the liver during the fetal phase and lastly in the bone marrow [10]. B cell development is dependent of several specific cytokines and contact with a favorable microenvironment [11, 12].

The hematopoietic progenitor cell differentiates into a CLP and depending on stimuli they will give rise to a T or a B lymphocyte [13]. B cell differentiation from a CLP is regulated by several factors such as E‐box binding protein 2A (E2A), early B cell factor‐1 (EBF1), purine box factor 1 (PU.1), Ikaros, paired box protein-5 (Pax5) and CXCL12. These factors are resulting from the interaction of interleukin 7 (IL‐7) and their receptor CD127 (IL‐7Rα) existent in the B lymphocyte. Together, these factors are crucial for the hematopoietic progenitor cell commitment to B lymphocyte differentiation and gene rearrangement to the immunoglobulin heavy chain [1421].

The first cell committed to B‐lymphoid differentiation is the pro‐B cell. The presence of stromal cells and IL‐7 also favors the B lymphocyte differentiation. These cells begin to express CD45dim, CD22, CD34, terminal deoxynucleotidyl transferase (TdT) and CD38high [19]. The Pax5 factor activates the CD19 expression, one of the most premature B lymphocyte antigens [18, 22]. In the next step of differentiation, these cells express CD10high, CD38high, CD34, CD79a, TdT and start being called pre‐B I [19, 20]. Immunoglobulin (Ig) gene recombination in the heavy chain locus starts in this phase. The heavy chain gene is present in segments that code for the variable (V), diversity (D), joining (J), and constant (C) regions [23, 24]. Gene recombination is initiated by recombinase activating gene proteins RAG1 and RAG2; these proteins have the ability to bind and cleave DNA at specific recombination signal sequences called RSSs.

These RSSs surround each genic segment V, D and J. During recombination, D and J gene segments are drawn closer, excluding the intermediary DNA, and this DJ segment is joined to a V segment originating VDJ rearranged exon. Pre-B I cells also express TdT, which is responsible for catalyzing the random addition of junctional (N) nucleotides [24, 25]. The VDJ rearrangement is now adjacent to the constant Cµ region and creates an active gene which codes for the heavy chain, whose synthesis originates the µ intracytoplasmic chain (IgM) and from now these cells become termed pre-B II [19].

During pre‐B II cells, they gain heterogeneous CD20 expression and lack the expression of CD34 and TdT [19, 20, 26]. Besides, B lymphocyte expresses a complex known as a pre‐B cell receptor (pre‐BCR) that is formed by the heavy chain μ (Igμ) associated to a light chain (λ5 and pre‐B V) joined to a heterodimer Igα (CD79a) and Igβ (CD79b) [27]. If the pre‐BCR is able to bind to the bone marrow microenvironment, the immune‐receptor tyrosine‐based activation motif (ITAMs) domains of Igα and Igβ are phosphorylated and their signaling rescues these cells from apoptosis. This positive selection confirms that the Igμ chain generated is functional, and the pre‐B II cells detected with Igμ not functional are deleted [19]. The signals generated by the pre‐BCR also stimulated the pre‐B II differentiation and are responsible for their proliferation in the bone marrow, inhibiting the heavy chain recombination and stimulating the light chain recombination [28, 29]. The light chain rearrangement comprises the junction of a V with a J segment forming a VJ exon associated with an Igμ chain. This association activates the translation of kappa and lambda proteins leading to a formation of a complete IgM molecule [30, 31]. The B cell that expresses an IgM molecule on the cell membrane is denominated immature B cell [19].

Immature B lymphocytes begin to express some immunophenotypic markers of still naïve but mature B lymphocytes; among these characteristics we can quote the CD20 expression, enhanced expression of CD45, CD10dim and CD38high, low expression of CD21, CD5 (homogeneous), and high levels of CD81 [19, 26].

These cells undergo a positive and negative selection process before completing their maturation status. During this process, the B lymphocytes that complete, successfully, the gene rearrangement program are positively selected, there are the shut‐down of RAG genes and these cells will receive survival signals to proceed in the maturation process [32]. Though, when the B lymphocyte recognizes self‐antigens, their receptor is modified, the genes RAG are reactivated and another rearrangement in the light chain V‐J is initiated allowing the B cell to develop a non‐self‐reactive BCR [3234]. However, if this rearrangement did not succeed this cell undergo through apoptosis in a process known as negative selection. The positively selected cells leave the bone marrow and complete their maturation status in the secondary lymphoid organs.

Mature B lymphocytes lack CD10 and CD38 and express IgD and IgM on their membrane (Figure 1) [19]. IgD expression happens when the VDJ segment is trasncribed with Cδ exon instead of Cµ [35]. B lymphocyte differentiation in the bone marrow is heterogeneous and goes through several maturation stages that can be observed by analysis of immunophenotypic characteristics (Figure 2).

Figure 1.

Schematic B cell regular development.

Figure 2.

Bone marrow expression profile of B cells in different stages of maturation. The B cells maturation states are represented by the colors displayed in the flow cytometry plots figures: light blue represents immature B cells, purple represents intermediate stage and brown the mature B cells. Analysis performed with the Infinicyt software (Cytognos).

1.1.2. Mature B lymphocytes

Mature B cells are usually divided into three subgroups known as follicular B cells, marginal zone (MZ) B‐cell and B1 cells. The follicular B lymphocytes are the majority of mature B cells and are located in the lymphoid follicle of the lymph node and spleen. These cells will stimulate the T lymphocyte response and this can occur in two different locations, extra‐follicular and in the germinal center [36]. B cells in the extra‐follicular can be activated by the T‐helper lymphocytes and differentiate into short‐lived plasmablasts which secrete antibodies. On the other hand, some activated B cells migrate back to the follicles and under follicular helper T cell (TFH) influence proliferate vigorously to form the germinal centers (GC) [37, 38]. Inside a GC, in the dark zone region, the B cells, now called centroblasts, goes through a rapid cellular division, somatic hypermutation, and isotypes class switching. Centroblasts express CD10 and Bcl-6. Next, these cells migrate to the light zone region and become centrocytes. These centrocytes in the light zone will be in contact with follicular dendritic cells (FDC) and TFH through the interactions between CD23 and CD40L (Figure 3). B cells with high affinity for the antigens in this microenvironment will differentiate into plasmablasts (plasma cells) or memory B cells and will express CD27. Plasma cells return to the bone marrow and display well‐defined characteristics expressing CD19, CD27, CD38, CD45, CD138 and intracytoplasmic Ig [10, 19, 3739].

Figure 3.

Schematic representation of B cell maturation in bone marrow and their differentiation in the lymph node [19, 35].

MZ B cells are located in the spleen MZ and are responsible by the T independent‐responses (polysaccharides, glycolipids, and nucleic acid). These cells express pan-B markers, lack CD10 and show weak IgD expression [39]. B-1 cells represent another B cell lineage located mainly in the peritonea and mucosa [40]. These cells are also responsive to T cell-independent antigens and can be recognized by the expression of CD27 and CD43, even though this phenotype remains controversial [40, 41].

1.2. T Lymphocytes

1.2.1. T Lymphocytes ontogeny

Currently, we will discuss the origin and differentiation of T and natural killer (NK) cells, as well the migration of T cell precursors from the bone marrow to the thymus. In addition, we will discuss stages of T cells maturation and immunological development such as differentiation and proliferation, T cell receptor (TCR) genic rearrangement, thymic positive and negative selection and T cells with different phenotypes. These steps summarize the requisites for T cells to become immunologically competent and populate the peripheral lymphoid tissues.

It is well known and scientifically accepted that all blood cells are derived from a hematopoietic stem cell (HSC), defined as pluripotent and capable of self‐renewal. HSCs express the CD34 antigen and account only for 0.1% of the all bone marrow nucleated cells. The first phase of HSC differentiation is the cell commitment to specific lineages. The lymphoid lineage commitment is dependent on several environmental factors such as stromal cells signaling, growth factors, cytokines, and kinases (Janus‐JAK), tyrosine kinases (Kit‐L) and surface molecules such as Notch‐1. Notch‐1 is very important since their collaboration with GATA‐3 will contribute to the T lymphocytes lineage commitment, mainly to the αβ receptor T cells [21]. Radtke et al. have shown that the deletion of Notch‐1 in murine models resulted in the T lymphocytes impaired development in the thymus [42].

The interleukins (IL) are also fundamental for HSC differentiation into lymphoid precursors and the key ILs involved in this process are IL‐1, IL‐2, IL‐3, IL6, and IL‐7. IL‐7 is produced by the bone marrow stromal cells and by the thymic epithelial cells, and IL‐7 plays a role in T cell development, proliferation and survival of lymphoid precursors. The IL‐7 receptor is composed of two chains, IL‐7Rα, and the gamma common (γc), the last is shared by several cytokines receptors such as IL‐2, IL‐4, IL‐9, IL15 and IL‐21. Alterations in the genes that codify IL‐7Rα or the γc result in an immunodeficiency X‐linked named severe combined immunodeficiency (SCID), which is characterized by the significant reduction or absence of T lymphocytes and NK cells, revealing the roles of IL‐7 in humans [4346].

Lymphoid progenitors can be characterized phenotypically by the expression of CD7 and CD34. Some studies define the lymphoid progenitors by the CD7 expression since this antigen has a lower expression in myeloid cells and is not expressed in other cell lineages [47].

A key event on T lymphocyte development is the gene rearrangement that is responsible for the generation of a diverse antigen receptor repertoire. The genes involved in the T cell receptor (TCR) rearrangement are present in the germ cell lineage and are located in the chromosomes 7 and 14. As seen in B lymphocytes, the heavy chain genes are present in the segments that code for the variable (V), diversity (D), joining (J) and constant (C) regions, and this recombination is known as V(D)J. In humans and mice, the γδ T cells have a limited repertoire of V and J segments that are involved in the TCR genic rearrangement [48, 49]. In humans, the δ‐locus is clustered inside the α‐locus, and there are only three true Vδ, they are Vδ1, Vδ2, and Vδ3. The human Vγ repertoire is situated in the γ‐locus with 12 Vγ genes, of which only seven are identified as functional, since Vγ1, Vγ5P, Vγ6, Vγ7, and Vγ10 are considered pseudogenes. The difference between the low diversity in γ-δ loci and the high diversity of the α-β loci suggest that the γδ TCRs low diversity accompanies their recognition of preserved self-proteins with low variability.

The TCR V(D)J recombination is very similar to the B lymphocytes BCR recombination, involving also the enzymes Rag‐1 and Rag‐2, endonuclease Artemis and terminal deoxynucleotidyl transferase (TdT), Ku70, Ku80, DNA‐dependent protein kinase (DNA‐PK) and XRCC4DNA ligase IV. Functional defects on these enzymes can result in immune deficiency such as SCID that can be derived of Rag‐1 or Rag‐2 mutations [5052]. The higher TCR diversity generated during V(D)J recombination is a result of two combined mechanisms of diversity that involve random combination of gene segments and junctional diversity that results from the nucleotides addition or removal, complementary or not, within the junctions between V(D)J segments. These mechanisms of diversity can generate around of 107 different T lymphocyte clones. Each clone represents a unique TCR, the number of T cells clones is not higher as expected, but can be explained by the large number of T cells depleted during the thymic selection [51, 53].

1.2.2. Thymocytes and thymic selection

Different from B lymphocytes, the precursors of T lymphocytes migrate from the bone marrow to the thymus to complete their maturation status and undergo the positive and negative selections. The thymus colonization by immature lymphocytes, also known as thymocytes, enrolls chemokines, mainly CC‐chemokine ligand 21 (CCL21) and CCL25, and their respective receptors CCR7 and CCR9 [54]. The thymic maturation is very important and only 1–3% of thymocytes that enter in the thymus survive the selection steps and gain the circulation [5557].

Initially, the T cell precursor migrates to the corticomedullary region of the thymus, then inside the cortex and the double negative cells (DN) CD4‐CD8‐ (Figures 4 and 5) [58]. The DN cells in development receive Notch‐1 and IL‐7 mediates signals, usually derived from the cortical thymic epithelial cells (cTECs) [59, 60]. Most thymocytes rearrange the V(D)J genes efficiently to express TCR αβ from DN3, at this stage, the TCR was not tested for their specificity and is termed pre‐TCRαβ, which is associated with the protein complex CD3/ζ for the signal transduction [61]. Following maturation, thymocytes begin to express CD4 and CD8 co‐receptors, initially expressing CD8, and then expressing CD4 to form the thymocyte double positives (DP) CD4+CD8+ (Figures 4 and 5). This DP population is present in the thymic cortex, expresses TCRαβ and consists of most lymphocytes inside the thymus of young individuals [62].

Figure 4.

Thymus expression profile of T cells in different stages of maturation. The maturation states are represented by the colors displayed in the flow cytometry plots figures: dark blue represents immature double negative T cells, orange represents double positive cells in intermediate stage and purple represents the single positive mature T cells. Analysis performed with Infinicyt software (Cytognos).

Figure 5.

Schematic representation of T lymphocytes thymic selection [54, 5963].

In the cortex, the TCRs from DP thymocytes interact with peptides via major histocompatibility complex (MHC) molecules expressed by the cTECs and dendritic cells and go through steps of positive and negative selections, thymocytes that interacting properly (low avidity interaction) are positively selected and receive survival signals (Figure 5). Thymocytes that fail in this interaction or interact with high avidity are selected negatively by the mechanism of apoptosis (death by neglect). This process is important so that thymocytes that continue their development are able to recognize foreign antigens but not self‐antigens, thus avoiding autoimmunity. Interestingly, approximately 90% of DP thymocytes express ineffective TCRs and do not pass through cortex checkpoints and end dying due to the absence of positive selection (Figure 5) [54, 61, 63]. The positively selected DP thymocytes migrate to the thymus medulla, guided primarily by the expression of CCR7 and the chemoattraction of CCL19 and CCL21 produced by the medullar thymic epithelial cells (mTECs) and are induced initiate differentiation for CD4+CD8‐ or CD8+CD4‐positive single (SP) thymocytes (Figure 4) per the MHC molecule involved (MHC class II for SP CD4+and MHC class I for SP CD8+) (Figure 5) [64].

The thymocytes that have escaped negative selection by cTECs and are self-reactive to tissue-specific antigens expressed by mTECs are also depleted [65, 66]. TThe expression of tissue-specific antigens by mTECs is controlled by the transcriptional factor autoimmune regulator (AIRE) and AIRE deficiency results in the disease, in humans, called autoimmune polyendocrinopathy – candidiasis - ectodermal dystrophy (APECED) [67, 68]. Interestingly, some thymocytes CD4+ that recognize self‐antigens with high avidity into the thymus may develop in a CD4+ population of peripheral regulatory lymphocytes, which have the function of controlling and preventing autoimmune reactions.

Phenotypically, SP thymocytes express CD62 L and CD69 and also acquire the functional capacity of mature T lymphocytes, but are still naïve, not having yet experienced antigens during an adaptive immune response [69, 70]. Expression of sphingosine‐1‐phosphate receptor 1 (S1P1), one of the S1P receptors, is required for the outflow of the mature T lymphocytes from the thymus and in addition to S1P1, CCR7, CCL19 and CXCL12 also participate in this process [71]. This stage of differentiation and maturation in the thymus lasts approximately 12 days and is critical for the establishment of central tolerance [56]. Finally, immunologically competent lymphocytes leave the thymus and become part of the pool of mature and naive peripheral T lymphocytes, which remain in the cell cycle interphase for extended periods until they encounter with specific antigens presented by antigen-presenting cells (APCs) via MHC in secondary lymphoid organs (Figure 5).

1.2.3. Mature T lymphocytes

Once lymphocytes have left the thymus, they are carried in the blood to the peripheral lymphoid tissues such as lymph nodes, where the cells organize themselves to facilitate the encounter with antigenic particles and consequent lymphocytic activation. Immune cell‐cell interactions are usually rolling interfaces that undergo continuous architectural change. The contact between T and B lymphocytes and the antigen‐presenting cells (APCs) favors the information exchange among the cells, contributing to the assembly, type, and scope of immune responses [72].

Now, we will describe different classes of lymphocytes based on transcription factors expression and their cytokine secretion profile, associated to their differentiation status, functional profile and plasticity. It is important to highlight that each lymphocyte has a unique antigen receptor that is generated by somatic recombination and recognizes a different foreign component, and thus collectively, lymphocytes provide an almost limitless defense against a wide range of antigens. Recent evidences have demonstrated that a lymphocyte can produce daughter cells with different fates that carry out different functions. Based on these indications, it seems as if a single lymphocyte typically produces a diversity of functional daughter cells and manages to renew itself [7376]. Successful immune responses often require more than one type of differentiated cell fate. Kelso and collaborators had already demonstrated in 1995 that biased T helper (Th) Th2 responses contained traces of Th1‐type cytokines expression [77].

Activated CD4+ T cells differentiate into immune suppressive regulatory T (Treg) cells or inflammatory T effector cells, such as T helper Th1, Th2, Th9, Th17, Th22 and follicular helper T (Tfh), each one with distinct characteristic metabolic programs (Figure 5). These different CD4+ subsets play a critical role in the immune and effector response functions of T cells [78]. In response to distinct antigen challenge and extracellular cytokines signals, each CD4+ T cell subset has distinct abilities in producing cytokine and chemokine receptors and expressing polarizing transcription factors, along with their association with specific forms of immune defense. New tools and techniques have revealed the capacity of polarized cells change their phenotype and repolarize towards mixed or alternative fates. The same cytokines that drive the polarization of each T helper cell subset during initial priming also drive the plasticity of established T helper cell subsets [79, 80].

1.2.3.1. CD4 Cell subsets

Since the establishment of the Th1‐Th2 dogma in the 1980s [81, 82], different lineages of effector T cells have been identified that not only promote but also suppress immune responses.

Th1 cells are defined based on the production of pro‐inflammatory cytokines such as interferon (IFN)‐γ, and tumor necrosis factor (TNF)‐α or TNF‐β to stimulate innate and T cell immune responses. These cells are induced by natural killer (NK) and/or dendritic cells, through IFN‐γ producing, which activate signal transducer and activator of transcription (STAT) STAT1, resulting in activation of lineage‐specific transcription factor encoded by T‐box transcription factor—TBX21 (T‐bet) [83]. IL‐27, a cytokine from IL‐12 family, contributes to STAT1 phosphorylation and T‐bet activation. T‐bet enhances the synthesis of the IL‐12 receptor, which activates STAT4 and consequent transcription and production of IFN‐γ. Th1‐type cytokines are responsible for the death of intracellular antigens and for the autoimmune response maintenance (Figure 6) [84, 85].

Figure 6.

CD4 subsets differentiation, cytokines and transcription factors [78, 8490, 121].

On the one hand, Th2 cells are defined as producers of IL‐4, IL‐5, and IL‐13, which are associated with the production of IgE and eosinophilic responses in atopy. Differentiation of the Th2 subset requires IL‐4 produced by Notch ligand activation of dendritic cells, inducing STAT6, which activates GATA‐3. This transcription factor activates the production of the Th2 the lineage‐specifying cytokines. Th2 cells control immunity to extracellular parasites and all forms of allergic inflammatory responses (Figure 6) [85, 86].

In recent years, it became evident that more functional subsets of T helper cells can be induced by various stimuli in vivo and in vitro. Induction of the Th17 lineage occurs when IL‐6, IL‐23, and TGF‐β are present in the inflammatory milieu without Th1 or Th2 cytokines (Figure 6) [87]. Toll‐like receptor signaling, leading to MyD88 signaling, is another innate immune signal fostering Th17 differentiation [88]. The cytokine IL‐6 promotes STAT3, which induces retinoic orphan receptor (ROR) transcription factors, RORα and RORγT, leading to production of Th17 cytokines IL‐17, IL‐17F, and IL‐22 [89, 90]. Functionally, Th17 cells play a role in host defense against extracellular pathogens by mediating the recruitment of neutrophils and macrophages to infected tissues. Moreover, it has become evident that abnormal regulation of Th17 cells may play a significant role in the pathogenesis of a variety of autoimmune and inflammatory diseases, because these cells can cause tissue injury when aber¬rantly regulated [85].

Another recently reported T helper population includes Th9 cells. This subset of cell undergoes a maturation program similar to Th2 cells, with IL‐4 inducing STAT6 activation, and produces the Th2 cytokines IL‐9 and IL‐10, but unlike Th2 cells, they require TGF‐β for maturation (Figure 6) [91, 92]. The IL‐2‐STAT5 signaling including interferon regulatory factor (IRF)4 expression is critical for Th9 cell differentiation. Additionally, the lineage‐specific transcription factor for Th9 development may be the activator protein 1 family transcription factor, BATF, leading to a transcriptional program, which results in increased IL‐9 and IL‐10 production [85, 91, 93]. There is a requirement as well for TGF‐β‐induced SMAD proteins and the SMAD‐independent induction the transcription factor PU.1 in the generation of IL‐9‐secreting T cells. Although the GATA3 expression is lower in Th9 cells than Th2 cells, STAT6 is an important target gene involved in Th9 differentiation. These cells can exacerbate the immune response by enhancing antibody production and increasing immune cell infiltration and activity within the respiratory tract, contributing to asthmatic disease. In addition, IL‐9 mediates anti‐parasitic activity by altering epithelial cell function, increasing immune cell infiltration into infected locations, and augmenting leukocyte immune function. Besides that, the production of IL‐9 by Th9 cells impairs tissue repair process during colitis, in contrast, can limit tumor growth by stimulating lymphocyte anti‐tumor activity (Figure 6).

Th22 cells are promoted by IL‐6 and TNF‐α, which induces STAT3, and expression of the aryl hydrocarbon receptor [94]. Th22 cells have a specific profile of Th1 and Th17‐associated genes, such as IFN‐γ, IL17a, T‐bet and RORγt [95]. In addition the counterpart maturation between Th17 and Th22 subsets, numerous phenotypic markers are expressed in both cell populations, including CCR6, CCR4, dipeptidyl peptidase IV, CD26, and CD90. But, differently from Th17, the Th22 cells expresses CCR10 and represents a distinct subtype of T cells that is involved in the epidermis immunity. IL‐22, a cytokine from the IL‐10 family, is not exclusively produced by the Th22 cells, then also by Th1 and Th17 cells (Figure 6) [85, 96].

A subset of human CD4+ T cells that specifically express IL‐22 has been identified in the skin where the synthesis of active vitamin D enhances IL‐22 expression, contributing to skin homeostasis, but also to the pathogenesis of skin disease, observed in psoriasis patients [97, 98].

Follicular helper CD4+T (Tfh) cells were first found in human tonsils, but now it is clear that are localized in the B‐cell follicle and germinal center (GC) and is specialized in facilitating B‐cell responses, enhancing immunoglobulin production [98, 99]. Tfh cells require a strong TCR signal for induction, which is also required for Treg responses [85, 100]. Tfh specification requires activation of the inducible co‐stimulator (ICOS), a CD28‐related co‐stimulatory signal provided by activated dendritic cells or B cells, which initiates transcription of MAF, one transcription factor that induces IL‐21 activation. The OX‐40/CD134 co‐stimulatory signal ligation is necessary to down‐regulates CTLA‐4, a dominant suppressor molecule of T cell activation (Figure 6) [101]. IL‐6 and STAT3 are required for Tfh development like Th17 cells, yet Tfh cells can be generated in the absence of Th17 cytokines, IL‐17, IL‐17F, or TGF‐β [102].

In order to understand how Tfh cells are identified, first is necessary to comprehend T and B cells migration for their interaction sites, which usually takes place in secondary lymphoid organs such as lymph nodes (LNs). Naive T cells migrate to the T cell zone in LNs responding to CCL19 and CCL21 gradients. After dendritic cells antigen stimulation, Tfh cells up‐regulate CXCR5, down‐regulate CCR7 and migrate to the interfollicular regions within LNs, where they interact with activated B cells. These interactions result in antibody production by the short‐lived plasmablasts, which take place in extrafollicular regions or in the germinal center. In both regions, Tfh cells support B‐cell maturation, class switch and affinity selection, via cytokines secretion or by expressing surface molecules. The germinal center responses drive memory B cell and plasma cell development. Then, Tfh cells are characterized according to patterns of receptor expression that enables their movements, as well as the expression of other surface proteins associated with migratory processes [103107].

Regulatory T‐cells (Treg) represent a heterogeneous population of CD4+T‐cells characterized by suppressive capacity, which can be generated in the thymus, termed natural Tregs (nTregs) or adaptive regulatory T cells, induced in the periphery, involved in maintaining oral tolerance (Th3 cells), and T regulatory type 1 cells (Tr1 cells), stimulated by IFN‐α secreted by neighboring plasmacytoid dendritic cells (pDCs). According to the literature, the nTregs need a strong TCR signal for their development. They are formed by low co‐stimulation, so the T cell antigen recognition without a robust second signal provide by the CD28 family members leads to tolerance [85, 108]. Differentiation of induced Tregs, Th3 cells, and Tr1 cells happens in the periphery and needs high TGF‐β concentrations and absence of pro‐inflammatory cytokines [109]. Cell‐cell interaction and IL‐10 secretion are essential for the Treg suppressor function, mediated by the transcription factor Foxp3 through STAT5 activation (Figure 6), and simultaneous RORγt down regulation, which is the Th17 transcription factor [85, 110, 111].

Human T cells can be divided into functionally distinct subsets. Two primary categories are naïve T cells (TN) that have not been exposed to antigen and those that are antigen‐experienced (memory). Naive T cells are usually characterized by the expression of CD45RA+, CD62L+ and CCR7+. CD45RA and CD45RO are high and low molecular weight protein derived from the CD45 gene splice variant, distinctly, with CD45RO being mainly expressed by memory cells. CD45RO+ cells are rarely found in neonates and gradually increased with age. The analysis of homing receptors revealed that T cells are heterogeneous and in particular naïve T cells express high levels of the lymph‐node homing receptor CD62 L (L‐selectin). Long‐lived memory CD4+ lymphocytes are a hallmark feature of the adaptive immune system in response to pathogens and tumors [112]. The memory T cell compartment is heterogeneous and has been conventionally divided into two subsets on the basis of the lymph node homing molecules CD62 L and CCR7 expression [113]. Central memory T cells (TCM cells) highly express CD45RO+, CD45RA−, CD62L+ and CCR7+, whereas CD45RO+, CD45RA−, CD62L− CCR7− effector memory T cells (TEM cells) are considered to be committed progenitor cells that undergo terminal differentiation after a limited number of divisions (Figure 7) [114]. CCR7 and CD62 L are mostly co‐expressed on the surface of CD4+ and CD8+T cells, and cells expressing these markers nearly uniformly express CD27 and CD28—but the inverse is not true. CD27 and CD28 are the main co‐stimulatory molecules required to induce T cell activation, although memory T cells seem to be less dependent on CD27 and CD28 for their reactivation than naïve T cells [115, 116]. CCR7−/CD62L−, CD28+ cells are found in the peripheral blood of healthy individuals and known as a subset of transitional memory (TM) cells. TM cells seem to be more mature than TCM cells, but not as totally mature as TEM cells [117]. IL‐15 administration increases a cell subset that re‐expresses CD45RA (named terminal effector cells–TEMRA). The TEMRA cells express senescence markers, such as KLRG‐1, CD57, and H2AX phosphorylation, have low functional and proliferative ability, indicating their terminal differentiation [118].

Figure 7.

CD4 and CD8 subsets of memory [113, 114, 155].

1.2.3.2. CD8+ cytotoxic T lymphocytes (CTLs) subsets

Similar to T CD4+ cells, naïve CD8+ T cells differentiate into effector T cells (CD62L‐ CD127‐) upon TCR engagement with antigen and costimulation by an APC, but the antigen recognition occurs by MHC class I in peripheral lymphatic organs. Additionally, CD8+ T cells also acquire different profiles according to co‐stimulatory molecules and cytokines presents in the environmental, contributing to transcription factors induction and specific differentiation into Tc1, Tc2, Tc9, Tc17 or CD8+ T regulatory fate, as we observe in CD4+ T cells [119].

Cytotoxic T lymphocytes (CTLs) also named Tc1 are the best‐characterized subset of CD8+ T cells that are responsible for the direct killing of infected, damaged, and dysfunctional cells, including tumor cells. Once differentiated, these cells are IL‐2 and IL‐12 dependent and highly cytotoxic, rapidly expressing high levels of IFN‐γ, TNF‐α, perforin, and granzymes, into immunological synapse, following activation [120]. IL‐12 promotes expression of T‐bet and Id2, and IL‐2 down‐regulates BCL‐6 [121]. The initial activation of CD8+ T cells is related with the up‐regulation of CD44 and CD69, killer cell lectin‐like receptor G1 (KLRG1) and IL‐2 receptor subunit‐α (CD25), though the L‐selectin (CD62 L), the IL‐7 receptor subunit‐α (CD127) and CD27 are diminished in comparison with naïve cells [119, 121].

While most CD8+ T cells die by apoptosis after antigen clearance, there are rare cells that survive as long-lived memory T cells. Memory CD8+ T cells were subdivided into two broad subsets [113, 122], central memory (CD62L+ CD127+ CCR7+) and effector memory (CD162L‐ CD127+CCR7‐), distinguished by the relative expression of two homing molecules, CD62 L and CCR7. T effector memory cells have a phenotype more similar to that of effector cells, characterized by a loss of CCR7 expression and intermediate to no CD62 L expression (Figure 7). These cells exhibit rapid effector function, readily differentiating into T effector cells that secrete high amounts of IFN‐γ and are highly cytotoxic upon re‐exposure to cognate antigen. In contrast, T central memory cells are less differentiated, have increased proliferative potential and greater self‐renewal capability, can produce high amounts of IL‐2, and acquire effector functions less rapidly [120, 123, 124].

Tc2 cells, similarly to Th2 cells, produce IL‐5, IL‐13, but the limited extent of IL‐4, besides granzymes and perforin, and express the lineage‐specific transcription factor GATA3. This profile is associated with propagation of Th2‐mediated allergy and probably contributes to rheumatoid arthritis [119, 125].

The differentiation of CD8+ T cells into IL‐9 producers (Tc9 cells), in agreement with low expression of granzyme B, occurs mostly in the intestinal epithelium by IL‐4 and TGF‐β induction, favoring their greater anti‐tumor activity. The transcription factors STAT6 and IRF4 are important for IL‐9 production while Foxp3 for inhibition [119, 126].

The IL‐17‐producing CD8+ T (Tc17) cells are differentiated by IL‐6 or IL‐21 along with TGF‐β while IL‐23 stabilizes their phenotype. Similar to Th17 cells, they produce IL-17 and IL-21, express the receptor for IL‐23 and the lineage‐specific transcription factors IRF‐4, RORγt and RORα [127]. Tc17 presents impaired cytotoxic activity because of a low IFN-𝛾, perforin, and granzyme B production. In contrast, they are able to enhance anti-tumor immunity due to their pro-inflammatory properties, which, on the other hand, may contribute to autoimmune processes [119, 128].

The suppressor CD8+ Treg cells restricted by the non‐classical MHC class Ib molecules Qa‐1 (mouse) or HLA‐E (human) represent a well‐defined subpopulation. These cells present CD44hi CD122+ Ly49+ Foxp3+ phenotype and IL‐15 is important to their activity [129]. For suppression, these CD8+ Treg cells rely on diverse mechanisms including TGF‐β, IL‐10, granzymes, perforin and indoleamine 2, 3‐dioxygenase (IDO) [129].

1.3. Natural killer cells (NK)

Natural killer (NK) cells are an important piece of the innate immunity and provide a first‐line defense against tumors and viral infections. NK cells were identified in the 1970s by the ability of kill tumor cells without previous activation [130]. The precursor cell able to differentiate towards NK cells was originally identified in the bone marrow, the main organ of hematopoiesis in adult life. However, subsequent experiments revealed that hematopoietic progenitor cell and/or NK cell committed precursors (NKPs) can traffic from bone marrow to peripheral sites, and it is now clear that the NK cell development occurs not only in the bone marrow but also in peripheral lymphoid and non‐lymphoid organs [131]. In addition, Yu et al. demonstrated that precursor cells isolated from different sites of tissues can differentiate in vitro into mature NK cells [132]. The HSC commitment to NK cells differentiation includes transcription factors such as ID2, PU.1, Ets‐1, TOX and NFIL3, and their maturation involves Eomes and Tbet [133136].

Later studies demonstrated the cytotoxic ability of these cells against virus‐infected cells and their participation into early inflammatory response secreting cytokines and chemokines [51, 137]. Particularly, upon activation, NK cells may also sense various bacterial products via toll‐like receptors, an event resulting in a significant and rapid increase in their cytolytic activity and cytokine production [138]. Human NK cell function is regulated by several inhibitory and activating receptors. Among the various inhibitory NK cell receptors, an important role is played by Killer Ig‐like Receptors (KIRs) that recognize allotypic determinants of HLA‐A, ‐B, ‐C molecules and by the heterodimer NKG2A specific for the non‐classical HLA‐E molecule [139]. Regarding the activating NK cell receptors, the examples are NKp46, NKp44, and NKp30 that have been together named Natural Cytotoxicity Receptors (NCRs). The NCRs ligands are only partially known and possibly include pathogen‐derived molecules and cellular ligands. In addition to NCR, other activating receptors and co‐receptors are involved in NK cell function. These include NKG2D (recognizing MICA/B and ULBPs molecules), DNAM‐1 (specific for CD155 and CD112), CD16 (FcRIII), NKp80 (specific for AICL), CD244 (that binds CD48), and NTBA (mediating hemophilic interaction) (Figure 8) [140].

Figure 8.

Developmental relationship between NK cells and other ILCs [137, 144, 156].

Peripheral blood NK cells are not a uniform population. Consequently, two main subsets are identified regarding levels of CD56 expression, CD56dim and CD56bright NK cells [141]. CD56dimNK cells are mainly in peripheral blood and show potent cytolytic activity and fast release of IFN‐γ and other cytokines or chemokines upon cell activation through NK receptors. A large fraction of CD56dim co‐expresses CD16. CD56bright NK cells represent a smaller population in peripheral blood while they are predominant in tissues and secondary lymphoid organs and are supposed to be responsible for the long‐lasting production of chemokines and cytokines [137]. Several soluble factors participate on the NK cells development, and they are listed in Table 1.

CytokineRoleReference
IL‐15Central role in the development, maturation, survival, proliferation and differentiation[142]
IL‐7, SCF and Flt3‐LNK cells differentiation[142]
IL‐21killer immunoglobulin‐like receptors (KIR) expression on final stage of NK cell differentiation[143]
IL‐8 and MIP‐1αFavor the precursor to differentiate into NK cell, inhibiting the development of myeloid precursors[144]

Table 1.

Soluble factors involved in the NK cell development.

1.4. Innate lymphoid cells (ILCs)

While NK cells have been known for almost four decades and have been extensively studied, other innate lymphoid cells (ILCs) have been better characterized in recent years. ILCs play a significant role in innate defenses against pathogens in different sites and in lymphoid tissue organization, primarily during fetal life. ILCs are emerging as a family of effectors and regulators of innate immunity and tissue remodeling and express neither somatically recombined antigen receptors nor phenotypical markers of myeloid cells [145]. These subsets of cells require the transcriptional repressor Id2 and the interleukin 7 (IL‐7) for their development, and they generate cytokine secretion patterns that mirror those of helper T cells of the adaptive immune system [146, 147]. ILCs have been classified in three main groups ILC1, ILC2, and ILC3 according to their cytokine profile and to the transcription factors required for their differentiation.

Similar to pro‐inflammatory T helper type 1 cells, ILCs of group 1 (ILC1) release IFN‐γ and require the transcription factor T‐bet for their development, as do NK cells of the innate immune system. ILC1 cells were distinct from natural killer (NK) cells as they lacked perforin, granzyme B and the NK cell markers CD56, CD16 and CD94 (Figure 8) [148].

ILCs of group 2, which include natural helper cells and nuocytes, secrete IL‐5 and IL‐13 and require the transcription factor GATA‐3 and thus resemble pro‐inflammatory T helper type 2 cells. ILC2s mediate parasite expulsion but also contribute to airway inflammation, emphasizing the functional similarity between these cells and Th2 cells (Figure 8) [149151]. Finally, ILCs of group 3 (ILC3 cells) require the transcription factors RORγt and AhR and include not only mucosal ‘NK‐22’ cells, which secrete IL‐22 and thus mimic non‐inflammatory cells of the Th22 subset of helper T cells. ILC22 cells include NKp46 (+) and lymphoid tissue inducer (LTi)‐like subsets that express the aryl hydrocarbon receptor (AHR). These cells were heterogeneous in their requirement for Notch and their effect on the generation of fetal and mucosal intestinal lymphoid tissues (LTi cells) (Figure 8) [152, 153], which produce IL‐22 and IL‐17 and thus resemble pro‐inflammatory cells of the Th17 subset of helper T cells. Interleukin 22 (IL‐22)‐ and IL‐17‐producing ILCs, which depend on the transcription factor RORγt, express CD127 (IL‐7 receptor α‐chain) and the natural killer cell marker CD161 [147, 154].

Advertisement

2. Concluding remarks

Herein, we have discussed B lymphocyte differentiation in the bone marrow and their stages of maturation in secondary lymphoid tissues and profiles in interfollicular, perifollicular, and follicular areas. In addition, we also have discussed derivation of T-cell precursors, natural killer cells, and other innate lymphoid cells. Moreover, T-cell precursor migration to thymus, differentiation, rearrangement, thymic selection, transcription factors, their profile and subsets in secondary lymphoid organs are also included. These topics are very important for the comprehension of the complex processes involved in forming a functional, consistently efficient immune response.

Advertisement

Acknowledgments

This work was funded by FAPESP, process number - 2014/15504-7 and by National Council of Technological and Scientific Development (CNPq) process number - 441665/2014-4. We are grateful to the contribution of Dr. Gilmar Santos, also to Dra. Anna Carla Goldberg for proofreading and to Sociedade Beneficente Israelita Brasileira Hospital Albert Einstein and the Clinical Pathology Laboratory for all their support.

References

  1. 1. Kondo M, Scherer DC, Miyamoto T, King AG, Akashi K, Sugamura K, Weissman IL. Cell‐fate conversion of lymphoid‐committed progenitors by instructive actions of cytokines. Nature. 2000;407(6802):383-386. DOI: 10.1038/35030112
  2. 2. Delogu A, Schebesta A, Sun Q, Aschenbrenner K, Perlot T, Busslinger M. Gene repression by Pax5 in B cells is essential for blood cell homeostasis and is reversed in plasma cells. Immunity. 2006;24(3):269-281. DOI: 10.1016/j.immuni.2006.01.012
  3. 3. Smith LG, Weissman IL, Heimfeld S. Clonal analysis of hematopoietic stem‐cell differentiation in vivo. Proceedings of the National Academy of Sciences of the United States of America. 1991;88(7):2788-92.
  4. 4. Osawa M, Hanada K, Hamada H, Nakauchi H. Long‐term lymphohematopoietic reconstitution by a single CD34‐low/negative hematopoietic stem cell. Science. 1996;273(5272):242-245. DOI: 10.1126/science.273.5272.242
  5. 5. Lai AY, Kondo MT, B. Lymphocyte differentiation from hematopoietic stem cell. Semin Immunol. 2008;20(4):207-212. DOI: 10.1016/j.smim.2008.05.002
  6. 6. Traver D, Miyamoto T, Christensen J, Iwasaki‐Arai J, Akashi K, Weissman IL. Fetal liver myelopoiesis occurs through distinct, prospectively isolatable progenitor subsets. Blood. 2001;98(3):627-635. DOI: doi.org/10.1182/blood. V98.3.627
  7. 7. Mebius RE, Miyamoto T, Christensen J, Domen J, Cupedo T, Weissman IL, Akashi K. The fetal liver counterpart of adult common lymphoid progenitors gives rise to all lymphoid lineages, CD45+CD4+CD3‐ cells, as well as macrophages. Journal of Immunology. 2001;166(11):6593-6601. DOI: https://doi.org/10.4049/jimmunol.166.11.6593
  8. 8. Kawamoto H, Ohmura K, Katsura Y. Presence of progenitors restricted to T, B, or myeloid lineage, but absence of multipotent stem cells, in the murine fetal thymus. Journal of Immunology. 1998; 161(8):3799-3780
  9. 9. Petrie HT, Kincade PW. Many roads, one destination for T cell progenitors. The Journal of Experimental Medicine. 2005;202(1):11-13. DOI: 10.1084/jem.20050990
  10. 10. Melchers F. Checkpoints that control B cell development. The Journal of Clinical Investigation. 2015;125(6):2203-2210. DOI: 10.1172/JCI78083
  11. 11. Dorshkind K. Regulation of hemopoiesis by bone marrow stromal cells and their products. Annual Review of Immunology. 1990;8:111-137. DOI: 10.1146/annurev.iy.08.040190.000551
  12. 12. Hentges F. B lymphocyte ontogeny and immunoglobulin production. Clinical and Experimental Immunology. 1994;97 Suppl 1:3-9. DOI: 10.1111/j.1365‐2249. 1994.tb06252.x
  13. 13. Ramirez J, Lukin K, Hagman J. From hematopoietic progenitors to B cells: mechanisms of lineage restriction and commitment. Current Opinion in Immunology. 2010;22(2):177-184. DOI: 10.1016/j.coi.2010.02.003
  14. 14. Nutt SL, Heavey B, Rolink AG, Busslinger M. Commitment to the B‐lymphoid lineage depends on the transcription factor Pax5. Nature. 1999;401(6753):556-562. DOI: 10.1038/44076
  15. 15. Busslinger M. Transcriptional control of early B cell development. Annual review of immunology. 2004; 22:55-79. DOI: 10.1146/annurev.immunol.22.012703.104807
  16. 16. Ikawa T, Kawamoto H, Wright LY, Murre C. Long‐term cultured E2A‐deficient hematopoietic progenitor cells are pluripotent. Immunity. 2004;20(3):349-360. DOI: http://dx.doi.org/10.1016/S1074‐7613(04)00049‐4
  17. 17. Yoshida T, Ng SY, Zuniga‐Pflucker JC, Georgopoulos K. Early hematopoietic lineage restrictions directed by Ikaros. Nature Immunology. 2006;7(4):382-391. DOI: 10.1038/ni1314
  18. 18. Hardy RR, Kincade PW, Dorshkind K. The protean nature of cells in the B lymphocyte lineage. Immunity. 2007;26(6):703-714. DOI: 10.1016/j.immuni.2007.05.013
  19. 19. Perez‐Andres M, Paiva B, Nieto WG, Caraux A, Schmitz A, Almeida J, et al. Human peripheral blood B‐cell compartments: a crossroad in B‐cell traffic. Cytometry Part B, Clinical Cytometry. 2010;78 Suppl 1:S47‐S60. DOI: 10.1002/cyto.b.20547
  20. 20. Nagasawa T. Microenvironmental niches in the bone marrow required for B‐cell development. Nature Reviews Immunology. 2006;6(2):107-116. DOI: 10.1038/nri1780
  21. 21. Blom B, Spits H. Development of human lymphoid cells. Annual Review of Immunology. 2006;24:287-320. DOI: 10.1146/annurev.immunol.24.021605.090612
  22. 22. Roessler S, Gyory I, Imhof S, Spivakov M, Williams RR, Busslinger M, Fisher AG, Grosschedl R. Distinct promoters mediate the regulation of Ebf1 gene expression by interleukin‐7 and Pax5. Molecular and Cellular Biology. 2007;27(2):579-594. DOI: 10.1128/MCB.01192‐06
  23. 23. Stewart AK, Schwartz RS. Immunoglobulin V regions and the B cell. Blood. 1994;83(7):1717-1730
  24. 24. Schatz DG, Ji Y. Recombination centres and the orchestration of V(D)J recombination. Nature Reviews Immunology. 2011;11(4):251-263. DOI: 10.1038/nri2941
  25. 25. Motea EA, Berdis AJ. Terminal deoxynucleotidyl transferase: the story of a misguided DNA polymerase. Biochimica et Biophysica acta. 2010;1804(5):1151-1166. DOI: 10.1016/j.bbapap.2009.06.030
  26. 26. van Lochem EG, van der Velden VH, Wind HK, Marvelde JG, Westerdaal NA, van Dongen JJ. Immunophenotypic differentiation patterns of normal hematopoiesis in human bone marrow: reference patterns for age‐related changes and disease‐induced shifts. Cytometry Part B, Clinical Cytometry. 2004;60(1):1-13. DOI: 10.1002/cyto.b.20008
  27. 27. Wang LD, Clark MR. B‐cell antigen‐receptor signalling in lymphocyte development. Immunology. 2003;110(4):411-420. DOI: 10.1111/j.1365‐2567.2003. 01756.x
  28. 28. Kitamura D, Rajewsky K. Targeted disruption of mu chain membrane exon causes loss of heavy‐chain allelic exclusion. Nature. 1992;356(6365):154-156. DOI: 10.1038/356154a0
  29. 29. Miosge LA, Goodnow CC. Genes, pathways and checkpoints in lymphocyte development and homeostasis. Immunology and Cell Biology. 2005;83(4):318-335. DOI: 10.1111/j.1440‐1711.2005. 01353.x
  30. 30. Brauninger A, Goossens T, Rajewsky K, Kuppers R. Regulation of immunoglobulin light chain gene rearrangements during early B cell development in the human. European Journal of Immunology. 2001;31(12):3631-3637. DOI: 10.1002/1521‐4141(200112)31:12<3631::AID‐IMMU3631>3.0.CO;2‐L
  31. 31. Pieper K, Grimbacher B, Eibel H. B‐cell biology and development. The Journal of Allergy and Clinical Immunology. 2013;131(4):959‐971. DOI: 10.1016/j.jaci.2013.01.046
  32. 32. Sandel PC, Monroe JG. Negative selection of immature B cells by receptor editing or deletion is determined by site of antigen encounter. Immunity. 1999;10(3):289-299. http://dx.doi.org/10.1016/S1074‐7613(00)80029‐1
  33. 33. Burrows PD, Cooper MD. B cell development and differentiation. Current Opinion in Immunology. 1997;9(2):239-244. DOI: 10.1016/S0952‐7915(97)80142‐2
  34. 34. Ghia P, ten Boekel E, Rolink AG, Melchers F. B‐cell development: a comparison between mouse and man. Immunology Today. 1998;19(10):480-485. DOI: http://dx.doi.org/10.1016/S0167‐5699(98)01330‐9
  35. 35. Geisberger R, Lamers M, Achatz G. The riddle of the dual expression of IgM and IgD. Immunology. 2006;118(4):429-437. DOI: 10.1111/j.1365‐2567.2006. 02386.x
  36. 36. Allman D, Pillai S. Peripheral B cell subsets. Current Opinion Immunology. 2008;20(2):149-157. DOI: 10.1016/j.coi.2008.03.014
  37. 37. Nutt SL, Hodgkin PD, Tarlinton DM, Corcoran LM. The generation of antibody‐secreting plasma cells. Nature Reviews Immunology. 2015;15(3):160-171. DOI: 10.1038/nri3795
  38. 38. Pillai S, Cariappa A. The folicular versus marginal zone B lymphocyte cell fate decision. Nature. 2009;9:767-777. DOI: 10.1038/nri2656
  39. 39. Cerutti A, Cols M, Puga I. Marginal zone B cells: virtues of innatelike antibody‐producing lymphocytes. Nature Reviews Immunology. 2013;13(2):118-132. DOI: 10.1038/nri3383
  40. 40. Bemark M. Translating transitions ‐ how to decipher peripheral human B cell development. Journal of Biomedical Research. 2015;29(4):264-284. DOI: 10.7555/JBR.29.20150035
  41. 41. Perez‐Andres M, Grosserichter‐Wagener C, Teodosio C, van Dongen JJ, Orfao A, van Zelm MC. The nature of circulating CD27+CD43+ B cells. The Journal of Experimental Medicine. 2011;208(13):2565-2566. DOI: 10.1084/jem.20112203
  42. 42. Radtke F, Wilson A, Stark G, Bauer M, van Meerwijk J, MacDonald HR, Aguet M. Deficient T cell fate specification in mice with an induced inactivation of Notch1. Immunity. 1999;10(5):547-558. DOI: http://dx.doi.org/10.1016/S1074‐7613(00)80054‐0
  43. 43. Noguchi M, Yi H, Rosenblatt HM, Filipovich AH, Adelstein S, Modi WS, McBride OW, Leonard WJ. Interleukin‐2 receptor gamma chain mutation results in X‐linked severe combined immunodeficiency in humans. Cell. 1993;73(1):147-157. DOI: http://dx.doi.org/10.1016/0092‐8674(93)90167‐O
  44. 44. Russell SM, Keegan AD, Harada N, Nakamura Y, Noguchi M, Leland P. et al. Interleukin‐2 receptor gamma chain: a functional component of the interleukin‐4 receptor. Science. 1993;262(5141):1880-1883. DOI: 10.1126/science.8266078
  45. 45. Puel A, Ziegler SF, Buckley RH, Leonard WJ. Defective IL7R expression in T (‐) B (+) NK(+) severe combined immunodeficiency. Nature Genetics. 1998;20(4):394-397. DOI: 10.1038/3877
  46. 46. Giliani S, Mori L, de Saint Basile G, Le Deist F, Rodriguez‐Perez C, Forino C, Mazzolari E, Dupuis S, Elhasid R, Kessel A, Galambrun C, Gil J, Fischer A, Etzioni A, Notarangelo LD. Interleukin‐7 receptor alpha (IL‐7Ralpha) deficiency: cellular and molecular bases. Analysis of clinical, immunological, and molecular features in 16 novel patients. Immunological Reviews. 2005;203:110-126. DOI: 10.1111/j.0105‐2896.2005. 00234.x
  47. 47. Barcena A, Muench MO, Galy AH, Cupp J, Roncarolo MG, Phillips JH, Spits H. Phenotypic and functional analysis of T‐cell precursors in the human fetal liver and thymus: CD7 expression in the early stages of T‐ and myeloid‐cell development. Blood. 1993;82(11):3401-3414
  48. 48. Schwarz K, Gauss GH, Ludwig L, Pannicke U, Li Z, Lindner D, Friedrich W, Seger RA, Hansen‐Hagge TE, Desiderio S, Lieber MR, Bartram CR. RAG mutations in human B cell‐negative SCID. Science. 1996;274(5284):97-99. DOI: 10.1126/science.274.5284.97
  49. 49. Santagata S, Gomez CA, Sobacchi C, Bozzi F, Abinun M, Pasic S, Cortes P, Vezzoni P, Villa A. N‐terminal RAG1 frameshift mutations in Omenn's syndrome: internal methionine usage leads to partial V(D)J recombination activity and reveals a fundamental role in vivo for the N‐terminal domains. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(26):14572-14577. DOI: 10.1073/pnas.97.26.14572
  50. 50. Villa A, Sobacchi C, Notarangelo LD, Bozzi F, Abinun M, Abrahamsen TG, et al. V(D)J recombination defects in lymphocytes due to RAG mutations: severe immunodeficiency with a spectrum of clinical presentations. Blood. 2001;97(1):81-88. DOI: https://doi.org/10.1182/blood.V97.1.81
  51. 51. Abbas AK, Lichtman AH, Pillai S. Cellular and Molecular Immunology. 7th ed; 2011
  52. 52. Kazen AR, Adams EJ. Evolution of the V, D, and J gene segments used in the primate gammadelta T‐cell receptor reveals a dichotomy of conservation and diversity. Proceedings of the National Academy of Sciences. U S A. 2011;108(29):E332-40. DOI: 10.1073/pnas.1105105108
  53. 53. Schlissel MS. Regulating antigen‐receptor gene assembly. Nature Reviews Immunology. 2003;3(11):890-899. Review. DOI: 10.1038/nri1225
  54. 54. Takahama Y. Journey through the thymus: stromal guides for T‐cell development and selection. Nature reviews Immunology. 2006;6(2):127-135. DOI: 10.1038/nri1781
  55. 55. Scollay RG, Butcher EC, Weissman IL. Thymus cell migration. Quantitative aspects of cellular traffic from the thymus to the periphery in mice. European Journal of Immunology. 1980;10(3):210-218. DOI: 10.1002/eji.1830100310
  56. 56. Egerton M, Scollay R, Shortman K. Kinetics of mature T‐cell development in the thymus. Proceedings of the National Academy of Sciences of the United States of America. 1990;87(7):2579-2582
  57. 57. Goldrath AW, Bevan MJ. Selecting and maintaining a diverse T‐cell repertoire. Nature. 1999;402(6759):255-262. DOI: 10.1038/46218
  58. 58. Godfrey DI, Kennedy J, Suda T, Zlotnik A. A developmental pathway involving four phenotypically and functionally distinct subsets of CD3‐CD4‐CD8‐ triple‐negative adult mouse thymocytes defined by CD44 and CD25 expression. Journal of Immunology. 1993;150(10):4244-4252
  59. 59. von Freeden‐Jeffry U, Vieira P, Lucian LA, McNeil T, Burdach SE, Murray R. Lymphopenia in interleukin (IL)‐7 gene‐deleted mice identifies IL‐7 as a nonredundant cytokine. The Journal of Experimental Medicine. 1995;181(4):1519-1526. DOI: 10.1084/jem.181.4.1519
  60. 60. Peschon JJ, Morrissey PJ, Grabstein KH, Ramsdell FJ, Maraskovsky E, Gliniak BC, Park LS, Ziegler SF, Williams DE, Ware CB, Meyer JD, Davison BL. Early lymphocyte expansion is severely impaired in interleukin 7 receptor‐deficient mice. The Journal of Experimental Medicine. 1994;180(5):1955-1960. DOI: 10.1084/jem.180.5.1955
  61. 61. Germain RN. T‐cell development and the CD4‐CD8 lineage decision. Nature reviews Immunology. 2002;2(5):309-322. DOI: 10.1038/nri798
  62. 62. Allman D, Sambandam A, Kim S, Miller JP, Pagan A, Well D, Meraz A, Bhandoola A. Thymopoiesis independent of common lymphoid progenitors. Nature Immunology. 2003;4(2):168-174. DOI: 10.1038/ni878
  63. 63. Zuniga‐Pflucker JC. T‐cell development made simple. Nature Reviews Immunology. 2004;4(1):67-72. DOI: 10.1038/nri1257
  64. 64. Ueno T, Saito F, Gray DH, Kuse S, Hieshima K, Nakano H, Kakiuchi T, Lipp M, Boyd RL, Takahama Y. CCR7 signals are essential for cortex‐medulla migration of developing thymocytes. The Journal of Experimental Medicine. 2004;200(4):493-505. DOI: 10.1084/jem.20040643
  65. 65. Zuklys S, Balciunaite G, Agarwal A, Fasler‐Kan E, Palmer E, Hollander GA. Normal thymic architecture and negative selection are associated with aire expression, the gene defective in the autoimmune‐polyendocrinopathy‐candidiasis‐ectodermal dystrophy (APECED). Journal of Immunology. 2000;165(4):1976-1983. DOI: https://doi.org/10.4049/jimmunol.165.4.1976
  66. 66. Derbinski J, Pinto S, Rosch S, Hexel K, Kyewski B. Promiscuous gene expression patterns in single medullary thymic epithelial cells argue for a stochastic mechanism. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(2):657-662. DOI: 10.1073/pnas.0707486105
  67. 67. Nagamine K, Peterson P, Scott HS, Kudoh J, Minoshima S, Heino M, Krohn KJ, Lalioti MD, Mullis PE, Antonarakis SE, Kawasaki K, Asakawa S, Ito F, Shimizu N. Positional cloning of the APECED gene. Nature Genetics. 1997;17(4):393-398. DOI: 10.1038/ng1297‐393
  68. 68. Finnish‐German AC. An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD‐type zinc‐finger domains. Nature Genetics. 1997;17(4):399-403. DOI: 10.1038/ng1297‐399
  69. 69. Reichert RA, Weissman IL, Butcher EC. Phenotypic analysis of thymocytes that express homing receptors for peripheral lymph nodes. Journal of Immunology. 1986;136(10):3521-3528
  70. 70. Ramsdell F, Jenkins M, Dinh Q, Fowlkes BJ. The majority of CD4+8‐ thymocytes are functionally immature. Journal of Immunology. 1991;147(6):1779-1785
  71. 71. Matloubian M, Lo CG, Cinamon G, Lesneski MJ, Xu Y, Brinkmann V, Allende ML, Proia RL, Cyster JG. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature. 2004;427(6972):355-360. DOI: 10.1038/nature02284
  72. 72. Basu R and Huse M. Mechanical Communication at the Immunological Synapse. Trends in Cell Biology. 2016. DOI: 10.1016/j.tcb.2016.10.005. [Epub ahead of print]
  73. 73. Tubo NJ, Pagán AJ, Taylor JJ, Nelson RW, Linehan JL, Ertelt JM, Huseby ES, Way SS, Jenkins MK.. Single naive CD4+ T cells from a diverse repertoire produce different effector cell types during infection. Cell. 2013;153:785-796. DOI: 10.1016/j.cell.2013.04.007
  74. 74. Gerlach, C. Rohr JC, Perié L, van Rooij N, van Heijst JW, Velds A, Urbanus J, Naik SH, Jacobs H, Beltman JB, de Boer RJ, Schumacher TN. Heterogeneous differentiation patterns of individual CD8+ T cells. Science. 2013;40:635-639. DOI: 10.1126/science.1235487
  75. 75. Gerlach, C. van Heijst JW, Swart E, Sie D, Armstrong N, Kerkhoven RM, Zehn D, Bevan MJ, Schepers K, Schumacher TN. One naive T cell, multiple fates in CD8+ T cell differentiation. The Journal of Experimental Medicine. 2010;207:1235-1246. DOI: 10.1084/jem.20091175
  76. 76. Reiner, SL, Adams, WC. Lymphocyte fate specification as a deterministic but highly plastic process. Nature Reviews Immunology. 2014;14(10):699-704. DOI: 10.1038/nri3734
  77. 77. Kelso A, Groves P, Troutt AB, Francis K. Evidence for the stochastic acquisition of cytokine profile by CD4+ T cells activated in a T helper type 2-like response in vivo. European Journal of Immunology. 1995;25:1168-1175. DOI: 10.1002/eji.1830250506
  78. 78. Raphael I, Nalawade S, Eagar TN, Forsthuber TG. T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine. 2015;74: 5-17. DOI: 10.1016/j.cyto.2014.09.011
  79. 79. DuPage M, Bluestone JA. Harnessing the plasticity of CD4+ T cells to treat immune‐mediated disease. Nature Reviews Immunology. 2016;16(3):149-163. DOI: 10.1038/nri.2015.18
  80. 80. Slack M, Wang T, Wang R. T cell metabolic reprogramming and plasticity. Molecular Immunology. 2015;68:507-512. DOI: 10.1016/j.molimm.2015.07.036
  81. 81. Coffman RL, Carty J. A T cell activity that enhances polyclonal IgE production and its inhibition by interferon‐γ. The Journal of Immunology. 1986;136:949-954
  82. 82. Mosmann T.R, Cherwinski H, Bond MW, Giedlin MA, Coffman RL. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. The Journal of Immunology. 1986;136:2348-2357
  83. 83. Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH. A novel transcription factor, T‐bet, directs Th1 lineage commitment. Cell. 2000;100 (6):655-669. DOI: http://dx.doi.org/10.1016/S0092‐8674(00)80702‐3
  84. 84. Amsen D, Antov A, Flavell RA. The different faces of Notch in T‐helper‐cell differentiation. Nature Reviews Immunology. 2009;9(2):116-124. DOI: 10.1038/nri2488
  85. 85. Caza T, Landas S. Functional and Phenotypic Plasticity of CD4(+) T Cell Subsets. Biomed Res Int. 2015; 2015:521957. doi: 10.1155/2015/521957. Epub 2015 Oct 25.
  86. 86. Amsen D, Spilianakis CG, Flavell RA. How are TH1 and TH2 effector cells made? Current Opinion in Immunology. 2009;21(2):153-160. DOI: 10.1016/j.coi.2009.03.010
  87. 87. Brucklacher‐Waldert V, Steinbach K, Lioznov M, Kolster M, Hölscher C, and Tolosa E. Phenotypical characterization of human Th17 cells unambiguously identified by surface IL‐17A expression. Journal of Immunology. 2009;183(9):5494-8501. DOI: 10.4049/jimmunol.0901000
  88. 88. Davila E, Kolls J. A ‘toll’ for Th17 cell expansion. Journal of Leukocyte Biology. 2010;88(1):5-7. DOI: 10.1189/jlb.0110057
  89. 89. Romano M, Sironi M, Toniatti C Polentarutti N, Fruscella P, Ghezzi P, Faggioni R, Luini W, van Hinsbergh V, Sozzani S, Bussolino F, Poli V, Ciliberto G, Mantovani A. Role of IL‐6 and its soluble receptor in induction of chemokines and leukocyte recruitment. Immunity. 1997;6(3):315-325. DOI: http://dx.doi.org/10.1016/S1074‐7613(00)80334‐9
  90. 90. Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, Chung Y, Ma L, Shah B, Panopoulos AD, Schluns KS, Watowich SS, Tian Q, Jetten AM, Dong C. T helper 17 lineage differentiation is programmed by orphan nuclear receptors RORα and RORγ. Immunity. 2008;28(1):29-39. DOI: 10.1016/j.immuni.2007.11.016
  91. 91. Veldhoen M, Uyttenhove C, van Snick J, Helmby H, Westendorf A, Buer J, Martin B, Wilhelm C, Stockinger B. Transforming growth factor‐β “reprograms” the differentiation of T helper 2 cells and promotes an interleukin 9‐producing subset. Nature Immunology. 2008;9(12):1341-1346. DOI: 10.1038/ni.1659
  92. 92. Dardalhon V, Awasthi A, Kwon H, Galileos G, Gao W, Sobel RA, Mitsdoerffer M, Strom TB, Elyaman W, Ho IC, Khoury S, Oukka M, Kuchroo VK. IL‐4 inhibits TGF‐β‐induced Foxp3+ T cells and, together with TGF‐β, generates IL‐9+ IL‐10+ Foxp3− effector T cells. Nature Immunology. 2008;9(12):1347-1355. DOI: 10.1038/ni.1677
  93. 93. Jabeen R, Goswami R, Awe O, Kulkarni A, Nguyen ET, Attenasio A, Walsh D, Olson MR, Kim MH, Tepper RS, Sun J, Kim CH, Taparowsky EJ, Zhou B, Kaplan MH. Th9 cell development requires a BATF‐regulated transcriptional network. Journal of Clinical Investigation. 2013;123(11):4641-4653. DOI: 10.1172/JCI69489
  94. 94. Pickert G, Neufert C, Leppkes M Zheng Y, Wittkopf N, Warntjen M, Lehr HA, Hirth S, Weigmann B, Wirtz S, Ouyang W, Neurath MF, Becker C. STAT3 links IL‐22 signaling in intestinal epithelial cells to mucosal wound healing. The Journal of Experimental Medicine. 2009;206(7):1465-1472. DOI: 10.1084/jem.20082683
  95. 95. Duhen T, Geiger R, Jarrossay D, Lanzavecchia A, Sallusto F. Production of interleukin 22 but not interleukin 17 by a subset of human skin‐homing memory T cells. Nature Immunology. 10:857-863, 2009. DOI: 10.1038/ni.1767
  96. 96. Trifari S, Kaplan CD, Tran EH, Crellin NK, Spits H. Identification of a human helper T cell population that has abundant production of interleukin 22 and is distinct from TH17, TH1 and TH2 cells. Nature Immunology. 2009;10(8):864-871. DOI: 10.1038/ni.1770
  97. 97. Kagami S, Rizzo HL, Lee JJ, Koguchi Y, Blauvelt A. Circulating Th17, Th22, and Th1 cells are increased in psoriasis. Journal of Investigative Dermatology. 2010;130:1373-1383. DOI: 10.1038/jid.2009.399
  98. 98. Sun B, Zhang Y. Overview of orchestration of CD4+ T cell subsets in immune responses. Advances in Experimental Medicine and Biology. 2014;841:1-235. DOI: 10.1007/978‐94‐017‐9487‐9_1
  99. 99. Breitfeld D, Ohl L, Kremmer E, Ellwart J, Sallusto F, Lipp M, Förster R. Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production. The Journal of Experimental Medicine. 2000;192:1545-1552. DOI: 10.1084/jem.192.11.1545
  100. 100. Hogquist KA. Signal strength in thymic selection and lineage commitment. Current Opinion in Immunology. 2001;13(2):225-31. DOI: http://dx.doi.org/10.1016/S0952‐7915(00)00208‐9
  101. 101. King C. New insights into the differentiation and function of T follicular helper cells. Nature Reviews Immunology. 2009;9(11):757-766. DOI: 10.1038/nri2644
  102. 102. Nurieva RI, Chung Y, Hwang D, Yang XO, Kang HS, Ma L, Wang YH, Watowich SS, Jetten AM, Tian Q, Dong C. Generation of T follicular helper cells is mediated by interleukin‐21 but independent of T helper 1, 2, or 17 cell lineages. Immunity. 2008;29(1):138-149. DOI: 10.1016/j.immuni.2008.05.009
  103. 103. Willimann K, Legler DF, Loetscher M, Roos RS, Delgado MB, Clark‐Lewis I, Baggiolini M, Moser B. The chemokine SLC is expressed in T cell areas of lymph nodes and mucosal lymphoid tissues and attracts activated T cells via CCR7. European Journal of Immunology. 1998;28:2025-2034. DOI: 10.1002/(SICI)1521‐4141(199806)28:06<2025::AID‐IMMU2025>3.0.CO;2‐C
  104. 104. Nagira M, Imai T, Yoshida R, Takagi S, Iwasaki M, Baba M, Tabira Y, Akagi J, Nomiyama H, Yoshie O. A lymphocyte‐specific CC chemokine, secondary lymphoid tissue chemokine (SLC), is a highly efficient chemoattractant for B cells and activated T cells. European Journal of Immunology. 1998;28:1516-1523. DOI: 10.1002/(SICI)1521‐4141(199805)28:05<1516::AID‐IMMU1516>3.0.CO;2‐J
  105. 105. Luther SA, Bidgol A, Hargreaves DC, Schmidt A, Xu Y, Paniyadi J, Matloubian M, Cyster JG. Differing activities of homeostatic chemokines CCL19, CCL21, and CXCL12 in lymphocyte and dendritic cell recruitment and lymphoid neogenesis. The Journal of Immunology. 2002;169:424-433. DOI: https://doi.org/10.4049/jimmunol.169.1.424
  106. 106. Haynes NM, Allen CD, Lesley R, Ansel KM, Killeen N, Cyster JG. Role of CXCR5 and CCR7 in follicular TH cell positioning and appearance of a programmed cell death gene‐1High germinal center‐associated subpopulation. The Journal of Immunology. 2007;179:5099-5108. DOI: https://doi.org/10.4049/jimmunol.179.8.5099.
  107. 107. Craft JE. Follicular helper T cells in immunity and systemic autoimmunity Nature Reviews Rheumatology. 2012;8:337-347. DOI: 10.1038/nrrheum.2012.58
  108. 108. Zhou X, Jeker LT, Fife BT, Zhu S, Anderson MS, McManus MT, Bluestone JA. Selective miRNAdisruption in T reg cells leads to uncontrolled autoimmunity. Journal of Experimental Medicine. 2008;205(9):1983-1991. DOI: 10.1084/jem.20080707
  109. 109. Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy KM.Th17: an effector CD4 T cell lineage with regulatory T cell ties. Immunity. 2006;24(6):677-688. DOI: 10.1016/j.immuni.2006.06.002
  110. 110. Zhou X, Bailey‐Bucktrout S, Jeker LT, Bluestone JA. Plasticity of CD4+ FoxP3+ T cells. Current Opinion in Immunology. 2009;21(3):281285. DOI: 10.1016/j.coi.2009.05.007
  111. 111. Lee YK, Mukasa R, Hatton RD, Weaver CT. Developmental plasticity of Th17 and Treg cells. Current Opinion in Immunology. 2009;21(3):274-280. DOI: 10.1016/j.coi.2009.05.021
  112. 112. Wakim LM, Bevan MJ. From the thymus to longevity in the periphery. Current Opinion in Immunology. 2010;22(3):274-278. DOI: 10.1016/j.coi.2010.03.003
  113. 113. Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401(6754):708-712. DOI:10.1038/44385
  114. 114. Fearon DT, Manders P, Wagner SD. Arrested differentiation, the self‐renewing memory lymphocyte, and vaccination. Science. 13 Jul 2001;293(5528):248-250. DOI: 10.1126/science.1062589
  115. 115. Tortorella C, Schulze‐Koops H, Thomas R, Splawski JB, Davis LS, Picker LJ, Lipsky PE. Expression of CD45RB and CD27 identifies subsets of CD4+ memory T cells with different capacities to induce B cell differentiation. The Journal of Immunology. 1995;155(1):149-162
  116. 116. Bryl E, Vallejo AN, Weyand CM, Goronzy JJ. Down‐regulation of CD28 expression by TNF‐alpha. Journal of Immunology. 2001;167(6):3231-3238. DOI: https://doi.org/10.4049/jimmunol.167.6.3231
  117. 117. Picker LJ, Reed‐Inderbitzin EF, Hagen SI, Edgar JB, Hansen SG, Legasse A, Planer S, Piatak M Jr, Lifson JD, Maino VC, Axthelm MK, Villinger F. IL‐15 induces CD4 effector memory T cell production and tissue emigration in nonhuman primates. Journal of Clinical Investigation. 2006;116(6):1514-1524. DOI: 10.1172/JCI27564
  118. 118. Geginat J, Lanzavecchia A, Sallusto F. Proliferation and differentiation potential of human CD8+ memory T‐cell subsets in response to antigen or homeostatic cytokines. Blood. 2003;101(11):4260-4266. DOI: 10.1182/blood‐2002‐11‐3577
  119. 119. Mittrücker HW, Visekruna A, Huber M. Heterogeneity in the Differentiation and Function of CD8+ T Cells. Archivum Immunologiae et Therapia Experimentalis. 2014;62:449-458. DOI: 10.1007/s00005‐014‐0293‐y
  120. 120. Reiser J, Banerjee A. Effector, Memory, and Dysfunctional CD8(+) T Cell Fates in the Antitumor Immune Response. J Immunol Res. 2016;2016:8941260. doi:10.1155/2016/8941260. Epub 2016 May 22.
  121. 121. Yang CY, Best JA, Knell J, Yang E, Sheridan AD, Jesionek AK, Li HS, Rivera RR, Lind KC, D'Cruz LM, Watowich SS, Murre C, Goldrath AW. The transcriptional regulators Id2 and Id3 control the formation of distinct memory CD8? T cell subsets. Nature Immunology. 2001;12:1221-1229. DOI: 10.1038/ni.2158
  122. 122. Förster R, Schubel A, Breitfeld D, Kremmer E, Renner‐Müller I, Wolf E, Lipp M. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell. 1999;99(1):23-33
  123. 123. Arbonés ML, Ord DC, Ley K, Ratech H, Maynard‐Curry C, Otten G, Capon DJ, Tedder TF. Lymphocyte homing and leukocyte rolling and migration are impaired in L‐selectin deficient mice. Immunity. 1994;1(4):247-260. DOI: 10.1016/1074‐7613(94)90076‐0
  124. 124. Bachmann MF, Wolint P, Schwarz K, Jäger P, Oxenius A. Functional properties and lineage relationship of CD8+ T cell subsets identified by expression of IL‐7 receptor α and CD62L. The Journal of Immunology. 2005;175(7):4686-4696. DOI: https://doi.org/10.4049/jimmunol.175.7.4686
  125. 125. Tang Y, Guan SP, Chua BY, Zhou Q, Ho AW, Wong KH, Wong KL, Wong WS, Kemeny DM. Antigen‐specific effector CD8 T cells regulate allergic responses via IFN‐gamma and dendritic cell function. Journal of Allergy and Clinical Immunology. 2012;129(1611-1620):e4. DOI: 10.1016/j.jaci.2011.12.976
  126. 126. Visekruna A, Ritter J, Scholz T, Campos L, Guralnik A, Poncette L, Raifer H, Hagner S, Garn H, Staudt V, Bopp T, Reuter S, Taube C, Loser K, Huber M. Tc9 cells, a new subset of CD8(+) T cells, support Th2‐mediated airway inflammation. European Journal of Immunology. 2013;43:606-618. DOI: 10.1002/eji.201242825
  127. 127. Huber M, Heink S, Pagenstecher A, Reinhard K, Ritter J, Visekruna A, Guralnik A, Bollig N, Jeltsch K, Heinemann C, Wittmann E, Buch T, Prazeres da Costa O, Brüstle A, Brenner D, Mak TW, Mittrücker HW, Tackenberg B, Kamradt T, Lohoff M. IL‐17A secretion by CD8+ T cells supports Th17‐mediated autoimmune encephalomyelitis. Journal of Clinical Investigation. 2013;123:247-260. DOI: 10.1172/JCI63681
  128. 128. Hamada H, Garcia‐Hernandez Mde L, Reome JB, Misra SK, Strutt TM, McKinstry KK, Cooper AM, Swain SL, Dutton RW. Tc17, a unique subset of CD8 T cells that can protect against lethal influenza challenge. The Journal of Immunology. 2009;182:3469-3481. DOI: 10.4049/jimmunol.0801814
  129. 129. Kim HJ, Cantor H. Regulation of self‐tolerancebyQa‐1‐restricted CD8+ regulatory T cells. Seminars in Immunology. 2011;23:446-452. DOI: 10.1016/j.smim.2011.06.001
  130. 130. Herberman RB, Nunn ME, Holden HT, Lavrin DH. Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic and allogeneic tumors. II. Characterization of effector cells. International Journal of Cancer. 1975;16(2):230-239
  131. 131. Di Santo JP. Natural killer cell developmental pathways: A question of balance. Annual Review of Immunology. 2006;24:257-286. DOI: 10.1146/annurev.immunol.24.021605.090700
  132. 132. Yu J, Freud AG, Caligiuri MA. Location and cellular stages of natural killer cell development. Trends in Immunology. 2013;34(12):573-582. DOI: 10.1016/j.it.2013.07.005
  133. 133. Yokota Y, Mansouri A, Mori S, Sugawara S, Adachi S, Nishikawa S, Gruss P. Development of peripheral lymphoid organs and natural killer cells depends on the helix‐loop‐helix inhibitor Id2. Nature. 1999;397(6721):702-706. DOI: 10.1038/17812
  134. 134. Colucci F, Samson SI, DeKoter RP, Lantz O, Singh H, Di Santo JP. Differential requirement for the transcription factor PU.1 in the generation of natural killer cells versus B and T cells. Blood. 2001; 97(9):2625-2632. DOI: https://doi.org/10.1182/blood.V97.9.2625
  135. 135. Yun S, Lee SH, Yoon SR, Kim MS, Piao ZH, Myung PK, Kim TD, Jung H, Choi I. TOX regulates the differentiation of human natural killer cells from hematopoietic stem cells in vitro. Immunology letters. 2011;136(1):29-36. DOI: 10.1016/j.imlet.2010.11.008
  136. 136. Gordon SM, Chaix J, Rupp LJ, Wu J, Madera S, Sun JC, Lindsten T, Reiner SL. The transcription factors T‐bet and Eomes control key checkpoints of natural killer cell maturation. Immunity. 2012;36(1):55-67. DOI: 10.1016/j.immuni.2011.11.016
  137. 137. Montaldo E, Vacca P, Moretta L, Mingari MC. Development of human natural killer cells and other innate lymphoid cells. Seminars in immunology. 2014;26(2):107-113. DOI: 10.1016/j.smim.2014.01.006
  138. 138. Marcenaro E, Ferranti B, Falco M, Moretta L, Moretta A. Human NK cells directly recognize Mycobacterium bovis via TLR2 and acquire the ability to kill monocyte‐derived DC. International Immunology. 2008;20(9):1155-1167. DOI: 10.1093/intimm/dxn073
  139. 139. Moretta A, Bottino C, Vitale M, Pende D, Biassoni R, Mingari MC, Moretta L. Receptors for HLA class‐I molecules in human natural killer cells. Annual Review of Immunology. 1996;14:619-648. DOI: 10.1146/annurev.immunol.14.1.619
  140. 140. Bottino C, Castriconi R, Pende D, Rivera P, Nanni M, Carnemolla B, Cantoni C, Grassi J, Marcenaro S, Reymond N, Vitale M, Moretta L, Lopez M, Moretta A. Identification of PVR (CD155) and Nectin‐2 (CD112) as cell surface ligands for the human DNAM‐1 (CD226) activating molecule. The Journal of Experimental Medicine. 2003;198(4):557-567. DOI: 10.1084/jem.20030788
  141. 141. Caligiuri MA. Human natural killer cells. Blood. 2008;112(3):461-469. DOI: 10.1182/blood‐2007‐09‐077438
  142. 142. Freud AG, Caligiuri MA. Human natural killer cell development. Immunological reviews. 2006;214:56-72. DOI: 10.1111/j.1600‐065X.2006. 00451.x
  143. 143. Sivori S, Cantoni C, Parolini S, Marcenaro E, Conte R, Moretta L, Moretta A. IL‐21 induces both rapid maturation of human CD34+ cell precursors towards NK cells and acquisition of surface killer Ig‐like receptors. European journal of immunology. 2003;33(12):3439-3447. DOI: 10.1002/eji.200324533
  144. 144. Montaldo E, Vitale C, Cottalasso F, Conte R, Glatzer T, Ambrosini P, Moretta L, Mingari MC. Human NK cells at early stages of differentiation produce CXCL8 and express CD161 molecule that functions as an activating receptor. Blood. 2012;119(17):3987-3996. DOI: 10.1182/blood‐2011‐09‐379693
  145. 145. Walker JA, Barlow JL, McKenzie AN. Innate lymphoid cells–how did we miss them? Nature Reviews Immunology. 2013;13:75-87. DOI: 10.1038/nri3349
  146. 146. Satoh‐Takayama N, Lesjean‐Pottier S, Vieira P, Sawa S, Eberl G, Vosshenrich CA, Di Santo JP. IL‐7 and IL‐15 independently program the differentiation of intestinal CD3−NKp46+ cell subsets from Id2‐dependent precursors. The Journal of Experimental Medicine. 2010;207:273-280. DOI: 10.1084/jem.20092029
  147. 147. Magri G, Miyajima M, Bascones S, Mortha A, Puga I, Cassis L, Barra CM et al. Innate lymphoid cells integrate stromal and immunological signals to enhance antibody production by splenic marginal zone B cells. Nature Immunology. 2014;15(4):354-364. DOI:10.1038/ni.2830
  148. 148. Bernink JH, Peters CP, Munneke M, te Velde AA, Meijer SL, Weijer K, Hreggvidsdottir HS, Heinsbroek SE, Legrand N, Buskens CJ, Bemelman WA, Mjösberg JM, Spits H. Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues. Nature Immunology. 2013;14:221-229. DOI: 10.1038/ni.2534
  149. 149. Neill DR, Wong SH, Bellosi A, Flynn RJ, Daly M, Langford TK, Bucks C, Kane CM, Fallon PG, Pannell R, Jolin HE, McKenzie AN. Nuocytes represent a new innate effector leukocyte that mediates type‐2 immunity. Nature. 2010; 464:1367-1370. DOI: 10.1038/nature08900
  150. 150. Mjösberg J, Bernink J, Golebski K, Karrich JJ, Peters CP, Blom B, te Velde AA, Fokkens WJ, van Drunen CM, Spits H. The transcription factor GATA3 is essential for the function of human type 2 innate lymphoid cells. Immunity. 2012;37:649-659. DOI: 10.1016/j.immuni.2012.08.015
  151. 151. Mjösberg JM, Trifari S, Crellin NK, Peters CP, van Drunen CM, Piet B, Fokkens WJ, Cupedo T, Spits H. Human IL‐25‐ and IL‐33‐responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nature Immunology. 2011;12:1055-1062. DOI: 10.1038/ni.2104
  152. 152. Cella M, Otero K, Colonna M. Expansion of human NK‐22 cells with IL‐7, IL‐2, and IL‐1β reveals intrinsic functional plasticity. Proceedings of the National Academy of Sciences. USA 2010;107:10961-10966. DOI: 10.1073/pnas.1005641107
  153. 153. Lee JS, Cella M, McDonald KG, Garlanda C, Kennedy GD, Nukaya M, Mantovani A, Kopan R, Bradfield CA, Newberry RD, Colonna M. AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nature Immunology. 2012;13:144-151. DOI: 10.1038/ni.2187
  154. 154. Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, Ivanov II, Littman DR, O'Shea JJ. Lymphoid tissue inducer‐like cells are an innate source of IL‐17 and IL‐22. The Journal of Experimental Medicine. 2009;206:35-41. DOI: 10.1038/nri1781
  155. 155. Golubovskaya V, Wu L. Different subsets of T cells, memory, effector functions, and CAR‐T immunotherapy. Cancers (Basel). 2016;8(3). pii: E36. DOI: 10.3390/cancers8030036
  156. 156. Hazenberg MD, Spits H. Human innate lymphoid cells. Blood. 2014;124(5):700-709. DOI: 10.1182/blood‐2013‐11‐427781

Written By

Luciana Cavalheiro Marti, Nydia Strachman Bacal, Laiz Camerão Bento, Rodolfo Patussi Correia and Fernanda Agostini Rocha

Reviewed: 07 April 2017 Published: 12 July 2017