Open access peer-reviewed chapter

Multiple Players in the Mechanical Control of T Cell Quiescence

Written By

Ahmed Fadhil Neama, Chung Yeng Looi and Won Fen Wong

Submitted: 13 March 2017 Reviewed: 29 March 2017 Published: 12 July 2017

DOI: 10.5772/intechopen.68869

From the Edited Volume

Lymphocyte Updates - Cancer, Autoimmunity and Infection

Edited by Gheorghita Isvoranu

Chapter metrics overview

1,753 Chapter Downloads

View Full Metrics

Abstract

Naive T cells are kept in a quiescence state, characterized by small cell size, with low proliferative and metabolic activities, until antigen engagement. T lymphocyte quiescence is a tightly controlled mechanism regulated by multiple quiescence‐associated factors. Loss or impaired functions of these factors regularly result in spontaneous activation of T cells that is ensured by fatal autoimmune diseases. Elucidating the mechanism to facilitate the switch on or off of T cells could be beneficial to ameliorate pathology triggered by T cell hyperactivation or dysfunction. In this chapter, we discuss multiple quiescence‐associated factors along with the mechanisms utilized to promote lymphocyte quiescence and longevity.

Keywords

  • T cell quiescence
  • Foxo
  • KLF2

1. Introduction

T lymphocytes are important players in adaptive immune responses to invading pathogens. In an individual, T lymphocyte repertoires are principally generated through a somatic recombinant process named as VDJ rearrangement in the thymus. Upon maturation, these T cells exit thymus to reside in the secondary lymph organs and patrol in the circulation. It is crucial to maintain T cell repertoires as standby for future assault by diverse types of antigens that might come from the massive array of microbes. Eventually, most of the lymphocyte reservoirs do not encounter their cognate (or specific) antigens throughout their lifetime. These virgin or naïve T cells must be kept in a state known as quiescence to prevent immune activation that is often ensued by activation‐induced cell exhaustion or death. At quiescence state, cells are sustained through homeostatic cell renewal without activation or expansion to maintain the size of the peripheral lymphocyte pool. Loss control of T cell quiescence has been associated with autoimmune diseases; hence, this stage is imperative to render an immune tolerance. In this chapter, we discuss the intricate transcriptional mechanism of T lymphocytes at quiescence stage to attain a long‐term standby status until they encounter the cognate antigens.

Lymphocyte quiescence refers to a state of inaction of cells characterized by small cell size with limited cytoplasmic region, low rates of cell metabolism, proliferation, transcription, and translation activities. Quiescence suppresses cell activation and prevents the unnecessary use of energy resources that will be consumed by the huge T lymphocyte repertoires in an individual. In addition, cells at quiescence state can reduce genetic damage due to repetitive replication therefore preventing development of malignancy [1] because constant replication of lymphocytes may increase the risk of leukemia and lymphoma [2]. Upon identification of a specific antigen, activation of a T cell beyond the cell signaling threshold triggers cells to exit quiescence state in a non‐reversible manner, thus, the T cell undergoes robust clonal expansion followed by cell cytotoxicity and cytokine‐secreting activities.

A quiescence state of lymphocytes was previously regarded as a default stage of cells in the absence of antigen recognition activation [3]. However, recent evidences accumulated suggest that quiescence is a steadily regulated stage by functionally diverse mechanisms [4], which include intrinsic control by gene expression programs as well as extrinsic suppression by regulatory T cells (Treg). Evidence derived from the microarray study demonstrates strikingly distinct expression patterns of diverse molecules in the quiescence versus stimulated T lymphocytes [5, 6]. Following T cell activation, the major change in gene expression profile is not limited to only increased expression of genes that promote growth and differentiation but also suppression of a group of genes that is linked to the quiescence program [6, 7]. Transcription factors and components in cell cycle control play a central role in the quiescence regulation. Recently, ubiquitination degradation pathway has also been added into the growing list of the quiescence‐associated factors [8].

A common characteristic shared by most of these quiescence‐associated molecules is their high abundance in the naïve T cell but the expression has rapidly vanished upon cell activation. Quiescence molecules are different from other negative regulators of T cells such as cytotoxic T lymphocyte antigen 4 (CTLA4) and program cell death protein 1 (PD‐1). Although both impose inhibitory signals on T cell proliferation and effector activity, it is important to note that the expression of quiescence molecules is usually high in naïve T cells but reduce upon cell activation. In contrast, CTLA4 and PD1‐1 are expressed only after T cell activation.

Deficiency or dysfunction of the quiescence molecules often results in loss of the quiescence control marked by the cell's semi‐activation to hyperactivation with robust proliferation, accompanied by exert activities such as cytokines secretion and predisposition to apoptotic cell death. The direct physical consequence to loss of T cells quiescence is impaired immune tolerance and development of autoimmune diseases in an individual. In this chapter, we will discuss several quiescence factors along with the mechanisms utilized to promote lymphocyte quiescence and longevity.

Advertisement

2. KLF2

The Sp/Kruppel‐like factor (KLF) family of zinc‐finger transcription factors contains at least 20 identified members, which include numerous Kruppel‐like factors that have different roles across the mammalian system [9]. Kruppel‐like family 2 (KLF2, also known as lung Kruppel‐like factor or LKLF) has been implicated in programming T cell quiescence [10]. KLF2 is highly expressed in mature CD4+ and CD8+ T cells. Similar to many other key players in quiescence control, its expression is rapidly switched off after T cell activation [11]. KLF2 can also be detected in thymocytes at single‐positive (SP) stage but not during earlier double‐negative (DN) or double‐positive (DP) stages [11]. KLF2 controls mature T cell egress from thymus [12] and recirculation through secondary lymphoid tissues [13] by regulating transcription of sphingosine‐1‐phosphate receptor 1 (S1Pr1). KLF2‐deficient thymocytes show impaired thymocyte emigration, whereas KLF2‐transduced T cells are prone to homing in lymphoid organs following adoptive transfer [14].

The conventional KLF2‐deficient mice died between embryonic days, from 12.5 to 14.5, due to severe intra‐embryonic and intra‐amniotic hemorrhage resulting from defects in the smooth muscle cells migration during blood vessel maturation [15, 16]. To study KLF2‐deficient T cells, a Rag‐2−/−KLF2−/− chimeric mouse system was applied, in which KLF2−/− embryonic stem cells were injected into RAG2−/− blastocyst to populate the T cell pool [11]. This model provides strong evidence supporting the role of KLF2 in the quiescence control. First, a massive loss (up to 90%) of the peripheral T cell is observed. Intriguingly, these KLF2‐deficient T cells displayed stigmata of activated phenotype, that is, an increased cell size and surface expression of activation markers (CD69hi CD44hi CD62Llo); however, these cells are non‐proliferative. A large number of KLF2‐deficient T cells are apoptotic, attributable to high surface expression of Fas ligand (FasL) [11].

In a different experiment using an overexpression model, in vitro forced expression of KLF2 in Jurkat T cells using doxycycline inducible system programs the cells into a quiescent phenotype [17]. KLF2 overexpression dramatically inhibits proliferation of Jurkat T cells and prevents synthesis of surface molecules such as CD30 and CD71, by which this effect can be reversible when the KLF2 expression is removed. KLF2‐mediated regulation of quiescent T cells is partially achieved through its suppression of c‐Myc. Conversely, transient expression of MadMyc, a dominant negative form of c‐Myc, recapitulates the phenotype produced by KLF2 overexpression [17]. On the other hand, expression of neurotransmitter dopamine D4 receptor on resting T cells promotes T cell quiescence by upregulating KLF2 expression and an administration of U101958; a D4 antagonist could diminish the effect [18]. KLF2‐deficient B cells showed increased apoptosis and impaired proliferation after B‐cell receptor cross‐linking [19]. B cell distribution and trafficking are disturbed due to low surface expression of CD62L and β7-integrin expression. Percentages of B cell subsets is also disturbed as B1 cells are almost disminished accompanied by increased in the number of MZ and transitional B cells. [19, 20]. These suggest a potential role of KLF2 in control of B‐cell quiescence.

Another KLF family member, KLF4, also known as gut‐enriched Kruppel‐like factor or GKLF, is also important in T cell biology by regulating thymocyte development and IL‐17 expression during Th17 differentiation [21]. Both thymocytes and mature T cells express high level of KLF4. In KLF4 knock‐out mice, the proliferation of thymocytes at double‐negative stage was significantly reduced, attributed to loss of KLF4 control on Cdkn1b, a cell cycle molecule. KLF4 is also involved in Th17 differentiation and IL‐17 expression by which its deficiency contributes to reduced IL‐17 production and thus ameliorates the severity of in vivo experimental autoimmune encephalomyelitis [21]. KLF4 is able to exert a global inhibitory effect on macromolecular biosynthesis, including protein biosynthesis, transcription, and cholesterol biosynthesis [22]. The expression pattern of KLF4 in B cells highly resembles those in T cells, whereby the expression is abundant in mature resting cells but rapidly decreased upon cell activation. In KLF4‐deficient mice, a modest decrease in the numbers of pre‐B cells in the bone marrow and mature B cells in the spleen can be observed [23]. Fewer B cells enter S phase of the cell cycle and complete cell division in response to BCR and/or CD40L engagement, in vitro, in the absence of KLF4, suggesting its role in maintaining quiescence in B cells. This could be a result of decreased expression of cyclin D2 in B cells because KLF4 regulates cyclin B2 through a direct binding to its promoter [23]. Thus, we can also postulate a potential role of KLF4 in controlling T or B lymphocytes quiescence.

Advertisement

3. Foxo

Forkhead box (Foxo) family genes are the orthologs of DAF‐16 gene identified in nematode worm Caenorhabditis elegans, which programs cells for resistance to oxidative stress and cell cycle control to maintain cells at a dauer (non‐action) state [24]. In mammals, there are three members in Foxo family, Foxo1, Foxo3, and Foxo4. The Foxo family of transcription factors triggers the induction or suppression of multiple target genes dependent on context molecules [25] and hence plays multiple functions in cell quiescence control [3, 2629], including maintenance of stem cells pluripotency [30], oxidative stress control [31], cell cycle, cancer progress [32], and others. In T cells, Foxo molecules have multiple roles by controlling cell‐surface molecules, signaling proteins, and nuclear factors that control gene expression [33]. Foxo1 is also detected in thymocyte subsets, dominant negative inhibition of Foxo1 causes increased proliferation capacity of thymocytes, thus interferes with central tolerance control [34].

In the animal model, Foxo1 deletion causes spontaneous T cell activation that leads to development of colitis [35]. Higher percentages of activated/memory T phenotypes have been reported in T cell‐specific Foxo1 knock‐out mice model [35, 36]. Besides, inflammatory bowel disease is also observed in the wildtype mice after receiving Foxo1‐deficient T cells. In a mouse model with CD4 promoter‐driven T cell‐specific deletion of Foxo1, mice develop exocrine pancreatitis, hind limb paralysis, and multiorgan lymphocyte infiltration. Anti‐nuclear antibodies and formation of germinal centers are detected in mice, suggesting Foxo1 suppresses cells from differentiating into follicular helper (TFH) subtypes [37]. Foxo1‐deficient T cells demonstrate a highly defective ability for cell homing to lymph nodes, due to impaired L‐selectin [38] and CCR7 expression [36]. Although some argue that Foxo is not a true quiescence factor stating the activated phenotype may be due to its functions like cell trafficking, these distinct roles of Foxo may directly or indirectly impose lymphocyte quiescence.

In contrast to Foxo1's specific expression in lymphocytes, Foxo3 is ubiquitously expressed in many tissues in the body. It mediates cell death in many cells including T and B lymphocytes. The role of Foxo3 in the regulation of cell quiescence remains controversial. An earlier report using Fox3TRAP mice (created by retroviral gene‐trap technique) demonstrates typical autoimmune characteristics including spontaneous lymphoproliferation, hyperactivation, and lymphocyte infiltration into multiple organs [39]. However, another two mice generated, using gene‐trap, show no such symptoms except for abnormal ovary development [40, 41]. Different mice generated with targeted recombinant techniques demonstrated only some decrease in the number of pre‐B and circulating B cells [42, 43]. Foxo3 protects quiescent cells from oxidative stress through regulation of antioxidant manganese superoxide dismutase (MnSOD) [44] and growth arrest, and damage response gene (Gadd45a) is a direct target of Foxo3a [45].

In the last decade, increasing numbers of target genes regulated by Foxo transcription factor have been identified. These include KLF2 [36], GTPase of immunity‐associated protein 5 (Gimap5) [46], IL‐7Ra, homing molecules (L‐selectin, CCR7, and Fam65b), CTLA4 [37], and shingosine‐1 phosphate receptor [38], among others. A major target of Foxo is KLF2, an essential transcription factor for quiescence control. Introduction of Foxo1 into T cells causes induction of KLF2 transcription factor while T cell's specific deletion of Foxo1 showed lower expression of KLF2 [36]. Foxo1 binds directly to promoter of KLF2 gene to induce its expression [47].

Advertisement

4. Foxp1

A member in the subfamily P of the large Fox family, Forkhead box protein P1 (Foxp1), has an essential role in B lymphopoiesis to control the expression of recombination‐activating genes 1 and 2 and transition from pro B to pre B cells [48]. Foxp1 has been implicated in the quiescence control of naïve T cells by inhibiting IL‐7Ra expression and diminishing signaling by the kinase Erk [49, 50]. Acute deletion of Foxp1 induces naïve T cells to gain effector phenotype. Homeostatic proliferation of quiescence cells is regulated by IL7 signaling pathway, which can be negatively regulated by autocrine feedback control of IL7Ra expression [51]. Transcription factor Foxp1 helps maintain the quiescence of naïve T cells by binding to 3.5 kb upstream of IL7R transcription start site and inhibiting IL‐7Ra expression [36]. Foxp1 is a negative regulator of Foxo1 as they compete with each other for the forkhead binding site at IL7R enhancer region [49].

Advertisement

5. Tob

A member of the Tob family shares a highly conserved NH2 terminal sequence. Tob is expressed in resting T cells as well as in anergic T cells [52]. Its expression is diminished upon T cell activation by anti‐CD3/anti‐CD28 or mitogen PMA stimulation. Forced expression of exogenous Tob molecule inhibits T cell proliferation. Tob interacts with Smad2 and Smad4 molecules and enhances Smad4, signaling to suppress the transcription of multiple cytokines including IL‐2. Overexpression of Tob also blocks cell cycle progression by promoting p27kip1, a negative regulator of cell cycle. In contrast, the positive regulator of cell cycle molecules including cyclin E, cyclin A, and Cdk2 was suppressed. Elimination of Tob protein synthesis using antisense oligonucleotide reduces the threshold of T cell activation.

Advertisement

6. Tsc1

Tuberous sclerosis 1 (Tsc1) functions as a GTPase‐activating protein (GAP) that binds small GTPase RHEB and negatively regulates mTOR1 signaling. Tsc1 is important in T cell biology in memory cell differentiation, effector, and regulatory functions [53, 54]. Tsc1 is also implicated in anergy T cells, and its expression is higher in anergy as compared to activated T cells [55]. Tsc1−/− T cells in mice model loss quiescence as demonstrated by increased cell surface marker (CD44hiCD122) and prominent upregulation of activation markers such as CD69, CD25 and CD71 [56]. Besides, Tsc1−/− T cells demonstrated increased cell size, proliferation, reactive oxygen species (ROS) generation, and susceptibility to apoptosis. Tsc1 deficiency also dampens anti‐bacterial immune response in animal model as reduced OVA‐reactive tetramer‐positive T cells and interferon‐producing CD8+ T cells. The effect of Tsc1 deficiency is attributed to its ability to inhibit mTORC1 as this effect can be reverted by rapamycin treatment.

Advertisement

7. Slfn2

The word “schlafen” means sleeping in German. Schlafen (Slfn) family of genes, so‐called owing to their ability to promote cells into an inactive state, consists of six genes with RNA helicase‐like motif in human. Schlafen proteins promote growth inhibitory responses and play roles in thymocytes development [57], effector, and regulatory T cells [58]. Slfn2 was added to the gaining list of quiescence factors coincidentally when scientists investigate the phenotype in elektra mouse, a G3 mice homozygous for chemically induced random germline mutation [59]. The eureka mice carry a single mutation in Slfn2, which results in isoleucine‐to‐asparagine substitution of amino acid residue 135, which is induced after exposure to N‐ethyl‐N‐nitrosourea. Eureka mice are defenseless and succumb to lymphocytic choriomeningitis virus and Listeria monocytogenes infection due to immunodeficient phenotype. This increased susceptibility to bacterial and viral infections can be reversed with bacteria artificial chromosome (BAC) transgenesis of Sfln2 gene, thus confirming the role of Sfln2. Interestingly, T cells from eureka mice exist in a semi‐activated state [59]. The T cells are generally lesser in amount but express higher surface activation marker (CD44hi) and proliferate strongly and are more prone to apoptosis in response to anti‐CD3/anti‐CD28 activation signal. A chronic ER stress under steady‐state conditions observed in Eureka T cells could explain the loss of immune cell quiescence [60]. Slfn2 has been suggested as a promising target for treating human T‐ALL malignancy [61]. In T‐ALL mice model, impaired Slfn2 functions rescue the mice from disease progress as the proliferation potential and survival of leukemic T cells are affected. In addition, the symptoms of severe lymphoproliferative disease in Fas deficient mice can also be rescued by Sfln2 loss-of-function mutation.

Advertisement

8. Runx1

Runx family comprise three members: Runx1 and Runx3 play crucial roles in T cell development and differentiation [62], whereas Runx2 is a key player in osteoblast differentiation during bone formation [63]. Runx1 knock‐out mice die of impaired fetal hematopoiesis at embryonic day E12.5 [64]. Conditional knock‐out mice driven by CD4‐Cre promoter lead to low number of CD4+ T cell population attributed to apoptotic cell death. When an anti‐apoptotic Bcl‐2 transgene is introduced to rescue the T cells, Runx1−/− T cells demonstrate spontaneous hyperactivation (CD44hiCD62Llo) phenotype. This mouse model displays a breakdown of immune tolerance as the signs and complications of systemic inflammatory response syndrome (SIRS) such as cytokine storm, monocytosis, blood coagulation, and muscle wasting syndrome can be observed. Infiltration of Runx1−/− cells into the lung causes autoimmune lung disease similar to human pulmonary alveolar proteinosis (PAP) [65]. In addition, increased surface activation markers, CD40L and CD69, were observed along with increased cytokine, chemokines, and other signaling molecules [66]. It is noteworthy that similar to other quiescence factors such as KLF2, Runx1 is abundantly expressed in naïve T cells, and TCR signaling results in rapid reduction of Runx1 expression [67]. One possible mechanism of Runx1 to exert quiescence is through silencing the expression of cytokines IL‐2. Besides, Runx1 may control quiescence indirectly through transcriptional control of Foxp1, Foxo1, and KLF2 by binding to their promoter region [66].

Advertisement

9. Peli

Ubiquitination is a post‐translational mechanism for protein degradation. During ubiquitination, small ubiquitin proteins attach to the lysine residues of substrate protein catalyzed by sequential action of E1, E2, and E3 ubiquitin‐activating enzymes. Ubiquitination process has been shown to involve in immune regulation [8, 68, 69]. Peli family is composed of three members, Peli1, Peli2, and Peli3 [70], among which E3 ligase Peli1 has been implicated in T cell quiescence control. CD4+ and CD8+ T cells from Peli‐deficient mice demonstrate hyperactivation and increased proliferation response upon TCR‐CD28 signaling [8]. Most of the Peli1−/− T cells turn into memory cells. Peli−/− mice develop autoimmune diseases, as demonstrated by multiorgan inflammation, detection of antinuclear autoantibody, and prominent immune complex deposition in kidney, and more pathogenic potentials are induced in experimental autoimmune encephalitis model [8].

Advertisement

10. Gimap5

Gimap5 stands for GTPase of immunity‐associated protein 5. A missense mutation in the Gimap5 results in abrogation of quiescence and reduced number and survival of lymphocytes [46]. Gimap5‐deficient CD4+ T cells from the mice are Th1/Th17 polarized and thus promotes colitis and early mortality. Gimap5 also plays a role in regulatory T cells because in its absence, regulatory T cells become reduced in frequency in the peripheral tissues and their immunosuppressive capacity becomes impaired.

11. Summary

Mature T lymphocytes in our body can remain in a quiescence state for a prolonged duration in the absence of infectious or stimulatory factors. Loss of T cell quiescence control leads to breakdown of immune tolerance and is the main causative factor for various types of autoimmune diseases, lymphoma, and leukemia. The understanding of the interaction among the multiple factors that are involved in intrinsic control of quiescence status is hence crucial to control the balance between immune tolerance and activation.

Acknowledgments

This study is supported by Fundamental Research Grant Scheme (FP030‐2014A) from Malaysia Ministry of Higher Education.

Abbreviations

VDJvariable, diversity and joining
SpSpecificity protein transcription factors
ILinterleukin
BCRreakpoint cluster region protein
PMAphorbol 12-myristate 13-acetate
OVAovalbumin
ERendoplasmic reticulum
CDcluster of differentiation
Th17helper T 17
RHEBRas homolog enriched in brain
CDKH1bCyclin-dependent kinase inhibitor 1B
CCR7C-C chemokine receptor type 7
Fam65bFamily With Sequence Similarity 65 Member B
T-ALLT-cell acute lymphoblastic leukaemia
RunxRunt-related transcription factor
PeliPellino E3 Ubiquitin Protein Ligase
BCL-2B-cell lymphoma 2

References

  1. 1. Tzachanis D, Boussiotis VA. Tob, a member of the APRO family, regulates immunological quiescence and tumor suppression. Cell Cycle. 2009;8:1019-1025. DOI: 10.4161/cc.8.7.8033
  2. 2. Horton MR, Powell JD. Quieting T cells with Slfn2. Nature Immunology. 2010;11:281-282. DOI: 10.1038/ni0410‐281
  3. 3. Yusuf I, Fruman DA. Regulation of quiescence in lymphocytes. Trends in Immunology. 2003;24:380‐386. DOI: S1471490603001418 [pii].
  4. 4. Tzachanis D, Lafuente EM, Li L, Boussiotis VA. Intrinsic and extrinsic regulation of T lymphocyte quiescence. Leukemia & Lymphoma. 2004;45:1959-1967. DOI: 10.1080/1042819042000219494
  5. 5. Feske S, Giltnane J, Dolmetsch R, Staudt LM, Rao A. Gene regulation mediated by calcium signals in T lymphocytes. Nature Immunology. 2001;2:316-324. DOI: 10.1038/86318
  6. 6. Glynne R, Ghandour G, Rayner J, Mack DH, Goodnow CC. B‐lymphocyte quiescence, tolerance and activation as viewed by global gene expression profiling on microarrays. Immunological Reviews. 2000;176:216-246
  7. 7. Teague TK, Hildeman D, Kedl RM, Mitchell T, Rees W, Schaefer BC, Bender J, Kappler J, Marrack P. Activation changes the spectrum but not the diversity of genes expressed by T cells. Proceedings of the National Academy of Sciences of the United States of America. 1999;96:12691-12696
  8. 8. Chang M, Jin W, Chang JH, Xiao Y, Brittain GC, Yu J, Zhou X, Wang YH, Cheng X, Li P, et al. The ubiquitin ligase Peli1 negatively regulates T cell activation and prevents autoimmunity. Nature Immunology. 2011;12:1002-1009. DOI: 10.1038/ni.2090
  9. 9. Black AR, Black JD, Azizkhan‐Clifford J. Sp1 and kruppel‐like factor family of transcription factors in cell growth regulation and cancer. Journal of Cellular Physiology. 2001;188:143-160. DOI: 10.1002/jcp.1111
  10. 10. Di Santo JP. Lung Krupple‐like factor: A quintessential player in T cell quiescence. Nature Immunology. 2001;2:667-668. DOI: 10.1038/90598
  11. 11. Kuo CT, Veselits ML, Leiden JM. LKLF: A transcriptional regulator of single‐positive T cell quiescence and survival. Science. 1997;277:1986-1990
  12. 12. Carlson CM, Endrizzi BT, Wu J, Ding X, Weinreich MA, Walsh ER, Wani MA, Lingrel JB, Hogquist KA, Jameson SC. Kruppel‐like factor 2 regulates thymocyte and T‐cell migration. Nature. 2006;442:299-302. DOI: 10.1038/nature04882
  13. 13. Yamada T, Park CS, Mamonkin M, Lacorazza HD. Transcription factor ELF4 controls the proliferation and homing of CD8+ T cells via the Kruppel‐like factors KLF4 and KLF2. Nature Immunology. 2009;10:618-626. DOI: 10.1038/ni.1730
  14. 14. Bai A, Hu H, Yeung M, Chen J. Kruppel‐like factor 2 controls T cell trafficking by activating L‐selectin (CD62L) and sphingosine‐1‐phosphate receptor 1 transcription. Journal of Immunology. 2007;178:7632-7639
  15. 15. Kuo CT, Veselits ML, Barton KP, Lu MM, Clendenin C, Leiden JM. The LKLF transcription factor is required for normal tunica media formation and blood vessel stabilization during murine embryogenesis. Genes & Development. 1997;11:2996-3006
  16. 16. Wu J, Bohanan CS, Neumann JC, Lingrel JB. KLF2 transcription factor modulates blood vessel maturation through smooth muscle cell migration. Journal of Biological Chemistry. 2008;283:3942-3950. DOI: 10.1074/jbc.M707882200
  17. 17. Buckley AF, Kuo CT, Leiden JM. Transcription factor LKLF is sufficient to program T cell quiescence via a c‐Myc‐dependent pathway. Nature Immunology. 2001;2:698-704. DOI: 10.1038/90633
  18. 18. Sarkar C, Das S, Chakroborty D, Chowdhury UR, Basu B, Dasgupta PS, Basu S. Cutting edge: Stimulation of dopamine D4 receptors induce T cell quiescence by up‐regulating Kruppel‐like factor‐2 expression through inhibition of ERK1/ERK2 phosphorylation. Journal of Immunology. 2006;177:7525-7529
  19. 19. Hart GT, Wang X, Hogquist KA, Jameson SC. Kruppel‐like factor 2 (KLF2) regulates B‐cell reactivity, subset differentiation, and trafficking molecule expression. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:716-721. DOI: 10.1073/pnas.1013168108
  20. 20. Winkelmann R, Sandrock L, Porstner M, Roth E, Mathews M, Hobeika E, Reth M, Kahn ML, Schuh W, Jack HM. B cell homeostasis and plasma cell homing controlled by Kruppel‐like factor 2. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:710-715. DOI: 10.1073/pnas.1012858108
  21. 21. An J, Golech S, Klaewsongkram J, Zhang Y, Subedi K, Huston GE, Wood WH 3rd, Wersto RP, Becker KG, Swain SL, et al. Kruppel‐like factor 4 (KLF4) directly regulates proliferation in thymocyte development and IL‐17 expression during Th17 differentiation. FASEB Journal. 2011;25:3634-3645. DOI: 10.1096/fj.11‐186924
  22. 22. Whitney EM, Ghaleb AM, Chen X, Yang VW. Transcriptional profiling of the cell cycle checkpoint gene kruppel‐like factor 4 reveals a global inhibitory function in macromolecular biosynthesis. Gene Expression. 2006;13:85-96
  23. 23. Klaewsongkram J, Yang Y, Golech S, Katz J, Kaestner KH, Weng NP. Kruppel‐like factor 4 regulates B cell number and activation‐induced B cell proliferation. Journal of Immunology. 2007;179:4679-4684
  24. 24. Burgering BM. A brief introduction to FOXOlogy. Oncogene. 2008;27:2258‐2262. DOI: 10.1038/onc.2008.29
  25. 25. van der Vos KE, Coffer PJ. FOXO‐binding partners: It takes two to tango. Oncogene. 2008;27:2289-2299. DOI: 10.1038/onc.2008.22
  26. 26. Coffer PJ. Transcriptional regulation of lymphocyte quiescence: As cunning as a FOX. Trends in Immunology. 2003;24:470-471; author reply 471
  27. 27. Eijkelenboom A, Burgering BM. FOXOs: Signalling integrators for homeostasis maintenance. Nature Reviews Molecular Cell Biology. 2013;14:83-97. DOI: 10.1038/nrm3507
  28. 28. Hedrick SM. The cunning little vixen: Foxo and the cycle of life and death. Nature Immunology. 2009;10:1057-1063. DOI: 10.1038/ni.1784
  29. 29. Ouyang W, Li MO. Foxo: In command of T lymphocyte homeostasis and tolerance. Trends in Immunology. 2011;32:26-33. DOI: 10.1016/j.it.2010.10.005
  30. 30. Zhang X, Yalcin S, Lee DF, Yeh TY, Lee SM, Su J, Mungamuri SK, Rimmele P, Kennedy M, Sellers R, et al. FOXO1 is an essential regulator of pluripotency in human embryonic stem cells. Nature Cell Biology. 2011;13:1092-1099. DOI: 10.1038/ncb2293
  31. 31. Sandri M. FOXOphagy path to inducing stress resistance and cell survival. Nature Cell Biology. 2012;14:786-788. DOI: 10.1038/ncb2550
  32. 32. Fu Z, Tindall DJ. FOXOs, cancer and regulation of apoptosis. Oncogene. 2008;27:2312‐2319. DOI: 10.1038/onc.2008.24
  33. 33. Hedrick SM, Hess Michelini R, Doedens AL, Goldrath AW, Stone EL. FOXO transcription factors throughout T cell biology. Nature Reviews Immunology. 2012;12:649-661. DOI: 10.1038/nri3278
  34. 34. Leenders H, Whiffield S, Benoist C, Mathis D. Role of the forkhead transcription family member, FKHR, in thymocyte differentiation. European Journal of Immunology. 2000;30:2980-2990. DOI: 10.1002/1521‐4141(200010)30:10<2980::AID‐IMMU2980>3.0.CO;2‐9
  35. 35. Ouyang W, Beckett O, Flavell RA, Li MO. An essential role of the Forkhead‐box transcription factor Foxo1 in control of T cell homeostasis and tolerance. Immunity. 2009;30:358-371. DOI: 10.1016/j.immuni.2009.02.003; S1074‐7613(09)00107‐1 [pii]
  36. 36. Kerdiles YM, Beisner DR, Tinoco R, Dejean AS, Castrillon DH, DePinho RA, Hedrick SM. Foxo1 links homing and survival of naive T cells by regulating L‐selectin, CCR7 and interleukin 7 receptor. Nature Immunology. 2009;10:176-184. DOI: 10.1038/ni.1689
  37. 37. Kerdiles YM, Stone EL, Beisner DR, McGargill MA, Ch’en IL, Stockmann C, Katayama CD, Hedrick SM. Foxo transcription factors control regulatory T cell development and function. Immunity. 2010;33:890-904. DOI: 10.1016/j.immuni.2010.12.002; S1074‐7613(10)00460‐7 [pii]
  38. 38. Fabre S, Lang V, Harriague J, Jobart A, Unterman TG, Trautmann A, Bismuth G. Stable activation of phosphatidylinositol 3‐kinase in the T cell immunological synapse stimulates Akt signaling to FoxO1 nuclear exclusion and cell growth control. Journal of Immunology. 2005;174:4161-4171. DOI: 174/7/4161 [pii].
  39. 39. Lin L, Hron JD, Peng SL. Regulation of NF‐kappaB, Th activation, and autoinflammation by the forkhead transcription factor Foxo3a. Immunity. 2004;21:203-213. DOI: 10.1016/j.immuni.2004.06.016; S1074761304002055 [pii].
  40. 40. Castrillon DH, Miao L, Kollipara R, Horner JW, DePinho RA. Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a. Science. 2003;301:215-218. DOI: 10.1126/science.1086336301/5630/215 [pii].
  41. 41. Hosaka T, Biggs WH 3rd, Tieu D, Boyer AD, Varki NM, Cavenee WK, Arden KC. Disruption of forkhead transcription factor (FOXO) family members in mice reveals their functional diversification. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:2975-2980. DOI: 10.1073/pnas.0400093101
  42. 42. Dengler HS, Baracho GV, Omori SA, Bruckner S, Arden KC, Castrillon DH, DePinho RA, Rickert RC. Distinct functions for the transcription factor Foxo1 at various stages of B cell differentiation. Nature Immunology. 2008;9:1388-1398. DOI: 10.1038/ni.1667
  43. 43. Hinman RM, Nichols WA, Diaz TM, Gallardo TD, Castrillon DH, Satterthwaite AB. Foxo3−/− mice demonstrate reduced numbers of pre‐B and recirculating B cells but normal splenic B cell sub‐population distribution. International Immunology. 2009;21:831-842. DOI: 10.1093/intimm/dxp049
  44. 44. Kops GJ, Dansen TB, Polderman PE, Saarloos I, Wirtz KW, Coffer PJ, Huang TT, Bos JL, Medema RH, Burgering BM. Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature. 2002;419:316-321. DOI: 10.1038/nature01036
  45. 45. Tran H, Brunet A, Grenier JM, Datta SR, Fornace AJ Jr, DiStefano PS, Chiang LW, Greenberg ME. DNA repair pathway stimulated by the forkhead transcription factor FOXO3a through the Gadd45 protein. Science. 2002;296:530-534. DOI: 10.1126/science.1068712
  46. 46. Aksoylar HI, Lampe K, Barnes MJ, Plas DR, Hoebe K. Loss of immunological tolerance in Gimap5‐deficient mice is associated with loss of Foxo in CD4+ T cells. Journal of Immunology. 2012;188:146-154. DOI: 10.4049/jimmunol.1101206
  47. 47. Fabre S, Carrette F, Chen J, Lang V, Semichon M, Denoyelle C, Lazar V, Cagnard N, Dubart‐Kupperschmitt A, Mangeney M, et al. FOXO1 regulates L‐selectin and a network of human T cell homing molecules downstream of phosphatidylinositol 3‐kinase. Journal of Immunology. 2008;181:2980-2989. DOI: 181/5/2980 [pii].
  48. 48. Hu H, Wang B, Borde M, Nardone J, Maika S, Allred L, Tucker PW, Rao A. Foxp1 is an essential transcriptional regulator of B cell development. Nature Immunology. 2006;7:819-826. DOI: 10.1038/ni1358
  49. 49. Feng X, Wang H, Takata H, Day TJ, Willen J, Hu H. Transcription factor Foxp1 exerts essential cell‐intrinsic regulation of the quiescence of naive T cells. Nature Immunology. 2011;12:544-550. DOI: 10.1038/ni.2034
  50. 50. Feng X, Ippolito GC, Tian L, Wiehagen K, Oh S, Sambandam A, Willen J, Bunte RM, Maika SD, Harriss JV, et al. Foxp1 is an essential transcriptional regulator for the generation of quiescent naive T cells during thymocyte development. Blood. 2010;115:510-518. DOI: 10.1182/blood‐2009‐07‐232694
  51. 51. Park JH, Yu Q, Erman B, Appelbaum JS, Montoya‐Durango D, Grimes HL, Singer A. Suppression of IL7Ralpha transcription by IL‐7 and other prosurvival cytokines: A novel mechanism for maximizing IL‐7‐dependent T cell survival. Immunity. 2004;21:289-302. DOI: 10.1016/j.immuni.2004.07.016; S1074761304002031 [pii].
  52. 52. Tzachanis D, Freeman GJ, Hirano N, van Puijenbroek AA, Delfs MW, Berezovskaya A, Nadler LM, Boussiotis VA. Tob is a negative regulator of activation that is expressed in anergic and quiescent T cells. Nature Immunology. 2001;2:1174-1182. DOI: 10.1038/ni730
  53. 53. Park Y, Jin HS, Lopez J, Elly C, Kim G, Murai M, Kronenberg M, Liu YC. TSC1 regulates the balance between effector and regulatory T cells. Journal of Clinical Investigation. 2013;123:5165-5178. DOI: 10.1172/JCI69751
  54. 54. Shrestha S, Yang K, Wei J, Karmaus PW, Neale G, Chi H. Tsc1 promotes the differentiation of memory CD8+ T cells via orchestrating the transcriptional and metabolic programs. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:14858-14863. DOI: 10.1073/pnas.1404264111
  55. 55. Xie DL, Wu J, Lou YL, Zhong XP. Tumor suppressor TSC1 is critical for T‐cell anergy. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:14152-14157. DOI: 10.1073/pnas.1119744109
  56. 56. Yang K, Neale G, Green DR, He W, Chi H. The tumor suppressor Tsc1 enforces quiescence of naive T cells to promote immune homeostasis and function. Nature Immunology. 2011;12:888-897. DOI: 10.1038/ni.2068
  57. 57. Schwarz DA, Katayama CD, Hedrick SM. Schlafen, a new family of growth regulatory genes that affect thymocyte development. Immunity. 1998;9:657-668
  58. 58. Condamine T, Le Luduec JB, Chiffoleau E, Beriou G, Louvet C, Heslan M, Tilly G, Cuturi MC. Characterization of Schlafen‐3 expression in effector and regulatory T cells. Journal of Leukocyte Biology. 2010;87:451-456. DOI: 10.1189/jlb.0609410
  59. 59. Berger M, Krebs P, Crozat K, Li X, Croker BA, Siggs OM, Popkin D, Du X, Lawson BR, Theofilopoulos AN, et al. An Slfn2 mutation causes lymphoid and myeloid immunodeficiency due to loss of immune cell quiescence. Nature Immunology. 2010;11:335-343. DOI: 10.1038/ni.1847
  60. 60. Omar I, Lapenna A, Cohen‐Daniel L, Tirosh B, Berger M. Schlafen2 mutation unravels a role for chronic ER stress in the loss of T cell quiescence. Oncotarget. 2016;7:39396-39407. DOI: 10.18632/oncotarget.9818
  61. 61. Goldshtein A, Zerbib SM, Omar I, Cohen‐Daniel L, Popkin D, Berger M. Loss of T‐cell quiescence by targeting Slfn2 prevents the development and progression of T‐ALL. Oncotarget. 2016;7:46835-46847. DOI: 10.18632/oncotarget.9390
  62. 62. Wong WF, Kohu K, Chiba T, Sato T, Satake M. Interplay of transcription factors in T‐cell differentiation and function: The role of Runx. Immunology. 2011;132:157-164. DOI: 10.1111/j.1365‐2567.2010.03381.x
  63. 63. Komori T. Regulation of osteoblast differentiation by Runx2. Advances in Experimental Medicine and Biology. 2010;658:43-49. DOI: 10.1007/978‐1‐4419‐1050‐9_5
  64. 64. Okuda T, van Deursen J, Hiebert SW, Grosveld G, Downing JR. AML1, the target of multiple chromosomal translocations in human leukemia, is essential for normal fetal liver hematopoiesis. Cell. 1996;84:321-330
  65. 65. Wong WF, Kohu K, Nakamura A, Ebina M, Kikuchi T, Tazawa R, Tanaka K, Kon S, Funaki T, Sugahara‐Tobinai A, et al. Runx1 deficiency in CD4+ T cells causes fatal autoimmune inflammatory lung disease due to spontaneous hyperactivation of cells. Journal of Immunology. 2012;188:5408-5420. DOI: 10.4049/jimmunol.1102991
  66. 66. Wong WF, Kohu K, Nagashima T, Funayama R, Matsumoto M, Movahed E, Tan GM, Yeow TC, Looi CY, Kurokawa M, et al. The artificial loss of Runx1 reduces the expression of quiescence‐associated transcription factors in CD4(+) T lymphocytes. Molecular Immunology. 2015;68:223-233. DOI: 10.1016/j.molimm.2015.08.012
  67. 67. Wong WF, Kurokawa M, Satake M, Kohu K. Down‐regulation of Runx1 expression by TCR signal involves an autoregulatory mechanism and contributes to IL‐2 production. Journal of Biological Chemistry. 2011;286:11110-11118. DOI: 10.1074/jbc.M110.166694
  68. 68. Chang M, Jin W, Sun SC. Peli1 facilitates TRIF‐dependent Toll‐like receptor signaling and proinflammatory cytokine production. Nature Immunology. 2009;10:1089-1095. DOI: 10.1038/ni.1777
  69. 69. Chen ZJ, Bhoj V, Seth RB. Ubiquitin, TAK1 and IKK: Is there a connection? Cell Death & Differentiation. 2006;13:687-692. DOI: 10.1038/sj.cdd.4401869
  70. 70. Schauvliege R, Janssens S, Beyaert R. Pellino proteins: Novel players in TLR and IL‐1R signalling. Journal of Cellular and Molecular Medicine. 2007;11:453-461. DOI: 10.1111/j.1582‐4934.2007.00040.x

Written By

Ahmed Fadhil Neama, Chung Yeng Looi and Won Fen Wong

Submitted: 13 March 2017 Reviewed: 29 March 2017 Published: 12 July 2017