activación de cels t

Upload: alejandra-nunez

Post on 02-Apr-2018

225 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/27/2019 Activacin de cels T

    1/31

  • 7/27/2019 Activacin de cels T

    2/31

    TCR: T cell antigenreceptor

    Signal transduction:biochemical eventslinking surface

    receptor engagementto cellular responses

    INTRODUCTION

    Twenty-five years ago, Annual Review of Im-munology published its first review describing

    features of the structure we now know as theT cell antigen receptor (TCR) (1). In recog-

    nition of this anniversary, this article begins byhighlighting a sampling of seminal observations

    made in thedecadefollowing theinitialdescrip-tion of the TCR. The discoveries made dur-

    ing this period established the basic paradigmfor how TCR engagement initiates the earli-

    est biochemical events leading to cellular ac-tivation, described nicely in an Annual Reviewof Immunology article in 1996 (2) (Figure 1a).

    This historical perspective sets the stage for adiscussion of our current state of understanding

    of the molecular and biochemical events criti-cal for T cell activation that have emerged from

    the work of multiple laboratories.

    DESCRIBING THETCR COMPLEX

    In the early 1980s, several groups began ex-periments to identify and characterize the anti-

    gen receptor on T cells. One approach usednewly developed molecular techniques, ulti-

    mately leading to the identification of the genesresponsible for the antigen-binding proteins

    (35). An even earlier approach relied on im-

    Figure 1

    TCR proximal signaling then and now: filling in the gaps. Then (a): TCR signaling mid-1990s (adapted from Reference 2). Ligationthe TCR/CD3 results in activation of Src and Syk family PTKs associated with the intracellular CD3 domains that then activatePLC1 and Ras-dependent pathways. PLC1 hydrolizes PIP2 to form IP3 and DAG. Now (bd): Current understanding of how thTCR couples to downstream pathways (b), the molecular basis for Ca2+ influx (c), and the positive feedback loop responsible for Rasactivation (d). (b) The link between PTKs and downstream pathways is a multimolecular signaling complex nucleated by the adapterproteins LAT, Gads, and SLP-76. Lck activates ZAP-70 to phosphorylate (p) tyrosine residues on LAT, which then recruits Gads anits constitutive binding partner SLP-76. ZAP-70-mediated phosphorylation of SLP-76 results in the recruitment of multiple SH2domaincontaining effector molecules (circles) and adapter proteins (octogons). SH3 domains (shaded overlapping areas) also link effecto

    to adapters and contribute to stabilization of the complex. (c) The link between depletion of ER Ca2+

    stores and CRAC channelactivation is STIM. TCR-induced IP3 production (see b) results in the activation of IP3 receptors (IP3R), which release Ca

    2+ from tER into the cytoplasm. STIM contains paired EF hands within the ER lumen, which bind a single Ca 2+ molecule. Upon depletion ER stores, Ca2+-free STIM molecules oligomerize and move to areas of ER/plasma membrane proximity, where they colocalize witand induce dimerization of Orai1 dimers, resulting in a functional CRAC channel and subsequent influx of Ca 2+. (d) Activation of Rinvolves two Ras GEFs (SOS and RasGRP) in a positive feedback loop. TCR-induced production of DAG (see b) results in themembrane recruitment of RasGRP, where it is phosphorylated and activated by PKC. RasGRP then facilitates the removal of GDPfrom Ras, which can then bind GTP and become activated. GTP-bound Ras then binds SOS, which is bound constitutively to GRBand is inducibly recruited to LAT, increasing its GEF activity, resulting in a positive feedback loop and robust Ras activity.

    munization of mice with T cell clones of a

    fined specificity, hybridomas, or clonal T ctumors in the hope that antibodies would

    generated that would react with the recepresponsible for binding to antigen (68). Su

    antibodies were produced and were then ufirst to demonstrate interference with antige

    responses and later to perform the initial bchemical characterization of the receptorself. These studies revealed a complicated c

    surface structure that included proteins retive with antibodies against the nonpolym

    phic CD3 proteins (initially thought of as thpolypeptides, , , and ) (9), as well as varia

    proteins designated and .The antigen-binding function of w

    obvious early on, by inference owing to thhighly polymorphic nature and similarity

    immunoglobulin and experimentally owingearly gene transfer experiments that demo

    strated that antigen/major histocompatibi

    complex (MHC) reactivity tracked with exprsion of these receptor components (10, 1

    However, it was less obvious what role the Cproteins played in TCR function. Several li

    of evidence suggested that the CD3 molecuwere critical for signal transduction: Unlikethe CD3 molecules had long cytoplasmic taand anti-CD3 antibodies resulted in T cell

    tivation, although it was difficult to demostratesignaling function conclusively. One ea

    592 Smith-Garvin Koretzky Jordan

  • 7/27/2019 Activacin de cels T

    3/31

    www.annualreviews.org T Cell Activation 593

  • 7/27/2019 Activacin de cels T

    4/31

    PKC: protein kinaseC

    PLC: phospholipaseC

    PTK: protein

    tyrosine kinaseImmunoreceptortyrosine-basedactivation motif(ITAM): a shortpeptide sequence inthe cytoplasmic tails ofkey surface receptorson hematopoietic cellsthat is characterized bytyrosine residues thatare phosphorylated bySrc family PTKs,

    enabling the ITAM torecruit activated Sykfamily kinases

    effort involved an attempt to create cell lines

    expressing without CD3 or CD3 without

    , thereby creating reagents to investigate the

    independent role of the receptor components.These experiments were not successful, as it

    became clear that there is an obligatory coex-pression of with CD3 (12). These studies

    did result, however, in the creation of the firstin a long line of genetically altered Jurkat Tcells that have been instrumental in our under-

    standingof how the TCR complex couplesto itssignaling machinery (13).

    EARLY SIGNALING STUDIES

    Initial investigations into how the TCR trans-

    duces its signals began with the observation thatTCR-deficient Jurkat T cells could be stimu-

    lated pharmacologically with the combinationof phorbol esters and Ca2+ ionophores (14).This observation led to speculation that en-

    gagement of the TCR might stimulate the samesignals that these reagents induced. This no-

    tion was tested directly in experiments demon-strating increases in intracellular free Ca2+

    following CD3 or TCR stimulation in bothJurkat cells and primary T cells. The source

    of the Ca2+ increase was shown to be a com-bination of Ca2+ released from an intracellular

    pool in response to inositol trisphosphate (IP3)production and influx of Ca2+ from outside of

    the cell (15). It was presumed that phorbol es-

    ters were important because of their ability toactivate protein kinase C (PKC, now known to

    be a family of enzymes), whose activity is regu-lated physiologically by diacylglycerol (DAG).

    Concurrent studies in other systems haddemonstrated that a single enzymatic reaction,

    hydrolysis of the membrane PI(4,5)P2 (phos-pholipid phosphatidylinositol 4,5 bisphos-

    phate) by phospholipase C (PLC), generatesboth IP3 and DAG, suggesting that the TCR

    may function to regulate PLC activity.Testing this notion proved somewhat

    difficult at first, as PLC regulation was initially

    described downstream of heterotrimericguanosine triphosphate (GTP)-binding pro-

    teins associated with seven-transmembrane

    domain receptors. An exhaustive but uns

    cessful search ensued for the GTP-bindprotein critical for coupling the TCR

    PLC activation. Insight into how the TCmay initiate PLC activation emerged from

    confluence of discoveries in immune cells aother lineages. For example, stimulation of

    TCR results in changes in protein phosphrylation, including inducible phosphorylatof the newly described chain of the C

    complex (16). Furthermore, growth facreceptors with intrinsic protein tyrosine kin

    (PTK) activity also stimulate PLC functiIn contrast to seven-transmembrane dom

    receptors, however, receptor PTKs stimulanother PLC isoform, PLC. Although no

    of the TCR components possessed enzymaactivity themselves, cytosolic PTKs of

    Src family (in particular Lck and Fyn) wbeing described in T cells. These PTKs w

    associated either with the TCR (17) or w

    the CD4 and CD8 coreceptors (18, 1both important for TCR signaling. Th

    observations, coupled with the new knowledthat T cell activation requires PTK funct

    (20), led to a new TCR signaling paradigin which the TCR recruits cytosolic PTKs

    activate key second messengers.

    HOW CD3 TRANSDUCESITS SIGNALS

    Testing this model led to a search for adtional substrates of the TCR-stimulated PTK

    Among the most attractive candidates were rosines within the CD3 molecules themsel

    that fell within a motif present once withCD3 , , and and in triplicate within eac

    chainandalsoinkeyimmunoreceptorsonothimmune cell lineages. The signature

    these motifs (21), eventually designated imunoreceptor tyrosine-based activation mo

    (ITAMs), is two tyrosines flanking a seriesamino acids, including key leucine/isoleucin

    with stereotypic spacing. Numerous labo

    tories demonstrated that the ITAM tyrosinare in fact phosphorylated upon TCR l

    ation, but defining the importance of t

    594 Smith-Garvin Koretzky Jordan

  • 7/27/2019 Activacin de cels T

    5/31

    posttranslational modification for TCR func-

    tion was not trivial. Because the CD3 moleculescould not be expressed without the chains,

    several groups created chimeric molecules fus-ing the cytoplasmic domains of individual CD3

    chains to extracellular and transmembrane do-mains from other proteins (2225). These

    cDNAs in chimeric proteins were then trans-fected into T cell lines selected for TCR loss.Antibody crosslinking of the extracellular do-

    main of the chimeras recapitulated all theknown TCR-mediated signaling events lead-

    ing to cellular activation. Mutation of the keyITAM tyrosines (or altering their spacing) abro-

    gated the ability of the chimeras to activate thecells, thus demonstrating that tyrosine phos-

    phorylation of the CD3 ITAMs is an earlyand requisite step for TCR-mediated T cell

    activation.Studies of the CD3 chimeras led naturally to

    the question of the purpose for ITAM phospho-

    rylation. Unlike growth factor receptors whoseintrinsic enzymatic activity is enhanced by tyro-

    sine phosphorylation, the CD3 molecules haveno such effector function on their own. Investi-

    gators speculated, therefore, that tyrosine phos-phorylation of the ITAMs might serve as dock-

    ing sites for interactions with other proteins.Indeed, it was soon shown that phosphory-

    lated CD3

    (and later other ITAM-containingproteins) is a recruitment site of a 70-kDa

    phosphoprotein, the Syk kinase family memberZAP-70 (-associated protein of 70 kDa) PTK

    (26). A model therefore emerged that engage-

    ment of the TCR leads to Src family PTK ac-tivity resulting in ITAM phosphorylation and

    recruitment of ZAP-70. This converted theTCR with no intrinsic enzymatic function to an

    active PTK able to phosphorylate a spectrumof substrates leading to a myriad of down-

    stream signals that, when integrated appropri-ately (along with signals from other corecep-

    tors), lead to T cell activation (27).The basic tenets of this model have stood the

    test of intensive investigation. In the 15 yearssince ZAP-70 was cloned, investigators have

    filled in many of the gaps between the TCR

    and initiation of effector functions. Much has

    ARGUING BY ANALOGY: COMPARINGAND CONTRASTING SIGNALING PATHWAYSBY MULTIPLE RECEPTORS AND LINEAGES

    Key insights important for understanding T cell activation havecome from studies in nonimmune mammalian cells and cells of

    lower organisms, including the observation that PTKs could linkto the phosphatidylinositol pathway and the paradigm describing

    adapter proteins as integrators of signal transduction. T cell biol-ogists have also provided unique insights to their nonimmunol-

    ogist colleagues. Examples include a mechanism for recruitment

    of nonreceptor PTKs to enzymatically inactive surface receptorsand the notion that protein tyrosine phosphatases may be positive

    as well as negative regulators of PTK pathways.Within the immune system, biologists studying TCR signal-

    ing have been the donors and recipients of information that hasbeen useful for investigators examining the signaling pathways of

    other cell surface receptors (e.g., integrins) and other hematopoi-etic lineages. It is clear that, although basic paradigms may be

    similar, each cell type and receptor utilizes unique ways to regu-late signal transduction cascades. Additionally, recent studies have

    demonstrated that pathways thought to be distinct (e.g., integrinsand immunoreceptors) instead intersect at multiple levels. Fu-

    ture insights into how diverse signaling pathways are integrated

    to result in the appropriate biologic response will undoubtedlycontinue to benefit from comparing and contrasting activation

    events downstream of multiple receptors in different immuneand nonimmune cell lineages.

    Adapter protein:cellular protein that

    functions to bridgemolecular interactiovia characteristicdomains able tomediateprotein/protein orprotein/lipidinteractions

    been learned about substrates of the PTKs (in-

    cluding Src, Syk, and more recently Tec fam-ily members) activated by the TCR and how

    these molecules participate in T cell activation,about how signaling complexes are organized

    by adapter proteins to bring effector proteinstogether, and about the unexpected intersec-

    tion of particular signaling pathways (see alsothe side bar, Arguing by Analogy: Comparing

    and Contrasting Signaling Pathways by Mul-tiple Receptors and Lineages). With the ac-

    cumulation of data, it has also become clearthat signaling via the TCR complex is not a

    linear event starting at the receptor and end-

    ing in the nucleus. Instead, there appears to becomplex feedback and feedforward regulation

    at each step. Ironically, one of the most central

    www.annualreviews.org T Cell Activation 595

  • 7/27/2019 Activacin de cels T

    6/31

  • 7/27/2019 Activacin de cels T

    7/31

    Table 1 Recent models for initial TCR triggering

    Model Description

    Piston-like movement An external force from the APC/T cell interaction pushes the TCR/CD3 complex in a piston-like

    fashion perpendicular to the plasma membrane, exposing activating motifs on the intracellular region

    of CD3 chains

    Receptor deformation The force of the motility of the T cell as it moves over an APC induces TCR triggering only when

    pMHC/TCR interactions are of a high enough affinity to withstand the force long enough to underg

    a conformational changePermissive geometry pMHC dimers bind TCR/CD3 dimers inducing a rotational scissor-like conformational change in CD

    chain(s) that reveal previously hidden intracellular activation motif(s)

    Kinetic segregation Signal initiation occurs because of exclusion of inhibitory molecules in the tight contact zone between

    the T cell and the APC, thereby shifting the enzymatic steady state toward an activating state

    Diffusion trapping Expanded kinetic segregation model in which the affinity/diffusion coefficient of the pMHC/TCR

    dictates the valency of TCRs required to trap the complex in the tight contact zone and therefore

    initiate a productive signal

    Pseudodimer Agonist pMHC/TCR recruits a second TCR via interaction with its associated CD4, the second TCR

    then binds a coagonist endogenous pMHC forming a stable pseudodimer that triggers signaling via

    proximity of ITAMs to CD4-associated Lck and/or by conformational change

    on ligand-induced conformational changes in

    the TCR and/or TCR aggregation have beenproposed (Table 1). The crystal structures

    of several pMHC/TCR complexes have re-vealed little conformational changes in the

    TCR heterodimer outside of the binding do-main (reviewed in 36). However, owing to the

    current technical limitations of crystallogra-phy and protein nuclear magnetic resonance

    spectroscopy, only pMHC/TCR structures in

    the absence of transmembrane domains andCD3 components have beenelucidated. There-

    fore, changes between the components of theTCR/CD3 complex in relation to one another

    or to the plasma membrane cannot be ruled out.The rigid, rod-like transmembrane domain of

    the CD3 could mediate a piston-like move-ment of the TCR complex perpendicular to

    the plasma membrane after ligand binding, ina manner similar to that reported for the as-

    partate receptor (37). Such a model would re-quire external forces that could be provided by

    cell-to-cell contact, but this possibility is dif-

    ficult to reconcile with the ability of solubleligands to activate the TCR. Similarly, an ex-

    ternal force is required for the recently pro-posed receptor deformation model, in which

    the movement of the T cell as it travels acrossthe antigen-presenting cell (APC) exerts a force

    on transient pMHC/TCR interactions. Only

    when the force required to induce conforma-tional changes in the TCR is less than the force

    required to break the pMHC/TCR interactionwill TCR triggering occur (38). Alternatively,

    the permissive geometry model for TCR trig-gering predicts that dimeric pMHCs bind two

    TCR/CD3 complexes (39). This interaction re-sults in a rotation of the TCR heterodimers

    around one another, displacing the extracellulardomains of the associated CD3 molecules. The

    transmembrane interactions between the CD3

    and TCR dimers serve as pivot points, and theCD3 movement is scissor-like, exposing previ-

    ouslyshielded ITAMs and/or other functionallyimportant domains. This model is consistent

    with studies that suggest that TCR aggregationis required for TCR triggering.

    TCR aggregation has long been impli-cated as a mechanism for TCR triggering. In

    the classic aggregation model, crosslinking ofTCR/CD3 complexes with multimeric pMHC

    enables close contact and transphosphorylationbetween the CD3 tails and associated PTKs.

    Clustering of several TCR complexes also may

    result in competition for membrane lipid be-tween the CD3 chains, resulting in dissocia-

    tion of the cytoplasmic domains from the mem-brane and subsequent ITAM phosphorylation

    www.annualreviews.org T Cell Activation 597

  • 7/27/2019 Activacin de cels T

    8/31

    PI3K:phosphoinositide3-kinase

    (28). Aggregation is supported strongly by

    observations that soluble multimeric but notmonomeric pMHC can trigger TCR activa-

    tion. However, recent biochemical and micro-scopic studies suggest that preformed TCR

    aggregates are present on nonactivated T cells(reviewed in 40). In the kinetic segregation

    model, adhesion and other accessory moleculeswith short extracellular domains such as CD2initiate close contact zones between an APC

    and a T cell (41). Inhibitory phosphatases withlong extracellular domains such as CD45 are

    excluded because of their size. TCR complexesthat engage pMHC on the APC surface remain

    in the close contact zone, where they are seg-regated from phosphatases and are able to ini-

    tiate signaling. TCR complexes that do not en-gage pMHC are free to diffuse outside of the

    close contact zone. Shortening the extracellulardomains of inhibitory molecules or lengthen-

    ing the extracellular domains of adhesion or ac-

    cessory molecules can abrogate TCR signaling,suggesting that kinetic segregation is an impor-

    tant aspect of TCR signal initiation (reviewedin 41). This notion was expanded with the dif-

    fusion trapping model in which immobilizationor trapping of pMHC/TCRs in the close con-

    tact zones is responsible for TCR signal initi-ation (42). The valency, or degree of aggrega-

    tion, of the pMHC/TCR required for trappingand therefore triggering depends on the affin-

    ity anddiffusion coefficient of thepMHC/TCRinteraction. Another model has been proposed

    that suggests that endogenous pMHCs amplify

    signals produced by the rare agonist pMHCsby promoting TCR aggregation and a subse-

    quent phosphorylation cascade (40). Similarly,the pseudodimer model proposes that an ago-

    nistpMHC/TCRcanrecruitasecondTCRinaCD4-dependent manner, and this second TCR

    binds a coagonist endogenous pMHC, forminga stable pseudodimer that could trigger signal-

    ing through ITAM/Lck proximity or throughCD3 conformational changes (43). Many of

    these models are not mutually exclusive, and itis likely that TCR aggregation, conformational

    changes within the TCR complex, and exclu-

    sion of inhibitory molecules are all required

    for TCR triggering, perhaps in a stepw

    fashion.

    PROXIMAL SIGNALINGCOMPLEX

    The earliest step in intracellular signaling f

    lowing TCR ligation is the activation of S(Lck and Fyn) PTKs, leading to phosphorytion of the CD3 ITAMs. Recruitment of ZA

    70 follows, leading to a cascade of phosphrylation events. The past decade has seen t

    description of a subcellular assembly and acvation of an adapter protein nucleated mu

    molecular signaling complex (Figure 1b). Tcomplex is responsible for propagating

    TCR/PTK signal into multiple and diverse dtal signaling pathways.

    Among the most important of the ZAPtargets are the transmembrane adapter protlinker for the activation of T cells (LAT) a

    the cytosolic adapter protein Src homolog(SH2) domaincontaining leukocyte phosph

    protein of 76 kDa (SLP-76) (44, 45). Thtwo adapters form the backbone of the co

    plex that organizes effector molecules in tcorrect spatiotemporal manner to allow for

    activation of multiple signaling pathways. Timportance of these adapters is underscored

    studies showing that the loss of either LATSLP-76 results in a near complete loss of TC

    signal transduction reminiscent of Syk/ZAPor Lck/Fyn double-deficient T cells (4648)

    LAT contains nine tyrosines that are ph

    phorylatedupon TCRengagement, which bthe C-terminal SH2domainof PLC1,thep

    subunit of phosphoinositide 3-kinase (PI3and the adapters growth factor receptor-bou

    protein 2 (GRB2) and GRB2-related adapdownstream of Shc (Gads) (reviewed in 4

    SLP-76 is then recruited to phosphorylaLAT via their mutual binding partner Ga

    (49). SLP-76 itself contains three modudomains: an N-terminal acidic domain w

    three phosphorylatable tyrosines that interwith the SH2 domains of Vav1, Nck, and I

    2-induced tyrosine kinase (Itk); a PRR t

    binds constitutively Gads and PLC1; an

    598 Smith-Garvin Koretzky Jordan

  • 7/27/2019 Activacin de cels T

    9/31

    C-terminal SH2 region that can bind adhesion

    and degranulationpromoting adapter protein(ADAP) and hematopoietic progenitor kinase

    1 (HPK1) (reviewed in 46). Although LAT andSLP-76 serve to nucleate this large signaling

    complex, the effector molecules themselvesalsoare important for stabilizing the complex. For

    example, the Tec family kinase Itk is requiredin a kinase-independent manner for the re-cruitment of the guanine nucleotide exchange

    factor (GEF) Vav1 to the APC contact site,whereas Vav1 is required for optimal SLP-76

    phosphorylation and recruitment to LAT aswell as for Itk activation (5052). These and

    other data suggest that the formation of thecomplex is more complicated than the linear

    modelmostofteninvokedforsimplicity.Forex-ample, PLC1 directly binds to SLP-76, LAT,

    and Vav1 as well as to its activating kinaseItk (reviewed in 53). It is thought that these

    interactions collectively are required to stabi-

    lize PLC1 in the correct conformation withinthe complex to allow for its optimal activity

    (54). Advances in biochemical and structuraltechniques are needed to elucidate the precise

    allosteric and perhaps stoichiometric changeswithin the multimolecular complex that allow

    for signal transduction.To investigate more precisely the impor-

    tance of these complex interactions in primaryT cells, several laboratories have generated

    mice expressing transgenic or knockin muta-tions in specific binding regions in various

    molecules involved in proximal signaling (55

    57). Tyrosine to phenylalanine mutations inSLP-76 at residues 112 and 128 or 145 in pri-

    mary thymocytes and T cells do not result ina loss of SLP-76/Vav1/Nck/Itk interactions, as

    would be expected from earlier phosphopeptidemapping studies and studies in cell lines (57).

    However, these tyrosine mutations still resultin severe defects in downstream signaling path-

    ways consistent with defective Vav1 or Itk activ-ity. Similarly, mutation of tyrosines of Vav1 does

    not result in a loss of interaction with their pro-posed binding partners, although it does result

    in dysregulated Vav1-dependent signaling (55).

    Although the continued interactions of these

    proteins seen by immunoprecipitation exper-

    iments are likely due to tertiary interactionswith other domains or other molecules, these

    studies suggest that SH2/phosphotyrosine in-teractions may play important regulatory roles

    for the activation of effector molecules. Indeed,structural studies have suggested that the in-

    teraction between the SH2 domain of Itk anda phosphotyrosine results in a conformational

    switch allowing kinase activity (58). Consistent

    with these data, investigators recently showedin Jurkat T cell lines that an Itk/SLP-76 in-

    teraction is required for Itk kinase activity, al-though we do not yet know if it is specifically

    the SH2/phosphotyrosine interaction that me-diates this kinase activity (59). Further stud-

    ies are required to determine how the activ-ities of molecules beyond Itk are affected by

    specific domain/domain interactions within thecomplex.

    The proximal signaling complex resultsin the activation of PLC1-dependent path-

    ways including Ca2+- and DAG-induced

    responses, cytoskeletal rearrangements, andintegrin activation pathways. Ligation of co-

    stimulatory receptors such as CD28 augmentsthese pathways. Below, we discuss the mecha-

    nismsbywhichthesepathwaysareactivatedandregulated.

    PLC1 ACTIVATIONAND SIGNAL TRANSDUCTION

    Following TCR ligation, PLC1 is found in

    the proximal signaling complex bound to SLP-76, Vav1, and LAT, where it is phosphorylated

    and activated by Itk. Activated PLC1 then hy-drolyzes the membrane lipidPI(4,5)P2, produc-

    ingthesecondmessengersIP3 andDAG. Thesetwomessengers areessential for T cell function,

    and therefore the regulation of PLC1 acti-vation has been the subject of intensive stud-

    ies. Localization of PLC1 to the proximalsignaling complex is dependent on LAT and

    the Gads-binding region of SLP-76 (54). Ac-

    tivation of PLC1 is dependent on Itk kinaseactivity that, in turn, is dependent on Vav1,

    Lck, ZAP-70, LAT, and SLP-76 (52, 60, 61).

    www.annualreviews.org T Cell Activation 599

  • 7/27/2019 Activacin de cels T

    10/31

    Following TCR ligation, Itk is recruited to the

    membrane through PH domain interactionswith PIP3, which has been locally generated

    by Lck-induced PI3K activity (reviewed in 61).At the membrane, Lck phosphorylates Itk, and

    the SH2 and SH3 domains of Itk interact withphosphorylated tyrosine 145 and the PRR of

    SLP-76, respectively (6264). The role of Vav1and ZAP-70 in Itk activation is not understoodbut may relate to theirinvolvement in the phos-

    phorylation of SLP-76 or PI3K activation (45,51,52). A secondTec family kinase, Rlk, canalso

    phosphorylate PLC1, resulting in a relativelymild defect in Itk-deficient mice and requiring

    the study of Rlk/Itk double-deficient mice tobetter understand the role of Tec kinases in T

    cell activation (reviewed in 61).

    DAG-MEDIATED SIGNALINGPATHWAYS

    TCR-induced production of DAG results inthe activation of two major pathways involving

    Ras and PKC. Ras is a guanine nucleotidebinding protein and is required for the acti-

    vation of the serine-threonine kinase Raf-1,which initiates a mitogen-associated protein

    kinase (MAPK) phosphorylation and activa-tion cascade. Raf-1 is a MAPK kinase kinase

    (MAPKKK) that phosphorylates and activatesMAPK kinases (MAPKKs), which in turn phos-

    phorylate and activate the MAPKs extracellular

    signal-regulated kinase 1 (Erk1) and Erk2. Erkkinase activity results in the activation of the

    transcription factor Elk1, which contributes tothe activation of the activator protein-1 (AP-1)

    ( Jun/Fos) transcription complex via regulationof Fos expression. Additionally, Erk activity can

    result in the transcriptional activation of sig-nal transducer and activator of transcription 3

    (STAT3) and in the serine phosphorylation ofLck (reviewed in 65).

    Ras is only active in the GTP-boundstate, and its activation is facilitated by GEFs

    and is suppressed by GTPase-activating pro-

    teins (GAPs). Two Ras GEFs are presentin T cells, son of sevenless (SOS) and Ras

    guanyl nucleotide-releasing protein (RasGRP).

    RasGRP appears more dominant for early

    tivation of Ras, as SOS cannot compensfor RasGRP deficiency (6668). RasGRP is

    ducibly recruited to the membrane througDAG-binding domain (69), where it is ph

    phorylated by PKC (70). SOS is constitively bound to the adapter protein GR

    and upon TCR stimulation, the GRB2 SHdomain is recruited to and binds phosph

    rylated tyrosines on LAT, thereby bring

    SOS into the proximal signaling compwhere it can facilitate the localized acti

    tion of Ras (71). The significance of thtwo modes of Ras activation was unclear u

    til recently, when it was shown that RasGRdependent RasGTP production catalyzes S

    activity, resulting in a positive feedback loand robust TCR-induced Ras activation (

    (Figure 1c). This unified model explains tRasGRP dominance, as RasGRP would be

    quired to initiate the production of RasGTwhich upon reaching threshold levels wo

    catalyze SOS activity and further amplify

    signal.The second major signaling pathway re

    lated by DAG is mediated by PKC, a PKfamily member that contains a lipid-bind

    domain specific for DAG, which is importfor recruiting PKC to the plasma membra

    following T cell activation. However, phosphrylation of PKC by Lck (reviewed in 73) m

    be required to induce a conformational chanthat enables binding to the lipid phosphati

    serine (PS), which in turn enhances bindingDAG, resulting in PKC activation (74). Ot

    proximal signaling molecules, including Va

    PI3K, and 3-phosphoinositide-dependent nase 1 (PDK1), also play roles in PKC loc

    ization, butdetails of their contributions arencompletely defined (73).

    One critical pathway that PKC regulais NF-B activation. Because both the cano

    cal and noncanonical activation of this pathwin T cells is described in detail in this volu

    ofAnnual Review of Immunology [see reviewKarin and colleagues (75)], we highlight o

    the major steps in the classical activationNF-B downstream of the TCR. The NF-

    600 Smith-Garvin Koretzky Jordan

  • 7/27/2019 Activacin de cels T

    11/31

    family of transcription factors consists of five

    members. In resting cells, NF-B is found inthe cytosol associated with inhibitor of NF-B

    (IB) family members that keep NF-B frommoving into the nucleus. Upon T cell activa-

    tion, IB is phosphorylated by the IB kinase(IKK) complex, ubiquitinylated, and degraded,

    allowing NF-B to translocate into the nucleus,where it activates genes involved in the func-tion, survival, and homeostasis of T cells (re-

    viewed in 76). Although we have known thegeneral pathway of NF-B activation for some

    time, the specifics of how PKC activation leadsto nuclear import of NF-B in T cells are still

    being elucidated.Over the past several years, the identifi-

    cation and characterization of a lymphocyte-specific activation complex have provided some

    insight into this question. Following TCRstimulation, a trimolecular complex forms be-

    tween CARMA1 [caspase recruitment domain

    (CARD) and membrane-associated guanylatekinase (MAGUK)-containing scaffold pro-

    tein], the CARD-containing adapter proteinBcl10, and mucosa-associated lymphoid tis-

    sue lymphoma translocation gene 1 (MALT1)(73, 76). The assembly of this CBM com-

    plex (CARMA1/Bcl10/MALT1) is regulatedby PKC through its phosphorylation of

    CARMA1, which is required for CARMA1oligomerization and association with Bcl10 (77,

    78). MALT1 binds to Bcl10 and contributes

    to the degradation of the regulatory subunitof the IKK complex (IKK) by facilitating its

    polyubiquitination, possibly via activation ofthe E3 ubiquitin ligase tumor necrosis factor

    receptor-associated factor 6 (TRAF6) (79, 80).Degradation of this regulatory subunit allows

    for phosphorylation of IB by the IKK cat-alytic subunits, subsequent IB degradation,

    and release of NF-B, resulting in NF-B nu-clear localization and gene activation. Recently,

    MALT1 was shown to enhance NF-B signal-ing through its ability to degrade the deubiq-

    uitinating enzyme A20, a negative regulator of

    NF-B activation (81, 82).One additional component of the complex

    that was recently identified as a CARMA1-

    associating protein is ADAP (83). ADAP,

    originally defined as a SLP-76-binding part-ner, associates with the MAGUK domain of

    CARMA1 following T cell stimulation. Inves-tigators proposed that this interaction might al-

    ter the conformation of CARMA1 and enhanceits association with Bcl10 and MALT1 and/or

    stabilize the CBM complex.

    Ca2+-MEDIATED SIGNALINGPATHWAYS

    Ca2+ ions are universal second messengers ineukaryotic cells. The IP3 generated by TCR-

    stimulated PLC1 activity stimulates Ca2+-permeable ion channel receptors (IP3R) on the

    endoplasmic reticulum (ER) membrane, lead-ing to the release of ER Ca2+ stores into

    the cytoplasm. Depletion of ER Ca

    2+

    trig-gers a sustained influx of extracellular Ca2+

    through the activation of plasma membrane

    Ca2+ release-activated Ca2+ (CRAC) channelsin a process known as store-operated Ca2+ en-

    try (SOCE) (reviewed in 84) (Figure 1d). Fordecades the CRAC channels had only been

    identified by their biophysical properties, andit has just been in the past few years that

    the pore-forming subunit of the channels wasidentified as the four-transmembrane domain

    containing molecule Orai1 (8587). Addition-ally, studies in the past few years have revealed

    the sensor for depleted ER Ca2+ stores and

    the activator of CRAC channels as stromal in-teraction molecule (STIM) (88, 89). STIM is

    an ER-resident transmembrane protein with aC-terminal cytoplasmic coiled-coil motif and,

    within the ER lumen, an N-terminal sterile

    motif (SAM) and paired EF hands, where

    one hand binds a single Ca2+ ion with lowaffinity (88, 89). Two STIM proteins, STIM1

    and STIM2, are found in mammals, and re-cent work has shown that STIM1 is important

    for the initial robust phase of SOCE, whereasSTIM2 is important for maintaining basal Ca2+

    levels and sustaining the late phase of SOCE

    (90, 91). Following ER Ca2+ depletion, STIM1molecules aggregate in clusters that preferen-

    tially localize to sites of ER plasma membrane

    www.annualreviews.org T Cell Activation 601

  • 7/27/2019 Activacin de cels T

    12/31

    NFAT: nuclear factorof activated T cells

    apposition, where they colocalize with clusters

    of Orai1, forming punctae (9294). Thebiochemical mechanism by which STIM1 cou-

    ples Ca2+ depletion to CRAC activation isnot yet fully understood and is an area of in-

    tense investigation. Recentwork hasshown that

    STIM1 oligomerization is sufficient to induce

    punctae formation and CRAC channel activa-tion independent of Ca2+ store depletion (95).Further studies showed that the C-terminal

    coiled-coil domain of STIM1 alone can inducedimerization of Orai1 dimers, which is suffi-

    cientfor CRACchannel activation independentof punctae formation and store depletion (96).

    How STIM1 oligomers translocate to areas ofER plasmamembraneappositionand how, once

    there, they induce theOrai1 tetramerization re-main unknown. Interestingly, the WASp family

    verprolin homologous protein (WAVE2) com-plex is also required for SOCE activity, througha mechanism that remains to be fully elucidated,

    although independent of its function in actinremodeling (97, 98). CRAC channels appear to

    be the dominant mode of Ca2+ entry in T cells,but other Ca2+ channels exist. Their relevance

    remains unclear (reviewed in 84).TCR-induced increases in intracellular

    Ca2+ levels result in the activation of Ca2+

    and calmodulin-dependent transcription fac-

    tors, including myocyte-enhancing factor 2(MEF2) and downstream regulatory element

    antagonist modulator (DREAM), as well as sig-

    naling proteins, including the phosphatase cal-cineurin and the Ca2+-calmodulin-dependent

    kinase (CaMK), that in turn activate a vari-ety of transcription programs (reviewed in 99).

    Activated calcineurin dephosphorylates mem-bers of the nuclear factor of activated T cells

    (NFAT) family, leading to their translocationto the nucleus. In the nucleus, NFAT iso-

    forms can form cooperative complexes with avariety of other transcription factors, thereby

    integrating signaling pathways, resulting in dif-ferential gene expression patterns and func-

    tional outcomes, depending on the context of

    the TCR signal. The most well-studied inter-action is NFAT/AP-1, which integrates Ca2+

    and Ras signals and results in the expression of

    genes important for T cell activation inclu

    ing IL-2. In contrast, NFAT activity in the sence of AP-1 activation induces a pattern

    gene expression that ultimately results in T canergy and a characteristic lack of IL-2 p

    duction (100). It is still unclear whether NFAisoforms are cooperating with other transcr

    tion factors or are functioning as dimersinduce the anergic transcriptional pattern (viewed in 101). The regulatory T cell lineag

    specific transcription factor forkhead bprotein 3 (FOXP3) also cooperates with NF

    and antagonizes NFAT/AP-1 gene transcrtion, resulting in Treg functional gene expr

    sionandalackofIL-2production(102).FinaNFAT family members can also cooperate w

    STAT proteins to induce either Th1 or Tdifferentiation through expression of T-bet

    GATA3, respectively (99).

    ACTIN AND CYTOSKELETALRESPONSES

    When a T cell is presented with cognate angen by an APC, signals from the TCR initi

    a program of actin cytoskeletal rearrangemethat results in polarization and activation of

    T cell (reviewed in 104). Actin reorganizatioessential for T cell function, as actin polym

    ization inhibitors impede T cell/APC intertions (105) and abolish proximal TCR sign

    (106).

    T cell/APC conjugation results in morphlogical changes, as the stimulated T cell roun

    up and accumulates filamentous actin (F-actat the stimulatory interface. These changes

    thought to depend on a TCR-induced increin plasma membrane fluidity and a decre

    in cellular motility. Cessation of motilityassociated with TCR-induced Ca2+-depend

    phosphorylation and deactivation of the myomotor protein MyH9; however, the signal

    pathway linking the TCR to this event has nyet been defined fully (107). Plasma membra

    fluidity is increased, in part, by the TCR- a

    Vav1-dependent transient dephosphorylatof ERM (ezrin, radixin, and moesin) prote

    resulting in the loss of their ability to link t

    602 Smith-Garvin Koretzky Jordan

  • 7/27/2019 Activacin de cels T

    13/31

    plasma membrane to the actin cytoskeleton

    (108). Ca2+ signaling and integrin activationdownstream of the TCR result in additional

    modifications of actin-associated proteins thatmay play roles in altering plasma membrane

    rigidity (104).Accumulation of F-actin at the T cell/APC

    interface is the result of TCR-induced lo-calized activation of multiple actin regulatoryand polymerizing pathways, the best stud-

    ied of which involves the actin-related pro-teins 2/3 (Arp2/3) complex, although Arp2/3-

    independent pathways also contribute to thisprocess (109). Activation of Arp2/3 requires its

    interaction with nucleation-promoting factors(NPF) including Wiskott-Aldrich syndrome

    protein (WASp), WAVE2, and hematopoieticcell lineagespecific protein 1 (HS1). WASp is

    recruited to the site of TCR activation throughits interaction with the SLP-76-associated

    adapter protein Nck, where it is activated via

    Vav1-dependent stimulation of the Rho fam-ily GTPase Cdc42 (110). Vav1-mediated acti-

    vation of a second Rho family GTPase, Rac1,results in the activation of WAVE2 (104). Given

    the proposed reliance of WASp and WAVE2on Vav1-mediated GTPase activation, it is in-

    teresting that actin-dependent processes thatare defective in Vav1-deficient T cells can

    be rescued with the expression of a GEF-inactive Vav1 mutant, suggesting that other

    Rac and Cdc42 GEFs may be able to sup-port TCR-induced actin changes (55). This

    result also suggests that other Vav1 functions

    are important for TCR-induced actin changes.Consistent with this is the observation that,

    through its protein interaction domains, Vav1may contribute to WAVE2 and WASp activa-

    tion through the recruitment of Dynamin2, aGTPase that is important for TCR-induced

    actin dynamics (111).Activation of the T cell in response to an

    APC also results in the polarization of the Tcell, whereby the microtubular organizing cen-

    ter (MTOC) moves toward the T cell/APCcontact site (112). Although polarization of the

    MTOC has long been observed as a hallmark of

    productive T cell/APC conjugation, the signal-

    cSMAC: centralsupramolecularactivation cluster

    pSMAC: peripherasupramolecular

    activation cluster

    ing mechanisms responsible for this movement

    remain undefined. Recent data suggest, how-ever, that the adapter protein ADAP (a com-

    ponent of the SLP-76-nucleated complex) mayplay a role through its interaction with the mi-

    crotubule motor protein dynein (113). Move-ment of the MTOC appears essential for the

    formation of the immunological synapse (IS).The IS is an organized structure that devel-opsatthecontactsitebetweentheTcellandthe

    APC. It is composed of two concentric regionsbased on molecular composition: the TCR-

    rich central supramolecular activation cluster(cSMAC), surrounded by the integrin-rich pe-

    ripheral SMAC (pSMAC) (114, 115). Althoughthe IS was described approximately 10 years

    ago, its precise role in T cell activation re-mains unclear. Initially, the concentration of

    receptors in the cSMAC led to the proposalthat the cSMAC is the site of enhanced recep-

    tor engagement and prolonged signaling (115).

    However, later studiesshowed that TCRsignalspeak prior to cSMAC formation and suggested

    that the cSMAC is primarily the site of TCRdegradation (116, 121, 122). More recently, it

    has been proposed that the cSMAC is the siteof both TCR signal enhancement and TCR

    degradation, andthe balance between these twoprocesses is determined by the quality of the

    antigen such that the cSMAC can serve to am-plify weak agonist signals (117). Most studies of

    the cSMAC have focused on the TCR and itsassociated molecules. However, a newly defined

    subregion of the cSMAC that is rich in CD28, a

    costimulatory molecule (see below), and PKCbut relatively devoid of the TCR points to the

    potential importance of the cSMAC for costim-ulatory molecule signal transduction (118).

    Although the precise roles of the cSMACremain controversial, it is now well accepted

    that the initiation of TCR signals occurs in pe-ripheral microclusters that begin to form prior

    to IS formation. pMHC/TCR ligation resultsin the formation of intracellular microclusters

    that contain the TCR complex and associatedsignaling molecules, including LAT and SLP-

    76 (120). These clusters initiate and can sus-

    tain Ca2+ signals (121). The clusters persist

    www.annualreviews.org T Cell Activation 603

  • 7/27/2019 Activacin de cels T

    14/31

    Inside-out signaling:signals initiated byengagement ofimmunoreceptors thatlead to conformationalchanges and clustering

    of integrins, therebyincreasing the affinityand avidity of theintegrins for theirligands

    for a short time, after which they converge

    toward the cSMAC (121, 122). Formation andtranslocation of the clusters are dependent on

    F-actin dynamics, and new clusters continue toform even after the mature IS is established

    (121123). Integrins play a key role in sus-taining microclusters, emphasizing their im-

    portance for T cell activation (124).Opposite the MTOC and IS, another or-dered structure forms, known as the distal pole

    complex (DPC). Although the role of the DPCis not known for certain, investigators specu-

    late that the DPC is critical for sequesteringnegative regulators away from the TCR acti-

    vation complex (104). Additionally, the DPCmay contribute to the polarization of key

    signaling molecules that may be required fordistinguishing memory versus effector fate de-

    cisions in recently divided cells (125). Forma-tion of this complex is dependent on F-actin

    and the rephosphorylation of ERM proteins

    that migrate to the distal pole linking signal-ing molecules to the cytoskeleton (126).

    TCR signaling cascades and pathwaysdownstream of actin reorganization are inter-

    twined and difficult to tease apart, as manyof the effector molecules involved have mul-

    tiple enzymatic and adapter functions. WASp,WAVE2, and Vav1 signals play roles in TCR-

    induced signalingthat appearto be independentof their roles in actin responses (52, 97, 127).

    Therefore, loss of different actin regulators mayresult in complex TCR signaling defects (128).

    Future studies are required to understand fully

    the feedforward and feedback mechanisms thatdefine the interdependence between cytoskele-

    tal dynamics and T cell activation.

    TCR INSIDE-OUT SIGNALINGTO INTEGRINS

    Integrins are heterodimeric receptors re-

    sponsible for mediating cell/cell or cell/matrixadhesions. Key T cell integrins include leuko-

    cyte functionassociated antigen-1 (LFA-1)

    and very late antigen-4 (VLA-4), which bindtheir respective ligands intercellular adhe-

    sion molecule (ICAM) and vascular cell ad-

    hesion molecule (VCAM) and fibronectin

    other immune cells, endothelial cells, fibrolasts, and extracellular matrix proteins. Acti

    tion of integrins (increasing their affinity aavidity for ligand) is critically dependent

    biochemical events initiated by the TCRprocess designated inside-out signaling (12

    Although the pathway from the TCR to ingrin activation has not been completely eludated, TCR-mediated activation of several k

    signaling molecules as well as TCR-inducactin/cytoskeletalchanges havebeen implica

    in this process.A central regulator of inside-out signal

    is the small GTPase Ras-proximity-1 (RapRap1 enhances T cell activation by mediati

    TCR-induced adhesion to ICAM-1. This coclusion is based on studies utilizing overexpr

    sion of dominant-negative and constitutivactive forms of Rap1 (130, 131) and more

    cently by analysis of mice deficient in Rap

    in which TCR-induced adhesion to ICAMis markedly reduced (132). The importance

    understanding therole of Rap1 is clear, as mutions in Rap1-mediatedintegrin activation h

    been linked to leukocyte adhesiondeficiensyndrome, a disease that can lead to severe b

    terial infections (reviewed in 129).Many proximal signaling molecules t

    comprise the early TCR signalosome, incling LAT, SLP-76, and PLC1, are required

    integrin and Rap1 activation (reviewed in 1(Figure 2). However, there are downstream

    fectors that have a more selective role in

    tegrin activation, including the adapter ptein ADAP. T cells from ADAP-deficient m

    are defective in TCR-induced LFA-1 clusting and adhesion to ICAM (133). The asso

    ation of ADAP with SLP-76 appears to be quiredfor integrin activation, as overexpress

    of ADAP but not a mutant of ADAP that canot bind to SLP-76 enhances integrin funct

    following TCR ligation (134, 135). Althoumice expressing the reciprocal mutation in

    SH2 domain of SLP-76 have been generaand share several characteristics with ADA

    deficient mice, TCR-induced integrin acti

    tion in these mice has not yet been reported

    604 Smith-Garvin Koretzky Jordan

  • 7/27/2019 Activacin de cels T

    15/31

  • 7/27/2019 Activacin de cels T

    16/31

  • 7/27/2019 Activacin de cels T

    17/31

    how Akt mediates T cell growth and survival

    downstream of CD28.The CD28-mediated generation of PIP3

    also serves as a docking site for the PHdomain of Itk.Although Itk inducibly associates

    with the LAT/SLP-76/Gads/PLC1 complex

    that forms following TCR ligation, its local-

    ization and activation also depends on PI3K-generated PIP3 (61). Itk can also associate di-rectly with CD28 via the CD28 proximal PxxP

    motif (61). It is possible that this interactionkeeps Itk close to Lck (which binds to the dis-

    tal PxxP motif of CD28), allowing for Lck-mediated phosphorylation and activation of Itk

    (149) and enhanced Ca2+ flux, another charac-teristic of CD28-mediated signaling. Despite

    the fact that Itk and CD28 associate with oneanother, studies in Itk-deficient T cells demon-

    strate that some CD28 signaling is still intact inthe absence of Itk, suggesting that this interac-tion may not be that critical for CD28 signal-

    ing (150). Although the proline motifs in thetail of CD28 are required for CD28-mediated

    proliferation and IL-2 production, these motifsare dispensable for Bcl-xl upregulation (151).

    This function appears to be more reliant on theproximal YMNM p85 binding site (151). Thus,

    CD28 can differentially regulate proliferationand survival in activated T cells.

    Another more recently described functionof CD28 is induction of arginine methyla-

    tion. Following CD28 ligation, protein argi-

    nine methyltransferase activity increases, andarginine methylation of multiple proteins, in-

    cluding Vav1, is induced. Vav1 arginine methy-lation appears to occur in thenucleusandcorre-

    lates with IL-2 production (152). Although theprecise biologic significance of this posttrans-

    lational modification is unknown, this pathwaymay provide yet another mechanism by which

    CD28 regulates TCR signaling.Many of the pathways described above are

    activated by TCR ligation alone; however, themagnitude of the response is considerably aug-

    mented with CD28 coligation. This observa-

    tion has led to speculation that CD28 engage-ment results primarily in a quantitative rather

    than a qualitative change in T cell activation

    parameters (141). Although this appears to be

    true, it is also true that quantitative differencesin signaling can result in qualitatively distinct

    functional outcomes.CD28-deficient mice exhibit dampened im-

    mune responses to a variety of infectious agents(reviewed in 141). Although these studies

    demonstrate the importance of costimulationby CD28, notall immuneresponses areseverely

    impacted by its loss (153). Such observations in-

    dicate that molecules other than CD28 can pro-vide costimulation for T cells. Indeed, multiple

    surface receptors have been described as havingcostimulatory functions. Included among these

    areCD2, CD5, CD30, 4-1BB, OX40, induciblecostimulator (ICOS), and LFA-1. For this re-

    view, we focus on the CD28-related proteinICOS and two members of the tumor necro-

    sis factor receptor (TNFR) family to provideexamples of how costimulatory molecules can

    link to downstream effectors, either directly orthrough adapter proteins.

    Unlike CD28, which is expressed at con-

    stantlevelsonbothrestingandactivatedTcells,ICOS is induciblyexpressed on activatedT cells

    (154). ICOS deficiency results in impaired im-mune responses, similar to yet not as severe

    as those observed in CD28 knockout models,suggesting that these two molecules may func-

    tion in similar pathways (155). Indeed, ICOSshares several structural features with CD28,

    including a YMXM motif in its cytoplasmictail that associates with p85 (155). This site

    is presumed to be responsible for PI3K-drivenAkt and/or Itk activation observed downstream

    of CD3/ICOS stimulation, which likely con-

    tributes to the similarities seen in CD3/CD28and CD3/ICOS-stimulated cells (156). ICOS

    does not induce IL-2 gene transcription asCD28 does. Failure to induce IL-2 has been

    attributed, at least in part, to the inability of theYxxM motif of ICOS to associate with Grb2,

    an association that is present via this motif inCD28 (157). Therefore, although ICOS acti-

    vates genes similar to those induced by CD28,there are differences in the degree to which par-

    ticular genes are expressed. These differenceshave in vivo relevance, as mice doubly deficient

    www.annualreviews.org T Cell Activation 607

  • 7/27/2019 Activacin de cels T

    18/31

  • 7/27/2019 Activacin de cels T

    19/31

  • 7/27/2019 Activacin de cels T

    20/31

    Dok-1 and -2, are expressed in T cells. Co-

    ordinated deletion or knockdown of theseproteins results in increased TCR cytokine

    production and proliferation; prolonged phos-phorylation of ZAP-70, LAT, SLP-76, and

    Akt; and the development of a lupus-like re-nal diseasewith highantidouble-stranded DNA

    antibody titers (167, 168). Dok can asso-ciate with several negative regulators includ-ing SHIP-1 (SH2 domaincontaining inositol

    phosphatase), Csk, and Ras GTPase-activatingprotein (RasGAP); however, how these associ-

    ations mediate the negative regulatory role ofDok proteins remains to fully elucidated. Re-

    cent structure/function studies in T cell linesshowed that the phosphotyrosine-binding do-

    main(PTB)ofDok-1and-2canbindtheITAMmotif in CD3, making it tempting to specu-

    late that Dok may compete with ZAP-70 forITAM binding (167). Whether this interaction

    is relevant in vivo remains to be shown. Dok

    proteins can also be recruited inducibly to aLAT-, Grb2-, and SHIP-1-containing molecu-

    lar complex (168). This finding is intriguing andbegs the question as to how LAT participates in

    both positive and negative signaling pathways.Determining the domains necessary for the as-

    sembly of this negative regulatory complex andwhether it is separate from or a part of the stim-

    ulatory LAT-containing complex may provideinsight into how early phosphorylation events

    are regulated.It has been suggested that SLP-76 can also

    be a target of negative regulation. The serine/

    threonine kinase HPK1 inducibly binds to theSH2 domain of SLP-76. This kinase has both

    positive and negative effects on TCR signaltransduction; however, its role as a promi-

    nent negative regulator was confirmed re-cently by the description of HPK1-deficient

    mice. HPK1-deficient T cells exhibit en-hanced phosphorylation of several early sig-

    naling molecules, and HPK1-deficient miceare more susceptible to experimental autoim-

    mune encephalomyelitis (EAE) than are wild-type mice (169). As a possible explanation for

    these phenotypes, two groups have shown that

    HPK1 can phosphorylate a serine residue

    SLP-76 that mediates recruitment of 14-family members (169, 170). 14-3-3 prote

    havebeenimplicatedintheregulationofsevesignaling pathways. Thus, although the p

    cise mechanism by which HPK1 disrupts TCsignal transduction remains unclear, identifi

    tion of a SLP-76/14-3-3 interaction may incate that this mechanism involves a conserv

    method of signal transduction regulation.

    The role of a novel family of proteins in rulating TCR signaling has become apprecia

    more recently. The suppressor of T cell rector signaling (Sts) family of proteins conta

    two members, Sts-1 (TULA2) and -2 (TULCombined deficiency in these proteins leads

    hyperproliferative T cells and an increased sceptibility to autoimmunity (171). When t

    phenotype of Sts-1 and -2 knockout mice wdescribed, the mechanism of negative regu

    tion was unclear. Although still not understofully, the C-terminal phosphoglycerate mut

    domain of Sts-1 acts as a phosphatase w

    specificity for Syk and, to a lesser degree, family members, thus providing a model

    how Sts-1 may negatively regulate TCR snal transduction (172). Interestingly, Sts-2 h

    very little phosphatase activity and, in cell lmodels, can enhance ZAP-70 activation (17

    Recent studies demonstrating that Sts-2 cbind to and induce the degradation of c-C

    (a negative regulator of TCR signaling; see blow) have provided a mechanism by which St

    may act as a positive regulator of T cell actition (174). Whether net ZAP-70 activity is u

    mately increased or decreased in response to

    in vivo challenge likely depends on the relatexpression of these two family members.

    Cbl proteins play an important role as native regulators of T cell signaling. Two fam

    members, c-Cbl and Cbl-b, are immune moulators, as deletion of each gene in vivo leads

    hypercellularity and, in the case of Cbl-b, sptaneous mulitorgan infiltration (175, 176). B

    c-Cbl and Cbl-b facilitate the ubiquitinationproteins, targeting them for degradation. Th

    also mediate the downregulation of the TC

    610 Smith-Garvin Koretzky Jordan

  • 7/27/2019 Activacin de cels T

    21/31

    following stimulation with antigenic peptides

    and have been recently shown to play a role inthe dissipation of the early signaling complex

    (176178). Thus, Cbl proteins, by way of theirability to regulate protein degradation, provide

    anexampleofyetanothermeansbywhichTCRsignaling is terminated.

    Similar to the necessity of signals providedby the TCR to be complemented by costim-ulatory molecules, TCR-generated regulatory

    signals are also aided by coreceptor signals.Cytotoxic T lymphocyte antigen-4 (CTLA-4)

    and programmed death-1 (PD-1) are two ex-amples of such receptors that limit the ex-

    pansion and activation of TCR-triggered Tcells. These molecules are found on activated

    T cells with peak expression 2448 h afterstimulation. Genetic studies have documented

    the importance of both for maintaining self-tolerance. By 56 days after birth, CTLA-4-

    deficient mice exhibit activated peripheral T

    cells, splenomegaly, and lymphocytic infiltratesinto nonlymphoid organs (179). Although less

    striking, PD-1-deficient mice also develop au-toimmune features, including the development

    of a lupus-like disease by 6 months of age (180).Like costimulatory receptors, inhibitory recep-

    tors utilizemotifs andmolecular pathways simi-lar to those used by the TCR when propagating

    a negative signal; they may even provide dock-ing sites for these shared molecules (181, 182).

    Another mode by which inhibitory receptorshave been proposed to function is to utilize the

    same ligand and/or signaling molecules as cos-

    timulatory receptors, thus setting up the poten-tial for competition or sequestration of ligands

    or key substrates. This means is best illustratedby CTLA-4 and CD28, which share the lig-

    ands CD80 and CD86 and counter each otherin the regulation of cell cycle proteins, cytokine

    expression, and Cbl-b expression.

    CONCLUSIONS

    As in the first 10 years after identification of

    the TCR, the past 15 years have seen a dra-

    matic expansion in our understanding of thebiochemical pathways triggered downstream ofthe TCR. We now appreciate more fully howPTKs couple to effectors and second messen-

    gers via the utilization of adapter proteins, howRas and NF-B pathways are activated, how

    Ca2+ ions are sensed, that TCRsignaling can be

    required for the activation of other cell surfacereceptors, and that the actin cytoskeleton does

    more than just dictate cell shape. Studies inves-tigating signal transduction by costimulatory

    molecules have highlighted the importance ofsignal amplification, and discoveries of spon-

    taneous or induced models of autoimmunityhave brought negative regulation of these sig-

    naling pathways to the forefront. However, asoften noted in this review, links between many

    signaling molecules and cellular outcomes re-main poorly defined. Future studies will be

    required to fill gaps in our knowledge andlikely will reveal new and unexpected interac-

    tions between currently known and unknown

    molecules.This review primarily focused on the signal

    transduction pathways in naive T cells. Overthe next 15 years, it will be important to ap-

    ply current and new knowledge to other T celllineages, including T regulatory cells, natural

    killer T cells, and memory T cells, to estab-lish whether these paradigms are universal. In

    the coming years, the population-based analy-ses that established the field of signal transduc-

    tion will be driven toward single cell analyses.We can anticipate that this technological shift

    will allow for better analysis of human T cells

    and the application of basic science to humandisease.

    DISCLOSURE STATEMENT

    The authors are not aware of any affiliations, memberships, funding, or financial holdings that

    might be perceived as affecting the objectivity of this review.

    www.annualreviews.org T Cell Activation 611

  • 7/27/2019 Activacin de cels T

    22/31

    ACKNOWLEDGMENTS

    We thank Christopher P. Garvin for figure design, Dr. Art Weiss for helpful comments, and JustStadanlick for editorial assistance.

    LITERATURE CITED

    1. Haskins K, Kappler J, Marrack P. 1984. The major histocompatibility complex-restricted antigen

    ceptor on T cells. Annu. Rev. Immunol. 2:51662. Cantrell D. 1996. T cell antigen receptor signal transduction pathways. Annu. Rev. Immunol. 14:259

    3. Yanagi Y, Yoshikai Y, Leggett K, Clark SP, Aleksander I, Mak TW. 1984. A human T cell-specific cD

    clone encodes a protein having extensive homology to immunoglobulin chains. Nature 308:14549

    4. Hedrick SM,Cohen DI,Nielsen EA,Davis MM. 1984. Isolationof cDNA clonesencoding T cell-spec

    membrane-associated proteins. Nature 308:14953

    5. Malissen M, Minard K, Mjolsness S, Kronenberg M, Goverman J, et al. 1984. Mouse T cell antigen

    ceptor: structure and organization of constant and joining gene segments encoding the beta polypept

    Cell37:110110

    6. Allison JP, McIntyre BW, Bloch D. 1982. Tumor-specific antigen of murine T-lymphoma defined w

    monoclonal antibody. J. Immunol. 129:2293300

    7. Haskins K, Kubo R, White J, Pigeon M, Kappler J, Marrack P. 1983. The major histocompatibi

    complex-restricted antigen receptor on T cells. I. Isolation with a monoclonal antibody. J. Exp. M157:114969

    8. Meuer SC, Fitzgerald KA, Hussey RE, Hodgdon JC, Schlossman SF, Reinherz EL. 1983. Clonoty

    structures involved in antigen-specifichumanT cell function. Relationship to the T3 molecular comp

    J. Exp. Med. 157:70519

    9. Borst J, Coligan JE, Oettgen H, Pessano S, Malin R, Terhorst C. 1984. The delta- and epsilon-cha

    of the human T3/T-cell receptor complex are distinct polypeptides. Nature 312:45558

    10. Dembic Z, Haas W, Weiss S, McCubrey J, Kiefer H, et al. 1986. Transfer of specificity by murine

    T-cell receptor genes. Nature 320:23238

    11. Saito T, Germain RN. 1987. Predictable acquisition of a new MHC recognition specificity follow

    expression of a transfected T-cell receptor -chain gene. Nature 329:25659

    12. Weiss A, Stobo JD. 1984. Requirement for the coexpression of T3 and the T cell antigen receptor o

    malignant human T cell line. J. Exp. Med. 160:12849913. Abraham RT, Weiss A. 2004. Jurkat T cells and development of the T-cell receptor signalling paradi

    Nat. Rev. Immunol. 4:3018

    14. Weiss A, Imboden JB. 1987. Cell surface molecules and early events involved in human T lymphoc

    activation. Adv. Immunol. 41:138

    15. Imboden JB, Stobo JD. 1985. Transmembrane signalling by the T cell antigen receptor. Perturbatio

    the T3-antigen receptor complex generates inositol phosphates and releases calcium ions from intra

    lular stores. J. Exp. Med. 161:44656

    16. Samelson LE, Patel MD, Weissman AM, Harford JB, Klausner RD. 1986. Antigen activation of mur

    T cells induces tyrosine phosphorylation of a polypeptide associated with the T cell antigen recep

    Cell46:108390

    17. Samelson LE, Phillips AF, Luong ET, Klausner RD.1990. Association of thefyn protein-tyrosine kin

    with the T-cell antigen receptor. Proc. Natl. Acad. Sci. USA 87:43586218. Veillette A, Bookman MA, Horak EM, Bolen JB. 1988. The CD4 and CD8 T cell surface antigens

    associated with the internal membrane tyrosine-protein kinase p56lck. Cell55:3018

    19. Barber EK, Dasgupta JD, Schlossman SF, Trevillyan JM, Rudd CE. 1989. The CD4 and CD8 antig

    are coupled to a protein-tyrosine kinase (p56lck) that phosphorylates the CD3 complex. Proc. Natl. A

    Sci. USA 86:327781

    20. June CH, Fletcher MC, Ledbetter JA, Schieven GL, Siegel JN, et al. 1990. Inhibition of tyrosine ph

    phorylation prevents T-cellreceptor-mediated signaltransduction.Proc. Natl. Acad. Sci. USA 87:7722

    612 Smith-Garvin Koretzky Jordan

  • 7/27/2019 Activacin de cels T

    23/31

    21. Reth M. 1989. Antigen receptor tail clue. Nature 338:38384

    2225. Four papers

    describing chimeric

    proteins that

    demonstrate the

    signaling capability of

    CD3, supporting the

    notion that CD3 is th

    signaling component

    the TCR complex.

    22. Irving BA, Weiss A. 1991. The cytoplasmic domain of the T cell receptor chain is sufficient to

    couple to receptor-associated signal transduction pathways. Cell64:891901

    23. Romeo C, Amiot M, Seed B. 1992. Sequence requirements for induction of cytolysis by the T

    cell antigen/Fc receptor chain. Cell68:88997

    24. Wegener AM, Letourneur F, Hoeveler A, Brocker T, Luton F, Malissen B. 1992. The T cell

    receptor/CD3 complex is composed of at least two autonomous transduction modules. Cell

    68:8395

    25. Letourneur F, Klausner RD. 1992. Activation of T cells by a tyrosine kinase activation domainin the cytoplasmic tail of CD3. Science 255:7982

    26. Describes the

    cloning and initial

    characterization of

    ZAP-70, the Syk fami

    PTK essential for

    coupling the TCR to

    downstream signaling

    machinery.

    26. Chan AC, Iwashima M, Turck CW, Weiss A. 1992. ZAP-70: a 70 kd protein-tyrosine kinase that

    associates with the TCR chain. Cell71:64962

    27. Iwashima M, Irving BA, van Oers NS, Chan AC, Weiss A. 1994. Sequential interactions of the TCR

    with two distinct cytoplasmic tyrosine kinases. Science 263:113639

    28. Aivazian D, Stern LJ.2000. Phosphorylation of T cell receptor is regulatedby a lipid dependent folding

    transition. Nat. Struct. Biol. 7:102326

    29. Xu C, Gagnon E, Call ME, Schnell JR, Schwieters CD, et al. 2008. Regulation of T cell receptor

    activation by dynamic membrane binding of the CD3epsilon cytoplasmic tyrosine-based motif. Cell

    135:70213

    30. The first ITAM-independent function

    for a CD3 molecule is

    identified in the PRR

    CD3 as a binding sit

    for Nck.

    30. Gil D, Schamel WW, Montoya M, Sanchez-Madrid F, Alarcon B. 2002. Recruitment of Nck by

    CD3 reveals a ligand-induced conformational change essential for T cell receptor signaling andsynapse formation. Cell109:90112

    31. Mingueneau M,Sansoni A, GregoireC, Roncagalli R,Aguado E, etal. 2008. Theproline-rich sequenceof

    CD3 controls T cell antigen receptor expression on and signaling potency in preselection CD4+CD8+

    thymocytes. Nat. Immunol. 9:52232

    32. Szymczak AL, Workman CJ, Gil D, Dilioglou S, Vignali KM, et al. 2005. The CD3 proline-rich

    sequence, and its interaction with Nck, is not required for T cell development and function. J. Immunol.

    175:27075

    33. Gil D, Schrum AG, Daniels MA, Palmer E. 2008. A role for CD8 in the developmental tuning of antigen

    recognition and CD3 conformational change. J. Immunol. 180:39009

    34. Tailor P, Tsai S, Shameli A, Serra P, Wang J, et al. 2008. The proline-rich sequence of CD3 as an

    amplifier of low-avidity TCR signaling. J. Immunol. 181:24355

    35. Takeuchi K, Yang H, Ng E, Park SY, Sun ZY, et al. 2008. Structural and functional evidence that Nckinteraction with CD3 regulates T-cell receptor activity. J. Mol. Biol. 380:70416

    36. Rudolph MG, Stanfield RL, Wilson IA. 2006. How TCRs bind MHCs, peptides, and coreceptors.Annu.

    Rev. Immunol. 24:41966

    37. Sun ZJ, Kim KS, Wagner G, Reinherz EL. 2001. Mechanisms contributing to T cell receptor signaling

    and assembly revealed by the solution structure of an ectodomain fragment of the CD3 heterodimer.

    Cell105:91323

    38. Ma Z, Janmey PA, Finkel TH. 2008. The receptor deformation model of TCR triggering. FASEB J.

    22:10028

    39. Minguet S, Schamel WW. 2008. A permissive geometry modelfor TCR-CD3 activation. Trends Biochem.

    Sci. 33:5157

    40. Alarcon B, Swamy M, van Santen HM, Schamel WW. 2006. T-cell antigen-receptor stoichiometry:

    preclustering for sensitivity. EMBO Rep. 7:4909541. Davis SJ, Van Der Merwe PA. 2006. The kinetic-segregation model: TCR triggering and beyond. Nat.

    Immunol. 7:8039

    42. Varma R. 2008. TCRtriggering by thepMHC complex: valency, affinity, anddynamics. Sci. Signal1:pe21

    43. Krogsgaard M, Li QJ, Sumen C, Huppa JB, Huse M, Davis MM. 2005. Agonist/endogenous peptide-

    MHC heterodimers drive T cell activation and sensitivity. Nature 434:23843

    44. Zhang W, Sloan-Lancaster J, Kitchen J, Trible RP, Samelson LE. 1998. LAT: the ZAP-70 tyrosine

    kinase substrate that links T cell receptor to cellular activation. Cell92:8392

    www.annualreviews.org T Cell Activation 613

  • 7/27/2019 Activacin de cels T

    24/31

  • 7/27/2019 Activacin de cels T

    25/31

    67. Egan SE, Giddings BW, Brooks MW, Buday L, Sizeland AM, Weinberg RA. 1993. Association of Sos

    Ras exchange protein with Grb2 is implicated in tyrosine kinase signal transduction and transformation.

    Nature 363:4551

    68. Dower NA, Stang SL, Bottorff DA, Ebinu JO, Dickie P, et al. 2000. RasGRP is essential for mouse

    thymocyte differentiation and TCR signaling. Nat. Immunol. 1:31721

    69. Ebinu JO,Bottorff DA, Chan EY,StangSL, Dunn RJ,Stone JC.1998. RasGRP, a Rasguanyl nucleotide-

    releasing protein with calcium- and diacylglycerol-binding motifs. Science 280:108286

    70. Roose JP,Mollenauer M, Gupta VA, Stone J, Weiss A. 2005. A diacylglycerol-protein kinaseC-RasGRP1

    pathway directs Ras activation upon antigen receptor stimulation of T cells. Mol. Cell. Biol. 25:44264171. Finco TS, Kadlecek T, Zhang W, Samelson LE, Weiss A. 1998. LAT is required for TCR-mediated

    activation of PLC-1 and the Ras pathway. Immunity 9:61726

    72. Roose JP, Mollenauer M, Ho M, Kurosaki T, Weiss A. 2007. Unusual interplay of two types of Ras

    activators, RasGRP and SOS, establishes sensitive and robust Ras activation in lymphocytes. Mol. Cell.

    Biol. 27:273245

    73. Hayashi K, Altman A. 2007. Protein kinase C theta (PKC): a key player in T cell life and death.

    Pharmacol. Res. 55:53744

    74. Melowic HR, Stahelin RV, Blatner NR, Tian W, Hayashi K, et al. 2007. Mechanism of diacylglycerol-

    induced membrane targeting and activation of protein kinase C. J. Biol. Chem. 282:2146776

    75. Vallabhapurapu S, Karin M. 2009. Regulation and function of NFB transcription factors in the immune

    system. Annu. Rev. Immunol. 27:693733

    76. Schulze-LuehrmannJ, Ghosh S. 2006. Antigen-receptor signaling to nuclear factorB.Immunity 25:70115

    77. Matsumoto R, Wang D, Blonska M, Li H, Kobayashi M, et al. 2005. Phosphorylation of CARMA1 plays

    a critical role in T cell receptor-mediated NFB activation. Immunity 23:57585

    78. SommerK, Guo B, Pomerantz JL,Bandaranayake AD,Moreno-Garcia ME,et al.2005. Phosphorylation

    of the CARMA1 linker controls NFB activation. Immunity 23:56174

    79. Sun L, Deng L, Ea CK, Xia ZP, Chen ZJ. 2004. The TRAF6 ubiquitin ligase and TAK1 kinase mediate

    IKK activation by BCL10 and MALT1 in T lymphocytes. Mol. Cell14:289301

    80. Zhou H, Wertz I, ORourke K, Ultsch M, Seshagiri S, et al. 2004. Bcl10 activates the NF-B pathway

    through ubiquitination of NEMO. Nature 427:16771

    81. Coornaert B, Baens M, Heyninck K, Bekaert T, Haegman M, et al. 2008. T cell antigen receptor

    stimulation induces MALT1 paracaspase-mediated cleavage of the NF-B inhibitor A20. Nat. Immunol.

    9:2637182. Rebeaud F, Hailfinger S, Posevitz-Fejfar A, Tapernoux M, Moser R, et al. 2008. The proteolytic activity

    of the paracaspase MALT1 is key in T cell activation. Nat. Immunol. 9:27281

    83. Medeiros RB,Burbach BJ,Mueller KL, Srivastava R, Moon JJ,et al.2007. Regulation of NFB activation

    in T cells via association of the adapter proteins ADAP and CARMA1. Science 316:75458

    84. Oh-Hora M, Rao A. 2008. Calcium signaling in lymphocytes. Curr. Opin. Immunol. 20:25058

    85. Zhang SL, Yeromin AV, Zhang XH, Yu Y, Safrina O, et al. 2006. Genome-wide RNAi screen of Ca2+

    influx identifies genes that regulate Ca2+ release-activated Ca2+ channel activity. Proc. Natl. Acad. Sci.

    USA 103:935762

    86. Feske S, Gwack Y, Prakriya M, Srikanth S, Puppel SH, et al. 2006. A mutation in Orai1 causes immune

    deficiency by abrogating CRAC channel function. Nature 441:17985

    87. Vig M, Peinelt C, Beck A, Koomoa DL, Rabah D, et al. 2006. CRACM1 is a plasma membrane protein

    essential for store-operated Ca2+ entry. Science 312:12202388. Roos J, DiGregorio PJ, Yeromin AV, Ohlsen K, Lioudyno M, et al. 2005. STIM1, an essential and

    conserved component of store-operated Ca2+ channel function. J. Cell Biol. 169:43545

    89. Liou J, Kim ML, Heo WD, Jones JT, Myers JW, et al. 2005. STIM is a Ca 2+ sensor essential for

    Ca2+-store-depletion-triggered Ca2+ influx. Curr. Biol. 15:123541

    90. Oh-Hora M, Yamashita M, Hogan PG, Sharma S, Lamperti E, et al. 2008. Dual functions for the

    endoplasmic reticulum calcium sensors STIM1 and STIM2 in T cell activation and tolerance. Nat.

    Immunol. 9:43243

    www.annualreviews.org T Cell Activation 615

  • 7/27/2019 Activacin de cels T

    26/31

    91. Brandman O, Liou J, Park WS, Meyer T. 2007. STIM2 is a feedback regulator that stabilizes b

    cytosolic and endoplasmic reticulum Ca2+ levels. Cell131:132739

    92. Liou J, Fivaz M, Inoue T, Meyer T. 2007. Live-cell imaging reveals sequential oligomerization and lo

    plasma membrane targeting of stromal interaction molecule 1 after Ca 2+ store depletion. Proc. N

    Acad. Sci. USA 104:93016

    93. Wu MM, Buchanan J, Luik RM, Lewis RS. 2006. Ca2+ store depletion causes STIM1 to accumulat

    ER regions closely associated with the plasma membrane. J. Cell Biol. 174:80313

    94. Luik RM, Wu MM, Buchanan J, Lewis RS. 2006. The elementary unit of store-operated Ca 2+ en

    local activation of CRAC channels by STIM1 at ER-plasma membrane junctions.J. Cell Biol. 174:81595. Luik RM, Wang B, Prakriya M, Wu MM, Lewis RS. 2008. Oligomerization of STIM1 couples

    calcium depletion to CRAC channel activation. Nature 454:53842

    96. Penna A, Demuro A, Yeromin AV, Zhang SL, Safrina O, et al. 2008. The CRAC channel consists

    tetramer formed by Stim-induced dimerization of Orai dimers. Nature 456:11620

    97. Nolz JC, Gomez TS, Zhu P, Li S, Medeiros RB, et al. 2006. The WAVE2 complex regulates ac

    cytoskeletal reorganizationand CRAC-mediatedcalcium entry during T cellactivation. Curr. Biol. 16:

    34

    98. Nolz JC, Medeiros RB, Mitchell JS, Zhu P, Freedman BD, et al. 2007. WAVE2 regulates high-affin

    integrin binding by recruiting vinculin and talin to the immunological synapse. Mol. Cell. Biol. 27:59

    6000

    99. Savignac M, Mellstrom B, Naranjo JR. 2007. Calcium-dependent transcription of cytokine genes in

    lymphocytes. Pflugers Arch. 454:52333100. Macian F, Garcia-Cozar F, Im SH, Horton HF, Byrne MC, Rao A. 2002. Transcriptional mechani

    underlying lymphocyte tolerance. Cell109:71931

    101. Bandyopadhyay S, Soto-Nieves N, Macian F. 2007. Transcriptional regulation of T cell tolerance. Sem

    Immunol. 19:18087

    102. Wu Y, Borde M, Heissmeyer V, Feuerer M, Lapan AD, et al. 2006. FOXP3 controls regulatory T

    function through cooperation with NFAT. Cell126:37587

    103. Dolmetsch RE, Lewis RS, Goodnow CC, Healy JI. 1997. Differential activation of transcription fac

    induced by Ca2+ response amplitude and duration. Nature 386:85558

    104. Burkhardt JK, Carrizosa E, Shaffer MH. 2008. The actin cytoskeleton in T cell activation. Annu. R

    Immunol. 26:23359

    105. Henney CS, Bubbers JE. 1973. Antigen-T lymphocyte interactions: inhibition by cytochalasin

    J. Immunol. 111:8590

    106. Holsinger LJ, Graef IA, Swat W, Chi T, Bautista DM, et al. 1998. Defects in actin-cap formation

    Vav-deficient mice implicate an actin requirement for lymphocyte signal transduction. Curr. Biol. 8:5

    72

    107. Jacobelli J, Chmura SA, Buxton DB, Davis MM, Krummel MF. 2004. A single class II myosin modul

    T cell motility and stopping, but not synapse formation. Nat. Immunol. 5:53138

    108. Faure S, Salazar-Fontana LI, Semichon M, Tybulewicz VL, Bismuth G, et al. 2004. ERM prote

    regulate cytoskeleton relaxation promoting T cell-APC conjugation. Nat. Immunol. 5:27279

    109. Gomez TS, Kumar K, Medeiros RB, Shimizu Y, Leibson PJ, Billadeau DD. 2007. Formins regu

    the actin-related protein 2/3 complex-independent polarization of the centrosome to the immunolog

    synapse. Immunity 26:17790

    110. Zeng R, Cannon JL, Abraham RT, Way M, Billadeau DD, et al. 2003. SLP-76 coordinates N

    dependent Wiskott-Aldrich syndrome protein recruitment with Vav-1/Cdc42-dependent Wisk

    Aldrich syndrome protein activation at the T cell-APC contact site. J. Immunol. 171:136068111. Gomez TS, Hamann MJ, McCarney S, Savoy DN, Lubking CM, et al. 2005. Dynamin 2 regulates T

    activation by controlling actin polymerization at the immunological synapse. Nat. Immunol. 6:2617

    112. Kupfer A, Swain SL, Singer SJ. 1987. The specific direct interaction of helper T cells and antig

    presenting B cells. II. Reorientation of the microtubule organizing center and reorganization of

    membrane-associated cytoskeleton inside the bound helper T cells. J. Exp. Med. 165:156580

    113. Combs J, Kim SJ, Tan S, Ligon LA, Holzbaur EL, et al. 2006. Recruitment of dynein to the Jur

    immunological synapse. Proc. Natl. Acad. Sci. USA 103:1488388

    616 Smith-Garvin Koretzky Jordan

  • 7/27/2019 Activacin de cels T

    27/31

    114. Monks CR, Freiberg BA, Kupfer H, Sciaky N, Kupfer A. 1998. Three-dimensional segregation of

    supramolecular activation clusters in T cells. Nature 395:8286

    115. Grakoui A, Bromley SK, Sumen C, Davis MM, Shaw AS, et al. 1999. The immunological synapse: a

    molecular machine controlling T cell activation. Science 285:22127

    116. Lee KH, Holdorf AD, Dustin ML, Chan AC, Allen PM, Shaw AS. 2002. T cell receptor signaling

    precedes immunological synapse formation. Science 295:153942

    117. Cemerski S, Das J, Giurisato E, Markiewicz MA, Allen PM, Chakraborty AK, Shaw AS. 2008. The

    balance between T cell receptor signaling and degradation at the center of the immunological synapse

    is determined by antigen quality. Immunity 29:41422118. Yokosuka T, Kobayashi W, Sakata-Sogawa K, Takamatsu M, Hashimoto-Tane A, et al. 2008. Spa-

    tiotemporal regulation of T cell costimulation by TCR-CD28 microclusters and protein kinase C

    translocation. Immunity 29:589601

    119. Dustin ML. 2008. T-cell activationthroughimmunological synapses and kinapses.Immunol. Rev. 221:77

    89

    120. Bunnell SC, Hong DI, Kardon JR, Yamazaki T, McGlade CJ, et al. 2002. T cell receptor ligation induces

    the formation of dynamically regulated signaling assemblies. J. Cell Biol. 158:126375

    121. Demonstrated th

    peripheral

    microclusters, not

    cSMAC, are the sites

    both TCR signalinitiation and

    maintenance.

    121. Yokosuka T, Sakata-Sogawa K, Kobayashi W, Hiroshima M, Hashimoto-Tane A, et al. 2005.

    Newly generated T cell receptor microclusters initiate and sustain T cell activation by recruit-

    ment of Zap70 and SLP-76. Nat. Immunol. 6:125362

    122. Varma R, Campi G,YokosukaT, Saito T, DustinML. 2006. T cell receptor-proximal signals aresustained

    in peripheral microclusters and terminated in the central supramolecular activation cluster. Immunity

    25:11727

    123. Campi G, Varma R, Dustin ML. 2005. Actin and agonist MHC-peptide complex-dependent T cell

    receptor microclusters as scaffolds for signaling. J. Exp. Med. 202:103136

    124. Nguyen K, Sylvain NR,Bunnell SC. 2008. T cell costimulation viathe integrin VLA-4 inhibits theactin-

    dependent centralization of signaling microclusters containing the adaptor SLP-76.Immunity 28:81021

    125. Chang JT, Palanivel VR, Kinjyo I, Schambach F, Intlekofer AM, et al. 2007. Asymmetric T lymphocyte

    division in the initiation of adaptive immune responses. Science 315:168791

    126. Allenspach EJ, Cullinan P, Tong J, Tang Q, Tesciuba AG, et al. 2001. ERM-dependent movement of

    CD43 defines a novel protein complex distal to the immunological synapse. Immunity 15:73950

    127. Zhang W, Irvin BJ, Trible RP, Abraham RT, Samelson LE. 1999. Functional analysis of LAT in TCR-

    mediated signaling pathways using a LAT-deficient Jurkat cell line. Int. Immunol. 11:94350128. Billadeau DD, Nolz JC, Gomez TS. 2007. Regulation of T-cell activation by the cytoskeleton. Nat. Rev.

    Immunol. 7:13143

    129. Menasche G, Kliche S, Bezman N, Schraven B. 2007. Regulation of T-cell antigen receptor-mediated

    inside-out signaling by cytosolic adapter proteins and Rap1 effector molecules. Immunol. Rev. 218:8291

    130. Katagiri K, Hattori M, MinatoN, Irie S, TakatsuK, Kinashi T. 2000. Rap1 is a potent activation signalfor

    leukocyte function-associated antigen 1 distinct from protein kinase C and phosphatidylinositol-3-OH

    kinase. Mol. Cell. Biol. 20:195669

    131. SebzdaE, BrackeM, Tugal T, Hogg N, Cantrell DA.2002. Rap1A positively regulatesT cells viaintegrin

    activation rather than inhibiting lymphocyte signaling. Nat. Immunol. 3:25158

    132. Duchniewicz M, Zemojtel T, Kolanczyk M, Grossmann S, Scheele JS, Zwartkruis FJ. 2006. Rap1A-

    deficient T and B cells show impaired integrin-mediated cell adhesion. Mol. Cell. Biol. 26:64353

    133. Peterson EJ, Woods ML, Dmowski SA, Derimanov G, Jordan MS, et al. 2001. Coupling of the TCR tointegrin activation by Slap-130/Fyb. Science 293:226365

    134. Kliche S, Breitling D, Togni M, Pusch R, Heuer K, et al. 2006. The ADAP/SKAP55 signaling module

    regulatesT-cell receptor-mediatedintegrinactivationthrough plasma membrane targeting of Rap1.Mol.

    Cell. Biol. 26:713044

    135. Wang H, McCann FE, Gordan JD, Wu X, Raab M, et al. 2004. ADAP-SLP-76 binding differen-

    tially regulates supramolecular activation cluster (SMAC) formation relative to T cell-APC conjugation.

    J. Exp. Med. 200:106374

    www.annualreviews.org T Cell Activation 617

  • 7/27/2019 Activacin de cels T

    28/31

    136. Menasche G, Kliche S, Chen EJ, Stradal TE, Schraven B, Koretzky G. 2007. RIAM links

    ADAP/SKAP-55 signaling module to Rap1, facilitating T-cell-receptor-mediated integrin activati

    Mol. Cell Biol. 27:407081

    137. Identified RIAM as

    a key adapter in Rap1

    plasma membrane

    localization and integrin

    activation in T cells.

    137. Lafuente EM, van Puijenbroek AA, Krause M, Carman CV, Freeman GJ, et al. 2004. RIAM,

    Ena/VASP and profilin ligand, interacts with Rap1-GTP and mediates Rap1-induced adhesi

    Dev. Cell7:58595

    138. Medeiros RB, Dickey DM, Chung H, Quale AC, Nagarajan LR, et al. 2005. Protein kinase D1 and

    1 integrin cytoplasmic domain control1 integrin function via regulation of Rap1 activation.Immu

    23:21326139. Katagiri K, Maeda A, Shimonaka M, Kinashi T. 2003. RAPL, a Rap1-binding molecule that media

    Rap1-induced adhesion through spatial regulation of LFA-1. Nat. Immunol. 4:74148

    140. Han J, Lim CJ, Watanabe N, Soriani A, Ratnikov B, et al. 2006. Reconstructing and deconstruct

    agonist-induced activation of integrin IIb3. Curr. Biol. 16:1796806

    141. Acuto O, Michel F. 2003. CD28-mediated co-stimulation: a quantitative support for TCR signalli

    Nat. Rev. Immunol. 3:93951

    142. Pages F, Ragueneau M, Rottapel R, Truneh A, Nunes J, et al. 1994. Binding of phosphatidylinosito

    OH kinase to CD28 is required for T-cell signalling. Nature 369:32729

    143. Narayan P, Holt B, Tosti R, Kane LP. 2006. CARMA1 is required for Akt-mediated NF-B activat

    in T cells. Mol. Cell. Biol. 26:232736

    144. Qiao G, Li Z, Molinero L, Alegre ML, Ying H, et al. 2008. T-cell receptor-induced NFB activatio

    negatively regulated by E3 ubiquitin ligase Cbl-b. Mol. Cell. Biol. 28:247080145. BealsCR, SheridanCM, TurckCW, Gardner P, CrabtreeGR. 1997.Nuclear export of NF-ATc enhan

    by glycogen synthase kinase-3. Science 275:193034

    146. Huang GN, Huso DL, Bouyain S, Tu J, McCorkell KA, et al. 2008. NFAT binding and regulation o

    cell activation by the cytoplasmic scaffolding Homer proteins. Science 319:47681

    147. CD28

    costimulation increases

    glucose uptake and

    glycolysis, via a

    PI3K/Akt pathway, to

    levels exceeding that

    required for immediate

    use.

    147. Frauwirth KA, Riley JL, Harris MH, Parry RV, Rathmell JC, et al. 2002. The CD28 signali

    pathway regulates glucose metabolism. Immunity 16:76977

    148. Jacobs SR, Herman CE, Maciver NJ, Wofford JA, Wieman HL, et al. 2008. Glucose uptake is limit

    in T cell activation and requires CD28-mediated Akt-dependent and independent pathways. J. Immu

    180:447686

    149. Heyeck SD, Wilcox HM, Bunnell SC, Berg LJ. 1997. Lck phosphorylates the activation loop tyros

    of the Itk kinase domain and activates Itk kinase activity. J. Biol. Chem. 272:254018

    150. Li CR, Berg LJ. 2005. Itk is not essential for CD28 signaling in naive T cells. J. Immunol. 174:4475151. Burr JS, Savage ND, Me