in mun ology

Upload: biol-miguel-angel-gutierrez-dominguez

Post on 02-Apr-2018

225 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/27/2019 In Mun Ology

    1/78

    Secondary bacterial pneumonia is a

    common complication of influenzavirus infection in humans. But whataccounts for the increased morbidity

    that is associated with viralbacterialco-infection? Now, Medzhitov andcolleagues describe a mouse model of

    co-infection with influenza virus andLegionella pneumophila that can beused to discriminate between the abil-

    ity of the host to detect and eliminatea pathogen (resistance) and the abilityto maintain homeostasis with a given

    level of pathogen burden (tolerance).All mice that had been infected

    intranasally with a sublethal

    dose of influenza virus followed3 days later by a sublethal dose ofL. pneumophila died within a week

    of co-infection, whereas all micesurvived a single infection with the

    virus or the bacteria. Importantly,

    there was no significant differencein the viral or bacterial burden after

    single infection or co-infection, andco-infection was not associated withsystemic dissemination of either

    pathogen. Furthermore, an attenu-ated strain ofL. pneumophila that isunable to secrete virulence factors

    still resulted in 100% mortalityin co-infected mice. So, bacterialgrowth or virulence is not required

    for the effects of co-infection, which

    confirms that this is not a failure of

    immune resistance.Using various mouse strains

    with genetic deletions of key immune

    molecules, as well as antibody-mediated depletion of neutrophilsor natural killer cells, the authors

    showed that co-infection still resultsin mortality in the absence of allmajor immune and inflammatory

    pathways that might be triggeredby the virus or bacteria. They sug-gest that these results show that an

    excessive inflammatory responseleading to immunopathology is notthe cause of death after co-infection.

    Even in the absence of all controllableimmunostimulatory signals whenimmunodeficient (Toll-like receptor 2

    (Tlr2)/Tlr4/) mice were infectedwith influenza virus and a severely

    attenuated L. pneumophila strain that

    lacked flagellin and was unable toreplicate or secrete effectors most

    mice died following co-infection butnot after a single infection with eitherthe virus or the bacteria.

    So, if resistance is not involved inco-infection-associated mortality in terms of the failure of the immune

    response to control pathogen growthand spread, increased pathogen

    virulence or the effects of immuno-

    pathology the alternative scenario

    is that the host is unable to tolerate

    the tissue damage that results fromco-infection. The lungs of co-infectedmice had significantly increased

    necrosis of airway epithelial cellscompared with singly infected mice,and genes involved in tissue protec-

    tion and repair were downregulatedin co-infected mice compared withsingly infected mice. Administration

    of amphiregulin an epidermalgrowth factor family member that hasa role in maintaining lung homeosta-

    sis during influenza virus infection to Tlr2/Tlr4/ mice that had beeninfected with influenza virus and the

    severely attenuated L. pneumophilastrain significantly decreased lungdamage and mortality but had no

    effect on viral or bacterial burden.Together, the results show that

    tolerance can determine the outcome

    of an infection independently ofresistance and is, thus, a bona fide

    host defence strategy that could betargeted for therapeutic purposes.

    Kirsty Minton

    I N F E C T I O N

    Resistance is futile

    ORIGINAL RESEARCH PAPER Jamieson, A. M. et al.

    Role of tissue protection in lethal respiratory viral-

    bacterial coinfection. Science 25 Apr 2013

    (doi:10.1126/science.1233632)

    FURTHER READING Schneider, D. S. & Ayres, J. S.

    Two ways to survive infection: what resistance and

    tolerance can teach us about treating infectious

    diseases. Nature Rev. Immunol.8, 889895 (2008)

    tolerance can

    determine the

    outcome ofan infection

    independently

    of resistance

    and is, thus,

    a bona fide

    host defence

    strategy

    NPG

    R E S E A R C H H I G H L I G H T S

    NATURE REVIEWS |IMMUNOLOGY VOLUME 13 | JUNE 2013

    Nature Reviews Immunology| AOP, published online 7 May 2013; doi:10.1038/nri3462

    2013 Macmillan Publishers Limited. All rights reserved

  • 7/27/2019 In Mun Ology

    2/78

  • 7/27/2019 In Mun Ology

    3/78

    During an adaptive immuneresponse, T follicular helper (T

    FH)

    cells enter germinal centres andsupport the affinity maturation,isotype class-switching and memory

    responses of antigen-specific B cells.Previous studies have focused on howcognate interactions shape germinal

    centre responses; Qi and colleaguesnow report that costimulatory signalsdelivered by bystander B cells are

    essential for TFH

    cell recruitment tothe germinal centre.

    Deficiency of inducible T cell

    costimulator (ICOS) leads to defec-tive germinal centre responses.This was thought to be owing to the

    inability of ICOS-deficient T cells toupregulate CXC-chemokine recep-tor 5 (CXCR5), which is required for

    migration into B cell follicles and,as such, for recruitment to germinal

    centres. The authors transduced

    ICOS-deficient ovalbumin (OVA)-specific CD4+ T cells (OT-II cells)

    with CXCR5 and found that theywere still unable to migrate into thefollicles after immunization with

    haptenated OVA. Instead, these cellsaccumulated at the T cellB cell(TB) border at the follicular edge.

    ICOS-deficient OT-II cells that weretransduced with the transcriptionfactor B cell lymphoma 6 (BCL-6; a

    master regulator of TFH

    cell develop-ment) also failed to enter the follicles.

    Therefore, ICOS signalling promotesT

    FHcell recruitment by a mechanism

    that is distinct from CXCR5 orBCL-6 upregulation.

    Surprisingly, recruitment of wild-

    type OT-II cells was not impairedif OVA-presenting dendritic cellsor OVA-presenting B cells were

    defective in ICOS ligand (ICOSL)expression. This suggested that ICOSsignalling promotes T cell entry into

    the follicles in an antigen-independentmanner. In support of this theory,T cells that had been activated in vitro

    could migrate deep into follicles,even if they were not specific for theimmunizing antigen; however, their

    entry was strictly dependent on ICOSexpression. Therefore, the authorsexamined whether ICOS signalling

    provided by bystander B cells pro-motes T cell entry into the follicles. In

    mice with a B cell-specific deficiency

    in ICOSL, activated OT-II cellswere unable to enter the follicles. By

    contrast, these T cells were recruitedinto follicles in control mice inwhich B cells expressed ICOSL,

    even if the B cells were deficient inMHC class II expression.

    Closer analyses showed that

    ICOSL-expressing bystanderB cells do not alter chemokinegradient sensing by T cells;

    instead, they promote persistentrandom motility in T cells in a

    phosphoinositide 3-kinase (PI3K)-dependent manner. Two-photon

    intravital imaging showed thatactivated T cells at the TBborder extended frequent pseudo-

    pods and maintained a polarizedstate, which is crucial for directionalcell migration. By contrast, in

    animals with ICOS-deficient T cellsor ICOSL-deficient B cells, T cellswere depolarized and showed

    decreased motility at the follicularborder. Finally, the authors showedthat the lack of expression of ICOSL

    by bystander B cells led to defectivegerminal centre responses followingimmunization, even when antigen-

    specific T cells and B cells werepresent and competent for ICOS andICOSL expression, respectively.

    The authors conclude thatbystander follicular B cells at the TB

    border provide an ICOS-dependent

    signal to activated CD4+ T cells,thereby promoting their eventual

    recruitment to germinal centres.Notably, this study shows thatstimulatory signals that are delivered

    to T cells by B cells in an antigen-independent manner also supportgerminal centre responses.

    Yvonne Bordon

    LYMPHOCYTE RESPONSES

    Stand by for action!

    ORIGINAL RESEARCH PAPER Xu, H. et al.

    Follicular T-helper cell recruitment governed by

    bystander B cells and ICOS-driven motility.Nature

    25 Apr 2013 (doi: 10.1038/nature12058)

    bystander

    B cells are

    essential

    for TFH

    cell

    recruitment to

    the germinal

    centre

    R E S E A R C H H I G H L I G H T S

    NATURE REVIEWS |IMMUNOLOGY VOLUME 13 | JUNE 2013

    Nature Reviews Immunology| AOP, published online 13 May 2013; doi:10.1038/nri3467

    2013 Macmillan Publishers Limited. All rights reserved

  • 7/27/2019 In Mun Ology

    4/78

    As well as detecting pathogenicorganisms, Toll-like receptors (TLRs)

    coordinate homeostatic responsesto commensal bacteria. Chambonand colleagues now report that TLR

    expression by intestinal epithelialcells (IECs) is under circadian con-trol, leading to oscillatory immune

    signalling in response to the micro-biota. Notably, in the absence of themicrobiota the circadian clock is

    disrupted in IECs and mice developmetabolic disturbances.

    To examine whether commensal

    bacteria are involved in circadianresponses, the authors compared tem-poral gene expression profiles in IECs

    from control and microbiota-depletedmice. IECs from microbiota-depletedmice showed disrupted circadian

    control of several nuclear receptors,including PPAR (peroxisomeproliferator-activated receptor-),

    REV-ERB and ROR (retinoic acidreceptor-related orphan receptor-),and of components of the circadian

    clock machinery. Notably, depletionof the microbiota also led to systemic

    metabolic effects, with animals show-ing increased blood levels of glucose,triglycerides and free fatty acids, and

    decreased production of insulin.Further analyses showed that

    these metabolic disturbances resulted

    from increased corticosterone syn-thesis by ileal IECs, which was causedby increased PPAR expression

    in these cells. Indeed, microbiota-depleted mice with an IEC-specificdeletion of PPAR showed normal

    regulation of circadian clockcomponents and did not show anymetabolic disturbances. Microbial

    products promoted the activationof JUN N-terminal kinase (JNK)and the activator protein 1 (AP-1)

    transcription factor JUN in IECs, andJUN was shown to repress PPAR.However, although JNK and JUN

    showed a circadian mode of activa-tion, their levels of gene and proteinexpression did not change over time.

    Instead, the authors found thatthe expression of TLR1 to TLR5

    and also TLR9 (but not TLR6 andTLR7) is under circadian control inIECs. This explains how microbiota-

    derived products can activate JNKand JUN and repress PPAR in acircadian manner. The temporal

    transcription of TLRs in IECs wasshown to be activated and repressedby the alternate binding of ROR or

    REV-ERB, respectively, to a con-served site in the TLR gene promot-ers. Using a bioinformatics approach,

    the authors found that a largenumber of genes (>2,000) that areexpressed by IECs contain the ROR

    and REV-ERB DNA-binding site.This suggests that many of the genesthat are involved in homeostatic IEC

    responses are controlled in a micro-biota-dependent circadian manner.Indeed, the authors confirmed that

    several immune mediators that areimportant for maintaining epithelialbarrier integrity are regulated in this

    way in IECs.Taken together, the data show

    a key role for the microbiota and

    TLRs in controlling circadianresponses in IECs. Such circadiancontrol of IEC responses could be

    important for coordinating homeo-static IEC functions with behavioural

    activities in mice.

    Yvonne Bordon

    MUCOSAL IMMUNOLOGY

    TLRs get rhythm

    ORIGINAL RESEARCH PAPER Mukherji, A. et al.

    Homeostasis in intestinal epithelium is

    orchestrated by the circadian clock and

    microbiota cues transduced by TLRs. Cell153,

    812827 (2013)

    FURTHER READING Sheiermann, C., Kunisaki, Y.

    & Frenette, P. S. Circadian control of the immune

    system.Nature Rev. Immunol.13, 190198 (2013)

    TLR

    expression

    by intestinal

    epithelial cells

    ... is undercircadian

    control

    COMSTOCKIMAGES

    R E S E A R C H H I G H L I G H T S

    NATURE REVIEWS |IMMUNOLOGY VOLUME 13 | JUNE 2013

    Nature Reviews Immunology| AOP, published online 17 May 2013; doi:10.1038/nri3472

    2013 Macmillan Publishers Limited. All rights reserved

  • 7/27/2019 In Mun Ology

    5/78

  • 7/27/2019 In Mun Ology

    6/78

    It has been previously suggested that

    thymus-derived regulatory T (TReg

    )cells primarily recognize self anti-gens, whereas peripherally derived

    TReg

    cells respond to foreign antigens.Indeed, conversion of forkhead boxP3 (FOXP3)CD4+ T cells to FOXP3+

    TReg

    cells is known to be favoured bythe tolerogenic microenvironmentof the gut, where there is an abun-

    dance of innocuous foreign antigens,including food- and microbiota-derived components. However,

    Cebula et al. now suggest that mostcolonic T

    Regcells, including T

    Regcells

    that recognize intestinal commensal

    bacteria, develop in the thymus.The authors used transgenic mice

    that express a limited but diverse

    T cell receptor (TCR) repertoire(TCRmini mice) to investigate the

    origin of colonic TReg

    cells onthe basis of high-throughput TCRsequencing analyses. T cells from

    TCRmini mice express a single TCR

    -chain combined with a variety of

    TCR -chains, and their develop-ment and function was found to becomparable to that of non-transgenic

    T cells.Sorted individual FOXP3+ T cells

    and FOXP3CD4+ T cells from the

    thymus, peripheral lymph nodesand intestines of TCRmini mice wereanalysed in terms of the comple-

    mentarity determining region 3(CDR3) sequence of the TCR-chain that they expressed. It was

    found that the most abundant TCRsdid not overlap between the FOXP3+and FOXP3CD4+ T cell subsets,

    irrespective of the organ of origin.Moreover, 86% of the TCRs from

    colonic TReg

    cells were identical to

    50% of the TCRs from FOXP3+CD4+thymocytes. This suggests that in all

    organs that were analysed, includingthe intestines, peripheral conversionof FOXP3CD4+ T cells to FOXP3+

    TReg

    cells did not account for the

    majority of TReg

    cells. A similar result

    was observed in transgenic mice thathad an even broader TCR repertoire.So, colonic T

    Regcells seem to derive

    predominantly from FOXP3+CD4+thymocytes.

    But do these thymus-derived

    intestinal TReg

    cells respond to foreignantigens such as those derived fromcommensal bacteria? Treatment of

    TCRmini mice with antibiotics alteredthe frequency of the most dominantcolonic T

    Regcell TCRs, which prob-

    ably reflects the antigen-specificexpansion or contraction of colonicT

    Regcell populations. However, the

    diversity of the colonic TReg

    cell TCRrepertoire was not reduced followingthe changes in the composition

    of colonic microbiota. Moreover,most colonic T

    Regcell TCRs were

    shared with FOXP3+CD4+ thymocytes

    even after treatment with antibiotics.This indicates that changes in thecomposition of the population of

    intestinal commensal bacteria didnot promote substantial recruitmentof peripherally derived T

    Regcells.

    Finally, hybridomas that weregenerated from colonic T

    Regcells

    showed responsiveness to sterile

    filtrates of caecal contents and tosonicates of individual commensalbacterial species. As 90% of the

    TCRs of hybridomas that respondedto caecal contents were expressedby FOXP3+CD4+ thymocytes, the

    authors conclude that thymus-derived T

    Regcells have a leading

    role in establishing intestinal T cell

    tolerance against microbiota.Maria Papatriantafyllou

    TOLERANCE

    The origins of colonic TReg

    cells

    ORIGINAL RESEARCH PAPER Cebula, A. et al.

    Thymus-derived regulatory T cells contribute to

    tolerance to commensal microbiota. Nature497,

    258262 (2013)

    colonic TReg

    cells seem

    to derive

    predominantly

    from

    FOXP3+CD4+

    thymocytes

    Ph

    oto

    dis

    c/G

    etty

    Im

    ages

    R E S E A R C H H I G H L I G H T S

    NATURE REVIEWS |IMMUNOLOGY VOLUME 13 | JUNE 2013

    Nature Reviews Immunology| AOP, published online 17 May 2013; doi:10.1038/nri3468

    2013 Macmillan Publishers Limited. All rights reserved

  • 7/27/2019 In Mun Ology

    7/78

  • 7/27/2019 In Mun Ology

    8/78

  • 7/27/2019 In Mun Ology

    9/78

    A major role of the immune system is to maintainhomeostatic tissue function. For example, sterile tis-sue damage that occurs after trauma needs to bedetected and repaired. Pathogens that can invade andcause harm to tissues should be eliminated while ourcommensal microbiome must be tolerated, as it fulfilsfunctions that are required for host survival. The innateimmune system has a number of signalling receptorsthat recognize foreign molecular structures as well asself molecules that are altered, that have become tooabundant or that emerge in areas normally devoid ofthese molecules1,2.

    Innate immune signalling receptors monitor theextracellular space as well as many subcellular com-partments for signs of infection, damage or othercellular stressors. The inflammasomes are a groupof multimeric protein complexes that consist of aninflammasome sensor molecule, the adaptor proteinASC and caspase 1. Inflammasome formation is trig-gered by a range of substances that emerge duringinfections, tissue damage or metabolic imbalances.Once the protein complexes have formed, the inflam-

    masomes activate caspase 1, which proteolytically acti-vates the pro-inflammatory cytokines interleukin-1(IL-1)3 and IL-18. In addition, inflammasome acti-

    vation causes a rapid, pro-inflammatory form of celldeath called pyroptosis4.

    With the discovery of pattern recognition receptors(PRRs), such as inflammasome sensor molecules, theirsignalling pathways and their ability to programme cel-lular immune responses, we are beginning to understandhow the immune system protects the host at a molecularlevel. At the same time, we have learnt that immune dys-regulation contributes to prevalent diseases in Westernsocieties such as atherosclerosis, type 2 diabetes, cancer

    and neurodegenerative diseases. Thus, a fine balancemust be maintained between the activation and inhi-bition of inflammation to allow the immune systemto remove any sources of danger without causing harm tothe host.

    In this Review, we present an overview of the currentunderstanding of inflammasome activation and regu-lation, and we discuss the recent findings about non-canonical processing of IL-1. We also compare thestructures and the regulation of inflammasomes withthat of the apoptosome.

    The inflammasomes and their coreceptors

    Inflammasome components. At first glance, the inflam-masomes are organized in a very simple manner: inflam-masome sensor molecules (see below) connect to caspase 1

    via ASC, which is an adaptor protein encoded byPYCARDthat is common to all inflammasomes. ASC consists of twodeath-fold domains: one pyrin domain and one caspase acti-

    vation and recruitment domain (CARD). ASC interactswith the upstream inflammasome sensor molecules viathe pyrin domain5. This interaction triggers the assem-

    bly of ASC into a large protein speck consisting mainlyof multimers of ASC dimers6,7. Using its CARD, ASCbrings monomers of pro-caspase 1 into close proximity,which initiates caspase 1 self-cleavage and the formationof the active heterotetrameric caspase 1. Active caspase 1proteolytically activates a number of proteins8, includingpro-IL-1 and pro-IL-18 (REFS 9,10), and induces theirrelease via a non-classical secretion pathway11.

    The transcription of pro-IL-1 is induced by theactivation of the transcription factor nuclear factor-B(NF-B), whereas pro-IL-18 is constitutively expressedand its expression is increased after cellular activation.Therefore, these potent pro-inflammatory cytokines are

    1Institute of Innate Immunity,

    University Hospital,

    University of Bonn,

    Bonn 53127, Germany.2Department of Medicine

    and Division of Infectious

    Diseases and Immunology,

    University of Massachusetts

    Medical School, Worcester

    01655, Massachusetts, USA.3Deutsches Zentrum fr

    Neurodegenerative

    Erkrankungen (DZNE),

    Bonn 53175, Germany.4Centre of Molecular

    Inflammation Research,Norwegian University of

    Science and Technology,

    Trondheim NO-7491,

    Norway.5Structural Immunobiology

    Unit, Laboratory of

    Immunology, National

    Institute of Allergy and

    Infectious Diseases,

    National Institutes of Health,

    Bethesda, Bethesda 20892

    Maryland, USA.

    Correspondence to E.L.

    e-mail: [email protected]

    doi:10.1038/nri3452

    ASC

    An adaptor protein that was

    originally found to form protein

    precipitates in apoptotic cells

    that are termed protein specks.

    Activation and regulation of theinflammasomesEicke Latz14, T. Sam Xiao5 and Andrea Stutz1

    Abstract | Inflammasomes are key signalling platforms that detect pathogenic microorganisms

    and sterile stressors, and that activate the highly pro-inflammatory cytokines interleukin-1

    (IL-1) and IL-18. In this Review, we discuss the complex regulatory mechanisms that facilitate

    a balanced but effective inflammasome-mediated immune response, and we highlight the

    similarities to another molecular signalling platform the apoptosome that monitorscellular health. Extracellular regulatory mechanisms are discussed, as well as the intracellular

    control of inflammasome assembly, for example, via ion fluxes, free radicals and autophagy.

    REVIEWS

    NATURE REVIEWS |IMMUNOLOGY VOLUME 13 | JUNE 2013 |397

    2013 Macmillan Publishers Limited. All rights reserved

    mailto:eicke.latz%40uni-bonn.de?subject=mailto:eicke.latz%40uni-bonn.de?subject=
  • 7/27/2019 In Mun Ology

    10/78

  • 7/27/2019 In Mun Ology

    11/78

  • 7/27/2019 In Mun Ology

    12/78

    IFNAR

    Type I IFN

    TRIF

    TNFR familymembersuch as CD95

    Dectin 1

    Mitochondrion

    Cytoplasm

    TLR4

    Bacterium

    BacterialmRNA

    Caspase 11

    ?

    Caspase 1

    Pro-IL-1

    NLRP3

    ASC

    IL-1

    Caspase 8

    Ripoptosome

    Fusion

    ?

    ?

    ?

    Non-canonical NLRP3inflammasome in responseto Gram-negative bacteria

    Non-canonicalcaspase 8inflammasome

    Auto-processing?

    Phagosome

    Lysosome

    Actinmobilization

    Negativefeedback

    Degradation Sequestration

    FADD

    RIP1

    XIAP

    IAP1

    RIP3

    Canonical NLRP3inflammasome

    Ripoptosome-induced IL-1processing

    TNFR-inducedIL-1 processing

    IL-1 secretion Pyroptosis

    Pro-IL-18 IL-18

    Pro-caspase 11

    Type IIFN

    IFN-induciblefactor

    Nucleus

    SYK

    ASCMALT1

    IAP2

    PRR

    ?

    ?

    ?

    ?

    ?

    formed, under which circumstances caspase 11 activa-tion will lead to NLRP3 activation and when it will causecell death independently of the canonical inflamma-somes. A recent review discusses caspase 11 activationin more detail48.

    Noninflammasome processing of IL1The inflammasomes and IL1 processingin vitro andin vivo. Studies that have been carried out in vitro usingcells that are deficient in inflammasome componentshave undoubtedly been instrumental in identifying

    several inflammatory triggers as activators of inflam-masomes. However, the contribution of inflammas-omes to IL-1 activation in vivo has not, at times, beennearly as convincing. The reduction in inflammationin mice that are deficient in the inflammasome compo-nents ASC or caspase 1 can be much less pronouncedthan that in mice lacking the IL-1 receptor (IL-1R)or IL-1. It is probable that inflammasome-independentIL-1 activation mechanisms are involved in vivo thatcould account for the observed differences betweenthe in vitro and in vivo studies. For example, the in vivo

    Figure 1 | Canonical and non-canonical activation of IL-1.NLRP3 (NOD-, LRR- and pyrin domain-containing 3) needsadditional cofactors for the processing of interleukin-1 (IL-1)in response to Gram-negative bacteria; this pathway has been

    termed the non-canonical NLRP3 inflammasome. Toll-like receptor 4 (TLR4) signalling via TIR domain-containing adaptorprotein inducing IFN (TRIF) induces the secretion of type I interferons (IFNs), which lead to the activation of caspase 11 viaautocrine signalling through the IFN/ receptor (IFNAR), possibly involving additional (not yet known) IFN-inducible factors.Caspase 11 is necessary for the activation of caspase 1 and has an independent role in IL-1 secretion and pyroptosis inresponse to Gram-negative bacteria. A possible mechanism for caspase 11 action might be its role in actin mobilization via

    cofilin, which might lead to increased fusion of lysosomes to phagosomes and, potentially, to increased leakage of bacterial

    mRNA (also termed vita-PAMPs) to the cytoplasm to activate NLRP3 (not shown). A platform consisting of mucosa-associated

    lymphoid tissue lymphoma translocation protein 1 (MALT1), caspase 8 and the adaptor protein ASC (termed the

    non-canonical caspase 8 inflammasome) is formed in response to stimulation of dectin 1. Caspase 8 might also be activated

    by TLRs using the signalling adaptor TRIF in the presence of cycloheximide. In addition, the formation of the ripoptosome is

    triggered by the loss of inhibitor of apoptosis proteins (IAPs) with concurrent TLR stimulation. The ripoptosome consisting

    of FAS-associated death domain protein (FADD) and receptor-interacting protein 1 (RIP1) activates caspase 8 via RIP3.

    However, caspase 8 can also limit ripoptosome action on NLRP3. Members of the tumour necrosis factor receptor (TNFR)

    family might also induce pro-IL-1 cleavage, as has been shown for CD95 (also known as FAS). Question marks showpathways that are still speculative. PRR, pattern recognition receptor; XIAP, X-linked inhibitor of apoptosis protein.

    R E V I E W S

    400 | JUNE 2013 | VOLUME 13 www.nature.com/reviews/immunol

    2013 Macmillan Publishers Limited. All rights reserved

  • 7/27/2019 In Mun Ology

    13/78

  • 7/27/2019 In Mun Ology

    14/78

    Cytoplasm

    Nucleus

    Activatory Inhibitory

    IFNGR IFNAR

    Type I IFN IL-1R PRR PRR

    ?

    ?

    CD40

    CD40L

    TNFR

    IFNAR

    Type I IFNIFN

    IFNGR

    IFN

    Priming viaexpressionlevels

    Degradation

    ? ?

    ?

    iNOS

    PKC

    NLRP3inflammasome

    Nitrosylation

    Nitrosylation

    NLRP3 mRNA

    miR-223

    NLRP3

    pro-IL-1

    AIM2

    NLRC4 NO

    P

    Caspase 1

    T cell

    Priming viadeubiquitylation

    ROS

    Ub

    NLRP3

    BRCC3

    Pro-IL-1 IL-1

    Amyloid-

    An endogenous peptide that is

    generated by proteases in the

    brain. It is prone to aggregation

    and plaque formation.

    Amyloid- plaques are a

    hallmark of Alzheimers disease

    and can activate the NLRP3

    (NOD-, LRR- and pyrindomain-containing 3)

    inflammasome.

    Cryopyrin-associated

    periodic syndrome

    (CAPS). Characterized by

    NLRP3 (NOD-, LRR- and pyrin

    domain-containing 3)

    inflammasome hyperactivity

    and the excessive release of

    interleukin-1, which leads to

    an autoinflammatory disease

    phenotype with periodic fever

    episodes, urticaria and often

    severe arthritis.

    byPRKCD), which depends on the recognition of bothflagellin and the type III secretion system. This indi-cates that PKC might be activated by PRR signalling67.Therefore, at least some inflammasomes are only fullycapable of responding to danger signals in situations in

    which either pro-inflammatory cytokines are present orinnate signalling molecules sense noxious signals.

    A few molecules, such as amyloid-, can induce bothNLRP3 priming through TLR activation and NLRP3inflammasome activation68. However, the response toaggregated amyloid- can be dramatically augmentedwhen innate sensors or cytokine receptors become acti-

    vated by additional stimuli. These priming mechanismsmight be of great relevance in determining the magnitudeof an inflammatory response to danger signals.

    Conversely, genetic differences that influence thethreshold of inflammasome activation might con-tribute to the development of chronic inflammatory

    or autoinflammatory diseases. It is known that about60% of patients with cryopyrin-associated periodicsyndrome (CAPS) carry activating mutations in thecoding sequence ofNLRP3 itself or in other inflamma-some components69. This indicates that inflammasome

    hyperactivity may be influenced by additional mecha-nisms. Indeed, single nucleotide polymorphisms in thepromoter region ofNLRP3 were found in patients withCAPS69. In these patients, an increase in NLRP3 expres-sion as opposed to the presence of a more active pro-tein (as a result of mutations in the coding sequence)could conceivably result in a lower threshold of NLRP3inflammasome activation in response to danger sig-nals and this could trigger the development of CAPS.Therefore, several mechanisms ensure that inflammas-omes are not accidentally triggered and that an appropri-ately balanced immune response to danger signals canoccur in a regulated manner.

    Figure 2 | Extracellular signals regulate the inflammasomes. Pro-interleukin-1 (pro-IL-1) and NLRP3 (NOD-, LRR-and pyrin domain-containing 3) expression are induced by transcriptionally active pattern recognition receptors (PRRs)

    or by cytokine receptors. Furthermore, NLRP3 deubiquitylation by the K63-specific deubiquitinase BRCC3 is crucial for its

    activation. Direct contact with mature or memory T cells inhibits the inflammasomes, probably via tumour necrosis factorreceptor (TNFR) superfamily interactions. Type I interferons (IFNs) inhibit the transcription of proIL1, but also upregulatethe expression of absent in melanoma 2 (AIM2). Both type I IFNs and IFN inhibit NLRP3 through the induction of nitricoxide (NO) via inducible nitric oxide synthase (iNOS), possibly with the requirement of concomitant priming by PRRs.

    Ub shows ubiquitylated proteins. Question marks show pathways that are still speculative. CD40L, CD40 ligand; IFNAR,

    interferon-/receptor; IFNGR, interferon- receptor; IL-1R, IL-1 receptor; miR-223, microRNA-223; NLRC4, NOD-,LRR- and CARD-containing 4; ROS, reactive oxygen species; PKC, protein kinase C.

    R E V I E W S

    402 | JUNE 2013 | VOLUME 13 www.nature.com/reviews/immunol

    2013 Macmillan Publishers Limited. All rights reserved

  • 7/27/2019 In Mun Ology

    15/78

  • 7/27/2019 In Mun Ology

    16/78

    through the hemichannel pannexin 1 (REFS 8284); how-ever, pannexin 1-deficient mice do not show diminishedcaspase 1 activation85. Thus, the mechanisms by whichseveral inflammasome triggers can induce K+ effluxremain unclear.

    In addition to the requirement for decreased K+ lev-els, osmotic pressure regulates NLRP3 inflammasomeactivation86. Cells that have been subjected to hypotonicsolutions undergo cellular swelling concomitant witha decrease in intracellular K+ and Cl concentrations.Interestingly, cell swelling induces a regulatory volumedecrease response through transient receptor poten-tial cation channels (TRPM7 and TRPV2) that triggersintracellular Ca2+ mobilization. The mobilized Ca2+has many molecular targets, including TGF-activatedkinase 1 (TAK1; also known as MAP3K7) (REF. 86).Although TAK1 might have a role in a PRR-dependentnon-transcriptional priming pathway87 that is, in thesignalling pathway that leads to the deubiquitylation, forexample, of NLRP3 further investigation into its roleis required. Another transient receptor potential channel

    TRPM2 has also been implicated in NLRP3 activa-tion in response to crystalline substances88. This channelsenses intracellular ROS and responds by opening itselfto facilitate Ca2+ influx into the cell; this is intriguing con-sidering that both ion fluxes and the oxidative state (seebelow) have important roles in NLRP3 inflammasomeactivation.

    Another regulator of Ca2+, namely C/EPB-homologousprotein (CHOP; also known as DDIT3), has been impli-cated in NLRP3 inflammasome activation89. However,the activation of CHOP following the induction of theunfolded protein response via inhibition of translation isinsufficient for NLRP3 inflammasome activation90 and,therefore, the exact role of CHOP in NLRP3 inflam-masome activation is unclear. Two recent studies haveimplicated calcium-sensing G-protein coupled receptors(GPCRs) in the activation of NLRP3: calcium-sensingreceptor (CASR) and GPRC6A signal via G

    iand G

    q,

    which inhibit adenylyl cyclase and activate phospho-lipase C, respectively91,92. Thus, sensing of extracellularCa2+ leads to reduced cyclic AMP (cAMP) levels throughadenylyl cyclase inhibition and to increased cytoplasmicCa2+ concentrations via the activation of phospholipase C.It is worth noting that the GPCR platelet activating factorreceptor can also stimulate inflammasome formation92,which indicates that other GPCR pathways could controlinflammasome activation. The role of cAMP in inflamma-

    some activation remains unclear as one study has reportedthat cAMP can directly inhibit NLRP3 (REF. 91), whereasanother study reported that cAMP levels had no directinfluence on inflammasome activation92. Taken together,these studies show that NLRP3 activation is regulated bythe ion content of the cell (FIG. 3); however, the moleculartargets and the mechanisms by which ion fluxes regulateNLRP3 remain to be fully elucidated.

    The redox state of the cell is another important indi-cator of the viability of cells, and many signalling path-ways are influenced by changes in the redox state. Inparticular, ROS facilitate the assembly of the apoptosomein several ways. For example, oxidative modifications to

    a b

    c90

    90

    90

    Cytochrome c

    dsDNA

    Pro-caspase 1

    Pro-caspase 9

    ASC

    CARDof APAF1

    APAF1

    AIM2

    Apoptosome

    Ligand-binding domain WD40 repeats HIN

    dsDNA

    PYD

    NACHT

    CARD

    Caspase 9 Caspase 1

    Oligomerization domain

    Signalling domain

    Catalytic domain

    AIM2 inflammasome

    Apoptosome AIM2 inflammasome

    Box 2 | Oligomerization of deathinducing molecular platforms

    Receptors of the death-inducing molecular platforms are activated by ligands such

    as cytochrome c for the apoptosome component apoptotic protease-activating

    factor 1 (APAF1), and double-stranded DNA (dsDNA) for the absent in melanoma 2

    (AIM2) inflammasome. By contrast, the NLRP1 (NOD-, LRR- and pyrin

    domain-containing 1) inflammasome can be activated by self-proteolysis in its

    function-to-find (FIIND) domain131133, which might be relevant to its proteolytic

    activation by the anthrax lethal toxin136

    . The cleaved fragments remain associatedwith each other but the proteolytic event enables NLRP1 to recruit the adaptor

    protein ASC and caspase 1. For the other receptors, ligand binding is thought to

    induce conformational changes, promoting oligomerization of the receptors into

    signalling platforms that recruit adaptors and activate effector caspases.

    Oligomerization of APAF1 and NOD-like receptors (NLRs) is mediated by their

    NACHT domains, which bind and hydrolyse deoxyadenosine triphosphate (dATP)

    or ATP146,147. However, AIM2 seems to use the multivalent ligand dsDNA as its

    oligomerization platform130. Both receptor oligomerization and ligand binding

    might be modulated by the regulatory domains of the receptors, including the

    WD40 repeats of APAF1, the HIN domain of AIM2 and the leucine-rich repeats

    (LRRs) of the NLRs. Two examples of the death-inducing platforms are shown

    in the box figure.

    The apoptosome consists of seven APAF1 molecules that are activated by

    cytochrome c; this then recruits pro-caspase 9 molecules137,148,149 (see figure, part a).

    Six- or eight-fold symmetry of the apoptosomes has also been observed, whichsuggests that there are variable oligomerization states137,148,149. Two hypothetical

    models of the AIM2 inflammasome highlight the positioning of the activating DNA

    either at the centre (see figure, part b) or the periphery (see figure, part c) of the

    complex. It is possible that the physiological AIM2 inflammasome may contain

    characteristics of both models, and the oligomerization states can be variable owing

    to the sequence-independent nature of the DNA-binding. For clarity, only one

    caspase dimer is shown for each oligomeric platform. According to a recent model,

    the pyrin domain (PYD) of ASC can interact with itself in a helical manner to form

    filamentous structures5, which might mediate ASC pyroptosome formation 6.

    CARD, caspase activation and recruitment domain.

    R E V I E W S

    404 | JUNE 2013 | VOLUME 13 www.nature.com/reviews/immunol

    2013 Macmillan Publishers Limited. All rights reserved

  • 7/27/2019 In Mun Ology

    17/78

  • 7/27/2019 In Mun Ology

    18/78

  • 7/27/2019 In Mun Ology

    19/78

    Regulation by interacting host proteins. In addition togeneral cellular conditions such as ion concentrationsand the nutritional state, specific regulatory proteinshave evolved that regulate inf lammasome formation(TABLE 1). The formation of the inflammasome can

    be controlled both at the level of inflammasome sen-sor molecules and, further downstream, at the level ofASC and caspase 1 interaction. As mentioned above,inflammasomes form large protein aggregates andthere are two levels at which death-fold domains areused to recruit effector inflammasome components.First, ASC is recruited to the inflammasome receptorsby homotypic pyrin domain interactions. Next, in a sec-ond death-fold domain interaction, ASC binds to andactivates pro-caspase 1 via its CARD. Studies have eluci-dated inflammasome regulation at the level of death-folddomain interactions and regulatory proteins.

    Caspase activity during cell death is regulated byCARD-containing proteins such as CARD8, whichhas also been shown to be a binding partner of NLRP3(REF. 108). CARD8 has multiple functions in regulat-ing apoptosis, one of which is to directly bind to pro-caspase 9 and to suppress its activation109. Simi larly,mouse caspase 12, which is a paralogue of caspase 1,interacts with caspase 1 to reduce its activity110. A poly-morphism in the caspase 12 (CASP12) gene in humansleads to either a truncated protein or to a full-lengthprotein (caspase 12L)111. Similarly to mouse caspase 12,caspase 12L reduces cytokine production in responseto LPS111, which at high doses can activate theNLRP3 inflammasome and thus caspase 1 without theneed for a second signal. Therefore, caspase 12L and

    mouse caspase 12 probably function as decoy proteinsfor caspase 1, thereby limiting its activation. CARD-only proteins (COPs) can also inhibit caspase 1 acti-

    vation112. CARD18 (also known as ICEBERG) andCARD16 (also known as pseudo-ICE and COP1)were the first decoy COPs to be described113. Throughtheir CARDCARD interaction, these two inhibitorssequester pro-caspase 1 and inhibit its activation by theinflammasome. CARD17 (also known as INCA), whichis another decoy protein, is upregulated by IFN to sup-press IL-1 generation114. Thus, proteins containing aCARD can sequester caspase 1, thereby blocking theformation of a functional inflammasome complex.

    An additional level of fine-tuning and regulationof the inflammasomes can be achieved by interferingwith the pyrinpyrin interaction in the inflamma-somes. Several pyrin domain-only proteins (POPs)and other pyrin-containing proteins have been char-

    acterized. POP1 (also known as PYDC1 and ASC2)and POP2 (also known as PYDC2) inhibit the inter-actions between the inflammasome sensor moleculesand ASC115117. However, COPs and POPs have not beenidentified in the mouse genome, which indicates thata more complex regulatory system might be presentin humans. The fact that COPs and POPs have notbeen identified in the mouse genome makes the studyof these proteins difficult, and many of the COPs andPOPs have only been investigated using overexpres-sion studies and not using genetic deletion or RNAinterference.

    Alternative splicing of ASC can provide an additionallevel of regulation by giving rise to ASC variants thatnegatively regulate the assembly of the inflamma-somes118. Furthermore, NLRP10 (also known as PYNOD)was suggested to negatively regulate inflammasomes bysequestering ASC119,120, but deletion ofNlrp10 in micedid not confirm this hypothesis121,122. NLRP7 has beensuggested to be a negative regulator of inflammasomes;however, more recent findings indicate that NLRP7might assemble an inflammasome in response tomicrobial acylated lipopeptides28,123.

    The role of pyrin, which is a protein that is encodedbyMEFV (Mediterranean fever gene), in inflam-masome regulation also remains unclear. One studyfound that Mefv deletion in mice lead to increased

    IL-1 release without influencing caspase 1 activityor inflammasome assembly. These findings suggestedthat pyrin can inhibit IL-1 release downstream of theinflammasomes124. However, another study found thatshort-term IL-1 release was not impaired after Mefvgene deletion and that IL-1 was only inhibited afterseveral days of stimulation125. In addition, mice carry-ingMefvmutations that have been identified in patientswith familial Mediterranean fever (FMF) showed ASC-dependent but NLRP3-independent release of IL-1125.Therefore, whether pyrin is an inhibitor at the level ofIL-1 release or whether it forms a pyrin inflammasomeremains to be determined.

    Table 1 (cont.) | Regulatory proteins of the inflammasomes

    Regulatoryprotein

    Inflammasomecomponenttargeted

    Mechanism of action Source of experimental evidence Refs

    Other regulators (cont.)

    HSP90 NLRP3 Necessary for activation, possibly owing to its chaperonefunction

    Biochemical evidence and inhibitor ofHSP90

    129

    SGT1 NLRP3 Necessary for activation, possibly owing to its chaperonefunction

    RNAi and biochemical evidence 129

    AIM2, absent in melanoma 2; BCL, B cell lymphoma; CARD, caspase activation and recruitment domain; FMF, familial Mediterranean fever; GBP5,guanylate-binding protein 5; HSP90, heat shock protein 90; IAP, inhibitor of apoptosis protein; IL-1, interleukin-1; NAIP, NLR family, apoptosis inhibitory protein;NLR, NOD-like receptor; NLRC4, NOD-, LRR- and CARD-containing 4; NLRP, NOD-, LRR- and pyrin domain-containing; NOD2, nucleotide-binding oligomerizationdomain-containing protein 2; PKC, protein kinase C; PKR, protein kinase R; POP, pyrin domain-only protein; PYD, pyrin domain; RNAi, RNA interference; XIAP,X-linked IAP. *Expressed only in mice. Expressed only in humans. Two contradictory studies have been published about this interactor.

    R E V I E W S

    NATURE REVIEWS |IMMUNOLOGY VOLUME 13 | JUNE 2013 |407

    2013 Macmillan Publishers Limited. All rights reserved

  • 7/27/2019 In Mun Ology

    20/78

  • 7/27/2019 In Mun Ology

    21/78

  • 7/27/2019 In Mun Ology

    22/78

    43. Gurung, P.et al. Toll or interleukin-1 receptor (TIR)

    domain-containing adaptor inducing interferon-(TRIF)-mediated caspase-11 protease production

    integrates Toll-like receptor 4 (TLR4) protein- and

    Nlrp3 inflammasome-mediated host defense against

    enteropathogens. J. Biol. Chem. 287, 3447434483

    (2012).

    44. Sander, L. E. et al. Detection of prokaryotic mRNA

    signifies microbial viability and promotes immunity.

    Nature 474, 385389 (2011).

    45. Akhter, A. et al. Caspase-11 promotes the fusion of

    phagosomes harboring pathogenic bacteria withlysosomes by modulating actin polymerization.

    Immunity 37, 3547 (2012).

    46. Broz, P.et al. Caspase-11 increases susceptibility to

    Salmonella infection in the absence of caspase-1.

    Nature 490, 288291 (2012).

    47. Aachoui, Y.et al. Caspase-11 protects against bacteria

    that escape the vacuole. Science 339, 975978

    (2013).48. Broz, P. & Monack, D. M. Noncanonical

    inflammasomes: caspase-11 activation and effector

    mechanisms. PLoS Pathog. 9, e1003144 (2013).

    49. Mayer-Barber, K. D. et al. Caspase-1 independent

    IL-1 production is critical for host resistance toMycobacterium tuberculosis and does not require

    TLR signaling in vivo.J. Immunol. 184, 33263330

    (2010).

    50. Provoost, S. et al. NLRP3/caspase-1-independent

    IL-1 production mediates diesel exhaust particle-induced pulmonary inflammation.J. Immunol. 187,

    33313337 (2011).

    51. Kono, H., Orlowski, G. M., Patel, Z. & Rock, K. L.

    The IL-1-dependent sterile inflammatory response has

    a substantial caspase-1-independent component that

    requires cathepsin C.J. Immunol. 189, 37343740

    (2012).52. Gross, O. et al. Inflammasome activators induce

    interleukin-1 secretion via distinct pathways withdifferential requirement for the protease function of

    caspase-1. Immunity 36, 388400 (2012).

    53. Gringhuis, S. I. et al. Dectin-1 is an extracellular

    pathogen sensor for the induction and processing of

    IL-1 via a noncanonical caspase-8 inflammasome.Nature Immunol. 13, 246254 (2012).

    54. Gross, O. et al. Syk kinase signalling couples to the

    Nlrp3 inflammasome for anti-fungal host defence.

    Nature 459, 433436 (2009).

    55. Maelfait, J. et al. Stimulation of Toll-like receptor 3

    and 4 induces interleukin-1 maturation by caspase-8.J. Exp. Med. 205, 19671973 (2008).

    56. Vince, J. E. et al. Inhibitor of apoptosis proteins limit

    RIP3 kinase-dependent interleukin-1 activation.Immunity 36, 215227 (2012).

    57. Kang, T. B., Yang, S. H., Toth, B., Kovalenko, A. &

    Wallach, D. Caspase-8 blocks kinase RIPK3-mediated

    activation of the NLRP3 inflammasome. Immunity 38,

    2740 (2013).

    58. Labbe, K., McIntire, C. R., Doiron, K., Leblanc, P. M. &

    Saleh, M. Cellular inhibitors of apoptosis proteins

    cIAP1 and cIAP2 are required for efficient caspase-1

    activation by the inflammasome. Immunity 35,

    897907 (2011).

    59. Miwa, K. et al. Caspase 1-independent IL-1 releaseand inflammation induced by the apoptosis inducer

    Fas ligand. Nature Med. 4, 12871292 (1998).

    60. Bossaller, L. et al. Cutting edge: FAS (CD95)

    mediates noncanonical IL-1 and IL-18 maturationvia caspase-8 in an RIP3-independent manner.

    J. Immunol. 189, 55085512 (2012).

    61. Lech, M., Avila-Ferrufino, A., Skuginna, V.,

    Susanti, H. E. & Anders, H. J. Quantitative expression

    of RIG-like helicase, NOD-like receptor andinflammasome-related mRNAs in humans and mice.

    Int. Immunol. 22, 717728 (2010).

    62. Juliana, C. et al. Non-transcriptional priming and

    deubiquitination regulate NLRP3 inflammasome

    activation.J. Biol. Chem. 287, 3661736622

    (2012).

    63. Py, B. F., Kim, M. S., Vakifahmetoglu-Norberg, H. &

    Yuan, J. Deubiquitination of NLRP3 by BRCC3

    critically regulates inflammasome activity.

    Mol. Cell49, 331338 (2013).

    64. Schroder, K. et al. Acute lipopolysaccharide priming

    boosts inflammasome activation independently of

    inflammasome sensor induction. Immunobiology 217,

    13251329 (2012).

    65. Burckstummer, T. et al. An orthogonal proteomic-

    genomic screen identifies AIM2 as a cytoplasmic

    DNA sensor for the inflammasome. Nature Immunol.

    10, 266272 (2009).

    66. DeYoung, K. L. et al. Cloning a novel member of the

    human interferon-inducible gene family associated

    with control of tumorigenicity in a model of human

    melanoma. Oncogene 15, 453457 (1997).

    67. Qu, Y. et al. Phosphorylation of NLRC4 is critical for

    inflammasome activation. Nature 490, 539542

    (2012).

    This study decribes the importance of a

    pathogen-induced post-translational modification

    event for the activation of NLRC4.68. Halle, A. et al. The NALP3 inflammasome is involved

    in the innate immune response to amyloid-.Nature Immunol. 9, 857865 (2008).69. Anderson, J. P. et al. Initial description of the human

    NLRP3 promoter. Genes Immun. 9, 721726 (2008).

    70. Guarda, G. et al. T cells dampen innate immune

    responses through inhibition of NLRP1 and NLRP3

    inflammasomes. Nature 460, 269273 (2009).

    71. Mishra, B. B. et al. Nitric oxide controls the

    immunopathology of tuberculosis by inhibiting

    NLRP3 inflammasome-dependent processing of IL-1.Nature Immunol. 14, 5260 (2013).

    72. Guarda, G. et al. Type I interferon inhibits

    interleukin-1 production and inflammasome

    activation. Immunity 34, 213223 (2011).

    73. Hernandez-Cuellar, E. et al. Cutting edge: nitric oxide

    inhibits the NLRP3 inflammasome.J. Immunol. 189,

    51135117 (2012).

    74. Haneklaus, M. et al. Cutting edge: miR-223 and EBV

    miR-BART15 regulate the NLRP3 inflammasome and

    IL-1 production.J. Immunol. 189, 37953799(2012).

    75. Bauernfeind, F. et al. NLRP3 inflammasome activity is

    negatively controlled by miR-223.J. Immunol. 189,

    41754181 (2012).

    76. Hornung, V. et al. Silica crystals and aluminum salts

    activate the NALP3 inflammasome through

    phagosomal destabilization. Nature Immunol. 9,

    847856 (2008).

    77. Cain, K., Langlais, C., Sun, X. M., Brown, D. G. &

    Cohen, G. M. Physiological concentrations of K+

    inhibit cytochrome c-dependent formation of the

    apoptosome.J. Biol. Chem. 276, 4198541990

    (2001).78. Purring-Koch, C. & McLendon, G. Cytochrome c binding

    to Apaf-1: the effects of dATP and ionic strength.

    Proc. Natl Acad. Sci. USA 97, 1192811931 (2000).

    79. Ptrilli, V. et al. Activation of the NALP3

    inflammasome is triggered by low intracellular

    potassium concentration. Cell Death Differ. 14,

    15831589 (2007).

    This study describes the role of K+ in NLRP1 and

    NLRP3 activation.80. Fernandes-Alnemri, T. et al. The AIM2 inflammasome

    is critical for innate immunity to Francisella tularensis.

    Nature Immunol. 11, 385393 (2010).

    81. Arlehamn, C. S., Petrilli, V., Gross, O., Tschopp, J. &

    Evans, T. J. The role of potassium in inflammasome

    activation by bacteria.J. Biol. Chem. 285,

    1050810518 (2010).

    82. Pelegrin, P. & Surprenant, A. Pannexin-1 couples to

    maitotoxin- and nigericin-induced interleukin-1release through a dye uptake-independent pathway.

    J. Biol. Chem. 282, 23862394 (2007).

    83. Pelegrin, P. & Surprenant, A. Pannexin-1 mediates

    large pore formation and interleukin-1 release by theATP-gated P2X7 receptor. EMBO J. 25, 50715082

    (2006).84. Locovei, S., Wang, J. & Dahl, G. Activation of

    pannexin 1 channels by ATP through P2Y receptors

    and by cytoplasmic calcium. FEBS Lett. 580,

    239244 (2006).

    85. Qu, Y. et al. Pannexin-1 is required for ATP releaseduring apoptosis but not for inflammasome activation.

    J. Immunol. 186, 65536561 (2011).86. Compan, V. et al. Cell volume regulation modulates

    NLRP3 inflammasome activation. Immunity 37,

    487500 (2012).

    87. Gong, Y. N. et al. Chemical probing reveals insights

    into the signaling mechanism of inflammasome

    activation. Cell Res. 20, 12891305 (2010).

    88. Zhong, Z. et al. TRPM2 links oxidative stress to

    NLRP3 inflammasome activation. Nature Commun.

    4, 1611 (2013).

    This study links Ca2+ mobilization to oxidative

    stress, both of which are important factors in

    NLRP3 activation.

    89. Murakami, T. et al. Critical role for calcium

    mobilization in activation of the NLRP3

    inflammasome. Proc. Natl Acad. Sci. USA 109,

    1128211287 (2012).

    90. Menu, P. et al. ER stress activates the NLRP3

    inflammasome via an UPR-independent pathway.

    Cell Death Dis. 3, e261 (2012).

    91. Lee, G. S. et al. The calcium-sensing receptor regulates

    the NLRP3 inflammasome through Ca2+ and cAMP.

    Nature 492, 123127 (2012).92. Rossol, M. et al. Extracellular Ca2+ is a danger signal

    activating the NLRP3 inflammasome through

    G protein-coupled calcium sensing receptors.

    Nature Commun. 3, 1329 (2012).

    References 91 and 92 describe that GPCRs,

    which sense extracellular Ca2+

    , activate the NLRP3inflammasome by activating phospholipase C and

    by inhibiting adenylyl cyclase.

    93. Zuo, Y.et al. Oxidative modification of caspase-9

    facilitates its activation via disulfide-mediated

    interaction with Apaf-1. Cell Res. 19, 449457 (2009).

    94. Zech, B., Kohl, R., von Knethen, A. & Brune, B.

    Nitric oxide donors inhibit formation of the Apaf-1/

    caspase-9 apoptosome and activation of caspases.

    Biochem. J. 371, 10551064 (2003).

    95. Kim, Y. M., Talanian, R. V., Li, J. & Billiar, T. R.

    Nitric oxide prevents IL-1 and IFN--inducing factor(IL-18) release from macrophages by inhibiting

    caspase-1 (IL-1-converting enzyme).J. Immunol.161, 41224128 (1998).

    96. Cruz, C. M. et al. ATP activates a reactive oxygen

    species-dependent oxidative stress response and

    secretion of proinflammatory cytokines in

    macrophages.J. Biol. Chem.282, 28712879 (2007).

    97. Dostert, C. et al. Innate immune activation through

    Nalp3 inflammasome sensing of asbestos and silica.

    Science 320, 674677 (2008).

    98. Cassel, S. L. et al. The Nalp3 inflammasome is

    essential for the development of silicosis. Proc. Natl

    Acad. Sci. USA 105, 90359040 (2008).

    99. van de Veerdonk, F. L. et al. Reactive oxygen species-

    independent activation of the IL-1 inflammasome incells from patients with chronic granulomatous disease.

    Proc. Natl Acad. Sci. USA 107, 30303033 (2010).

    100. Meissner, F. et al. Inflammasome activation in

    NADPH oxidase defective mononuclear phagocytes

    from patients with chronic granulomatous disease.

    Blood116, 15701573 (2010).101. van Bruggen, R. et al. Human NLRP3 inflammasome

    activation is Nox14 independent. Blood115,

    53985400 (2010).

    102. Zhou, R., Yazdi, A. S., Menu, P. & Tschopp, J.

    A role for mitochondria in NLRP3 inflammasome

    activation. Nature 469, 221225 (2011).

    103. Bauernfeind, F. et al. Cutting edge: reactive oxygen

    species inhibitors block priming, but not activation,

    of the NLRP3 inflammasome.J. Immunol. 187,613617 (2011).

    104. Gordy, C. & He, Y. W. The crosstalk between

    autophagy and apoptosis: where does this lead?

    Protein Cell3, 1727 (2012).

    105. Saitoh, T. et al. Loss of the autophagy protein

    Atg16L1 enhances endotoxin-induced IL-1production. Nature 456, 264268 (2008).

    This is the first study to describe that autophagy

    limits IL-1 production.106. Shi, C. S. et al. Activation of autophagy by

    inflammatory signals limits IL-1 production bytargeting ubiquitinated inflammasomes for

    destruction. Nature Immunol. 13, 255263 (2012).

    107. Harris, J. et al. Autophagy controls IL-1 secretion bytargeting pro-IL-1 for degradation.J. Biol. Chem.286, 95879597 (2011).

    108. Agostini, L. et al. NALP3 forms an IL-1-processinginflammasome with increased activity in MuckleWells

    autoinflammatory disorder. Immunity 20, 319325

    (2004).109. Pathan, N. et al. TUCAN, an antiapoptotic caspase-

    associated recruitment domain family protein

    overexpressed in cancer.J. Biol. Chem. 276,

    3222032229 (2001).

    110. Saleh, M. et al. Enhanced bacterial clearance and

    sepsis resistance in caspase-12-deficient mice.

    Nature 440, 10641068 (2006).

    111. Saleh, M. et al. Differential modulation of endotoxin

    responsiveness by human caspase-12 polymorphisms.

    Nature 429, 7579 (2004).

    112. Stehlik, C. & Dorfleutner, A. COPs and POPs:

    modulators of inflammasome activity.J. Immunol.

    179, 79937998 (2007).

    113. Druilhe, A. Srinivasula, S. M., Razmara, M.,

    Ahmad, M. & Alnemri, E. S. Regulation of IL-1generation by pseudo-ICE and ICEBERG, two

    dominant negative caspase recruitment domain

    proteins. Cell Death Differ. 8, 649657 (2001).

    R E V I E W S

    410 | JUNE 2013 | VOLUME 13 www.nature.com/reviews/immunol

    2013 Macmillan Publishers Limited. All rights reserved

  • 7/27/2019 In Mun Ology

    23/78

    114. Lamkanfi, M. et al. INCA, a novel human caspase

    recruitment domain protein that inhibits

    interleukin-1 generation.J. Biol. Chem. 279,5172951738 (2004).

    115. Stehlik, C. et al. The PAAD/PYRIN-only protein

    POP1/ASC2 is a modulator of ASC-mediated nuclear-

    factor-B and pro-caspase-1 regulation. Biochem. J.373, 101113 (2003).

    116. Dorfleutner, A. et al. Cellular pyrin domain-only

    protein 2 is a candidate regulator of inflammasome

    activation. Infect. Immun. 75, 14841492 (2007).

    117. Bedoya, F., Sandler, L. L. & Harton, J. A. Pyrin-onlyprotein 2 modulates NF-B and disrupts ASC:CLRinteractions.J. Immunol. 178, 38373845 (2007).

    118. Bryan, N. B. et al. Differential splicing of the

    apoptosis-associated speck like protein containing a

    caspase recruitment domain (ASC) regulates

    inflammasomes.J. Inflamm. 7, 23 (2010).

    119. Imamura, R. et al. Anti-inflammatory activity of

    PYNOD and its mechanism in humans and mice.

    J. Immunol. 184, 58745884 (2010).

    120. Wang, Y. et al. PYNOD, a novel Apaf-1/CED4-like

    protein is an inhibitor of ASC and caspase-1.

    Int. Immunol. 16, 777786 (2004).

    121. Eisenbarth, S. C. et al. NLRP10 is a NOD-like

    receptor essential to initiate adaptive immunity by

    dendritic cells. Nature 484, 510513 (2012).

    122. Joly, S. et al. Cutting edge: Nlrp10 is essential for

    protective antifungal adaptive immunity against

    Candida albicans.J. Immunol. 189, 47134717(2012).

    123. Kinoshita, T., Wang, Y., Hasegawa, M., Imamura, R. &

    Suda, T. PYPAF3, a PYRIN-containing APAF-1-like

    protein, is a feedback regulator of caspase-1-

    dependent interleukin-1 secretion.J. Biol. Chem.280, 2172021725 (2005).

    124. Hesker, P. R., Nguyen, M., Kovarova, M., Ting, J. P. &

    Koller, B. H. Genetic loss of murine pyrin, the familial

    Mediterranean fever protein, increases interleukin-1levels. PLoS ONE7, e51105 (2012).

    125. Chae, J. J. et al. Gain-of-function pyrin mutations

    induce NLRP3 protein-independent interleukin-1activation and severe autoinflammation in mice.

    Immunity 34, 755768 (2011).126. Vyleta, M. L., Wong, J. & Magun, B. E. Suppression of

    ribosomal function triggers innate immune signaling

    through activation of the NLRP3 inflammasome.

    PLoS ONE7, e36044 (2012).

    127. Lu, B. et al. Novel role of PKR in inflammasome

    activation and HMGB1 release. Nature 488,

    670674 (2012).

    128. He, Y., Franchi, L. & Nunez, G. The protein kinase

    PKR is critical for LPS-induced iNOS production butdispensable for inflammasome activation in

    macrophages. Eur. J. Immunol.43, 11471152 (2013).

    129. Mayor, A., Martinon, F., De Smedt, T., Petrilli, V. &

    Tschopp, J. A crucial function of SGT1 and HSP90 in

    inflammasome activity links mammalian and plant

    innate immune responses. Nature Immunol. 8,

    497503 (2007).

    130. Jin, T. et al. Structures of the HIN domain:DNA

    complexes reveal ligand binding and activation

    mechanisms of the AIM2 inflammasome and IFI16

    receptor. Immunity 36, 561571 (2012).

    This is the first crystal structure of an

    inflammasome sensor with its ligand.

    131. DOsualdo, A. et al. CARD8 and NLRP1 undergoautoproteolytic processing through a ZU5-like domain.

    PLoS ONE6, e27396 (2011).

    132. Frew, B. C., Joag, V. R. & Mogridge, J. Proteolytic

    processing of Nlrp1b is required for inflammasome

    activity. PLoS Pathog. 8, e1002659 (2012).

    133. Finger, J. N. et al. Autolytic proteolysis within the

    function to find domain (FIIND) is required for

    NLRP1 inflammasome activity. J. Biol. Chem. 287,

    2503025037 (2012).

    134. Martin, A. G. & Fearnhead, H. O. Apocytochrome c

    blocks caspase-9 activation and Bax-induced

    apoptosis.J. Biol. Chem. 277, 5083450841 (2002).

    135. Nakagawa, H. et al. Nitration of specific tyrosine

    residues of cytochrome c is associated with caspase-

    cascade inactivation. Biol. Pharm. Bull. 30, 1520

    (2007).

    136. Levinsohn, J. L. et al. Anthrax lethal factor cleavage of

    Nlrp1 is required for activation of the inflammasome.

    PLoS Pathog. 8, e1002638 (2012).

    137. Acehan, D. et al. Three-dimensional structure of the

    apoptosome: implications for assembly, procaspase-9

    binding, and activation. Mol. Cell9, 423432

    (2002).

    138. Llambi, F. & Green, D. R. Apoptosis and oncogenesis:

    give and take in the BCL-2 family. Curr. Opin. Genet.

    Dev. 21, 1220 (2011).

    139. Bruey, J. M. et al. Bcl-2 and Bcl-XL regulate

    proinflammatory caspase-1 activation by interaction

    with NALP1. Cell129, 4556 (2007).

    140. Faustin, B. et al. Mechanism of Bcl-2 and Bcl-XL

    inhibition of NLRP1 inflammasome: loop domain-

    dependent suppression of ATP binding and

    oligomerization. Proc. Natl Acad. Sci. USA 106,

    39353940 (2009).

    141. Nunnari, J. & Suomalainen, A. Mitochondria: in

    sickness and in health. Cell148, 11451159 (2012).

    142. Feldman, J. L., Dittenhafer-Reed, K. E. & Denu, J. M.

    Sirtuin catalysis and regulation.J. Biol. Chem. 287,

    4241942427 (2012).143. Misawa, T. et al. Microtubule-driven spatial

    arrangement of mitochondria promotes activation ofthe NLRP3 inflammasome. Nature Immunol. 14,

    454460 (2013).

    144. Subramanian, N., Natarajan, K., Clatworthy, M. R.,

    Wang, Z. & Germain, R. N. The adaptor MAVS

    promotes NLRP3 mitochondrial localization and

    inflammasome activation. Cell153, 348361 (2013).

    145. Tannahill, G. M. et al. Succinate is an inflammatory

    signal that induces IL-1 through HIF-1. Nature 496,238242 (2013).

    146. Kim, H. E., Du, F., Fang, M. & Wang, X. Formation of

    apoptosome is initiated by cytochrome c-induced

    dATP hydrolysis and subsequent nucleotide exchange

    on Apaf-1. Proc. Natl Acad. Sci. USA 102,

    1754517550 (2005).147. Duncan, J. A. et al. Cryopyrin/NALP3 binds ATP/dATP,

    is an ATPase, and requires ATP binding to mediate

    inflammatory signaling. Proc. Natl Acad. Sci. USA

    104, 80418046 (2007).

    148. Yuan, S. et al. Structure of an apoptosome-

    procaspase-9 CARD complex. Structure 18, 571583

    (2010).

    149. Qi, S. et al. Crystal structure of the Caenorhabditis

    elegans apoptosome reveals an octameric assembly

    of CED-4. Cell141, 446457 (2010).

    150. Jha, S. et al. The inflammasome sensor, NLRP3,

    regulates CNS inflammation and demyelination via

    caspase-1 and interleukin-18.J. Neurosci. 30,

    1581115820 (2010).

    151. Inoue, M. et al. Interferon- therapy against EAE iseffective only when development of the disease

    depends on the NLRP3 inflammasome. Sci. Signal.

    5,ra38 (2012).152. Lamkanfi, M. et al. Glyburide inhibits the cryopyrin/

    Nalp3 inflammasome.J. Cell Biol. 187, 6170 (2009).

    153. Juliana, C. et al. Anti-inflammatory compounds

    parthenolide and Bay 117082 are direct inhibitors of

    the inflammasome.J. Biol. Chem. 285, 97929802

    (2010).

    AcknowledgementsThe authors would like to thank C. M. De Nardo and B. G.

    Monks for critical reading of the manuscript. This work was

    supported by grants from the US National Institutes of

    Health (NIH) and the Deutsche Forschungsgemeinschaft

    (DFG), Germany (to E.L.), and by the Division of Intramural

    Research, National Institute of Allergy and Infectious

    Diseases, NIH, USA (to T.S.X.). E.L. is a member of the

    excellence cluster ImmunoSensation in Bonn, Germany.

    Competing interests statementThe authors declare no competing financial interests.

    FURTHER INFORMATION

    Eicke Latzs homepage: http://www.iii.uni-bonn.de/

    ALL LINKS ARE ACTIVE IN THE ONLINE PDF

    R E V I E W S

    NATURE REVIEWS |IMMUNOLOGY VOLUME 13 | JUNE 2013 |411

    2013 Macmillan Publishers Limited. All rights reserved

    http://www.iii.uni-bonn.de/http://www.iii.uni-bonn.de/http://www.iii.uni-bonn.de/
  • 7/27/2019 In Mun Ology

    24/78

  • 7/27/2019 In Mun Ology

    25/78

    on the basis of their surface phenotype. Consequently, inboth humans and mice810,1417, T

    FHcells are considered to

    be CD4+ T cells that express the highest levels of CXCR5,together with the surface receptors inducible T cell costimulator (ICOS) and programmed cell death protein 1(PD1; also known as PDCD1), the transcriptional repressor B cell lymphoma 6 (BCL6) and the cytokine IL21(BOX 3), and that have downregulated the T cell zonehoming receptor CCchemokine receptor 7 (CCR7) andIL7 receptor (IL7R) (TABLE 1).

    This phenotypic approach to defining TFH

    cells hasfacilitated their molecular and cellular characterization.However, it needs to be appreciated that, just as B cellsundergo important differentiation events at the T cellB cell border (such as the formation of extrafollicular

    plasmablasts) and in GCs (such as the formation ofmemory and plasma cells), CD4+ T cells can differentiate

    into TFH cell subsets that are strategically located at theseregions to facilitate distinct phases of a T celldependentB cell response6,7,18,19. These T

    FHcell subsets probably pro

    vide early B cell help at the T cellB cell border, and/orthey represent precursor cells that differentiate into GCT

    FHcells following receipt of appropriate signals from

    inside the active B cell follicle and that guide the differentiation of GC B cells into memory or plasma cells.In this Review, we discuss recent developments in theinvestigation of the mechanisms that underlie T

    FHcell

    development and function, the discovery of specializedsubsets of T

    FHcells and how perturbations to T

    FHcells

    potentially contribute to numerous human diseases.

    Table 1 | Effector subsets of CD4+ T cells: ontogenic and functional requirements, and roles in disease

    CD4+T cell

    subset

    Inducingcytokines

    ActivatedSTATs

    Transcriptionfactors

    Suppressingcytokines

    Canonicalcytokinesproduced

    Roles in hostprotection

    Associatedpathologies

    Refs

    TH1 cells IL-12

    IFNSTAT4STAT1

    T-bet IL-4 and IL-10 IFN Antiviral andantimicrobialimmunity

    Cell-mediatedimmunity

    Mendeliansusceptibility tomycobacterialdisease (decrease inT

    H1 cells)

    Multiple sclerosis(increase in T

    H1 cells)

    1,121,122

    TH2 cells IL-4 STAT6 GATA3 and

    MAFIFN IL-4, IL-5

    and IL-13Immunity to

    extracellular parasitesAllergy, asthma or

    eczema (increase inT

    H2 cells)

    1,123

    TH17 cells IL-23 and

    IL-1IL-6 and

    IL-1TGF

    STAT3 RORt andROR

    IL-4, IFN,IL-27 andIL-2

    TGF(suppressesIL-22expression)

    IL-17A,IL-17F,IL-21, IL-22and IL-26*

    Protection atmucocutaneous sites

    Antimicrobialimmunity (forexample, againstCandida spp. andStaphylococcusspp.)

    Inflammatory boweldisease

    Inflammatory boweldisease (increase inT

    H17 cells)

    Susceptibility tofungal infections(decrease in T

    H17

    cells)

    1,121,122,124

    TH9 cells TGF

    IL-4STAT6 PU-1 and

    IRF4IFN andIL-27

    IL-9 Protection againsthelminth infections

    Allergy (atopicdermatitis) andasthma (increase inT

    H9 cells)

    125

    TH22 cells TNF

    IL-6STAT1STAT3STAT5

    RORt andAHR

    High doses ofTGF

    IL-22 Barrier immunity(skin, intestines andairways)

    Enhancement ofinnate immunity

    Tissue regeneration

    Allergy (atopicdermatitis) (increasein T

    H22 cells)

    Inflammation atjoints and barriers(increase in T

    H22

    cells in mice)

    126

    TReg

    cells TGF andIL-2

    STAT5 FOXP3 IL-6 TGF andIL-10

    Immune suppression IPEX syndrome(decrease in T

    Reg

    cells)

    75

    TFHcells IL-6, IL-21and/or IL-27IL-12

    STAT3STAT4STAT1

    BCL-6,IRF4, MAF andBATF

    IL-2 and IL-10 IL-21, IL-4and IL-10 Help for B cellactivation ordifferentiation

    Generation oflong-lived antibodyresponses

    Humoralimmunodeficiency(decrease in T

    FHcells)

    Autoimmunity(increase in T

    FHcells)

    T cell lymphoma

    (increase in TFH

    cells)(see TABLE 2)

    2325,45

    AHR, aryl hydrocarbon receptor; BATF, basic leukine zipper transcriptional factor ATF-like; BCL-6, B cell lymphoma 6; FOXP3, forkhead box P3; GATA3,GATA-binding protein 3; IFN, interferon-; IL, interleukin; IPEX, immunodysregulation, polyendocrinopathy and enteropathy X-linked; IRF4, interferon-regulatoryfactor 4; ROR, retinoid-related orphan receptor; STAT, signal transducer and activator of transcription; TGF, transforming growth factor-; T

    FH, T follicular helper;

    TH, T helper; TNF, tumour necrosis factor; T

    Reg, T regulatory. *Human-specific cytokine. Reported in mice.

    R E V I E W S

    NATURE REVIEWS |IMMUNOLOGY VOLUME 13 | JUNE 2013 |413

    2013 Macmillan Publishers Limited. All rights reserved

  • 7/27/2019 In Mun Ology

    26/78

    B cell lymphoma 6

    (BCL-6). A transcriptional

    repressor identified as beingcrucial for the formation of

    T follicular helper (TFH

    ) cells.

    Several mechanisms have been

    proposed for the role of BCL-6

    in TFH

    cell commitment,

    including suppression of the

    expression of transcription

    factors that are required for the

    generation of alternative TH

    fates, suppression of

    microRNAs and cooperation

    with other transcriptional

    regulators to induce the

    expression of important TFH

    cell-related genes.

    Requirements for TFH

    cell formation

    TFH

    cell differentiation requires input from several surfacereceptors (including CD28, ICOS, CD40 ligand (CD40L)and signalling lymphocytic activation molecule (SLAM)family members), as well as from cytokines and theirassociated signalling pathways (for example, signal transducer and activator of transcription 3 (STAT3) or SLAMassociated protein (SAP; also known as SH2D1A)), whichall culminate in the induction of BCL6, an important

    regulator of the TFH cell lineage2022. However, additionaltranscription factors (including interferonregulatoryfactor 4 (IRF4), basic leucine zipper transcriptional factorATFlike (BATF) and MAF) and microRNAs also haveimportant regulatory functions during T

    FHcell develop

    ment2225 (FIG. 1; TABLE 1; Supplementary information S1(table)). The requirements for T

    FHcell formation have

    been extensively reviewed in the literature2326 and aresummarized in Supplementary information S1 (table). Inthis Review, we discuss the most recent studies that haveidentified pathways that positively and negatively influence T

    FHcell generation and function, and that clarify

    previous inconsistencies.

    The TNF receptor superfamily. B cellactivating factor(BAFF; also known as TNFSF13B) and a proliferationinducing ligand (APRIL; also known as TNFSF13) areligands of the tumour necrosis factor (TNF) superfamilythat regulate B cell survival and differentiation11. Bothligands bind to the common receptors transmembraneactivator and CAML interactor (TACI; also known asTNFRSF13B) and B cell maturation antigen (BCMA;also known as TNFRSF17), and BAFF also binds to the

    BAFF receptor (BAFFR; also known as TNFRSF13C);receptors which are predominantly expressed onB cells11. NFBinducing kinase (NIK; also known asMAP3K14) is a component of the BAFFR signallingpathway11. Expression of NIK and BAFFR by B cells wasfound to be required for their constitutive expression ofICOS ligand (ICOSL). The significance of this is thatNIK deficiency in B cells compromises T

    FHcell induc

    tion27. Thus, sustained BAFFBAFFRNIK signalling inB cells maintains ICOSL expression, thereby facilitatingcognate ICOSICOSL interactions between activatedCD4+ T cells and B cells, which result in optimal T

    FHcell

    differentiation.

    Box 1 | TFH

    cells and affinity maturation of GC B cells

    Affinitymaturationoftheantibody

    responseisbasedontheselective

    perpetuation(knownaspositive

    selection)ofgerminalcentre(GC)B cells

    thathaveacquiredincreasedaffinityfor

    foreignantigensthroughsomatic

    hypermutation(SHM)oftheirimmunoglobulinvariable-regiongenes.

    WhereasSHMitselftakesplaceinB cells

    thatoccupytheGCdarkzone,

    high-affinityGCB cellsonlyundergo

    positiveselectionafterreturningtothe

    lightzonewheretheygainpreferential

    accesstoforeignantigensthatare

    expressedonthesurfaceoffollicular

    dendriticcells(seethefigure).However,

    theprecisemechanismsbywhich

    preferentialaccesstoantigenstranslates

    intopositiveselectionofhigh-affinityGC

    B cellshavenotbeenclearlyestablished.

    ThespecificprovisionofTfollicular

    helper(TFH)cell-derivedhelpersignalstohigh-affinityGCB cellsisproposedtobe

    oneof,ifnottheonly,majordriversof

    antibodyaffinitymaturation12,13.In

    theory,thegreaterpropensityof

    high-affinityGCB cellstoaccessforeign

    antigensshouldaugmenttheirabilityto

    internalize,processandpresentforeignpeptidestotheTFH

    cellsthatresideinthelightzoneofGCs(seethefigure).

    However,althoughitisclearthatTFH

    cellsarerequiredtosupporttheGCresponse,ithasbeendifficulttoestablishtheir

    preciseroleindrivingaffinitymaturation.Inarecentstudy,aGCresponsewasmanipulatedinmicesuchthatantigen

    presentationbyGCB cellstoTFH

    cellswasdecoupledfromB cellreceptor(BCR)-mediatedantigenrecognition.This

    resultedinthedeliveryofTFH

    cellhelptoallGCB cellsandtheproliferativeexpansionofthepopulationofTFH

    cells

    regardlessoftheiraffinityforantigen12.ThisresultdoesnotprovethatpreferentialdeliveryofTFH

    cellhelpisthe

    fundamentaldriverofaffinitymaturationbutitclearlydemonstratesthatTFH

    cellshavethepotentialtoperformthis

    roleifhigh-affinityGCB cellsareindeedsuperioratpresentingantigentoTFH

    cells.

    CD40L,CD40ligand;CR2,complementreceptor2;FcRII,lowaffinityFcreceptorforimmunoglobulin;Ig,immunoglobulin;

    TCR,Tcellreceptor.

    Folliculardendritic cell

    Germinal centre

    BCR

    Cell death

    Affinity-maturedantibodies

    High affinity

    B cell TFH cell

    BCR

    CR2 or FcRII

    Ig

    Foreign antigen

    CD40LCD40

    Adhesionmolecules

    TCR

    MHC class IIpeptide complex

    B cell

    Low affinity

    R E V I E W S

    414 | JUNE 2013 | VOLUME 13 www.nature.com/reviews/immunol

    2013 Macmillan Publishers Limited. All rights reserved

    http://www.nature.com/nri/journal/v13/n6/suppinfo/nri3447.htmlhttp://www.nature.com/nri/journal/v13/n6/suppinfo/nri3447.html
  • 7/27/2019 In Mun Ology

    27/78

  • 7/27/2019 In Mun Ology

    28/78

    SH2 domaincontaining

    protein tyrosine

    phosphatase 1

    (SHP1). A protein tyrosine

    phosphatase that is involved in

    suppressing intracellular

    signals delivered via numerous

    activating receptors, including

    T cell and B cell antigen

    receptors, as well as members

    of the signalling lymphocytic

    activation molecule (SLAM)

    family of surface receptors.

    One proposed mechanism of

    action is the direct or indirect

    dephosphorylation of

    components of the T cell

    receptor signalling pathway,

    such as CD3, LCK,

    -chain-associated protein

    kinase of 70 kDa (ZAP70) and

    phosphoinositide 3-kinase.

    Follicular T regulatory cells

    A subset of T regulatory (TReg

    )

    cells that co-opts the

    transcriptional machinery of

    T follicular helper (TFH

    ) cells

    to facilitate their migration to

    germinal centres, where they

    can appropriately restrain

    humoral immune responses,thereby potentially preventing

    overzealous antibody

    responses. Follicular TReg

    cells

    can be identified by the

    expression of typical

    TFH

    cell surface markers

    (CXC-chemokine receptor 5

    (CXCR5), inducible T cell

    co-stimulator (ICOS) and

    programmed cell death

    protein 1 (PD1)) along with

    the TReg

    transcription factor

    forkhead box P3. Their

    mechanism of action remains

    to be completely elucidated.

    SAP) have poor humoral immunity31. A role for CD84 inSAPdependent T

    FHcell generation following immuniza

    tion with protein antigen was recently demonstrated33.However, the T

    FHcell deficiency in Cd84/ mice was

    less severe than in SAPdeficient mice33, and TFH

    cellformation following viral infection was unaffected bythe absence of CD84 (REF. 35).

    Analysis of genetargeted mice has failed to showa requirement for SLAM family receptors other thanCD84 in T

    FHcell formation (reviewed in REF. 23),

    although SLAM has been shown to be required for IL4expression by GC T

    FHcells18. One interpretation of these

    observations is that the severe effect of SAP deficiency inT

    FHcells reflects a requirement for numerous SLAM

    receptors during the differentiation of TFH

    cells.Alternatively, as SLAM receptors can also recruitinhibitors of signalling (such as lipid and tyrosine phosphatases), SAP deficiency might exacerbate negativesignals that are delivered through one or more of theSLAM receptors. Consistent with this idea, loss of LY108reversed the inability of SAPdeficient CD4+ T cells toform T

    FHcells and to support B cell responses35. This

    was due to a reduced recruitment ofSH2 domain-containingprotein tyrosine phosphatase 1 (SHP1; also known asPTPN6) to the immune synapse by LY108 (REF. 35). Thus,LY108 functions as a rheostat that is capable of deliveringpositive SAPdependent and negative SHP1dependent

    signals that dynamically regulate TFH cells (FIG. 1).

    The role of PD1. An important phenotypic determinant of T

    FHcells is their high expression of PD1

    (REF. 14). PD1 has an inhibitory role in TFH

    cell differentiation, as mice with impaired PD1 function have moreT

    FHcells (CCR7lowICOShi cells in Pd1/ mice) as a result

    of increased proliferation and reduced apoptosis3639.Expression of PD1 ligand 1 (PDL1; also known asCD274), rather than PDL2, on B cells constrains T

    FHcell

    formation via the PD1 pathway38(FIG. 1). Although thesestudies showed that ablating PD1 signalling increasedT

    FHcell numbers, they yielded conflicting results about

    how this affected the outcome of the GC response3639.For instance, some groups reported increased antigenspecific antibody responses in PDL1deficient mice38or in Plasmodiuminfected mice that had been treatedwith a PDL1specific mAb39. These findings in miceare consistent with data demonstrating that ligation ofPD1 suppresses the proliferation, activation and function of human T

    FHcells in vitro40. However, other groups

    have reported impaired plasma cell and GC responsesin the absence of PD1 signalling 36,37. These differencesmight reflect nuances in the experimental systems thathave been used, but they might also result from distinct functions of PD1 in the development and functionof not only T

    FHcells but also follicular T regulatory cells

    (follicular TReg

    cells), which express higher levels of PD1than T

    FHcells41.

    Cytokines. Numerous cytokines have been shownto be important for T

    FHcell generation in vivo and

    in vitro. The initial cytokines that were identified toinduce T

    FHcelllike features in cultured CD4+ T cells

    were IL6 and IL21 (TABLE 1). However, subsequentanalyses of IL6 and IL21 or IL21Rdeficient miceyielded conflicting results regarding the necessity ofthese cytokines in regulating T

    FHcell formation in vivo

    (reviewed in REFS 23,24). Furthermore, recent studieshave provided greater insights into the roles of these

    cytokines in TFH cell commitment. Using several mousemodels of viral infection, investigators found varying and transient degrees of impairment in T

    FHcell

    numbers in the absence of IL6, but they observedconsistently reduced levels of virusspecific IgG4245.A more severe decrease in T

    FHcell formation and pro

    tective IgG production occurred in the absence of bothIL6 and IL21 (REFS 42,44)(FIG. 1). This requirementfor IL6 and IL21 is consistent with reductions in thenumber of T

    FHcells in the absence of functional STAT3

    (REFS 30,46), which acts downstream of both cytokines(TABLE 1). Interestingly, the early reduction in the number of mouse T

    FHcells in the absence of STAT3 was

    Box 3 | IL21 is a potent differentiation factor for human B cells

    Interleukin-21(IL-21)wasdiscoveredin2000asapleiotropiccytokinethatiscapableofactivatingmostlymphocyte

    populations116.TheinitialdescriptionreportedthatIL-21stronglyinducedtheproliferationofCD40-stimulatedhuman

    B cells,butthatitinhibitedIL-4-inducedBcellproliferation116.Sincethen,manystudieshaveestablishedthepotency

    ofIL-21asagrowthanddifferentiationfactorforhumanB cells.WhenhumanB cellsareprimedwithTcellhelpinthe

    formofaCD40-specificmonoclonalantibodyorCD40ligand(CD40L),IL-21inducesrobustBcellproliferationaswell

    astheexpressionofactivation-inducedcytidinedeaminase( AICDA;requiredforimmunoglobulinclassswitching),

    Blymphocyte-inducedmaturationprotein1( BLIMP1;alsoknownasPRDM1)andXbox-bindingprotein1(XBP1)83,117,118

    ,allofwhichmediatethedifferentiationofB cellsintoplasmacells.Consequently,atleastin vitro,IL-21efficiently

    inducessubsetsofactivatedB cellstoundergoclassswitchingeithertobecomeIgG-orIgA-expressingcells,orto

    becomeplasmablastssecretingIgM,IgG,IgAorIgE83,117120.IL-21predominantlyinducesswitchingtoIgG3,IgG1

    andIgA1subclasses117,119,120,whereasIL-21-stimulatednaive,GCormemoryB cellsproducelargequantitiesof

    immunoglobulins83,117,119.IL-21canalsoinducetheexpressionofBcelllymphoma6(BCL6)inhumannaiveB cells117,

    whichisconsistentwithIL-21havingaroleinestablishingGCs.BeforethediscoveryofIL-21,itwaswellrecognizedthat

    classswitchingbyhumanB cellswasregulatedbyIL-4(whichpromotesclassswitchingtoIgG4andIgE),IL-10(which

    promotesclassswitchingtoIgG1andIgG3),IL-13(whichpromotesclassswitchingtoIgG4andIgE)andtransforming

    growthfactor-(TGF; whichpromotesclassswitchingtoIgA),andIL-10wasalsoconsideredtobeastronginducerofimmunoglobulinsecretion.Furthermore,B cellsurvivalwasshowntobepositivelyregulatedbyIL-4orIL-10(reviewed

    inREF. 82).StudiesoverthepastdecadehavehighlightedtheimportanceofIL-21inhumoralimmunityinhumansby

    demonstratingthatithastheremarkableabilitytoexertallofthesefunctionsonhumanB cells.

    R E V I E W S

    416 | JUNE 2013 | VOLUME 13 www.nature.com/reviews/immunol

    2013 Macmillan Publishers Limited. All rights reserved

  • 7/27/2019 In Mun Ology

    29/78

  • 7/27/2019 In Mun Ology

    30/78

  • 7/27/2019 In Mun Ology

    31/78

    T cells

    T cells that express the T cell

    receptor. These T cells are

    present in the skin, vagina

    and intestinal epithelium as

    intraepithelial lymphocytes.

    Although the exact function of

    T cells is unknown, it has

    been suggested that mucosal

    T cells are involved in innate

    immune responses.

    Recent studies have reassessed the nature of NKTcells that provide B cell help. It was shown that following immunization with GalCerantigen conjugates,a small proportion of mouse NKT cells acquired aT

    FHlike phenotype62,63. These NKT follicular helper

    (NKTFH

    ) cells were detected in human tonsils62 andtheir development depended on the same factors thatare important for conventional T

    FHcells62. By produc

    ing IL21, NKTFH

    cells supported the rapid formation ofGCs, yielding detectable levels of antigenspecific IgG,with some evidence of affinity maturation6264(FIG. 2).

    Although NKTFH

    cells resembled TFH

    cells, themost striking difference was their inability to invokelonglived memory responses to lipid antigens (FIG. 2).Furthermore, the magnitude of the NKT

    FHcellinduced

    antibody responses to lipid antigens was inferior tothose driven by conventional T

    FHcells6264. Despite this,

    administering GalCer as an adjuvant or as a conjugated component of immunizing antigens increasedthe production of antigenspecific antibodies. Thisprobably reflects the direct actions of NKT

    FHcells on

    antigenspecific CD1d+ B cells, as well as the indirectactions of NKT

    FHcells, such as cytokine production, on

    other cell types5861. It might also indicate that there issynergy between NKT

    FHand T

    FHcells. Taken together,

    these findings support the inclusion of GalCer invaccine adjuvants.

    TFH

    cells.The finding that mice and humans lacking the conventional TCR have T

    Hcells that elicit

    humoral responses against T celldependent antigensled to the realization that T cells can provide helpto B cells to generate GCs65. Similar to conventionalT

    FHcells, some human T

    FHcells express CXCR5

    and localize to follicles and GCs6668. T cells expressa semiinvariant TCR repertoire and recognize nonpeptidic phosphoantigens that are derived from microbial metabolites65. These antigens rapidly activate T cells, which can subsequently acquire T

    FHcell fea

    tures66,68,69; the differentiation of T cells to TFH

    likecells is increased by exogenous IL21 (REFS 68,69) (FIG. 2).As T cells do not produce IL21 (REFS 68,69), theyare dependent on extrinsic sources of this cytokine todifferentiate into T

    FHlike cells.

    As conventional CD4+ T cells, T cells and NKTcells recognize different repertoires of microbial antigens, their ability to differentiate into T

    FHlike effec

    tor cells provides a mechanism whereby humoral

    immunity can be generated against a broad range ofpathogenassociated antigens (FIG. 2). This expandsthe number of molecular targets that initiate protective immunity, but it might also contribute to postinfection autoimmunity by generating crossreactiveautoantibodies13,70(BOX 2).

    Follicular TReg

    cells. Recent studies have proposed thatT

    FHcells are controlled by follicular T

    Regcells a spe

    cialized subset of TReg

    cells that colocalize within B cellfollicles. Follicular T

    Reglike cells were first described

    in human tonsils in 2004 (REF. 71), but it took another78 years for them to be examined in greater detail.

    Follicular TReg

    cells comprise approximately 1015%of the T

    FHcell population in human and murine lym

    phoid tissues7274. They show characteristics of bothT

    FHcells and T

    Regcells, but they lack expression of CD40L,

    IL4 and IL21 (REF. 73). Abrogating either follicular TReg

    cell development or their follicular localizati