inmunologia_de_la_piel__br_j_dermatol._2007_dec_157_suppl_2_1_7[1]

Upload: matrixx15

Post on 06-Apr-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/3/2019 inmunologia_de_la_piel__Br_J_Dermatol._2007_Dec_157_Suppl_2_1_7[1]

    1/7

    D O I 1 0 . 1 1 1 1 / j . 1 3 6 5- 2 1 3 3 . 2 0 0 7 . 0 8 2 6 4. x

    Immune response profiles in human skinT. Meyer, * E. Stockfleth and E. Christophers

    *Institute for Immunology, Clinical Pathology and Molecular Medicine, Lademannbogen 61, 22339 Hamburg, Germany

    Department of Dermatology, University of Berlin, HTCC, Chariteplatz 1, 10117 Berlin, Germany

    Department of Dermatology, University of Kiel, Schittenhelmstrae 7, 24105 Kiel, Germany

    CorrespondenceThomas Meyer.

    E-mail: [email protected]

    Accepted for publication4 September 2007

    Key wordsadaptive immunity, epithelial defense, innate

    immunity, TH17 cell, toll-like receptor

    Conflicts of interestES acts as a consultant to Meda Pharma. TM and

    EC declare no conflicts of interest.

    Summary

    In addition to the function as a physical barrier human skin has been shown to

    be an important immune organ displaying various defense mechanisms, which

    can be divided into three major functional compartiments: (i) Epithelial defense,

    which is characterized by antimicrobial peptides and proteins (AP) and which

    can be induced in inflammatory lesions but also in the absence of inflammation.

    (ii) Innate-inflammatory immunity, which involves recognition of microbial

    compounds by particular receptors like Toll-like receptors (TLR) and subsequent

    activation of signalling pathways resulting in expression of pro-inflammatory

    cytokines and interferons, as well as genes of adaptive immunity. Interferon a

    (IFNa) produced by plasmacytoid dendritic cells (DC) may stimulate myeloid DC

    to produce IL-12 resulting in classical T-cell activation or to produce IL-23 acti-

    vating IL-17 producing T-cells (IL-23IL-17 pathway). (iii) Adaptive immunity,

    which is based on antigen presenting cells, T-cells and B-cells and which is char-

    acterized by specificity and memory. In contrast to epithelial defense and innate-

    inflammatory immunity, adaptive immune functions provide slowly reacting pro-

    tection. Recent improvements of our knowledge of dysregulated immune path-

    ways associated with inflammatory skin diseases represent an important basis of

    novel immunomodulatory treatment modalities.

    With a surface area of approximately 17.000 cm2 human skin

    is one of the largest organs of the body. Located at the inter-

    face between environment and living organ systems skin

    serves a variety of functions. These include

    1 maintaining an effective barrier against loss of body fluids

    and protection against chemical and physical injury,

    2 protection against UV injury together with adequate radia-

    tion damage repair and

    3 the capacity for rapid wound healing along with a powerful

    defense armentarium against invading microorganisms.

    Human skin is covered by the epidermis, which partly con-

    sists of 1720 layers of coherent, flattened cells, the Stratum

    corneum.1,2 As skin is constantly exposed to the outside world

    physical injury is among the most common threats eventually

    leading toward lethal outcomes. Thus, during phylogeny avariety of defense systems has emerged which actively serve to

    maintain cutaneous integrity and immunity. Players participat-

    ing in this scenario consist of protective antimicrobial peptides

    (APs) as well as inflammatory cells and signal substances

    in conjunction with sessile skin cells forming a skin immune

    armentarium. Functionally this consists of three major

    compartments:

    1 Epithelial defense

    2 Innate-inflammatory immunity and

    3 Antigen-elicited, adaptive immunity.

    Epithelial defense has only recently been discovered in

    human skin3 whereas the leukocyte driven inflammatory

    response has been known since a long time as an innate (pre-

    viously called nonspecific) arm.4

    The third arm (adaptive immunity) has by far been given

    greatest attention in medicine. With specificity and memory as

    outstanding features this system provides powerful, however,

    slowly reacting protection of unlimited diversity. In addition,

    adaptive immunity is of principal importance in immune sur-

    veillance against viruses and transformed cells. On the other

    hand in humans this (adaptive) skin immune system5

    appears responsible for a greater number of diseases than any

    of the other two defense systems.

    In the following some aspects of this skin immune system

    will be briefly discussed.

    Epithelial defense

    Chemical protection by peptides and proteins has first been

    observed in insects (Drosophila) and plants.6,7 The first antimi-

    crobial peptide (AP) found in human skin was Lysozyme.8

    At present about ten APs have been detected in human skin

    (Table 1) and more of these compounds will be discovered.

    RNAse 7 and psoriasin (S100A7) represent constitutively

    expressed APs, whereas human beta defensin (hBD) 24 as

    2007 The Authors

    Journal Compilation 2007 British Association of Dermatologists British Journal of Dermatology 2007, 157 (Suppl. 2), 17 1

  • 8/3/2019 inmunologia_de_la_piel__Br_J_Dermatol._2007_Dec_157_Suppl_2_1_7[1]

    2/7

    well as LL-37 are inducible. As skin is practically soaked with

    (mostly keratinocyte derived) defense molecules (within

    microgram range per g tissue)4 their immediate availability

    provides a powerful chemical shield for instance in wounding.

    In fact most of the peptides and proteins are discovered in

    inflamed skin, notably in psoriatic scales.9

    Interestingly, only few of the APs exhibit broad antimicro-

    bial activities (e.g. hBD3), instead the majority is directed

    against either gram-negative (e.g. hBD1, hBD2) or gram-posi-

    tive bacteria or fungi (Lysozyme, RNAse7, CAP 18LL-37). Adegree of specificity is also seen with psoriasin which primar-

    ily acts against E. coli.10

    Principal producers of APs are keratinocytes. They respond

    to a variety of stimuli including IL-1, TNFa (Tumour necrosis

    factor alpha), IGF-1 (Insulin-like growth factor 1) as well as

    bacterial membrane substances.3 Recently, IL-22 has been

    detected as potent cytokine stimulating keratinocytes to pro-

    duce defense molecules, proteases and growth factors (see

    below).

    While regulatory pathways have become better under-

    stood,3 so far receptors for AP induction have been hiding

    from detection. One may assume that receptors different from

    known innate receptors e.g. Toll-like receptors (TLRs)11 are

    active. So far studies have shown induction of hBD2 via

    TLR4.12

    In certain situations APs may participate in the adaptive

    immune response by serving as chemoattractants for immature

    dendritic cells (DC) and memory T cells (seen with hBD2 and

    hBD3) as well as neutrophils, T cells and monocytes (seen

    with LL-37).13,14

    Innate (inflammatory) immune response

    In order to elicit inflammatory responses innate immunity

    employs germline-encoded pattern recognition receptors to

    recognize microbial compounds. TLRs have been shown to act

    as primary sensors by binding of particular structures present

    on bacteria, viruses, fungi as well as protozoa [also called

    pathogen associated molecular patterns (PAMPs)15]. There are

    10 members of the TLR family in humans (Table 2).

    Organ systems exposed to environmental influences e.g.

    skin, lung, and intestines show different colonisation with

    microbial symbionts. In consequence, gut epithelium as com-

    pared to skin or lung shows different defense strategies in

    host-bacterial interaction.16 For these nonsterile organs with

    permanent contact to micro-organisms it is important to pre-

    vent constant activation of innate immune functions, which

    would potentially compromise organ function. To maintainhomeostasis several regulatory mechanisms have evolved that

    control innate immunity. Due to the heterogeneity of micro-

    bial colonization homeostasis is achieved through different,

    organ-specific mechanisms involving TLRs. This includes dif-

    ferences in TLR activation thresholds, tolerance, and varying

    receptor profiles in distinct organs, as well as different expres-

    sion of negative regulators of TLR signalling and environmen-

    tal molecules regulating TLR function (e.g. IFNa, IL-10 or

    TGFb [Transforming growth factor beta]).16

    Binding of PAMPs to TLRs reflects early infection with acti-

    vation of cellular signalling pathways including transcription

    factor NFkB (Nuclear factor kappa B) and activation of MAP

    (mitogen-activated protein) kinases p38 and JNK (jun amino

    terminal kinase). These factors regulate expression of many

    genes involved in inflammation and immunity. Thus, pro-

    inflammatory cytokines and interferons, but also adaptive

    immune functions such as MHC (major histocompatibility

    Table 1 Antimicrobial peptides in human skin

    Lysozyme

    Human beta defensin 1 (hBD1)

    hBD 24

    RNAse 7, 8

    Psoriasin

    CAP 18 LL-37

    Dermcidin

    Table 2 Toll-like receptors and their ligands

    Receptor Ligands

    TLR 1 Triacylated lipopeptidesa

    TLR 2 Peptidoglycan, bacterial lipoprotein, zymosan, lipoteichoic acid, LPS

    (Porphyromonas gingivalis, Leptospira interrogans), GPI-anchor proteins (Trypanosoma cruzi.)

    TLR 3 ds RNA

    TLR 4 LPS (gram-negative bacteria), F-Protein (RSV), Hsp60, Fibronectin domain ATLR 5 Flagellin

    TLR 6 MALP-2 (Mycoplsma)b, phenol-soluble modulin (Staphylococcus epidermidis)b

    TLR 7 ssRNA, Loxoribine, Bropirimine, Guanosine analogs, Imiquimod, Resiquimod

    TLR 8 ss RNA, Resiquimod, Loxoribine, Bropirimine

    TLR 9 Unmethylated CpG-DNA (bacteria and viruses)

    TLR10 No ligands found yet

    LPS, Lipopolysaccharid; GPI, glycosylphosphatidylinositol; MALP, macrophage activating

    lipopeptide.aLigands recognized by TLR1 + TLR2bLigands recognized by TLR2 + TLR6

    2007 The Authors

    Journal Compilation 2007 British Association of Dermatologists British Journal of Dermatology 2007, 157 (Suppl. 2), 17

    2 Immune response profiles in human skin, T. Meyer et al.

  • 8/3/2019 inmunologia_de_la_piel__Br_J_Dermatol._2007_Dec_157_Suppl_2_1_7[1]

    3/7

    complex)-proteins, co-stimulatory signals and adhesion mole-

    cules become activated.17,18

    Furthermore, signal transduction induced upon ligand bind-

    ing to TLR favours a complex interplay of a growing number

    of adaptor proteins.19 MyD88 was the first adaptor molecule

    identified. TLR and MyD88 associate via common Toll-IL1-

    receptor domain, which results in recruitment of kinases

    (IRAK1 and IRAK4). Phosphorylated IRAK1 then leads to acti-

    vation of protein kinase TAK1 (TGF-activated kinase) which

    finally liberates NFkB from the Inhibitor of NFkB kinase com-

    plex. This pathway of NFkB activation has been seen with

    IL1R, but also represents the main pathway of TLR signalling,

    except for TLR3.

    Next to MyD88 several other adaptor proteins like MyD88

    adaptor-like, TIR-related adaptor protein inducing interferon

    (TRIF) and TRIF-related adaptor molecule have been identified

    to be involved in TLR signalling. Importantly, different TLRs

    signal through different combinations of adaptors, resulting in

    activation of different transcription factors and diverse gene

    induction (see Fig. 1).

    Within the context of this report it is of relevance that

    ligand binding to intracellular receptors TLR7 and TLR 9

    mainly results in activation of IRF7 and induction of IFNa, an

    important factor for antiviral defense and as shown below in acertain group of diseases.20

    Toll-like receptor expression in different cellsand tissues

    Expression of TLRs has been detected in various human tissues

    with varying expression levels.21 In general, organs involved in

    immune response or exposed to environment, e.g. spleen, skin

    and lungs were found to have significantly higher TLR expres-

    sion, than for instance the brain, liver and skeletal muscles.21

    TLR expression is also increased in peripheral blood mono-

    nuclear cells with large differences of TLR expression levels

    between different cell types. For example, TLR 2, 4 and 8 are

    expressed predominantly in monocytes and macrophages22,23

    with the highest level shown for TLR2 in monocytes. Blood-

    derived myeloid dendritic cells (mDC) express TLR 2, 3, 4, 5

    and 8,2325 while TLR 7, 9 and 10 were found mainly on

    plasmacytoid dendritic cells (pDC) and B-cells.22,26 Of impor-

    tance pDCs are the primary class I IFN producing cells.27

    Interestingly, Imiquimod and Resiquimod act as TLR7 agon-

    ists28 and were shown to induce IFNa and IFNx in pDCs iso-

    lated from blood.29

    In human skin TLRs are expressed in both DC and keratino-

    cytes. Human keratinocytes constitutively express mRNA of

    TLR 1, 2, 3, 4, 5, 6, 9 and 10 not, however, of TLR 7 and

    8.3032 Functionality was also shown for TLR 2, 3, 4, 5 and

    9.30,31,33 Langerhans cells (LC) were also shown to express

    TLR 1, 2, 3, 5, 6, and 10.34

    In another study LCs also respond to TLR 4, TLR 7 and TLR

    9 ligands.35 Among DCs, however, TLR 8 is usually expressed

    in mDCs, while TLR 7 was found predominantly in pDCs.23

    Recently, the accumulation of pDC-like cells has been

    described in the dermis of mice topically treated with TLR 7

    agonist Imiquimod.36

    pDCs were also identified in psoriaticskin lesion.37 Importantly, topical application of TLR7 agonist

    Imiquimod exacerbates the psoriatic lesion,38 indicating that

    pDCs are major effector cells of the immune response induced

    by Imiquimod.

    Th17-induced inflammation

    Recently, further differentiation of immune defense has been

    elaborated.39,40 This arm involves cytokines as well a distinct

    set of lymphocyte subpopulations. Central role in this pathway

    TIR

    TLR4

    TLR7,8,9TIR

    TIR

    TRIFMyD88

    MyD88

    MyD88

    TLR2MD2/CD14

    LPS (E. coli)

    MAL

    TRAM

    MAL

    LPS (P. gingivalis)

    PG (S. aureus)

    NFkB

    ssRNA, CpG-DNA

    Imiquimod, Resiquimod

    IRF7

    TNF, IL1, IL6, IL12IL8, MIP1a RANTESCD40, CD80, CD86

    TNF, IL1, IL6, IL12IL8, MIP1a RANTESCD40, CD80, CD86

    IFN

    TIR

    TLR3

    TRIF

    dsRNA

    IFN

    IRF3NFkB

    IKK complex IKK complexTBK-1 IRAK-1Fig 1. Differential gene expression of TLR-

    signalling. LPS, Lipopolysaccharid; PG,

    Proteoglycan; TIR, Toll-IL1-receptor; MAL,

    MyD88 adaptor-like; TRIF, TIR-related adaptor

    protein inducing interferon; TRAM, TRIF-

    related adaptor molecule; IKK, Inhibitor of

    NFkB kinase; TBK, TANK-binding kinase;

    IRAK, Interleukin-1 receptor associated kinase;

    IRF, Interferon regulatory factor.

    2007 The Authors

    Journal Compilation 2007 British Association of Dermatologists British Journal of Dermatology 2007, 157 (Suppl. 2), 17

    Immune response profiles in human skin, T. Meyer et al. 3

  • 8/3/2019 inmunologia_de_la_piel__Br_J_Dermatol._2007_Dec_157_Suppl_2_1_7[1]

    4/7

    is played by pDC and a new T cell subtype called Th17.40

    As shown by Nestle et al.37 pDC can be activated by a variety

    of ligands including virus RNA, HSP (Heat shock protein) or

    bacterial products to produce IFNa with production being

    1000 times higher compared to any other cell. The responder

    cell for this excess IFNa is noted as mDC.

    Following stimulation via IFNa these latter cells produce

    IL-12, whereby classical T cell activation, marked by secretion

    of IFNc, granzyme and preforin and others, proceeds.41 mDC

    are also seen to produce IL-23 which is structurally related to

    IL-12, sharing a common subunit (p40). IL-12 is known as

    the key promoter of naive T cells to multiply and differentiate

    into Th1 cells and cytotoxic T cells, while IL-23 causes activa-

    tion of a IL-17 producing T cell, which is hence called

    Th17.39,40

    Recent evidence suggests that under the co-stimulatory

    effect of TGFb and IL6 Th17 cells develop from nave T cells.

    Th17 cells once activated by IL-23 become important actors in

    the ensuing inflammatory program in that IL-17 stimulates

    various cells to produce pro-inflammatory cytokines: under

    the stimulus of IL-17 stromal cells, endothelial cells and others

    produce IL-1, TNFa, IL-6, IL-8 and GM-CSF [Granulocyte-

    macrophage colony stimulating factor].42 These mediators are

    chemokines themselves (e.g. IL-8) or powerful stimulators of

    leukocyte activation and migration.43

    The IL-23IL-17 pathway represents an important pro-inflammatory arm of the innate defense with leukocyte infil-

    tration and phagocytosis as ultimate goals. It bears relevance

    to psoriasis and it has been suggested that psoriasis rather than

    being a Th1 disease actually is a Th17 disease.40

    Th17 cells have recently been shown also to produce IL-22

    and further, a receptor for IL-22 is expressed by keratinocytes

    and other epithelial cell e.g. in the gut.4446 This ligand-recep-

    tor binding in keratinocytes causes up-regulation of defense

    molecules (e.g. defensins), but also proteases, S100 proteins

    and chemokines.4749

    Apparently by these two arms of the innate defense armen-tarium (IL-17 and IL-22) not only inflammation is initiated

    but also keratinocytes become active players (see Fig. 2). In

    this context keratinocytes not only respond to environmental

    pathogens by producing antimicrobial defense molecules and

    also, via the IL-22 receptor, interact with endogenous den-

    dritic cell derived signals.

    One of the key TLRs capable for sensing viral nucleotides

    on pDCs is TLR7, which also binds Imiquimod. Several reports

    have shown that Imiquimod applied to skin can exacerbate

    psoriasis.50,51 As IL-23 and IL-17 are found to be up-regulated

    in psoriasis52,53 this IL-17 regulated pathway could play a sig-

    nificant role in initiating psoriatic lesions.

    Attraction of pDCs, which are almost absent in normal skin,

    to inflamed skin areas is mediated by chemotactic peptides like

    IL8 and complement split products, like C5a and its desarginated

    form C5a-des-Arg. These act as powerful chemoattractants and

    are present in increased amounts in psoriatic skin lesions.54,55

    The complement cascade consists of a set of proteolytic

    enzymes and represents a component of the humoral innate

    immune system. Complement can be activated by three sepa-

    rate pathways (classical antibody-mediated, alternate proper-

    dine-induced, and lectinMASP (Mannose binding lectin-

    associated serine protease)-mediated).56 Activation finally leads

    to formation of membrane attack complex, which kills bacte-

    ria by cell lysis. During complement activation the split prod-ucts C3a and C5a are generated, which have anaphylactic and

    chemotactic properties. In serum both are des-arginated by

    carboxypeptidase N to less potent fragments C3a-des-Arg and

    C5a-des-Arg. C5a-des-Arg, like C5a binds to C5a receptor

    (C5aR).57 Recently, expression of C5aR and C3aR was shown

    in immature pDCs of skin lesions from patients with LE and

    contact dermatitis.58 Considering significant amounts of C5a

    and C5a-des-Arg in psoriatic skin lesions complement medi-

    ated attraction might be relevant for infiltration of pDCs in

    psoriatic skin.

    Th1

    Th17

    PMN

    KC KCMP

    G-CSF, IL8, NO

    TNF, IL1, IL6

    pDC mDC

    IL23, TGF, IL6

    IL12

    APsProteases

    IFN

    IFN

    IL17IL22

    FG-CSF

    E

    EC

    Fig 2. The IL-23 Th17 pathway in

    inflammation. pDC, plasmacytoid dendritic

    cell; mDC, myeloid dendritic cell; Th17,

    IL-17 producing T cell; EC, endothelial cell;

    PMN, polymorphonuclear cell; MP,

    macrophage; F, fibroblast; E, epithelial cell;

    KC, keratinocyte; APs, antimicrobial peptides.

    2007 The Authors

    Journal Compilation 2007 British Association of Dermatologists British Journal of Dermatology 2007, 157 (Suppl. 2), 17

    4 Immune response profiles in human skin, T. Meyer et al.

  • 8/3/2019 inmunologia_de_la_piel__Br_J_Dermatol._2007_Dec_157_Suppl_2_1_7[1]

    5/7

    Induction of adaptive immune functions

    A crucial step of the induction of adaptive immune reactions

    is reflected by activation of DC, which are professional antigen

    processing and presenting cells. In skin epidermal LC and der-

    mal dendritic cells (DDC) represent the most important types

    of DC. Resident epidermal LCs are characterized by expression

    of CD1a, CD1c, Langerin, E-cadherin and membrane ATPase

    and by the presence of Birbeck granules. These rod-shaped

    membrane structures are absent in resident DDC. Also, in con-

    trast to LC, DDC do not express Langerin, E-cadherin, and

    membrane ATPase, but do express clotting factor FXIIIa, as

    well as CD1b and CD1c.59

    Dendritic cells take up and process peripheral antigens, and

    after migration to lymphoid organs, present them to nave T

    cells. In addition binding of PAMPs to TLRs results in activa-

    tion of DC by NFkB-induced expression of chemokines, cyto-

    kines, MHC class I and II antigens, co-stimulatory signals, and

    adhesion molecules.19

    Migration of skin DCs to draining lymph nodes through

    afferent lymphatics is associated with functional and pheno-typic changes. While migrating centrally LCs loose their Bir-

    beck granules and downregulate CD1, langerin and E-

    cadherin. In contrast, due to TLR activation, surface proteins

    important for T-cell activation like MHC-proteins, CD40,

    CD58, ICAM-1, CD80, and CD86, are induced.59

    During interaction with nave CD4 T-cells, which have

    entered the lymph nodes through high endothelial venules,

    DCs can induce differentiation of these cells. Usually, differen-

    tiation into two subsets is distinguished: Th1, induced by IL-

    12 and promoting cellular immunity and Th2, induced by IL-

    4 and promoting humoral immunity.41

    Two pathways of antigen presentation in DCs have been

    described: an endocytic pathway, which results in binding of

    exogenous antigens into MHC II molecules for presentation to

    CD4+ T cells, and an endogenous pathway which results in

    association of endogenous antigens to MHC I molecules for

    presentation to CD8+ T cells.60 Moreover, a third pathway of

    antigen uptake and presentation exists, which depends on

    cross presentation and represents an important mechanism for

    presentation of exogenous antigens to CD8 T cells through a

    process called cross priming.61 In addition, nonprotein anti-

    gens (mycobacterial glycolipids) can be internalized after

    binding to langerin. After loading to CD1 molecules in Bir-

    beck granules glycolipids are recycled to cell membrane and

    presented to CD4 and CD8 T cells and NK (Natural Killer)cells.62,63

    In addition to immunostimulatory functions of LCs and

    DDCs in response to potential pathogens skin DCs exert regu-

    latory functions to maintain homeostasis of the skin and to

    prevent excessive inflammatory reactions.64,65 In the absence

    of danger signals under steady state conditions, LCs take up

    epidermal antigens and transport them to local lymph nodes.

    Here they act as promoters of peripheral tolerance to skin

    antigens by inducing anergy or apoptosis of nave T cells or

    by stimulation of regulatory T cells (Treg, CD4+, CD25+).66

    Effectors of adaptive immunity

    T cells become activated by interaction with DC in local drain-

    ing lymph nodes. Only nave T cells with TCRs, specific for

    the antigens presented, are stimulated. In addition to TCR-

    MHC-antigen interaction (tri-molecular complex) co-stimula-

    tory signals are necessary to induce clonal expansion of appro-

    priate T cells (defined by antigen specificity). Co-stimulatory

    signals are represented by interactions of surface molecules,

    like CD40-CD40L, CD28-CD8086 and CD2-CD58, as well as

    cytokines (IL-1, IL-6 TNFa). Absence of these secondary sig-

    nals results in nonreactivity (anergy) to the respective antigens.

    Activated T cells leave the lymph node and enter the skin

    by interaction with specific homing receptors. Most important

    is the interaction of CLA (cutaneous lymphocyte antigen) with

    E-selectin expressed on endothelial cells of inflamed skin. CLA

    is not expressed in nave T cells, but found in about 30% of

    circulating memory T cells and T cells in inflamed skin are

    mainly CD45Ro- and CLA-positive.67,68 Next to CLA, several

    chemokine receptors (CCR4, CCR10) are also associated with

    skin homing of T cells.69,70

    Among T cells, subpopulations with different functions are

    distinguished. CD4+ T cells (T helper cells) are mainly

    involved in immune responses to foreign antigens, while

    CD8+ T cells (cytotoxic T cells) provide antiviral and antitu-

    mor immune reactions. According to the cytokines produced,

    T helper cells were divided into Th1 and Th2 cells: Th1 cells

    produce IL-2, TNFa and IFNc and induce cell mediated

    immune responses. Th2 cells produce IL-4, IL-5, IL-10 and IL-

    13 and promote humoral immune responses, as well as growth

    of eosinophils and IgE production. Th2 responses are fre-

    quently associated with allergic diseases, like atopic dermatitis.

    Th17 cells represent another subset of CD4 and CD8 T cells,

    which are generated different from Th1 and Th2 cells, as

    described above. As shown Th17 cells were suggested to be

    involved in the pathogenesis of psoriasis and other autoim-

    mune diseases.46,71

    Other populations of T cells with suppressiveregulatory

    properties, important to control autoreactive immune

    responses, were recently identified. CD4+ CD25+ regulatory T

    cells (Tregs) have suppressive effects and induce anergy and

    tolerance by cell contacts probably via membrane-bound fac-

    tors. The exact mechanism is still unknown, but does not

    require soluble suppressive cytokines. Th3 and type 1 T regu-

    latory cells (Tr1) are also immunosuppressive, which, in con-

    trast to Tregs, depends on production of immunosuppressivecytokines (IL-10, TGFb).72

    Outlook

    In human approximately 2000 separate disease entities are

    presently recognized and nearly half of these are inflamma-

    tory. Thus the identification of inflammatory pathways in

    skinincluding innate and adaptive, as well as stimulatory

    and regulatoryhas been revolutionized by the rapidly

    increasing knowledge especially of innate immunity. All of

    2007 The Authors

    Journal Compilation 2007 British Association of Dermatologists British Journal of Dermatology 2007, 157 (Suppl. 2), 17

    Immune response profiles in human skin, T. Meyer et al. 5

  • 8/3/2019 inmunologia_de_la_piel__Br_J_Dermatol._2007_Dec_157_Suppl_2_1_7[1]

    6/7

    this makes the skin an appropriate target for immunomodula-

    tory therapy. As our knowledge of underlying immune dysre-

    gulations in skin diseases is improving, activation or

    suppression of distinct immune functions by topical applica-

    tion of immune response modifiers (the number of which is

    steadily increasing) will provide extending treatment modali-

    ties in near time.

    References

    1 Christophers E. Cellular architecture of the stratum corneum. J Invest

    Dermatol 1971; 56:1659.

    2 Elias PM. Stratum corneum architecture, metabolic activity and

    interactivity with subjacent cell layers. Exp Dermatol 1996; 5:191

    201.

    3 Schroder JM, Harder J. Antimicrobial skin peptides and proteins.

    Cell Mol Life Sci 2006; 63:46986.

    4 Janeway CA, Medzhitov R. Innate immune recognition. Annu Rev

    Immunol 2002; 20:197216.

    5 Bos JD, Kapsenberg ML. The skin immune system: progress in

    cutaneous biology. Immunol Today 1993; 14:758.

    6 Boman HG. Antimicrobial peptides: basic facts and emerging con-

    cepts. J Intern Med 2003; 254:197215.

    7 Lehrer RI, Ganz T. Antimicrobial peptides in mammalian and

    insect host defense. Curr Opin Immunol 1999; 11:237.

    8 Ogawa H, Miyazaki H, Kimura M. Isolation and characterization of

    human skin lysozyme. J Invest Dermatol 1971; 57:11116.

    9 Harder J, Schroder JM. Psoriatic scales: a promising source for the

    isolation of human skin-derived antimicrobial proteins. J Leukoc Biol

    2005; 77:47686.

    10 Glaser R, Harder J, Lange H et al. Antimicrobial psoriasin (S100A7)

    protects human skin from Escherichia coli infection. Nat Immunol

    2005; 6:5764.

    11 Sorensen OE, Thapa DR, Rosenthal A et al. Differential regulation

    of beta defensin expression in human skin by microbial stimuli.

    J Immunol 2005; 174:48709.

    12 Nagy I, Pivarcsi A, Koreck A et al. Distinct strains of Propionibacte-rium acnes induce selective human beta-defensin 2 and interleukin

    8 expression in human keratinocytes through toll-like receptors.

    J Invest Dermatol 2005; 124:9318.

    13 Yang D, Chertov O, Bykovskaia SN et al. Beta-defensis: liniking

    innate and adaptive immunity through dendritic and T cell CCR6.

    Science 1999; 286:5258.

    14 Bals R, Wilson JM. Cathelicidins a family of multifunctional anti-

    microbial peptides. Cell Mol Life Sci 2003; 60:71120.

    15 Medzhitov R, Janeway CA. Innate immunity. N Engl J Med 2000;

    343:33844.

    16 Raz E. Organ-specific regulation of innate immunity. Nat Immunol

    2007; 8:34.

    17 Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins

    linking innate and acquired immunity. Nat Immunol 2001; 2:67580.

    18 Barton GM, Medzhitov R. Toll-like receptors and their ligands. Curr

    Top Microbiol Immunol 2002; 270:8192.

    19 ONeill L. How Toll-like receptors signal: what we know and what

    we dont know. Curr Opin Immunol 2006; 18:39.

    20 Uematsu S, Sato S, Yamamoto M et al. Interleukin-1 receptor-asso-

    ciated kinase-1 plays an essential role for Toll-like receptor (TLR)

    7- and TLR 9-mediated interferon-(alpha) induction. J Exp Med

    2005; 201:91523.

    21 Zarember KA, Godowski PJ. Tissue expression of human Toll-like

    receptors and differential regulation of Toll-like receptor mRNAs

    in leukocytes in response to microbes, their products, and cyto-

    kines. J Immunol 2002; 168:55461.

    22 Hornung V, Rothenfusser S, Britsch S et al. Quantitative expression

    of Toll-like receptor 1-10 mRNA in cellular subsets of human

    peripheral blood momonuclear cells and sensitivity to CpG oligo-

    deoxynucleotides. J Immunol 2002; 168:45317.

    23 Gorden KB, Gorski KS, Gibson SJ et al. Synthetic TLR agonists reveal

    functional differences between human TLR7 and TLR8. J Immunol

    2005; 174:125968.

    24 Jarrossay D, Napolitani G, Colonna M et al. Specialization andcomplementarity in microbial molecule recognition by human

    myeloid and plasmacytoid dendritic cells. Eur J Immunol 2001;

    31:338893.

    25 Kadowaki N, Ho S, Antonenko S et al. Subsets of human dendritic

    cell precursors express different Toll-like receptorsand respond to

    different microbial antigens. J Exp Med 2001; 194:8639.

    26 Hasan U, Chaffois C, Gaillard C et al. Human TLR10 is a functional

    receptor, expressed by B cells and plasmacytoid dendritic cells,

    which activates gene transcription through MyD88. J Immunol

    2005; 174:294250.

    27 Akira S, Hemmi H. Recognition of pathogen-associated molecular

    patterns by TLR family. Immunol Lett 2003; 85:8595.

    28 Hemmi H, Kaisho T, Takeuchi O et al. Small anti-viral compounds

    activate immune cells via the TLR7 MyD88-dependent signalingpathway. Nat Immunol 2002; 3:196200.

    29 Gibson SJ, Lindh JM, Riter TR et al. Plasmacytoid dendritic cells

    produce cytokines and mature in response to the TLR7 agonists,

    imiquimod and resiquimod. Cell Immunol 2002; 218:7486.

    30 Lebre MC, van der Aar AMG, van Baarsen L et al. Human keratino-

    cytes express functional Toll-like receptor 3, 4, 5, and 9. J Invest

    Dermatol 2007; 127:33141.

    31 Kollisch G, Kalali BN, Voelcker V et al. Various members of the

    Toll-like receptor family contribute to the innate immune response

    of human epidermal keratinocytes. Immunology 2005; 114:53141.

    32 Baker BS, Ovigne JM, Powles AV et al. Normal keratinocytes express

    Toll-like receptors (TLRs) 1, 2, and 5: modulation of TLR expres-

    sion in chronic plaque psoriasis. Br J Dermatol 2003; 148:6709.

    33 Song PI, Park YM, Abraham T et al. Human keratinocytes expressfunctional CD14 and Toll-like receptor 4. J Invest Dermatol 2002;

    119:42434.

    34 Flacher V, Bouschbacher M, Verronese E et al. Human Langerhans

    cells express a specific TLR profile and differentially respond to

    viruses and Gram-positive bacteria. J Immunol 2006; 177:795967.

    35 Waibler Z, Kalinke U, Will J et al. TLR-ligand stimulated interleu-

    kin 23 subunit expression and assembly is regulated differentially

    in murine plasmacytoid and myeloid dendritic cells. Mol Immunol

    2007; 44:14839.

    36 Palamara F, Meindl S, Holcmann M et al. Identification and charac-

    terization of pDC-like cells in normal mouse skin and melanomas

    treated with imiquimod. J Immunol 2004; 173:305161.

    37 Nestle FO, Conrad C, Tun-Kyi A. et al. Plasmacytoid predendritic

    cells (PDC) initiate psoriasis through interferon-a production. J Exp

    Med 2005; 202:13543.

    38 Gilliet M, Conrad C, Geiges M et al. Psoriasis triggered by Toll-like

    recptor agonist imiquimod in the presence of dermal plasmacytoid

    dendritic cell precursors. Arch Dermatol 2004; 140:14905.

    39 McKenzie BS, Kastelein RA, Cua DJ. Understanding the IL-23-IL17

    immune pathway. Trends Immunol 2006; 27:1723.

    40 Blauvelt A. New concepts in the pathogenesis and treatment of

    psoriasis: key roles for Il-17 and TGFb. Exp Rev Dermatol 2007;

    2:6978.

    41 Murphy KM, Reiner SL. The lineage decision of helper T cells. Nat

    Rev Immunol 2002; 2:93344.

    2007 The Authors

    Journal Compilation 2007 British Association of Dermatologists British Journal of Dermatology 2007, 157 (Suppl. 2), 17

    6 Immune response profiles in human skin, T. Meyer et al.

  • 8/3/2019 inmunologia_de_la_piel__Br_J_Dermatol._2007_Dec_157_Suppl_2_1_7[1]

    7/7

    42 Kolls JK, Linden A. Interleukin 17 family members and inflamma-

    tion. Immunity 2004; 21:46776.

    43 Luster AD. The role of chemokines in linking innate and adaptive

    immunity. Curr Opin Immunol 2002; 14:12935.

    44 Nagalakshmi ML, Rascle A, Zurawski S et al. Interleukin 22 activates

    STAT3 and induces IL-10 by colon epithelial cells. Int Immunopharma-

    col 2004; 4:67991.

    45 Chung Y, Yang X, Chang SH et al. Expression and regulation of

    IL-22 in the IL-17 producing T lymphocytes. Cell Res 2006;

    16:9027.46 Zheng Y, Danilenko DM, Valdez P et al. Interleukin 22, a Th17

    cytokine, mediates IL-23-induced dermal inflammation and acan-

    thosis. Nature 2007; 445:64851.

    47 Sa SM, Valdez PA, Wu J et al. The effects of IL-20 subfamily cyto-

    kines in reconstituted human epidermis suggest potential roles in

    cutaneous innate defense and pathogenetic adaptive immunity in

    psoriasis. J Immunol 2007; 178:222940.

    48 Wolk K, Kunz S, Witte E et al. IL-22 increaes the innate immunity

    of tissues. Immunity 2004; 21:24154.

    49 Liang SC, Tan XY, Luxenberg DP et al. Interleukin (IL)-22 and

    Il-17 are coexpressed by Th17 cells and cooperatively enhance

    expression of antimicrobial peptides. J Exp Med 2006; 203:

    22719.

    50 Rajan N, Langtry JA. Generalized exacerbation of psoriasis associ-ated with imiquimod cream treatment of superficial basal cell car-

    cinomas. Clin Exp Dermatol 2006; 31:1401.

    51 Fanti PA, Dika E, Vaccari S et al. Generalized psoriasis induced by

    topical treatment of actinic keratosis with imiquimod. Int J Dermatol

    2006; 45:14645.

    52 Lee E, Trepicchio WL, Oestreicher JL et al. Increased expression of

    interleukin 23 p19 and p40 in lesional skin of patients with psori-

    asis vulgaris. J Exp Med 2004; 199:12530.

    53 Toichi E, Torres G, McCormick TS et al. An anti-IL12-p40 antibody

    down-regulates type 1 cytokines, chemokines, and IL12 Il23 in

    psoriasis. J Immunol 2006; 177:491726.

    54 Schroder JM, Christophers E. Identification of C5a des arg and an

    anionic neutrophil-activating peptide (ANAP) in psoriatic scales.

    J Invest Dermatol 1986;87

    :538.55 Takematsu H, Tagami H. Quantification of chemotactic peptides

    (C5a anaphylatoxin and IL8) in psoriatic lesional skin. Arch Dermatol

    1993; 129:7480.

    56 Beutler B. Innate immunity: an overview. Mol Immunol 2004;

    40:84559.

    57 Cain SA, Monk PN. The orphan receptor C5L2 has high affinity

    binding sites for complement fragments C5a and C5a des-Arg.

    J Biol Chem 2002; 277:71659.

    58 Gutzmer R, Kother B, Zwirner J et al. Human plasmacytoid den-

    dritic cells express receptors for anaphylatoxins C3a and C5a and

    are chemoattracted to C3a and C5a. J Invest Dermatol 2006;

    126:24229.

    59 Larregina AT, Falo LD. Changing paradigms in cutaneous immu-

    nology: adapting with dendritic cells. J Invest Dermatol 2005; 124:

    112.

    60 Larregina AT, Falo LD. Dendritic cells in the context of skin immu-

    nity. In: Dendritic Cell Biology and Clinical Applications. (Lotze M, Thomas

    AW eds), AP London UK: Academic Press, 2001; chapter 23:30114.

    61 Kovacsovics-Bankowski M, Rock K. A phagosome to cytosol path-

    wayfor exogenous antigens presented on MHC class I molecules.

    Science 1995; 267:2436.

    62 Hunger RE, Sieling PA, Ochoa MT. Langerhans cells utilize CD1a

    and langerin to efficiently present non-peptide antigens to T cells.

    J Clin Invest 2004; 113:7018.

    63 Brigl M, Brenner MB. CD1: antigen presentation and T cell func-

    tion. Annu Rev Immunol 2004; 22:81790.

    64 Steinmann RM, Hawiger D, Nussenzweig MC. Tolerogenic den-

    dritic cells. Annu Rev Immunol 2003; 21:685711.

    65 Nickoloff BJ. Cutaneous dendritic cells in the crossfire between

    innate and adaptive immunity. J Dermatol Sci 2002; 29:15965.

    66 Morelli AE, Hackstein H, Thomson AW. Potential tolerogenic den-dritic cells for transplantation. Semin Immunol 2001; 13:32335.

    67 Picker LJ, Michie SA, Rott LS, Butcher ECA. A unique phenotype of

    skin-associated lymphocytes in humans. Preferential expression of

    the HECA-452 epitope by benign and malignant T cells at cutane-

    ous sites. Am J Pathol 1990; 138:105368.

    68 Pitzalis C, Cauli A, Pipitone N et al. Cutaneous lymphocyte antigen-

    positive T lymphocytes preferentially migrate to skin but not to

    the joint in psoriatic arthritis. Arthritis Rheum 1996; 39:13745.

    69 Biedermann T, Schwarzler C, Lametschwandtner G et al. Targeting

    CLA E-selectin interactions prevents CCR4-mediated recruitment of

    human Th2 memory cells to human skin in vivo. Eur J Immunol

    2002; 32:317180.

    70 Hudak S, Hagen M, Liu Y et al. Immune surveillance and effector

    functions of CCR10+ skin homing T cells. J Immunol 2002;169:118996.

    71 Kikly K, Liu L, Na S, Sedgwick JD. The IL-23 Th(17) axis: thera-

    peutic targets for autoimmune inflammation. Curr Opin Immunol

    2006; 18:6705.

    72 Jonuleit H, Schmitt E. The regulatory T cell family: distinct subsets

    and their interrelations. J Immunol 2003; 171:63237.

    2007 The Authors

    Journal Compilation 2007 British Association of Dermatologists British Journal of Dermatology 2007, 157 (Suppl. 2), 17

    Immune response profiles in human skin, T. Meyer et al. 7