Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Dec 14.
Published in final edited form as: Nat Med. 2012 May 4;18(5):10.1038/nm.2760. doi: 10.1038/nm.2760

T cell homing to epithelial barriers in allergic disease

Sabina A Islam 1, Andrew D Luster 1
PMCID: PMC3863331  NIHMSID: NIHMS534816  PMID: 22561834

Abstract

Allergic inflammation develops in tissues that have large epithelial surface areas that are exposed to the environment, such as the lung, skin and gut. In the steady state, antigen-experienced memory T cells patrol these peripheral tissues to facilitate swift immune responses against invading pathogens. In at least two allergy-prone organs, the skin and the gut, memory T cells are programmed during the initial antigen priming to express trafficking receptors that enable them to preferentially home to these organs. In this review we propose that tissue-specific memory and inflammation-specific T cell trafficking facilitates the development of allergic disease in these organs. We thus review recent advances in our understanding of tissue-specific T cell trafficking and how regulation of T cell trafficking by the chemokine system contributes to allergic inflammation in mouse models and in human allergic diseases of the skin, lung and gut. Inflammation- and tissue-specific T lymphocyte trafficking pathways are currently being targeted as new treatments for non-allergic inflammatory diseases and may yield effective new therapeutics for allergic diseases.

Sensitization to allergens and epithelial-barrier dysfunction

Atopic sensitization commonly occurs early in life if epithelial barrier integrity is compromised and the epithelium becomes aberrantly activated, which may occur through a complex interplay of environmental insults and host factors1. For example, filaggrin loss-of-function mutations that compromise epithelial barrier integrity have been associated with an increased risk for developing eczema and asthma1,2. Barrier dysfunction in the lung and skin allows allergens to activate the epithelium and produce cytokines that are permissive for the induction and development of T helper type 2 (TH2) responses, such as interleukin-33 (IL-33), thymic stromal lymphopoietin (TSLP) and IL-25 (Fig. 1)110. A dysfunctional epithelial barrier also allows antigen-sampling dendritic cells to become directly activated by allergens and undergo maturation in a TH2-permissive milieu such that they subsequently prime allergen-specific TH2 responses1,6,8.

Figure 1.

Figure 1

Generation of allergen-specific effector and memory T cells during primary mmune responses at the epithelial barrier. During the primary immune response to allergens, disruption of the epithelial barrier eads to epithelial cell production of cytokines, such as TSLP, IL-25 and IL-33. Allergens and other inflammatory cues in the presence of TSLP, IL-25 and IL-33 induce dendritic cell (DC) maturation and trafficking to tissue draining lymph nodes (LN) where dendritic cells activate naive T (TN) cells to differentiate into TH2-type effector (TEFF) cells. Allergen-specific TEFF cells undergo clonal proliferation in the lymph nodes, and TEFF cells exit the ymph nodes through efferent lymphatics. TEFF cells re-enter the systemic circulation through the thoracic duct and enter inflamed tissue through postcapillary venules. The majority of TEFF cells eventually die, but a minority differentiate into memory T (TM) cells several weeks after the resolution of nflammation. Depending on the profile of trafficking receptors expressed, different subsets of TM cells perform distinct types of mmune surveillance in the steady state. Naive T cells and central memory T (TCM) cells equipped with L-selectin (CD62L) and the chemokine receptor CCR7 have restricted and specific access to secondary ymphoid tissue through specialized high endothelial venules (HEVs), which express their respective ligands (PNAd and CCL21). T cells recirculate between lymphoid tissue and blood. Effector memory T (TEM) cells primarily recirculate between the blood and peripheral tissue. TRM cells persist in peripheral tissue and do not recirculate. TSLPR, TSLP receptor; IL-33R, IL-33 receptor.

Programmed T cell trafficking and allergic disease

Accurate and efficient tissue-specific trafficking between the circulation, lymphoid organs and peripheral tissues is a fundamental prerequisite for effector T cell function and results in either host immunity or tissue immunopathology. Leukocytes coordinately use adhesion molecules in a highly regulated process to directionally extra-vasate from the blood into target peripheral tissues11. These adhesion molecules include selectins, integrins and chemoattractant receptors. Selectins are a conserved family of C-type lectins and include L-selectin and P-selectin glycoprotein ligand 1 (PSGL1) and promote the rolling movement of leukocytes along the surfaces of endothelial cells11. Integrins are a family of two-chain type I transmembrane receptors and include leukocyte function-associated antigen 1 LFA-1 (CD11a–CD18 or αLβ2) and α4β7 and are involved in the firm adhesion of leukocytes to endothelial cells11. Chemoattractant receptors are a family of G protein–coupled receptors (GPCRs) that activate integrins through inside-out signaling to initiate firm adhesion and induce directed cell migration through tissue gradients of their ligands and include chemokine and lipid chemoattractant receptors11. During inflammation, ligands for selectins, integrins and chemoattractant receptors are upregulated in tissue and vascular beds and provide directional cues for inflammatory T cells, on which the corresponding receptors are upregulated, to enter inflamed tissue from the blood. In addition to inflammation-regulated trafficking cues, allergy-prone organs such as the skin and the small intestine provide an additional level of specificity in T cell trafficking to these organs through a process known as ‘imprinting’1113.

The profile of integrins, selectins and chemokine receptors expressed by T cells is determined by their states of activation, polarization and differentiation, as well as their initial tissue site of antigen priming, leading to tissue-specific imprinting in some organs1316. Tissue-specific imprinting is best characterized in the small intestine and the skin, as preferential homing to these organs was discerned more than two decades ago1721. Naive, effector and memory T cells have distinct trafficking patterns within the circulation, lymphoid organs and peripheral tissue (Fig. 1). After T cell receptor activation, naive T cells differentiate into effector T cells (Fig. 1), which express an altered profile of trafficking receptors; some of these trafficking receptors are maintained in steady-state memory T cells after inflammation is resolved14,15. During the initial priming of naïve T cells, some allergy-prone organs that are exposed to the environment ‘program’, or imprint, preferential tissue-specific inflammatory effector T cell and memory T cell entry from the circulation to streamline immune surveillance, as many microbes are tissue tropic in virulence12,17,22.

T cells that are programmed to home to the small intestine are imprinted during the initial antigen priming to specifically express integrin α4β7 and the chemokine receptor chemokine (C-C motif) receptor 9 (CCR9), expression of which enables restricted and regulated entry into the small intestine23,24 (Fig. 2). In contrast, the signature-homing receptors of skin-tropic T cells are the lectin cutaneous leukocyte antigen (CLA), which is derived from the glycosylation of PSGL1, in combination with the chemokine receptors CCR4 and CCR10 and, for some T cell subsets, CCR8 and CCR6 (Fig. 2)20,2532. Low levels of the ligands for these receptors are constitutively expressed in the steady state to facilitate tissue-specific memory T cell homing and immune surveillance30,3336. Higher levels of the ligands induced during inflammation promote increased tissue-specific effector T cell homing. Notably, mechanisms that facilitate gut- and skin-specific imprinting are dependent on tissue-specific dendritic cells and tissue-specific environmental cues12,22. In the gut, CD103+Eβ7 integrin) lamina propria and Peyer’s patch dendritic cells produce high concentrations of retinoic acid; retinoic acid enables these dendritic cells to imprint expression of the gut-tropic receptors α4β7 and CCR9 on naive T cells3742. Likewise, skin dendritic cells can generate high concentrations of the active vitamin D3 metabolite, 1,25(OH)2D3, and then use it to induce CCR10 expression on T cells in higher mammals22,43.

Figure 2.

Figure 2

Tissue-specific imprinting and steady-state programmed memory T cell trafficking to atopy-prone organs. Dendritic cells of the skin and the gut imprint T cells during the primary immune response to express trafficking receptors that enable tissue-specific homing. Skin T cells express CLA, CCR10, CCR4 and CCR8, which enable skin TEM cell entry into the skin in the steady state, as postcapillary venules in the skin express CCL17, E-selectin and CCL1 (the respective igands for CCR4, CLA and CCR8) and epidermal cells expresses CCL27 (the ligand for CCR10) and mCCL8 (the ligand for CCR8 in the mouse). Gut TEM cells are programmed to express CCR9 and α4β7, which enable the entry of gut TEM cells into the small intestine through postcapillary venules that express CCL25 and MADCAM1, which are the respective ligands for CCR9 and α4β7. Skin- and gut-specific programmed lymphocyte homing pathways are well characterized in human and mouse studies. Human studies also suggest that programmed tissue-specific homing of lung TEM cells may also occur.

Studies suggest that the lungs and nasal-associated lymphoid tissue (NALT) also program T cells for tissue-specific homing12,44. High endothelial venules in NALT express high amounts of peripheral node addressin (PNAd), the ligand for CD62L, the expression of which distinguishes NALT from other mucosal-associated tissues, such as Peyer’s patches. Programmed allergen-specific T cell entry into NALT through interactions with PNAd promotes allergic rhinitis44. Though not definitive, lung-specific imprinting is suggested by human studies in which CD8+ T cells specific for respiratory, but not systemic, viruses were selectively enriched in human lungs, analogous to the enrichment of CLAhi T cells specific for skin-tropic but not systemic viruses in the skin45,46. Steady-state human lung-resident T cells are also reported to be enriched for CCR5, CCR6, chemokine (C-X-C motif) receptor 3 (CXCR3) and the integrins VLA1 (α1β1), αEEβ7 or CD103) and VLA4 (α4β1)4749. However, the cellular mechanisms and environmental cues that promote lung-specific T cell imprinting have not been elucidated.

A consequence of naive T cell priming at any tissue site is the generation of memory T cells after the resolution of inflammation; recently a category of memory T cells known as tissue-resident memory T cells (TRM) has been characterized50 (Fig. 1). Based on functional traits that are linked to the ability to traffic to either secondary lymphoid tissue or peripheral tissue, memory T cells are classified into ‘central memory’ and ‘peripheral memory’ T cells51 (Fig. 1). Central memory T cells preferentially traffic between secondary lymphoid tissue and the systemic circulation, whereas effector memory T cells preferentially home to peripheral tissue from the blood14,15,51 (Fig. 1). A fraction of antigen-primed effector T cells differentiate into long-lived antigen-specific memory T cells, some of which persist throughout the lifetime of the host50. Recent studies have suggested that large pools of tissue TRM cells with enhanced effector capabilities persist in uninflamed peripheral organs, such as the lung, skin and gut, and provide a first line of defense against pathogens (Fig. 1)32,48,50,5258.

In the context of allergic diseases, programmed homing pathways increase the likelihood that tissue-patrolling memory T cells will encounter activated dendritic cells that present a cognate allergen in the tissue as well as in the draining lymph nodes. Sensitization to environmental allergens at the epithelial surfaces will generate long-lived allergen-specific memory T cells that are programmed to home to the mucosal and epithelial surfaces for immune surveillance. Concordantly, allergen-induced pathological activation of the epithelial barrier, through the production of cytokines such as IL-33, TSLP and IL-25, enhances the activation and maturation of TH2-priming dendritic cells. Prior and recurrent allergen exposure will also generate higher frequencies of recirculating allergen-specific memory T cells, as well as long-lived allergen-specific TRM cells at the tissue sites of allergen exposure (Fig. 1). Thus, instead of being a very rare event, programmed homing pathways increase the likelihood that after allergen re-exposure, tissue-patrolling recirculating memory T cells and TRM cells encounter cognate allergen presented by dendritic cells, particularly if the epithelial barrier is compromised59.

Programmed regulatory T cell trafficking to the epithelial barrier

Programmed trafficking pathways that promote T cell–mediated allergic inflammation are also used by regulatory T (Treg) cells to rein in inappropriate responses at mucosal and epithelial surfaces. The dendritic-cell–derived factors retinoic acid and 1,25(OH)2D3 can be either proinflammatory or anti-inflammatory6065. In the intestine, retinoic acid derived from CD103+ mucosal dendritic cells induces forkhead box P3 (Foxp3)+ Treg cell differentiation from naive T cells after in vivo activation63,65,66. Retinoic acid also imprints Foxp3+ adaptive Treg cells in the mesenteric lymph node (MLN) to acquire expression of α4β7 and CCR9 to enable homing to the small intestine from the MLN61. In mice deficient for integrin β7 or the integrin ligand mucosal vascular addressin cell adhesion molecule 1 (MADCAM1), oral tolerance is abrogated, as Treg cells fail to traffic from the MLN to the lamina propria67. In contrast, in the presence of pro-inflammatory cytokines, such as IL-15, retinoic acid can act as an adjuvant to promote inflammatory responses to dietary antigen68. In the skin, skin-tropic effector memory T cells and skin-tropic Treg cells share receptor profiles comprised of CCR4, CLA and CCR8, which suggests that common environmental cues may imprint proinflammatory and anti-inflammatory T cell homing pathways to the skin13,69. Treg cells exert their suppressive effects by localizing to lymphoid organs during the induction of the immune response and to peripheral tissues during the ongoing immune response13,67,7072. For example, oral tolerance to ingested soluble antigen is initiated during the inductive phase of the immune response in the MLN, and oral tolerance is abrogated after mesenteric lymphadenectomy and in CCR7-deficient mice71. An altered epithelial barrier may allow proinflammatory cytokines or persistent allergen exposure to tip a programmed tissue-tropic pathway that promotes tolerance to innocuous allergens toward one that induces allergic inflammation.

Tissue-resident memory T cells and allergic disease

A convincing body of work over the past decade has shown that antigen-experienced effector memory CD4 and CD8 T cells persist for long periods of time in uninflamed peripheral tissue as functional TRM cells32,48,50,5258. Steady-state migration of pathogen-specific memory CD8 T cells from the circulation into the lung, skin and gut is more restricted compared to other peripheral organs55,56. In infectious models, memory CD8 T cell persistence is antigen dependent in the lung and is antigen independent in the skin and intestine55,73. Re-exposure to antigen can also increase the size of the TRM cell pool57. TRM CD4 and CD8 T cells can be activated by antigen-presenting dendritic cells in tissue before activation by dendritic cells in the draining lymph nodes54,58,74. In addition to a functional role for TRM cells in host immunity to pathogens, a functional role for TRM cells has been shown in chronic inflammatory disease53,75. Memory CD4 T cells derived from human psoriasis skin and grafted into mouse dermis are sufficient for induction of disease in a xenotransplanation model of psoriasis75.

In humans, large reservoirs of TRM cells have been reported in the skin and lungs32,48. Twice as many TRM cells are present in healthy skin compared to blood, and 98% of all CLA+ T cells are found in the skin in the steady state32. Likewise, steady-state TRM cells in the human lung are equivalent in number to the circulating steady-state memory T cells in the blood48. Consistent with in vivo mouse studies, these lung TRM cells are not anergic, and they produce effector cytokines after activation48. Moreover, only lung TRM cells robustly proliferated ex vivo in response to influenza virus, a respiratory pathogen, in an antigen-specific manner, whereas skin-resident and blood T cells did not48.

It is unclear whether and how TRM cells modulate allergic inflammation. The traditional thinking has been that the hugely expanded pool of allergen-specific effector T cells derived from dendritic-cell–activated central memory T cells express inflammatory chemokine receptors that enable their exit from lymphoid organs and their entry into peripheral sites of inflammation from the circulation76. Although the majority of effector T cells in a recall response are probably derived from central memory T cells, this paradigm may require revision with the appreciation that pre-existing tissue pools of TRM cells can be activated by tissue-resident dendritic cells. TRM cells may locally modulate allergic responses during the initiation phase of an allergen-specific recall response within the first few days after exposure, before lymph-node–derived amplified allergen-specific effector T cell entry into inflamed tissue from the bloodstream.

Inflammatory TH2 cell trafficking

CD4 TH cells have a pivotal role in mediating adaptive immunity to diverse pathogens and in promoting autoimmune diseases and allergic inflammation77. During inflammation and after T cell receptor (TCR) activation, specific cytokines derived from the innate immune system and distinct master transcription factors program naive TH cells to differentiate into TH1, TH2, TH17, Treg and follicular T helper (TFH) cell subsets77. The master transcriptional regulators T-box 21 (T-bet), GATA binding protein 3 (GATA3), RAR-related orphan receptor γ (ROR-γT), FOXP3 and B cell CLL/lymphoma 6 (BCL6) drive TH cell subset differentiation and impart specificity of TH cell lineage function through the production of signature effector cytokines77 (Fig. 3). Repeated antigen exposure and progressive lineage polarization also promote terminal TH1 and TH2 cell differentiation through epigenetic modifications at the Ifng and Il4 loci to enable rapid effector cytokine production after antigen rechallenge78. Polarizing cytokines and master transcription factors that promote TH1, TH2 and TH17 cell lineage differentiation also program the expression of unique combinations of inflammation-specific chemokine receptors on TH cell lineages during priming13,15,79 (Fig. 3). Thus, TH2 cells are programmed during lineage differentiation to selectively express the chemokine receptors CCR4, CCR8 and, in humans, the prostaglandin D2 chemoattractant receptor DP2 or CRTH2 (refs. 79–81), which are associated with TH2-type inflammation.

Figure 3.

Figure 3

Chemokine receptors associated with CD4+ helper T cell subsets. (a) During the primary immune response, cytokines (indicated above the arrow) and transcription factors (T-bet, GATA3 and ROR-γT) nstruct naive T cells to differentiate into TH cell subsets. After TCR activation, specific cytokines derived from the innate immune system and unique master transcription factors direct naive CD4 TH cells to differentiate into TH1, TH2, TH17, Treg and TFH cell subsets. Differentiated TH cell subsets produce lineage-specific effector cytokines and express characteristic chemokine receptors. (b) After recall stimulation with an antigen, allergen-specific memory TH2 cells rapidly produce higher amounts of effector cytokines, such as IL-4, on rechallenge with the cognate allergen as compared to naive T cells. Repeated allergen-induced TH2 cell differentiation induces a highly differentiated, CCR8+IL-5hi TH2 cell subset that coexpresses the indicated receptors. Memory TH2 cells may not be fixed in differentiation along the TH2 lineage and may adopt features of TH17 and TH1 cells on rechallenge with allergen in the context of inflammatory cues derived from fungi or viruses, respectively. TGF-β, transforming growth factor β; IL-25R, IL-25 receptor.

TH2 cell heterogeneity

TH2 cells can also be divided into an IL-4hiIL-5lo subset and a more differentiated IL-4loIL-5hi subset, which can be distinguished by their expression of CCR4 or CCR8, respectively29,82. Upregulation of CCR4 expression on TH2 cells occurs rapidly after TCR ligation irrespective of prior antigen exposure and the extent of TH2 differentiation29,83. However, CCR8 expression and robust expression of IL-5 in TH2 cells is restricted to more terminally differentiated TH2 cells that have previously been exposed to antigen29,69,82,84,85. Other examples of lineage-specific gene expression in more terminally differentiated T helper subsets include the IL-33 receptor (also called T1ST2 or ST2) in more terminally differentiated TH2 cells and hepatitis A virus cellular receptor 2 (TIM3) in highly differentiated TH1 cells86,87.

Although it was previously believed that highly differentiated T helper subsets are fixed with regard to lineage differentiation, plasticity in TH17, Treg, TFH and TH2 cell lineage differentiation is now appreciated, as is the fact that additional TH2 cell subsets exist in vivo during chronic inflammation8890. Virus infection and the induction of interferons reprogram antigen-specific, IL-4–producing GATA3+ TH2 memory cells to become TH2+1 cells that coexpress GATA3 and T-bet and secrete both IL-4 and interferon γ (IFN-γ)88. Likewise, TH2 memory cells differentiate into a subset of IL-17–producing TH2 cells that coexpress GATA3 and ROR-γT in severe chronic allergic inflammation that manifests a robust mixed neutrophilic and eosinophilic infiltrate in vivo89.

This heterogeneity of TH2 cells should be kept in mind, as the original studies that elucidated TH cell subset biology were primarily conducted in tissue culture in the presence of neutralizing antibodies to generate pure TH cell lineages. During chronic inflammation in the absence of neutralizing antibodies in vivo and in the setting of genetic and environmental heterogeneity, human allergic disease is more complex and involves the simultaneous production of multiple cytokines. This is concordant with the current appreciation that human asthma is a heterogeneous disease and that therapeutic strategies that target a single TH2 cell cytokine may only be effective in subpopulations of patients with asthma in which the therapeutic intervention is matched to the underlying pathophysiology91,92. Thus, ex vivo studies of human peripheral blood T cells have not provided a clear cut correlate of TH1 and TH2 effector cell cytokine production and the signature chemokine receptors that are associated with pure TH1 and TH2 lineage-differentiated cells16,93. It therefore follows that T cell–mediated allergic disease probably involves multiple chemokines and their receptors, the relative roles of which will vary depending on the affected organ and the temporal phase of the disease. We illustrate these points in the following sections.

T lymphocyte trafficking in atopic disease

Atopic dermatitis

The role of CD4+ T cell effector functions in the pathogenesis of atopic dermatitis has been established in both mouse and human studies94,95. In human atopic dermatitis, a number of skin-homing trafficking receptors, such as CLA, CCR4, CCR10 and CCR8, as well as their ligands, have been implicated and reported to correlate with disease activity94,9699. Although the contributions of CCL17 and CCL27 in the recruitment of human memory CD4 T cells from the blood to the sites of skin inflammation are well established, a correlation of skin CCL17 and CCL27 transcripts with specific inflammatory human disease phenotypes has not been found100. In contrast, comparative analyses of all known chemokine RNA transcripts in healthy skin from control individuals and lesional skin from individuals with different chronic inflammatory skin disorders found significant and specific induction of the chemokines CCL1 and CCL18 in inflamed atopic dermatitis skin34,101. Currently, the receptor for CCL18 is not known, although CCL18 binds skin-tropic CLA+ memory T cells in patients with atopic dermatitis and induces in vivo migration of human memory T cells102. Additionally, microarray analyses have revealed that expression of the enzyme fucosyltransferase VII (FucT-VII), which regulates CLA expression, as well as the chemokine receptors CCR4 and CCR10, was significantly elevated in blood CD4+ T cells from individuals with atopic dermatitis compared to healthy controls103,104. Notably, the frequency of CLA+ blood T cells correlates with disease activity and skin inflammation in both contact hyper-sensitivity and atopic dermatitis105. Additional studies have suggested a role for chemokine (C-X3-C) receptor 1 (CX3CR1) and its ligand, CX3CL1, in active atopic dermatitis and are concordant with studies that suggest a role for CX3CR1 in asthma106108.

The in vivo role of trafficking receptors that regulate T cell–mediated atopic skin inflammation has recently been clarified in two complementary studies that used a 7-week treatment schedule in a mouse model of chronic atopic dermatitis109. CCL27 promotes atopic dermatitis in this model28. Recent studies highlighted how the kinetics of chemokine and chemokine-receptor expression, tissue-specific T cell trafficking and tissue reservoirs of antigen-specific T cells might regulate cutaneous inflammation during acute, recurrent or chronic allergen exposure (Fig. 4)29,110. CCR4 is essential for the acute entry of T cells into the skin through the cutaneous vasculature and drives atopic inflammation in antigen-naive skin, in which pre-existing skin-resident reservoirs of antigen-specific memory T cells are absent110. Thus CCR4 is crucial for T cell entry into the skin during the initiation phase of cutaneous allergic inflammation, which is concordant with the rapid kinetics of the induction of its ligand, CCL17, in skin endothelium and with findings in other models of acute cutaneous inflammation25,111. However, after repeated cutaneous allergen sensitization, CCR4 is not necessary for the entry of TH2 cells from cutaneous vascular beds into the skin to sustain chronic eosinophilic atopic inflammation29. In this setting, pre-existing skin pools of allergen-specific memory TH2 cells are probably sufficient to initiate and drive local inflammatory responses through effector cytokine production after activation by skin-resident antigen-presenting cells. In contrast, in this setting, both CCR8 and its newly described second ligand, mCCL8 (a ligand for CCR8 in the mouse), are essential for promoting IL-5–driven eosinophilic inflammation in chronic atopic dermatitis29. After 7 weeks of treatment, when peak atopic inflammation is manifest, mCCL8 is the only T cell–attracting chemokine that is significantly induced in sensitized skin, and its expression is several folds higher than those of CCL1, CCL17 and CCL22. Moreover, trafficking of CCR8+ TH2 cells, which are enriched for IL-5 and the IL-25 receptor, into allergen sensitized skin is essential for driving skin inflammation, as both IL-5 and IL-25 are cytokines that amplify eosinophilic inflammation (Fig. 4)5,112.

Figure 4.

Figure 4

Trafficking of TH2 cell subsets into the skin during the acute and chronic phases of atopic dermatitis. During the acute phase of atopic dermatitis, the initiation of inflammation is dependent on TH2 cell entry into inflamed skin by CCR4 and the CCR4 ligand CCL17, which is upregulated in inflamed postcapillary venules. During the chronic phase of atopic dermatitis, the pool of allergen-specific skin tissue TRM cells is increased as compared to in the acute phase, and these allergen-specific TRM cells can initiate inflammation independent of CCR4-dependent TH2 cell entry from the blood. Repeated prior allergen exposure also leads to the in vivo generation of the more differentiated CCR8+IL-5hi TH2 cell subset in the chronic phase. Allergen challenge induces production of the CCR8 ligand, mCCL8, in the skin. mCCL8-dependent trafficking of L-5hi CCR8+ TH2 cells into the skin drives eosinophilic skin inflammation in the chronic phase of atopic dermatitis in the mouse model.

Asthma

The mechanisms of T cell–mediated inflammation have been extensively characterized in human asthma and in mouse models of allergic pulmonary inflammation and have identified CD4+ TH2 cells as being central to the pathogenesis of human asthma. This concept has been established through studies of the specific effects of TH2 cytokines and the in vivo findings that TH2 cells are sufficient to initiate and drive asthma pathogenesis59,77,113,114. However, it is now well appreciated that other T cell subtypes, such as TH1, TH17, Treg, natural killer T (NKT) and innate helper type 2 cells coexist with TH2 cells and contribute to the pathogenesis of asthma113,114. Furthermore, the plasticity of TH2 lineage differentiation, such that TH2 cells may undergo additional differentiation to coproduce IFN-γ or IL-17, has been shown in mouse models of allergic pulmonary inflammation and, for IL-17, in human asthma88,89. Given the chronicity of human asthma, the possibility that various environmental exposures that induce recurrences and exacerbations throughout the course of disease cumulatively modulate the phenotype of an individual’s allergic inflammation is not surprising. For example, respiratory viral infections and environmental mold exposure have been shown to be key variables in the exacerbation of allergic asthma115118.

The clinical presentation of asthma led us to focus our efforts on dissecting the mechanisms that underlie T cell trafficking and inflammation in the lung in the context of the asthmatic response, which can be divided into an early and a late phase119. The early phase of the asthmatic response occurs rapidly (within 5–60 min of allergen exposure) as a result of mast cell degranulation and mediator release and is characterized by airway swelling and bronchoconstriction, which results in a corollary decrease in airway airflow (Fig. 5)119. The early phase response often resolves but is followed several hours (>4) later by a more severe late phase response, which is induced by the infiltration of inflammatory cells, such as T cells and eosinophils in the airways, and is also associated with reduced airway airflow119. The late phase asthmatic response can last for extended periods of time120. Moreover, recurrent and constant exposure to inciting allergens drives inflammation and permanent airway remodeling in individuals with chronic asthma119. We first showed that T cell trafficking is necessary for asthmatic inflammation by establishing the essential requirement of GPCR signaling in allergic airway inflammation, for which we used an adoptive transfer model of asthma121,122. T cell trafficking during the early and late phase asthmatic response involves multiple trafficking receptors, which are regulated by both innate and adaptive pathways and are summarized below and reviewed in more detail in ref. 123 (Fig. 5).

Figure 5.

Figure 5

Chemokine-regulated TH cell trafficking into the lung during the early and late phases of the asthmatic response. During the early phase allergic airway response (left), allergen-IgE complexes stimulate airway mast cells to degranulate and release acute mediators such as histamine, tumor necrosis factor α (TNF-α), LTB4, PGD2 and the chemokine CCL1. LTB4, PGD2 and CCL1 recruit TH2 cells into the airways through BLT1, CRTH2 and CCR8, respectively. T lymphocyte trafficking during the late phase allergic airway response (right) is facilitated by innate cells, such as macrophages and myeloid dendritic cells (mDCs). Allergen-induced TLR activation induces macrophage and epithelial cell release of chemokines such as CXCL10 and CCL20. These chemokines recruit an initial wave of T cells into the airways through their receptors, CXCR3 and CCR6. IL-4 and IL-13 produced by activated lung allergen-specific TRM cells stimulate mDCs to release the chemokines CCL17 and CCL22 in a STAT6-dependent fashion. These chemokines amplify the subsequent recruitment of TH2 effector cells, which are generated in lung draining lymph nodes, into the airways from the circulation by the TH2 cell receptor CCR4. IL-4 and IL-13 also induce the differentiation of alternatively activated macrophages, which produce CCL17 to promote CCR4-dependent TH2 cell recruitment. CCR8 and CX3CR1 are also probably involved in T cell recruitment into the lung during the late phase allergic airway response. CCR8 and CX3CR1 are implicated in human asthma and in mouse models of airway inflammation. The CCR8 ligand mCCL8 is upregulated in mouse models of allergic airway inflammation, but the cellular sources of mCCL8 in the lung have not been defined. The CX3CR1 ligand, CX3CL1, is induced in airway smooth muscle cells, lung endothelium and epithelial cells after allergen challenge.

Airway mast cells are the innate cells that are crucial for the early phase recruitment of T cells and initiation of the asthmatic response. When sensitized individuals inhale an allergen, pre-existing allergen-specific immunoglobulin E (IgE) leads to the crosslinking of mast cell IgE Fc receptors and the release of TH2-type effector cytokines, inflammatory mediators, leukotrienes, prostaglandins and inflammatory chemokines by mast cells into the airways (Fig. 5). Among the leukotrienes released by mast cells, leukotriene B4 (LTB4) is rapidly generated within minutes of exposure from membrane lipids and is the lipid chemoattractant ligand for the GPCR leukotriene B4 receptor 1 (BLT1), which is expressed by TH1, TH2 and TH17 cells and induces their migration (unpublished observations and ref. 124). A deficiency of BLT1 leads to a defect in early T cell accumulation in the airways in a model of allergic pulmonary inflammation that involves active allergen immunization and the generation of allergen-specific IgE124,125. Mast cells also produce the chemokines CCL1, CCL17 and CCL22 (refs. 34,126,127). Mast cell release of CCL1 may be key for early TH2 cell recruitment through the CCR8 receptor, as was shown in a mast-cell–dependent model of chronic allergic inflammation126. The prostaglandins released include prostaglandin D2 (PGD2), the receptors for which are the GPCRs DP1 and DP2. DP2 is also called CRTH2 (chemo-attractant receptor-homologous molecule expressed on TH2 cells) and is expressed by human TH2 cells. PGD2 induces migration and activation of TH2 cells through CRTH2, and polymorphisms of CRTH2 are associated with an increased risk for asthma in Chinese children and African Americans but a reduced risk for asthma in German children81,128,129. The rapid kinetics of mast cell–mediator release suggests that the recruitment of T cells in this early phase probably involves lung-resident allergen-specific memory cells.

During the late, T cell–mediated effector phase of asthma, initial T cell recruitment is probably facilitated by allergen- and damage-associated molecular pattern-mediated activation of pattern recognition receptors, such as Toll-like receptors (TLRs) or C-type lectins on innate immune cells, or by direct activation of epithelial cells by allergens. This induces the production of TH2-promoting cytokines and chemokines by epithelial cells and innate tissue leukocytes and an initial phase of STAT6-independent recruitment of a small number of allergen-specific T cells into the lungs121. The kinetics of T cell recruitment in this initial phase suggests that it may also involve lung-resident allergen-specific memory and effector cells. For example, TLR activation and allergen exposure are reported to induce CXCL10 and CCL20 production by macrophages and epithelial cells123,130. The cognate receptors of CXCL10 and CCL20, CXCR3 and CCR6, respectively, are expressed by human T cells that infiltrate the lungs during allergic inflammation, particularly the airways 24 h after allergen challenge49,131,132. Moreover Ccr6−/− mice have diminished airway hyper-responsiveness and allergic inflammation, and adoptive transfer of Ccr6+/+ T cells into Ccr6−/− mice increases allergic lung inflammation133,134. Additionally, tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) enhances the production of CCL20 by epithelial cells, perpetuating allergic inflammation through increased recruitment of CCR6+ memory and effector TH2 cells into the airways135. Likewise, findings with Cxcl10−/− and CXCL10 transgenic mice suggest that CXCR3+ T cell recruitment enhances allergic pulmonary inflammation136.

After the initial trafficking of T cells into the airways, TH2 cell recruitment into the lungs is markedly amplified after T cell activation by mature dendritic cells in the draining lymph node, where T cells also undergo rapid proliferation76. The chemokine receptors CCR4, CCR8 and, possibly, CX3CR1 are the likely key chemokine receptors that regulate TH2 cell recruitment into the lung during this phase. Concentrations of CCR4, CCR8 and their ligands are elevated in the lung or the bronchoalveolar lavage fluid of patients with asthma and in mouse models after allergen challenge137140. However, deletion of CCR4 and CCR8 receptors in mouse models of asthma and in human studies has yielded conflicting results126,141146. Thus, aside from in two studies126,141, CCR8 has not been shown to drive TH2-type effector cell recruitment in models of allergic airway inflammation126,141,143,145,146. Likewise, 24 h after allergen challenge of steroid-naive subjects with mild asthma, there was no difference in the expression of CCR4 or its ligands in the bronchoalveolar lavage fluid, although CCR4 was required for efficient tissue entry of TH2 cells in an adoptive transfer model of acute allergic pulmonary inflammation49,146. In a recent study, CCR4+ T cells in the peripheral blood positively correlated with asthma severity in a cohort that included subjects with mild asthma as well as steroid-dependent subjects with moderate to severe asthma, and ex vivo production of CCL17 was higher in bronchial biopsy explants from subjects with asthma compared to healthy controls147. Additionally, studies of airway allergen challenge and genome-wide association have implicated CX3CR1 and its ligand CX3CL1 in human asthma108,148. A recent study provided a possible mechanism for this association by showing that CX3CR1 signaling aided TH2 cell survival and maintenance to drive allergic pulmonary inflammation107. Facilitating the long-term persistence of allergen-specific memory and effector T cells is also a mechanism by which the CCR8 pathway may promote chronic allergic inflammation149.

To define the transcriptional signals that regulate the induction of inflammatory chemokines and amplify T cell recruitment into the asthmatic lung, we used an adoptive transfer model of acute allergic inflammation and showed that it is STAT6 dependent121. T cell recruitment was dependent on STAT6 expression by a resident lung cell, which we subsequently showed to be from a subset of CD11b+CD11c+ myeloid pulmonary dendritic cells, concordant with findings from other groups150152. Thus, in this model of acute asthma, Stat6−/− mice had significantly lower production of the TH2 cell–attracting chemokines CCL17 and CCL22, as well as decreased induction of the eosinophil-attracting chemokines CCL11 and CCL24. Moreover, CCL17, CCL22 and CCL11 were produced in a STAT6-dependent fashion in a 4-week active immunization model of allergic pulmonary inflammation153. In contrast, the transcriptional signals that regulate the induction of CCR8 and CX3CR1 in the allergic lung are not as well defined. Lung mCCL8 induction was STAT6 independent in a chronic 4-week active immunization model153. With respect to other T helper subsets, it is notable that Treg cells express the chemokine receptors CCR4 and CCR8 and, thus, may be recruited by the release of CCL1 during the early phase asthmatic response and the release of CCL17, CCL22 and mCCL8 during the late phase asthmatic response69.

Food allergy

In humans, allergen-specific TH2 cells are essential for triggering non–IgE-mediated food allergies as well as maintaining type I IgE-mediated allergic responses. In allergic individuals, allergen-specific T cells in the blood proliferate more in response to food allergens compared to in non-allergic individuals, and human milk-specific T cells derived from the duodenum have a dominant TH2-type cytokine secretion profile154,155. After food challenges, the duodena of symptomatic individuals contain increased numbers of IL-4+CD4+ T cells associated with IgE+ cells that were mostly mast cells despite lacking food-allergen–specific serum IgE156. With regard to programmed organ-specific trafficking molecules, the integrin α4β7 is selectively increased in lymphocytes stimulated with β-lactoglobulin in children allergic to cow’s milk157. Peach-protein-allergen–specific T cell clones are also selectively enriched for α4β7 and lack expression of CLA158. Thus food-allergen–specific T cells in humans are imprinted with gut-specific homing receptors. Milk-induced skin urticaria is also a common manifestation of milk allergy in young children, and milk exposure induces symptoms of eczema in individuals with atopic dermatitis159,160. Notably, milk-induced urticaria and atopic eczema are both associated with increased circulating concentrations of allergen-specific T lymphocytes, which express CLA159,160. Additionally, a recent human study found that anaphylactic peanut allergy is associated with an increased frequency in the number of blood IL-5IL-4+ peanut-specific TH2 cells and IgE responses161. Conversely, blood IL-5+IL-4+ peanut-specific TH2 cells are associated with IgE-independent allergic eosinophilic gastroenteritis161. Extending these observations, the majority of peanut-allergen–specific blood CD4 T cells express CCR4 in individuals with a history of peanut anaphylaxis and peanut-specific IgE, and peanut-allergen–specific cell lines from these individuals primarily produce IL-4 (ref. 162).

In the mouse, CCR7 expression on Treg cells is crucial for the induction of oral tolerance in the MLN, and Treg cells are directed to home to the gut lamina propria by MADCAM1 (refs. 67,71). In a mast-cell–dependent mouse model of chronic food allergy in which inflammation is induced in the small intestine and manifests as allergic diarrhea, allergen-specific T cells are induced in the MLN and traffic to the small intestine163. In this model, increased numbers of TH2 cytokines and the CCR8 and CCR4 ligands CCL1, CCL17 and CCL22 are upregulated in the inflamed small intestine163. In this same model, CCR6 is crucial for T cell trafficking to the small intestine and for driving IgE-independent TH2 cell inflammation and allergic diarrhea164. Thus, chemokine receptors associated with allergic airway inflammation also seem to drive allergic inflammation in the gut.

Therapeutic targeting of leukocyte trafficking in allergy

We suggest that programmed tissue-specific T cell trafficking pathways during immune surveillance and inflammation contribute to the propensity of the skin, gut and lung for developing allergic diseases. Blocking tissue- and inflammation-specific trafficking pathways provides an opportunity for developing new targeted therapies for inflammation and allergic diseases and is actively being pursued (Table 1). Integrins were the first category of trafficking receptors to be targeted for therapy. Efalizumab, an antibody to CD11a that inhibits skin-homing CLA+ T cell entry into the skin, was the first migration-inhibitory drug to be approved by the US Food and Drug Administration, and although it was initially approved for the treatment of psoriasis, it was also reported to be effective in some individuals with atopic dermatitis165. Among the chemoattractant receptors, the first drug to successfully reach phase 3 clinical trials for an inflammatory disease was, in fact, an antagonist for another tissue-specific homing molecule, CCR9, for the treatment of Crohn’s disease. With regard to allergic inflammation, based on population-based genetic studies of an association of CRTH2 polymorphisms with allergic disease, many companies are actively pursuing inhibitors of CRTH2, some of which have recently completed phase 2 trials81,128,129,166. Other chemokine receptors being targeted for anti-allergic therapy are CCR3 and CCR4. An advantage of anti-migration–based therapy is that the likelihood of toxicity is less than that of immunosuppressive therapy. However, a caveat of extended anti-integrin–based therapies has been an unpredicted increased risk for developing fatal progressive multifocal leukoencephalopathy167.

Table 1.

Drugs in development or currently available that inhibit leukocyte migration

Target molecule Target cell Compound or drug Company Clinical indication and stage of development
CRTH2 (DP2) T cell, eosinophil and basophil QAW039, oral antagonist Novartis Moderate to severe asthma with sputum eosinophilia; phase 2, currently recruiting
ADC3680B, oral antagonist Pulmagen Partly controlled mild to moderate atopic asthma; phase 2 completed February 2012
MK-7246, oral antagonist Merck In development
AZD1981, oral antagonist AstraZeneca In development
OC000459, oral antagonist Oxagen In development
AM156, topical antagonist Amira Allergic conjunctivitis; in development
Antagonist Pfizer/Wyeth In development
Antagonist Millenium In development

CRTH2 and DP1 T cell, eosinophil and basophil AMG 853, oral antagonist Amgen Asthma; phase 2 completed March 2011
AMG-009, oral antagonist Amgen Asthma and allergic rhinitis; phase 1 planned

CCR3 Eosinophil GW-766904, oral antagonist GlaxoSmithKline Mild to moderate asthmatics with sputum eosinophilia; phase 2, currently recruiting

CCR4 T cell KW-0761, intravenous antibody Kyowa Hakko Allergic rhinitis and healthy volunteers; phase 1 completed
Kyowa Hakko Adult T cell leukemia-lymphoma; phase 2 completed April 2011
Amgen Non-oncology, asthma and inflammatory indications; in development
Antagonist Astellas In development
Antagonist Bristol-Meyers Squibb In development

CCR9 T cell GSK1605786A ChemoCentryx Crohn’s Disease; phase 1 and 2 completed
Oral antagonist GlaxoSmithKline Crohn’s Disease; phase 3 recruiting

LFA1 (CD11a–CD18; αLβ2 integrin) T cell Efalizumab, subcutaneous antibody Genentech US Food and Drug Administration (FDA) approved in 2003 for treatment of moderate to severe psoriasis. Voluntarily withdrawn in 2009 because of association with fatal progressive multifocal leukoencephalopathy (PML)

VLA-4 (α4β1 integrin) T cell Natal izumab, intravenous antibody Biogen Idec FDA approved in 2004 for relapsing-remitting multiple sclerosis. FDA black box warning: increased risk of PML. FDA approved in 2008 for moderate to severe Crohn’s disease

In developing therapies based on anti-chemokine receptors, as we highlight in this review, the kinetics of inflammation-driven chemokine induction and T cell chemokine receptor expression during chronic allergic disease dictates the relative roles of different ligand-receptor pairs during distinct phases of disease and in different compartments. Large pre-existing pools of tissue-resident allergen-specific memory T cell pools also probably influence migration-inhibitor–based therapies. Though classical TH2 cells are the main orchestrators of allergic inflammation through the production of TH2 effector cytokines, other T helper subsets contribute to allergic inflammation. These factors reflect the importance of multiple T cell trafficking receptor pathways in allergic inflammation. Thus, the strategic design of new therapeutic agents directed against trafficking-receptor pathways for the treatment of allergic inflammation requires an appreciation of these multiple factors.

ACKNOWLEDGMENTS

The authors were supported by US National Institutes of Health grants R37AI040618 and U19AI095261 to A.D.L.

Footnotes

COMPETING FINANCIAL INTERESTS

The authors declare no competing financial interests.

References

  • 1.Cookson W. The immunogenetics of asthma and eczema: a new focus on the epithelium. Nat. Rev. Immunol. 2004;4:978–988. doi: 10.1038/nri1500. [DOI] [PubMed] [Google Scholar]
  • 2.Rodriguez E, et al. Meta-analysis of filaggrin polymorphisms in eczema and asthma: robust risk factors in atopic disease. J. Allergy Clin. Immunol. 2009;123:1361–1370. doi: 10.1016/j.jaci.2009.03.036. [DOI] [PubMed] [Google Scholar]
  • 3.Al-Shami A, et al. A role for TSLP in the development of inflammation in an asthma model. J. Exp. Med. 2005;202:829–839. doi: 10.1084/jem.20050199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Eiwegger T, Akdis CA. IL-33 links tissue cells, dendritic cells and Th2 cell development in a mouse model of asthma. Eur. J. Immunol. 2011;41:1535–1538. doi: 10.1002/eji.201141668. [DOI] [PubMed] [Google Scholar]
  • 5.Fort MM, et al. IL-25 induces IL-4, IL-5, and IL-13 and Th2-associated pathologiesin vivo . Immunity. 2001;15:985–995. doi: 10.1016/s1074-7613(01)00243-6. [DOI] [PubMed] [Google Scholar]
  • 6.Hammad H, et al. House dust mite allergen induces asthma via Toll-like receptor 4 triggering of airway structural cells. Nat. Med. 2009;15:410–416. doi: 10.1038/nm.1946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hvid M, et al. IL-25 in atopic dermatitis: a possible link between inflammation and skin barrier dysfunction? J. Invest. Dermatol. 2011;131:150–157. doi: 10.1038/jid.2010.277. [DOI] [PubMed] [Google Scholar]
  • 8.Paul WE, Zhu J. How are TH2-type immune responses initiated and amplified? Nat. Rev. Immunol. 2010;10:225–235. doi: 10.1038/nri2735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Préfontaine D, et al. Increased expression of IL-33 in severe asthma: evidence of expression by airway smooth muscle cells. J. Immunol. 2009;183:5094–5103. doi: 10.4049/jimmunol.0802387. [DOI] [PubMed] [Google Scholar]
  • 10.Rank MA, et al. IL-33–activated dendritic cells induce an atypical TH2-type response. J. Allergy Clin. Immunol. 2009;123:1047–1054. doi: 10.1016/j.jaci.2009.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Luster AD, Alon R, von Andrian UH. Immune cell migration in inflammation: present and future therapeutic targets. Nat. Immunol. 2005;6:1182–1190. doi: 10.1038/ni1275. [DOI] [PubMed] [Google Scholar]
  • 12.Agace WW. Tissue-tropic effector T cells: generation and targeting opportunities. Nat. Rev. Immunol. 2006;6:682–692. doi: 10.1038/nri1869. [DOI] [PubMed] [Google Scholar]
  • 13.Bromley SK, Mempel TR, Luster AD. Orchestrating the orchestrators: chemokines in control of T cell traffic. Nat. Immunol. 2008;9:970–980. doi: 10.1038/ni.f.213. [DOI] [PubMed] [Google Scholar]
  • 14.Sallusto F, Mackay CR. Chemoattractants and their receptors in homeostasis and inflammation. Curr. Opin. Immunol. 2004;16:724–731. doi: 10.1016/j.coi.2004.09.012. [DOI] [PubMed] [Google Scholar]
  • 15.Sallusto F, Mackay CR, Lanzavecchia A. The role of chemokine receptors in primary, effector, and memory immune responses. Annu. Rev. Immunol. 2000;18:593–620. doi: 10.1146/annurev.immunol.18.1.593. [DOI] [PubMed] [Google Scholar]
  • 16.Sallusto F, et al. Flexible programs of chemokine receptor expression on human polarized T helper 1 and 2 lymphocytes. J. Exp. Med. 1998;187:875–883. doi: 10.1084/jem.187.6.875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Cahill RN, et al. Two distinct pools of recirculating T lymphocytes: migratory characteristics of nodal and intestinal T lymphocytes. J. Exp. Med. 1977;145:420–428. doi: 10.1084/jem.145.2.420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Berg EL, et al. The cutaneous lymphocyte antigen is a skin lymphocyte homing receptor for the vascular lectin endothelial cell-leukocyte adhesion molecule 1. J. Exp. Med. 1991;174:1461–1466. doi: 10.1084/jem.174.6.1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Berlin C, et al. alpha 4 integrins mediate lymphocyte attachment and rolling under physiologic flow. Cell. 1995;80:413–422. doi: 10.1016/0092-8674(95)90491-3. [DOI] [PubMed] [Google Scholar]
  • 20.Picker LJ, et al. ELAM-1 is an adhesion molecule for skin-homing T cells. Nature. 1991;349:796–799. doi: 10.1038/349796a0. [DOI] [PubMed] [Google Scholar]
  • 21.Picker LJ, et al. Differential expression of homing-associated adhesion molecules by T cell subsets in man. J. Immunol. 1990;145:3247–3255. [PubMed] [Google Scholar]
  • 22.Sigmundsdottir H, Butcher EC. Environmental cues, dendritic cells and the programming of tissue-selective lymphocyte trafficking. Nat. Immunol. 2008;9:981–987. doi: 10.1038/ni.f.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Briskin M, et al. Human mucosal addressin cell adhesion molecule-1 is preferentially expressed in intestinal tract and associated lymphoid tissue. Am. J. Pathol. 1997;151:97–110. [PMC free article] [PubMed] [Google Scholar]
  • 24.Kunkel EJ, et al. Lymphocyte CC chemokine receptor 9 and epithelial thymus-expressed chemokine (TECK) expression distinguish the small intestinal immune compartment: epithelial expression of tissue-specific chemokines as an organizing principle in regional immunity. J. Exp. Med. 2000;192:761–768. doi: 10.1084/jem.192.5.761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Campbell JJ, et al. The chemokine receptor CCR4 in vascular recognition by cutaneous but not intestinal memory T cells. Nature. 1999;400:776–780. doi: 10.1038/23495. [DOI] [PubMed] [Google Scholar]
  • 26.Duhen T, et al. Production of interleukin 22 but not interleukin 17 by a subset of human skin-homing memory T cells. Nat. Immunol. 2009;10:857–863. doi: 10.1038/ni.1767. [DOI] [PubMed] [Google Scholar]
  • 27.Fuhlbrigge RC, et al. Cutaneous lymphocyte antigen is a specialized form of PSGL-1 expressed on skin-homing T cells. Nature. 1997;389:978–981. doi: 10.1038/40166. [DOI] [PubMed] [Google Scholar]
  • 28.Homey B, et al. CCL27-CCR10 interactions regulate T cell–mediated skin inflammation. Nat. Med. 2002;8:157–165. doi: 10.1038/nm0202-157. [DOI] [PubMed] [Google Scholar]
  • 29.Islam SA, et al. Mouse CCL8, a CCR8 agonist, promotes atopic dermatitis by recruiting IL-5+ TH2 cells. Nat. Immunol. 2011;12:167–177. doi: 10.1038/ni.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Schaerli P, et al. A skin-selective homing mechanism for human immune surveillance T cells. J. Exp. Med. 2004;199:1265–1275. doi: 10.1084/jem.20032177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Trifari S, et al. Identification of a human helper T cell population that has abundant production of interleukin 22 and is distinct from TH-17, TH1 and TH2 cells. Nat. Immunol. 2009;10:864–871. doi: 10.1038/ni.1770. [DOI] [PubMed] [Google Scholar]
  • 32.Clark RA, et al. The vast majority of CLA+ T cells are resident in normal skin. J. Immunol. 2006;176:4431–4439. doi: 10.4049/jimmunol.176.7.4431. [DOI] [PubMed] [Google Scholar]
  • 33.Charbonnier AS, et al. Macrophage inflammatory protein 3a is involved in the constitutive trafficking of epidermal langerhans cells. J. Exp. Med. 1999;190:1755–1768. doi: 10.1084/jem.190.12.1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Gombert M, et al. CCL1-CCR8 interactions: an axis mediating the recruitment of T cells and Langerhans-type dendritic cells to sites of atopic skin inflammation. J. Immunol. 2005;174:5082–5091. doi: 10.4049/jimmunol.174.8.5082. [DOI] [PubMed] [Google Scholar]
  • 35.Morales J, et al. CTACK, a skin-associated chemokine that preferentially attracts skin-homing memory T cells. Proc. Natl. Acad. Sci. USA. 1999;96:14470–14475. doi: 10.1073/pnas.96.25.14470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Weninger W, et al. Specialized contributions by α (1,3)-fucosyltransferase-IV and FucT-VII during leukocyte rolling in dermal microvessels. Immunity. 2000;12:665–676. doi: 10.1016/s1074-7613(00)80217-4. [DOI] [PubMed] [Google Scholar]
  • 37.Annacker O, et al. Essential role for CD103 in the T cell-mediated regulation of experimental colitis. J. Exp. Med. 2005;202:1051–1061. doi: 10.1084/jem.20040662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Edele F, et al. Cutting edge: instructive role of peripheral tissue cells in the imprinting of T cell homing receptor patterns. J. Immunol. 2008;181:3745–3749. doi: 10.4049/jimmunol.181.6.3745. [DOI] [PubMed] [Google Scholar]
  • 39.Johansson-Lindbom B, et al. Functional specialization of gut CD103+ dendritic cells in the regulation of tissue-selective T cell homing. J. Exp. Med. 2005;202:1063–1073. doi: 10.1084/jem.20051100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Mora JR, et al. Selective imprinting of gut-homing T cells by Peyer’s patch dendritic cells. Nature. 2003;424:88–93. doi: 10.1038/nature01726. [DOI] [PubMed] [Google Scholar]
  • 41.Iwata M, et al. Retinoic acid imprints gut-homing specificity on T cells. Immunity. 2004;21:527–538. doi: 10.1016/j.immuni.2004.08.011. [DOI] [PubMed] [Google Scholar]
  • 42.Mora JR, et al. Reciprocal and dynamic control of CD8 T cell homing by dendritic cells from skin- and gut-associated lymphoid tissues. J. Exp. Med. 2005;201:303–316. doi: 10.1084/jem.20041645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sigmundsdottir H, et al. DCs metabolize sunlight-induced vitamin D3 to ‘program’ T cell attraction to the epidermal chemokine CCL27. Nat. Immunol. 2007;8:285–293. doi: 10.1038/ni1433. [DOI] [PubMed] [Google Scholar]
  • 44.Ohmichi Y, et al. Essential role of peripheral node addressin in lymphocyte homing to nasal-associated lymphoid tissues and allergic immune responses. J. Exp. Med. 2011;208:1015–1025. doi: 10.1084/jem.20101786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.de Bree GJ, et al. Selective accumulation of differentiated CD8+ T cells specific for respiratory viruses in the human lung. J. Exp. Med. 2005;202:1433–1442. doi: 10.1084/jem.20051365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Koelle DM, et al. Expression of cutaneous lymphocyte–associated antigen by CD8+ T cells specific for a skin-tropic virus. J. Clin. Invest. 2002;110:537–548. doi: 10.1172/JCI15537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Campbell JJ, et al. Expression of chemokine receptors by lung T cells from normal and asthmatic subjects. J. Immunol. 2001;166:2842–2848. doi: 10.4049/jimmunol.166.4.2842. [DOI] [PubMed] [Google Scholar]
  • 48.Purwar R, et al. Resident memory T cells (T(RM)) are abundant in human lung: diversity, function, and antigen specificity. PLoS ONE. 2011;6:el6245. doi: 10.1371/journal.pone.0016245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Thomas SY, et al. Multiple chemokine receptors, including CCR6 and CXCR3, regulate antigen-induced T cell homing to the human asthmatic airway. J. Immunol. 2007;179:1901–1912. doi: 10.4049/jimmunol.179.3.1901. [DOI] [PubMed] [Google Scholar]
  • 50.Sheridan BS, Lefrancois L. Regional and mucosal memory T cells. Nat. Immunol. 2011;121:485–491. doi: 10.1038/ni.2029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Sallusto F, et al. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–712. doi: 10.1038/44385. [DOI] [PubMed] [Google Scholar]
  • 52.Reinhardt RL, et al. Visualizing the generation of memory CD4 T cells in the whole body. Nature. 2001;410:101–105. doi: 10.1038/35065111. [DOI] [PubMed] [Google Scholar]
  • 53.Conrad C, et al. α1β1 integrin is crucial for accumulation of epidermal T cells and the development of psoriasis. Nat Med. 2007;13:836–842. doi: 10.1038/nm1605. [DOI] [PubMed] [Google Scholar]
  • 54.Gebhardt T, et al. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat. Immunol. 2009;10:524–530. doi: 10.1038/ni.1718. [DOI] [PubMed] [Google Scholar]
  • 55.Klonowski KD, et al. Dynamics of blood-borne CD8 memory T cell migration in vivo . Immunity. 2004;20:551–562. doi: 10.1016/s1074-7613(04)00103-7. [DOI] [PubMed] [Google Scholar]
  • 56.Masopust D, et al. Dynamic T cell migration program provides resident memory within intestinal epithelium. J. Exp. Med. 2010;207:553–564. doi: 10.1084/jem.20090858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Vezys V, et al. Memory CD8 T-cell compartment grows in size with immunological experience. Nature. 2009;457:196–199. doi: 10.1038/nature07486. [DOI] [PubMed] [Google Scholar]
  • 58.Wakim LM, et al. Dendritic cell-induced memory T cell activation in nonlymphoid tissues. Science. 2008;319:198–202. doi: 10.1126/science.1151869. [DOI] [PubMed] [Google Scholar]
  • 59.Lambrecht BN, Hammad H. Biology of lung dendritic cells at the origin of asthma. Immunity. 2009;31:412–424. doi: 10.1016/j.immuni.2009.08.008. [DOI] [PubMed] [Google Scholar]
  • 60.Adorini L. Tolerogenic dendritic cells induced by vitamin D receptor ligands enhance regulatory T cells inhibiting autoimmune diabetes. Ann. NY Acad. Sci. 2003;987:258–261. doi: 10.1111/j.1749-6632.2003.tb06057.x. [DOI] [PubMed] [Google Scholar]
  • 61.Benson MJ, et al. All-trans retinoic acid mediates enhanced Treg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation. J. Exp. Med. 2007;204:1765–1774. doi: 10.1084/jem.20070719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Boonstra A, et al. 1α,25-Dihydroxyvitamin d3 has a direct effect on naive CD4+ T cells to enhance the development of Th2 cells. J. Immunol. 2001;167:4974–4980. doi: 10.4049/jimmunol.167.9.4974. [DOI] [PubMed] [Google Scholar]
  • 63.Coombes JL, et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-β and retinoic acid-dependent mechanism. J. Exp. Med. 2007;204:1757–1764. doi: 10.1084/jem.20070590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Iwata M, Eshima Y, Kagechika H. Retinoic acids exert direct effects on T cells to suppress Th1 development and enhance Th2 development via retinoic acid receptors. Int. Immunol. 2003;15:1017–1025. doi: 10.1093/intimm/dxg101. [DOI] [PubMed] [Google Scholar]
  • 65.Sun CM, et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 Treg cells via retinoic acid. J. Exp. Med. 2007;204:1775–1785. doi: 10.1084/jem.20070602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mucida D, et al. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science. 2007;317:256–260. doi: 10.1126/science.1145697. [DOI] [PubMed] [Google Scholar]
  • 67.Hadis U, et al. Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity. 2011;34:237–246. doi: 10.1016/j.immuni.2011.01.016. [DOI] [PubMed] [Google Scholar]
  • 68.DePaolo RW, et al. Co-adjuvant effects of retinoic acid and IL-15 induce inflammatory immunity to dietary antigens. Nature. 2011;471:220–224. doi: 10.1038/nature09849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Iellem A, et al. Unique chemotactic response profile and specific expression of chemokine receptors CCR4 and CCR8 by CD4+CD25+ regulatory T cells. J. Exp. Med. 2001;194:847–853. doi: 10.1084/jem.194.6.847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Siegmund K, et al. Migration matters: regulatory T-cell compartmentalization determines suppressive activity in vivo . Blood. 2005;106:3097–3104. doi: 10.1182/blood-2005-05-1864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Worbs T, et al. Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells. J. Exp. Med. 2006;203:519–527. doi: 10.1084/jem.20052016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Worbs T, Forster R. A key role for CCR7 in establishing central and peripheral tolerance. Trends Immunol. 2007;28:274–280. doi: 10.1016/j.it.2007.04.002. [DOI] [PubMed] [Google Scholar]
  • 73.Zammit DJ, et al. Residual antigen presentation after influenza virus infection affects CD8 T cell activation and migration. Immunity. 2006;24:439–449. doi: 10.1016/j.immuni.2006.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.McLachlan JB, et al. Dendritic cell antigen presentation drives simultaneous cytokine production by effector and regulatory T cells in inflamed skin. Immunity. 2009;30:277–288. doi: 10.1016/j.immuni.2008.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Boyman O, et al. Spontaneous development of psoriasis in a new animal model shows an essential role for resident T cells and tumor necrosis factor-α. J. Exp. Med. 2004;199:731–736. doi: 10.1084/jem.20031482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Harris NL, et al. Differential T cell function and fate in lymph node and nonlymphoid tissues. J. Exp. Med. 2002;195:317–326. doi: 10.1084/jem.20011558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell populations. Annu. Rev. Immunol. 2010;28:445–489. doi: 10.1146/annurev-immunol-030409-101212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Ansel KM, Lee DU, Rao A. An epigenetic view of helper T cell differentiation. Nat Immunol. 2003;4:616–623. doi: 10.1038/ni0703-616. [DOI] [PubMed] [Google Scholar]
  • 79.Sallusto F, Lanzavecchia A, Mackay CR. Chemokines and chemokine receptors in T-cell priming and Th1/Th2-mediated responses. Immunol. Today. 1998;19:568–574. doi: 10.1016/s0167-5699(98)01346-2. [DOI] [PubMed] [Google Scholar]
  • 80.Abe H, et al. Molecular cloning, chromosome mapping and characterization of the mouse CRTH2 gene, a putative member of the leukocyte chemoattractant receptor family. Gene. 1999;227:71–77. doi: 10.1016/s0378-1119(98)00599-x. [DOI] [PubMed] [Google Scholar]
  • 81.Hirai H, et al. Prostaglandin D2 selectively induces chemotaxis in T helper type 2 cells, eosinophils, and basophils via seven-transmembrane receptor CRTH2. J. Exp. Med. 2001;193:255–261. doi: 10.1084/jem.193.2.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Upadhyaya B, et al. Hierarchical IL-5 expression defines a subpopulation of highly differentiated human Th2 cells. J. Immunol. 2011;187:3111–3120. doi: 10.4049/jimmunol.1101283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kim CH, Nagata K, Butcher EC. Dendritic cells support sequential reprogramming of chemoattractant receptor profiles during naive to effector T cell differentiation. J. Immunol. 2003;171:152–158. doi: 10.4049/jimmunol.171.1.152. [DOI] [PubMed] [Google Scholar]
  • 84.D’Ambrosio D, et al. Selective up-regulation of chemokine receptors CCR4 and CCR8 upon activation of polarized human type 2 Th cells. J. Immunol. 1998;161:5111–5115. [PubMed] [Google Scholar]
  • 85.Wei L, et al. Discrete roles of STAT4 and STAT6 transcription factors in tuning epigenetic modifications and transcription during T helper cell differentiation. Immunity. 2010;32:840–851. doi: 10.1016/j.immuni.2010.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Meisel C, et al. Regulation and function of T1/ST2 expression on CD4+ T cells: induction of type 2 cytokine production by T1/ST2 cross-linking. J. Immunol. 2001;166:3143–3150. doi: 10.4049/jimmunol.166.5.3143. [DOI] [PubMed] [Google Scholar]
  • 87.Monney L, et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature. 2002;415:536–541. doi: 10.1038/415536a. [DOI] [PubMed] [Google Scholar]
  • 88.Hegazy AN, et al. Interferons direct Th2 cell reprogramming to generate a stable GATA-3+T-bet+ cell subset with combined Th2 and Th1 cell functions. Immunity. 2010;32:116–128. doi: 10.1016/j.immuni.2009.12.004. [DOI] [PubMed] [Google Scholar]
  • 89.Wang YH, et al. A novel subset of CD4+ T(H)2 memory/effector cells that produce inflammatory IL-17 cytokine and promote the exacerbation of chronic allergic asthma. J. Exp. Med. 2010;207:2479–2491. doi: 10.1084/jem.20101376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zhu J, Paul WE. CD4+ T cell plasticity-Th2 cells join the crowd. Immunity. 2010;32:11–13. doi: 10.1016/j.immuni.2010.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Corren J, et al. Lebrikizumab treatment in adults with asthma. N. Engl. J. Med. 2011;365:1088–1098. doi: 10.1056/NEJMoa1106469. [DOI] [PubMed] [Google Scholar]
  • 92.Kraft M. Asthma phenotypes and interleukin-13—moving closer to personalized medicine. N. Engl. J. Med. 2011;365:1141–1144. doi: 10.1056/NEJMe1108666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kim CH, et al. Rules of chemokine receptor association with T cell polarization. in vivo. J. Clin. Invest. 2001;108:1331–1339. doi: 10.1172/JCI13543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Leung DY, Bieber T. Atopic dermatitis. Lancet. 2003;361:151–160. doi: 10.1016/S0140-6736(03)12193-9. [DOI] [PubMed] [Google Scholar]
  • 95.Woodward AL, et al. An obligate role for T-cell receptor αβ+ T cells but not T-cell receptor γδ+ T cells, B cells, or CD40/CD40L interactions in a mouse model of atopic dermatitis. J. Allergy Clin. Immunol. 2001;107:359–366. doi: 10.1067/mai.2001.112695. [DOI] [PubMed] [Google Scholar]
  • 96.Kakinuma T, et al. Increased serum cutaneous T cell-attracting chemokine (CCL27) levels in patients with atopic dermatitis and psoriasis vulgaris. J. Allergy Clin. Immunol. 2003;111:592–597. doi: 10.1067/mai.2003.114. [DOI] [PubMed] [Google Scholar]
  • 97.Leung TF, et al. Plasma concentration of thymus and activation-regulated chemokine is elevated in childhood asthma. J. Allergy Clin. Immunol. 2002;110:404–409. doi: 10.1067/mai.2002.126378. [DOI] [PubMed] [Google Scholar]
  • 98.Shimada Y, Takehara K, Sato S. Both Th2 and Th1 chemokines (TARC/CCL17, MDC/CCL22, and Mig/CXCL9) are elevated in sera from patients with atopic dermatitis. J. Dermatol. Sci. 2004;34:201–208. doi: 10.1016/j.jdermsci.2004.01.001. [DOI] [PubMed] [Google Scholar]
  • 99.Vestergaard C, et al. Expression of the T-helper 2–specific chemokine receptor CCR4 on CCR10-positive lymphocytes in atopic dermatitis skin but not in psoriasis skin. Br. J. Dermatol. 2003;149:457–463. doi: 10.1046/j.1365-2133.2003.05505.x. [DOI] [PubMed] [Google Scholar]
  • 100.Lonsdorf AS, Hwang ST, Enk AH. Chemokine receptors in T-cell-mediated diseases of the skin. J. Invest. Dermatol. 2009;129:2552–2566. doi: 10.1038/jid.2009.122. [DOI] [PubMed] [Google Scholar]
  • 101.Pivarcsi A, et al. CC chemokine ligand 18, an atopic dermatitis-associated and dendritic cell-derived chemokine, is regulated by staphylococcal products and allergen exposure. J. Immunol. 2004;173:5810–5817. doi: 10.4049/jimmunol.173.9.5810. [DOI] [PubMed] [Google Scholar]
  • 102.Günther C, et al. CCL18 is expressed in atopic dermatitis and mediates skin homing of human memory T cells. J. Immunol. 2005;174:1723–1728. doi: 10.4049/jimmunol.174.3.1723. [DOI] [PubMed] [Google Scholar]
  • 103.Hijnen D, et al. Differential expression of genes involved in skin homing, proliferation, and apoptosis in CD4+ T cells of patients with atopic dermatitis. J. Invest. Dermatol. 2005;125:1149–1155. doi: 10.1111/j.0022-202X.2005.23932.x. [DOI] [PubMed] [Google Scholar]
  • 104.Hijnen D, et al. Serum thymus and activation-regulated chemokine (TARC) and cutaneous T cell- attracting chemokine (CTACK) levels in allergic diseases: TARC and CTACK are disease-specific markers for atopic dermatitis. J. Allergy Clin. Immunol. 2004;113:334–340. doi: 10.1016/j.jaci.2003.12.007. [DOI] [PubMed] [Google Scholar]
  • 105.Santamaria Babi LF, et al. Circulating allergen-reactive T cells from patients with atopic dermatitis and allergic contact dermatitis express the skin-selective homing receptor, the cutaneous lymphocyte-associated antigen. J. Exp. Med. 1995;181:1935–1940. doi: 10.1084/jem.181.5.1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Echigo T, et al. Expression of fractalkine and its receptor, CX3CR1, in atopic dermatitis: possible contribution to skin inflammation. J. Allergy Clin. Immunol. 2004;113:940–948. doi: 10.1016/j.jaci.2004.02.030. [DOI] [PubMed] [Google Scholar]
  • 107.Mionnet C, et al. CX3CR1 is required for airway inflammation by promoting T helper cell survival and maintenance in inflamed lung. Nat. Med. 2010;16:1305–1312. doi: 10.1038/nm.2253. [DOI] [PubMed] [Google Scholar]
  • 108.Tremblay K, et al. Association study between the CX3CR1 gene and asthma. Genes Immun. 2006;7:632–639. doi: 10.1038/sj.gene.6364340. [DOI] [PubMed] [Google Scholar]
  • 109.Spergel JM, et al. Roles of TH1 and TH2 cytokines in a murine model of allergic dermatitis. J. Clin. Invest. 1999;103:1103–1111. doi: 10.1172/JCI5669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Oyoshi MK, et al. Epicutaneous challenge of orally immunized mice redirects antigen-specific gut-homing T cells to the skin. J. Clin. Invest. 2011;121:2210–2220. doi: 10.1172/JCI43586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Campbell JJ, O’Connell DJ, Wurbel MA. Cutting edge: chemokine receptor CCR4 is necessary for antigen-driven cutaneous accumulation of CD4 T cells under physiological conditions. J. Immunol. 2007;178:3358–3362. doi: 10.4049/jimmunol.178.6.3358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Takatsu K, Nakajima H. IL-5 and eosinophilia. Curr. Opin. Immunol. 2008;20:288–294. doi: 10.1016/j.coi.2008.04.001. [DOI] [PubMed] [Google Scholar]
  • 113.Lloyd CM, Hessel EM. Functions of T cells in asthma: more than just TH2 cells. Nat Rev. Immunol. 2010;10:838–848. doi: 10.1038/nri2870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Robinson DS. The role of the T cell in asthma. J. Allergy Clin. Immunol. 2010;126:1081–1091. doi: 10.1016/j.jaci.2010.06.025. quiz 1092–1093. [DOI] [PubMed] [Google Scholar]
  • 115.Fairs A, et al. IgE sensitization to Aspergillus fumigatus is associated with reduced lung function in asthma. Am. J. Respir. Crit. Care Med. 2010;182:1362–1368. doi: 10.1164/rccm.201001-0087OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Kheradmand F, et al. A protease-activated pathway underlying Th cell type 2 activation and allergic lung disease. J. Immunol. 2002;169:5904–5911. doi: 10.4049/jimmunol.169.10.5904. [DOI] [PubMed] [Google Scholar]
  • 117.Lamhamedi-Cherradi SE, et al. Fungal proteases induce Th2 polarization through limited dendritic cell maturation and reduced production of IL-12. J. Immunol. 2008;180:6000–6009. doi: 10.4049/jimmunol.180.9.6000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Wark PA, et al. Asthmatic bronchial epithelial cells have a deficient innate immune response to infection with rhinovirus. J. Exp. Med. 2005;201:937–947. doi: 10.1084/jem.20041901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Muro S, Minshall EM, Hamid QA. The pathology of chronic asthma. Clin. Chest Med. 2000;21:225–244. doi: 10.1016/s0272-5231(05)70263-x. [DOI] [PubMed] [Google Scholar]
  • 120.Bentley AM, Kay AB, Durham SR. Human late asthmatic reactions. Clin. Exp. Allergy. 1997;27(suppl. 1):71–86. doi: 10.1111/j.1365-2222.1997.tb01830.x. [DOI] [PubMed] [Google Scholar]
  • 121.Mathew A, et al. Signal transducer and activator of transcription 6 controls chemokine production and T helper cell type 2 cell trafficking in allergic pulmonary inflammation. J. Exp. Med. 2001;193:1087–1096. doi: 10.1084/jem.193.9.1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Mathew A, et al. Cutting edge: Th2 cell trafficking into the allergic lung is dependent on chemoattractant receptor signaling. J. Immunol. 2002;169:651–655. doi: 10.4049/jimmunol.169.2.651. [DOI] [PubMed] [Google Scholar]
  • 123.Medoff BD, Thomas SY, Luster AD. T cell trafficking in allergic asthma: the ins and outs. Annu. Rev. Immunol. 2008;26:205–232. doi: 10.1146/annurev.immunol.26.021607.090312. [DOI] [PubMed] [Google Scholar]
  • 124.Tager AM, et al. Leukotriene B4 receptor BLT1 mediates early effector T cell recruitment. Nat Immunol. 2003;4:982–990. doi: 10.1038/ni970. [DOI] [PubMed] [Google Scholar]
  • 125.Medoff BD, et al. Antibody-antigen interaction in the airway drives early granulocyte recruitment through BLT1. Am. J. Physiol. Lung Cell. Mol. Physiol. 2006;290:L170–L178. doi: 10.1152/ajplung.00212.2005. [DOI] [PubMed] [Google Scholar]
  • 126.Gonzalo JA, et al. Coordinated involvement of mast cells and T cells in allergic mucosal inflammation: critical role of the CC chemokine ligand 1:CCR8 axis. J. Immunol. 2007;179:1740–1750. doi: 10.4049/jimmunol.179.3.1740. [DOI] [PubMed] [Google Scholar]
  • 127.Oliveira SH, Lukacs NW. Stem cell factor and igE-stimulated murine mast cells produce chemokines (CCL2, CCL17, CCL22) and express chemokine receptors. Inflamm. Res. 2001;50:168–174. doi: 10.1007/s000110050741. [DOI] [PubMed] [Google Scholar]
  • 128.Cameron L, et al. Genetic variation in CRTh2 influences development of allergic phenotypes. Allergy. 2009;64:1478–1485. doi: 10.1111/j.1398-9995.2009.02053.x. [DOI] [PubMed] [Google Scholar]
  • 129.Huang JL, et al. Sequence variants of the gene encoding chemoattractant receptor expressed on Th2 cells (CRTH2) are associated with asthma and differentially influence mRNA stability. Hum. Mol. Genet. 2004;13:2691–2697. doi: 10.1093/hmg/ddh279. [DOI] [PubMed] [Google Scholar]
  • 130.Pichavant M, et al. Asthmatic bronchial epithelium activated by the proteolytic allergen Der p 1 increases selective dendritic cell recruitment. J. Allergy Clin. Immunol. 2005;115:771–778. doi: 10.1016/j.jaci.2004.11.043. [DOI] [PubMed] [Google Scholar]
  • 131.Kawai T, et al. Lipopolysaccharide stimulates the MyD88-independent pathway and results in activation of IFN-regulatory factor 3 and the expression of a subset of lipopolysaccharide-inducible genes. J. Immunol. 2001;167:5887–5894. doi: 10.4049/jimmunol.167.10.5887. [DOI] [PubMed] [Google Scholar]
  • 132.Reibman J, et al. Airway epithelial cells release MIP-3α/CCL20 in response to cytokines and ambient particulate matter. Am. J. Respir. Cell Mol. Biol. 2003;28:648–654. doi: 10.1165/rcmb.2002-0095OC. [DOI] [PubMed] [Google Scholar]
  • 133.Lukacs NW, et al. Requirement for the chemokine receptor CCR6 in allergic pulmonary inflammation. J. Exp. Med. 2001;194:551–555. doi: 10.1084/jem.194.4.551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Lundy SK, et al. Attenuation of allergen-induced responses in CCR6 −/− mice is dependent upon altered pulmonary T lymphocyte activation. J. Immunol. 2005;174:2054–2060. doi: 10.4049/jimmunol.174.4.2054. [DOI] [PubMed] [Google Scholar]
  • 135.Weckmann M, et al. Critical link between TRAIL and CCL20 for the activation of TH2 cells and the expression of allergic airway disease. Nat. Med. 2007;13:1308–1315. doi: 10.1038/nm1660. [DOI] [PubMed] [Google Scholar]
  • 136.Medoff BD, et al. IFN-γ-inducible protein 10 (CXCL10) contributes to airway hyperreactivity and airway inflammation in a mouse model of asthma. J. Immunol. 2002;168:5278–5286. doi: 10.4049/jimmunol.168.10.5278. [DOI] [PubMed] [Google Scholar]
  • 137.Panina-Bordignon P, et al. The C-C chemokine receptors CCR4 and CCR8 identify airway T cells of allergen-challenged atopic asthmatics. J. Clin. Invest. 2001;107:1357–1364. doi: 10.1172/JCI12655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Nouri-Aria KT, et al. CCR4 in human allergen-induced late responses in the skin and lung. Eur. J. Immunol. 2002;32:1933–1938. doi: 10.1002/1521-4141(200207)32:7<1933::AID-IMMU1933>3.0.CO;2-R. [DOI] [PubMed] [Google Scholar]
  • 139.Pilette C, et al. CCR4 ligands are up-regulated in the airways of atopic asthmatics after segmental allergen challenge. Eur. Respir. J. 2004;23:876–884. doi: 10.1183/09031936.04.00102504. [DOI] [PubMed] [Google Scholar]
  • 140.Mutalithas K, et al. Expression of CCR8 is increased in asthma. Clin. Exp. Allergy. 2010;40:1175–1185. doi: 10.1111/j.1365-2222.2010.03504.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Chensue SW, et al. Aberrant in vivo T helper type 2 cell response and impaired eosinophil recruitment in CC chemokine receptor 8 knockout mice. J. Exp. Med. 2001;193:573–584. doi: 10.1084/jem.193.5.573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Schuh JM, et al. Airway hyperresponsiveness, but not airway remodeling, is attenuated during chronic pulmonary allergic responses to Aspergillus in CCR4 −/− mice. FASEB J. 2002;16:1313–1315. doi: 10.1096/fj.02-0193fje. [DOI] [PubMed] [Google Scholar]
  • 143.Chung CD, et al. CCR8 is not essential for the development of inflammation in a mouse model of allergic airway disease. J. Immunol. 2003;170:581–587. doi: 10.4049/jimmunol.170.1.581. [DOI] [PubMed] [Google Scholar]
  • 144.Conroy DM, et al. CCR4 blockade does not inhibit allergic airways inflammation. J. Leukoc. Biol. 2003;74:558–563. doi: 10.1189/jlb.0103030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Goya I, et al. Absence of CCR8 does not impair the response to ovalbumin-induced allergic airway disease. J. Immunol. 2003;170:2138–2146. doi: 10.4049/jimmunol.170.4.2138. [DOI] [PubMed] [Google Scholar]
  • 146.Mikhak Z, et al. Contribution of CCR4 and CCR8 to antigen-specific TH2 cell trafficking in allergic pulmonary inflammation. J. Allergy Clin. Immunol. 2009;123:67–73. doi: 10.1016/j.jaci.2008.09.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Vijayanand P, et al. Chemokine receptor 4 plays a key role in T cell recruitment into the airways of asthmatic patients. J. Immunol. 2010;184:4568–4574. doi: 10.4049/jimmunol.0901342. [DOI] [PubMed] [Google Scholar]
  • 148.El-Shazly A, et al. Fraktalkine produced by airway smooth muscle cells contributes to mast cell recruitment in asthma. J. Immunol. 2006;176:1860–1868. doi: 10.4049/jimmunol.176.3.1860. [DOI] [PubMed] [Google Scholar]
  • 149.Van Snick J, et al. 1-309/T cell activation gene-3 chemokine protects murine T cell lymphomas against dexamethasone-induced apoptosis. J. Immunol. 1996;157:2570–2576. [PubMed] [Google Scholar]
  • 150.Medoff BD, et al. CD11b+ myeloid cells are the key mediators of Th2 cell homing into the airway in allergic inflammation. J. Immunol. 2009;182:623–635. doi: 10.4049/jimmunol.182.1.623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Perros F, et al. Blockade of CCR4 in a humanized model of asthma reveals a critical role for DC-derived CCL17 and CCL22 in attracting Th2 cells and inducing airway inflammation. Allergy. 2009;64:995–1002. doi: 10.1111/j.1398-9995.2009.02095.x. [DOI] [PubMed] [Google Scholar]
  • 152.Voehringer D, et al. Type 2 immunity is controlled by IL-4/IL-13 expression in hematopoietic non-eosinophil cells of the innate immune system. J. Exp. Med. 2006;203:1435–1446. doi: 10.1084/jem.20052448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Fulkerson PC, et al. Pulmonary chemokine expression is coordinately regulated by STAT1, STAT6, and IFN-γ. J. Immunol. 2004;173:7565–7574. doi: 10.4049/jimmunol.173.12.7565. [DOI] [PubMed] [Google Scholar]
  • 154.Reekers R, et al. The role of circulating food antigen-specific lymphocytes in food allergic children with atopic dermatitis. Br. J. Dermatol. 1996;135:935–941. doi: 10.1046/j.1365-2133.1996.d01-1098.x. [DOI] [PubMed] [Google Scholar]
  • 155.Beyer K, et al. Human milk-specific mucosal lymphocytes of the gastrointestinal tract display a TH2 cytokine profile. J. Allergy Clin. Immunol. 2002;109:707–713. doi: 10.1067/mai.2002.122503. [DOI] [PubMed] [Google Scholar]
  • 156.Lin XP, et al. Local allergic reaction in food-hypersensitive adults despite a lack of systemic food-specific IgE. J. Allergy Clin. Immunol. 2002;109:879–887. doi: 10.1067/mai.2002.123238. [DOI] [PubMed] [Google Scholar]
  • 157.Eigenmann PA, Tropia L, Hauser C. The mucosal adhesion receptor α4β7 integrin is selectively increased in lymphocytes stimulated with β-lactoglobulin in children allergic to cow’s milk. J. Allergy Clin. Immunol. 1999;103:931–936. doi: 10.1016/s0091-6749(99)70441-0. [DOI] [PubMed] [Google Scholar]
  • 158.Schulten V, et al. Characterization of the allergic T-cell response to Pru p 3, the nonspecific lipid transfer protein in peach. J. Allergy Clin. Immunol. 2009;124:100–107. doi: 10.1016/j.jaci.2009.02.010. [DOI] [PubMed] [Google Scholar]
  • 159.Abernathy-Carver KJ, et al. Milk-induced eczema is associated with the expansion of T cells expressing cutaneous lymphocyte antigen. J. Clin. Invest. 1995;95:913–918. doi: 10.1172/JCI117743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Beyer K, et al. Milk-induced urticaria is associated with the expansion of T cells expressing cutaneous lymphocyte antigen. J. Allergy Clin. Immunol. 2002;109:688–693. doi: 10.1067/mai.2002.123235. [DOI] [PubMed] [Google Scholar]
  • 161.Prussin C, Lee J, Foster B. Eosinophilic gastrointestinal disease and peanut allergy are alternatively associated with IL-5+ and IL-5− TH2 responses. J. Allergy Clin. Immunol. 2009;124:1326–1332. doi: 10.1016/j.jaci.2009.09.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Delong JH, et al. Ara h 1-reactive T cells in individuals with peanut allergy. J. Allergy Clin. Immunol. 2011;127:1211–1218. doi: 10.1016/j.jaci.2011.02.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Knight AK, et al. CD4 T cells activated in the mesenteric lymph node mediate gastrointestinal food allergy in mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2007;293:G1234–G1243. doi: 10.1152/ajpgi.00323.2007. [DOI] [PubMed] [Google Scholar]
  • 164.Blazquez AB, et al. A functional role for CCR6 on proallergic T cells in the gastrointestinal tract. Gastroenterology. 2010;138:275–284. doi: 10.1053/j.gastro.2009.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Harper EG, et al. Efalizumab therapy for atopic dermatitis causes marked increases in circulating effector memory CD4+ T cells that express cutaneous lymphocyte antigen. J. Invest. Dermatol. 2008;128:1173–1181. doi: 10.1038/sj.jid.5701169. [DOI] [PubMed] [Google Scholar]
  • 166.Pettipher R, Hansel TT, Armer R. Antagonism of the prostaglandin D2 receptors DP1 and CRTH2 as an approach to treat allergic diseases. Nat. Rev Drug Discov. 2007;6:313–325. doi: 10.1038/nrd2266. [DOI] [PubMed] [Google Scholar]
  • 167.Carson KR, et al. Monoclonal antibody-associated progressive multifocal leucoencephalopathy in patients treated with rituximab, natalizumab, and efalizumab: a Review from the Research on Adverse Drug Events and Reports (RADAR) Project. Lancet Oncol. 2009;10:816–824. doi: 10.1016/S1470-2045(09)70161-5. [DOI] [PubMed] [Google Scholar]

RESOURCES