Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2011 Jun 23.
Published in final edited form as: Trends Immunol. 2005 Mar;26(3):150–156. doi: 10.1016/j.it.2004.11.014

A stromal address code defined by fibroblasts

Greg Parsonage 1, Andrew D Filer 1, Oliver Haworth 1, Gerard B Nash 2, G Ed Rainger 2, Michael Salmon 1, Christopher D Buckley 1
PMCID: PMC3121558  EMSID: UKMS35689  PMID: 15745857

Abstract

To navigate into and within tissues, leukocytes require guidance cues that enable them to recognize which tissues to enter and which to avoid. Such cues are partly provided at the time of extravasation from blood by an endothelial address code on the luminal surface of the vascular endothelium. Here, we review the evidence that fibroblasts help define an additional stromal address code that directs leukocyte behaviour within tissues. We examine how this stromal code regulates site-specific leukocyte accumulation, differentiation and survival in a variety of physiological stromal niches, and how the aberrant expression of components of this code in the wrong tissue at the wrong time contributes to the persistence of chronic inflammatory diseases.


The recruitment of cells into tissue occurs across vascular endothelium and requires a combinatorial set of cellular interactions involving capture receptors (selectins), activation molecules (chemokines) and adhesion receptors (integrins). Collectively, these comprise an endothelial address code, which provides a regulatory mechanism for organ-specific trafficking of leukocytes (reviewed in Ref. [1]). Although there is overwhelming experimental support for this site-specific recruitment model, it does not address how leukocyte navigation is regulated beyond the vascular endothelium, within the tissue itself.

Once a cell has been recruited into tissue, it must make several decisions – should it briefly scan the stromal environment on its way out via draining lymphatic vessels or should it remain in the interstitium for an extended period? Should it differentiate, proliferate or die? Recent evidence suggests that stromal cells, such as fibroblasts, directly affect the behaviour of infiltrating cells by providing retention, differentiation and exit codes analogous to the endothelial entry code.

Vascular address codes

Naïve lymphocytes continuously cycle through the vascular and lymphatic circulation until they encounter their cognate antigen, displayed by professional antigen-presenting cells, in lymph nodes. They gain entry to lymph nodes because they express the correct address code: high levels of L-selectin and the chemokine receptor CCR7 [2-5]. These molecules recognize ligands, which are found on the lumenal side of lymph node high endothelial venules (HEVs). CCR7 engagement causes cells to adhere firmly to the HEVs through integrins, enabling their transit through the endothelium into the lymph node (Figure 1).

Figure 1.

Figure 1

Vascular codes provide for compartmentalization within the immune system. During the development of an immune response, immature naïve T cells (red dotted arrows) and B cells (green dotted arrows), which have been educated in the thymus and bone marrow, respectively, enter lymph nodes through the high endothelial venules (HEVs) using the endothelial entry code CCR7 and L-selectin (area codes are shown in grey). Antigen-experienced B cells (green arrows) leave the lymph node and migrate into specific niches, such as the red pulp of the spleen and the bone marrow, using the endothelial entry code CXCR4. Antigen-experienced central memory T cells continue to recirculate back to the lymph nodes via the efferent lymphatics and thoracic duct (red arrows; dot and dash). Effector memory T cells preferentially recirculate to the tissue in which they were initially activated; for example, if they are activated in a lymph node draining the skin they acquire the skin entry code CCR4, CCR10, CLA (grey) and if activated in a lymph node draining the gut they acquire the gut entry code CCR9, α4β7. At the end of immune surveillance within tissues, memory T cells can recirculate back to the lymph nodes via the afferent lymphatics (red arrows).

Antigen-experienced B cells become unresponsive to lymph node cues and cease to recirculate, partitioning into specific niches, such as the red pulp of the spleen and the bone marrow [6]. By contrast, central memory T cells (which retain CCR7 expression) continue to recirculate like naïve cells to lymph nodes, whereas effector memory T cells traffic through tissues [7]. Some memory T cells preferentially recirculate to the tissue in which they were initially activated – a site where they are most likely to re-encounter their cognate antigen. However, such well-defined patterns of homing have thus far only been described for the skin and the gut [8] (Figure 1).

Dendritic cells (DCs) communicate tissue identity to T cells

There is now good evidence to suggest that T cells are imprinted with an endothelial address code as they undergo activation in the lymph node, and that a cognate interaction with interdigitating DCs is sufficient for this process (reviewed in Ref. [9]).

DCs isolated from various lymphoid organs all share the ability to activate CD8+ T cells but only those taken from Peyer’s patches (PPs) imprint them with the gut-homing code (α4β7, CCR9+) [10]. The positional identity of DCs appears stable enough to withstand ex vivo culture. Furthermore, Stagg et al. showed that DCs possess an instructive accessory component that is not dependent on cognate T-cell receptor (TCR)–MHC–peptide interaction and can act in trans [11]. Recently, Iwata et al. have suggested that retinoic acid might be such an accessory signal generated by intestinal DCs to induce a gut-homing phenotype in T cells [12].

An elegant study by Dudda et al. demonstrated that bone marrow-derived DCs injected intracutaneously can induce CD8+ T cells that express the cutaneous vascular entry code [CLA+ (cutaneous leukocyte antigen)]. T cells induced after intraperitoneal injection of the same bone marrow-derived DCs, however, expressed the intestinal entry code (integrin α4β7) [13]. Although the possibility that endogenous tissue resident cells have a direct role in imprinting T cells to return to the injection site cannot be formally excluded, this strongly suggests that DCs are plastic enough to acquire tissue-specific properties from their local microenvironment. So what tells a DC, or any other bone marrow-derived cell, that it is in a particular tissue microenvironment?

Address codes are sequentially encountered

Leukocytes arriving in interstitial tissue exhibit different properties to their blood-borne counterparts. In particular, their ability to respond to stromally derived cues for proliferation, differentiation, positioning and survival is likely to be radically altered [14]. Despite the inherently sequential nature of leukocyte trafficking, the effects of transendothelial migration on leukocyte behaviour within tissues remain poorly characterized.

For obvious experimental and technical reasons, in vitro studies of leukocyte-stromal interactions have neglected the effects of prior vascular and lymphatic endothelial transit on the subsequent behaviour of leukocytes. Likewise, the cellular conversations that take place between endothelia and underlying stroma remain largely uncharacterized, although in vivo data suggest that they could be crucial to defining the vascular address code, by transcytosis and vascular expression of stromally derived chemokines [15]. There is now a large body of evidence to support the concept that the proinflammatory behaviour of endothelial cells is affected by the type of matrix on which they grow and the nature of the underlying stromal cells (reviewed in Ref. [16]).

Rainger et al. have recently developed a novel flow-based co-culture system, which directly allows the effects of stromal cells on overlying endothelium to be described [17]. This model has shown that stromal cells, such as smooth muscle cells and fibroblasts, prime the overlying endothelium and lead to an alteration in the pattern of leukocyte recruitment across the endothelium [16,17].

Leukocyte migration from peripheral tissues to local lymph nodes requires movement through the lymphatic endothelium in a baso-lumenal direction. The important discovery of unique markers for lymphatic endothelium, such as the hyaluronic acid receptor LYVE-1 and vascular endothelial growth factor receptor 3 (VEGFR3), has facilitated the isolation and molecular characterization of lymphatic endothelial cells [18,19]. This now provides an opportunity to further investigate their contribution to stromal address codes experimentally, and to determine if an exit code exists for the lymphatic system analogous to the entry code on vascular endothelium (reviewed in Ref. [20]) (Figure 2).

Figure 2.

Figure 2

Leukocyte–stromal interactions are involved at every stage in lymphocyte recirculation. The molecular basis by which leukocytes leave the circulation and migrate across endothelium has been well studied (step 1, endothelium). Leukocytes also interact with the basal lamina, matrix proteins and pericytes surrounding the endothelium (black dots), before entering interstitial tissue (step 2, stroma), where they interact with tissue stromal cells, such as fibroblasts. How leucocytes exit tissue into the lymphatics (step 3, lymphatics) remains poorly understood. Each step in the process delivers different types of instructions to the leukocyte. Step 1 regulates entry; step 2 regulates proliferation, survival and differentiation and step 3 regulates exit.

Fibroblast diversity, autonomy and positional identity

Fibroblasts isolated from different tissues display different functional properties (reviewed in Ref. [21]). Consistent with the varying biophysical requirements of different tissues, phenotypic differences in the well-known structural functions of fibroblasts, such as migratory capacity, extra cellular matrix (ECM) production and degradation and contractility, have been reported. The less well-known immunomodulatory functions of fibroblasts are also known to vary according to anatomical site and disease status [22].

The molecular basis for such fibroblast diversity has only recently been addressed. Global transcriptome analysis of a panel of human fibroblasts has been performed by several independent research groups [23,24]. Comparative analysis revealed that the transcriptional profiles of fibroblasts can be clustered into well-defined groups defined by anatomical site. Furthermore, specific homeobox (HOX) gene expression is very strongly associated with the cluster groups [24]. HOX genes encode important transcription factors that confer regional identity to tissues [25]. This provides support for the concept that fibroblasts possess positional identity that is transcriptionally imprinted, and mechanistic evidence to explain long-standing observations of stable fibroblast diversity in the adult, even within a single tissue type.

Such diversity is not unique to the fibroblast element of tissue stroma. Vascular endothelial cells cultured from different anatomical sites also show a wide diversity of transcriptional and proteomic profiles [26,27]. In addition, a similar approach has revealed important biological differences between vascular and lymphatic endothelium [28]. Other stromal elements, such as epithelial cells, smooth muscle cells and pericytes, are likely to be as varied as fibroblasts.

It seems plausible that tissue-transiting cells, such as lymphocytes and DCs, receive instruction about their anatomical position, at least in part, from endothelial cells, stromal fibroblasts and their secreted products. Equally, tissue-specific epithelia and other specialist cells with positional identity, such as macrophages, might contribute to the instructional process. Fibroblasts are particularly well-suited to define positional identity because they exhibit considerable autonomy. Members of the fibroblast family are known to function as key organizers at the heart of the immune system, for example, during lymph node development and the presentation of antigen by follicular DCs in germinal centers [29].

Stromal address codes in leukocyte development

Fibroblast-like stromal cells are required to support effective haematopoiesis and help define the bone marrow stromal niche [30]. Recent work has revealed that the expression of a few crucial molecules can support leukocyte survival, regulate leukocyte position and control leukocyte differentiation in a variety of stem-cell niches, including the bone marrow, thymus and lymph node. These molecules appear to constitute part of a stromal address code that consists of a homeostatic or constitutive chemokine {CXCL12 (SDF-1), CXCL13 [B cell-attracting chemokine-1 (BCA-1)], CCL19 [EBI1 ligand chemokine (ELC)] or CCL21 [secondary lymphoid-tissue chemokine (SLC)]} together with VCAM-1 (CD106), in the presence of a cytokine or growth factor [(interleukin-6 (IL-6), IL-7 and fibroblast growth factor-7 (FGF-7) or FGF-10] (Figure 3).

Figure 3.

Figure 3

(a) Stromal area codes regulate leukocyte accumulation, differentiation and survival in the thymus, bone marrow and lymph node. Homeostatic chemokines (CXCL12, CXCL13, CCL19, CCL21), adhesion molecules [vascular cell adhesion molecule-1 (VCAM-1)] and cytokines and growth factors [interleukin-6 (IL-6), IL-7, fibroblast growth factor-7 (FGF-7), FGF-10] are components of the stromal code that help define stromal niches (bone marrow, thymus and lymph node) involved in leukocyte accumulation, differentiation and survival. Fibroblasts produce the appropriate cytokine or chemokine and express the appropriate adhesion receptor that is recognized by cognate receptors on the infiltrating leukocytes. In the case of the developing lymph node, ‘inducer’ lymphocytes produce lymphotoxin-α and IL-7, which induce the secretion of constitutive chemokines from the stromal ‘organizer’ cells, leading to the development of lymphoid aggregates with a lymph node structure. (b) Components of the stromal area code are aberrantly expressed in disease. During physiological inflammation, inflammatory chemokines (CCL2-CCL5, CX3CL1 and CXCL1-CXCL11) and inflammatory mediators, such as interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α) and IL-1, are produced by stromal cells and lead to the recruitment of inflammatory cells (lymphocytes, neutrophils and monocytes). During the resolution phase of inflammation, these proinflammatory stimuli are removed. However, in chronic persistent inflammation (pathological inflammation), stromal cells in tissues, such as the synovium, salivary gland, thyroid and liver, begin to aberrantly produce or express components of the physiological stromal code normally associated with primary and secondary lymphoid tissue, leading to the generation of pathogenic tertiary lymphoid tissue.

(i) Bone marrow

In the bone marrow, CXCL12 is expressed by a subset of fibroblasts that co-express VCAM-1 but not IL-7 [31]. The interplay of CXCL12 and VCAM-1 is essential in regulating the release of B cells into the bloodstream and their maturation. Genetic ablation or functional blockade of the unique CXCL12 receptor, CXCR4, leads to premature release of both granulocyte and B-cell precursors [32,33], whereas genetic deletion of the α4 integrin subunit prevents early B-cell lymphopoiesis, suggesting that VCAM-1 is also likely to be essential in retaining cells in the bone marrow [34].

Tokoyoda et al. have elegantly demonstrated that multipotent haematopoietic stem cells and pre-pro-B cells associate specifically with the CXCL12+ fibroblast subset and that CXCR4 engagement on the pre-pro-B cells increases their adhesiveness to VCAM-1 through α4β1 integrin [31]. Interestingly, CXCL12+ and IL-7+ stromal subsets are spatially separated but do not have a clearly compartmentalized distribution. Upon maturation from pre-pro-B cell to pro-B cell, migration away from CXCL12+ towards IL-7+ stromal cells is observed. A similar role for CXCL12+ in the retention of immature neutrophils and the subsequent recall of senescent neutrophils to the bone marrow has recently been demonstrated [33].

The CXCL12+ bone marrow fibroblast niche also provides a resting place for terminally differentiated B cells (plasma cells). Long-lived plasma cell survival and function are highly dependent on their close association with bone marrow fibroblasts [35]. An α4β1 integrin-mediated interaction, which does not appear to use VCAM-1 as the ligand, is reported as essential to the secretion of IL-6 from fibroblasts. Together with engagement of plasma cell CD44, a receptor for the extracellular matrix component hyaluronic acid (HA), IL-6 promotes the antibody-secreting function of long-lived plasma cells. CXCL12 is responsible for attracting the plasma cells back to their particular stromal niche, and like IL-6, is a potent survival factor for them [36].

(ii) Thymus

Mesenchymal fibroblasts are now known to have at least two vital roles in thymic function. First, they are required to support the proliferation of thymic epithelial cells through the release of FGF-7 and FGF-10 [37]. Second, in early T-cell development they are necessary for the appropriate presentation of IL-7 on the extracellular matrix [38].

In the postnatal thymus, CXCL12+ and VCAM-1+ cortical fibroblasts are crucial for attracting and supporting the migration of early T-cell progenitors from the thymic medulla to the cortex [39,40]. Repositioning of thymic progenitors is necessary for their maturation, which relies on sequential signals from phenotypically distinct thymic epithelia. After positive selection in the cortex, T cells in neonatal mice depend on CCR7 ligands to reverse their direction of movement, back into the medulla where negative selection can take place [41]. Thus, stromal fibroblasts and epithelia provide adhesive substrates, chemotactic cues, differentiation stimuli and proliferative signals to developing T cells. The positional identity of these cells is clearly pivotal to compartmentalizing the cortical and medullary microenvironments of the thymus.

(iii) Peripheral lymph nodes

Although the fundamental role of the fibroblast as choreographer is similar, the stromal address code necessary for the generation of secondary lymphoid tissue differs mechanistically from that required for leukocyte retention and differentiation in primary lymphoid tissues, such as bone marrow and thymus. VCAM-1+ LTbR+ fibroblastic ‘organizer’ cells secrete the homeostatic chemokine CXCL13, attracting and activating α4β1 integrin on CXCR5+LTα1β2+CD3CD4+CD45+ ‘inducer’ cells. When these cells come into contact a positive feedback loop is initiated whereby lymphotoxin-β (LT-β) receptor engagement, stabilized by VCAM-1 ligation, leads to upregulation of CXCL13 and VCAM-1 expression, and consequently an increased recruitment of inducer cells [42-44] (reviewed in Ref. [29]).

Transgenic mouse studies have revealed a hierarchy among the homeostatic chemokines in their ability to support lymphoid neogenesis. Ectopic expression of CXCL13 in pancreatic islet cells suggested it was the most efficient, followed by CCL21, CCL19 and finally CXCL12 [45]. However, the choice of tissue for ectopic expression affects the outcome, suggesting that certain microenvironments (pancreas) are more plastic than others (brain and skin) [46,47].

Pathological stromal codes

An increasing number of studies suggest a key pathological role for the ectopic temporal and spatial expression of cytokines and chemokines in diseases, such as rheumatoid arthritis (RA), autoimmune liver disease, thyroid disease and diabetes [48-50]. These chronic immune-mediated inflammatory conditions are characterized by the abnormal persistence and continued episodic recruitment of infiltrating inflammatory cells, which is accompanied by a local expansion and activation of fibroblasts in the diseased tissue [48]. The fibrotic tissue gradually acquires an extremely stable state that leads inexorably to loss of tissue function and in several diseases the tissue becomes organized into lymphoid-like structures that have been called ‘tertiary lymphoid tissue’ [51].

These observations, in conjunction with data from transgenic mice that overexpress chemokines involved in lymphoid organogenesis, have led to the proposal that chronic inflammation is lymphoid neogenesis in the wrong place at the wrong time [52,53]. Applying this concept more globally predicts that the aberrant expression of other components of the stromal address code involved in defining other stromal niches, such as the bone marrow and thymus, might also contribute to pathogenesis in chronic inflammation.

Many research groups have now reported findings that support this concept of an ectopic, aberrant stromal code. Fibroblast-derived CXCL12 and CXCL13 are overexpressed in the rheumatoid arthritis joint [53,54] and in a range of other chronic inflammatory immune mediated diseases [55-57]. Transforming growth factor-β (TGF-β) found at high levels in the inflamed synovium, induces the expression of CXCR4 on highly differentiated T lymphocytes and this might provide a mechanism for their retention at this site [54]. The prolonged survival of these highly differentiated T cells is promoted by the secretion of type-I IFNs by synovial fibroblasts and macrophages [58]. Of interest, synovial fibroblasts isolated from the rheumatoid synovium also express high levels of VCAM-1 and IL-6 [59,60].

Dysregulated expression of constitutive chemokines has also been reported in the salivary glands in Sjogren’s syndrome, in the liver in cirrhosis and in the thyroid in Hashimoto’s thyroiditis [49,61,62].In a considerable proportion of cases, organized lymphoid aggregates with lymphocytes expressing the appropriate cognate chemokine receptor are also found. In other words, the diseased stromal tissue expresses an inappropriate lymphoid stromal code. Why this occurs remains unclear but recent advances in our understanding of the origins of fibroblasts have begun to provide some tantalizing clues.

Origins of fibroblasts: mesenchymal proliferation, local transdifferentiation from epithelium or seeding from bone marrow?

Despite increasing evidence for their crucial immunomodulatory functions, fibroblasts are still widely thought of as primary mesenchymal cells that are deposited in tissue interstitia, simply as a consequence of organ development. It is not disputed that their developmental origin is the primary mesenchyme, and that fibroblasts can proliferate to generate new fibroblasts, however, emerging evidence suggests that immature fibroblasts are not phenotypically equivalent to primary mesenchyme. This might imply that they are not functionally equivalent either.

After unsuccessful attempts to identify fibroblast-specific markers by a conventional antibody approach, an elegant subtractive hybridization strategy using transcripts from fibroblasts and epithelial cells from the same microenvironment, identified fibroblast-specific protein (FSP1), also called S100A4 (Mts1) [63]. Iwano et al. demonstrated that FSP1+ fibroblasts can be derived from local secondary epithelia in response to physical injury or inflammation in adult mice; a phenomenon well known to developmental and cancer biologists, called epithelial to mesenchymal transition [64]. This has led this group and others to propose that, in several tissues (lung, gut, kidneys), new fibroblasts can arise from epithelial structures.

A third pathway by which fibroblasts can be generated is from precursor cells called fibrocytes. These represent a small population (~0.5%) of circulating blood cells that co-express markers of the haematopoietic (CD34+CD45+) and mesenchymal (vimentin+ collagen I+) lineages and appear to be derived primarily from CD14+ peripheral blood monocytes [65]. Fibrocytes rapidly gain entry to sites of tissue damage, where they exhibit the contractile function of myofibroblasts and secrete extracellular matrix proteins to promote wound healing [14,65-67]. In vivo the homeostatic chemokines CCL21 and CXCL12 regulate fibrocyte traffic into tissues in the context of wound healing and pulmonary fibrosis [65,68]. Interestingly, there is substantial evidence to suggest that fibroblast proliferation and matrix deposition occur independently in idiopathic pulmonary fibrosis. This is also the case in rheumatoid arthritis, suggesting that tissue damage and inflammation can be uncoupled (reviewed in Refs [48,69]).

Therefore, it is now clear that fibroblasts can be derived in at least three different ways: locally (by proliferation or transdifferentiation) and distally (from the bone marrow). In some organs and models of inflammation (e.g. experimentally induced renal fibrosis), all of these processes appear to contribute to the pathological accumulation of fibroblasts [64]. These findings need to be reconciled with the work of Chang et al., who found stable transcriptionally imprinted positional identity in fibroblasts from normal human tissue [25]. Do fibroblasts from different sources have intrinsic positional imprints or are they adaptable to new microenvironments, as DCs appear to be? As the lineage relationships and differentiation potentials of human fibroblasts become more clearly defined, it will be important to determine whether distally or locally derived fibroblasts predominate in different fibrotic diseases, and whether this depends on the presence or absence of epithelium in target organs.

Concluding remarks

The prevailing paradigm accounting for the accumulation of specific leukocyte subsets in an inflamed tissue is based on endothelial selectivity at the point of recruitment. However, this ignores the role of selective retention within the tissue, mediated by a stromal address code that is defined, at least in part, by fibroblasts. A combinatorial stromal address code involving homeostatic or constitutive chemokines (CXCL12, CXCL13, CCL19, CCL21), adhesion molecules (VCAM-1) and cytokines (IL-6 and IL-7) is found at key sites of the immune system, in the bone marrow, thymus and lymph node, where it regulates leukocyte maturation and lymphoid organogenesis. The inappropriate expression of some or all of the components of this physiological stromal address code is likely to be involved in the switch from resolving to persistent inflammatory disease. Fibroblasts are able to impose a stromal address code on tissue-infiltrating leukocytes, which governs their accumulation, survival and differentiation. Exactly how the stromal address code becomes stably altered is unclear, although it is possible that transdifferentiating epithelial cells and infiltrating fibrocytes mistakenly acquire the stromal address code of lymphoid tissues, rather than the appropriate code for the microenvironment they have entered. Episodes of injury and inflammation are traumatic times for fibroblast regional identity – particularly if the appropriate stromal area code is not re-established correctly.

References

  • 1.Campbell DJ, et al. Chemokines in the systemic organization of immunity. Immunol. Rev. 2003;195:58–71. doi: 10.1034/j.1600-065x.2003.00067.x. [DOI] [PubMed] [Google Scholar]
  • 2.Warnock RA, et al. Molecular mechanisms of lymphocyte homing to peripheral lymph nodes. J. Exp. Med. 1998;187:205–216. doi: 10.1084/jem.187.2.205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Nakano H, et al. A novel mutant gene involved in T-lymphocyte-specific homing into peripheral lymphoid organs on mouse chromosome 4. Blood. 1998;91:2886–2895. [PubMed] [Google Scholar]
  • 4.Nakano H, et al. Genetic defect in T lymphocyte-specific homing into peripheral lymph nodes. Eur. J. Immunol. 1997;27:215–221. doi: 10.1002/eji.1830270132. [DOI] [PubMed] [Google Scholar]
  • 5.Gunn MD, et al. Mice lacking expression of secondary lymphoid organ chemokine have defects in lymphocyte homing and dendritic cell localization. J. Exp. Med. 1999;189:451–460. doi: 10.1084/jem.189.3.451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hargreaves DC, et al. A coordinated change in chemokine responsiveness guides plasma cell movements. J. Exp. Med. 2001;194:45–56. doi: 10.1084/jem.194.1.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.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]
  • 8.Campbell DJ, Butcher EC. Rapid acquisition of tissue-specific homing phenotypes by CD4+ T cells activated in cutaneous or mucosal lymphoid tissues. J. Exp. Med. 2002;195:135–141. doi: 10.1084/jem.20011502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Dudda JC, Martin SF. Tissue targeting of T cells by DCs and microenvironments. Trends Immunol. 2004;25:417–421. doi: 10.1016/j.it.2004.05.008. [DOI] [PubMed] [Google Scholar]
  • 10.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]
  • 11.Stagg AJ, et al. Intestinal dendritic cells increase T cell expression of alpha4beta7 integrin. Eur. J. Immunol. 2002;32:1445–1454. doi: 10.1002/1521-4141(200205)32:5<1445::AID-IMMU1445>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
  • 12.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]
  • 13.Dudda JC, et al. Dendritic cell immunization route determines CD8+ T cell trafficking to inflamed skin: role for tissue microenvironment and dendritic cells in establishment of T cell-homing subsets. J. Immunol. 2004;172:857–863. doi: 10.4049/jimmunol.172.2.857. [DOI] [PubMed] [Google Scholar]
  • 14.Buckley CD, et al. Fibroblasts regulate the switch from acute resolving to chronic persistent inflammation. Trends Immunol. 2001;22:199–204. doi: 10.1016/s1471-4906(01)01863-4. [DOI] [PubMed] [Google Scholar]
  • 15.Middleton J, et al. Transcytosis and surface presentation of IL-8 by venular endothelial cells. Cell. 1997;91:385–395. doi: 10.1016/s0092-8674(00)80422-5. [DOI] [PubMed] [Google Scholar]
  • 16.Nash GB, et al. The local physicochemical environment conditions the proinflammatory response of endothelial cells and thus modulates leukocyte recruitment. FEBS Lett. 2004;569:13–17. doi: 10.1016/j.febslet.2004.05.040. [DOI] [PubMed] [Google Scholar]
  • 17.Rainger GE, et al. A novel system for investigating the ability of smooth muscle cells and fibroblasts to regulate adhesion of flowing leukocytes to endothelial cells. J. Immunol. Methods. 2001;255:73–82. doi: 10.1016/s0022-1759(01)00427-6. [DOI] [PubMed] [Google Scholar]
  • 18.Banerji S, et al. LYVE-1, a new homologue of the CD44 glycoprotein, is a lymph-specific receptor for hyaluronan. J. Cell Biol. 1999;144:789–801. doi: 10.1083/jcb.144.4.789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jussila L, et al. Lymphatic endothelium and Kaposi’s sarcoma spindle cells detected by antibodies against the vascular endothelial growth factor receptor-3. Cancer Res. 1998;58:1599–1604. [PubMed] [Google Scholar]
  • 20.Jackson DG. The lymphatics revisited: new perspectives from the hyaluronan receptor LYVE-1. Trends Cardiovasc. Med. 2003;13:1–7. doi: 10.1016/s1050-1738(02)00189-5. [DOI] [PubMed] [Google Scholar]
  • 21.Fries KM, et al. Evidence of fibroblast heterogeneity and the role of fibroblast subpopulations in fibrosis. Clin. Immunol. Immunopathol. 1994;72:283–292. doi: 10.1006/clin.1994.1144. [DOI] [PubMed] [Google Scholar]
  • 22.Brouty-Boye D, et al. Chemokines and CD40 expression in human fibroblasts. Eur. J. Immunol. 2000;30:914–919. doi: 10.1002/1521-4141(200003)30:3<914::AID-IMMU914>3.0.CO;2-D. [DOI] [PubMed] [Google Scholar]
  • 23.Parsonage G, et al. Global gene expression profiles in fibroblasts from synovial, skin and lymphoid tissue reveals distinct cytokine and chemokine expression patterns. Thromb. Haemost. 2003;90:688–697. doi: 10.1160/TH03-04-0208. [DOI] [PubMed] [Google Scholar]
  • 24.Chang HY, et al. Diversity, topographic differentiation, and positional memory in human fibroblasts. Proc. Natl. Acad. Sci. U. S. A. 2002;99:12877–12882. doi: 10.1073/pnas.162488599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gilbert SF. The genetics of axis specification in Drosophila. In: Gilbert SF, editor. Developmental Biology. 4th edn Sinauer Associates Inc.; 1994. pp. 531–574. [Google Scholar]
  • 26.Chi JT, et al. Endothelial cell diversity revealed by global expression profiling. Proc. Natl. Acad. Sci. U. S. A. 2003;100:10623–10628. doi: 10.1073/pnas.1434429100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Oh P, et al. Subtractive proteomic mapping of the endothelial surface in lung and solid tumours for tissue-specific therapy. Nature. 2004;429:629–635. doi: 10.1038/nature02580. [DOI] [PubMed] [Google Scholar]
  • 28.Podgrabinska S, et al. Molecular characterization of lymphatic endothelial cells. Proc. Natl. Acad. Sci. U. S. A. 2002;99:16069–16074. doi: 10.1073/pnas.242401399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Mebius RE. Organogenesis of lymphoid tissues. Nat. Rev. Immunol. 2003;3:292–303. doi: 10.1038/nri1054. [DOI] [PubMed] [Google Scholar]
  • 30.Dexter TM. Stromal cell associated haemopoiesis. J. Cell. Physiol. Suppl. 1982;1:87–94. doi: 10.1002/jcp.1041130414. [DOI] [PubMed] [Google Scholar]
  • 31.Tokoyoda K, et al. Cellular niches controlling B lymphocyte behavior within bone marrow during development. Immunity. 2004;20:707–718. doi: 10.1016/j.immuni.2004.05.001. [DOI] [PubMed] [Google Scholar]
  • 32.Ma Q, et al. The chemokine receptor CXCR4 is required for the retention of B lineage and granulocytic precursors within the bone marrow microenvironment. Immunity. 1999;10:463–471. doi: 10.1016/s1074-7613(00)80046-1. [DOI] [PubMed] [Google Scholar]
  • 33.Martin C, et al. Chemokines acting via CXCR2 and CXCR4 control the release of neutrophils from the bone marrow and their return following senescence. Immunity. 2003;19:583–593. doi: 10.1016/s1074-7613(03)00263-2. [DOI] [PubMed] [Google Scholar]
  • 34.Arroyo AG, et al. Differential requirements for alpha4 integrins during fetal and adult hematopoiesis. Cell. 1996;85:997–1008. doi: 10.1016/s0092-8674(00)81301-x. [DOI] [PubMed] [Google Scholar]
  • 35.Minges Wols HA, et al. The role of bone marrow-derived stromal cells in the maintenance of plasma cell longevity. J. Immunol. 2002;169:4213–4221. doi: 10.4049/jimmunol.169.8.4213. [DOI] [PubMed] [Google Scholar]
  • 36.Cassese G, et al. Plasma cell survival is mediated by synergistic effects of cytokines and adhesion-dependent signals. J. Immunol. 2003;171:1684–1690. doi: 10.4049/jimmunol.171.4.1684. [DOI] [PubMed] [Google Scholar]
  • 37.Jenkinson WE, et al. Differential requirement for mesenchyme in the proliferation and maturation of thymic epithelial progenitors. J. Exp. Med. 2003;198:325–332. doi: 10.1084/jem.20022135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Banwell CM, et al. Studies on the role of IL-7 presentation by mesenchymal fibroblasts during early thymocyte development. Eur. J. Immunol. 2000;30:2125–2129. doi: 10.1002/1521-4141(2000)30:8<2125::AID-IMMU2125>3.0.CO;2-H. [DOI] [PubMed] [Google Scholar]
  • 39.Plotkin J, et al. Critical role for CXCR4 signaling in progenitor localization and T cell differentiation in the postnatal thymus. J. Immunol. 2003;171:4521–4527. doi: 10.4049/jimmunol.171.9.4521. [DOI] [PubMed] [Google Scholar]
  • 40.Prockop SE, et al. Stromal cells provide the matrix for migration of early lymphoid progenitors through the thymic cortex. J. Immunol. 2002;169:4354–4361. doi: 10.4049/jimmunol.169.8.4354. [DOI] [PubMed] [Google Scholar]
  • 41.Ueno T, et al. CCR7 signals are essential for cortex-medulla migration of developing thymocytes. J. Exp. Med. 2004;200:493–505. doi: 10.1084/jem.20040643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.de Togni P, et al. Abnormal development of peripheral lymphoid organs in mice deficient in lymphotoxin. Science. 1994;264:703–707. [PubMed] [Google Scholar]
  • 43.Rennert PD, et al. Lymph node genesis is induced by signaling through the lymphotoxin β receptor. Immunity. 1998;9:71–79. doi: 10.1016/s1074-7613(00)80589-0. [DOI] [PubMed] [Google Scholar]
  • 44.Ansel KM, et al. A chemokine-driven positive feedback loop organizes lymphoid follicles. Nature. 2000;406:309–314. doi: 10.1038/35018581. [DOI] [PubMed] [Google Scholar]
  • 45.Luther SA, et al. Differing activities of homeostatic chemokines CCL19, CCL21, and CXCL12 in lymphocyte and dendritic cell recruitment and lymphoid neogenesis. J. Immunol. 2002;169:424–433. doi: 10.4049/jimmunol.169.1.424. [DOI] [PubMed] [Google Scholar]
  • 46.Chen SC, et al. Ectopic expression of the murine chemokines CCL21a and CCL21b induces the formation of lymph node-like structures in pancreas, but not skin, of transgenic mice. J. Immunol. 2002;168:1001–1008. doi: 10.4049/jimmunol.168.3.1001. [DOI] [PubMed] [Google Scholar]
  • 47.Chen SC, et al. Central nervous system inflammation and neurological disease in transgenic mice expressing the CC chemokine CCL21 in oligodendrocytes. J. Immunol. 2002;168:1009–1017. doi: 10.4049/jimmunol.168.3.1009. [DOI] [PubMed] [Google Scholar]
  • 48.Buckley CD. Michael Mason prize essay 2003. Why do leucocytes accumulate within chronically inflamed joints? Rheumatology (Oxford) 2003;42:1433–1444. doi: 10.1093/rheumatology/keg413. [DOI] [PubMed] [Google Scholar]
  • 49.Armengol MP, et al. Chemokines determine local lymphoneogenesis and a reduction of circulating CXCR4+ T and CCR7 B and T lymphocytes in thyroid autoimmune diseases. J. Immunol. 2003;170:6320–6328. doi: 10.4049/jimmunol.170.12.6320. [DOI] [PubMed] [Google Scholar]
  • 50.Green EA, Flavell RA. The temporal importance of TNFα expression in the development of diabetes. Immunity. 2000;12:459–469. doi: 10.1016/s1074-7613(00)80198-3. [DOI] [PubMed] [Google Scholar]
  • 51.Ruddle NH. Lymphoid neo-organogenesis: lymphotoxin’s role in inflammation and development. Immunol. Res. 1999;19:119–125. doi: 10.1007/BF02786481. [DOI] [PubMed] [Google Scholar]
  • 52.Hjelmstrom P, et al. Lymphoid tissue homing chemokines are expressed in chronic inflammation. Am. J. Pathol. 2000;156:1133–1138. doi: 10.1016/S0002-9440(10)64981-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hjelmstrom P. Lymphoid neogenesis: de novo formation of lymphoid tissue in chronic inflammation through expression of homing chemokines. J. Leukoc. Biol. 2001;69:331–339. [PubMed] [Google Scholar]
  • 54.Buckley CD, et al. Persistent induction of the chemokine receptor CXCR4 by TGF-β1 on synovial T cells contributes to their accumulation within the rheumatoid synovium. J. Immunol. 2000;165:3423–3429. doi: 10.4049/jimmunol.165.6.3423. [DOI] [PubMed] [Google Scholar]
  • 55.Nanki T, et al. Stromal cell-derived factor-1-CXC chemokine receptor 4 interactions play a central role in CD4+ T cell accumulation in rheumatoid arthritis synovium. J. Immunol. 2000;165:6590–6598. doi: 10.4049/jimmunol.165.11.6590. [DOI] [PubMed] [Google Scholar]
  • 56.Bradfield PF, et al. Rheumatoid fibroblast-like synoviocytes overexpress the chemokine stromal cell-derived factor 1 (CXCL12), which supports distinct patterns and rates of CD4+ and CD8+ T cell migration within synovial tissue. Arthritis Rheum. 2003;48:2472–2482. doi: 10.1002/art.11219. [DOI] [PubMed] [Google Scholar]
  • 57.Lande R, et al. Characterization and recruitment of plasma-cytoid dendritic cells in synovial fluid and tissue of patients with chronic inflammatory arthritis. J. Immunol. 2004;173:2815–2824. doi: 10.4049/jimmunol.173.4.2815. [DOI] [PubMed] [Google Scholar]
  • 58.Pilling D, et al. Interferon-β mediates stromal cell rescue of T cells from apoptosis. Eur. J. Immunol. 1999;29:1041–1050. doi: 10.1002/(SICI)1521-4141(199903)29:03<1041::AID-IMMU1041>3.0.CO;2-#. [DOI] [PubMed] [Google Scholar]
  • 59.Morales-Ducret J, et al. α4/β1 integrin (VLA-4) ligands in arthritis. Vascular cell adhesion molecule-1 expression in synovium and on fibroblast-like synoviocytes. J. Immunol. 1992;149:1424–1431. [PubMed] [Google Scholar]
  • 60.Firestein GS, et al. Quantitative analysis of cytokine gene expression in rheumatoid arthritis. J. Immunol. 1990;144:3347–3353. [PubMed] [Google Scholar]
  • 61.Amft N, et al. Ectopic expression of the B cell-attracting chemokine BCA-1 (CXCL13) on endothelial cells and within lymphoid follicles contributes to the establishment of germinal center-like structures in Sjogren’s syndrome. Arthritis Rheum. 2001;44:2633–2641. doi: 10.1002/1529-0131(200111)44:11<2633::aid-art443>3.0.co;2-9. [DOI] [PubMed] [Google Scholar]
  • 62.Grant AJ, et al. Hepatic expression of secondary lymphoid chemokine (CCL21) promotes the development of portal-associated lymphoid tissue in chronic inflammatory liver disease. Am. J. Pathol. 2002;160:1445–1455. doi: 10.1016/S0002-9440(10)62570-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Strutz F, et al. Identification and characterization of a fibroblast marker: FSP1. J. Cell Biol. 1995;130:393–405. doi: 10.1083/jcb.130.2.393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Iwano M, et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J. Clin. Invest. 2002;110:341–350. doi: 10.1172/JCI15518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Abe R, et al. Peripheral blood fibrocytes: differentiation pathway and migration to wound sites. J. Immunol. 2001;166:7556–7562. doi: 10.4049/jimmunol.166.12.7556. [DOI] [PubMed] [Google Scholar]
  • 66.Bucala R, et al. Circulating fibrocytes define a new leukocyte subpopulation that mediates tissue repair. Mol. Med. 1994;1:71–81. [PMC free article] [PubMed] [Google Scholar]
  • 67.Pilling D, et al. Inhibition of fibrocyte differentiation by serum amyloid P. J. Immunol. 2003;171:5537–5546. doi: 10.4049/jimmunol.171.10.5537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Phillips RJ, et al. Circulating fibrocytes traffic to the lungs in response to CXCL12 and mediate fibrosis. J. Clin. Invest. 2004;114:438–446. doi: 10.1172/JCI20997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Selman M, et al. Idiopathic pulmonary fibrosis: prevailing and evolving hypotheses about its pathogenesis and implications for therapy. Ann. Intern. Med. 2001;134:136–151. doi: 10.7326/0003-4819-134-2-200101160-00015. [DOI] [PubMed] [Google Scholar]

RESOURCES