GET THE APP

Regulation of T-cell Tolerance by Lymphatic Endothelial Cells
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2014) Volume 0, Issue 0

Regulation of T-cell Tolerance by Lymphatic Endothelial Cells

Sherin J. Rouhani, Jacob D. Eccles, Eric F. Tewalt and Victor H. Engelhard*
Carter Immunology Center, Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
*Corresponding Author: Victor H. Engelhard, Carter Immunology Center, University of Virginia School of Medicine, Box 801386, Charlottesville VA 22908, USA, Tel: 434-924-2423, Fax: 434-924-1221 Email:

Abstract

Lymphatic endothelial cells are most often thought of as structural cells that form the lymphatic vasculature, which transports fluid out of peripheral tissues and transports antigens and antigen presenting cells to lymph nodes. Recently, it has been shown that lymphatic endothelial cells also dynamically respond to and influence the immune response in several ways. Here, we describe how lymphatic endothelial cells induce peripheral T-cell tolerance and how this relates to tolerance induced by other types of antigen presenting cells. Furthermore, the ability of lymphatic endothelial cells to alter immune responses under steady-state or inflammatory conditions is explored, and the therapeutic potential of bypassing lymphatic endothelial cell -induced tolerance to enhance cancer immunotherapy is discussed.

Keywords: Lymphatic endothelial cells; Peripheral tissue antigens; T-cell tolerance; Inflammation; Lymph node stromal cells; PD-L1

Antigen Presenting Cells and T-cell Tolerance Induction in the Thymus and Lymph Nodes

T-cell receptors (TCR) are stochastically generated in the thymus, which enables the immune system to recognize a tremendous diversity of foreign antigens. However, it also creates T-cells with the potential to recognize and attack host tissues expressing normal self-antigens. To prevent autoimmunity, T-cells that recognize self-antigens are tolerized through intrinsic mechanisms such as deletion, anergy, and induction of a regulatory T-cell (Treg) phenotype, or through extrinsic suppression by already differentiated Treg [1]. Tolerance induction occurs in the thymus at the time of T-cell development, but also occurs in lymph nodes (LN) draining peripheral tissues. Deletion and Treg induction are thought to be the dominant mechanisms of tolerance in the thymus, while anergy, deletion and Treg induction all occur in LN [2]. In all cases, tolerance induction is driven by antigen presenting cells (APC) that present ubiquitously expressed and tissue-restricted antigens. While it was initially thought that dendritic cells (DC) were the primary tolerance-inducing APC, it has become clear recently that several other cell types can also serve this role. Among them are the lymphatic endothelial cells (LEC) that reside in LN.

Direct Expression of Peripheral Tissue Antigens by LEC and Other APC

All APC have the ability to present antigens derived from ubiquitously expressed proteins, but vary in their ability to present peripheral tissue antigens (PTA), which are defined as antigens from proteins normally expressed in fewer than 5 tissues [3]. A particular challenge has been to understand how tolerance to PTA would develop, since it is expected that these antigens will be largely absent from the thymus. DC are particularly efficient at acquiring and presenting antigens, including PTA, that are originally derived from other cells, both in the thymus [4] and LN [5-7]. This led to early models suggesting that tolerance to PTA occurred primarily in LN, and was based on DC acquisition of PTA derived from the tissues drained by individual LN [8]. However, it was subsequently shown that medullary thymic epithelial cells (mTEC) transcriptionally express PTA in the thymus [9,10]. More recently, several groups including our own, have shown that multiple subpopulations in LN also transcriptionally express PTA. PTA are expressed by extrathymic Aire expressing cells (eTAC) and several LN stromal cell (LNSC) subsets including LEC, blood endothelial cells (BEC), and fibroblastic reticular cells (FRC) [11-13]. Interestingly, each of these subpopulations presents distinct PTA, although the overall size and overlap of their PTA repertoires has not been determined. Nonetheless, this mechanism broadens the presentation of PTA to non-draining LN, and enables efficient system-wide peripheral tolerance induction.

The transcriptional regulation of PTA expression is best understood in mTEC in the thymus, where it is controlled by the autoimmune regulatory element (Aire) [10]. Using single cell PCR assays, it was demonstrated that Aire stochastically induces expression of a PTA in 1-3% of total mTEC [14,15]. Limiting the number of PTA expressed in each cell is likely advantageous to ensure that each antigen is adequately represented on the limited number of MHC molecules on the cell surface, and avoid possible deleterious consequences for mTEC functionality from expressing a large number of functionally specialized proteins. Some PTA transcripts have different start sites in mTEC compared to peripheral tissues, and Aire-regulated genes within a cell are clustered based on chromosomal position rather than by cell of origin [3,14,16]. These results suggest than Aire operates by opening up a region of the chromosome to additional transcriptional regulators, rather than inducing mTEC to differentiate towards and express PTA from an alternative cell lineage. Interestingly, while Aire also regulates PTA expression in eTAC, the PTA repertoires of mTEC and eTAC are distinct [11], suggesting that these additional transcriptional regulators may differ between the two cell types.

The transcriptional regulation of PTA in LNSC is not well understood. Aire is not expressed in LEC, FRC, or BEC [12,13], and expression of PTA in these subsets does not change in Aire-/- mice [12]. Aire is a member of the SAND family of transcription factors [17], and Yip et al. [18] demonstrated that Deaf1, another SAND family member, controls the expression of 600 genes in LNSC of pancreatic LN. Most of these genes were distinct from genes regulated by Aire in eTAC. Deaf-1 regulated genes included pancreatic polypeptide (Ppy) and insulin 2 (Ins2), which are PTA normally expressed in pancreatic islets. Ppy and Ins2 are primarily expressed in LEC and FRC, respectively, and at much lower levels in CD31negpodoplaninneg cells [12,19]. However, Deaf1 is expressed in all LNSC subsets [13] suggesting that additional transcriptional regulators that differ among these subpopulations control the specificity of PTA expression. Deaf1 also controls PTA translation by regulating the transcription of the eukaryotic translation initiation factor Eif4g3 [19]. Further work is needed to determine the extent to which Deaf1 controls expression of PTA in non-pancreatic LN, and whether other members of the SAND family also play a role in controlling PTA expression in LEC and other LNSC. The overall PTA repertoire of LEC and other LNSC remains to be determined, as does the pattern of PTA expression in individual LECs. While PTA expression in mTEC provides a logical model for how PTA expression in LEC may operate, the master transcriptional regulator is different and future studies will illuminate what other similarities and differences exist.

Cross-presentation of Soluble and Tumor Derived Antigens Acquired by LEC

In addition to transcriptionally expressed PTA, LEC can also acquire and cross-present exogenously derived antigens. Lund et al. [20] demonstrated that LEC in tumor-draining LN can acquire antigens derived from VEGF-C overexpressing tumors, and present them via their MHC I molecules. Further work from the same group demonstrated that LEC engulf intradermally injected OVA, and present OVA antigen to T-cells in vitro [21]. Previous work has shown that soluble antigens travel to the LN through the lymphatics. While large antigens are taken up by macrophages in the subcapsular and medullary sinuses, antigens smaller than 70 kDa travel through FRC-lined conduits into the paracortex and B cell follicles where they are engulfed by DC and B cells [22-24]. LEC were found to engulf intradermally injected OVA as efficiently as DC [21], although LEC comprise only 0.5% of the total OVA+ cells in LN. The presentation of both tumor-derived and soluble antigens by LEC led to dysfunctional T-cell activation and increased apoptosis in vitro [20,21]. However, the in vivo consequences were not examined. In addition, while the induction of dysfunctional T-cells in the context of tumor outgrowth is intriguing and may illuminate another aspect of the immunosuppressive microenvironment in and around tumors, the results with OVA suggest that LEC might also limit responses to foreign antigens. Further work is needed to fully understand the impact of cross-presentation of acquired antigen by LEC on both immunity and tolerance.

Mechanisms of T-cell Tolerance Induction by LEC

The demonstration that LEC express PTA and function as tolerance-inducing APC in LN was the culmination of our work over several years to understand tolerance to PTA expressed in both melanocytes and melanomas, termed melanocyte differentiation antigens, which are targets in both autoimmune vitiligo and melanoma immunotherapy. Using a model system based on recognition of one such melanocyte differentiation antigen, tyrosinase, we showed that tyrosinase-specific CD8 T-cells do not undergo central tolerance in the thymus [25]. In addition, under steady state conditions, DC in LN do not present tyrosinase. Instead, tolerance to tyrosinase is strictly due to direct expression of tyrosinase mRNA and display of tyrosinase antigen by MHC I molecules on LEC [12]. This antigen presentation leads to activation and initial proliferation of tyrosinase-specific CD8 T-cells, but these cells undergo apoptosis and deletion rather than accumulating [12,25,26].

While this process of abortive proliferation has been shown in many models of CD8 deletional tolerance, the mechanisms involved in driving this outcome have been somewhat unclear. Some previous work had established that peripheral tolerance could be induced by antigen engagement in the absence of costimulation [27-31], while other studies pointed to the engagement of inhibitory molecules [32-36]. While investigating the mechanisms involved in LEC-induced deletion, we found that both of these processes were involved and interdependent [26]. LEC do not express any of the costimulatory molecules that normally drive immunogenic accumulation of activated T-cells, such as CD80, CD86, OX40L, 4-1BBL, or CD70. However, they express multiple ligands that can activate inhibitory pathways, and express a particularly high level of PD-L1. Indeed, deletion of tyrosinase-specific CD8 T-cells is strictly dependent on engagement of the PD-1/PD-L1 pathway [26]. However, it is antigen activation in the absence of costimulation that leads to rapid, high-level upregulation of PD-1 on tyrosinase-specific CD8 T-cells, which is required for deletion to occur. This can be overcome by administration of exogenous anti-4-1BB costimulation. Importantly, tyrosinase-specific CD8 T-cells that are rescued from LEC-mediated deletion gain effector function and induce autoimmune vitiligo. Given that LEC express multiple PTA, these results suggest that impairment of LEC-induced tolerance could have a role in the induction of numerous autoimmune disorders. While there is considerable interest in PD-1/PD-L1 as a mechanism to suppress pathology in peripheral tissue and in the genesis of clonal exhaustion in tumors [37-39], these results establish a central role of this pathway in peripheral tolerance induction.

LEC that have acquired soluble OVA cross-present it to CD8 T cells in a TAP1-dependent manner, and in vitro, this leads to decreased OT-I IFN? and IL-2 production and increased expression of Annexin V, PD-1, CD80, CTLA-4 [21]. Cross-presentation of tumor antigens by LEC from the tumor draining LN also leads to increased Annexin V staining on co-cultured tumor-specific CD8 T-cells ex vivo [20]. This demonstrates that cross-presentation of soluble antigens by LEC in vitro leads to dysfunctional T-cell activation, similar to the phenotype seen when LEC directly present the PTA tyrosinase in vivo [26]. This suggests that under steady-state conditions, the constellation of inhibitory molecules expressed by LEC combined with their lack of costimulatory molecules predisposes LEC to induce dysfunctional T-cell activation, regardless of the source of the antigen. This raises important questions about whether the tolerance-inducing state of LEC can be modulated, such as during infection when the LEC may acquire foreign antigens draining through the lymphatics.

T-cell tolerance can take many forms, including deletion, anergy, or Treg induction. The form of tolerance induced has been shown to depend on several factors, including TCR avidity, availability of costimulatory or inhibitory pathways, and the cytokine environment [40-45]. mTEC, DC, and eTAC have all been shown to induce multiple forms of tolerance [2,11,46,47], while to date LEC have only been shown to induce deletion in vivo [26]. A question of some interest is whether LEC can also induce other forms of tolerance, based on the molecules they express or the particular microenvironmental niche in the LN that they occupy.

LEC Interactions with DC Leading to CD4 T-cell Tolerance Induction

LEC express intermediate levels of MHC II molecules, suggesting they might also play a role in CD4 T-cell tolerance. MHC II expression is under the control of the class II transactivator (CIITA). In mice, CIITA is regulated by three different promoters (pI, pIII, and pIV) which are differentially expressed in different cell types [48]. It was recently demonstrated that LEC only express pIV mRNA [49], suggesting that endogenous expression of MHC II on LEC is controlled by CIITA driven off of this promoter. Surprisingly, LEC from pIV-/- mice still express approximately half the level of MHC II as LEC from wild-type mice, suggesting that some of the MHC II on the cell surface is not endogenously synthesized by LEC. Instead, these investigators found that LEC acquire MHC II from DC in vitro and in vivo. This is highly selective, as MHC II is not acquired from macrophages or B cells, and LEC do not acquire costimulatory molecules from DC. The mechanism of transfer was found to involve cell contact and/or exosomes. Interestingly, CD4 T-cells specific for antigens displayed by the MHC II molecules on these LEC did not proliferate in vitro, but showed an increase in Annexin V staining, suggesting an unusual form of deletional tolerance. Furthermore, when these T-cells were subsequently restimulated with aCD3/aCD28 for an additional 3 days, they showed a reduced proliferative response. However, it is not clear whether the decrease in proliferation is due to the increased level of T-cell death observed in the first 3 days of co-culture, or if LEC additionally suppress T-cell proliferation through an antigen-dependent mechanism. The induction of tolerance induced by peptide:MHC II complexes acquired by LEC remains to be established in vivo, as does the importance of this modality relative to presentation of these same complexes by DC in the same LN.

While some of the MHC II expressed on LEC is acquired from DC [49], LEC also express endogenous MHC II [49, unpublished data]. To test whether LEC directly present PTA on their own MHC II molecules and induce CD4 T-cell tolerance, we developed model systems in which the model antigens ß-galactosidase (ß-gal) and influenza hemagglutinin (HA) were conditionally expressed under the control of the Prox-1 [50] and Lyve-1 [51] promoters (unpublished). An advantage of these models is that both CD8 and CD4 T-cell receptor transgenic mice are available, enabling us to comparatively evaluate the ability of LEC to induce tolerance to epitopes from the same protein presented by either MHC I or MHC II molecules. We found that although LEC directly present ß-gal and HA epitopes on MHC I to CD8 T-cells, they do not present either antigen on MHC II to CD4 T-cells in vivo or in vitro. Our data suggest that the lack of direct presentation of MHC II epitopes by LEC is due to a defect in the MHC II processing and presentation pathway. However, we also found that these antigens were transferred to DC in vivo, and subsequent presentation by DC resulted in the induction of CD4 T-cell anergy. Interestingly, this division of labor between LEC and DC is analogous to what occurs in the thymus, where some PTA expressed in mTEC are not directly presented on MHC II, but are instead transferred to DC for the induction of tolerance [52,53]. However, other PTA are directly presented by mTEC on MHC II [52-55]. It is not understood what determines whether or not mTEC will directly present a particular antigen on MHC II. However, mTEC constitutively undergo high levels of autophagy [56,57], suggesting that the efficiency with which antigen can access the autophagy pathway may be a contributing factor [58]. The level of autophagy occurring in LEC at the steady-state has not been examined. Given the ability of LEC to acquire and present tumor-derived and soluble antigens on MHC I [20,21], an important direction for future work is to determine whether these antigens can also be directly loaded onto MHC II in LEC. Regardless, the work performed to date indicates multiple mechanisms by which LECs and DC can cooperate to induce CD4 T-cell tolerance in LN (Figure 1). While DC share pre-formed peptide:MHC II complexes with LEC, the large variety of PTA transcribed by LEC serves as a reservoir and repertoire of antigens that may be acquired by DC for tolerance induction.

clinical-cellular-immunology-LEC-transcriptionally

Figure 1: Roles of LEC in CD4 and CD8 T-cell tolerance. LEC transcriptionally express a variety of PTA, which are directly presented to CD8 T-cells, leading to CD8 T-cell proliferation and death through the combined effects of PD-1/PD-L1 signaling and a lack of costimulation. LEC do not directly present PTA on MHC II, but instead transfer the PTA to DC, which induce CD4 T-cell anergy. LEC can acquire peptide/MHC II complexes generated by DC, which are then presented by the LEC, leading to CD4 T-cell apoptosis. Finally, LEC can cross-present soluble antigens from the lymph, leading to dysfunctional CD8 activation and increased apoptosis. It is unknown whether soluble antigens can be presented by LEC on MHC II to CD4 T-cells.

LEC Localized in Peripheral Tissue and LN Subregions Differ in Their Ability to Induce Tolerance

LEC form lymphatic vessels in peripheral tissues in addition to lymphatic sinuses in the LN, raising the question of whether tissue lymphatics share the tolerogenic properties of LEC in the LN (LN-LEC). We found that LEC that form vessels in the diaphragm (D-LEC) and colon (C-LEC) express substantially less tyrosinase mRNA than LN-LEC, and do not induce proliferation of tyrosinase-specific CD8 T-cells in vitro [59]. Additionally, 6 out of 7 other PTA tested were more highly expressed in LN-LEC compared to D-LEC or C-LEC. Furthermore, D-LEC and CLEC express substantially less PD-L1 than LN-LEC. Collectively, these results suggest that tolerance induction is a specialized property of LN-LEC not shared by those in tissue lymphatics.

Within the LN, LEC are found in the subcapsular sinus, the cortical sinus, and the medullary sinus. The afferent lymphatics drain into the subcapsular sinus, which forms a thin structure at the outer edge of the LN [60]. DC enter the LN parenchyma through the floor of the subcapsular sinus, while T-cells in the afferent lymph pass through the subcapsular sinus to the medullary sinus, where they enter the LN [61]. To exit the LN, lymphocytes first enter blunt-ended cortical sinuses, which are interspersed throughout the T and B cell zone [62]. Lymphatic fluid in the cortical sinuses flows towards the medullary sinus, and lymphocytes ultimately leave the LN through the medullary sinus in the efferent lymph [62-66]. We showed that LEC in these different sinuses can be distinguished by differential expression of PD-L1, ICAM-1, MAdCAM-1, and LTßR: subcapsular sinus LEC are PD-L1hiICAM-1hiMAdCAM-1+LTßRlo, medullary sinus LEC are PD-L1hiICAM-1hiMAdCAM-1negLTßR+, and cortical sinus LEC are PD-L1intICAM-1intMAdCAM-1negLTßR+ [59]. In addition to expressing high levels of PD-L1, medullary LEC are the only subset that expressed a sufficient level of tyrosinase to activate tyrosinase-specific CD8 T-cells. Since the medullary sinus is an exit from the LN, this suggests a model in which LEC function as gatekeepers, engaging and inducing deletion of activated self-reactive CD8 T-cells as they attempt to leave. This model is also intriguing because LEC express the highest level of PD-L1 of any LNSC. A question that remains unaddressed is whether LEC may exert a more generalized quality control role, inducing the deletion of suboptimally activated T-cells, regardless of whether or not the LEC display the cognate antigen for T-cell recognition.

The specific microenvironmental influences that control the phenotypic distinctions between LEC in the periphery and in different LN sinuses remain to be fully understood. Within the LN, we have shown that high level expression of PD-L1 on medullary LEC and MAdCAM-1 on subcapsular sinus LEC is dependent on LTßR signaling and B-cells, but not DC, and that these two signals are independent of one another [59]. Interestingly, the presence of T-cells showed the opposite effect. Tyrosinase expression was not affected by any of these manipulations. We also found that, while a medullary region is present by postnatal day 7, LEC from these neonatal mice do not present tyrosinase and the expression of PD-L1 on LEC is substantially lower than that in adult mice. Combined, these results indicate that the tolerogenic phenotype of LN-LEC develops after the neonatal period in a way that is influenced by, but not entirely dependent on, the effects of lymphocytes and LTßR signaling. Previous work has shown that thymic tolerance is most critical during the perinatal period [67] and that naïve neonatal T-cells directly access peripheral tissues and are tolerized there, instead of in draining LN [68]. This suggests that the relative importance of each tolerogenic site and the associated tolerogenic APC shifts from neonatal to adult animals [69]: during the initial waves of neonatal T-cell development the majority of tolerance occurs in the thymus, and T-cells specific for PTA not expressed in the thymus can be tolerized directly in the peripheral tissues. Later in life, as thymic output decreases and the peripheral tissues become inaccessible to naïve T-cells, peripheral tolerance by LNSC, eTAC, and DC in LN becomes relatively more important to ensure continual tolerance of circulating T-cells.

Homeostatic Functions of LEC in Immunity

In addition to tolerizing self-reactive T-cells, LEC also influence several other aspects of the immune response. LN and tissue LEC are important sources of IL-7 [70-73], which is an homeostatic survival cytokine for naïve and memory T-cells. IL-7 secretion by LEC is enhanced during lymphopenia, LN remodeling, and revascularization after transplant, and this assists in re-establishing normal LN architecture and cellularity after an infection or other perturbation [71,72]. IL-7 promotes survival and differentiation of memory CD8 T-cells [74,75], suggesting that increased IL-7 production during LN remodeling in the resolution phase of an infection could potentially play a role in enhancing T-cell memory. Additionally, both LN-based and tissue-based LECs express IL-7R and respond to IL-7 in an autocrine fashion, which is required for normal lymphangiogenesis and efficient lymphatic drainage [73]. IL-7R-/- animals have thin and highly branched lymph vessels, suggesting IL-7 might stabilize larger lymphatic vessels. IL-7 also induces VEGF-D secretion in cancer cells, suggesting IL-7 may enhance canonical lymphangiogenesis through the VEGF pathway [76,77]. Through these mechanisms, LEC help control the size of the T-cell compartment.

LEC also control T-cell egress from the LN. Lymphocytes express a receptor for sphingosine-1-phosphate (S1P), a lipid that is present at a relatively high concentration in plasma, but is generally at a low concentration in LN. However, LEC synthesize S1P, providing a high local concentration in their vicinity [51]. Expression of the S1P receptor, S1PR1, on lymphocytes is endocytically downregulated by prolonged exposure to S1P in plasma and by the activation marker CD69 [78,79], but is re-expressed upon entry into LN. Signaling through S1PR1 as lymphocytes encounter LEC allows them to overcome CCR7-mediated retention signals and leave the LN [63,65]. S1P is upregulated when tenascin C, a marker of inflammation induced by a variety of pathogen or damage associated molecular patterns, binds to a9-integrin on LN cortical and medullary sinus LEC, and blockade of a9-integrin inhibits lymphocyte egress [80]. Reciprocal regulation of CD69 and S1P1 prevents activated T-cells from leaving the LN during an immune response, and may also serve a similar function during self-tolerance. Tyrosinase specific T-cells tolerized by LEC undergo several rounds of proliferation before expressing the high-level of PD-1 that mediates deletion. Therefore, retention in the LN may encourage the Tcells to remain in proximity with PD-L1+ LEC. Alternatively, self-reactive T-cells may leave their initial activating LN, and encounter PD-L1 on LEC in a downstream LN to complete the tolerogenic process. Regardless, production of IL-7 and S1P are two additional ways that LEC regulate the immune response, by controlling the homeostasis of naïve and memory lymphocytes as well as their ability to exit the LN.

LEC Restrain T-cell Proliferation in Response to Inflammation

Inflammation has a myriad of effects on lymphatics and the immune response. TLR ligation induces macrophages to secrete VEGF-C/D, which binds to VEGFR-3, triggering proliferation of tissue LEC and lymphangiogenesis [81,82]. IFN?, TNFa, and TLR ligands increase expression of chemokines and adhesion molecules on LEC, thereby enhancing cell recruitment and migration towards the LN [83,84]. These processes have been extensively reviewed elsewhere [84-87]. Inflammation has the potential to adversely affect tolerance, as inflammatory cytokines such as IFN? and TNFa can mature DC, leading to the upregulation of costimulatory molecules and the potential for immunogenic presentation of self-antigens acquired in the periphery. LEC help dampen cytokine-induced DC maturation, as TNFa stimulated LEC decrease the expression of the costimulatory molecule CD86 on immature or TNFa stimulated DC and decrease the ability of the DC to stimulate T-cell proliferation [88]. Although the exact mechanism is uncertain, it requires adhesion of the DC to the LEC through ICAM-1/Mac-1 interactions. TNFa stimulated LEC do not affect DC matured with LPS, suggesting that this mechanism only occurs in the absence of pathogen associated molecular patterns, thus potentially contributing to the resolution of inflammation after clearance of infection. LN LEC and FRC also respond to the pro-inflammatory cytokines IFN? and TNFa by secreting nitric oxide (NO), which limits the proliferation but not effector activity of already activated T-cells [89,90]. Additionally, IFN?-stimulated cultured human LN-LEC produce indoleamine 2,3 dioxygenase and suppress CD4 T-cell proliferation [91]. Both of these may curtail excessive T-cell expansion to prevent disruption of LN architecture. Interestingly, NO production by FRC also reduced proliferation of self-reactive OT-I CD8 T-cells in vivo [90]. Although the effects on deletion were not investigated, this mechanism may also ensure that the proliferating self-reactive T-cells do not expand too rapidly and potentially overwhelm the tolerogenic capacity of the LNSC. However, self-reactive CD8 T-cells generally do not produce IFN? or TNFa after tolerogenic activation [92] (unpublished data), so the relevance of this mechanism may also vary depending on the characteristics and activation state of the T-cells.

TLR ligation also alters the ability of LEC and FRC to induce tolerance. Primary murine LEC and FRC express TLR3, and treatment with the TLR3 ligand Poly(I:C) upregulates PD-L1 on LEC and FRC but does not change expression of CD80 or CD86 [13]. Poly(I:C) downregulates the PTA OVA in FRC of iFABP-OVA mice, leading to reduced OVA specific CD8 T-cell proliferation in vitro; however, the functional consequences in vivo were not evaluated. Interestingly, other PTA were either down or up-regulated by TLR3 signaling in both LEC and FRC. PTA downregulation may be an attempt to maintain the ignorance of self-reactive T-cells until after inflammatory conditions have passed, while the upregulation of PD-L1 may help enforce tolerance of any self-reactive T-cells that get activated. The significance of PTA upregulation remains unclear. Additionally, although Poly(I:C) does not upregulate costimulatory molecules on LEC, future work is needed to determine if other inflammatory circumstances can lead to immunogenic activation of T-cells recognizing PTA or endocytosed antigens presented by LEC. Combined, these studies suggest that LN-LEC respond to inflammation by dampening T-cell proliferation, which likely helps ensure continued T-cell tolerance and protects LNSC from damage during an overly vigorous immune response.

LEC-induced tolerance: a new target for cancer immunotherapy?

Studies performed to date suggest that LEC can enhance tumor growth by either increasing tumor metastasis to the LN through the formation of tumor draining lymphatics [93], or by inducing tolerance of tumor-reactive T-cells [12,20,26,94,95]. This suggests that inhibiting LEC-induced tolerance may provide a method of boosting anti-tumor immunotherapy. Indeed, while tyrosinase is overexpressed in melanoma and is a target of melanoma immunotherapy, LEC-mediated self-tolerance to tyrosinase limits active immunotherapy [95-97]. PD-1 inhibitory antibodies represent one approach to mitigating these effects, and incidentally already show great promise as a monotherapy independently of cancer vaccines in clinical trials [39,98]. These antibodies are currently being tested for their ability to revitalize exhausted effector T-cells and prevent tumor immune evasion [98]. However, our work has established that PD-1 blockade also inhibits LEC-induced tolerance, and this suggests that anti-PD-1 blockade may particularly complement efforts to specifically target tyrosinase using cancer vaccines or T-cell adoptive therapy. This combination therapy may provide a synergistic benefit by inhibiting tolerance and simultaneously preventing T-cell exhaustion. Increasing our understanding of the role of LEC in T-cell tolerance may provide new opportunities to enhance cancer immunotherapies.

Technical Challenges and Opportunities in LEC Research

Studying the role of LEC in tolerance and immunity presents a number of challenges due to the rarity and fragility of this cell type. LEC represent about 0.2% of the cells in the LN, and on average less than 50,000 LEC are isolated from the major LN per mouse [13,59]. The small numbers of LECs makes many biochemical techniques impractical on ex vivo samples. As a result, several groups have optimized protocols to expand LNSC and LEC in culture [99,100]. However, the phenotype of LEC changes rapidly in culture, as the surrounding microenvironment contributes to the maintenance of LEC differentiation in vivo [59,101-103]. Notably, we have found that mRNA levels of the PTA tyrosinase and Mart1 diminish more than 50-fold, and expression of PD-L1 and HVEM is lost after 5 days in culture (unpublished). Additionally, the geography of the LN and the localization of LEC at the LN entrances and exits may affect how LEC interact with other cells, and the physiological roles of LEC in the LN. This information is lost in vitro. As a result, it is important to confirm the physiological relevance of all in vitro observations with an in vivo system. Fortunately, a number of mouse models have recently become available to facilitate in vivo research. LEC are a radioresistant cell, so bone marrow chimeras can be used to localize genetic knockouts to either the hematopoietic or radioresistant compartments. Several groups have also developed mice expressing Cre-recombinase in LEC, under either the control of the Lyve-1 [51], Prox1 [50,104], or podoplanin [105] promoters. These mice can be used to specifically delete floxed genes in LEC. Additionally, reporter mice expressing tdTomato [106], mOrange2 [107], or GFP [108] under the control of the Prox1 promoter allows for intravital two-photon imaging and lymphatic tracing. These tools will provide new opportunities to identify, track, and manipulate LEC in vivo.

Acknowledgements

This work was supported in part by USPHS grant AI099033 and AI109250 to V.H.E, USPHS training grants AI07496 to S.J.R and E.F.T and GM007267 to S.J.R and PF-10-156-01-LIB from the American Cancer Society to E.F.T.

References

  1. Walker LS, Abbas AK (2002) The enemy within: keeping self-reactive T cells at bay in the periphery. Nat Rev Immunol 2: 11-19.
  2. Xing Y, Hogquist KA (2012) T-cell tolerance: central and peripheral. Cold Spring Harb Perspect Biol 4.
  3. Derbinski J, Gäbler J, Brors B, Tierling S, Jonnakuty S, et al. (2005) Promiscuous gene expression in thymic epithelial cells is regulated at multiple levels. J Exp Med 202: 33-45.
  4. Bonasio R, Scimone ML, Schaerli P, Grabie N, Lichtman AH, et al. (2006) Clonal deletion of thymocytes by circulating dendritic cells homing to the thymus. Nat Immunol 7: 1092-1100.
  5. Kurts C, Kosaka H, Carbone FR, Miller JFAP, Heath WR (1997) Class I-restricted crosspresentation of exogenous self-antigens leads to deletion of autoreactive CD8+ T Cells. J Exp Med 186: 239-245.
  6. Adler AJ, Marsh DW, Yochum GS, Guzzo JL, Nigam A, et al. (1998) CD4+ T cell tolerance to parenchymal self-antigens requires presentation by bone marrow-derived antigen-presenting cells. J Exp Med 187: 1555-1564.
  7. Steinman RM, Turley S, Mellman I, Inaba K (2000) The induction of tolerance by dendritic cells that have captured apoptotic cells. J Exp Med 191: 411-416.
  8. Heath WR, Kurts C, Miller JF, Carbone FR (1998) Cross-tolerance: a pathway for inducing tolerance to peripheral tissue antigens. J Exp Med 187: 1549-1553.
  9. Derbinski J, Schulte A, Kyewski B, Klein L (2001) Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self. Nat Immunol 2: 1032-1039.
  10. Anderson MS, Venanzi ES, Klein L, Chen Z, Berzins SP, et al. (2002) Projection of an immunological self shadow within the thymus by the aire protein. Science 298: 1395-1401.
  11. Gardner JM, Devoss JJ, Friedman RS, Wong DJ, Tan YX, et al. (2008) Deletional tolerance mediated by extrathymic Aire-expressing cells. Science 321: 843-847.
  12. Cohen JN, Guidi CJ, Tewalt EF, Qiao H, Rouhani SJ, et al. (2010) Lymph node-resident lymphatic endothelial cells mediate peripheral tolerance via Aire-independent direct antigen presentation. J Exp Med 207: 681-688.
  13. Fletcher AL, Lukacs-Kornek V, Reynoso ED, Pinner SE, Bellemare-Pelletier A, et al. (2010) Lymph node fibroblastic reticular cells directly present peripheral tissue antigen under steady-state and inflammatory conditions. J Exp Med 207: 689-697.
  14. Villaseñor J, Besse W, Benoist C, Mathis D (2008) Ectopic expression of peripheral-tissue antigens in the thymic epithelium: probabilistic, monoallelic, misinitiated. Proc Natl Acad Sci U S A 105: 15854-15859.
  15. Derbinski J, Pinto S, Rösch S, Hexel K, Kyewski B (2008) Promiscuous gene expression patterns in single medullary thymic epithelial cells argue for a stochastic mechanism. Proc Natl Acad Sci U S A 105: 657-662.
  16. Johnnidis JB, Venanzi ES, Taxman DJ, Ting JP, Benoist CO, et al. (2005) Chromosomal clustering of genes controlled by the aire transcription factor. Proc Natl Acad Sci U S A 102: 7233-7238.
  17. Gibson TJ, Ramu C, Gemünd C, Aasland R (1998) The APECED polyglandular autoimmune syndrome protein, AIRE-1, contains the SAND domain and is probably a transcription factor. Trends Biochem Sci 23: 242-244.
  18. Yip L, Su L, Sheng D, Chang P, Atkinson M, et al. (2009) Deaf1 isoforms control the expression of genes encoding peripheral tissue antigens in the pancreatic lymph nodes during type 1 diabetes. Nat Immunol 10: 1026-1033.
  19. Yip L, Creusot RJ, Pager CT, Sarnow P, Fathman CG (2013) Reduced DEAF1 function during type 1 diabetes inhibits translation in lymph node stromal cells by suppressing Eif4g3. J Mol Cell Biol 5: 99-110.
  20. Lund AW, Duraes FV, Hirosue S, Raghavan VR, Nembrini C, et al. (2012) VEGF-C promotes immune tolerance in B16 melanomas and cross-presentation of tumor antigen by lymph node lymphatics. Cell Rep 1: 191-199.
  21. Hirosue S, Vokali E, Raghavan VR, Rincon-Restrepo M, Lund AW, et al. (2014) Steady-state antigen scavenging, cross-presentation, and CD8+ T cell priming: a new role for lymphatic endothelial cells. J Immunol 192: 5002-5011.
  22. Sixt M, Kanazawa N, Selg M, Samson T, Roos G, et al. (2005) The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity 22: 19-29.
  23. Roozendaal R, Mempel TR, Pitcher LA, Gonzalez SF, Verschoor A, et al. (2009) Conduits mediate transport of low-molecular-weight antigen to lymph node follicles. Immunity 30: 264-276.
  24. Gretz JE, Norbury CC, Anderson AO, Proudfoot AEI, Shaw S (2000) Lymph-Borne Chemokines and Other Low Molecular Weight Molecules Reach High Endothelial Venules via Specialized Conduits While a Functional Barrier Limits Access to the Lymphocyte Microenvironments in Lymph Node Cortex. J Exp Med 192: 1425-1440.
  25. Nichols LA1, Chen Y, Colella TA, Bennett CL, Clausen BE, et al. (2007) Deletional self-tolerance to a melanocyte/melanoma antigen derived from tyrosinase is mediated by a radio-resistant cell in peripheral and mesenteric lymph nodes. J Immunol 179: 993-1003.
  26. Tewalt EF, Cohen JN, Rouhani SJ, Guidi CJ, Qiao H, et al. (2012) Lymphatic endothelial cells induce tolerance via PD-L1 and lack of costimulation leading to high-level PD-1 expression on CD8 T cells. Blood 120: 4772-4782.
  27. Harding FA, McArthur JG, Gross JA, Raulet DH, Allison JP (1992) CD28-mediated signalling co-stimulates murine T cells and prevents induction of anergy in T-cell clones. Nature 356: 607-609.
  28. Hawiger D, Inaba K, Dorsett Y, Guo M, Mahnke K, et al. (2001) Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J Exp Med 194: 769-779.
  29. Hernández J, Aung S, Marquardt K, Sherman LA (2002) Uncoupling of proliferative potential and gain of effector function by CD8(+) T cells responding to self-antigens. J Exp Med 196: 323-333.
  30. Keller AM, Schildknecht A, Xiao Y, van den Broek M, Borst J (2008) Expression of costimulatory ligand CD70 on steady-state dendritic cells breaks CD8+ T cell tolerance and permits effective immunity. Immunity 29: 934-946.
  31. Bandyopadhyay S, Long M, Qui HZ, Hagymasi AT, Slaiby AM, et al. (2008) Self-antigen prevents CD8 T cell effector differentiation by CD134 and CD137 dual costimulation. J Immunol 181: 7728-7737.
  32. Martin-Orozco N, Wang YH, Yagita H, Dong C (2006) Cutting Edge: Programmed death (PD) ligand-1/PD-1 interaction is required for CD8+ T cell tolerance to tissue antigens. J Immunol 177: 8291-8295.
  33. Liu X, Alexiou M, Martin-Orozco N, Chung Y, Nurieva RI, et al. (2009) Cutting edge: A critical role of B and T lymphocyte attenuator in peripheral T cell tolerance induction. J Immunol 182: 4516-4520.
  34. Reynoso ED, Elpek KG, Francisco L, Bronson R, Bellemare-Pelletier A, et al. (2009) Intestinal tolerance is converted to autoimmune enteritis upon PD-1 ligand blockade. J Immunol 182: 2102-2112.
  35. Goldberg MV, Maris CH, Hipkiss EL, Flies AS, Zhen L, et al. (2007) Role of PD-1 and its ligand, B7-H1, in early fate decisions of CD8 T cells. Blood 110: 186-192.
  36. Tsushima F, Yao S, Shin T, Flies A, Flies S, et al. (2007) Interaction between B7-H1 and PD-1 determines initiation and reversal of T-cell anergy. Blood 110: 180-185.
  37. Keir ME, Liang SC, Guleria I, Latchman YE, Qipo A, et al. (2006) Tissue expression of PD-L1 mediates peripheral T cell tolerance. J Exp Med 203: 883-895.
  38. Pedoeem A, Azoulay-Alfaguter I, Strazza M, Silverman GJ, Mor A (2014) Programmed death-1 pathway in cancer and autoimmunity. Clin Immunol 153: 145-152.
  39. Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, et al. (2014) Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 32: 1020-1030.
  40. Hinterberger M, Aichinger M, Prazeres da Costa O, Voehringer D, Hoffmann R, et al. (2010) Autonomous role of medullary thymic epithelial cells in central CD4(+) T cell tolerance. Nat Immunol 11: 512-519.
  41. Lee HM, Bautista JL, Scott-Browne J, Mohan JF, Hsieh CS (2012) A broad range of self-reactivity drives thymic regulatory T cell selection to limit responses to self. Immunity 37: 475-486.
  42. Ouyang W, Beckett O, Ma Q, Li MO (2010) Transforming growth factor-beta signaling curbs thymic negative selection promoting regulatory T cell development. Immunity 32: 642-653.
  43. Coombes JL, Siddiqui KR, Arancibia-Cárcamo CV, Hall J, Sun CM, et al. (2007) A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med 204: 1757-1764.
  44. Redmond WL, Marincek BC, Sherman LA (2005) Distinct requirements for deletion versus anergy during CD8 T cell peripheral tolerance in vivo. J Immunol 174: 2046-2053.
  45. Kretschmer K, Apostolou I, Hawiger D, Khazaie K, Nussenzweig MC, et al. (2005) Inducing and expanding regulatory T cell populations by foreign antigen. Nat Immunol 6: 1219-1227.
  46. Manicassamy S, Pulendran B (2011) Dendritic cell control of tolerogenic responses. Immunol Rev 241: 206-227.
  47. Gardner JM, Metzger TC, McMahon EJ, Au-Yeung BB, Krawisz AK, et al. (2013) Extrathymic Aire-expressing cells are a distinct bone marrow-derived population that induce functional inactivation of CD4⁺ T cells. Immunity 39: 560-572.
  48. Reith W, LeibundGut-Landmann S, Waldburger JM (2005) Regulation of MHC class II gene expression by the class II transactivator. Nat Rev Immunol 5: 793-806.
  49. Dubrot J, Duraes FV1, Potin L2, Capotosti F2, Brighouse D1, et al. (2014) Lymph node stromal cells acquire peptide-MHCII complexes from dendritic cells and induce antigen-specific CD4+ T cell tolerance. J Exp Med 211: 1153-1166.
  50. Bazigou E, Lyons OT, Smith A, Venn GE, Cope C, et al. (2011) Genes regulating lymphangiogenesis control venous valve formation and maintenance in mice. J Clin Invest 121: 2984-2992.
  51. Pham TH, Baluk P, Xu Y, Grigorova I, Bankovich AJ, et al. (2010) Lymphatic endothelial cell sphingosine kinase activity is required for lymphocyte egress and lymphatic patterning. J Exp Med 207: 17-27.
  52. Taniguchi RT, DeVoss JJ, Moon JJ, Sidney J, Sette A, et al. (2012) Detection of an autoreactive T-cell population within the polyclonal repertoire that undergoes distinct autoimmune regulator (Aire)-mediated selection. Proc Natl Acad Sci U S A 109: 7847-7852.
  53. Hubert FX, Kinkel SA, Davey GM, Phipson B, Mueller SN, et al. (2011) Aire regulates the transfer of antigen from mTECs to dendritic cells for induction of thymic tolerance. Blood 118: 2462-2472.
  54. Aschenbrenner K, D'Cruz LM, Vollmann EH, Hinterberger M, Emmerich J, et al. (2007) Selection of Foxp3+ regulatory T cells specific for self antigen expressed and presented by Aire+ medullary thymic epithelial cells. Nat Immunol 8: 351-358.
  55. Koble C, Kyewski B (2009) The thymic medulla: a unique microenvironment for intercellular self-antigen transfer. J Exp Med 206: 1505-1513.
  56. Nedjic J, Aichinger M, Emmerich J, Mizushima N, Klein L (2008) Autophagy in thymic epithelium shapes the T-cell repertoire and is essential for tolerance. Nature 455: 396-400.
  57. Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y (2004) In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell 15: 1101-1111.
  58. Aichinger M, Wu C, Nedjic J, Klein L (2013) Macroautophagy substrates are loaded onto MHC class II of medullary thymic epithelial cells for central tolerance. J Exp Med 210: 287-300.
  59. Cohen JN, Tewalt EF, Rouhani SJ, Buonomo EL, Bruce AN, et al. (2014) Tolerogenic properties of lymphatic endothelial cells are controlled by the lymph node microenvironment. PLoS One 9: e87740.
  60. Roozendaal R, Mebius RE, Kraal G (2008) The conduit system of the lymph node. Int Immunol 20: 1483-1487.
  61. Braun A, Worbs T, Moschovakis GL, Halle S, Hoffmann K, et al. (2011) Afferent lymph-derived T cells and DCs use different chemokine receptor CCR7-dependent routes for entry into the lymph node and intranodal migration. Nat Immunol 12: 879-887.
  62. Grigorova IL, Panteleev M, Cyster JG (2010) Lymph node cortical sinus organization and relationship to lymphocyte egress dynamics and antigen exposure. Proc Natl Acad Sci U S A 107: 20447-20452.
  63. Pham TH, Okada T, Matloubian M, Lo CG, Cyster JG (2008) S1P1 receptor signaling overrides retention mediated by G alpha i-coupled receptors to promote T cell egress. Immunity 28: 122-133.
  64. Sinha RK, Park C, Hwang I-Y, Davis MD, Kehrl JH (2009) B Lymphocytes Exit Lymph Nodes through Cortical Lymphatic Sinusoids by a Mechanism Independent of Sphingosine-1-Phosphate-Mediated Chemotaxis. Immunity 30: 434-446.
  65. Grigorova IL, Schwab SR, Phan TG, Pham TH, Okada T, et al. (2009) Cortical sinus probing, S1P1-dependent entry and flow-based capture of egressing T cells. Nat Immunol 10: 58-65.
  66. Wei SH, Rosen H, Matheu MP, Sanna MG, Wang SK, et al. (2005) Sphingosine 1-phosphate type 1 receptor agonism inhibits transendothelial migration of medullary T cells to lymphatic sinuses. Nat Immunol 6: 1228-1235.
  67. Guerau-de-Arellano M, Martinic M, Benoist C, Mathis D (2009) Neonatal tolerance revisited: a perinatal window for Aire control of autoimmunity. J Exp Med 206: 1245-1252.
  68. Alferink J, Tafuri A, Vestweber D, Hallmann R, Hämmerling GJ, et al. (1998) Control of neonatal tolerance to tissue antigens by peripheral T cell trafficking. Science 282: 1338-1341.
  69. Metzger TC, Anderson MS (2011) Control of central and peripheral tolerance by Aire. Immunol Rev 241: 89-103.
  70. Hara T, Shitara S, Imai K, Miyachi H, Kitano S, et al. (2012) Identification of IL-7-producing cells in primary and secondary lymphoid organs using IL-7-GFP knock-in mice. J Immunol 189: 1577-1584.
  71. Miller CN, Hartigan-O'Connor DJ, Lee MS, Laidlaw G, Cornelissen IP, et al. (2013) IL-7 production in murine lymphatic endothelial cells and induction in the setting of peripheral lymphopenia. Int Immunol 25: 471-483.
  72. Onder L, Narang P, Scandella E, Chai Q, Iolyeva M, et al. (2012) IL-7-producing stromal cells are critical for lymph node remodeling. Blood 120: 4675-4683.
  73. Iolyeva M, Aebischer D, Proulx ST, Willrodt AH, Ecoiffier T, et al. (2013) Interleukin-7 is produced by afferent lymphatic vessels and supports lymphatic drainage. Blood 122: 2271-2281.
  74. Schluns KS, Kieper WC, Jameson SC, Lefrançois L (2000) Interleukin-7 mediates the homeostasis of naïve and memory CD8 T cells in vivo. Nat Immunol 1: 426-432.
  75. Schluns KS, Lefrançois L (2003) Cytokine control of memory T-cell development and survival. Nat Rev Immunol 3: 269-279.
  76. Al-Rawi MA, Watkins G, Mansel RE, Jiang WG (2005) Interleukin 7 upregulates vascular endothelial growth factor D in breast cancer cells and induces lymphangiogenesis in vivo. Br J Surg 92: 305-310.
  77. Ming J, Zhang Q, Qiu X, Wang E (2009) Interleukin 7/interleukin 7 receptor induce c-Fos/c-Jun-dependent vascular endothelial growth factor-D up-regulation: a mechanism of lymphangiogenesis in lung cancer. Eur J Cancer 45: 866-873.
  78. Shiow LR, Rosen DB, Brdicková N, Xu Y, An J, et al. (2006) CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature 440: 540-544.
  79. Bankovich AJ, Shiow LR, Cyster JG (2010) CD69 suppresses sphingosine 1-phosophate receptor-1 (S1P1) function through interaction with membrane helix 4. J Biol Chem 285: 22328-22337.
  80. Ito K, Morimoto J, Kihara A, Matsui Y, Kurotaki D, et al. (2014) Integrin α9 on lymphatic endothelial cells regulates lymphocyte egress. Proc Natl Acad Sci U S A 111: 3080-3085.
  81. Kataru RP, Jung K, Jang C, Yang H, Schwendener RA, et al. (2009) Critical role of CD11b+ macrophages and VEGF in inflammatory lymphangiogenesis, antigen clearance, and inflammation resolution. Blood 113: 5650-5659.
  82. Kim KE, Koh Y-J, Jeon B-H, Jang C, Han J, et al. (2009) Role of CD11b+ macrophages in intraperitoneal lipopolysaccharide-induced aberrant lymphangiogenesis and lymphatic function in the diaphragm. Am J Pathol 175: 1733-1745.
  83. Pegu A, Qin S, Fallert Junecko BA, Nisato RE, Pepper MS, et al. (2008) Human lymphatic endothelial cells express multiple functional TLRs. J Immunol 180: 3399-3405.
  84. Aebischer D, Iolyeva M, Halin C (2014) The inflammatory response of lymphatic endothelium. Angiogenesis 17: 383-393.
  85. Liao S, von der Weid PY (2014) Inflammation-induced lymphangiogenesis and lymphatic dysfunction. Angiogenesis 17: 325-334.
  86. Johnson LA, Jackson DG (2014) Control of dendritic cell trafficking in lymphatics by chemokines. Angiogenesis 17: 335-345.
  87. Tan KW, Chong SZ, Angeli V (2014) Inflammatory lymphangiogenesis: cellular mediators and functional implications. Angiogenesis 17: 373-381.
  88. Podgrabinska S, Kamalu O, Mayer L, Shimaoka M, Snoeck H, et al. (2009) Inflamed lymphatic endothelium suppresses dendritic cell maturation and function via Mac-1/ICAM-1-dependent mechanism. J Immunol 183: 1767-1779.
  89. Khan O, Headley M, Gerard A, Wei W, Liu L, et al. (2011) Regulation of T cell priming by lymphoid stroma. PLoS One 6: e26138.
  90. Lukacs-Kornek V, Malhotra D, Fletcher AL, Acton SE, Elpek KG, et al. (2011) Regulated release of nitric oxide by nonhematopoietic stroma controls expansion of the activated T cell pool in lymph nodes. Nat Immunol 12: 1096-1104.
  91. Nörder M, Gutierrez MG, Zicari S, Cervi E, Caruso A, et al. (2012) Lymph node-derived lymphatic endothelial cells express functional costimulatory molecules and impair dendritic cell-induced allogenic T-cell proliferation. FASEB J 26: 2835-2846.
  92. Hernandez J, Aung S, Redmond WL, Sherman LA (2001) Phenotypic and functional analysis of CD8(+) T cells undergoing peripheral deletion in response to cross-presentation of self-antigen. J Exp Med 194: 707-717.
  93. Karaman S, Detmar M (2014) Mechanisms of lymphatic metastasis. J Clin Invest 124: 922-928.
  94. Kittlesen DJ, Thompson LW, Gulden PH, Skipper JC, Colella TA, et al. (1998) Human melanoma patients recognize an HLA-A1-restricted CTL epitope from tyrosinase containing two cysteine residues: implications for tumor vaccine development. J Immunol 160: 2099-2106.
  95. Colella TA, Bullock TN, Russell LB, Mullins DW, Overwijk WW, et al. (2000) Self-tolerance to the murine homologue of a tyrosinase-derived melanoma antigen: implications for tumor immunotherapy. J Exp Med 191: 1221-1232.
  96. Slingluff CL Jr, Chianese-Bullock KA, Bullock TN, Grosh WW, Mullins DW, et al. (2006) Immunity to melanoma antigens: from self-tolerance to immunotherapy. Adv Immunol 90: 243-295.
  97. Ferguson AR, Nichols LA, Zarling AL, Thompson ED, Brinkman CC, et al. (2008) Strategies and challenges in eliciting immunity to melanoma. Immunol Rev 222: 28-42.
  98. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, et al. (2012) Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366: 2443-2454.
  99. Jordan-Williams KL, Ruddell A (2014) Culturing Purifies Murine Lymph Node Lymphatic Endothelium. Lymphat Res Biol .
  100. Fletcher AL, Malhotra D, Acton SE, Lukacs-Kornek V, Bellemare-Pelletier A, et al. (2011) Reproducible Isolation of Lymph Node Stromal Cells Reveals Site-Dependent Differences in Fibroblastic Reticular Cells. Front Immunol 2: 35
  101. Lee S, Choi I, Hong YK (2010) Heterogeneity and plasticity of lymphatic endothelial cells. Semin Thromb Hemost 36: 352-361.
  102. Amatschek S, Kriehuber E, Bauer W, Reininger B, Meraner P, et al. (2007) Blood and lymphatic endothelial cell-specific differentiation programs are stringently controlled by the tissue environment. Blood 109: 4777-4785.
  103. Wick N, Saharinen P, Saharinen J, Gurnhofer E, Steiner CW, et al. (2007) Transcriptomal comparison of human dermal lymphatic endothelial cells ex vivo and in vitro. Physiol Genomics 28: 179-192.
  104. Srinivasan RS, Dillard ME, Lagutin OV, Lin FJ, Tsai S, et al. (2007) Lineage tracing demonstrates the venous origin of the mammalian lymphatic vasculature. Genes Dev 21: 2422-2432.
  105. Onder L, Scandella E, Chai Q, Firner S, Mayer CT, et al. (2011) A Novel Bacterial Artificial Chromosome-Transgenic Podoplanin-Cre Mouse Targets Lymphoid Organ Stromal Cells in vivo. Front Immunol 2: 50.
  106. Truman LA, Bentley KL, Smith EC, Massaro SA, Gonzalez DG, et al. (2012) ProxTom lymphatic vessel reporter mice reveal Prox1 expression in the adrenal medulla, megakaryocytes, and platelets. Am J Pathol 180: 1715-1725.
  107. Hägerling R, Pollmann C, Kremer L, Andresen V, Kiefer F (2011) Intravital two-photon microscopy of lymphatic vessel development and function using a transgenic Prox1 promoter-directed mOrange2 reporter mouse. Biochem Soc Trans 39: 1674-1681.
  108. Choi I, Chung HK, Ramu S, Lee HN, Kim KE, et al. (2011) Visualization of lymphatic vessels by Prox1-promoter directed GFP reporter in a bacterial artificial chromosome-based transgenic mouse. Blood 117: 362-365.
Citation: Rouhani SJ, Eccles JD, Tewalt EF, Engelhard VH (2014) Regulation of T-cell Tolerance by Lymphatic Endothelial Cells. J Clin Cell Immunol 5:242.

Copyright: © 2014 Rouhani SJ, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top