Abstract
High endothelial venules (HEVs) are blood vessels especially adapted for lymphocyte trafficking which are normally found in secondary lymphoid organs such as lymph nodes (LN) and Peyer's patches. It has long been known that HEVs develop in non-lymphoid organs during chronic inflammation driven by autoimmunity, infection or allografts. More recently, HEVs have been observed in solid, vascularized tumors and their presence correlated with reduced tumor size and improved patient outcome. It is proposed that newly formed HEV promote antitumor immunity by recruiting naive lymphocytes into the tumor, thus allowing the local generation of cancerous tissue-destroying lymphocytes. Understanding how HEVs develop and function are therefore important to unravel their role in human cancers. In LN, HEVs develop during embryonic and early post-natal life and are actively maintained by the LN microenvironment. Systemic blockade of lymphotoxin-β receptor leads to HEV de-differentiation, but the LN components that induce HEV differentiation have remained elusive. Recent elegant studies using gene-targeted mice have demonstrated clearly that triggering the lymphotoxin-β receptor in endothelial cells (EC) induces the differentiation of HEV and that CD11c+ dendritic cells play a crucial role in this process. It will be important to determine whether lymphotoxin-β receptor-dependent signaling in EC drives the development of HEV during tumorigenesis and which cells have HEV-inducer properties. This may reveal therapeutic approaches to promote HEV neogenesis and determine the impact of newly formed HEV on tumor immunity.
Keywords: dendritic cells, high endothelial venules, lymphotoxin-β receptor, T cell homing, tumor immunotherapy
Abbreviations
- EC
endothelial cells
- FRC
fibroblast reticular cells
- HEC
high endothelial cells
- HEV
high endothelial venules
- LN
lymph nodes
- LPA
lysophosphatidic acid
- LT
lymphotoxin
- LT-βR
lymphotoxin-β receptor
- MAdCAM
mucosal cell adhesion molecule
- PNAd
peripheral node addressin
- SIP
sphingosine-1-phosphate
- TLO
tertiary lymphoid organ
- VE-cadherin
vascular endothelial cadherin
- VEGF
vascular endothelial growth factor
Introduction
The raison d'etre of a LN is to filter lymph that passes through it, sequester incoming antigen and mount an appropriate response, be it activation, tolerance or homeostatic proliferation of lymphocytes. To do this, LN must sample the full repertoire of naive and memory lymphocytes in the body.1,2 Specialized blood vessels called HEVs are key players in this process because they extract naive and memory lymphocytes from the bloodstream, regardless of antigen receptor specificity, and deliver them into the node under homeostatic conditions.3 Here, lymphocytes scan dendritic cells as well as the supporting fibroblast reticular cell (FRC) network for activating, tolerogenic and homeostatic stimuli.4-7 Lymphocytes that do not encounter a cognate antigen leave the node within hours and re-enter the same or a different LN during lymphocyte recirculation, which is a fundamental for effective immunosurveillance.8 Following activation and differentiation, precursors of effector T lymphocytes, such as T helper cells and cytotoxic T cells, exit the LN via efferent lymphatics after 2–3 d, re-enter the bloodstream and are recruited to sites of inflammation by cytokine-activated blood vessels (which are not HEVs) to clear infection and repair damaged tissues (Fig. 1).
Figure 1.
The role of high endothelial venules in T lymphocyte dependent immunity. HEV recruit naive and central memory T lymphocytes from the bloodstream into lymph nodes where they scan antigen loaded dendritic cells that have migrated from infected, damaged or cancerous tissues (1). Following activation by antigen, activated T lymphocytes exit the lymph node via lymphatics and re-enter the bloodstream (2). Activated T lymphocytes are recruited to sites of inflammation by cytokine-activated blood vessels (which are not HEVs) to clear infected or cancerous tissue (3).
What are High Endothelial Venules (HEVs)?
HEVs form a branching network of post-capillary venules which is fully integrated into the normal blood vascular bed of all secondary lymphoid organs except the spleen. The HEV network is highly spatially organized, controlling both the site of lymphocyte entry and contributing to the structural organization of LN (Fig. 2). Incoming arteries arborize into a capillary bed in the outer cortex or B cell area of the LN and feed directly into the post-capillary venular network where HEVs are found. HEVs gradually increase in size from the smallest at the cortical–paracortical junction and largest vessels in the paracortex or T cell area of the node. HEVs merge with larger flat-walled venules in the medulla which drain into the collecting vein which exits the LN.9 A combination of ultrastructural and histochemical studies has been used to identify and characterize HEV within LN. In histological preparations, EC lining HEV have a characteristic cuboidal morphology which distinguishes HEV from other post-capillary venules (Fig. 3); it is this morphology that has engendered the name HEVs. Other characteristic features of HEV include a thickened apical glycocalyx and a thickened basal lamina.10 The endothelial lining is enveloped in overlapping layers of pericytes and pericyte-like FRC which form a prominent perivascular sheath that is part of the thickened basal lamina.11,12
Figure 2.
The migration of immune cells in and out of lymph nodes via high endothelial venules and lymphatics. The main artery into the node arborizes into a capillary bed in the outer cortex that leads directly into the post-capillary venular network where HEVs are located. HEVs increase in size as they traverse the paracortex or T cell area of the node and merge with flat-walled venules in the medulla. HEV are ensheathed by fibroblast reticular cells (FRC) that are continuous with the FRC-coated conduits that form the supporting internal scaffold on which lymphocytes and antigen presenting cells crawl during immunosurveillance (insert). Under homeostatic conditions HEV are major portals for entry of naive (TN), central memory (TCM) T and B cells as well as precursors of conventional dendritic cells (pre-DCs), natural killer (NK) cells and plasmacytoid dendritic cells (pDCs). Effector T cells (TE), NK cells, pDCs, neutrophils (PMN) and monocytes can be recruited by HEV in activated LN. Lymphatic vessels form a separate vascular system. Afferent lymphatics drain the surrounding area and deliver tissue-derived dendritic cells (DCs) to the FRC network and <70 kDa solutes to the basal lamina of HEV via the conduit system. Recirculating and activated lymphocytes leave via efferent lymphatics to re-enter the bloodstream.
Figure 3.
(See previous page). Distinguishing properties of high endothelial venules. (A) High endothelial venules (HEV) are lined with plump high endothelial cells (HEC) which contrast with flat endothelial cells (EC) lining non-specialized post-capillary venules. HEC are supported by a thick basal lamina and perivascular sheath of fibroblast reticular cells (FRC). HEV are also characterized by the presence of lymphocytes (Ly) within the endothelial cell lining and basal lamina as shown by transmission electron micrography. (B) HEV in subcutaneous (peripheral) lymph nodes of mice such as axillary LN selectively express peripheral LN addressin (PNAd) and HEV in mucosal associated lymphoid organs such as Peyer's patches selectively express the mucosal addressin MAdCAM-1. However HEV in other mucosal associated lymphoid organs such as mesenteric LN co-express PNAd and MAdCAM-1. C57BL/6 mice were injected with anti-PNAd (MECA-79) or anti-MAdCAM-1 (MECA-89) antibody and vibratome sections processed for whole mount immunohistochemistry. Scale bar is 50 μM for LN and 100 μM for Peyer's patches.
Although a defining feature, the characteristic endothelial morphology on its own cannot be relied on to identify HEV. The height of EC varies significantly between strains of mice, animal species and the method of tissue collection.10,13,14 High endothelial cells (HEC) express differentiation markers such as vascular endothelial (VE)-cadherin/CD144 and CD31 that confirm their endothelial identity; however, these markers are expressed by all vascular EC and are not specific for HEC.15 A more reliable marker for HEV is the expression of peripheral and/or mucosal addressin (Fig. 3). Addressins are expressed on the inner, apical surface of EC lining HEV and are ligands for homing receptors on lymphocytes. Thus, addressins identify the functional capacity of HEV to recruit lymphocytes from the bloodstream into LN.
In adult mice, expression of peripheral node addressin (PNAd), a ligand for L-selectin/CD62L, is a defining feature of HEV since it is not normally expressed by other types of blood vessel inside or outside of lymphoid organs,16 although PNAd staining is detected in some activated epithelia.17 PNAd expressing HEV are identified by immunohistochemical staining using the rat monoclonal antibody MECA-79 (Fig. 3). MECA-79 identifies 6-sulpho sialyl Lewisx, (a functional carbohydrate epitope that binds L-selectin) on extended core-1 branched O-linked sugars and detects HEV in human and murine tissues as well as in sheep LN which do not have characteristic high-walled HEV.10 The MECA-79 epitope is displayed on a number of serine/threonine-rich mucin domain containing proteins including CD34, GlyCAM-1, podocalyxin, endomucin and nepmucin.17 L-selectin also binds 6-sulpho sialyl Lewisx on core-2 branched O-linked sugars as well as N-linked sugars but these are not identified by MECA-79.18 Monoclonal antibodies that identify 6-sulpho sialyl LewisX on both O- and N-linked sugars in mice and humans (including those identified by MECA-79) have recently been described.19,20 Interestingly, PNAd is also expressed at the basolateral or ablumenal surface of HEC but its expression is regulated independently of apical PNAd. Basolaterally expressed PNAd has been shown to contribute to lymphocyte homing to LN but its precise role is not fully understood.17
The mucosal addressin (MAdCAM-1), a ligand for α4β7 integrin, is used to identify HEV in mucosal lymphoid organs (mesenteric LN and Peyer's patches) of adult mice. However, MAdCAM-1 is not a specific marker of HEV since it is also expressed by blood vessels in the gastro-intestinal lamina propria and the spleen; MAdCAM-1 is also expressed by stromal cells in embryonic LNs.21 The serine/threonine-rich mucin domain in murine MAdCAM-1 can be modified with the MECA-79 epitope and bind L-selectin as well as α4β7 integrin22; it is not clear if the less conserved mucin domain in human MAdCAM-1 binds L-selectin.23,24
The distribution of addressins in human lymphoid organs is similar to that reported in mice. PNAd positive, structurally distinct HEV have been reported in peripheral LN.25 MAdCAM-1 is preferentially expressed by HEV in mucosal associated lymphoid tissues, such as the appendix, but is also expressed by non-HEV blood vessels in the lamina propria and submucosa of the gastro-intestinal tract as well as marginal sinus lining cells of the spleen.25-27
In adults, PNAd and MAdCAM-1 were originally described to distinguish between HEV in peripheral (subcutaneous) and mucosal LN (mesenteric LN and Peyer's patches). However, expression of these two addressins overlaps in some LN. For example, PNAd is co-expressed by MAdCAM-1 positive HEV in mucosal associated lymphoid organs such as mouse mesenteric LN (Fig. 3) and human tonsils16,25 and PNAd dominates over MAdCAM-1 in HEV of mucosal lymphoid tissues that develop post-natally, such as nasal- , bronchial- and ocular-associated lymphoid tissues.28-30 Addressin expression is also developmentally regulated. PNAd and MAdCAM-1 are co-expressed by peripheral LN HEV in utero and neonatally, however PNAd expression is restricted to the baslolateral surface of HEC. During the first weeks of life, MAdCAM-1 expression is downregulated and PNAd is expressed at the apical surface as HEV complete maturation.31,32 Addressin expression is also regulated by immune activation; MAdCAM-1 can be re-expressed by peripheral LN HEV and PNAd expression can be downregulated in antigen-reactive LNs of adult mice with consequent changes to the homing properties of HEV.33,34
How Do HEVs Work?
Although widely used to identify HEV, PNAd is only one component of the molecular address required for lymphocytes to home to peripheral LN under homeostatic conditions. The role of apically expressed PNAd is to support the capture and rolling of L-selectin positive blood-borne leucocytes on the endothelial cell lining of HEV. Additional requirements are luminal expression of an arrest chemokine such as CCL21 (or CXCL13 for B cells)35 and ICAM-1/CD54, which supports LFA-1 integrin dependent arrest of rolling lymphocytes on the inner, luminal surface of HEV (Fig. 4).36,37 Naive and central memory T cells as well as B cells are recruited into peripheral LN under homeostatic conditions using this address code. Recent studies have shown that some innate immune cells enter LN under homeostatic conditions using, at least in part, L-selectin and/or CCR7. For example, precursors of classical dendritic cells (pre-DCs),38 natural killer (NK) cells39,40 and plasmacytoid dendritic cells (pDCs)41 have all been shown to enter peripheral LN in unperturbed mice, although in much lower numbers than T and B lymphocytes.
Figure 4.
Lymphocyte transmigration across high endothelial venules is a multistage process. High endothelial cells express a molecular address that captures and arrests blood-borne lymphocytes on the inner, luminal surface (1). Arrested lymphocytes crawl over the endothelial lining before transmigrating across the wall of HEV. Transmigration can be separated into distinct stages according to the location of migrating lymphocytes. Lymphocytes first transmigrate the endothelial lining where they can accumulate in HEV pockets (2). Lymphocytes can be retained in the sub-endothelial space (3) before completing diapedesis by crossing the basal lamina and perivascular sheath to enter the LN parenchyma (4). Inhibition of ADAM/MMPs arrests lymphocytes within the endothelial lining (stage 2) and the endothelial lining is thickened due to accumulated lymphocytes as shown by transmission electron micrography.52
A defining histological feature of HEV is the presence of lymphocytes within the endothelial cell lining and the surrounding basal lamina (Figs. 3 and 4)37 which suggests that transmigration across the HEV wall is regulated and rate-limiting. This is a complex event involving sequential interactions between migrating immune cells, EC, pericytes, and FRC which is only just starting to be understood. Intravital microscopic analysis of lymphocyte transmigration across HEV has shown that the first step of transendothelial migration from the apical to basolateral endothelial surface takes as little as 3 min.4 Although lymphocytes have been reported to penetrate the endothelial cell cytoplasm (transcellular migration) in vitro, they also take the conventional route between adjacent EC (paracellular migration).42 Lymphocytes take 10 min to migrate across the underlying basal lamina and the surrounding perivascular sheath, however, the close apposition of FRC to the basal lamina of HEV makes it difficult to separate these stages.36 Some progress has been made in identifying signaling pathways in lymphocytes that control migration across HEV. Studies using genetically modified mice and pharmacological agents have shown that cooperative signaling downstream of L-selectin and CCR7,43 downregulation of cell surface L-selectin by ectodomain proteolysis,44,45 affinity regulation of LFA-146 and the intermediate filament vimentin47 all regulate transendothelial migration but how these events are integrated is not understood. An additional key regulator of transendothelial migration is lysophosphatidic acid (LPA) which is generated locally by HEV-derived autotaxin and promotes transendothelial migration by inducing polarization and motility in lymphocytes.48,49 Lymphocytes must also be able to deform sufficiently to penetrate the HEV wall and this is achieved by contraction of the tail (uropod).50 During transmigration, lymphocytes can accumulate apparently within the endothelial lining of HEV in so-called “pockets” but these are extracellular, not intracellular.51 The lysophophospholipids LPA and sphingosine-1-phosphate (S1P) may control accumulation in HEV pockets49,51 but there are other mechanisms since lymphocytes accumulate within the endothelial lining of HEV in mice treated systemically with a dual metalloproteinase disintegrin (ADAM) and matrix metalloproteinase (MMP) inhibitor (Fig. 4), although HEV pockets were not identified in this study.52 Identifying the signaling pathways in high and flat EC that control lymphocyte transmigration53 may help in unraveling the role of HEV pockets in regulating lymphocyte entry into LN.
Compared to lymphocytes, pre-dendritic cells spend considerably longer within the walls of HEV before entering the LN parenchyma (5 h vs. 10 min for lymphocytes); whether dendritic cells reside in HEV pockets alongside lymphocytes with the potential for cellular cross-talk remains to be determined. What determines residence time within the HEV wall is also an important question to address. Adhesion molecules such as the leucocyte integrins are strong candidates since they switch rapidly between inactive and activate conformations during leucocyte recruitment. For example, VLA-3 integrin at the leading edge of transmigrating leucocytes binding to laminin in the basal lamina and LFA-1 at the trailing edge (uropod) binding to EC regulates retention vs. release of leucocytes in inflamed blood vessels.54 Other candidates include activated VLA-4 integrin which binds to fibronectin, another component of the basal lamina.55 L-selectin proteolysis may also regulate residence within the walls of HEV45 since PNAd is expressed at the basolateral endothelial cell surface17 and lymphocytes unable to downregulate L-selectin take longer to transmigrate HEV.44,52 The chemokine-rich basal lamina is also likely to control the onward migration of lymphocytes into the LN parenchyma.43,56
A unique feature of HEV which is extremely important for controlling lymphocyte recruitment is the connection with afferent lymph. The perivascular FRC sheath that surrounds HEV is continuous with the FRC coated conduit system within LN and forms a communicating unit that delivers incoming lymph-borne soluble factors, such as chemokines and cytokines, directly to the basal lamina of HEV (Fig. 2). Button-like attachments between adjacent HEC and reverse transcytosis allow access of chemokines to the luminal surface of HEV where they arrest rolling leucocytes.57 The connection with afferent lymph is also important to maintain fully differentiated HEV since expression of PNAd and CCL21 depend on continual stimulation by, as yet, unidentified components in afferent lymph (see below).33,58-60
The molecular address on HEC changes dramatically under inflammatory conditions, in part due to the HEV-afferent lymphatic connection which delivers inflammatory mediators from infected or damaged tissues directly to the basolateral surfaces of HEV. De novo expression of endothelial E- and P-selectins, increased expression of VCAM-1, presentation of inflammatory chemokines and binding of blood cells or microparticles allows recruitment of blood-borne leucocytes which are normally excluded under homeostatic conditions because they lack L-selectin and/or CCR7.61 Interactions between HEV and activated platelets are important to prevent blood loss in inflamed LN by maintaining vascular endothelial cadherin (VE-cadherin) expression on HEC34 and HEV bound platelets can also recruit L-selectin negative lymphocytes into LN.62 Depending on the infection or inflammatory stimulus, effector T cells,63 effector memory T cells,64 NK cells 39,65 pDCs,66 monocytes.67,68 and neutrophils69 can be recruited by HEV into activated LN and have divergent effects on the immune response. For example, effector T cell recruitment by activated HEV can progress or resolve ongoing immunity depending on whether antigen presenting cells are killed or primed by incoming effector T cells.63,64
HEVs in Cancer
The presence and precise location of tumor-infiltrating lymphocytes, particularly cytotoxic and memory T cells, is a predictor of clinical outcome in several vascularized tumors including colorectal, lung and ovarian cancer.70-75 Conventionally it is thought that effector T cells are generated in organized lymphoid tissues, such as draining LN, and recruited to tumor tissue from the bloodstream (Fig. 1). However, the recent finding of HEVs in a number of different human cancers is important since it raises the possibility that naive lymphocytes could be recruited into the tumor tissue via these newly formed blood vessels where an appropriate pro-inflammatory environment would allow the generation of cancerous tissue-destroying effector lymphocytes within the tumor tissue, thus avoiding the dilution associated with their redistribution from draining LN via the bloodstream.
PNAd expressing blood vessels with structural features of HEV have been reported in primary tumors of breast, lung and ovary, as well as in melanoma.76 The density of HEVs correlated with the extent of T- and B-lymphocyte infiltration of the tumor suggesting that, as in LN, HEVs are entry point for lymphocytes. In a detailed study of resected tumor tissue from 146 primary, invasive, non-metastatic breast cancers, the density of HEVs (number of vessels/tumor area) correlated with the numbers of infiltrating naive, memory and granzyme+ CD8+ T cells as well as a gene expression profile typical of Tbet, Th1, CD8+, and IFNγ+ cells.76 The clinical impact of HEVs in these patients following surgery for primary breast cancer was analyzed retrospectively and the density of HEVs correlated with disease-free, metastasis-free and overall survival rates for both global and node-positive breast cancers.76 In a study of 225 primary melanomas, the density of HEVs correlated strongly with reduced tumor size, expression of naive T- and Th1-associated genes and the presence of DC-LAMP+ dendritic cells.77 Although relatively small numbers of patients have been analyzed, these clinical data linking development of HEV to improved tumor immunity are supported by experimental studies in mice where the development of PNAd and/or MAdCAM-1 expressing vessels correlates with reduced tumor growth, increased recruitment of naive/central memory T cells and/or local expansion of T cells within the tumor.78-80 However, in a mouse model of infection (Helicobacter pylori)-induced carcinogenesis, the development of PNAd expressing HEV preceded adenocarcinoma formation.81 The impact of newly formed HEV on tumor outcome will also depend on whether functionally mature dendritic cells are present in sufficient numbers within the tumor tissue to present tumor-derived peptides to naive T cells and induce full T cell activation. Newly formed HEVs may also recruit immunosuppressive cells, such as regulatory T cells, which will limit effective antitumor immunity. It will, therefore, be important to determine the mechanisms underlying the antitumoral effects of HEV reported in breast cancer and melanoma and whether they operate in other types of clinical cancer.
Tumor Induced HEV in the Absence and Presence of Extra-Lymphoid Structures
It has long been known that PNAd expressing blood vessels develop at sites of chronic inflammation associated with autoimmunity, infection, allergic inflammation, or graft rejection in experimental mice and patients.17,28,82-84 These vessels show histological features characteristic of HEV such as cuboidal endothelium and lymphocytes transmigrating the vessel wall.25 In addition, they are most often surrounded by dense lymphocytic infiltrates organized into lymph-node like, T- and B-cell areas which are called tertiary lymphoid organs (TLOs) because of their resemblance to secondary lymphoid organs.
TLO have been reported in resected tumor tissues from a range of different cancers,85 including non-small cell lung cancer patients,72 metastatic melanomas86 and breast cancer.87,88 In colorectal cancer tissue, tumor-induced lymphoid follicles often containing germinal centers (Crohn's-like aggregates) are found not in the tumor tissue but in the surrounding peritumoral area (Fig. 5).89 The role of tumor-induced TLOs in regulating tumor progression is just starting to be analyzed. In retrospective studies of lung, breast and colorectal cancer, the presence of these structures is associated with improved patient survival in some studies.72,90-92 TLO that support somatic hypermutation and oligoclonal B cell expansion are found in invasive ductal breast carcinomas93 and in metastatic, but not primary melanomas,86 but the role of locally produced antibodies in tumor progression has not been determined. Experimental studies in mice have highlighted a protumoral, rather than an antitumoral, effect of tumor-induced TLOs. For instance, B16F10 melanoma cells expressing the CCR7 ligand, CCL21, induced the formation of lymphoid tissues at the tumor site, recruited Tregs and myeloid-derived suppressor cells (MDSC) and promoted tumor growth.94 The study by Martinet et al.76 is the first to measure the density of HEVs in clinical cancers. Although the co-localization of HEV with T- and B-lymphocyte infiltrates was reported, the density of lymphocytic infiltrates or whether they were organized into TLOs was not reported. In a recent study of colorectal cancer, HEV were rarely observed within tumor tissue but were found within organized lymphoid structures in the surrounding peritumoral area.95 As reported recently,96 the density of HEV containing TLOs did not correlate with improved survival for all stages of colorectal cancer.95 The types of immune cell recruited by tumor-derived HEV will be regulated by the local inflammatory environment as well as the organization of stromal cells during the development of TLO. Therefore, the impact of HEV on tumor immunity may change during progression of the disease. It will be interesting to determine whether the antitumoral effects of HEV reported in breast cancer and melanoma are modified by the formation of tumor-induced TLO.
Figure 5.
Tumor-infiltrating lymphocytes and tertiary lymphoid organs in colorectal cancer. The location and phenotype of CD3+ lymphocytes infiltrating the tumor tissue has been correlated with patient outcome (A).73 Lymphocytes are also found in tumor-induced tertiary lymphoid organs/lymphoid follicles in the peritumoral area (B). Tumor-infiltrating lymphocytes could be recruited directly from the bloodstream following their activation in draining LN or in peritumoral TLO and release into the circulation, as outlined in Figure. 1. Lymphocytes activated in peritumoural TLOs could bypass the bloodstream and migrate directly into the adjacent tumor tissue. Lymphocytes in cryostat sections of tumors were stained either for CD3 (A) or mismatch repair enzyme MLH1 (B).
Interestingly, PNAd expressing blood vessels have been reported in tumors in the apparent absence of TLO. For example, PNAd expressing HEV have been reported in primary melanoma in the absence of organized B-cell follicles86 and HEVs that form in tumor bearing mice following Treg depletion are not located within highly organized, LN-like T- and B cell infiltrates.80 PNAd expressing blood vessels that do not adopt the conventional structure of HEV have also been described in cancer97 and interestingly, tumor regression in primary cutaneous melanoma correlated better with the presence of PNAd+ vessels lined with flat as opposed to cuboidal EC.98 Flat-walled PNAd expressing blood vessels have been observed as early as 3 d following an inflammatory insult99 which suggests that HEV development outside of LN can be initiated independently of tertiary lymphoid organogenesis.
What Drives the Formation of HEV?
In the recent study of primary breast cancer, the density of PNAd expressing blood vessels was associated with longer disease-free survival,76 however tumor-induced blood vessel growth is thought to correlate with poor outcome. It is, therefore, important to identify components of the tumor microenvironment that control the development of PNAd expressing blood vessels. Currently, the development of PNAd expressing blood vessels is best understood in mouse LN.
PNAd expressing HEV develop in mouse LN during early neonatal life (Fig. 6). Since EC lining HEV are of vascular origin HEV neogenesis may represent differentiation of the LN post-capillary network under the influence of, as yet, unknown factors within the LN microenvironment. Several different approaches have demonstrated clearly that once formed, fully differentiated HEV are actively maintained by an intact stromal compartment. Following isolation of HEC from adult mice, PNAd expression, the distinct endothelial cell morphology and lymphocyte transmigration are downregulated within days,100 although rat HEC retain some differentiated properties in culture.101,102 Ligation of afferent lymphatics in mice and rats also results in HEV de-differentiation.58,103 Administration of a lymphotoxin-β decoy receptor (LTβR-Ig) phenocopies afferent lymphatic ligation in that PNAd expression and HEV function are downregulated in adult mice.104 Whether this was a direct effect of blocking LTβR signaling in EC or in other LTβR-expressing stromal cells such as pericytes, FRC or lymphatic EC was not determined.105-107
Figure 6.
The development of high endothelial venules inside and outside of lymph nodes. Lymph node: HEV develop as an integral part of the blood vasculature during embryonic and early post-natal life. Mucosal addressin (MAdCAM-1) is expressed on blood vessel endothelial cells in the late embryo. Luminal expression of peripheral node addressin (PNAd) is induced on MAdCAM-1 expressing blood vessels early in post-natal life and MAdCAM-1 expression is either maintained or downregulated. Engagement of lymphotoxin-β receptor (LT-βR) on endothelial cells drives the development of PNAd expressing HEV. Dendritic cells (DCs) and lymphatics vessels are required to maintain fully differentiated PNAd+ HEV and the size of the HEV network is regulated by CCR7+ DCs. The stimuli that organize the surrounding basal lamina, perivascular sheath and connecting conduits are not known. Tumor: Tumor-derived factors, such as vascular endothelial growth factor, stimulate the growth of new blood vessels to nourish the growing tumor. In mice, tumor-derived ligands for LT-βR stimulate HEV neogenesis and in primary non-invasive breast cancer, dendritic cells (DC) are a candidate HEV-inducer cell since they are a major source of lymphotoxin-β. Whether tumor-derived HEV arise from pre-existing blood vessels during tumor angiogenesis or develop from circulating endothelial progenitor cells during tumor vasculogenesis remains to be determined.
Several recent papers have started to unravel the complex relationship between LTβR signaling and the development of HEV and have highlighted important roles for dendritic cells in the differentiation and growth of HEV. Selective ablation of LTβR expression by vascular EC prevented the development of fully functional, PNAd expressing HEV able to support high levels of lymphocyte trafficking in peripheral LN of mice.108 Engagement of endothelial LTβR by CD11c+ cells is important in maintaining HEV differentiation since depletion of CD11c+ cells results in loss of HEV structure and function in a similar manner to afferent lymphatic ligation.109 A separate approach by the Forster lab identified a role for CCR7 expressing CD11c+ cells, not in maintaining HEV differentiation, but in regulating the overall size of the HEV network110 which suggests that HEV growth and differentiation may be regulated by different types of dendritic cell. Previous studies had shown that tissue-derived dendritic cells stimulate expansion of the HEV network via LTβR dependent release of vascular endothelial growth factor (VEGF)-A from the FRC network.105,111
Since the known LTβR ligands are membrane bound, an important question is which LT-αβ expressing dendritic cells make contact with EC to induce and/or maintain HEV differentiation. The study by Moussion and Girard109 showed that subcutaneously administered dendritic cells entering via the afferent lymphatics were able to maintain fully differentiated HEV in CD11c+ depleted mice.109 However, tissue-derived dendritic cells are attached to the FRC network inside LN and have not been reported to make contact with HEC.112-114 In contrast, pre-DCs contact HEC during recruitment from the bloodstream and reside within the HEV wall for up to 5 h but whether they express LT-αβ has not been determined.38 Studies using mice deficient in different types of dendritic cell will be useful to unravel the impact of dendritic cells and LTβR signaling on the development and growth of HEV.38,115 Other LT-αβ expressing cells which are important for LN organogenesis and remodeling should also be considered as HEV-inducer cells, such as RORγt+ lymphoid tissue inducer cells and B cells. It is also interesting to consider that the effect of afferent lymph on HEV differentiation may not be to deliver LT-αβ expressing HEV-inducer cells, such as tissue-derived dendritic cells, but rather as a source of chemokines which gain access to HEV via the conduit system where they position HEV-inducer cells alongside HEC to deliver the contact signals necessary for HEV differentiation.
Whether HEV development and growth are driven by dendritic cells and/or LTβR signaling in tumor blood vessel EC in clinical cancer remains to be determined (Fig. 6). Evidence in support of this hypothesis comes from a study of primary non-invasive breast cancer patients which showed that DC-LAMP+ dendritic cells represent the major source of LT-β in tumor tissues and their presence correlates with the density of HEV. In primary melanoma, the density of HEV also correlated with the presence of DC-LAMP+ dendritic cells.77 However, as in LN the majority of dendritic cells are localized outside the basal lamina of HEV and very few are in direct contact with HEC.116 LTβR dependent HEV neogenesis in seen in experimental animals in which LT-α or LIGHT are directly targeted to tumor cells,78 raising the possibility that cells other than dendritic cells could drive HEV neogenesis in cancer. Interestingly, direct intratumoural injection of CCL21 secreting dendritic cells recruited and primed naive tumor reactive T cells within the tumor and resulted in reduced tumor growth. In view of the findings of dendritic cell dependent HEV differentiation, it will be interesting to determine whether the effect of dendritic cells in controlling tumor growth depends on HEV neogenesis.117,118
Can Tumor Blood Vessels Be Manipulated to Promote HEV Dependent Lymphocyte Homing?
If the induction of HEVs in tumor tissue correlates with reduced tumor progression, an obvious goal would be to stimulate HEV neogenesis in tumors but we know very little about the factors that control HEV neogenesis outside of LN. Some clues have come from studies in which cytokines were ectopically expressed in pancreatic islets of mice. Expression of lymphotoxin-α induced MAdCAM-1 but both lymphotoxin-α and lymphotoxin-β were required to induce PNAd expressing blood vessels.119 The balance of LT-α vs. LT-αβ expressing cells may therefore drive the development of MAdCAM-1 and PNAd expressing HEV in non-lymphoid tissues.
LT-α and LT-αβ both activate the classical NF-κB pathway characterized by nuclear translocation of p50-RelA complexes. However, LT-αβ also activates the alternative, non-canonical NF-κB pathway that is hallmarked by NF-κB-inducing Kinase (NIK)-dependent activation of IκB kinase (IKK)-α and nuclear translocation of p52-RelB complexes.120 Non-canonical NF-κB signaling is thought to play a dominant role in HEV neogenesis since blockade of LTβR, but not TNFR, downregulates several HEV-specific markers such as GlyCAM-1, MAdCAM-1, CCL21 and HEC-6ST (the sulphotransferase that generates apically expressed PNAd).104 In addition, IKKα(AA) mutant mice with defective non-canonical NF-κB signaling lack functional HEV.28 Conversely, mice lacking full-length p100 protein which express constitutively active p52, display aberrant PNAd expressing HEV in the spleen.121 However, recent studies suggest that there is considerable overlap between classical and non-canonical NFκB signaling in driving the expression of HEV-associated genes.122,123 For example, recombinant TNF-α, LT-α and LT-αβ all induce expression of MAdCAM-1 in EC isolated from human and mouse tissues. HEC-6ST gene expression is induced in EC by soluble recombinant LT-αβ and by TNF-α in human, but not mouse, EC (MJM, unpublished).124-126 However, induction of PNAd glycoproteins has not been reported in EC isolated from non-lymphoid tissues, indicating that stimuli in addition to activation of NF-κB signaling are required for HEV neogenesis.
If HEV neogenesis could be induced how might this impact tumor immunity? Clinical data and experimental studies in mice suggest that the tumor microenvironment restricts the recruitment of cytotoxic, effector T lymphocytes from tumor blood vessels.127 This restriction could be considered an immune checkpoint which needs to be overcome for effective immunotherapy (Fig. 7). For example, following vaccination or adoptive T cell therapy of tumor-bearing patients, even when tumor-specific T cells comprise 20% of circulating lymphocytes, the outcome on tumor growth can be small.128 In mice, tumor blood vessels are anergic to inflammatory cytokines that upregulate CD8+ T cell homing in non-involved peritumoral vessels129 and tumor-derived factors, such as endothelin-B, suppress T cell recruitment by limiting endothelial expression of homing-associated molecules such as ICAM-1 and VCAM-1.130 Endothelial cell anergy may be related to the pro-angiogenic tumor environment; VEGF and FGF prevent cytokine induced homing molecule expression by EC.131 Another potential mechanism of limiting cytotoxic T cell entry is the induction of FasL on tumour EC.132 The recent finding that Foxp3 expressing Tregs suppress blood vessel differentiation by limiting HEV neogenesis in tumors is yet another strategy by which tumors restrict lymphocyte entry from bloostream.80
Figure 7.
Manipulating tumor blood vessels to promote T lymphocyte homing in cancer immunotherapy. Left Tumor blood vessels are anergic to inflammatory cytokines that normally upregulate endothelial cell (EC) expression of homing-associated molecules for T lymphocytes. Tumor-derived factors such as endothelin-B and vascular endothelial growth factor also limit the expression of homing-associated molecules thereby restricting the recruitment of T lymphocytes. Right The recruitment of pericytes to immature tumor blood vessels leads to vessel normalization which is associated with increased immune cell infiltration and reduce tumor growth. Vessel normalization reverses EC anergy and upregulates expression of homing-associated molecules which recruit cancer-destroying T lymphocyte. Tumor-derived HEV may recruit naive and central memory lymphocytes and allow the generation of tissue-destroying lymphocytes within the tumor tissue. The development of HEV in tumours may occur independently of vessel normalization.
Different experimental approaches have been explored to increase the recruitment of effector CD8+ T cells by tumor blood vessels. Targeted delivery of TNF-α using monoclonal antibodies that bind to tumor vessels promotes T lymphocyte infiltration of tumors indicating that endothelial cell anergy can be overcome.133 Expression of ICAM-1 and VCAM-1 can be induced by irradiation134 and hyperthermia induced IL-6 trans-signaling leads to increased effector T cell tumor infiltration and a reduction in tumor growth.129 Interestingly and somewhat counter-intuitively, anti-angiogenic therapy promotes CD8+ T cell infiltration of tumors131 and increases the efficacy of adoptive CD8+ T cell therapy in experimental mouse models.135 This could be a direct consequence of increased homing molecule expression on tumor vessels. However, rather than inhibit tumor angiogenesis, anti-angiogenic therapy has been reported to “normalize” tumor blood vessels by promoting pericyte recruitment and increasing tumor vessel perfusion.136 Pericyte maturation also promotes tumor blood vessel normalization and has been shown to increase immune cell infiltration and reduce tumor growth.137-139 It is possible that normalized tumor blood vessels recruit more lymphocytes because they can support the shear stresses required to maintain lymphocyte rolling140 and transmigration.141 The development of HEV could promote antitumor immunity by recruiting naive lymphocytes into the tumor, thus allowing the local generation of cancerous tissue-destroying lymphocytes as shown in mice.79 It is also possible that, as in LN, locally produced inflammatory mediators activate HEV to recruit effector cells which counteract the panoply of immunosuppressive cells which are enriched in vascularized tumors.142 It will, therefore, be important to determine which populations of immune cells are recruited by tumor-induced HEV to dissect their impact on tumor immunity.
Summary
The recent reports of HEVs in tumor tissue and a correlation with reduced tumor progression has generated interest in how these specialized blood vessels form and their impact on immune responses to tumors. The rise of cancer immunotherapy has re-focused attention on the tumor vasculature and the necessary role that it plays in recruiting effector lymphocytes able to destroy tumor cells. HEV neogenesis would represent a novel approach to cancer therapy which is diametrically opposed to the long-standing goal to block tumor angiogenesis. However, anti-angiogenesis therapies have not performed as well as first hoped. Although many aspects of HEV biology are still to be unraveled, the recent findings that lymphotoxin-β receptor-dependent signaling in EC is critical for the development and function of HEV are significant advances in our understanding and may provide therapeutic approaches to promote HEV neogenesis in tumors and determine the impact on HEV on tumor immunity.
Acknowledgments
We thank Diana Costa Bento, Emily Colbeck, Dr Angharad Watson and Professor Geraint Williams for their very helpful comments on the manuscript and Janice Sharp for the figures. The images of vascular addressin expression were generously provided by Lisa Newman and Dr Mark Coles of the University of York, UK and those of tumor infiltrating lymphocytes by Professor Mark Arends, University of Edinburgh.
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Funding
AA is supported by project grants from the Wellcome Trust (094511/Z/10/Z) and the Medical Research Council UK (MR/L008742/1) and MJM is supported by NIH RO1HL096642.
References
- 1.Girard JP, Moussion C, Forster R. HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes. Nat Rev Immunol 2012; 12:762-73; PMID:23018291; http://dx.doi.org/ 10.1038/nri3298 [DOI] [PubMed] [Google Scholar]
- 2.Masopust D, Schenkel JM. The integration of T cell migration, differentiation and function. Nat Rev Immunol 2013; 13:309-20; PMID:23598650; http://dx.doi.org/ 10.1038/nri3442 [DOI] [PubMed] [Google Scholar]
- 3.Gowans JL, Knight EJ. The route of recirculation of lymphocytes in the rat. Proc R Soc Series B 1964; 159:257-82; http://dx.doi.org/ 10.1098/rspb.1964.0001 [DOI] [PubMed] [Google Scholar]
- 4.Bajenoff M, Egen JG, Koo LY, Laugier JP, Brau F, Glaichenhaus N, Germain RN. Stromal cell networks regulate lymphocyte entry, migration, and territoriality in lymph nodes. Immunity 2006; 25:989-1001; PMID:17112751; http://dx.doi.org/ 10.1016/j.immuni.2006.10.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Mueller SN, Germain RN. Stromal cell contributions to the homeostasis and functionality of the immune system. Nat Rev Immunol 2009; 9:618-29; PMID:19644499; http://dx.doi.org/ 10.1038/nri2588 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Turley SJ, Fletcher AL, Elpek KG. The stromal and haematopoietic antigen-presenting cells that reside in secondary lymphoid organs. Nat Rev Immunol 2010; 10:813-25; PMID:21088682; http://dx.doi.org/ 10.1038/nri2886 [DOI] [PubMed] [Google Scholar]
- 7.Link A, Vogt TK, Favre S, Britschgi MR, Acha-Orbea H, Hinz B, Cyster JG, Luther SA. Fibroblastic reticular cells in lymph nodes regulate the homeostasis of naive T cells. Nat Immunol 2007; 8:1255-65; PMID:17893676; http://dx.doi.org/ 10.1038/ni1513 [DOI] [PubMed] [Google Scholar]
- 8.Gowans JL. The recirculation of lymphocytes from blood to lymph in the rat. J Physiol 1959; 146:54-69; PMID:13655215; http://dx.doi.org/ 10.1113/jphysiol.1959.sp006177 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Warnock RA, Askari S, Butcher EC, von Andrian UH. Molecular mechanisms of lymphocyte homing to peripheral lymph nodes. J Exp Med 1998; 187:205-16; PMID:9432978; http://dx.doi.org/ 10.1084/jem.187.2.205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Rosen SD. Ligands for L-selectin: homing, inflammation, and beyond. Annu Rev Immunol 2004; 22:129-56; PMID:15032576; http://dx.doi.org/ 10.1146/annurev.immunol.21.090501.080131 [DOI] [PubMed] [Google Scholar]
- 11.Anderson AO, Anderson ND. Lymphocyte emigration from high endothelial venules in rat lymph nodes. Immunol 1976; 31:731-48; PMID:992709 [PMC free article] [PubMed] [Google Scholar]
- 12.Anderson ND, Anderson AO, Wyllie RG. Specialized structure and metabolic activities of high endothelial venules in rat lymphatic tissues. Immunol 1976; 31:455-73; PMID:1027726 [PMC free article] [PubMed] [Google Scholar]
- 13.Belisle C, Sainte-Marie G. The narrowing of high endothelial venules of the rat lymph node. Anat Rec 1985; 211:184-91; PMID:3883847; http://dx.doi.org/ 10.1002/ar.1092110210 [DOI] [PubMed] [Google Scholar]
- 14.Fossum S, Smith ME, Ford WL. The migration of lymphocytes across specialized vascular endothelium VII. The migration of T and B lymphocytes from the blood of the athymic, nude rat. Scand J Immunol 1983; 17:539-50; PMID:6603012; http://dx.doi.org/ 10.1111/j.1365-3083.1983.tb00822.x [DOI] [PubMed] [Google Scholar]
- 15.Pfeiffer F, Kumar V, Butz S, Vestweber D, Imhof BA, Stein JV, Engelhardt B. Distinct molecular composition of blood and lymphatic vascular endothelial cell junctions establishes specific functional barriers within the peripheral lymph node. Eur J Immunol 2008; 38:2142-55; PMID:18629939; http://dx.doi.org/ 10.1002/eji.200838140 [DOI] [PubMed] [Google Scholar]
- 16.Streeter PR, Rouse BT, Butcher EC. Immunohistologic and functional characterization of a vascular addressin involved in lymphocyte homing into peripheral lymph nodes. J Cell Biol 1988; 107:1853-62; PMID:2460470; http://dx.doi.org/ 10.1083/jcb.107.5.1853 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Uchimura K, Rosen SD. Sulfated L-selectin ligands as a therapeutic target in chronic inflammation. Trends Immunol 2006; 27:559-65; PMID:17049924; http://dx.doi.org/ 10.1016/j.it.2006.10.007 [DOI] [PubMed] [Google Scholar]
- 18.Mitoma J, Bao X, Petryanik B, Schaerli P, Gauguet JM, Yu SY, Kawashima H, Saito H, Ohtsubo K, Marth JD et al.. Critical functions of N-glycans in L-selectin-mediated lymphocyte homing and recruitment. Nat Immunol 2007; 8:409-18; PMID:17334369; http://dx.doi.org/ 10.1038/ni1442 [DOI] [PubMed] [Google Scholar]
- 19.Hirakawa J, Tsuboi K, Sato K, Kobayashi M, Watanabe S, Takakura A, Imai Y, Ito Y, Fukuda M, Kawashima H. Novel anti-carbohydrate antibodies reveal the cooperative function of sulfated N- and O-glycans in lymphocyte homing. J Biol Chem 2010; 285:40864-78; PMID:20929857; http://dx.doi.org/ 10.1074/jbc.M110.167296 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Arata-Kawai H, Singer MS, Bistrup A, Zante A, Wang YQ, Ito Y, Bao X, Hemmerich S, Fukuda M, Rosen SD. Functional contributions of N- and O-glycans to L-selectin ligands in murine and human lymphoid organs. Am J Pathol 2011; 178:423-33; PMID:21224079; http://dx.doi.org/ 10.1016/j.ajpath.2010.11.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Streeter PR, Berg EL, Rouse BT, Bargatze RF, Butcher EC. A tissue-specific endothelial cell molecule involved in lymphocyte homing. Nature 1988; 331:41-6; PMID:3340147; http://dx.doi.org/ 10.1038/366695a0 [DOI] [PubMed] [Google Scholar]
- 22.Berg EL, McEvoy LM, Berlin C, Bargatze RF, Butcher EC. L-selectin-mediated lymphocyte rolling on MAdCAM-1. Nature 1993; 366:695-8; PMID:7505053; http://dx.doi.org/ 10.1038/366695a0 [DOI] [PubMed] [Google Scholar]
- 23.Shyjan AM, Bertagnolli M, Kenney CJ, Briskin MJ. Human mucosal addressin cell adhesion molecule-1 (MAdCAM-1) demonstrates structural and functional similarities to the alpha 4 beta 7-integrin binding domains of murine MAdCAM-1, but extreme divergence of mucin-like sequences. J Immunol 1996; 156:2851-7. [PubMed] [Google Scholar]
- 24.Briskin MJ, McEvoy LM, Butcher EC. MAdCAM-1 has homology to immunoglobulin and mucin-like adhesion receptors and to IgA1. Nature 1993; 363:461-4; PMID:8502297; http://dx.doi.org/ 10.1038/363461a0 [DOI] [PubMed] [Google Scholar]
- 25.Michie SA, Streeter PR, Bolt PA, Butcher EC, Picker LJ. The human peripheral lymph node vascular addressin. An inducible endothelial antigen involved in lymphocyte homing. Am J Pathol 1993; 143:1688-98. [PMC free article] [PubMed] [Google Scholar]
- 26.Briskin M, Winsor-Hines D, Shyjan A, Cochran N, Bloom S, Wilson J, McEvoy LM, Butcher EC, Kassam N, Mackay CR et al.. Human mucosal addressin cell adhesion molecule-1 is preferentially expressed in intestinal tract and associated lymphoid tissue. Am J Patho 1997; 151:97-110; PMCID: [PMC free article] [PubMed] [Google Scholar]
- 27.Pullen N, Molloy E, Carter D, Syntin P, Clemo F, Finco-Kent D, Reagan W, Zhao S, Kawabata T, Sreckovic S. Pharmacological characterization of PF-00547659, an anti-human MAdCAM monoclonal antibody. Br J Pharmacol 2009; 157:281-93; PMID:19366349; http://dx.doi.org/ 10.1111/j.1476-5381.2009.00137.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Drayton DL, Bonizzi G, Ying X, Liao S, Karin M, Ruddle NH. I kappa B kinase complex alpha kinase activity controls chemokine and high endothelial venule gene expression in lymph nodes and nasal-associated lymphoid tissue. J Immunol 2004; 173:6161-8; PMID:15528353; http://dx.doi.org/ 10.4049/jimmunol.173.10.6161 [DOI] [PubMed] [Google Scholar]
- 29.Xu B, Wagner N, Pham LN, Magno V, Shan Z, Butcher EC, Michie SA. Lymphocyte homing to bronchus-associated lymphoid tissue (BALT) is mediated by L-selectin/PNAd, alpha4beta1 integrin/VCAM-1, and LFA-1 adhesion pathways. J Exp Med 2003; 197:1255-67; PMID:12756264; http://dx.doi.org/ 10.1084/jem.20010685 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Nagatake T, Fukuyama S, Kim DY, Goda K, Igarashi O, Sato S, Nochi T, Sagara H, Yokota Y, Jetten AM. Id2-, RORgammat-, and LTbetaR-independent initiation of lymphoid organogenesis in ocular immunity. J Exp Med 2009; 206:2351-64; PMID:19822644; http://dx.doi.org/ 10.1084/jem.20091436 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Salmi M, Alanen K, Grenman S, Briskin M, Butcher EC, Jalkanen S. Immune cell trafficking in uterus and early life is dominated by the mucosal addressin MAdCAM-1 in humans. Gastroenterology 2001; 121:853-64; PMID:11606499; http://dx.doi.org/ 10.1053/gast.2001.27968 [DOI] [PubMed] [Google Scholar]
- 32.Mebius RE, Streeter PR, Michie S, Butcher EC, Weissman IL. A developmental switch in lymphocyte homing receptor and endothelial vascular addressin expression regulates lymphocyte homing and permits CD4+ CD3- cells to colonize lymph nodes. Proc Natl Acad Sci USA 1996; 93:11019-24; PMID:8855301; http://dx.doi.org/ 10.1073/pnas.93.20.11019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Liao S, Ruddle NH. Synchrony of high endothelial venules and lymphatic vessels revealed by immunization. J Immunol 2006; 177:3369-79; PMID:16920978; http://dx.doi.org/ 10.4049/jimmunol.177.5.3369 [DOI] [PubMed] [Google Scholar]
- 34.Herzog BH, Fu J, Wilson SJ, Hess PR, Sen A, McDaniel JM, Pan Y, Sheng M, Yago T, Silasi-Mansat R et al.. Podoplanin maintains high endothelial venule integrity by interacting with platelet CLEC-2. Nature 2013; 502:105-9; PMID:23995678; http://dx.doi.org/ 10.1038/nature12501 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Carlsen HS, Haraldsen G, Brandtzaeg P, Baekkevold ES. Disparate lymphoid chemokine expression in mice and men: no evidence of CCL21 synthesis by human high endothelial venules. Blood 2005; 106:444-6; PMID:15863780; http://dx.doi.org/ 10.1182/blood-2004-11-4353 [DOI] [PubMed] [Google Scholar]
- 36.Boscacci RT, Pfeiffer F, Gollmer K, Sevilla AI, Martin AM, Soriano SF, Natale D, Henrickson S, von Andrian UH, Fukui Y et al.. Comprehensive analysis of lymph node stroma-expressed Ig superfamily members reveals redundant and nonredundant roles for ICAM-1, ICAM-2, and VCAM-1 in lymphocyte homing. Blood 2010; 116:915-25; PMID:20395417; http://dx.doi.org/ 10.1182/blood-2009-11-254334 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Miyasaka M, Tanaka T. Lymphocyte trafficking across high endothelial venules: dogmas and enigmas. Nat Rev Immunol 2004; 4:360-70; PMID:15122201; http://dx.doi.org/ 10.1038/nri1354 [DOI] [PubMed] [Google Scholar]
- 38.Liu K, Victora GD, Schwickert TA, Guermonprez P, Meredith MM, Yao K, Chu FF, Randolph GJ, Rudensky AY, Nussenzweig M. In vivo analysis of dendritic cell development and homeostasis. Science 2009; 324:392-7; PMID:19286519; http://dx.doi.org/ 10.1126/science.1171243 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Diacovo TG, Blasius AL, Mak TW, Cella M, Colonna M. Adhesive mechanisms governing interferon-producing cell recruitment into lymph nodes. J Exp Med 2005; 202:687-96; PMID:16147979; http://dx.doi.org/ 10.1084/jem.20051035 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Garrod KR, Wei SH, Parker I, Cahalan MD. Natural killer cells actively patrol peripheral lymph nodes forming stable conjugates to eliminate MHC-mismatched targets. Proc Natl Acad Sci USA 2007; 104:12081-6; PMID:17609379; http://dx.doi.org/ 10.1073/pnas.0702867104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Seth S, Oberdorfer L, Hyde R, Hoff K, Thies V, Worbs T, Schmitz S, Förster R. CCR7 essentially contributes to the homing of plasmacytoid dendritic cells to lymph nodes under steady-state as well as inflammatory conditions. J Immunol 2011; 186:3364-72; PMID:21296980; http://dx.doi.org/ 10.4049/jimmunol.1002598 [DOI] [PubMed] [Google Scholar]
- 42.Dejana E. The transcellular railway: insights into leukocyte diapedesis. Nat Cell Biol 2006; 8:105-7; PMID:16450004; http://dx.doi.org/ 10.1038/ncb0206-105 [DOI] [PubMed] [Google Scholar]
- 43.Subramanian H, Grailer JJ, Ohlrich KC, Rymaszewski AL, Loppnow JJ, Kodera M, Conway RM, Steeber DA. Signaling through L-selectin mediates enhanced chemotaxis of lymphocyte subsets to secondary lymphoid tissue chemokine. J Immunol 2012; 188:3223-36; PMID:22387549; http://dx.doi.org/ 10.4049/jimmunol.1101032 [DOI] [PubMed] [Google Scholar]
- 44.Galkina E, Tanousis K, Preece G, Tolaini M, Kioussis D, Florey O, Haskard DO, Tedder TF, Ager A. L-selectin shedding does not regulate constitutive T cell trafficking but controls the migration pathways of antigen-activated T lymphocytes. J Exp Med 2003; 198:1323-35; PMID:14597735; http://dx.doi.org/ 10.1084/jem.20030485 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Klinger A, Gebert A, Bieber K, Kalies K, Ager A, Bell EB, Westermann J . Cyclical expression of L-selectin (CD62L) by recirculating T cells. Int Immunol 2009; 21:443-55; PMID:19240088; http://dx.doi.org/ 10.1093/intimm/dxp012 [DOI] [PubMed] [Google Scholar]
- 46.Park EJ, Peixoto A, Imai Y, Goodarzi A, Cheng G, Carman CV, von Andrian UH, Shimaoka M. Distinct roles for LFA-1 affinity regulation during T-cell adhesion, diapedesis, and interstitial migration in lymph nodes. Blood 2010; 115:1572-81; PMID:20023213; http://dx.doi.org/ 10.1182/blood-2009-08-237917 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Nieminen M, Henttinen T, Merinen M, Marttila-Ichihara F, Eriksson JE, Jalkanen S. Vimentin function in lymphocyte adhesion and transcellular migration. Nat Cell Biol 2006; 8:156-62; PMID:16429129; http://dx.doi.org/ 10.1038/ncb1355 [DOI] [PubMed] [Google Scholar]
- 48.Kanda H, Newton R, Klein R, Morita Y, Gunn MD, Rosen SD. Autotaxin, an ectoenzyme that produces lysophosphatidic acid, promotes the entry of lymphocytes into secondary lymphoid organs. Nat Immunol 2008; 9:415-23; PMID:18327261; http://dx.doi.org/ 10.1038/ni1573 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bai Z, Cai L, Umemoto E, Takeda A, Tohya K, Komai Y, Veeraveedu PT, Hata E, Sugiura Y, Kubo A et al.. Constitutive lymphocyte transmigration across the basal lamina of high endothelial venules is regulated by the autotaxin/lysophosphatidic acid axis. J Immunol 2013; 190:2036-48; PMID:23365076; http://dx.doi.org/ 10.4049/jimmunol.1202025 [DOI] [PubMed] [Google Scholar]
- 50.Soriano SF, Hons M, Schumann K, Kumar V, Dennier TJ, Lyck R, Sixt M, Stein JV. In vivo analysis of uropod function during physiological T cell trafficking. J Immunol 2011; 187:2356-64; PMID:21795598; http://dx.doi.org/ 10.4049/jimmunol.1100935 [DOI] [PubMed] [Google Scholar]
- 51.Mionnet C, Sanos SL, Mondor I, Jorquera A, Laugier JP, Germain RN, Bajénoff M. High endothelial venules as traffic control points maintaining lymphocyte population homeostasis in lymph nodes. Blood 2011; 118:6115-22; PMID:21937697; http://dx.doi.org/ 10.1182/blood-2011-07-367409 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Faveeuw C, Preece G, Ager A. Transendothelial migration of lymphocytes across high endothelial venules into lymph nodes is affected by metalloproteinases. Blood 2001; 98:688-95; PMID:11468168; http://dx.doi.org/ 10.1182/blood.V98.3.688 [DOI] [PubMed] [Google Scholar]
- 53.Lee M, Kiefel H, LaJevic MD, Macauley MS, Kawashima H,O'Hara E, Pan J,Paulson JC, Butcher EC . Transcriptional programs of lymphoid tissue capillary and high endothelium reveal control mechanisms for lymphocyte homing. Nat Immunol 2014; 15(10):982-995; PMID:4222088; http://dx.doi.org/ 10.1038/ni.2983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hyun YM, Sumagin R, Sarangi PP, Lomakina E, Overstreet MG, Baker CM, Fowell DJ, Waugh RE, Sarelius IH, Kim M. Uropod elongation is a common final step in leukocyte extravasation through inflamed vessels. J Exp Med 2012; 209:1349-62; PMID:22711877; http://dx.doi.org/ 10.1084/jem.20111426 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hourihan H, Allen TD, Ager A. Lymphocyte migration across high endothelium is associated with increases in alpha 4 beta 1 integrin (VLA-4) affinity. J Cell Sci 1993; 104:1049-59; http://dx.doi.org/ 10.1002/eji.1830130811 [DOI] [PubMed] [Google Scholar]
- 56.Yang BG, Tanaka T, Jang MH, Bai Z, Hayasaka H, Miyasaka M. Binding of lymphoid chemokines to collagen IV that accumulates in the basal lamina of high endothelial venules: its implications in lymphocyte trafficking. J Immunol 2007; 179:4376-82; http://dx.doi.org/ 10.4049/jimmunol.179.7.4376 [DOI] [PubMed] [Google Scholar]
- 57.Gretz JE, Norbury CC, Anderson AO, Proudfoot AE, Shaw S. 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 2000; 192:1425-40; PMID:11085745; http://dx.doi.org/ 10.1084/jem.192.10.1425 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hendricks HR, Eestermans IL. Disappearance and reappearance of high endothelial venules and immigrating lymphocytes in lymph nodes deprived of afferent lymphatic vessels: a possible regulatory role of macrophages in lymphocyte migration. Eur J Immunol 1983; 13:663-9; PMID:6884423; http://dx.doi.org/ 10.1002/eji.1830130811 [DOI] [PubMed] [Google Scholar]
- 59.Mebius RE, Streeter PR, Breve J, Duijvestijn AM, Kraal G. The influence of afferent lymphatic vessel interruption on vascular addressin expression. J Cell Biol 1991; 115:85-95; PMID:1918141; http://dx.doi.org/ 10.1083/jcb.115.1.85 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Drayson MT, Ford WL. Afferent lymph and lymph borne cells: their influence on lymph node function. Immunobiology 1984; 168:362-79; PMID:6530238; http://dx.doi.org/ 10.1016/S0171-2985(84)80123-0 [DOI] [PubMed] [Google Scholar]
- 61.Pruenster M, Mudde L, Bombosi P, Dimitrova S, Zsak M, Middleton J, Richmond A, Graham GJ, Segerer S, Nibbs RJ et al.. The Duffy antigen receptor for chemokines transports chemokines and supports their promigratory activity. Nat Immunol 2009; 10:101-8; PMID:19060902; http://dx.doi.org/ 10.1038/ni.1675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Diacovo TG, Catalina MD, Siegelman MH, von Andrian UH. Circulating activated platelets reconstitute lymphocyte homing and immunity in L-selectin-deficient mice. J Exp Med 1998; 187:197-204; PMID:9432977; http://dx.doi.org/ 10.1084/jem.187.2.197 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Guarda G, Hons M, Soriano SF, Huang AY, Polley R, Martin-Fontecha A, Stein JV, Germain RN, Lanzavecchia A, Sallusto F. L-selectin-negative CCR7- effector and memory CD8+ T cells enter reactive lymph nodes and kill dendritic cells. Nat Immunol 2007; 8:743-52; PMID:17529983; http://dx.doi.org/ 10.1038/ni1469 [DOI] [PubMed] [Google Scholar]
- 64.Martin-Fontecha A, Baumjohann D, Guarda G, Reboldi A, Hons M, Lanzavecchia A, Sallusto F. CD40L+ CD4+ memory T cells migrate in a CD62P-dependent fashion into reactive lymph nodes and license dendritic cells for T cell priming. J Exp Med 2008; 205:2561-74; PMID:18838544; http://dx.doi.org/ 10.1084/jem.20081212 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Martin-Fontecha A, Thomsen LL, Brett S, Gerard C, Lipp M, Lanzavecchia A, Sallusto F. Induced recruitment of NK cells to lymph nodes provides IFN-gamma for T(H)1 priming. Nat Immunol 2004; 5:1260-5; PMID:15531883; http://dx.doi.org/ 10.1038/ni1138 [DOI] [PubMed] [Google Scholar]
- 66.Yoneyama H, Matsuno K, Zhang Y, Nishiwaki T, Kitabatake M, Ueha S, Narumi S, Morikawa S, Ezaki T, Lu B et al.. Evidence for recruitment of plasmacytoid dendritic cell precursors to inflamed lymph nodes through high endothelial venules. Int Immunol 2004; 16:915-28; PMID:15159375; http://dx.doi.org/ 10.1093/intimm/dxh093 [DOI] [PubMed] [Google Scholar]
- 67.Palframan RT, Jung S, Cheng G, Weninger W, Luo Y, Dorf M, Littman DR, Rollins BJ, Zweerink H, Rot A et al.. Inflammatory chemokine transport and presentation in HEV: a remote control mechanism for monocyte recruitment to lymph nodes in inflamed tissues. J Exp Med 2001; 194:1361-73; PMID:11696600; http://dx.doi.org/ 10.1084/jem.194.9.1361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Janatpour MJ, Hudak S, Sathe M, Sedgwick JD, McEvoy LM. Tumor necrosis factor-dependent segmental control of MIG expression by high endothelial venules in inflamed lymph nodes regulates monocyte recruitment. J Exp Med 2001; 194:1375-84; PMID:11696601; http://dx.doi.org/ 10.1084/jem.194.9.1375 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Marchesi VT. The site of leucocyte emigration during inflammation. Quart J Exp Physiol 1961; 46:115-8; PMID:13766496; http://dx.doi.org/ 10.1113/expphysiol.1961.sp001522 [DOI] [PubMed] [Google Scholar]
- 70.Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pages C, Tosolini M, Camus M, Berger A, Wind P. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006; 313:1960-4; PMID:17008531; http://dx.doi.org/ 10.1126/science.1129139 [DOI] [PubMed] [Google Scholar]
- 71.Pages F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R, Mlecnik B, Kirilovsky A, Nilsson M, Damotte D et al.. Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med 2005; 353:2654-66; PMID:16371631; http://dx.doi.org/ 10.1056/NEJMoa051424 [DOI] [PubMed] [Google Scholar]
- 72.Dieu-Nosjean MC, Antoine M, Danel C, Heudes D, Wislez M, Poulot V, Rabbe N, Laurans L, Tartour E, de Chaisemartin L et al.. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol 2008; 26:4410-7; PMID:18802153; http://dx.doi.org/ 10.1200/JCO.2007.15.0284 [DOI] [PubMed] [Google Scholar]
- 73.Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN et al.. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 2003; 348:203-13; PMID:12529460; http://dx.doi.org/ 10.1056/NEJMoa020177 [DOI] [PubMed] [Google Scholar]
- 74.Fridman WH, Galon J, Pages F, Tartour E, Sautes-Fridman C, Kroemer G. Prognostic and predictive impact of intra- and peritumoral immune infiltrates. Cancer Res 2011; 71:5601-5; PMID:21846822; http://dx.doi.org/ 10.1158/0008-5472.CAN-11-1316 [DOI] [PubMed] [Google Scholar]
- 75.Disis ML. Immune regulation of cancer. J Clin Oncol 2010; 28:4531-8; PMID:20516428; http://dx.doi.org/ 10.1200/JCO.2009.27.2146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Martinet L, Garrido I, Filleron T, Le Guellec S, Bellard E, Fournie JJ, Rochaix P, Girard JP. Human solid tumors contain high endothelial venules: association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res 2011; 71:5678-87; PMID:21846823; http://dx.doi.org/ 10.1158/0008-5472.CAN-11-0431 [DOI] [PubMed] [Google Scholar]
- 77.Martinet L, Le Guellec S, Filleron T, Lamant L, Meyer N, Rochaix P, Garrido I, Girard JP. High endothelial venules (HEVs) in human melanoma lesions: Major gateways for tumor-infiltrating lymphocytes. Oncoimmunology 2012; 1:829-39; PMID:23162750; http://dx.doi.org/ 10.4161/onci.20492 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Schrama D, thor Straten P, Fischer WH, McLellan AD, Brocker EB, Reisfeld RA, Becker JC. Targeting of lymphotoxin-alpha to the tumor elicits an efficient immune response associated with induction of peripheral lymphoid-like tissue. Immunity 2001; 14:111-21; PMID:11239444; http://dx.doi.org/ 10.1016/S1074-7613(01)00094-2 [DOI] [PubMed] [Google Scholar]
- 79.Yu P, Lee Y, Liu W, Chin RK, Wang J, Wang Y, Schietinger A, Philip M, Schreiber H, Fu YX. Priming of naive T cells inside tumors leads to eradication of established tumors. Nat Immunol 2004; 5:141-9; PMID:14704792; http://dx.doi.org/ 10.1038/ni1029 [DOI] [PubMed] [Google Scholar]
- 80.Hindley JP, Jones E, Smart K, Bridgeman H, Lauder SN, Ondondo B, Cutting S, Ladell K, Wynn KK, Withers D et al.. T cell trafficking facilitated by high endothelial venules is required for tumor control after regulatory T cell depletion. Cancer Res 2012; 72:5473-82; PMID:22962270; http://dx.doi.org/ 10.1158/0008-5472.CAN-12-1912 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kobayashi M, Lee H, Schaffer L, Gilmartin TJ, Head SR, Takaishi S, Wang TC, Nakayama J, Fukuda M. A distinctive set of genes is upregulated during the inflammation-carcinoma sequence in mouse stomach infected by Helicobacter felis. J Histochem Cytochem 2007; 55:263-74; PMID:17101721; http://dx.doi.org/ 10.1369/jhc.6A7097.2006 [DOI] [PubMed] [Google Scholar]
- 82.Drayton DL, Liao S, Mounzer RH, Ruddle NH. Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol 2006; 7:344-53; PMID:16550197; http://dx.doi.org/ 10.1038/ni1330 [DOI] [PubMed] [Google Scholar]
- 83.Suzawa K, Kobayashi M, Sakai Y, Hoshino H, Watanabe M, Harada O, Ohtani H, Fukuda M, Nakayama J. Preferential induction of peripheral lymph node addressin on high endothelial venule-like vessels in the active phase of ulcerative colitis. Am J Gastroenterol 2007; 102:1499-509; PMID:17459027; http://dx.doi.org/ 10.1111/j.1572-0241.2007.01189.x [DOI] [PubMed] [Google Scholar]
- 84.Kobayashi M, Mitoma J, Nakamura N, Katsuyama T, Nakayama J, Fukuda M. Induction of peripheral lymph node addressin in human gastric mucosa infected by Helicobacter pylori. Proc Natl Acad Sci USA 2004; 101:17807-12; PMID:15591109; http://dx.doi.org/ 10.1073/pnas.0407503101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Goc J, Fridman WH, Sautes-Fridman C, Dieu-Nosjean MC. Characteristics of tertiary lymphoid structures in primary cancers. Oncoimmunology 2013; 2:e26836; PMID:24498556; http://dx.doi.org/ 10.4161/onci.26836 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Cipponi A, Mercier M, Seremet T, Baurain JF, Theate I, van den Oord J, Stas M, Boon T, Coulie PG, van Baren N. Neogenesis of lymphoid structures and antibody responses occur in human melanoma metastases. Cancer Res 2012; 72:3997-4007; PMID:22850419; http://dx.doi.org/ 10.1158/0008-5472.CAN-12-1377 [DOI] [PubMed] [Google Scholar]
- 87.Coronella JA, Spier C, Welch M, Trevor KT, Stopeck AT, Villar H, Hersh EM. Antigen-driven oligoclonal expansion of tumor-infiltrating B cells in infiltrating ductal carcinoma of the breast. J Immunol 2002; 169:1829-36; PMID:12165506; http://dx.doi.org/ 10.4049/jimmunol.169.4.1829 [DOI] [PubMed] [Google Scholar]
- 88.Bell D, Chomarat P, Broyles D, Netto G, Harb GM, Lebecque S, Valladeau J, Davoust J, Palucka KA, Banchereau J. In breast carcinoma tissue, immature dendritic cells reside within the tumor, whereas mature dendritic cells are located in peritumoral areas. J Exp Med 1999; 190:1417-26; PMID:10562317; http://dx.doi.org/ 10.1084/jem.190.10.1417 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Banner BF, Savas L, Baker S, Woda BA. Characterization of the inflammatory cell populations in normal colon and colonic carcinomas. Virchows Arch B Cell Pathol Incl Mol Pathol 1993; 64:213-20; PMID:8287117; http://dx.doi.org/ 10.1007/BF02915115 [DOI] [PubMed] [Google Scholar]
- 90.Goc J, Germain C, Vo-Bourgais TK, Lupo A, Klein C, Knockaert S, de Chaisemartin L, Ouakrim H, Becht E, Alifano M et al.. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res 2014; 74:705-15; PMID:24366885; http://dx.doi.org/ 10.1158/0008-5472.CAN-13-1342 [DOI] [PubMed] [Google Scholar]
- 91.Ogino S, Nosho K, Irahara N, Meyerhardt JA, Baba Y, Shima K, Glickman JN, Ferrone CR, Mino-Kenudson M, Tanaka N et al.. Lymphocytic reaction to colorectal cancer is associated with longer survival, independent of lymph node count, microsatellite instability, and CpG island methylator phenotype. Clin Cancer Res 2009; 15:6412-20; PMID:19825961; http://dx.doi.org/ 10.1158/1078-0432.CCR-09-1438 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Coppola D, Nebozhyn M, Khalil F, Dai H, Yeatman T, Loboda A, Mulé JJ. Unique ectopic lymph node-like structures present in human primary colorectal carcinoma are identified by immune gene array profiling. Am J Pathol 2011; 179:37-45; PMID:21703392; http://dx.doi.org/ 10.1016/j.ajpath.2011.03.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Nzula S, Going JJ, Stott DI. Antigen-driven clonal proliferation, somatic hypermutation, and selection of B lymphocytes infiltrating human ductal breast carcinomas. Cancer Res 2003; 63:3275-80 [PubMed] [Google Scholar]
- 94.Shields JD, Kourtis IC, Tomei AA, Roberts JM, Swartz MA. Induction of lymphoidlike stroma and immune escape by tumors that express the chemokine CCL21. Science 2010; 328:749-52; http://dx.doi.org/ 10.1126/science.1185837 [DOI] [PubMed] [Google Scholar]
- 95.Bento DC, Jones E, Junaid S, Tull J, Williams GT, Godkin A, Ager A, Gallimore A High endothelial venules are rare in colorectal cancers but accumulate in extra-tumoral areas with disease progression, OncoImmunology, 2014; 4(3):e974374; http://dx.doi.org/ 10.4161/2162402X.2014.974374 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Di Caro G, Bergomas F, Grizzi F, Doni A, Bianchi P, Malesci A, Laghi L, Allavena P, Mantovani A, Marchesi F. Occurrence of tertiary lymphoid tissue is associated with T-cell infiltration and predicts better prognosis in early-stage colorectal cancers. Clin Cancer Res 2014; 20:2147-58; PMID:24523438; http://dx.doi.org/ 10.1158/1078-0432.CCR-13-2590 [DOI] [PubMed] [Google Scholar]
- 97.Rosen SD. Endothelial ligands for L-selectin: from lymphocyte recirculation to allograft rejection. Am J Pathol 1999; 155:1013-20; PMID:10514381; http://dx.doi.org/ 10.1016/S0002-9440(10)65201-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Avram G, Sanchez-Sendra B, Martin JM, Terradez L, Ramos D, Monteagudo C. The density and type of MECA-79-positive high endothelial venules correlate with lymphocytic infiltration and tumour regression in primary cutaneous melanoma. Histopathology 2013; 63:852-61; PMID:24102908; http://dx.doi.org/ 10.1111/his.12235 [DOI] [PubMed] [Google Scholar]
- 99.Turunen JP, Majuri ML, Seppo A, Tiisala S, Paavonen T, Miyasaka M, Lemström K, Penttilä L, Renkonen O, Renkonen R. De novo expression of endothelial sialyl Lewisa and sialyl Lewis(x) during cardiac transplant rejection: Superior capacity of a tetravalent sialyl lewis(x) oligosaccharide in inhibiting L-selectin-dependent lymphocyte adhesion. J Exp Med 1995; 182:1133-42; PMID:7561686; http://dx.doi.org/ 10.1084/jem.182.4.1133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Baekkevold ES, Jahnsen FL, Johansen F-E, Bakke O, Gaudernack G, Brandtzaeg P, Haraldsen G. Culture characterization of differentiated high endothelial venule cells from human tonsils. Lab Invest 1999; 79:327-36; PMID:10092069 [PubMed] [Google Scholar]
- 101.Tamatani T, Kuida K, Watanabe T, Koike S, Miyasaka M. Molecular mechanisms underlying lymphocyte recirculation: III. Characterization of the LECAM-1 (L-selectin)-dependent adhesion pathway in rats. J Immunol 1993; 150:1735-45. [PubMed] [Google Scholar]
- 102.Derry CJ, Faveeuw C, Mordsley KR, Ager A. Novel chondroitin sulfate-modified ligands for L-selectin on lymph node high endothelial venules. Eur J Immunol 1999; 29:419-30; PMID:10064057; http://dx.doi.org/ 10.1002/(SICI)1521-4141(199902)29:02%3c419::AID-IMMU419%3e3.0.CO;2-A [DOI] [PubMed] [Google Scholar]
- 103.Hendriks HR, Duijvestijn AM, Kraal G. Rapid decrease in lymphocyte adherence to high endothelial venules in lymph nodes deprived of afferent lymphatic vessels. Eur J Immunol 1987; 17:1691-5; PMID:3500859; http://dx.doi.org/ 10.1002/eji.1830171203 [DOI] [PubMed] [Google Scholar]
- 104.Browning JL, Allaire N, Ngam-Ek A, Notidis E, Hunt J, Perrin S, Fava RA. Lymphotoxin-beta receptor signaling is required for the homeostatic control of HEV differentiation and function. Immunity 2005; 23:539-50; PMID:16286021; http://dx.doi.org/ 10.1016/j.immuni.2005.10.002 [DOI] [PubMed] [Google Scholar]
- 105.Webster B, Ekland EH, Agle LM, Chyou S, Ruggieri R, Lu TT. Regulation of lymph node vascular growth by dendritic cells. J Exp Med 2006; 203:1903-13; PMID:16831898; http://dx.doi.org/ 10.1084/jem.20052272 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Malhotra D, Fletcher AL, Astarita J, Lukacs-Kornek V, Tayalia P, Gonzalez SF, Elpek KG, Chang SK, Knoblich K, Hemler ME et al.. Transcriptional profiling of stroma from inflamed and resting lymph nodes defines immunological hallmarks. Nat Immunol 2012; 13:499-510; PMID:22466668; http://dx.doi.org/ 10.1038/ni.2262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Benezech C, White A, Mader E, Serre K, Parnell S, Pfeffer K, Ware CF, Anderson G, Caamaño JH. Ontogeny of stromal organizer cells during lymph node development. J Immunol 2010; 184:4521-30; PMID:20237296; http://dx.doi.org/ 10.4049/jimmunol.0903113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Onder L, Danuser R, Scandella E, Firner S, Chai Q, Hehlgans T, Stein JV, Ludewig B. Endothelial cell-specific lymphotoxin-beta receptor signaling is critical for lymph node and high endothelial venule formation. J Exp Med 2013; 210:465-73; PMID:23420877; http://dx.doi.org/ 10.1084/jem.20121462 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Moussion C, Girard JP. Dendritic cells control lymphocyte entry to lymph nodes through high endothelial venules. Nature 2011; 479:542-6; PMID:22080953; http://dx.doi.org/ 10.1038/nature10540 [DOI] [PubMed] [Google Scholar]
- 110.Wendland M, Willenzon S, Kocks J, Davalos-Misslitz AC, Hammerschmidt SI, Schumann K, Kremmer E, Sixt M, Hoffmeyer A, Pabst O et al.. Lymph node T cell homeostasis relies on steady state homing of dendritic cells. Immunity 2011; 35:945-57; PMID:22195748; http://dx.doi.org/ 10.1016/j.immuni.2011.10.017 [DOI] [PubMed] [Google Scholar]
- 111.Chyou S, Ekland EH, Carpenter AC, Tzeng TC, Tian S, Michaud M, Madri JA, Lu TT. Fibroblast-type reticular stromal cells regulate the lymph node vasculature. J Immunol 2008; 181:3887-96; PMID:18768843; http://dx.doi.org/ 10.4049/jimmunol.181.6.3887 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Lindquist RL, Shakhar G, Dudziak D, Wardemann H, Eisenreich T, Dustin ML, Nussenzweig MC. Visualizing dendritic cell networks in vivo. Nat Immunol 2004; 5:1243-50; PMID:15543150; http://dx.doi.org/ 10.1038/ni1139 [DOI] [PubMed] [Google Scholar]
- 113.Kissenpfennig A, Henri S, Dubois B, Laplace-Builhe C, Perrin P, Romani N, Tripp CH, Douillard P, Leserman L, Kaiserlian D. Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells. Immunity 2005; 22:643-54; PMID:15894281; http://dx.doi.org/ 10.1016/j.immuni.2005.04.004 [DOI] [PubMed] [Google Scholar]
- 114.Randolph GJ, Ochando J, Partida-Sanchez S. Migration of dendritic cell subsets and their precursors. Annu Rev Immunol 2008; 26:293-316; PMID:18045026; http://dx.doi.org/ 10.1146/annurev.immunol.26.021607.090254 [DOI] [PubMed] [Google Scholar]
- 115.Swiecki M, Gilfillan S, Vermi W, Wang Y, Colonna M. Plasmacytoid dendritic cell ablation impacts early interferon responses and antiviral NK and CD8(+) T cell accrual. Immunity 2010; 33:955-66; PMID:21130004; http://dx.doi.org/ 10.1016/j.immuni.2010.11.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Martinet L, Filleron T, Le Guellec S, Rochaix P, Garrido I, Girard JP. High endothelial venule blood vessels for tumor-infiltrating lymphocytes are associated with lymphotoxin beta-producing dendritic cells in human breast cancer. J Immunol 2013; 191:2001-8; PMID:23825314; http://dx.doi.org/ 10.4049/jimmunol.1300872 [DOI] [PubMed] [Google Scholar]
- 117.Sharma S, Stolina M, Luo J, Strieter RM, Burdick M, Zhu LX, Batra RK, Dubinett SM. Secondary lymphoid tissue chemokine mediates T cell-dependent antitumor responses in vivo. J Immunol 2000; 164:4558-63; PMID:10779757; http://dx.doi.org/ 10.4049/jimmunol.164.9.4558 [DOI] [PubMed] [Google Scholar]
- 118.Kirk CJ, Hartigan-O'Connor D, Mule JJ. The dynamics of the T-cell antitumor response: chemokine-secreting dendritic cells can prime tumor-reactive T cells extranodally. Cancer Res 2001; 61:8794-802. [PubMed] [Google Scholar]
- 119.Cuff CA, Sacca R, Ruddle NH. Differential induction of adhesion molecule and chemokine expression by LTalpha3 and LTalphabeta in inflammation elucidates potential mechanisms of mesenteric and peripheral lymph node development. J Immunol 1999; 162:5965-72. [PubMed] [Google Scholar]
- 120.Sun SC. The noncanonical NF-kappaB pathway. Immunol Rev 2012; 246:125-40; PMID:22435551; http://dx.doi.org/ 10.1111/j.1600-065X.2011.01088.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Guo F, Weih D, Meier E, Weih F Constitutive alternative NF-kappaB signaling promotes marginal zone B-cell development but disrupts the marginal sinus and induces HEV-like structures in the spleen. Blood 2007; 110(7):2381-9; http://dx.doi.org/ 10.1182/blood-2007-02-075143 [DOI] [PubMed] [Google Scholar]
- 122.Madge LA, May MJ. Classical NF-kappaB activation negatively regulates noncanonical NF-kappaB-dependent CXCL12 expression. J Biol Chem; 285:38069-77; PMID:20923761; http://dx.doi.org/ 10.1074/jbc.M110.147207 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Lovas A, Weidemann A, Albrecht D, Wiechert L, Weih D, Weih F. p100 Deficiency is insufficient for full activation of the alternative NF-kappaB pathway: TNF cooperates with p52-RelB in target gene transcription. PLoS One 2012; 7:e42741; PMID:22880094; http://dx.doi.org/ 10.1371/journal.pone.0042741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Pablos JL, Santiago B, Tsay D, Singer MS, Palao G, Galindo M, Rosen SD. A HEV-restricted sulfotransferase is expressed in rheumatoid arthritis synovium and is induced by lymphotoxin-alpha/beta and TNF-alpha in cultured endothelial cells. BMC Immunol 2005; 6:6; PMID:15752429; http://dx.doi.org/ 10.1186/1471-2172-6-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Cuff CA, Schwartz J, Bergman CM, Russell KS, Bender JR, Ruddle NH. Lymphotoxin alpha3 induces chemokines and adhesion molecules: insight into the role of LT alpha in inflammation and lymphoid organ development. J Immunol 1998; 161:6853-60; PMID:9862717 [PubMed] [Google Scholar]
- 126.Sikorski EE, Hallmann R, Berg EL, Butcher EC. The Peyer's patch high endothelial receptor for lymphocytes, the mucosal vascular addressin, is induced on a murine endothelial cell line by tumor necrosis factor-alpha and IL-1. J Immunol 1993; 151:5239-50; PMID:7693807 [PubMed] [Google Scholar]
- 127.Joyce JA and Fearon DT T cell exclusion, immune privilege, and the tumor microenvironment. Science 2015; 348:74-80; http://dx.doi.org/ 10.1126/science.aaa6204 [DOI] [PubMed] [Google Scholar]
- 128.Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, Royal RE, Topalian SL, Kammula US, Restifo NP et al.. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 2006; 314:126-9; PMID:16946036; http://dx.doi.org/ 10.1126/science.1129003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Fisher DT, Chen Q, Skitzki JJ, Muhitch JB, Zhou L, Appenheimer MM, Vardam TD, Weis EL, Passanese J, Wang WC et al.. IL-6 trans-signaling licenses mouse and human tumor microvascular gateways for trafficking of cytotoxic T cells. J Clin Invest 2011; 121:3846-59; PMID:21926464; http://dx.doi.org/ 10.1172/JCI44952 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Buckanovich RJ, Facciabene A, Kim S, Benencia F, Sasaroli D, Balint K, Katsaros D, O'Brien-Jenkins A, Gimotty PA, Coukos G. Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Nat Med 2008; 14:28-36; PMID:18157142; http://dx.doi.org/ 10.1038/nm1699 [DOI] [PubMed] [Google Scholar]
- 131.Dirkx AE, oude Egbrink MG, Castermans K, van der Schaft DW, Thijssen VL, Dings RP, Kwee L, Mayo KH, Wagstaff J, Bouma-ter Steege JC et al.. Anti-angiogenesis therapy can overcome endothelial cell anergy and promote leukocyte-endothelium interactions and infiltration in tumors. FASEB J 2006; 20:621-30; PMID:16581970; http://dx.doi.org/ 10.1096/fj.05-4493com [DOI] [PubMed] [Google Scholar]
- 132.Motz GT, Santoro SP, Wang LP, Garrabrant T,Lastra RR, Hagemann IS, Lal P, Feldman MD, Benencia F, Coukos.. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat Med 2014; 20(6):607-15; http:dx.doi.org/ 10.1038/nm.3541 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Calcinotto A, Grioni M, Jachetti E, Curnis F, Mondino A, Parmiani G, Corti A, Bellone M. Targeting TNF-alpha to neoangiogenic vessels enhances lymphocyte infiltration in tumors and increases the therapeutic potential of immunotherapy. J Immunol 2012; 188:2687-94; PMID:22323546; http://dx.doi.org/ 10.4049/jimmunol.1101877 [DOI] [PubMed] [Google Scholar]
- 134.Quezada SA, Peggs KS, Simpson TR, Shen Y, Littman DR, Allison JP. Limited tumor infiltration by activated T effector cells restricts the therapeutic activity of regulatory T cell depletion against established melanoma. J Exp Med 2008; 205:2125-38; PMID:18725522; http://dx.doi.org/ 10.1084/jem.20080099 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Shrimali RK, Yu Z, Theoret MR, Chinnasamy D, Restifo NP, Rosenberg SA. Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer. Cancer Res 2010; 70:6171-80; PMID:20631075; http://dx.doi.org/ 10.1158/0008-5472.CAN-10-0153 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Huang Y, Yuan J, Righi E, Kamoun WS, Ancukiewicz M, Nezivar J, Santosuosso M, Martin JD, Martin MR, Vianello F et al.. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci USA 2012; 109:17561-6; PMID:23045683; http://dx.doi.org/ 10.1073/pnas.1215397109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hamzah J, Jugold M, Kiessling F, Rigby P, Manzur M, Marti HH, Rabie T, Kaden S, Gröne HJ, Hämmerling GJ et al.. Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature 2008; 453:410-4; PMID:18418378; http://dx.doi.org/ 10.1038/nature06868 [DOI] [PubMed] [Google Scholar]
- 138.Berger M, Bergers G, Arnold B, Hammerling GJ, Ganss R. Regulator of G-protein signaling-5 induction in pericytes coincides with active vessel remodeling during neovascularization. Blood 2005; 105:1094-101; PMID:15459006; http://dx.doi.org/ 10.1182/blood-2004-06-2315 [DOI] [PubMed] [Google Scholar]
- 139.Ganss R, Ryschich E, Klar E, Arnold B, Hammerling GJ. Combination of T-cell therapy and trigger of inflammation induces remodeling of the vasculature and tumor eradication. Cancer Res 2002; 62:1462-70; PMID:11888921 [PubMed] [Google Scholar]
- 140.Finger EB, Puri KD, Alon R, Lawrence MB, von Andrian UH, Springer TA. Adhesion through L-selectin requires a threshold hydrodynamic shear. Nature 1996; 379:266-9; PMID:8538793; http://dx.doi.org/ 10.1038/379266a0 [DOI] [PubMed] [Google Scholar]
- 141.Cinamon G, Shinder V, Alon R. Shear forces promote lymphocyte migration across vascular endothelium bearing apical chemokines. Nat Immunol 2001; 2:515-22; PMID:11376338; http://dx.doi.org/ 10.1038/88710 [DOI] [PubMed] [Google Scholar]
- 142.Lanca T, Silva-Santos B. The split nature of tumor-infiltrating leukocytes: Implications for cancer surveillance and immunotherapy. Oncoimmunology 2012; 1:717-25; PMID:22934263; http://dx.doi.org/ 10.4161/onci.20068 [DOI] [PMC free article] [PubMed] [Google Scholar]