Abstract
In organ transplantation, blood borne cells and macromolecules (e.g. antibodies) of the host immune system are brought into direct contact with the endothelial cell (EC) lining of graft vessels. In this location, graft ECs play several roles in allograft rejection, including the initiation of rejection responses by presentation of alloantigen to circulating T cells; the development of inflammation and thrombosis; and as targets of injury and agents of repair.
Keywords: antigen presentation, inflammation, leukocyte, lymphocyte, antibody, complement
Introduction
In this review we will summarize the roles that graft endothelial cells (ECs) play in various stages of the rejection of allografts.
Endothelial Cells as Initiators of Rejection
T cells recognize allografts as non-self either by direct or indirect recognition of alloantigens (signal 1) (1, 2). Direct recognition is initially dominant, involving many more T cell clones, whereas indirect recognition may become more important at later times. The adaptive immune response is altered by experience and at least half of all circulating T cells in an adult human are memory cells (3, 4). Naïve and memory T cells have different requirements for activation (5–7) and patterns of recirculation (8). Memory cells are further subdivided into effector memory T cells that home to sites of inflammation and central memory T cells that recirculate through secondary lymphoid organs (8). Memory T cells generally have lesser requirements for costimulation (signal 2) and are less subject to modulation of their patterns of response by cytokines (signal 3) (9). They are also harder to immunosuppress, and to regulate (10). There is a high frequency of alloreactive memory T cells in humans that correlates with the outcome of clinical transplantation (11). As we will discuss shortly, memory T cells have a special relationship with ECs that will be described below.
Signals 1 and 2 are normally typically presented to naïve T cells by dendritic cells (DCs), referred to as “professional” antigen presenting cells (APCs). DCs increase their expression of MHC peptide complexes and co-stimulators (undergo “maturation”) in response to microbe-derived “pathogen-associated molecular patterns” (12) or signals released by injured cells (“damage-associated molecular patterns” or “alarmins”) (13). The latter may be more relevant in transplantation because ischemia/reperfusion (I/R) injury during organ transplantation releases alarmins, maturing DCs within the graft. Alarmins may also directly influence alloreactive memory T cell responses independently of DCs (14). In a rat kidney transplant model, graft-derived DCs (“passenger leukocytes") may be required to initiate graft rejection (15). In mice, allograft rejection by adoptively transferred naïve CD8+ T cells requires their activation within secondary lymphoid organs by graft-derived DCs (16), but rejection by adoptively transferred memory CD8+ T cells may occur in the absence of secondary lymphoid organs (17) and without a need for graft DCs (18). Passenger leukocyte depletion with antibody failed to prevent rejection of human kidneys although it is difficult to ascertain the completeness of depletion in this case. Nevertheless, these observations suggest that cells other than DCs may initiate allograft rejection perhaps through activation of alloreactive memory T cells (19). Several lines of evidence suggest that human (or mouse) ECs serve as this alternative initiator of rejection:
(1). In human allografts, vascular ECs basally express both class I and class II MHC molecules (20, 21). (see Table I). In culture, human ECs reduce class I and completely lose expression of class II molecules; cytokines, including TNF, IFN-α, IFN-β and IFN-γ, can restore class I MHC molecule expression, but only IFN-γ restores class II expression (22). Human ECs also process protein antigens to peptides that can be recognized by T cell clones (23). It is unknown if human ECs process antigens differently from other cell types, although this has been suggested for viral antigens (24). However, CTL produced by culturing human CD8+ T cells with allogeneic B lymphoblastoid cells readily recognize allogeneic ECs, implying that common peptides are generated (25, 26). Human ECs differ from mouse ECs in several respects relevant for antigen presentation (see Table II), and this must be considered when interpreting rodent transplant experiments.
Table I.
Category | Molecule | Comment | Rels |
---|---|---|---|
MHC | HLA-A,B,C | Constitutive; increased by IFN-α,β,γ TNF | (20, 21) |
TAP1,2 | Constitutive; increased by IFN-α,β,γ TNF | (105) | |
LMP2,7 | Constitutive; increased by IFN-α,β,γ TNF | (105) | |
HLA-DR, DP, DQ | Basal and induced further by IFN-γ | (106) | |
Invariant Chain | Basal and induced further by IFN-γ | (106) | |
Costimulators | LFA-3 (CD58) | Constitutive (Ig superfamily) | (107) |
PDL-1 | Constitutive; increased by IFN-γ (Ig superfamily) | (108) | |
PDL-2 | Constitutive; increased by IFN-γ (Ig superfamily) | (108) | |
ICOS-Ligand | Constitutive; increased by TNF (Ig superfamily) | (109) | |
4-1BB Ligand | Constitutive; increased by IFN-γ (TNF superfamily) | (7) | |
CD40 | Constitutive; increased by IFN-γ and TNF (TNFR superfamily) | (110) | |
Ox40-Ligand | Constitutive (TNF superfamily) | (7) | |
GITR-Ligand | Induced by TNF (TNF superfamily) | (108) | |
Cytokines | IL-1α | Constitutive; increased by IL-1, TNF | (111) |
IL-6 | Induced by IL-1, TNF | (112) | |
Other | Indolamine 2,3 dioxygenase | Induced by IFN αβγ | (113) |
Table II.
Molecule | Human | Mouse | Rel |
---|---|---|---|
Class II MHC | basal and inducible | absent | (114) |
LFA-3 | constitutive | absent | (114) |
CD80 | absent | constitutive and inducible | (114) |
P-selectin | constitutive and mobilizable | cytokine inducible | (114) |
IL-8 | inducible | absent | (114) |
Activation of CD4+ T cells | central and effector memory cells | regulatory cells only | (29, 32) |
Activation of CD8+ T cells | memory cells only | naïve and memory cells | (4, 31) |
(2), Cultured human ECs display functional co-stimulators that enhance T cell IL-2 production (27). While unable to express either CD80 or CD86, the principal costimulators required by naïve T cells, human ECs express other costimulators that are relevant for the activation of memory T cells. (see Table I).
(3). Human ECs cultured with resting allogeneic T cells in the absence of professional APCs stimulate IL-2 production by and proliferation of memory but not naïve T cells (4, 7, 27). This capacity distinguishes human ECs from most other cell types in peripheral tissues (28) as well as from DCs (see Table III). Once activated by ECs, memory T cells can subsequently respond to other cell types expressing relevant MHC molecules (28). Activation by ECs causes effector cytokine production from memory CD4+ T cells (29) and maturation of memory CD8+ T cells into cytolytic T lymphocytes (CTL), some of which are specific for EC targets (30). In mice, CD8+ but not CD4+ T cells proliferate to cultured allogeneic ECs (31, 32). CD4+ T cell responses to cultured mouse ECs may largely involve activation of regulatory T cell populations (33). Significantly for transplant rejection, adoptive transfer of resting human effector memory T cells to an immunodeficient mouse host in the absence of professional APCs can injure allogeneic human ECs within a human skin graft (29).
Table III.
Property | Endothelial Cell | Dendritic Cell |
---|---|---|
Phagocytosis | absent (in vitro only) | present (when immature) |
Pinocytosis | active | active |
MHC expression | basal; regulated by cytokines | regulated by maturation |
Cross presentation | probable | yes |
Costimulation | LFA-3 dominant; no B7 (CD80, CD86); | B7 (CD80, CD86) dominant |
Adhesion | selective for memory T cells | Cluster with naïve T cells |
Activation of T cells | selective for (effector) memory T cells | Preferential with naïve T cells |
Although human graft ECs are capable of directly activating T cells, ECs may play additional roles in the process of alloantigen recognition. First, EC may promote differentiation of monocytes into competent APCs (34) and may actually contribute to their differentiation into DCs (35). Second, host ECs may pick up graft antigens and present them to CD8+ T cells, displaying the property of "cross-presentation" sometimes proposed as unique to DCs (36).
The ability of ECs to stimulate alloreactive effector memory CD4+ T cells to secrete IFN-γ may have a special role in chronic allograft vasculopathy. This lesion, which is the major cause of late cardiac graft loss (and probably late renal graft loss as well) may depend on local IFN-γ production within the vessel wall (37). Histological analysis of these lesions in allograft coronary arteries suggests the persistence of graft ECs (38) in direct contact with subjacent host T cells (39). ECs may further contribute to chronic allograft vasculopathy by acting as a source of signals that induce bystander T cells to express inducible nitric oxide synthase, a signal that may help sustain alloresponses (40).
ECs as Participants in Rejection
Allograft rejection involves recruitment and activation of circulating leukocytes in response to co-ordinated changes in microvascular ECs (reviewed in (41)). Pro-inflammatory alterations in ECs is termed “activation” (41). Type I activation is mediated in response to ligands such as histamine or thrombin, and occurs independently of protein synthesis within minutes. Examples are NO or prostaglandin production or P-selectin translocation from storage organelles (Weibel-Palade bodies) to the cell surface. Type II activation is induced by cytokines such as TNF or IL-1, and involves transcription of new genes such as the adhesion molecules E-selectin, ICAM-1 and VCAM-1. Both responses may contribute to vascular leakiness, a cardinal feature of inflammation, although type II activation causes more sustained junctional changes and vascular leak. Key changes in activated venular ECs that contribute to leukocyte recruitment are expression of adhesion molecules that bind leukocytes and display of chemokines that activate the captured leukocytes. (see Table IV). The relevant EC adhesion molecules and activating chemokines responsible for leukocyte recruitment vary with particular leukocyte populations. Monocyte recruitment into rejecting human renal allografts may depend particularly upon EC selectins and the chemokines MCP-1 (CCL2) and RANTES (CCL5) that bind to chemokine receptors CCR1, CCR2 and CCR5 (42). Inflammatory T cells, especially those that have differentiated into effector memory T cells, more readily interact with VCAM-1 and with chemokines that bind to the receptor designated CXCR3, namely Mig (CXCL9) IP-10 (CXCL10) and ITAC (CXCL11) (43) and ENA-78 (CXCL5) (44). Both RANTES and IP-10 expression correlate with T cell infiltration into rejecting renal allografts (45, 46). In vitro, T cells, but not monocytes, require shear stress to initiate transmigration (43, 47). ECs may further contribute to leukocyte recruitment by forming a cup-like extension of the plasma membrane at sites of transmigration (48). This structure may form away from intercellular junctions allowing leukocytes to pass through the EC body.
Table IV.
Category | Molecules | Comment | Rels |
---|---|---|---|
Leukocyte Adhesion | E-selectin | Induced by TNF, IL-1 | (117) |
ICAM-1 | Constitutive; increased by TNF, IL-1 IFN-γ | (118) | |
VCAM-1 | Induced by TNF, IL-1, IL-4, IL-13 | (119, 120) | |
P-selectin | Constitutive: sequestered until translocated | (121) | |
ICAM-2 | Constitutive | (122) | |
Leukocyte Transmigration | PECAM-1 | Constitutive | (123) |
CD99 | Constitutive | (124) | |
Leukocyte Activation | Platelet activating factor | Synthesized in response to thrombin, histamine | (125) |
IL-8, Gro-α | Induced by TNF, IL-1 | (126, 127) | |
IP-10 | Induced by IFN-γ | (129) | |
ITAC | Induced by IFN-γ | (129) | |
Mig | Induced by IFN-γ | (130) | |
ENA-78 | Induced by IL-1 | (44) | |
MCP-1 | Induced by TNF, IL-1, IFN-γ IL-4, IL-13 | (131, 132) | |
Rantes | Induced by TNF plus IFN-γ | (133) | |
Eotaxin 3 | Induced by IL-4 | (134) | |
Fractalkine | Induced by TNF, IL-1, IFN-γ | (135) | |
Vasoactive | Cyclo-oxygenase 2 | Induced by TNF, IL-1 | (136) |
Endothelial NOS | Constitutive; decreased by TNF | (137) | |
Procoagulant | Tissue factor | Induced by TNF, IL-1, CD40L | (138, 139) |
Heparan sulfate | Shed by Ab + C' | (84) | |
Thrombomodulin | Constitutive; decreased by TNF, CD40L | (139, 140) |
EC functions related to antigen presentation and leukocyte recruitment may be linked. Antigen-activated T cells are a major source of cytokines that mediate type II EC activation, including TNF and lymphotoxin (LT, also called LTα or TNFβ). Antigen-activated T cells may also activate ECs through a contact-dependent pathway utilizing CD154 (49). EC antigen presentation to effector memory T cells may trigger T cell diapedesis involving the cytokine-inducible transmembrane chemokine fractalkine (CX3CL1) and its receptor (CX3CR1) on effector memory T cells (50).
ECs may also contribute to graft vessel thrombosis that can exacerbate graft injury (51). Resting ECs keep blood fluid (41) by preventing platelet activation and displaying tissue factor pathway inhibitors and inhibitors of thrombin, such as heparan sulfates (51). If thrombin is generated, resting ECs capture the active enzyme on thrombomodulin and use it to activate protein C, which degrades clotting factors. ECs also produce plasminogen activators that lead to lysis of fibrin thrombi. Type II activated EC synthesize express pro-coagulant molecules. At the same time EC injury (see below) abrogates EC anti-coagulant properties. Although thrombosis may be observed in acute cell-mediated rejection (ACR), it is more characteristically associated with antibody-mediated rejection (AMR) (52).
ECs as Targets of Rejection
Allograft ECs are a primary target of rejection. Cells and molecules that contribute to rejection through their actions on ECs include:
(1). Cytolytic T Lymphocytes (CTL)
Direct recognition of a specific complex of peptide and non-self class I MHC molecules on graft EC by host CTL is a major mechanism of ACR in immunosuppressed patients, leading to graft cell apoptosis (53, 54). The presence of CTL and CTL-specific mRNAs in biopsies correlates with ACR (55). CTL generated from CD8+ memory T cells by co-culture with allogeneic ECs may only kill ECs (30), and EC-selective CTL have been recovered from rejecting grafts (56). EC lysis by CTL involves either T cell ligands that engage death receptors (DRs, see below) on ECs or granule exocytosis of effector molecules (57). Both pathways require cell-cell contact and are initiated by recognition of non-self class I MHC/peptide complexes. Granule exocytosis deposits perforin and granzyme B from the CTL on the target cell surface. Perforin facilitates entry of granzyme B, a serine protease that cleaves and activates pro-capsase 3, an effector of apoptosis. The activation and activity of caspase 3 may be held in check by X-linked Inhibitor of Apoptosis Protein (x-IAP). Granzyme B also cleaves and activates pro-caspase 8, which may cleave and activate pro-caspase 3, and may also cleave and activate the cytosolic protein Bid. Activated Bid interacts with Bax, a pro-apoptotic member of the Bcl-2 family, inducing Bax to dimerize in the outer mitochondrial membrane and initiate release of proteins that promote cell death, including cytochrome c, Apoptosis Inducing Factor (AIF) and Second Mediator of Apoptotic Cell death (SMAC, also known as Diablo). This Bax-mediated release reaction is antagonized by anti-apoptotic members of the Bcl-2 family, including Bcl-2 itself. SMAC/Diablo prevents x-IAP from inhibiting caspase 3 activation. Cytochrome c interacts with Apoptotic Protein Activating Factor-1 (APAF-1) to activate procaspase 9, an alternative activator of caspase-3. AIF leads to cell death independent of caspases. CTL-mediated killing of cultured human ECs depends granule exocytosis and can be inhibited by overexpression of Bcl-2, implying a role for mitochondrial release of SMAC-Diablo in this cell type (58).
(2). Natural Killer (NK) cells
NK cells infiltrate grafts shortly after transplantation (59) and typically recognize targets by a combination of activating and inhibitory receptors (60). Activating receptors recognize ligands such as the Fc portions of antibodies, enabling antibody-dependent cell-mediated cytotoxicity (ADCC), or molecules such as MHC class I-related antigen-A-1 (MICA-1). ADCC may be a major component of acute AMR in which ECs are primary targets of alloantibody (see below). NK cell inhibitory receptors often recognize specific allelic forms of class I MHC molecules complexed to self-peptides but cannot recognize allogeneic cells that lack these molecules (“absent self”). In the absence of inhibitory signals, host NK cells will recognize and kill allogeneic ECs (61). NK cell-mediated EC killing has been invoked as a primary pathway of hyperacute and accelerated acute rejection of xenografts (62) and has been proposed to play a role in chronic allograft vasculopathy (63). A role for NK cells in ACR of allografts is less clear. NK cell killing utilizes the same mechanisms as CTL, namely engaging DR ligands and granule exocytosis of effector molecules.
(3). Macrophages
Monocytes infiltrate kidneys undergoing ACR or AMR and differentiate into macrophages, which contribute to rejection (64). Macrophages have been associated with capillary regression during development by inducing EC apoptosis (65); similar events may occur during rejection. EC injury by macrophages may involve multiple effector mechanisms including elaboration of TNF, TRAIL, reactive oxygen species (ROS) and NO. The expression of these effectors can be enhanced by IFN-γ-mediated macrophage activation (66). Macrophages may undergo an alternative pathway of activation, differentiating into “M2 cells” that are distinct from IFN-γ-activated “M1 cells”. M2 cells are associated with tissue repair but may also promote fibrosis in late graft loss (66).
(4). Neutrophils
Neutrophils are usually the first cell type recruited into inflammatory responses and their paucity in the infiltrates associated with ACR is striking. It is possible that they are transiently recruited at the very earliest stages of the acute rejection process and have disappeared by the time biopsies are taken. This idea is supported by the observation that E-selectin expression by ECs in human cardiac biopsies, which is important for neutrophil recruitment, is transient and predictive of imminent rejection and absent during rejection responses (67, 68). Neutrophils are more common in AMR or in non-immune injury to ECs (see below) and reduction of neutrophil infiltrates in the peri-operative period reduces subsequent T cell-mediated rejection of mouse cardiac allografts (69). Injury of EC depends on neutrophil activation and neutrophils may be activated by cytokines, chemokines, antibodies and complement products. Activated neutrophils may release both ROS and degradative enzymes that contribute to killing of microbes; these effector molecules are also capable of injuring or killing ECs.
(5). Death receptor ligands
DRs are members of the TNF receptor superfamily that contain an intracellular protein-protein interaction domain called a death domain (DD) (70, 71). DRs expressed on ECs include TNFR1 (also known as DR1), Fas (also known as DR2), DR3, TRAIL-R1 (also known as DR4) and TRAIL-R2 (also known as DR5). TNFR1 binds both TNF and lymphotoxin, made primarily by activated leukocytes, whereas DR3 binds TNF-like molecule 1A (TL1A), which is made by ECs and activated leukocytes (72). TNF, LT and TL1A are found in rejecting allografts (73). Fas binds FasL, whereas the two TRAIL receptors bind TRAIL. These membrane-associated ligands are expressed on activated effector cells, such as CD4+ effector T cells, CD8+ CTL, NK cells or macrophages, and can mediate cell-cell contact-dependent killing. In most cells, DR-initiated killing involves activation of DR-linked pro-caspase 8. This has been shown to occur in cultured human ECs and is enhanced by IFN-γ treatment which increases pro-caspase 8 expression (74). IFN-γ also allows TNFR1 to initiate a caspase-independent death pathway in ECs that is initiated by the lysosomal enzyme cathepsin B and involves Bid-independent release of AIF from mitochondria (75).
In the kidney TNFR1 is expressed basally on glomerular and vascular ECs and is downregulated during ACR or ischemia/reperfusion (I/R) injury (76). Transcripts and protein for DR3 are induced in vascular ECs (and renal tubular epithelial cells) in human kidney allografts undergoing either ACR or I/R injury (77). TNFR1 signaling appears responsible for TNFR1 downregulation (78). Signaling through TNFR2, which is not a DR and which is minimally expressed in normal kidney, upregulates TNFR2 expression on ECs and on tubular epthelial cells (78). Whereas TNFR1 signals lead to inflammation and apoptosis, TNFR2 signals promote proliferation and repair (78). Signals that induce DR3 are unknown and signals through DR3 are initiated by binding of TL1A (72). TL1A protein is synthesized by ECs in normal human kidney and synthesis is increased in rejection (73). In organ cultures of normal human kidney and wild-type mice, TL1A activates NF-κB, induces caspase-3 and apoptosis, and upregulates TNFR2 in TEC. In DR3 null mice, TL1A does not induce NF-κB or caspase-3 activation but still upregulates TNFR2, suggesting there is a second receptor for TL1A (73).
(6). Antibodies and complement
AMR is characterized by monocyte margination against EC and by capillaries containing thrombi and fibrinoid arterial necrosis (52). Antibody and complement components are present on the EC surface, and C4d deposition is now taken as evidence of AMR (79) but the lectin pathway of complement activation, which can be triggered by ischemia, could also lead to endothelial C4d deposits. The diagnosis of AMR is supported by detection of circulating antibodies that recognize graft ECs (54). Such antibodies most often react with HLA-A, B, C and DR antigens, but circulating anti-EC antibodies that react with non-HLA alloantigens have been detected in patients (80). Some non-HLA alloantigens are shared by ECs and monocytes but not lymphocytes (E-M antigens), and some are only found on ECs (E antigens). MICA on renal endothelium is also a potential target for AMR. AMR to ECs may involve complement and complement-induced inflammation and/or NK cells, which recognize bound antibodies via Fc receptor and mediate ADCC (81).
In addition to killing ECs via complement activation or targeting NK cells for ADCC, alloantibodies may activate ECs. Anti-MHC antibodies activate ECs to degranulate Weibel-Palade bodies, leading to release of IL-8 and display of P-selectin. In culture and in humans skin grafts placed on immunodeficient mice, F(ab)’ fragments of an HLA-A,B,C antibody can exert such effects despite their inability to engage Fc receptors or active complement (82). Anti-class I antibodies may also be mitogenic for ECs, although the role of this response to allograft rejection is unclear. Activation of the complement system may also affect ECs independent of antibody, causing perturbation in EC barrier function retraction of EC plasma membrane from the underlying substrate, release of preformed von Willebrand factor and P-selectin from Weibel-Palade bodies to the cell surface, and production of cytokines including IL-α, IL-8 and MCP-1 (83). The combination of C5a and antibody binding can cause ECs to shed anti-coagulant heparan sulfates (84). ECs express a variety of complement receptors and complement regulatory proteins that may modulate these responses (85, 86). There is little evidence to date that any of these are actively regulated in allograft rejection. Deposition of complement membrane attack complex on ECs usually does not, by itself, kill ECs but may induce endothelial microparticle (EMP) formation and thrombosis (see below) (87).
(7). Non-immune injuries
Hemodynamic factors combined with mediators such as cytokines have profound effects on EC injury. Disturbances in flow patterns in atherosclerosis, post-surgical, intimal hyperplasia and I/R injury or infection (e.g., with cytomegalovirus) can influence EC-T cell interactions through expression of alarmins and adhesion molecules promoting rejection (88–90). These responses may explain why renal allografts from living-related donors which are healthy, generally have a higher graft survival than cadaver donor transplants, involving injured kidneys.
(8). Thrombosis
Injury to ECs from whatever cause may lead to graft vessel thrombosis. Circulating procoagulant EMPs, which are shed plasma membrane components, are markers of vascular damage produced by EC apoptosis or deposition of the complement membrane attack complex, but are also generated in response to thrombin, collagen, and/or shear stress (91, 92). EMPs impair EC function in vitro, diminishing acetylcholine-induced vasorelaxation and NO production, and increasing superoxide production (93). At the same time, cytokine-activated EC lose some anti-coagulant properties, such as expression of thrombomodulin and acquire pro-coagulant properties such as expression of tissue factor and fibrinogen-like protein 2 (41). As noted earlier, EC injury and type II-activation may thus combine to promote thrombosis.
ECs Resistance to Injury and Repair
ECs can acquire resistance to injury by up-regulating a number of cytoprotective genes such as hemoxygenase-1 (HO-1), A20, Bcl-2, and Bcl-xL, which protect cells from cytokine-mediated apoptosis (94). Many of these gene products are regulated by NF-κB and are induced by TNF. Induction of protective genes may contribute to resistance against AMR, a phenomenon called “accommodation.” (94). Accommodated grafts function without rejection despite the presence of recipient-specific antibodies. Although many genes contributing to resistance to injury are regulated by NF-κB, activation of the serine/threonine kinase Akt via the PI3K signaling pathway and/or of ERK-1,2 in response to TNF (95) or growth factors, (96–98) can also contribute to cytoprotection. IL-11 and IL-6, which signal via STAT3, have also been shown to render EC resistant to immune-mediated injury (99, 100).
Graft vessels containing injured ECs may undergo repair. For injuries of limited extent, neighboring ECs may simply spread to cover the defect and then divide. In cases of more extensive vessel injury (e.g., vascular rejection), vessels of transplant recipients may display endothelial chimerism through replacement of damaged donor endothelium by host-derived precursors (101). The percentage of recipient ECs in the peritubular capillaries correlates with the type of renal transplant rejection, being predominantly found in vascular rejection. It is likely that ECs are lost from the vessel as a result of rejection and are replaced by circulating endothelial progenitor cells (EPCs) that have the capacity to differentiate into mature ECs. The markers that define circulating EPCs are controversial and true EPCs must be distinguished from hematopoietic cells that stimulate angiogenesis but do not give rise to long-lived ECs (102). The signals that recruit EPCs are also unknown, but both VEGF (103) and SDF-1 (104) are thought to be important. Both may be detected in allografts.
Conclusions
We have reviewed evidence that graft ECs are critical players in all stages of the host response to allografts, contributing to the initiation and effector phases of rejection. We have also noted that ECs may resist injury or contribute to repair. Despite these central roles performed by ECs, it is striking that none of the existing therapeutic strategies to improve the outcome of transplantation have focused on improving EC health or function. Development of such strategies may not only protect the graft, but may also improve the cardiovascular health of the host, a major complication of current transplantation medicine.
Acknowledgments
Supported by grants from National Institute for Health Research and British Heart Foundation to JRB and grants from the NIH (HL036003, HL051014, HL062188 and HL070295) to JSP.
Abbreviations
- ACR
acute cell-mediated rejection
- ADCC
antibody-dependent cell-mediated cytotoxicity
- AIF
apoptosis inducing factor
- AMR
antibody-mediated rejection
- APC
antigen presenting cell(s)
- APAF-1
apoptotic protease activating factor-1
- CTL
cytolytic T cell(s)
- DC
dendritic cell(s)
- DD
death domain
- DR
death receptor
- EC
endothelial cell(s)
- EMP
EC-derived microparticles
- EPC
endothelial progenitor cell(s)
- HO
hemoxygenase
- ICAM-1
intercellular adhesion molecule-1
- IL
interleukin
- MCP-1
monocyte chemoattractant protein-I
- MICA-1
MHC class I-related antigen A-1
- NK
natural killer
- ROS
reactive oxygen species
- SMAC
second mediator of apoptotic cell death
- TL1A
TNF-like molecule 1A
- TNF
tumor necrosis factor
- TNFR
TNF receptor
- TRAIL
TNF-related apoptosis-inducing ligand
- VCAM-1
vascular cell adhesion molecule-1
- x-IAP
X-linked inhibitor of apoptosis protein
Footnotes
The authors have no conflicts of interest.
References
- 1.Game DS, Lechler RI. Pathways of allorecognition: implications for transplantation tolerance. Transpl Immunol. 2002;10:101. doi: 10.1016/s0966-3274(02)00055-2. [DOI] [PubMed] [Google Scholar]
- 2.Felix NJ, Allen PM. Specificity of T-cell alloreactivity. Nat Rev Immunol. 2007;7:942. doi: 10.1038/nri2200. [DOI] [PubMed] [Google Scholar]
- 3.Lombardi G, Sidhu S, Daly M, Batchelor JR, Makgoba W, Lechler RI. Are primary alloresponses truly primary? Int Immunol. 1990;2:9. doi: 10.1093/intimm/2.1.9. [DOI] [PubMed] [Google Scholar]
- 4.Epperson DE, Pober JS. Antigen-presenting function of human endothelial cells. Direct activation of resting CD8 T cells. J Immunol. 1994;153:5402. [PubMed] [Google Scholar]
- 5.de Jong R, Brouwer M, Miedema F, van Lier RA. Human CD8+ T lymphocytes can be divided into CD45RA+ and CD45RO+ cells with different requirements for activation and differentiation. J Immunol. 1991;146:2088. [PubMed] [Google Scholar]
- 6.de Jong R, Brouwer M, Kuiper HM, Hooibrink B, Miedema F, van Lier RA. Maturation- and differentiation-dependent responsiveness of human CD4+ T helper subsets. J Immunol. 1992;149:2795. [PubMed] [Google Scholar]
- 7.Shiao SL, McNiff JM, Pober JS. Memory T cells and their costimulators in human allograft injury. J Immunol. 2005;175:4886. doi: 10.4049/jimmunol.175.8.4886. [DOI] [PubMed] [Google Scholar]
- 8.Lanzavecchia A, Sallusto F. Understanding the generation and function of memory T cell subsets. Curr Opin Immunol. 2005;17:326. doi: 10.1016/j.coi.2005.04.010. [DOI] [PubMed] [Google Scholar]
- 9.London CA, Lodge MP, Abbas AK. Functional responses and costimulator dependence of memory CD4+ T cells. J Immunol. 2000;164:265. doi: 10.4049/jimmunol.164.1.265. [DOI] [PubMed] [Google Scholar]
- 10.Yang J, Brook MO, Carvalho-Gaspar M, et al. Allograft rejection mediated by memory T cells is resistant to regulation. Proc Natl Acad Sci U S A. 2007;104:19954. doi: 10.1073/pnas.0704397104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Heeger PS, Greenspan NS, Kuhlenschmidt S, et al. Pretransplant frequency of donor-specific, IFN-gamma-producing lymphocytes is a manifestation of immunologic memory and correlates with the risk of posttransplant rejection episodes. J Immunol. 1999;163:2267. [PubMed] [Google Scholar]
- 12.Kabelitz D, Medzhitov R. Innate immunity--cross-talk with adaptive immunity through pattern recognition receptors and cytokines. Curr Opin Immunol. 2007;19:1. doi: 10.1016/j.coi.2006.11.018. [DOI] [PubMed] [Google Scholar]
- 13.Oppenheim JJ, Tewary P, de la Rosa G, Yang D. Alarmins initiate host defense. Adv Exp Med Biol. 2007;601:185. doi: 10.1007/978-0-387-72005-0_19. [DOI] [PubMed] [Google Scholar]
- 14.Rao DA, Tracey KJ, Pober JS. IL-1alpha and IL-1beta are endogenous mediators linking cell injury to the adaptive alloimmune response. J Immunol. 2007;179:6536. doi: 10.4049/jimmunol.179.10.6536. [DOI] [PubMed] [Google Scholar]
- 15.Lechler RI, Batchelor JR. Restoration of immunogenicity to passenger cell-depleted kidney allografts by the addition of donor strain dendritic cells. J Exp Med. 1982;155:31. doi: 10.1084/jem.155.1.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lakkis FG, Arakelov A, Konieczny BT, Inoue Y. Immunologic 'ignorance' of vascularized organ transplants in the absence of secondary lymphoid tissue. Nat Med. 2000;6:686. doi: 10.1038/76267. [DOI] [PubMed] [Google Scholar]
- 17.Chalasani G, Dai Z, Konieczny BT, Baddoura FK, Lakkis FG. Recall and propagation of allospecific memory T cells independent of secondary lymphoid organs. Proc Natl Acad Sci U S A. 2002;99:6175. doi: 10.1073/pnas.092596999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kreisel D, Krupnick AS, Gelman AE, et al. Non-hematopoietic allograft cells directly activate CD8+ T cells and trigger acute rejection: an alternative mechanism of allorecognition. Nat Med. 2002;8:233. doi: 10.1038/nm0302-233. [DOI] [PubMed] [Google Scholar]
- 19.Brewer Y, Palmer A, Taube D, et al. Effect of graft perfusion with two CD45 monoclonal antibodies on incidence of kidney allograft rejection. Lancet. 1989;2:935. doi: 10.1016/s0140-6736(89)90951-3. [DOI] [PubMed] [Google Scholar]
- 20.Hart DN, Fuggle SV, Williams KA, Fabre JW, Ting A, Morris PJ. Localization of HLA-ABC and DR antigens in human kidney. Transplantation. 1981;31:428. doi: 10.1097/00007890-198106000-00005. [DOI] [PubMed] [Google Scholar]
- 21.Hancock WW, Kraft N, Atkins RC. The immunohistochemical demonstration of major histocompatibility antigens in the human kidney using monoclonal antibodies. Pathology. 1982;14:409. doi: 10.3109/00313028209092120. [DOI] [PubMed] [Google Scholar]
- 22.Lapierre LA, Fiers W, Pober JS. Three distinct classes of regulatory cytokines control endothelial cell MHC antigen expression. Interactions with immune gamma interferon differentiate the effects of tumor necrosis factor and lymphotoxin from those of leukocyte alpha and fibroblast beta interferons. J Exp Med. 1988;167:794. doi: 10.1084/jem.167.3.794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Savage CO, Brooks CJ, Harcourt GC, et al. Human vascular endothelial cells process and present autoantigen to human T cell lines. Int Immunol. 1995;7:471. doi: 10.1093/intimm/7.3.471. [DOI] [PubMed] [Google Scholar]
- 24.Kummer M, Lev A, Reiter Y, Biedermann BC. Vascular endothelial cells have impaired capacity to present immunodominant, antigenic peptides: a mechanism of cell type-specific immune escape. J Immunol. 2005;174:1947. doi: 10.4049/jimmunol.174.4.1947. [DOI] [PubMed] [Google Scholar]
- 25.Biedermann BC, Pober JS. Human endothelial cells induce and regulate cytolytic T cell differentiation. J Immunol. 1998;161:4679. [PubMed] [Google Scholar]
- 26.Dengler TJ, Pober JS. Human vascular endothelial cells stimulate memory but not naive CD8+ T cells to differentiate into CTL retaining an early activation phenotype. J Immunol. 2000;164:5146. doi: 10.4049/jimmunol.164.10.5146. [DOI] [PubMed] [Google Scholar]
- 27.Hughes CC, Savage CO, Pober JS. Endothelial cells augment T cell interleukin 2 production by a contact-dependent mechanism involving CD2/LFA-3 interaction. J Exp Med. 1990;171:1453. doi: 10.1084/jem.171.5.1453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Pober JS, Collins T, Gimbrone MA, Jr, et al. Lymphocytes recognize human vascular endothelial and dermal fibroblast Ia antigens induced by recombinant immune interferon. Nature. 1983;305:726. doi: 10.1038/305726a0. [DOI] [PubMed] [Google Scholar]
- 29.Shiao SL, Kirkiles-Smith NC, Shepherd BR, McNiff JM, Carr EJ, Pober JS. Human effector memory CD4+ T cells directly recognize allogeneic endothelial cells in vitro and in vivo. J Immunol. 2007;179:4397. doi: 10.4049/jimmunol.179.7.4397. [DOI] [PubMed] [Google Scholar]
- 30.Biedermann BC, Pober JS. Human endothelial cells induce and regulate cytolytic T cell differentiation. J.Immunol. 1998;161:4679. [PubMed] [Google Scholar]
- 31.Kreisel D, Krupnick AS, Balsara KR, et al. Mouse vascular endothelium activates CD8+ T lymphocytes in a B7-dependent fashion. J Immunol. 2002;169:6154. doi: 10.4049/jimmunol.169.11.6154. [DOI] [PubMed] [Google Scholar]
- 32.Kreisel D, Krasinskas AM, Krupnick AS, et al. Vascular endothelium does not activate CD4+ direct allorecognition in graft rejection. J Immunol. 2004;173:3027. doi: 10.4049/jimmunol.173.5.3027. [DOI] [PubMed] [Google Scholar]
- 33.Krupnick AS, Gelman AE, Barchet W, et al. Murine vascular endothelium activates and induces the generation of allogeneic CD4+25+Foxp3+ regulatory T cells. J Immunol. 2005;175:6265. doi: 10.4049/jimmunol.175.10.6265. [DOI] [PubMed] [Google Scholar]
- 34.Denton MD, Geehan CS, Alexander SI, Sayegh MH, Briscoe DM. Endothelial cells modify the costimulatory capacity of transmigrating leukocytes and promote CD28-mediated CD4(+) T cell alloactivation. J Exp Med. 1999;190:555. doi: 10.1084/jem.190.4.555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Randolph GJ, Beaulieu S, Lebecque S, Steinman RM, Muller WA. Differentiation of monocytes into dendritic cells in a model of transendothelial trafficking. Science. 1998;282:480. doi: 10.1126/science.282.5388.480. [DOI] [PubMed] [Google Scholar]
- 36.Bagai R, Valujskikh A, Canaday DH, et al. Mouse endothelial cells cross-present lymphocyte-derived antigen on class I MHC via a TAP1- and proteasome-dependent pathway. J Immunol. 2005;174:7711. doi: 10.4049/jimmunol.174.12.7711. [DOI] [PubMed] [Google Scholar]
- 37.Tellides G, Pober JS. Interferon-gamma axis in graft arteriosclerosis. Circ Res. 2007;100:622. doi: 10.1161/01.RES.0000258861.72279.29. [DOI] [PubMed] [Google Scholar]
- 38.Minami E, Laflamme MA, Saffitz JE, Murry CE. Extracardiac progenitor cells repopulate most major cell types in the transplanted human heart. Circulation. 2005;112:2951. doi: 10.1161/CIRCULATIONAHA.105.576017. [DOI] [PubMed] [Google Scholar]
- 39.Salomon RN, Hughes CC, Schoen FJ, Payne DD, Pober JS, Libby P. Human coronary transplantation-associated arteriosclerosis. Evidence for a chronic immune reaction to activated graft endothelial cells. Am J Pathol. 1991;138:791. [PMC free article] [PubMed] [Google Scholar]
- 40.Choy JC, Wang Y, Tellides G, Pober JS. Induction of inducible NO synthase in bystander human T cells increases allogeneic responses in the vasculature. Proc Natl Acad Sci U S A. 2007;104:1313. doi: 10.1073/pnas.0607731104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pober JS, Sessa WC. Evolving functions of endothelial cells in inflammation. Nat Rev Immunol. 2007;7:803. doi: 10.1038/nri2171. [DOI] [PubMed] [Google Scholar]
- 42.Grandaliano G, Gesualdo L, Ranieri E, Monno R, Stallone G, Schena FP. Monocyte chemotactic peptide-1 expression and monocyte infiltration in acute renal transplant rejection. Transplantation. 1997;63:414. doi: 10.1097/00007890-199702150-00015. [DOI] [PubMed] [Google Scholar]
- 43.Manes TD, Pober JS, Kluger MS. Endothelial cell-T lymphocyte interactions: IP-10 stimulates rapid transendothelial migration of human effort but not central memory CD4+ T cells. Requirements for shear stress and adhesion molecules. Transplantation. 2006;82:S9. doi: 10.1097/01.tp.0000231356.57576.82. [DOI] [PubMed] [Google Scholar]
- 44.Imaizumi T, Albertine KH, Jicha DL, McIntyre TM, Prescott SM, Zimmerman GA. Human endothelial cells synthesize ENA-78: relationship to IL-8 and to signaling of PMN adhesion. Am J Respir Cell Mol Biol. 1997;17:181. doi: 10.1165/ajrcmb.17.2.2818. [DOI] [PubMed] [Google Scholar]
- 45.Panzer U, Reinking RR, Steinmetz OM, et al. CXCR3 and CCR5 positive T-cell recruitment in acute human renal allograft rejection. Transplantation. 2004;78:1341. doi: 10.1097/01.tp.0000140483.59664.64. [DOI] [PubMed] [Google Scholar]
- 46.Hoffmann U, Segerer S, Rummele P, et al. Expression of the chemokine receptor CXCR3 in human renal allografts--a prospective study. Nephrol Dial Transplant. 2006;21:1373. doi: 10.1093/ndt/gfk075. [DOI] [PubMed] [Google Scholar]
- 47.Cinamon G, Shinder V, Alon R. Shear forces promote lymphocyte migration across vascular endothelium bearing apical chemokines. Nat Immunol. 2001;2:515. doi: 10.1038/88710. [DOI] [PubMed] [Google Scholar]
- 48.Carman CV, Springer TA. A transmigratory cup in leukocyte diapedesis both through individual vascular endothelial cells and between them. J Cell Biol. 2004;167:377. doi: 10.1083/jcb.200404129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Karmann K, Hughes CC, Schechner J, Fanslow WC, Pober JS. CD40 on human endothelial cells: inducibility by cytokines and functional regulation of adhesion molecule expression. Proc Natl Acad Sci U S A. 1995;92:4342. doi: 10.1073/pnas.92.10.4342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Manes T, Pober JS. Antigen presentation by human microvascular endothelial cells triggers ICAM-1-independent transndothelial protrusion by and factalkine-dependent transendothelial migration of effector memory CD4+ T cells. J Immunol. 2008 doi: 10.4049/jimmunol.180.12.8386. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Manes TD, Pober JS, Kluger MS. Endothelial cell-T lymphocyte interactions: IP[corrected]-10 stimulates rapid transendothelial migration of human effort but not central memory CD4+ T cells. Requirements for shear stress and adhesion molecules. Transplantation. 2006;82:S9. doi: 10.1097/01.tp.0000231356.57576.82. [DOI] [PubMed] [Google Scholar]
- 52.Shimizu A, Colvin RB. Pathological features of antibody-mediated rejection. Curr Drug Targets Cardiovasc Haematol Disord. 2005;5:199. doi: 10.2174/1568006054064744. [DOI] [PubMed] [Google Scholar]
- 53.Meehan SM, McCluskey RT, Pascual M, et al. Cytotoxicity and apoptosis in human renal allografts: identification, distribution, and quantitation of cells with a cytotoxic granule protein GMP-17 (TIA-1) and cells with fragmented nuclear DNA. Lab Invest. 1997;76:639. [PubMed] [Google Scholar]
- 54.Colvin RB, Cohen AH, Saiontz C, et al. Evaluation of pathologic criteria for acute renal allograft rejection: reproducibility, sensitivity, and clinical correlation. J.Am.Soc.Nephrol. 1997;8:1930. doi: 10.1681/ASN.V8121930. [DOI] [PubMed] [Google Scholar]
- 55.Strehlau J, Pavlakis M, Lipman M, et al. Quantitative detection of immune activation transcripts as a diagnostic tool in kidney transplantation. Proc.Natl.Acad.Sci.U.S.A. 1997;94:695. doi: 10.1073/pnas.94.2.695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Jutte NH, Knoop CJ, Heijse P, et al. Human heart endothelial-cell-restricted allorecognition. Transplantation. 1996;62:403. doi: 10.1097/00007890-199608150-00017. [DOI] [PubMed] [Google Scholar]
- 57.Bleackley RC. A molecular view of cytotoxicT lymphocyte induced killing. Biochem Cell Biol. 2005;83:747. doi: 10.1139/o05-146. [DOI] [PubMed] [Google Scholar]
- 58.Zheng L, Dengler TJ, Kluger MS, et al. Cytoprotection of human umbilical vein endothelial cells against apoptosis and CTL-mediated lysis provided by caspase-resistant Bcl-2 without alterations in growth or activation responses. J Immunol. 2000;164:4665. doi: 10.4049/jimmunol.164.9.4665. [DOI] [PubMed] [Google Scholar]
- 59.Hsieh CL, Obara H, Ogura Y, Martinez OM, Krams SM. NK cells and transplantation. Transpl.Immunol. 2002;9:111. doi: 10.1016/s0966-3274(02)00033-3. [DOI] [PubMed] [Google Scholar]
- 60.Bottino C, Moretta L, Pende D, Vitale M, Moretta A. Learning how to discriminate between friends and enemies, a lesson from Natural Killer cells. Mol.Immunol. 2004;41:569. doi: 10.1016/j.molimm.2004.04.004. [DOI] [PubMed] [Google Scholar]
- 61.Bender JR, Pardi R, Engleman E. T-cell receptor-negative natural killer cells display antigen-specific cytotoxicity for microvascular endothelial cells. Proc Natl Acad Sci U S A. 1990;87:6949. doi: 10.1073/pnas.87.18.6949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Candinas D, Belliveau S, Koyamada N, et al. T cell independence of macrophage and natural killer cell infiltration, cytokine production, and endothelial activation during delayed xenograft rejection. Transplantation. 1996;62:1920. doi: 10.1097/00007890-199612270-00042. [DOI] [PubMed] [Google Scholar]
- 63.Uehara S, Chase CM, Kitchens WH, et al. NK cells can trigger allograft vasculopathy: the role of hybrid resistance in solid organ allografts. J Immunol. 2005;175:3424. doi: 10.4049/jimmunol.175.5.3424. [DOI] [PubMed] [Google Scholar]
- 64.Wyburn KR, Jose MD, Wu H, Atkins RC, Chadban SJ. The role of macrophages in allograft rejection. Transplantation. 2005;80:1641. doi: 10.1097/01.tp.0000173903.26886.20. [DOI] [PubMed] [Google Scholar]
- 65.Diez-Roux G, Lang RA. Macrophages induce apoptosis in normal cells in vivo. Development. 1997;124:3633. doi: 10.1242/dev.124.18.3633. [DOI] [PubMed] [Google Scholar]
- 66.Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453. doi: 10.2741/2692. [DOI] [PubMed] [Google Scholar]
- 67.Briscoe DM, Schoen FJ, Rice GE, Bevilacqua MP, Ganz P, Pober JS. Induced expression of endothelial-leukocyte adhesion molecules in human cardiac allografts. Transplantation. 1991;51:537. [PubMed] [Google Scholar]
- 68.Ferran C, Peuchmaur M, Desruennes M, et al. Implications of de novo ELAM-1 and VCAM-1 expression in human cardiac allograft rejection. Transplantation. 1993;55:605. doi: 10.1097/00007890-199303000-00026. [DOI] [PubMed] [Google Scholar]
- 69.Morita K, Miura M, Paolone DR, et al. Early chemokine cascades in murine cardiac grafts regulate T cell recruitment and progression of acute allograft rejection. J Immunol. 2001;167:2979. doi: 10.4049/jimmunol.167.5.2979. [DOI] [PubMed] [Google Scholar]
- 70.Ashkenazi A, Dixit VM. Death receptors: signaling and modulation. Science. 1998;281:1305. doi: 10.1126/science.281.5381.1305. [DOI] [PubMed] [Google Scholar]
- 71.Locksley RM, Killeen N, Lenardo MJ. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell. 2001;104:487. doi: 10.1016/s0092-8674(01)00237-9. [DOI] [PubMed] [Google Scholar]
- 72.Migone TS, Zhang J, Luo X, et al. TL1A is a TNF-like ligand for DR3 and TR6/DcR3 and functions as a T cell costimulator. Immunity. 2002;16:479. doi: 10.1016/s1074-7613(02)00283-2. [DOI] [PubMed] [Google Scholar]
- 73.Al-Lamki RS, Wang J, Tolkovsky AM, et al. TL1A Both Promotes and Protects from Renal Inflammation and Injury. J.Am.Soc.Nephrol. 2008 doi: 10.1681/ASN.2007060706. (Epub ahead of print) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Li JH, Kluger MS, Madge LA, Zheng L, Bothwell AL, Pober JS. Interferon-gamma augments CD95(APO-1/Fas) and pro-caspase-8 expression and sensitizes human vascular endothelial cells to CD95-mediated apoptosis. Am J Pathol. 2002;161:1485. doi: 10.1016/s0002-9440(10)64424-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Li JH, Pober JS. The cathepsin B death pathway contributes to TNF plus IFN-gamma-mediated human endothelial injury. J Immunol. 2005;175:1858. doi: 10.4049/jimmunol.175.3.1858. [DOI] [PubMed] [Google Scholar]
- 76.Al-Lamki RS, Wang J, Skepper JN, Thiru S, Pober JS, Bradley JR. Expression of tumor necrosis factor receptors in normal kidney and rejecting renal transplants. Lab Invest. 2001;81:1503. doi: 10.1038/labinvest.3780364. [DOI] [PubMed] [Google Scholar]
- 77.Al-Lamki RS, Wang J, Thiru S, et al. Expression of silencer of death domains and death-receptor-3 in normal human kidney and in rejecting renal transplants. Am.J.Pathol. 2003;163:401. doi: 10.1016/S0002-9440(10)63670-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Al-Lamki RS, Wang J, Vandenabeele P, et al. TNFR1- and TNFR2-mediated signaling pathways in human kidney are cell type-specific and differentially contribute to renal injury. FASEB J. 2005;19:1637. doi: 10.1096/fj.05-3841com. [DOI] [PubMed] [Google Scholar]
- 79.Kato M, Morozumi K, Takeuchi O, et al. Complement fragment C4d deposition in peritubular capillaries in acute humoral rejection after ABO blood group-incompatible human kidney transplantation. Transplantation. 2003;75:663. doi: 10.1097/01.TP.0000053402.87256.6B. [DOI] [PubMed] [Google Scholar]
- 80.Tinckam KJ, Chandraker A. Mechanisms and role of HLA and non-HLA alloantibodies. Clin J Am Soc Nephrol. 2006;1:404. doi: 10.2215/CJN.00270106. [DOI] [PubMed] [Google Scholar]
- 81.Akalin E, Watschinger B. Antibody-mediated rejection. Semin Nephrol. 2007;27:393. doi: 10.1016/j.semnephrol.2007.05.001. [DOI] [PubMed] [Google Scholar]
- 82.Yamakuchi M, Kirkiles-Smith NC, Ferlito M, et al. Antibody to human leukocyte antigen triggers endothelial exocytosis. Proc Natl Acad Sci U S A. 2007;104:1301. doi: 10.1073/pnas.0602035104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wasowska BA, Lee CY, Halushka MK, Baldwin WM., 3rd New concepts of complement in allorecognition and graft rejection. Cell Immunol. 2007;248:18. doi: 10.1016/j.cellimm.2007.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Platt JL, Dalmasso AP, Lindman BJ, Ihrcke NS, Bach FH. The role of C5a and antibody in the release of heparan sulfate from endothelial cells. Eur J Immunol. 1991;21:2887. doi: 10.1002/eji.1830211135. [DOI] [PubMed] [Google Scholar]
- 85.Brooimans RA, van Wieringen PA, van Es LA, Daha MR. Relative roles of decayaccelerating factor, membrane cofactor protein, and CD59 in the protection of human endothelial cells against complement-mediated lysis. Eur.J.Immunol. 1992;22:3135. doi: 10.1002/eji.1830221216. [DOI] [PubMed] [Google Scholar]
- 86.Brooimans RA, Van der Ark AA, Tomita M, van Es LA, Daha MR. CD59 expressed by human endothelial cells functions as a protective molecule against complement-mediated lysis. Eur.J.Immunol. 1992;22:791. doi: 10.1002/eji.1830220324. [DOI] [PubMed] [Google Scholar]
- 87.Sims PJ, Wiedmer T. Induction of cellular procoagulant activity by the membrane attack complex of complement. Semin Cell Biol. 1995;6:275. doi: 10.1006/scel.1995.0037. [DOI] [PubMed] [Google Scholar]
- 88.Methe H, Balcells M, Alegret MC, et al. Vascular bed origin dictates flow pattern regulation of endothelial adhesion molecule expression. Am.J.Physiol Heart Circ.Physiol. 2007;292:H2167. doi: 10.1152/ajpheart.00403.2006. [DOI] [PubMed] [Google Scholar]
- 89.Koo DD, Welsh KI, McLaren AJ, Roake JA, Morris PJ, Fuggle SV. Cadaver versus living donor kidneys: impact of donor factors on antigen induction before transplantation. Kidney Int. 1999;56:1551. doi: 10.1046/j.1523-1755.1999.00657.x. [DOI] [PubMed] [Google Scholar]
- 90.Schlichting CL, Schareck WD, Weis M. Renal ischemia-reperfusion injury: new implications of dendritic cell-endothelial cell interactions. Transplant.Proc. 2006;38:670. doi: 10.1016/j.transproceed.2006.01.059. [DOI] [PubMed] [Google Scholar]
- 91.Brodsky SV, Zhang F, Nasjletti A, Goligorsky MS. Endothelium-derived microparticles impair endothelial function in vitro. Am.J.Physiol Heart Circ.Physiol. 2004;286:H1910. doi: 10.1152/ajpheart.01172.2003. [DOI] [PubMed] [Google Scholar]
- 92.Densmore JC, Signorino PR, Ou J, et al. Endothelium-derived microparticles induce endothelial dysfunction and acute lung injury. Shock. 2006;26:464. doi: 10.1097/01.shk.0000228791.10550.36. [DOI] [PubMed] [Google Scholar]
- 93.Mezentsev A, Merks RM, O'Riordan E, et al. Endothelial microparticles affect angiogenesis in vitro: role of oxidative stress. Am.J.Physiol Heart Circ.Physiol. 2005;289:H1106. doi: 10.1152/ajpheart.00265.2005. [DOI] [PubMed] [Google Scholar]
- 94.Soares MP, Lin Y, Sato K, Stuhlmeier KM, Bach FH. Accommodation. Immunol Today. 1999;20:434. doi: 10.1016/s0167-5699(99)01530-3. [DOI] [PubMed] [Google Scholar]
- 95.Madge LA, Pober JS. A phosphatidylinositol 3-kinase/Akt pathway, activated by tumor necrosis factor or interleukin-1, inhibits apoptosis but does not activate NFkappaB in human endothelial cells. J Biol Chem. 2000;275:15458. doi: 10.1074/jbc.M001237200. [DOI] [PubMed] [Google Scholar]
- 96.Lemos FB, Ijzermans JN, Zondervan PE, et al. Differential expression of heme oxygenase-1 and vascular endothelial growth factor in cadaveric and living donor kidneys after ischemia-reperfusion. J Am Soc Nephrol. 2003;14:3278. doi: 10.1097/01.asn.0000098683.92538.66. [DOI] [PubMed] [Google Scholar]
- 97.House SL, Bolte C, Zhou M, et al. Cardiac-specific overexpression of fibroblast growth factor-2 protects against myocardial dysfunction and infarction in a murine model of low-flow ischemia. Circulation. 2003;108:3140. doi: 10.1161/01.CIR.0000105723.91637.1C. [DOI] [PubMed] [Google Scholar]
- 98.d'Uscio LV, Smith LA, Santhanam AV, Richardson D, Nath KA, Katusic ZS. Essential role of endothelial nitric oxide synthase in vascular effects of erythropoietin. Hypertension. 2007;49:1142. doi: 10.1161/HYPERTENSIONAHA.106.085704. [DOI] [PubMed] [Google Scholar]
- 99.Mahboubi K, Biedermann BC, Carroll JM, Pober JS, et al. IL-11 activates human endothelial cells to resist immune-mediated injury. J Immunol. 2000;164:3837. doi: 10.4049/jimmunol.164.7.3837. [DOI] [PubMed] [Google Scholar]
- 100.Waxman AB, Mahboubi K, Knickelbein RG, et al. Interleukin-11 and interleukin-6 protect cultured human endothelial cells from H2O2-induced cell death. Am J Respir Cell Mol Biol. 2003;29:513. doi: 10.1165/rcmb.2002-0044OC. [DOI] [PubMed] [Google Scholar]
- 101.Lagaaij EL, Cramer-Knijnenburg GF, van Kemenade FJ, van Es LA, Bruijn JA, van Krieken JH. Endothelial cell chimerism after renal transplantation and vascular rejection. Lancet. 2001;357:33. doi: 10.1016/S0140-6736(00)03569-8. [DOI] [PubMed] [Google Scholar]
- 102.Sieveking DP, Buckle A, Celermajer DS, Ng MK. Strikingly different angiogenic properties of endothelial progenitor cell subpopulations: insights from a novel human angiogenesis assay. J.Am.Coll.Cardiol. 2008;51:660. doi: 10.1016/j.jacc.2007.09.059. [DOI] [PubMed] [Google Scholar]
- 103.Wijelath ES, Rahman S, Murray J, Patel Y, Savidge G, Sobel M. Fibronectin promotes VEGF-induced CD34 cell differentiation into endothelial cells. J.Vasc.Surg. 2004;39:655. doi: 10.1016/j.jvs.2003.10.042. [DOI] [PubMed] [Google Scholar]
- 104.Zemani F, Silvestre JS, Fauvel-Lafeve F, et al. Ex vivo priming of endothelial progenitor cells with SDF-1 before transplantation could increase their proangiogenic potential. Arterioscler.Thromb.Vasc.Biol. 2008;28:644. doi: 10.1161/ATVBAHA.107.160044. [DOI] [PubMed] [Google Scholar]
- 105.Min W, Pober JS, Johnson DR. Kinetically coordinated induction of TAP1 and HLA class I by IFN-gamma: the rapid induction of TAP1 by IFN-gamma is mediated by Stat1 alpha. J Immunol. 1996;156:3174. [PubMed] [Google Scholar]
- 106.Collins T, Korman AJ, Wake CT, et al. Immune interferon activates multiple class II major histocompatibility complex genes and the associated invariant chain gene in human endothelial cells and dermal fibroblasts. Proc Natl Acad Sci U S A. 1984;81:4917. doi: 10.1073/pnas.81.15.4917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Pober JS, Gimbrone MA, Jr, Collins T, et al. Interactions of T lymphocytes with human vascular endothelial cells: role of endothelial cells surface antigens. Immunobiology. 1984;168:483. doi: 10.1016/s0171-2985(84)80132-1. [DOI] [PubMed] [Google Scholar]
- 108.Suarez Y, Shepherd BR, Rao DA, Pober JS. Alloimmunity to human endothelial cells derived from cord blood progenitors. J Immunol. 2007;179:7488. doi: 10.4049/jimmunol.179.11.7488. [DOI] [PubMed] [Google Scholar]
- 109.Khayyamian S, Hutloff A, Buchner K, et al. ICOS-ligand, expressed on human endothelial cells, costimulates Th1 and Th2 cytokine secretion by memory CD4+ T cells. Proc Natl Acad Sci U S A. 2002;99:6198. doi: 10.1073/pnas.092576699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Karmann K, Hughes CC, Schechner J, Fanslow WC, Pober JS. CD40 on human endothelial cells: inducibility by cytokines and functional regulation of adhesion molecule expression. Proc Natl Acad Sci U S A. 1995;92:4342. doi: 10.1073/pnas.92.10.4342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kurt-Jones EA, Fiers W, Pober JS. Membrane interleukin 1 induction on human endothelial cells and dermal fibroblasts. J Immunol. 1987;139:2317. [PubMed] [Google Scholar]
- 112.May LT, Torcia G, Cozzolino F, et al. Interleukin-6 gene expression in human endothelial cells: RNA start sites, multiple IL-6 proteins and inhibition of proliferation. Biochem Biophys Res Commun. 1989;159:991. doi: 10.1016/0006-291x(89)92206-7. [DOI] [PubMed] [Google Scholar]
- 113.Beutelspacher SC, Tan PH, McClure MO, Larkin DF, Lechler RI, George AJ. Expression of indoleamine 2,3-dioxygenase (IDO) by endothelial cells: implications for the control of alloresponses. Am J Transplant. 2006;6:1320. doi: 10.1111/j.1600-6143.2006.01324.x. [DOI] [PubMed] [Google Scholar]
- 114.Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol. 2004;172:2731. doi: 10.4049/jimmunol.172.5.2731. [DOI] [PubMed] [Google Scholar]
- 115.Choi J, Enis DR, Koh KP, Shiao SL, Pober JS. T lymphocyte-endothelial cell interactions. Annu Rev Immunol. 2004;22:683. doi: 10.1146/annurev.immunol.22.012703.104639. [DOI] [PubMed] [Google Scholar]
- 116.Gatti E, Velleca MA, Biedermann BC, et al. Large-scale culture and selective maturation of human Langerhans cells from granulocyte colony-stimulating factor-mobilized CD34+ progenitors. J Immunol. 2000;164:3600. doi: 10.4049/jimmunol.164.7.3600. [DOI] [PubMed] [Google Scholar]
- 117.Pober JS, Bevilacqua MP, Mendrick DL, Lapierre LA, Fiers W, Gimbrone MA., Jr Two distinct monokines, interleukin 1 and tumor necrosis factor, each independently induce biosynthesis and transient expression of the same antigen on the surface of cultured human vascular endothelial cells. J Immunol. 1986;136:1680. [PubMed] [Google Scholar]
- 118.Pober JS, Gimbrone MA, Jr, Lapierre LA, et al. Overlapping patterns of activation of human endothelial cells by interleukin 1, tumor necrosis factor, and immune interferon. J Immunol. 1986;137:1893. [PubMed] [Google Scholar]
- 119.Carlos T, Kovach N, Schwartz B, et al. Human monocytes bind to two cytokine-induced adhesive ligands on cultured human endothelial cells: endothelial-leukocyte adhesion molecule-1 and vascular cell adhesion molecule-1. Blood. 1991;77:2266. [PubMed] [Google Scholar]
- 120.McCarty JM, Yee EK, Deisher TA, Harlan JM, Kaushansky K. Interleukin-4 induces endothelial vascular cell adhesion molecule-1 (VCAM-1) by an NF-kappa b-independent mechanism. FEBS Lett. 1995;372:194. doi: 10.1016/0014-5793(95)00976-g. [DOI] [PubMed] [Google Scholar]
- 121.Hattori R, Hamilton KK, Fugate RD, McEver RP, Sims PJ. Stimulated secretion of endothelial von Willebrand factor is accompanied by rapid redistribution to the cell surface of the intracellular granule membrane protein GMP-140. J Biol Chem. 1989;264:7768. [PubMed] [Google Scholar]
- 122.de Fougerolles AR, Stacker SA, Schwarting R, Springer TA. Characterization of ICAM-2 and evidence for a third counter-receptor for LFA-1. J Exp Med. 1991;174:253. doi: 10.1084/jem.174.1.253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Muller WA, Weigl SA, Deng X, Phillips DM. PECAM-1 is required for transendothelial migration of leukocytes. J Exp Med. 1993;178:449. doi: 10.1084/jem.178.2.449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Schenkel AR, Mamdouh Z, Chen X, Liebman RM, Muller WA. CD99 plays a major role in the migration of monocytes through endothelial junctions. Nat Immunol. 2002;3:143. doi: 10.1038/ni749. [DOI] [PubMed] [Google Scholar]
- 125.Cabre F, Tost D, Suesa N, et al. Synthesis and release of platelet-activating factor and eicosanoids in human endothelial cells induced by different agonists. Agents Actions. 1993;38:212. doi: 10.1007/BF01976213. [DOI] [PubMed] [Google Scholar]
- 126.Strieter RM, Kunkel SL, Showell HJ, et al. Endothelial cell gene expression of a neutrophil chemotactic factor by TNF-alpha, LPS, and IL-1 beta. Science. 1989;243:1467. doi: 10.1126/science.2648570. [DOI] [PubMed] [Google Scholar]
- 127.Wen DZ, Rowland A, Derynck R. Expression and secretion of gro/MGSA by stimulated human endothelial cells. Embo J. 1989;8:1761. doi: 10.1002/j.1460-2075.1989.tb03569.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Luster AD, Unkeless JC, Ravetch JV. Gamma-interferon transcriptionally regulates an early-response gene containing homology to platelet proteins. Nature. 1985;315:672. doi: 10.1038/315672a0. [DOI] [PubMed] [Google Scholar]
- 129.Kao J, Kobashigawa J, Fishbein MC, et al. Elevated serum levels of the CXCR3 chemokine ITAC are associated with the development of transplant coronary artery disease. Circulation. 2003;107:1958. doi: 10.1161/01.CIR.0000069270.16498.75. [DOI] [PubMed] [Google Scholar]
- 130.Koga S, Kobayashi H, Novick AC, Toma H, Fairchild RL. Alloantigen-specific CD8(+) T cells stimulate endothelial cells to produce the T-cell chemoattractants IP-10 and Mig. Transplant Proc. 2001;33:459. doi: 10.1016/s0041-1345(00)02093-5. [DOI] [PubMed] [Google Scholar]
- 131.Rollins BJ, Yoshimura T, Leonard EJ, Pober JS. Cytokine-activated human endothelial cells synthesize and secrete a monocyte chemoattractant, MCP-1/JE. Am J Pathol. 1990;136:1229. [PMC free article] [PubMed] [Google Scholar]
- 132.Rollins BJ, Pober JS. Interleukin-4 induces the synthesis and secretion of MCP-1/JE by human endothelial cells. Am J Pathol. 1991;138:1315. [PMC free article] [PubMed] [Google Scholar]
- 133.Marfaing-Koka A, Devergne O, Gorgone G, et al. Regulation of the production of the RANTES chemokine by endothelial cells. Synergistic induction by IFN-gamma plus TNF-alpha and inhibition by IL-4 and IL-13. J Immunol. 1995;154:1870. [PubMed] [Google Scholar]
- 134.Shinkai A, Yoshisue H, Koike M, et al. A novel human CC chemokine, eotaxin-3, which is expressed in IL-4-stimulated vascular endothelial cells, exhibits potent activity toward eosinophils. J Immunol. 1999;163:1602. [PubMed] [Google Scholar]
- 135.Imaizumi T, Matsumiya T, Fujimoto K, et al. Interferon-gamma stimulates the expression of CX3CL1/fractalkine in cultured human endothelial cells. Tohoku J Exp Med. 2000;192:127. doi: 10.1620/tjem.192.127. [DOI] [PubMed] [Google Scholar]
- 136.Maier JA, Hla T, Maciag T. Cyclooxygenase is an immediate-early gene induced by interleukin-1 in human endothelial cells. J Biol Chem. 1990;265:10805. [PubMed] [Google Scholar]
- 137.Yoshizumi M, Perrella MA, Burnett JC, Jr, Lee ME. Tumor necrosis factor downregulates an endothelial nitric oxide synthase mRNA by shortening its half-life. Circ Res. 1993;73:205. doi: 10.1161/01.res.73.1.205. [DOI] [PubMed] [Google Scholar]
- 138.Bevilacqua MP, Pober JS, Majeau GR, Fiers W, Cotran RS, Gimbrone MA., Jr Recombinant tumor necrosis factor induces procoagulant activity in cultured human vascular endothelium: characterization and comparison with the actions of interleukin 1. Proc Natl Acad Sci U S A. 1986;83:4533. doi: 10.1073/pnas.83.12.4533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Miller DL, Yaron R, Yellin MJ. CD40L-CD40 interactions regulate endothelial cell surface tissue factor and thrombomodulin expression. J Leukoc Biol. 1998;63:373. doi: 10.1002/jlb.63.3.373. [DOI] [PubMed] [Google Scholar]
- 140.Conway EM, Rosenberg RD. Tumor necrosis factor suppresses transcription of the thrombomodulin gene in endothelial cells. Mol Cell Biol. 1988;8:5588. doi: 10.1128/mcb.8.12.5588. [DOI] [PMC free article] [PubMed] [Google Scholar]