Abstract
CD8 T cells play a critical role in several pathological conditions affecting the liver, most notably viral hepatitis. Accordingly, understanding the mechanisms that modulate the intrahepatic recruitment of CD8 T cells is of paramount importance. Some of the rules governing the behavior of these cells in the liver have been characterized at the population level, or have been inferred by studying the intrahepatic behavior of other leukocyte subpopulations. In contrast to most microvascular beds where leukocyte adhesion is restricted to the endothelium of post-capillary venules, it is now becoming clear that in the liver leukocytes, including CD8 T cells, can efficiently interact with the endothelium of hepatic capillaries (i.e. the sinusoids). While physical trapping has been proposed to play an important role in leukocyte adhesion to hepatic sinusoids, there is mounting evidence that T cell recruitment to the liver is highly regulated and depends on recruitment signals that are either constitutive or induced by inflammation. We review here several specific adhesive mechanisms that have been shown to regulate CD8 T cell trafficking within the liver, as well as highlight recent data that establish platelets as key cellular regulators of intrahepatic CD8 T cell accumulation.
Keywords: Liver, CD8, Hepatitis B virus, Hepatitis C virus, hepatocellular carcinoma, platelets, selectins, integrins, chemokines, VAP-1, CD44
1. Introduction
CD8 T cells play a fundamental role in the pathogenesis of liver disease and viral clearance during acute, self-limited hepatitis B virus (HBV) and hepatitis C virus (HCV) infection(Guidotti and Chisari, 2006; Iannacone et al., 2006). Moreover, the pathogenesis of chronic HBV or HCV infection is thought to involve functionally inefficient CD8 T cells that do not eradicate the infection but sustain repetitive cycles of immune-mediated hepatocellular necrosis, hepatocellular regeneration and inflammation that are likely to precipitate random genetic damage and promote HCC development(Guidotti and Chisari, 2006). Both CD8 T cells’ defensive and destructive functions are mediated by antigen(Ag)-experienced effector cells and depend on these cells’ ability to migrate from the blood to the liver. Understanding the signals that modulate the intrahepatic recruitment of CD8 T cells is therefore critical to get insight into the pathogenesis of acute and chronic viral hepatitis.
The classic paradigm for leukocyte migration from blood vessels to interstitial tissues involves a multistep process that occurs in post-capillary venules(Springer, 1994) but not in arterioles or capillaries (where leukocyte adhesion may limit gas exchange and tissue perfusion(Andrian and Mackay, 2000)). The initial weak rolling interactions between leukocytes and endothelial cells are mediated by a family of proteins called selectins(Kansas, 1996). There are three types of selectins: one expressed on leukocytes (L-selectin), one on endothelial cells (E-selectin), and one on platelets and on endothelial cells (P-selectin). The ligands for selectins are sialylated oligosaccharides bound to mucin-like glycoprotein backbones(Kansas, 1996). Firm adhesion of leukocytes to endothelial cells is mediated by a family of heterodimeric leukocyte surface proteins called integrins(Hynes, 1992; Springer, 1994). The combination of cytokine-induced endothelial expression of integrin ligands, mainly vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1), and chemokine-mediated conversion of integrins to a high-affinity state on leukocytes(Hynes, 2002) results in firm adhesion of leukocytes to the endothelium at sites of inflammation.
The liver represents an exception to this leukocyte migration paradigm in several respects(Lee and Kubes, 2008). First, leukocyte adhesion is not restricted to the endothelium of post-capillary venules, but it also occurs in sinusoids(Lee and Kubes, 2008); indeed, in response to a chemotactic stimulus such as N-formyl-methionyl-leucyl-phenylalanine (fMLP), the majority of leukocytes have been shown to adhere to the sinusoidal bed, with only a small fraction of leukocytes adhering to post-sinusoidal venules(Wong et al., 1997). It is of note, however, that the quantitative importance of sinusoidal adhesion is less established for CD8 T cells, particularly in the context of intrahepatic Ag recognition. Second, visualization of leukocyte behavior in the liver microvasculature revealed that while in post-sinusoidal vessels rolling precedes adhesion, leukocyte adhesion to liver sinusoidal endothelial cells (LSEC) often occurs independent of any notable rolling(Lee and Kubes, 2008). It is also of note that LSEC are morphologically unique and characterized by the absence of tight junctions between cells and the lack of a basal membrane (Wisse et al., 1985). This is in contrast to most vascular beds in other tissues and organs, where a continuous endothelial cell layer and a basement membrane physically separate parenchymal cells from circulating leukocytes (Wisse et al., 1985). Moreover, hepatocyte membranes often protrude from the fenestrated endothelial barrier of sinusoids, thus providing the opportunity for direct interaction of circulating cells with the underlying hepatocytes(Warren et al., 2006). For all these reasons, the molecular mechanisms leading to leukocyte adhesion to LSEC appear to be somewhat different from those occurring in post-capillary venules of other vascular districts(Lee and Kubes, 2008). We will review below our current understanding of the molecular and cellular mechanisms mediating CD8 T cell homing to the liver, focusing, when possible, on effector CD8 T cell trafficking in the context of Ag recognition.
2. Selectins
A mentioned earlier, the selectin family has three members: L-selectin (CD62L), E-selectin (CD62E) and P-selectin (CD62P). Selectins are the quintessential adhesion molecules: they are highly efficient mediators of tethering and rolling(Kansas, 1996), and they do so constitutively, i.e. they do not require an activating stimulus to bind to a carbohydrate ligand through their N-terminal, Ca2+-dependent lectin domain. The role of selectins in leukocyte recruitment into organs such as lymph nodes, peritoneal cavity, mesentery, muscle and skin has been extremely well characterized (Lee and Kubes, 2008).
Consistently with the idea that leukocyte adhesion to LSEC occurs independently of any notable rolling (see above), selectins were shown to be dispensable for leukocyte adhesion in liver sinusoids (Wong et al., 1997; Essani et al., 1998; Fox-Robichaud and Kubes, 2000; Bowen et al., 2004). Similarly, migration of virus-specific CD8 T cells to lymphocytic choriomeningitis virus (LCMV)-infected liver was shown to occur in the absence of endothelial (E/P) selectins(Bartholdy et al., 2000).
3. Integrins
Integrins are a large family of heterodimeric glycoproteins(Hynes, 1992; Springer, 1994; Hynes, 2002) that are found on most cell types. Two subfamilies are most important for leukocyte migration: the α4- (CD49) and the β2- (CD18) integrins. Endothelial ligands for these molecules are members of the immunoglobulin superfamily (IgSF). Arguably, the most important ligand for β2-integrins is the IgSF member ICAM-1. In most vascular districts ICAM-1 is constitutively expressed on post-capillary venules and only minimally on capillaries; by contrast, the density of ICAM-1 in hepatic sinusoids is comparable to that of central venules(Iigo et al., 1997). The IgSF member VCAM-1 (the ligand for the α4-integrin VLA-4) is not expressed in normal liver tissue but it is markedly upregulated on sinusoidal endothelium when inflammation is present(Volpes et al., 1992). During some inflammatory conditions the hepatic endothelium can be induced to also express the mucosal addressin cell adhesion molecule-1 (MAdCAM-1; the ligand for α4β7), which is normally confined to mucosal endothelium in the bowel (see later).
An important feature of integrins is their “tunability”(Hynes, 2002). While selectins are always active, integrins must first assume an activated state to mediate adhesion and their affinity and/or avidity towards the respective ligands can be rapidly modified in response to stimuli such as chemokines (see later). We will review below our current understanding of the role that specific integrins play in the recruitment of CD8 T cells to both uninflamed and inflamed liver microvasculature.
3.1. β2-integrins
The leucocyte-restricted β2-integrins comprise four members, namely αLβ2 (LFA-1), αMβ2 (Mac-1), αxβ2 (p150, 95) and αDβ2. Each of the four known β2-integrin heterodimers has a different cellular distribution, with LFA-1 expressed on all leukocytes, including CD8 T cells(Luo et al., 2007; Tan, 2012). The absence of β2-integrins in humans results in leukocyte adhesion deficiency type 1 (LAD-1), a syndrome that manifests itself with increased susceptibility to infections and impaired capacity of wound healing(Anderson and Springer, 1987). β2-integrin knockout mice have a phenotype similar to that of LAD-1 patients, and leukocytes derived from these animals show diminished ability to extravasate at sites of infection or injury(Grabbe et al., 2002).
Although it is now well appreciated that β2-integrin mediates firm adhesion of leukocytes in many tissues, the evidence for a role for this integrin in the liver is less compelling(Lee and Kubes, 2008). LFA-1 has been proposed to mediate both naïve(Bertolino et al., 2005) and effector(John and Crispe, 2004; Sato et al., 2006) CD8 T cell adhesion to LSEC, but this may occur only in the context of antigen presentation in the liver, possibly because of a TCR-mediated increase in LFA-1 affinity for ICAM-1.
3.2. α4-integrins
The α4-integrin family includes α4β1 (VLA-4) and α4β7, two molecules that are expressed on lymphocytes and monocytes(Springer, 1994). VLA-4 and α4β7 bind to VCAM-1 and MAdCAM-1, respectively, on endothelial cells, and this process regulates the trafficking of different leukocyte subsets in mucosal tissues, especially the gut(Springer, 1994). Antigen non-specific adhesion of activated CD8 T cells to LSEC has been proposed to occur via VCAM-1/α4-integrin(John and Crispe, 2004). In a graft versus host disease model, however, α4-integrin was found to be dispensable for the recruitment of activated CD8+ T cells into the liver(Sato et al., 2006). Also, naïve CD8 T cells appear not to need α4-integrin to be recruited to the uninflamed liver (Bertolino et al., 2005).
As mentioned earlier, when the hepatic vasculature is inflamed, additional adhesion molecules are expressed(Crispe, 2012). Of particular interest is MAdCAM-1, which engages the α4β7 integrin that is prominently expressed on intestinal lymphocytes. This interaction might account for the hepatic trapping of activated gut-derived T cells in inflammatory bowel disease(Grant et al., 2001).
3.3. αVβ3
Activated lymphocytes express the integrin αVβ3, which binds several extracellular matrix molecules and PECAM-1 (CD31), an IgSF molecule that has been implicated in leukocyte migration across endothelial cells(Muller et al., 1993). Although the role of αVβ3 in CD8 T cell trafficking to the liver has never been tested, in vivo blockade of this molecule via specific antibodies had no effect on hepatic neutrophil accumulation(Chosay et al., 1998). This is consistent with the observation that LSEC express little or no PECAM-1(Chosay et al., 1998).
4. Chemokines
Chemokines (chemotactic cytokines) are secreted polypeptides that bind to surface receptors and transmit signals through Gαi proteins(Rot and Andrian, 2004). Just like adhesion molecules, chemokine receptors can be upregulated or lost as cells differentiate, allowing leukocytes to coordinate migratory routes and biological function(Rot and Andrian, 2004). After secretion into extracellular spaces, chemokines bind to heparin-like glycosaminoglycans on cell surfaces and in the extracellular matrix; leukocytes can track down these immobilized chemokines, which may persist in tissues longer and at higher concentrations than freely diffusible molecules. Since lymphocytes must be positioned correctly to interact with other cells, the pattern of chemokine receptors and the type and distribution of chemokines in tissues critically influence immune responses(Rot and Andrian, 2004).
The chemokine molecular signature includes four conserved cysteine residues that form two disulfide bonds pairing the first with the third and the second with the fourth cysteines(Zlotnik and Yoshie, 2012). Based on the arrangement of the N-terminal two cysteine residues, chemokines are grouped into four subfamilies: CXC, CC, (X)C, and CX3C. In the CXC chemokines, one amino acid separates the first two cysteines, whereas in CC chemokines, these two cysteines are adjacent. A single member of the CX3C subfamily, CX3CL1 or fractalkine, has three amino acids between the two cysteines, whereas the first and third cysteines are missing in the (X)C subfamily(Zlotnik and Yoshie, 2012).
Several lines of evidence from human studies suggest that the intrahepatic recruitment of CD8 T cells and other inflammatory cells may be promoted by chemokines. Most effector T cells infiltrating the chronically inflamed human liver express high levels of CXCR3, CXCR6, CCR1 and CCR5(Shields et al., 1999; Kunkel et al., 2002; Boisvert et al., 2003; Leroy et al., 2003; Heydtmann et al., 2006);(Dumoulin et al., 1997; Apolinario et al., 2002; Arai et al., 2002; Diago et al., 2006; Larrubia et al., 2007), with CCR5-bearing T cells preferentially accumulating around portal tracts and CXCR3-bearing T cells distributing more evenly throughout the liver lobule(Murai et al., 1999; Harvey et al., 2003; Curbishley et al., 2005). We review below examples of specific chemokine/chemokine receptor interactions that appear to be predominant at mediating CD8 T cell recruitment into the liver.
4.1. CXCR3 and its ligands CXCL9, CXCL10 and CXCL11
Using flow-based adhesion assays it has been shown that the CXCR3 ligands CXCL9, CXCL10 and CXCL11 are important not only in adhesion, but also in transmigration of human effector T lymphocytes through the hepatic endothelium(Curbishley et al., 2005). Additional evidence for a role of CXCR3 and its ligands in liver T cell trafficking comes from studies in which HBV-replication competent transgenic mice were used as recipients of HBV-specific effector CD8 T cells. In those studies it was shown that CXCL9 and CXCL10 are rapidly and strongly induced in the liver after T cell transfer(Kakimi et al., 2001); the transferred T cells produce neither chemokine but, rather, they activate (via the secretion of IFN-γ) liver non-parenchymal cells and especially hepatocytes to produce them (Kakimi et al., 2001); importantly, blocking CXCL9 and CXCL10 in vivo reduces the recruitment of host-derived mononuclear cells into the liver, particularly those subsets that are known to express CXCR3 (NK cells, myeloid dendritic cells and effector CD8 T cells)(Kakimi et al., 2001). It is also important to note that CXCL9 and CXCL10 neutralization only partially reduces the recruitment of virus-specific effector CD8 T cells(Kakimi et al., 2001), suggesting that other receptor/ligand pairs play a role in the intrahepatic recruitment of these cells.
4.2. CXCR6 and its ligand CXCL16
Both human(Heydtmann et al., 2005) and mouse(Sato et al., 2005) studies have shown that CXCR6/CXCL16 can regulate the recruitment of activated CD8 T cells to the inflamed liver. Along these lines, Klenerman and colleagues have recently reported a unique subset of HCV-specific CXCR6+ liver-inflitrating CD8 T cells that express the C-type lectin CD161 and secrete IL-17 and IFN-γ(Northfield et al., 2008). Of note, CXCR6 was also shown to be required for the hepatic homing of NK and NKT cells(Geissmann et al., 2005).
4.3. CCR5 and its ligand CCL3
In murine models of graft-versus-host disease CCR5 and CCL3 have been shown to support effector CD8 T cell recruitment to portal tracts(Murai et al., 1999; 2003).
4.4. CCR9 and its ligand CCL25
During primary sclerosing cholangitis, a chronic inflammatory liver disease characterized by progressive bile duct destruction and developing as an extra-intestinal complication of inflammatory bowel disease, liver-infiltrating lymphocytes include CCR9+ mucosal T cells(Eksteen et al., 2004). It has been suggested that the hepatic recruitment of CCR9+ T cells depends on the aberrant liver expression of the gut-specific chemokine CCL25(Eksteen et al., 2004).
5. Other adhesion molecules (VAP-1, CD44)
5.1. VAP-1
VAP-1 is a 170-kDa homodimeric glycoprotein that is expressed by endothelial cells and mediates lymphocyte binding to high endothelial venules under shear conditions(Salmi and Jalkanen, 1996; Salmi et al., 1997). VAP-1 is expressed at high levels in the human liver and it promotes lymphocyte adhesion and transmigration across hepatic sinusoidal endothelial cells in vitro(Lalor et al., 2002). In humans, LSEC have been shown to constitutively express VAP-1 and to up-regulate this protein during inflammatory responses(McNab et al., 1996). In mice, LSEC express very little VAP-1 under basal conditions, but they do significantly express it when inflammation is present(Bonder et al., 2005).
Although CD4 T cells polarized to a Th2 phenotype have been shown to require VAP-1 for efficient homing to the inflamed liver(Bonder et al., 2005), CD8 T cell recruitment into the liver has been shown to be independent of VAP-1(Bertolino et al., 2005). Of note, the ligand for VAP-1 remains to be identified, but it has been postulated that, upon activation, this glycoprotein leads to the upregulation of E-selectin, ICAM-1 and VCAM-1 on LSEC and the secretion of CXCL8, thus supporting leukocyte homing indirectly (Lalor et al., 2007).
5.2. CD44
Interaction of CD44 with sinusoid-expressed hyaluronan has been recently proposed to be the dominant mechanism for neutrophil sequestration in inflamed liver sinusoids(Mcdonald et al., 2008). Antigen-experienced CD8 T cells are known to express high levels of CD44, but whether this molecule can also support lymphocyte adhesion to hepatic sinusoids is yet to be defined.
6. Kupffer cells and platelets
So far we discussed molecular interactions between CD8 T cells and the sinusoidal endothelium that have been shown to regulate T cell trafficking to the liver. Lymphocytes and endothelial cells, however, exist in a complex multi-cellular microenvironment where other cells types might influence their behavior via paracrine interaction. One example of this is provided by Kupffer cells, liver-resident intravascular macrophages. Although Kupffer cells can transiently interact with T cells (Bertolino et al., 2002), their contribution to the intrahepatic accumulation of effector CD8 T cells was shown to be negligible(Sitia et al., 2011). Another example is provided by platelets, anucleated blood cells that have been shown to interact with leukocytes and modulate their function(Ruggeri, 2009; Vieira-de-Abreu et al., 2012).
Several recent studies have demonstrated that intrahepatic recruitment of antigen-specific effector CD8 T cells is critically dependent on platelets(Iannacone et al., 2005; 2007a; 2007b; Lang et al., 2008; Iannacone et al., 2009; Sitia et al., 2012). Indeed, in mouse models of CD8 T cell-mediated acute viral hepatitis, we recently showed that platelet depletion is associated with a profound reduction in the intrahepatic accumulation of virus-specific effector CD8 T cells and a proportional reduction in liver disease severity, both of which are restored upon reconstitution with normal platelets, but not upon reconstitution with platelets treated with prostaglandin (PG)E1, a known inhibitor of platelet activation(Iannacone et al., 2005). In vitro findings also indicate that, under the low shear flow conditions likely occurring in the venous circulation of the liver, antigen-specific effector CD8 T cells tightly interact with platelets and, again, this process is inhibited when platelets are treated with PGE1(Iannacone et al., 2005). In the ongoing effort to explain mechanistically why platelets are required to support CD8+ T cell-induced liver pathology, we also found that this process is influenced by two specific inhibitors of platelet activation pathways, aspirin that blocks thromboxane (TX) A2 production and clopidogrel that blocks the P2Y12 ADP receptor(Cattaneo, 2004). Indeed, treating mice with aspirin, clopidogrel, or a combination of the two, attenuates acute liver injury by reducing the hepatic accumulation of antigen-specific CD8+ T cells and antigen-nonspecific inflammatory cells(Iannacone et al., 2007a). Of note, platelet activation follows adhesion to activated endothelium and/or exposed subendothelial matrix and is mediated primarily by two receptors, GPIb-α and GPVI, which bind to von Willebrand factor (vWF) and collagen, respectively(Ruggeri, 2002). Platelet activation induces cytoskeletal assembly and shape changes, secretion of agonists promoting further activation and aggregation, and functional expression of molecules such as P-selectin or GPIIbIIIa(Weyrich and Zimmerman, 2004) that could be involved in the interaction with effector CD8 T cells. Pertinent to this, platelet P-selectin has been shown to interact with PSGL-1 on leukocytes (including T cells) and promote their rolling along the endothelium of lymph nodes(Diacovo et al., 1996). Upon interaction with platelets, leukocytes are also thought to roll on the endothelium of cutaneous post-capillary venules thanks to platelet expression of GPIIbIIIa, which may secondarily interact with endothelial ICAM-1(Ludwig et al., 2004). Along these lines, intravital microscopy studies in mesenteric venules have recently suggested that, after directly supporting an initial rolling of leukocytes in a P-selectin-dependent manner, platelets stimulate endothelial cells to become activated, express P-selectin themselves, and further sustain leukocyte rolling(Dole et al., 2005). Based on the aforementioned evidence, it is possible that the expression of P-selectin or GPIIbIIIa on platelets and PSGL-1 on effector CD8 T cells(Borges et al., 1997) may promote interaction between these cell types.
If a functional connection between platelets and T cells depends on direct and/or indirect intercellular interactions within the liver remains to be demonstrated. We have proposed that the activation-dependent expression of platelet CD40 ligand contributes to the expansion phase of virus-specific CD8+ T cells, resulting in their accumulation at sites of infection(Iannacone et al., 2008); this effect may reflect direct interaction of activated platelets with CD8+ T cells that express CD40(Bourgeois et al., 2002; Meunier et al., 2012). Others have indicated that platelet CD40 ligand has the potential to enhance virus-specific CD8+ T cell responses indirectly, mostly by promoting the maturation of dendritic cells(Elzey et al., 2003; Li, 2008).
While the exact molecular mechanisms by which platelets support CD8 T cell-mediated liver immunopathology remains ill-defined, we recently adapted a mouse model of chronic immune-mediated hepatitis B that progresses to HCC(Nakamoto et al., 1998; 2004) to evaluate whether aspirin and clopidogrel may also blunt the hepatic accumulation of pathogenic effector CD8 T cells under conditions of sustained liver injury. We were able to show that anti-platelet therapy suppresses hepatic immunopathology overtime, thus preventing/delaying the development of HCC and improving overall survival(Sitia et al., 2012).
7. Conclusions and future directions
Significant advances have been made in our comprehension of hepatic CD8 T cell recruitment, how it differs from the recruitment of these cells to other tissues or organs and how the process is modulated by inflammation. While some of the rules that govern CD8 T cell homing to the liver have started to be clarified at the population level, we still have limited knowledge of the precise dynamics of intrahepatic CD8 T cell migration and interaction with other cell types at the single-cell level, particularly in the context of intrahepatic antigen recognition. We believe that recent advances in the field of live imaging, coupled with animal models that express viral antigens in the hepatocyte, will provide the opportunity to tackle some of these questions directly in the living animal. This will not only greatly improve our understanding of CD8 T cell trafficking within the liver but it may also provide tools for the design of new immune therapeutic strategies for the treatment of chronic viral hepatitis and liver cancer.
Highlights.
In addition to postcapillary venules, CD8 T cells adhere to liver sinusoids
Integrins and chemokines, not selectins, contribute to CD8 T cell adhesion
Platelets are key cellular regulators of intrahepatic CD8 T cell accumulation
Acknowledgments
We thank all members of the Guidotti and the Iannacone laboratories for helpful discussions.
This work was supported by European Research Council Grants 281648 (to M.I.) and 250219 (to L.G.G.), a Career Development Award from the Giovanni Armenise – Harvard Foundation (to M.I.), National Institutes of Health Grant R01-AI40696 (to L.G.G), Italian Association for Cancer Research Grants 4643 and 6278 (to L.G.G.).
Abbreviations
- HBV
Hepatitis B virus
- HCV
Hepatitis C virus
- HCC
hepatocellular carcinoma
- Ag
antigen
- VCAM-1
vascular cell adhesion molecule 1
- ICAM-1
intercellular adhesion molecule 1
- fMLP
N-formyl-methionyl-leucyl-phenylalanine
- LSEC
liver sinusoidal endothelial cells
- LCMV
lymphocytic choriomeningitis virus
- IgSF
immunoglobulin superfamily
- VLA-4
very late antigen 4
- MAdCAM-1
mucosal addressin cell adhesion molecule 1
- LFA-1
lymphocyte function-associated antigen 1
- LAD-1
leukocyte adhesion deficiency type 1
- TCR
T cell receptor
- PECAM-1
platelet endothelial cell adhesion molecule 1
- IFN
Interferon
- NK
natural killer
- IL
interleukin
- VAP
vascular adhesion protein
- Th
T helper
- PG
prostaglandin
- TX
thromboxane
- ADP
adenosine diphosphate
- GP
glycoprotein
- vWF
von Willebrand factor
- PSGL-1
P-selectin glycoprotein ligand 1
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Anderson DC, Springer TA. Leukocyte adhesion deficiency: an inherited defect in the Mac-1, LFA-1, and p150,95 glycoproteins. Annual Review of Medicine. 1987;38:175–194. doi: 10.1146/annurev.me.38.020187.001135. [DOI] [PubMed] [Google Scholar]
- von Andrian UH, Mackay CR. T-cell function and migration. Two sides of the same coin. N Engl J Med. 2000;343:1020–1034. doi: 10.1056/NEJM200010053431407. [DOI] [PubMed] [Google Scholar]
- Apolinario A, Majano PL, Alvarez-Pérez E, Saez A, Lozano C, Vargas J, García-Monzón C. Increased expression of T cell chemokines and their receptors in chronic hepatitis C: relationship with the histological activity of liver disease. Am J Gastroenterol. 2002;97:2861–2870. doi: 10.1111/j.1572-0241.2002.07054.x. [DOI] [PubMed] [Google Scholar]
- Arai K, Liu ZX, Lane T, Dennert G. IP-10 and Mig facilitate accumulation of T cells in the virus-infected liver. Cell Immunol. 2002;219:48–56. doi: 10.1016/s0008-8749(02)00584-1. [DOI] [PubMed] [Google Scholar]
- Bartholdy C, Marker O, Thomsen AR. Migration of activated CD8(+) T lymphocytes to sites of viral infection does not require endothelial selectins. Blood. 2000;95:1362–1369. [PubMed] [Google Scholar]
- Bertolino P, McCaughan GW, Bowen DG. Role of primary intrahepatic T-cell activation in the ‘liver tolerance effect’. Immunol Cell Biol. 2002;80:84–92. doi: 10.1046/j.0818-9641.2001.01048.x. [DOI] [PubMed] [Google Scholar]
- Bertolino P, Schrage A, Bowen DG, Klugewitz K, Ghani S, Eulenburg K, Holz L, Hogg N, McCaughan GW, Hamann A. Early intrahepatic antigen-specific retention of naïve CD8+ T cells is predominantly ICAM-1/LFA-1 dependent in mice. Hepatology. 2005;42:1063–1071. doi: 10.1002/hep.20885. [DOI] [PubMed] [Google Scholar]
- Boisvert J, Kunkel EJ, Campbell JJ, Keeffe EB, Butcher EC, Greenberg HB. Liver-infiltrating lymphocytes in end-stage hepatitis C virus: subsets, activation status, and chemokine receptor phenotypes. J Hepatol. 2003;38:67–75. doi: 10.1016/s0168-8278(02)00328-8. [DOI] [PubMed] [Google Scholar]
- Bonder CS, Norman MU, Swain MG, Zbytnuik LD, Yamanouchi J, Santamaria P, Ajuebor M, Salmi M, Jalkanen S, Kubes P. Rules of recruitment for Th1 and Th2 lymphocytes in inflamed liver: a role for alpha-4 integrin and vascular adhesion protein-1. Immunity. 2005;23:153–163. doi: 10.1016/j.immuni.2005.06.007. [DOI] [PubMed] [Google Scholar]
- Borges E, Tietz W, Steegmaier M, Moll T, Hallmann R, Hamann A, Vestweber D. P-selectin glycoprotein ligand-1 (PSGL-1) on T helper 1 but not on T helper 2 cells binds to P-selectin and supports migration into inflamed skin. J Exp Med. 1997;185:573–578. doi: 10.1084/jem.185.3.573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bourgeois C, Rocha B, Tanchot C. A role for CD40 expression on CD8+ T cells in the generation of CD8+ T cell memory. Science. 2002;297:2060–2063. doi: 10.1126/science.1072615. [DOI] [PubMed] [Google Scholar]
- Bowen DG, Zen M, Holz L, Davis T, McCaughan GW, Bertolino P. The site of primary T cell activation is a determinant of the balance between intrahepatic tolerance and immunity. J Clin Invest. 2004;114:701–712. doi: 10.1172/JCI21593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cattaneo M. Aspirin and clopidogrel: efficacy, safety, and the issue of drug resistance. Arterioscler Thromb Vasc Biol. 2004;24:1980–1987. doi: 10.1161/01.ATV.0000145980.39477.a9. [DOI] [PubMed] [Google Scholar]
- Chosay JG, Fisher MA, Farhood A, Ready KA, Dunn CJ, Jaeschke H. Role of PECAM-1 (CD31) in neutrophil transmigration in murine models of liver and peritoneal inflammation. Am J Physiol. 1998;274:G776–G782. doi: 10.1152/ajpgi.1998.274.4.G776. [DOI] [PubMed] [Google Scholar]
- Crispe IN. Migration of lymphocytes into hepatic sinusoids. J Hepatol. 2012;57:218–220. doi: 10.1016/j.jhep.2011.12.035. [DOI] [PubMed] [Google Scholar]
- Curbishley SM, Eksteen B, Gladue RP, Lalor P, Adams DH. CXCR 3 activation promotes lymphocyte transendothelial migration across human hepatic endothelium under fluid flow. Am J Pathol. 2005;167:887–899. doi: 10.1016/S0002-9440(10)62060-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diacovo TG, Puri KD, Warnock RA, Springer TA, von Andrian UH. Platelet-mediated lymphocyte delivery to high endothelial venules. Science. 1996;273:252–255. doi: 10.1126/science.273.5272.252. [DOI] [PubMed] [Google Scholar]
- Diago M, Castellano G, García-Samaniego J, Pérez C, Fernández I, Romero M, Iacono OL, García-Monzón C. Association of pretreatment serum interferon gamma inducible protein 10 levels with sustained virological response to peginterferon plus ribavirin therapy in genotype 1 infected patients with chronic hepatitis C. Gut. 2006;55:374–379. doi: 10.1136/gut.2005.074062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dole VS, Bergmeier W, Mitchell HA, Eichenberger SC, Wagner DD. Activated platelets induce Weibel-Palade-body secretion and leukocyte rolling in vivo: role of P-selectin. Blood. 2005;106:2334–2339. doi: 10.1182/blood-2005-04-1530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dumoulin FL, Bach A, Leifeld L, El-Bakri M, Fischer HP, Sauerbruch T, Spengler U. Semiquantitative analysis of intrahepatic cytokine mRNAs in chronic hepatitis C. J Infect Dis. 1997;175:681–685. doi: 10.1093/infdis/175.3.681. [DOI] [PubMed] [Google Scholar]
- Eksteen B, Grant AJ, Miles A, Curbishley SM, Lalor PF, Hubscher SG, Briskin M, Salmon M, Adams DH. Hepatic endothelial CCL25 mediates the recruitment of CCR9+ gut-homing lymphocytes to the liver in primary sclerosing cholangitis. J Exp Med. 2004;200:1511–1517. doi: 10.1084/jem.20041035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elzey B, Tian J, Jensen R, Swanson A, Lees J, Lentz S, Stein C, Nieswandt B, Wang Y, Davidson B, et al. Platelet-mediated modulation of adaptive immunity. A communication link between innate and adaptive immune compartments. Immunity. 2003;19:9–19. doi: 10.1016/s1074-7613(03)00177-8. [DOI] [PubMed] [Google Scholar]
- Essani NA, Fisher MA, Simmons CA, Hoover JL, Farhood A, Jaeschke H. Increased P-selectin gene expression in the liver vasculature and its role in the pathophysiology of neutrophil-induced liver injury in murine endotoxin shock. J Leukoc Biol. 1998;63:288–296. doi: 10.1002/jlb.63.3.288. [DOI] [PubMed] [Google Scholar]
- Fox-Robichaud A, Kubes P. Molecular mechanisms of tumor necrosis factor alpha-stimulated leukocyte recruitment into the murine hepatic circulation. Hepatology. 2000;31:1123–1127. doi: 10.1053/he.2000.6961. [DOI] [PubMed] [Google Scholar]
- Geissmann F, Cameron TO, Sidobre S, Manlongat N, Kronenberg M, Briskin MJ, Dustin ML, Littman DR. Intravascular immune surveillance by CXCR6+ NKT cells patrolling liver sinusoids. PLoS Biol. 2005;3:e113. doi: 10.1371/journal.pbio.0030113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grabbe S, Varga G, Beissert S, Steinert M, Pendl G, Seeliger S, Bloch W, Peters T, Schwarz T, Sunderkötter C, et al. Beta2 integrins are required for skin homing of primed T cells but not for priming naive T cells. J Clin Invest. 2002;109:183–192. doi: 10.1172/JCI11703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grant AJ, Lalor PF, Hubscher SG, Briskin M, Adams DH. MAdCAM-1 expressed in chronic inflammatory liver disease supports mucosal lymphocyte adhesion to hepatic endothelium (MAdCAM-1 in chronic inflammatory liver disease) Hepatology. 2001;33:1065–1072. doi: 10.1053/jhep.2001.24231. [DOI] [PubMed] [Google Scholar]
- Guidotti L, Chisari F. Immunobiology and pathogenesis of viral hepatitis. Annu Rev Pathol. 2006;1:23–61. doi: 10.1146/annurev.pathol.1.110304.100230. [DOI] [PubMed] [Google Scholar]
- Harvey CE, Post JJ, Palladinetti P, Freeman AJ, Ffrench RA, Kumar RK, Marinos G, Lloyd AR. Expression of the chemokine IP-10 (CXCL10) by hepatocytes in chronic hepatitis C virus infection correlates with histological severity and lobular inflammation. J Leukoc Biol. 2003;74:360–369. doi: 10.1189/jlb.0303093. [DOI] [PubMed] [Google Scholar]
- Heydtmann M, Hardie D, Shields PL, Faint J, Buckley CD, Campbell JJ, Salmon M, Adams DH. Detailed analysis of intrahepatic CD8 T cells in the normal and hepatitis C-infected liver reveals differences in specific populations of memory cells with distinct homing phenotypes. J Immunol. 2006;177:729–738. doi: 10.4049/jimmunol.177.1.729. [DOI] [PubMed] [Google Scholar]
- Heydtmann M, Lalor PF, Eksteen JA, Hubscher SG, Briskin M, Adams DH. CXC chemokine ligand 16 promotes integrin-mediated adhesion of liver-infiltrating lymphocytes to cholangiocytes and hepatocytes within the inflamed human liver. J Immunol. 2005;174:1055–1062. doi: 10.4049/jimmunol.174.2.1055. [DOI] [PubMed] [Google Scholar]
- Hynes RO. Integrins: versatility, modulation, and signaling in cell adhesion. Cell. 1992;69:11–25. doi: 10.1016/0092-8674(92)90115-s. [DOI] [PubMed] [Google Scholar]
- Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell. 2002;110:673–687. doi: 10.1016/s0092-8674(02)00971-6. [DOI] [PubMed] [Google Scholar]
- Iannacone M, Sitia G, Guidotti LG. Pathogenetic and antiviral immune responses against hepatitis B virus. Future Virology. 2006;1:189–196. [Google Scholar]
- Iannacone M, Sitia G, Guidotti LG. On the role of platelets in the pathogenesis of viral hepatitis. J Hepatol. 2009;51:599–600. doi: 10.1016/j.jhep.2009.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iannacone M, Sitia G, Isogawa M, Marchese P, Castro MG, Lowenstein PR, Chisari FV, Ruggeri ZM, Guidotti LG. Platelets mediate cytotoxic T lymphocyte-induced liver damage. Nat Med. 2005;11:1167–1169. doi: 10.1038/nm1317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iannacone M, Sitia G, Isogawa M, Whitmire JK, Marchese P, Chisari FV, Ruggeri ZM, Guidotti LG. Platelets prevent IFN-alpha/beta-induced lethal hemorrhage promoting CTL-dependent clearance of lymphocytic choriomeningitis virus. Proc Natl Acad Sci USA. 2008;105:629–634. doi: 10.1073/pnas.0711200105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iannacone M, Sitia G, Narvaiza I, Ruggeri ZM, Guidotti LG. Antiplatelet drug therapy moderates immune-mediated liver disease and inhibits viral clearance in mice infected with a replication-deficient adenovirus. Clin Vaccine Immunol. 2007a;14:1532–1535. doi: 10.1128/CVI.00298-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iannacone M, Sitia G, Ruggeri ZM, Guidotti LG. HBV pathogenesis in animal models: recent advances on the role of platelets. J Hepatol. 2007b;46:719–726. doi: 10.1016/j.jhep.2007.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iigo Y, Suematsu M, Higashida T, Oheda J, Matsumoto K, Wakabayashi Y, Ishimura Y, Miyasaka M, Takashi T. Constitutive expression of ICAM-1 in rat microvascular systems analyzed by laser confocal microscopy. Am J Physiol. 1997;273:H138–H147. doi: 10.1152/ajpheart.1997.273.1.H138. [DOI] [PubMed] [Google Scholar]
- John B, Crispe IN. Passive and active mechanisms trap activated CD8+ T cells in the liver. J Immunol. 2004;172:5222–5229. doi: 10.4049/jimmunol.172.9.5222. [DOI] [PubMed] [Google Scholar]
- Kakimi K, Lane TE, Wieland S, Asensio VC, Campbell IL, Chisari FV, Guidotti LG. Blocking chemokine responsive to gamma-2/interferon (IFN)-gamma inducible protein and monokine induced by IFN-gamma activity in vivo reduces the pathogenetic but not the antiviral potential of hepatitis B virus-specific cytotoxic T lymphocytes. J Exp Med. 2001;194:1755–1766. doi: 10.1084/jem.194.12.1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kansas GS. Selectins and their ligands: current concepts and controversies. Blood. 1996;88:3259–3287. [PubMed] [Google Scholar]
- Kunkel EJ, Boisvert J, Murphy K, Vierra MA, Genovese MC, Wardlaw AJ, Greenberg HB, Hodge MR, Wu L, Butcher EC, et al. Expression of the chemokine receptors CCR4, CCR5, and CXCR3 by human tissue-infiltrating lymphocytes. Am J Pathol. 2002;160:347–355. doi: 10.1016/S0002-9440(10)64378-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lalor PF, Edwards S, McNab G, Salmi M, Jalkanen S, Adams DH. Vascular adhesion protein-1 mediates adhesion and transmigration of lymphocytes on human hepatic endothelial cells. J Immunol. 2002;169:983–992. doi: 10.4049/jimmunol.169.2.983. [DOI] [PubMed] [Google Scholar]
- Lalor PF, Sun PJ, Weston CJ, Martin-Santos A, Wakelam MJO, Adams DH. Activation of vascular adhesion protein-1 on liver endothelium results in an NF-kappaB-dependent increase in lymphocyte adhesion. Hepatology. 2007;45:465–474. doi: 10.1002/hep.21497. [DOI] [PubMed] [Google Scholar]
- Lang PA, Contaldo C, Georgiev P, El-Badry AM, Recher M, Kurrer M, Cervantes-Barragan L, Ludewig B, Calzascia T, Bolinger B, et al. Aggravation of viral hepatitis by platelet-derived serotonin. Nat Med. 2008;14:756–761. doi: 10.1038/nm1780. [DOI] [PubMed] [Google Scholar]
- Larrubia JR, Calvino M, Benito S, Sanz-de-Villalobos E, Perna C, Pérez-Hornedo J, González-Mateos F, García-Garzón S, Bienvenido A, Parra T. The role of CCR5/CXCR3 expressing CD8+ cells in liver damage and viral control during persistent hepatitis C virus infection. J Hepatol. 2007;47:632–641. doi: 10.1016/j.jhep.2007.04.009. [DOI] [PubMed] [Google Scholar]
- Lee WY, Kubes P. Leukocyte adhesion in the liver: distinct adhesion paradigm from other organs. J Hepatol. 2008;48:504–512. doi: 10.1016/j.jhep.2007.12.005. [DOI] [PubMed] [Google Scholar]
- Leroy V, Vigan I, Mosnier JF, Dufeu-Duchesne T, Pernollet M, Zarski JP, Marche PN, Jouvin-Marche E. Phenotypic and functional characterization of intrahepatic T lymphocytes during chronic hepatitis C. Hepatology. 2003;38:829–841. doi: 10.1053/jhep.2003.50410. [DOI] [PubMed] [Google Scholar]
- Li N. Platelet-lymphocyte cross-talk. J Leukoc Biol. 2008;83:1069–1078. doi: 10.1189/jlb.0907615. [DOI] [PubMed] [Google Scholar]
- Ludwig R, Schultz J, Boehncke W, Podda M, Tandi C, Krombach F, Baatz H, Kaufmann R, von Andrian U, Zollner T. Activated, not resting, platelets increase leukocyte rolling in murine skin utilizing a distinct set of adhesion molecules. J Invest Dermatol. 2004;122:830–836. doi: 10.1111/j.0022-202X.2004.22318.x. [DOI] [PubMed] [Google Scholar]
- Luo BH, Carman CV, Springer TA. Structural basis of integrin regulation and signaling. Annu Rev Immunol. 2007;25:619–647. doi: 10.1146/annurev.immunol.25.022106.141618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mcdonald B, McAvoy EF, Lam F, Gill V, de la Motte C, Savani RC, Kubes P. Interaction of CD44 and hyaluronan is the dominant mechanism for neutrophil sequestration in inflamed liver sinusoids. J Exp Med. 2008;205:915–927. doi: 10.1084/jem.20071765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McNab G, Reeves JL, Salmi M, Hubscher S, Jalkanen S, Adams DH. Vascular adhesion protein 1 mediates binding of T cells to human hepatic endothelium. Gastroenterology. 1996;110:522–528. doi: 10.1053/gast.1996.v110.pm8566600. [DOI] [PubMed] [Google Scholar]
- Meunier S, Rapetti L, Beziaud L, Pontoux C, Legrand A, Tanchot C. Synergistic CD40 signaling on APCs and CD8 T cells drives efficient CD8 response and memory differentiation. J Leukoc Biol. 2012;91:859–869. doi: 10.1189/jlb.0611292. [DOI] [PubMed] [Google Scholar]
- Muller WA, Weigl SA, Deng X, Phillips DM. PECAM-1 is required for transendothelial migration of leukocytes. J Exp Med. 1993;178:449–460. doi: 10.1084/jem.178.2.449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murai M, Yoneyama H, Ezaki T, Suematsu M, Terashima Y, Harada A, Hamada H, Asakura H, Ishikawa H, Matsushima K. Peyer’s patch is the essential site in initiating murine acute and lethal graft-versus-host reaction. Nat Immunol. 2003;4:154–160. doi: 10.1038/ni879. [DOI] [PubMed] [Google Scholar]
- Murai M, Yoneyama H, Harada A, Yi Z, Vestergaard C, Guo B, Suzuki K, Asakura H, Matsushima K. Active participation of CCR5(+)CD8(+) T lymphocytes in the pathogenesis of liver injury in graft-versus-host disease. J Clin Invest. 1999;104:49–57. doi: 10.1172/JCI6642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakamoto Y, Guidotti LG, Kuhlen CV, Fowler P, Chisari FV. Immune pathogenesis of hepatocellular carcinoma. J Exp Med. 1998;188:341–350. doi: 10.1084/jem.188.2.341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakamoto Y, Suda T, Momoi T, Kaneko S. Different procarcinogenic potentials of lymphocyte subsets in a transgenic mouse model of chronic hepatitis B. Cancer Res. 2004;64:3326–3333. doi: 10.1158/0008-5472.can-03-3817. [DOI] [PubMed] [Google Scholar]
- Northfield JW, Kasprowicz V, Lucas M, Kersting N, Bengsch B, Bengsh B, Kim A, Phillips RE, Walker BD, Thimme R, et al. CD161 expression on hepatitis C virus-specific CD8+ T cells suggests a distinct pathway of T cell differentiation. Hepatology. 2008;47:396–406. doi: 10.1002/hep.22040. [DOI] [PubMed] [Google Scholar]
- Rot A, von Andrian UH. Chemokines in innate and adaptive host defense: basic chemokinese grammar for immune cells. Annu Rev Immunol. 2004;22:891–928. doi: 10.1146/annurev.immunol.22.012703.104543. [DOI] [PubMed] [Google Scholar]
- Ruggeri ZM. Platelets in atherothrombosis. Nat Med. 2002;8:1227–1234. doi: 10.1038/nm1102-1227. [DOI] [PubMed] [Google Scholar]
- Ruggeri ZM. Platelet adhesion under flow. Microcirculation (New York, NY: 1994) 2009;16:58–83. doi: 10.1080/10739680802651477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salmi M, Jalkanen S. Human vascular adhesion protein 1 (VAP-1) is a unique sialoglycoprotein that mediates carbohydrate-dependent binding of lymphocytes to endothelial cells. J Exp Med. 1996;183:569–579. doi: 10.1084/jem.183.2.569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salmi M, Tohka S, Berg EL, Butcher EC, Jalkanen S. Vascular adhesion protein 1 (VAP-1) mediates lymphocyte subtype-specific, selectin-independent recognition of vascular endothelium in human lymph nodes. J Exp Med. 1997;186:589–600. doi: 10.1084/jem.186.4.589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sato T, Habtezion A, Beilhack A, Schulz S, Butcher E, Thorlacius H. Short-term homing assay reveals a critical role for lymphocyte function-associated antigen-1 in the hepatic recruitment of lymphocytes in graft-versus-host disease. J Hepatol. 2006;44:1132–1140. doi: 10.1016/j.jhep.2005.11.042. [DOI] [PubMed] [Google Scholar]
- Sato T, Thorlacius H, Johnston B, Staton TL, Xiang W, Littman DR, Butcher EC. Role for CXCR6 in recruitment of activated CD8+ lymphocytes to inflamed liver. J Immunol. 2005;174:277–283. doi: 10.4049/jimmunol.174.1.277. [DOI] [PubMed] [Google Scholar]
- Shields PL, Morland CM, Salmon M, Qin S, Hubscher SG, Adams DH. Chemokine and chemokine receptor interactions provide a mechanism for selective T cell recruitment to specific liver compartments within hepatitis C-infected liver. J Immunol. 1999;163:6236–6243. [PubMed] [Google Scholar]
- Sitia G, Aiolfi R, Di Lucia P, Mainetti M, Fiocchi A, Mingozzi F, Esposito A, Ruggeri ZM, Chisari FV, Iannacone M, et al. Antiplatelet therapy prevents hepatocellular carcinoma and improves survival in a mouse model of chronic hepatitis B. Proc Natl Acad Sci USA. 2012;109:E2165–E2172. doi: 10.1073/pnas.1209182109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sitia G, Iannacone M, Aiolfi R, Isogawa M, van Rooijen N, Scozzesi C, Bianchi ME, von Andrian UH, Chisari FV, Guidotti LG. Kupffer Cells Hasten Resolution of Liver Immunopathology in Mouse Models of Viral Hepatitis. PLoS Pathog. 2011;7:e1002061. doi: 10.1371/journal.ppat.1002061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Springer TA. Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm. Cell. 1994;76:301–314. doi: 10.1016/0092-8674(94)90337-9. [DOI] [PubMed] [Google Scholar]
- Tan SM. The leucocyte β2 (CD18) integrins: the structure, functional regulation and signalling properties. Biosci Rep. 2012;32:241–269. doi: 10.1042/BSR20110101. [DOI] [PubMed] [Google Scholar]
- Vieira-de-Abreu A, Campbell RA, Weyrich AS, Zimmerman GA. Platelets: versatile effector cells in hemostasis, inflammation, and the immune continuum. Semin Immunopathol. 2012;34:5–30. doi: 10.1007/s00281-011-0286-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Volpes R, Van Den Oord JJ, Desmet VJ. Vascular adhesion molecules in acute and chronic liver inflammation. Hepatology. 1992;15:269–275. doi: 10.1002/hep.1840150216. [DOI] [PubMed] [Google Scholar]
- Warren A, Le Couteur DG, Fraser R, Bowen DG, McCaughan GW, Bertolino P. T lymphocytes interact with hepatocytes through fenestrations in murine liver sinusoidal endothelial cells. Hepatology. 2006;44:1182–1190. doi: 10.1002/hep.21378. [DOI] [PubMed] [Google Scholar]
- Weyrich A, Zimmerman G. Platelets: signaling cells in the immune continuum. Trends Immunol. 2004;25:489–495. doi: 10.1016/j.it.2004.07.003. [DOI] [PubMed] [Google Scholar]
- Wisse E, De Zanger RB, Charels K, Van Der Smissen P, McCuskey RS. The liver sieve: considerations concerning the structure and function of endothelial fenestrae, the sinusoidal wall and the space of Disse. Hepatology. 1985;5:683–692. doi: 10.1002/hep.1840050427. [DOI] [PubMed] [Google Scholar]
- Wong J, Johnston B, Lee SS, Bullard DC, Smith CW, Beaudet AL, Kubes P. A minimal role for selectins in the recruitment of leukocytes into the inflamed liver microvasculature. J Clin Invest. 1997;99:2782–2790. doi: 10.1172/JCI119468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zlotnik A, Yoshie O. The Chemokine Superfamily Revisited. Immunity. 2012;36:705–716. doi: 10.1016/j.immuni.2012.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]