Abstract
Programmed Death-1 (PD-1)/Programmed Death-Ligand 1 (PD-L1) interactions are critical for dampening the immune response to both self and foreign antigens. The signaling of PD-L1 via its cytoplasmic domain, rather than through its interactions with PD-1 via the extracellular domain, has been termed PD-L1 reverse signaling. While this signaling is beneficial for cancer progression, little is understood about the consequences of PD-L1 reverse signaling in immune cells that express PD-L1 at steady state or in response to infection. Loss of PD-L1 during infection leads to unchecked T cell proliferation and increased autoimmune T cell responses. While the T cell intrinsic role of PD-1 for inhibiting T cell responses has been well explored, little to no effort has been directed at investigating the consequences of PD-L1 reverse signaling on the DCs interacting with PD-1+ T cells. We recently reported a defect in dendritic cell trafficking from the skin to the draining lymph node following immunization or infection in the absence of PD-L1. We demonstrated that a region within the cytoplasmic tail was responsible for the defect in dendritic cell trafficking. Here, we review the processes involved in dendritic cell trafficking and highlight what we know about PD-L1 expression, PD-L1 post-translational modifications, PD-L1 intracellular interactions and PD-L1 extracellular interactions.
Keywords: Dendritic cell migration, Dendritic cell trafficking, PD-L1 reverse signaling, CD80 signaling, interferon
Graphical abstract
PD-L1 forward/reverse signals influence cellular functions. PD-L1 can be expressed by cancer cells to prevent T cell activity or to promote cancer cell survival via PD-L1 reverse signaling. PD-L1 can be expressed by many cells in chronic infection/autoimmune disease and promote T cell exhaustion/dysfunction and is highly expressed by dendritic cells during infection, promoting dendritic cell migration from the skin to the lymph node via reverse signaling. Created with BioRender.com
PD-L1 interacts with PD-1 to inhibit the T cell response.
Our current understanding of PD-L1/PD-1 signaling comes largely from normal regulation of the immune response. During infection PD-1 is upregulated and remains high on effector T cells while PD-L1 is upregulated on macrophages and dendritic cells. PD-1 and PD-L1 interactions have been demonstrated to be critically important for mitigating T cell responses and inducing immune cell tolerance [1–13]. This interaction has been well documented to down modulate the expression of the T cell receptor (TCR) [14] and the cell cycle to dampen the T cell response via direct PD-1 signaling into the T cell [15–17]. In instances of cancer or chronic infection PD-1 and PD-L1 expression is often dysregulated resulting in immune dysfunction and chronic exhaustion [15–21]. As such, PD-1, PD-L1 and PD-L2 are designated as part of an immune checkpoint pathway which limits T cell responses. High expression of PD-L1 has been observed in tumors and demonstrates the immune inhibitory microenviroment within the tumor. PD-1 and PD-L1 have been targeted extensively with immunotherapies [22]. These immunotherapies are designed to block PD-1/PD-L1 interactions to promote T cell responses against the tumor and have been successful in patients.
Programmed death ligand 1 (PD-L1) reverse signaling.
Many of the studies described in the above section have documented the importance of PD-L1 as the ligand for PD-1 and demonstrated the signaling events within T cells that are mediated by this interaction. However, over 10 years ago it was demonstrated that PD-L1 reverse signaling could act as a molecular shield against cytotoxic T cells in vitro [23]. This study showed that removal of the cytoplasmic domain of PD-L1 in target cells left them vulnerable to killing by CD8 T cells [23]. Following this finding, several investigators began to identify targets of PD-L1 reverse signaling in cancer cells [24]. In these studies, PD-L1 was shown to mediate the epithelial to mesenchymal transition through the RAS/ERK/MEK pathway [25], AKT phosphorylation was demonstrated in PD-L1hi compared to PD-L1lo melanoma cells during autophagy [26] and more recently STAT3 phosphorylation in the context of type 1 interferon (IFN) was shown to protect melanoma cells from apoptosis [27]. In glioblastoma multiforme (GBM), PD-L1, via its cytoplasmic domain, was shown to interact with H-RAS [25]. This interaction resulted in MAPK activation and lead to enhanced GBM motility and invasion [25]. This study provided the first evidence that loss of PD-L1 expression by a cancer cell could not only protect the cell from death, but could also promote cellular movement and metastasis. Thus far, PD-L1 reverse signaling has largely been studied in cancer cells and has significantly improved our understanding of how the intracellular domain of PD-L1 can impact cellular responses. In recent work performed by our lab, we demonstrated that normal lymphatic endothelial cell (LEC) expression of PD-L1 also promotes LEC survival during viral infection or polyI:C injection [28]. We went on to investigate the requirement for PD-L1 reverse signaling in dendritic cells and the involvement of PD-L1 in dendritic cell (DC) migration from the skin to the draining lymph node (LN) in the setting of viral infection or polyI:C injection [29]. In this review we will focus less on the implications of PD-L1 expression by cancer cells (reviewed in [24]) and instead focus on what is and is unknown about how PD-L1 reverse signaling and PD-L1 interactions may affect dendritic cell migration and subsequent T cell activation.
Dermal dendritic cell (DC) migration through lymphatics.
Upon infection, tissue resident DCs must leave the tissue and migrate to the LN for initiation of the adaptive immune response. Disruption of E-cadherin junctions between DCs and keratinocytes, DC release of matrix metalloproteinases (MMP)2 and 9, downregulation of tissue inhibitors of metalloproteinases (TIMP) [30–32] and digestion of large hyaluronan (HA) non-sulfated glycosaminoglycans are required for tissue release [33] (Figure 1). These processes occur concurrently with the upregulation of CC chemokine receptor (CCR)7 and major histocompatibility complex (MHC) Class II [34]. As the DCs migrate through the extracellular matrix (ECM) via “amoeboid movement” to the lymphatic capillaries, the DCs are guided by CC chemokine ligand (CCL)21 secreted from the lymphatics [35, 36]. Once the DCs reach the lymphatic capillaries they enter at specialized transmigratory cups coated with intracellular adhesion molecule 1 (ICAM1), vascular cellular adhesion molecule 1 (VCAM1) and lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1) where local release of CCL21 facilitates DC transit [37–40](Figure 1). The DCs are guided by lymphatic derived chemotactic CCL21 gradients, until they reach the downstream lymphatic collector where they are propelled to the lymph node by lymphatic flow [41]. Once in the LN the conventional DC subsets, cDC1 and cDC2 are further guided by CCL19 and CCL21 expressed by the fibroblastic reticular network [42–44] (Figure 1). As such, XCR1+ cDC1s express the most CCR7 and travel the furthest to interact with CD8 T cells deep in the cortex [45] while XCR1− CD11b+ cDC2s remain near the B cell follicles and interact predominantly with CD4+ T cells [46]. The migration of antigen bearing dermal DCs from the dermis through the lymphatics is required to prime naïve T cells [47–51]. This is specifically important in response to localized skin infections like Leishmania [52], bacterial infections like methicillin-resistant Staphylococcus aureus (MRSA)[53] and pox viruses[54] which require DC trafficking to prime naïve T cells in the lymph node [47]. Indeed, DC migration to the LN is of critical importance for immune system activation in response to most bacterial infections (400-2000nm) as they are too large to pass through the lymphatic capillaries [49]. However, protein immunization (30-50kD) and virus given subcutaneously [55] pass through the lymphatics directly to the LN resulting in antigen presentation by LN resident cDCs [49]. DC migration is also required to interact with antigen bearing LECs in the subcapsular sinus of the LN [56, 57] where the process of antigen exchange occurs for the benefit of protective immunity [56, 57].
Figure 1. Dendritic cell migration from the skin or tissue to the draining lymph node.
Upon infection with a pathogen dendritic cells (DC) become activated and must leave the tissue in order to present antigens to naïve T cells within the draining lymph node (LN) . DC migration from the skin requires the release of dermal DCs from the extracellular matrix (ECM) via matrix metalloproteinases (MMP) [30–32] and hyaluronidase (HA) [33]. The DCs upregulate the chemokine receptor CCR7 and migrate toward the chemokine ligand CCL21 produced by the lymphatic capillaries comprised of oak leaf shaped LECs with highly permeable button like junctions [35, 36]. Upon reaching the lymphatic capillaries the DCs enter at transmigratory cups via interactions with integrins an cellular adhesion molecules such as ICAM1 and VCAM1. DC entry also requires interactions with LYVE-1 expressed by the lymphatic endothelial cells via interaction with Hyaluronan on the DC [37–40]. Once the DCs have squeezed through the lymphatic capillaries via actin myosin contractility they crawl along the luminal surface of the lymphatic capillaries until they enter the collecting vessels where they are propelled by lymphatic flow. Entry into the LN cortex where the T cells reside involves the expression of CCL19 and CCL21 by the fibroblastic reticular cell network [42–44].
DC activation and PD-L1.
The process of DC migration described above occurs following immunization or infection and occurs concurrently with DC activation [58, 59]. This activation includes, in addition to the upregulation of CCR7 and MHC class II, upregulation of CD80, CD86 and CD40 [34]. Upregulation of the activation markers and PD-L1 expression on DCs can result of IFN receptor signaling [60–62]. Migratory DCs present antigens acquired in the tissue providing at least three signals to initiate a productive T cell response. These three signals include recognition of peptide MHC by the T cell receptor, CD28-CD80/86 (B7.1/2) interactions between the T cell and DC, and cytokine production by the DC (IL-12, CD70) [63–65]. PD-L1 (B7-H1) and other B7-H family members B7-H3 and B7-H4 act as co-inhibitory receptors and unlike CD80/86 (B7.1/2), B7-H family members can impede T cell responses [66, 67]. As such, loss of PD-L1 during infection leads to unchecked T cell proliferation and increased autoimmune T cell responses [15–17]. Whether PD-L1 intracellular signaling in DCs also affects T cell responses has not been well explored.
PD-L1 expression in the steady state is limited to cDC2s while PD-L1 expression is induced on both cDC1 and cDC2s [60] during infection as a consequence of induction of IFNα/β, IFNγ or tumor necrosis factor (TNF)α. As a result of IFN receptor engagement, IRF1 and STAT3 can localize to the PD-L1 promoter [61, 62, 68] (Figure 2). PD-L1 upregulation occurs within hours in response to lipopolysaccharide (LPS) or polyI:C on DCs [29, 60]. Why PD-L1 upregulation by DCs occurs early on has been elusive, however recent reports have demonstrated that CD80 interactions with PD-L1 prevent PD-1 access to PD-L1 [9, 12, 13, 69]. These reports highlight how CD80 - PD-L1 extracellular interactions could block PD-L1 interaction with PD-1 to promote T cell activation. Further, these studies demonstrated that in the absence of DC activation, and thus in the absence of CD80, PD-L1 - PD-1 interactions took place and T cells were de-activated. Interestingly, cell types that do not express CD80 also upregulate PD-L1 in response to IFNα/β, IFNγ, and TNFα [3, 28, 70]. One cell type of particular interest to DC activation and migration is the lymphatic endothelial cell (LEC). As mentioned above DC migration from the skin to the draining lymph node must occur through the lymphatic vasculature which is comprised of LECs. Following immune activation lymphatic vessels must expand and remodel to allow for DC entry and trafficking into the draining lymph node [71, 72]. LECs immediately and dramatically upregulate PD-L1 as a result of type 1 and type 2 IFN signaling [3, 28, 70]. How PD-L1 upregulation by LECs during infection or immunization may influence lymphatic remodeling and DC migration is not yet understood. The requirement for PD-L1 upregulation on cell types that interact with DCs may be an important path of investigation to truly understand the consequences of PD-L1 reverse signaling during the initial stages of an immune response.
Figure 2. Dendritic cell signaling events during activation.
Upon reception of type 1 IFNs dendritic cells will upregulate the transcription of CCR7, CD80, CD86, MHC Class II and PD-L1 [58, 59] [34]. Activation of PD-L1 has been demonstrated to be through STAT3 and IRF1 [61, 62]. The upregulation of CD80, PD-L1 and CCR7 lead to chemokine receptor signaling events. CCR7, a G-protein coupled receptor, releases the Gα and Gβγ subunits of the large G-proteins. Gα activation leads to phospholipase C activity and ERK phosphorylation. PLC activity also results in calcium flux and and actin remodeling [78]. All of these activities are required for DC migration and are impeded by the loss of cytoplasmic PD-L1 [29]. How PD-L1 impedes PLC activity is unknown. The extracellular interaction of PD-L1 with CD80 could be an important extracellular interaction required for these migratory signaling events.
PD-L1 promotes dendritic cell migration.
As noted above, PD-L1 has been demonstrated in a glioblastoma multiforme cell line to increase migration and pseudopodia length compared to short hairpin (sh)-PD-L1 cells where PD-L1 expression was minimal [25]. The expression of PD-L1 was important for ERK phosphorylation as they showed decreased ERK phosphorylation in the sh-PD-L1 cells[25]. As dendritic cells express relatively high levels of PD-L1 which is dramatically upregulated following immunization or infection [29, 60] we investigated whether dendritic cell migration may also be affected by PD-L1 expression. Using both bone marrow chimeras to eliminate hematopoietic but not stromal PD-L1 expression followed by FITC painting or in vivo DC transfer assays we determined that loss of PD-L1 expression by DCs resulted in decreased DC migration from the skin to the draining lymph node [29]. This loss of DC migration in the absence of PD-L1 occurred in response to polyI:C and listeria monocytogenes, but was not affected by contact hypersensitivity [29]. To investigate whether the intracellular domain or the extracellular domain was contributing to defects in DC migration we created a mouse model in which just three residues, TSS, of the cytoplasmic domain were mutated to alanine [29]. The extracellular domain of PD-L1 remained intact and compared to the Pdl1−/− DCs, the cytoplasmic mutant Pdl1CyMt mouse had WT levels of CD80 and PD-L1 expression following polyI:C injection [29]. We showed that the Pdl1CyMt mice phenocopied the defect in DC migration we showed in the Pdl1−/− mice and again confirmed this defect was DC intrinsic as bone marrow derived DCs were unable to migrate in vitro or in vivo. While this was the first time that PD-L1 reverse signaling in DCs was demonstrated to have an effect on DC migration, many others had demonstrated significant impacts on T cell responses caused by PD-L1 forward signaling [1–13]. We demonstrated no differences in T cell priming in the Pdl1CyMt mouse compared to WT except in infections where DC migration was required [29]. The defect in DC migration was caused by the loss of chemokine receptor signaling. The exact mechanism of how PD-L1 reverse signaling in dendritic cells results in lost chemokine receptor signaling or why DC migration is only impeded following interferon inducing stimuli (polyI:C, LPS, listeria, but not contact hypersensitivity) is not yet resolved, however, several considerations are outlined below.
Cytokine signaling and PD-L1.
Type 1 IFN induction in DCs occurs following viral infection, listeria infection and vaccination with several different adjuvants that signal through TLR3, 4, 7, 9, among others. Type 1 IFN signals are received via the IFNαβ receptor which results in activation of JAK1 and TYK2 causing phosphorylation of the receptor (reviewed in [73]). Phosphorylation of the IFNαβR recruits and activates STAT1 via phosphorylation and the subsequent upregulation of interferon regulatory factor (IRF) and interferon stimulated genes (ISG) [73]. To prevent over activation of an IFN response, STAT3 activation, also a result of IFN receptor signaling, triggers the transcription of Suppressor of Cytokine Signaling (SOCS) proteins. Of the SOCS proteins, SOCS3 is upregulated by IL-10 while SOCS1 becomes upregulated by IL-6 [74]. Induction of the SOCS proteins results in a positive feedback loop, causing increased expression of IL-6 or IL-10 while decreasing IFNαβR expression [74]. Increased IL-6 and IL-10 expression both lead to STAT3 phosphorylation. In Pdl1−/− mice, mice with a cytoplasmic mutation in PD-L1, or cells lacking PD-L1; STAT3, but not STAT1 phosphorylation is significantly increased compared to WT as a result of type 1 IFN receptor signaling [27, 29]. The mechanism behind this difference in STAT3 phosphorylation in the absence of PD-L1 reverse signaling is not understood. Furthermore, whether these differences in STAT3 phosphorylation are a result of increased IL6 or IL10 expression or secretion, as a result of PD-L1 reverse signaling, remains to be investigated.
Interestingly, several reports have demonstrated that type 1 IFN can inhibit DC migration from the skin to the draining LN [75–77]. In one of these reports IFNβ treatment decreased DC migration as a result of decreased CCR7 and MMP9 expression [76]. The decrease in CCR7 and MMP9 expression was a result of STAT1 upregulation by IFNβ [76]. As PD-L1 had been shown to be protective against IFNβ induced cell death it was possible that PD-L1 could act to modulate IFNβ regulation of CCR7 expression. In the absence of PD-L1 no differences in CCR7 expression were observed following polyI:C injection in vivo or after LPS stimulation in vitro when DC migration was impeded [29]. Together this predicts that PD-L1 could act as a rheostat for either type 1 IFN signaling or STAT signaling. This rheostat activity could be direct or via manipulation of the expression of other cytokines that downregulate IFN signaling, such as IL-6 or IL-10, as described above. Further investigation as to how loss of migration could occur revealed that downstream chemokine receptor signaling in the Pdl1−/− or Pdl1CyMt mouse was impeded, in contrast to S1P1R [29]. Chemokine receptors are G protein coupled receptors that release and activate Gα and Gβγ subunits upon ligation. Activation and release of the Gα subunit causes PLC activation and downstream MAPK activity, calcium flux and actin polymerization while Gβγ activation and release leads to PI3K activity and changes in cell viability [78] (Figure 2). Mutation of the TSS region of the cytoplasmic domain of PD-L1 led to decreased chemokine receptor mediated PLC activation, ERK phosphorylation and actin polymerization [29]. How exactly PD-L1 promotes chemokine signaling following DC activation and migration by polyI:C compared to contact hypersensitivity is still unclear. However, it remains a possibility that the modifications within the cytoplasmic tail of PD-L1 could contribute to stabilization of the chemokine receptor, the Gα protein or PLC via direct or indirect interactions at the membrane (Figure 2). Further, PD-L1 may regulate type 1 IFN receptor signaling thereby changing the cytokine milieu and modulating the DC chemokine receptor signaling events. Finally, while CCR7 surface and transcript expression was normal in the absence of PD-L1 reverse signaling it is possible that CCR7 localization is impaired. Thus, understanding what interacts with PD-L1, and what modifications occur within the cytoplasmic domain, in different cell types will be critical to our understanding of PD-L1 reverse signaling.
PD-L1 modifications and the cytoplasmic domain.
The murine cytoplasmic region of PD-L1 spans 30 amino acids from arginine (R) 271 to threonine (T) 290. The murine cytoplasmic domain is similar to the human with a lysine residue found at amino acid 280, immediately following a threonine-serine-serine (TSS) motif found at amino acids 277-279. PD-L1 stabilization is regulated by a cysteine residue, 5 amino acids upstream of the TSS motif in the cytoplasmic region of PD-L1, that is palmitoylated and anchored to the membrane [79] (Figure 3). Indeed, loss of this cysteine residue along with the 7 amino acids before it mimics loss of PD-L1 in response to type 1 IFN sensitivity in melanoma cells [27]. Additionally, PD-L1 mono- and poly-Ubiquitination occurs both before and after epidermal growth factor (EGF) stimulation. Predicted ubiquitination sites are at amino acids lysine (K) K178 and K281 in human PD-L1 [80]. Only residue K281 of the predicted ubiquitination sites in human PD-L1 resides in the cytoplasmic domain (Figure 3). Other modifications such as phosphorylation (T80,S184), acetylation and glycosylation (asparagine (N) 35, N192, N200, N219) have been shown, but primarily occur within the extracellular region of PD-L1 [81]. Intriguingly, phosphorylation of residue S284 in humans, homologous to murine S279, has been detected in several mass spectrometry studies in cancer cells and cancer cell lines [82–87] (Figure 3). While the implications of this phosphorylation have not been readily addressed, mutation of the TSS region in mouse has severe implications for DC migration while maintaining protein stability and expression [29]. Deletion of the DTSSK residues in melanoma cells appears to make them less sensitive to type 1 interferon induced cell death, however this mutation includes the ubiquitinated lysine reside which could have additional effects [27]. These findings implicate phosphorylation and/or ubiquitination of PD-L1 cytoplasmic residues in the regulation of motility and potentially survivability. Intriguingly, PD-L1 has been demonstrated to interact with RAS to induce ERK and MAPK signaling events in GBM cells[25]. It is still unclear which region or if the cytoplasmic domain of PD-L1 interacts with RAS in GBM cells or if PD-L1 interacts with RAS in dendritic cells. However, loss of the TSS domain significantly impeded ERK phosphorylation in dendritic cells [29]. Therefore, it seems likely that the short cytoplasmic domain of PD-L1 may signal via either or both post-translational modifications and/or direct protein-protein interactions.
Figure 3. PD-L1 domains.
A. Upon activation DCs upregulate CD80 and PD-L1 [34]. The interaction between PD-L1 and CD80 requires Y56 on PD-L1 and L107 on CD80 and does not impede the interaction between CD80 and the CDR3 region of CD28, but does prevent PD-L1 – PD-1 interactions [9, 12, 13, 69]. This is likely because CD80 and PD-1 interactions with PD-L1 require the Y56 residue of PD-L1 where mutation of PD-1 residues K78, I126 and E136 blocks interaction of PD-1 with PD-L1. Interaction of CD28 with CD80 leads to CD80 reverse signaling that causes increased IL-6 and IFNg production [93]. Stabilization of PD-L1 requires palmitoylation of the cysteine residue [79] within the cytoplasmic tail while actin polymerization and signaling events downstream of chemokine receptors that result in cellular migration require the TSS motif [29]. Phosphorylation and Ubiquitination are likely on the serine and lysine residues nearby, but their function is unknown [79]. The TSS region may also inhibit the RMLDVEFLKC domain to prevent STAT3 activation [27]. B. During de-activation of the DC CD80 becomes down regulated and PD-L1 is now allowed to interact with PD-1 [9, 12, 13, 69]. Whether PD-1 – PD-L1 interactions result in PD-L1 reverse signaling events is unknown. PD-1 ligation to PD-L1 results in downregulation of the T cell receptor and apoptosis.
PD-L1 extracellular interactions and reverse signaling.
Like CD80 (B7.1) and CD86 (B7.2), PD-L1 (B7-H1) is a B7 family member belonging to the immunoglobulin superfamily containing both IgC-like and IgV-like domains. These family members have limited homology and diverge dramatically in their cytoplasmic regions [88]. While CD80 and CD86 interact with CD28, CTLA4 and ICOS, PD-L1 interacts with PD-1 via PD-1 residues K78, I126 and E136 [89] and CD80 via residue L107 [9] and not CD28, CTLA4 or ICOS [90]. PD-L1 interactions with PD-1 are most well described to occur in trans with T cells, however, PD-L1 can also interact with PD-1 expressed on the DC in cis [12]. CD80 interactions with PD-L1 were demonstrated to only occur in cis on DCs via residue Y56 on PD-L1 and residue L107 on CD80 [9] (Figure 2,3). Evidence for this was provided several years ago now when CD80-PD-L1 interactions were evaluated by biacore analysis. This study found that only when CD80 was immobilized to a plate with a flexible linker was PD-L1 able to bind to CD80, demonstrating that PD-L1 interaction with CD80 likely did not occur between two cells [91]. Interestingly, reverse signaling through CD80 and CD86 as a result of CD28 interactions at the CDR3 region of CD28 [92] can lead to upregulation of interleukin (IL)-6 and IFNγ [93] to promote T cell responses (Figure 3). Whether PD-L1 interactions with CD80 also encourage the upregulation of IL-6 and IFNγ is still unknown. However, based on the similarity between B7 family members it is not surprising that cytoplasmic signaling events occur with any or all members. Still, it remains undefined whether interactions with the extracellular domain of PD-L1 can influence intracellular signaling events within dendritic cells. Indeed, we and others have demonstrated that loss of PD-L1 significantly impedes CD80 upregulation by dendritic cells [29, 94]. This upregulation appears to be dependent on the extracellular interaction of CD80 with PD-L1 rather than the intracellular interactions as mutation of a TSS motif within the cytoplasmic domain of PD-L1 does not impede CD80 expression or upregulation, but does impair DC migration [29]. Whether this loss of DC migration is related to IL6 or IFNγ mis-regulation is still unknown, but an intriguing path to pursue based on recent findings that IL6 interactions are necessary for DC migration [95].
As PD-L1-CD80 interactions occur in cis and block PD-L1-PD-1 interactions [9, 12, 13, 69] it is possible that upregulation of CD80 and PD-L1 on DCs could mediate the signaling events that occur downstream of PD-L1 (Figure 3). While extracellular regions of PD-L1 and CD80 interact, it has yet to be shown whether intracellular interactions between PD-L1 and CD80 occur [91]. If interactions between PD-L1 and CD80 are necessary for PD-L1 or CD80 reverse signaling, then inhibiting this interaction would impede PD-L1 or CD80 reverse signaling. If this was the case, PD-L1 antibodies, which block both PD-1 and CD80 interactions with PD-L1, would have significant consequences for T cell responses that require DC migration. However, it has been demonstrated that blocking PD-L1 interactions enhance T cell responses by maintaining T cell activation [5, 96, 97]. It remains to be seen whether blocking PD-L1 - CD80 interactions influence DC migration. However, in the event that CD80 – PD-L1 interactions are important for DC migration this could confound the observed T cell responses in the presence of PD-L1 blocking antibodies. Finally, what, if any, role PD-L1 reverse signaling has in memory T cell responses or during chronic infection is completely unexplored. Future studies will be important to parse out differences in PD-L1 forward and reverse signaling and could be performed using genetic mouse models where either the extracellular or intracellular domain of PD-L1 or the binding domains found within each are mutated.
Conclusions.
PD-L1, a relatively well-known transmembrane protein, has a complex array of functions that modulate immune responses. The hijacking of this protein by cancer cells thus has implications in different cellular functions from increasing cell viability to increasing migration and invasion. However, the function of PD-L1 reverse signaling in cells that constitutively express PD-L1 and dramatically upregulate PD-L1 during activation [28, 29, 60, 70] is not well understood. Our understanding of what PD-L1 interacts with and the post-translational modifications that occur during dendritic cell activation, aside from interactions with PD-1 and CD80 is insufficient. Therefore, to move this field forward, significant work must be done to evaluate both intracellular and extracellular events controlled by PD-L1, particularly in situations where blocking interactions are involved.
Acknowledgements:
I thank Matthew Burchill for critical reading of this manuscript. BT was supported by NIH awards R01AI121209, R01AI155474, R21AI155929, the University of Colorado Outstanding Early Career Scholar and RBI Clinical Scholar Award and the Waterman Family Foundation for Liver Research.
Abbreviations:
- PD-L1
programmed death ligand 1
- PD-1
programmed death 1
- TCR-T
cell receptor
- DC
dendritic cell
- cDC
conventional dendritic cell
- LEC
lymphatic endothelial cell
- RAS
rat sarcoma
- ERK
extracellular signal related kinase
- MEK
MAPK/ERK kinase
- AKT
Ak strain transforming
- STAT
signal transducer and activator or transcription
- IFN
interferon
- GBM
glioblastoma multiforme
- MAPK
mitogen activated protein kinase
- LN
lymph node
- MMP
matrix metalloproteinase
- CCR-C
C chemokine receptor
- ECM
extracellular matrix
- HA
hyaluronic acid
- ICAM
intercellular adhesion molecule
- VCAM
vascular cell adhesion molecule
- Lyve
lymphatic vessel endothelial hyaluronan receptor
- CCL-C
C chemokine ligad
- XCR1
X-C motif chemokine receptor
- MHC
major histocompatibility complex
- IRF
interferon regulatory factor
- WT
wild-type
- S1P1R
sphingosine-1-phosphate receptor 1
- PI3K
phosphatidylinositol-3 kinase
- PLC
phospholipase C
- TNF
tumor necrosis factor
- FITC
fluorescein isothiocyanate
- EGF
epidermal growth factor
- Ig
immunoglobulin
- IL-6
interleukin 6
- CDR3
complementarity-determining regions 3
- CTLA4
cytotoxic T-lymphocyte-associated protein 4
- ICOS
inducible T-cell costimulator
- CyMt
cytoplasmic mutant
Footnotes
Conflict of interest: I declare no conflicts of interst.
References:
- 1.Francisco LM, Sage PT & Sharpe AH (2010) The PD-1 pathway in tolerance and autoimmunity, Immunol Rev. 236, 219–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Grabie N, Gotsman I, DaCosta R, Pang H, Stavrakis G, Butte MJ, Keir ME, Freeman GJ, Sharpe AH & Lichtman AH (2007) Endothelial programmed death-1 ligand 1 (PD-L1) regulates CD8+ T-cell mediated injury in the heart, Circulation. 116, 2062–71. [DOI] [PubMed] [Google Scholar]
- 3.Lane RS, Femel J, Breazeale AP, Loo CP, Thibault G, Kaempf A, Mori M, Tsujikawa T, Chang YH & Lund AW (2018) IFNgamma-activated dermal lymphatic vessels inhibit cytotoxic T cells in melanoma and inflamed skin, J Exp Med. 215, 3057–3074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Mueller SN, Vanguri VK, Ha SJ, West EE, Keir ME, Glickman JN, Sharpe AH & Ahmed R (2010) PD-L1 has distinct functions in hematopoietic and nonhematopoietic cells in regulating T cell responses during chronic infection in mice, J Clin Invest. 120, 2508–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Peng Q, Qiu X, Zhang Z, Zhang S, Zhang Y, Liang Y, Guo J, Peng H, Chen M, Fu YX & Tang H (2020) PD-L1 on dendritic cells attenuates T cell activation and regulates response to immune checkpoint blockade, Nat Commun. 11, 4835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Rouhani SJ, Eccles JD, Riccardi P, Peske JD, Tewalt EF, Cohen JN, Liblau R, Makinen T & Engelhard VH (2015) Roles of lymphatic endothelial cells expressing peripheral tissue antigens in CD4 T-cell tolerance induction, Nat Commun. 6, 6771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Rouhani SJ, Eccles JD, Tewalt EF & Engelhard VH (2014) Regulation of T-cell Tolerance by Lymphatic Endothelial Cells, J Clin Cell Immunol. 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Shi J, Hou S, Fang Q, Liu X, Liu X & Qi H (2018) PD-1 Controls Follicular T Helper Cell Positioning and Function, Immunity. 49, 264–274 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sugiura D, Maruhashi T, Okazaki IM, Shimizu K, Maeda TK, Takemoto T & Okazaki T (2019) Restriction of PD-1 function by cis-PD-L1/CD80 interactions is required for optimal T cell responses, Science. 364, 558–566. [DOI] [PubMed] [Google Scholar]
- 10.Sun C, Mezzadra R & Schumacher TN (2018) Regulation and Function of the PD-L1 Checkpoint, Immunity. 48, 434–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Tewalt EF, Cohen JN, Rouhani SJ, Guidi CJ, Qiao H, Fahl SP, Conaway MR, Bender TP, Tung KS, Vella AT, Adler AJ, Chen L & Engelhard VH (2012) Lymphatic endothelial cells induce tolerance via PD-L1 and lack of costimulation leading to high-level PD-1 expression on CD8 T cells, Blood. 120, 4772–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zhao Y, Harrison DL, Song Y, Ji J, Huang J & Hui E (2018) Antigen-Presenting Cell-Intrinsic PD-1 Neutralizes PD-L1 in cis to Attenuate PD-1 Signaling in T Cells, Cell Rep. 24, 379–390 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zhao Y, Lee CK, Lin CH, Gassen RB, Xu X, Huang Z, Xiao C, Bonorino C, Lu LF, Bui JD & Hui E (2019) PD-L1:CD80 Cis-Heterodimer Triggers the Co-stimulatory Receptor CD28 While Repressing the Inhibitory PD-1 and CTLA-4 Pathways, Immunity. 51, 1059–1073 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Karwacz K, Bricogne C, MacDonald D, Arce F, Bennett CL, Collins M & Escors D (2011) PD-L1 co-stimulation contributes to ligand-induced T cell receptor down-modulation on CD8+ T cells, EMBO Mol Med. 3, 581–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Fife BT, Pauken KE, Eagar TN, Obu T, Wu J, Tang Q, Azuma M, Krummel MF & Bluestone JA (2009) Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal, Nat Immunol. 10, 1185–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Iwai Y, Terawaki S & Honjo T (2005) PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells, Int Immunol. 17, 133–44. [DOI] [PubMed] [Google Scholar]
- 17.Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, Hashimoto-Tane A, Azuma M & Saito T (2012) Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2, J Exp Med. 209, 1201–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, Kobayashi SV, Linsley PS, Thompson CB & Riley JL (2005) CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms, Molecular and cellular biology. 25, 9543–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Patsoukis N, Brown J, Petkova V, Liu F, Li L & Boussiotis VA (2012) Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation, Sci Signal. 5, ra46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Attanasio J & Wherry EJ (2016) Costimulatory and Coinhibitory Receptor Pathways in Infectious Disease, Immunity. 44, 1052–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bengsch B, Johnson AL, Kurachi M, Odorizzi PM, Pauken KE, Attanasio J, Stelekati E, McLane LM, Paley MA, Delgoffe GM & Wherry EJ (2016) Bioenergetic Insufficiencies Due to Metabolic Alterations Regulated by the Inhibitory Receptor PD-1 Are an Early Driver of CD8(+) T Cell Exhaustion, Immunity. 45, 358–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Jiang Y, Chen M, Nie H & Yuan Y (2019) PD-1 and PD-L1 in cancer immunotherapy: clinical implications and future considerations, Hum Vaccin Immunother. 15, 1111–1122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Azuma T, Yao S, Zhu G, Flies AS, Flies SJ & Chen L (2008) B7-H1 is a ubiquitous antiapoptotic receptor on cancer cells, Blood. 111, 3635–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Escors D, Gato-Canas M, Zuazo M, Arasanz H, Garcia-Granda MJ, Vera R & Kochan G (2018) The intracellular signalosome of PD-L1 in cancer cells, Signal Transduct Target Ther. 3, 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Qiu XY, Hu DX, Chen WQ, Chen RQ, Qian SR, Li CY, Li YJ, Xiong XX, Liu D, Pan F, Yu SB & Chen XQ (2018) PD-L1 confers glioblastoma multiforme malignancy via Ras binding and Ras/Erk/EMT activation, Biochim Biophys Acta Mol Basis Dis. 1864, 1754–1769. [DOI] [PubMed] [Google Scholar]
- 26.Clark CA, Gupta HB, Sareddy G, Pandeswara S, Lao S, Yuan B, Drerup JM, Padron A, Conejo-Garcia J, Murthy K, Liu Y, Turk MJ, Thedieck K, Hurez V, Li R, Vadlamudi R & Curiel TJ (2016) Tumor-Intrinsic PD-L1 Signals Regulate Cell Growth, Pathogenesis, and Autophagy in Ovarian Cancer and Melanoma, Cancer Res. 76, 6964–6974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Gato-Canas M, Zuazo M, Arasanz H, Ibanez-Vea M, Lorenzo L, Fernandez-Hinojal G, Vera R, Smerdou C, Martisova E, Arozarena I, Wellbrock C, Llopiz D, Ruiz M, Sarobe P, Breckpot K, Kochan G & Escors D (2017) PDL1 Signals through Conserved Sequence Motifs to Overcome Interferon-Mediated Cytotoxicity, Cell Rep. 20, 1818–1829. [DOI] [PubMed] [Google Scholar]
- 28.Lucas ED, Finlon JM, Burchill MA, McCarthy MK, Morrison TE, Colpitts TM & Tamburini BAJ (2018) Type 1 IFN and PD-L1 Coordinate Lymphatic Endothelial Cell Expansion and Contraction during an Inflammatory Immune Response, J Immunol. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lucas ED, Schafer JB, Matsuda J, Kraus M, Burchill MA & Tamburini BAJ (2020) PD-L1 Reverse Signaling in Dermal Dendritic Cells Promotes Dendritic Cell Migration Required for Skin Immunity, Cell Rep. 33, 108258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Darmanin S, Chen J, Zhao S, Cui H, Shirkoohi R, Kubo N, Kuge Y, Tamaki N, Nakagawa K, Hamada J, Moriuchi T & Kobayashi M (2007) All-trans retinoic acid enhances murine dendritic cell migration to draining lymph nodes via the balance of matrix metalloproteinases and their inhibitors, J Immunol. 179, 4616–25. [DOI] [PubMed] [Google Scholar]
- 31.Ratzinger G, Stoitzner P, Ebner S, Lutz MB, Layton GT, Rainer C, Senior RM, Shipley JM, Fritsch P, Schuler G & Romani N (2002) Matrix metalloproteinases 9 and 2 are necessary for the migration of Langerhans cells and dermal dendritic cells from human and murine skin, J Immunol. 168, 4361–71. [DOI] [PubMed] [Google Scholar]
- 32.Yen JH, Khayrullina T & Ganea D (2008) PGE2-induced metalloproteinase-9 is essential for dendritic cell migration, Blood. 111, 260–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Muto J, Morioka Y, Yamasaki K, Kim M, Garcia A, Carlin AF, Varki A & Gallo RL (2014) Hyaluronan digestion controls DC migration from the skin, J Clin Invest. 124, 1309–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Cabeza-Cabrerizo M, Cardoso A, Minutti CM, Pereira da Costa M & Reis ESC (2021) Dendritic Cells Revisited, Annu Rev Immunol. [DOI] [PubMed] [Google Scholar]
- 35.Lammermann T, Bader BL, Monkley SJ, Worbs T, Wedlich-Soldner R, Hirsch K, Keller M, Forster R, Critchley DR, Fassler R & Sixt M (2008) Rapid leukocyte migration by integrin-independent flowing and squeezing, Nature. 453, 51–5. [DOI] [PubMed] [Google Scholar]
- 36.Schumann K, Lammermann T, Bruckner M, Legler DF, Polleux J, Spatz JP, Schuler G, Forster R, Lutz MB, Sorokin L & Sixt M (2010) Immobilized chemokine fields and soluble chemokine gradients cooperatively shape migration patterns of dendritic cells, Immunity. 32, 703–13. [DOI] [PubMed] [Google Scholar]
- 37.Jackson DG (2019) Leucocyte Trafficking via the Lymphatic Vasculature-Mechanisms and Consequences, Front Immunol. 10, 471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Johnson LA, Banerji S, Lawrance W, Gileadi U, Prota G, Holder KA, Roshorm YM, Hanke T, Cerundolo V, Gale NW & Jackson DG (2017) Dendritic cells enter lymph vessels by hyaluronan-mediated docking to the endothelial receptor LYVE-1, Nat Immunol. 18, 762–770. [DOI] [PubMed] [Google Scholar]
- 39.Rouzaut A, Garasa S, Teijeira A, Gonzalez I, Martinez-Forero I, Suarez N, Larrea E, Alfaro C, Palazon A, Dubrot J, Hervas-Stubbs S & Melero I (2010) Dendritic cells adhere to and transmigrate across lymphatic endothelium in response to IFN-alpha, Eur J Immunol. 40, 3054–63. [DOI] [PubMed] [Google Scholar]
- 40.Teijeira A, Garasa S, Pelaez R, Azpilikueta A, Ochoa C, Marre D, Rodrigues M, Alfaro C, Auba C, Valitutti S, Melero I & Rouzaut A (2013) Lymphatic endothelium forms integrin-engaging 3D structures during DC transit across inflamed lymphatic vessels, J Invest Dermatol. 133, 2276–85. [DOI] [PubMed] [Google Scholar]
- 41.Schineis P, Runge P & Halin C (2019) Cellular traffic through afferent lymphatic vessels, Vascul Pharmacol. 112, 31–41. [DOI] [PubMed] [Google Scholar]
- 42.Bajenoff M, Egen JG, Koo LY, Laugier JP, Brau F, Glaichenhaus N & Germain RN (2006) Stromal cell networks regulate lymphocyte entry, migration, and territoriality in lymph nodes, Immunity. 25, 989–1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Forster R, Schubel A, Breitfeld D, Kremmer E, Renner-Muller I, Wolf E & Lipp M (1999) CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs, Cell. 99, 23–33. [DOI] [PubMed] [Google Scholar]
- 44.Link A, Vogt TK, Favre S, Britschgi MR, Acha-Orbea H, Hinz B, Cyster JG & Luther SA (2007) Fibroblastic reticular cells in lymph nodes regulate the homeostasis of naive T cells, Nat Immunol. 8, 1255–65. [DOI] [PubMed] [Google Scholar]
- 45.Eisenbarth SC (2019) Dendritic cell subsets in T cell programming: location dictates function, Nat Rev Immunol. 19, 89–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gerner MY, Casey KA, Kastenmuller W & Germain RN (2017) Dendritic cell and antigen dispersal landscapes regulate T cell immunity, J Exp Med. 214, 3105–3122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dejani NN, Brandt SL, Pineros A, Glosson-Byers NL, Wang S, Son YM, Medeiros AI & Serezani CH (2016) Topical Prostaglandin E Analog Restores Defective Dendritic Cell-Mediated Th17 Host Defense Against Methicillin-Resistant Staphylococcus Aureus in the Skin of Diabetic Mice, Diabetes. 65, 3718–3729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Johansen P, Storni T, Rettig L, Qiu Z, Der-Sarkissian A, Smith KA, Manolova V, Lang KS, Senti G, Mullhaupt B, Gerlach T, Speck RF, Bot A & Kundig TM (2008) Antigen kinetics determines immune reactivity, Proc Natl Acad Sci U S A. 105, 5189–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Manolova V, Flace A, Bauer M, Schwarz K, Saudan P & Bachmann MF (2008) Nanoparticles target distinct dendritic cell populations according to their size, Eur J Immunol. 38, 1404–13. [DOI] [PubMed] [Google Scholar]
- 50.Tomura M, Hata A, Matsuoka S, Shand FH, Nakanishi Y, Ikebuchi R, Ueha S, Tsutsui H, Inaba K, Matsushima K, Miyawaki A, Kabashima K, Watanabe T & Kanagawa O (2014) Tracking and quantification of dendritic cell migration and antigen trafficking between the skin and lymph nodes, Sci Rep. 4, 6030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Chu CC, Di Meglio P & Nestle FO (2011) Harnessing dendritic cells in inflammatory skin diseases, Semin Immunol. 23, 28–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Leon B, Lopez-Bravo M & Ardavin C (2007) Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania, Immunity. 26, 519–31. [DOI] [PubMed] [Google Scholar]
- 53.Wu X & Xu F (2014) Dendritic cells during Staphylococcus aureus infection: subsets and roles, J Transl Med. 12, 358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Seneschal J, Jiang X & Kupper TS (2014) Langerin+ dermal DC, but not Langerhans cells, are required for effective CD8-mediated immune responses after skin scarification with vaccinia virus, J Invest Dermatol. 134, 686–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hickman HD, Takeda K, Skon CN, Murray FR, Hensley SE, Loomis J, Barber GN, Bennink JR & Yewdell JW (2008) Direct priming of antiviral CD8+ T cells in the peripheral interfollicular region of lymph nodes, Nat Immunol. 9, 155–65. [DOI] [PubMed] [Google Scholar]
- 56.Kedl RM, Lindsay RS, Finlon JM, Lucas ED, Friedman RS & Tamburini BAJ (2017) Migratory dendritic cells acquire and present lymphatic endothelial cell-archived antigens during lymph node contraction, Nat Commun. 8, 2034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Tamburini BA, Burchill MA & Kedl RM (2014) Antigen capture and archiving by lymphatic endothelial cells following vaccination or viral infection, Nat Commun. 5, 3989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Luft T, Luetjens P, Hochrein H, Toy T, Masterman KA, Rizkalla M, Maliszewski C, Shortman K, Cebon J & Maraskovsky E (2002) IFN-alpha enhances CD40 ligand-mediated activation of immature monocyte-derived dendritic cells, Int Immunol. 14, 367–80. [DOI] [PubMed] [Google Scholar]
- 59.Padovan E, Spagnoli GC, Ferrantini M & Heberer M (2002) IFN-alpha2a induces IP-10/CXCL10 and MIG/CXCL9 production in monocyte-derived dendritic cells and enhances their capacity to attract and stimulate CD8+ effector T cells, J Leukoc Biol. 71, 669–76. [PubMed] [Google Scholar]
- 60.Brown CC, Gudjonson H, Pritykin Y, Deep D, Lavallee VP, Mendoza A, Fromme R, Mazutis L, Ariyan C, Leslie C, Pe’er D & Rudensky AY (2019) Transcriptional Basis of Mouse and Human Dendritic Cell Heterogeneity, Cell. 179, 846–863 e24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Bazhin AV, von Ahn K, Fritz J, Werner J & Karakhanova S (2018) Interferon-alpha Up-Regulates the Expression of PD-L1 Molecules on Immune Cells Through STAT3 and p38 Signaling, Front Immunol. 9, 2129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, Zaretsky JM, Sun L, Hugo W, Wang X, Parisi G, Saus CP, Torrejon DY, Graeber TG, Comin-Anduix B, Hu-Lieskovan S, Damoiseaux R, Lo RS & Ribas A (2017) Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression, Cell Rep. 19, 1189–1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Curtsinger JM, Schmidt CS, Mondino A, Lins DC, Kedl RM, Jenkins MK & Mescher MF (1999) Inflammatory cytokines provide a third signal for activation of naive CD4+ and CD8+ T cells, J Immunol. 162, 3256–62. [PubMed] [Google Scholar]
- 64.Curtsinger JM, Valenzuela JO, Agarwal P, Lins D & Mescher MF (2005) Type I IFNs provide a third signal to CD8 T cells to stimulate clonal expansion and differentiation, J Immunol. 174, 4465–9. [DOI] [PubMed] [Google Scholar]
- 65.Sanchez PJ & Kedl RM (2011) An alternative signal 3: CD8(+) T cell memory independent of IL-12 and type I IFN is dependent on CD27/OX40 signaling, Vaccine. 30, 1154–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Sica GL, Choi IH, Zhu G, Tamada K, Wang SD, Tamura H, Chapoval AI, Flies DB, Bajorath J & Chen L (2003) B7-H4, a molecule of the B7 family, negatively regulates T cell immunity, Immunity. 18, 849–61. [DOI] [PubMed] [Google Scholar]
- 67.Yang S, Wei W & Zhao Q (2020) B7-H3, a checkpoint molecule, as a target for cancer immunotherapy, Int J Biol Sci. 16, 1767–1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Michalska A, Blaszczyk K, Wesoly J & Bluyssen HAR (2018) A Positive Feedback Amplifier Circuit That Regulates Interferon (IFN)-Stimulated Gene Expression and Controls Type I and Type II IFN Responses, Front Immunol. 9, 1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Butte MJ, Keir ME, Phamduy TB, Sharpe AH & Freeman GJ (2007) Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses, Immunity. 27, 111–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Lucas ED & Tamburini BAJ (2019) Lymph Node Lymphatic Endothelial Cell Expansion and Contraction and the Programming of the Immune Response, Front Immunol. 10, 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Angeli V, Ginhoux F, Llodra J, Quemeneur L, Frenette PS, Skobe M, Jessberger R, Merad M & Randolph GJ (2006) B cell-driven lymphangiogenesis in inflamed lymph nodes enhances dendritic cell mobilization, Immunity. 24, 203–15. [DOI] [PubMed] [Google Scholar]
- 72.Chyou S, Benahmed F, Chen J, Kumar V, Tian S, Lipp M & Lu TT (2011) Coordinated regulation of lymph node vascular-stromal growth first by CD11c+ cells and then by T and B cells, J Immunol. 187, 5558–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ivashkiv LB & Donlin LT (2014) Regulation of type I interferon responses, Nat Rev Immunol. 14, 36–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Niemand C, Nimmesgern A, Haan S, Fischer P, Schaper F, Rossaint R, Heinrich PC & Muller-Newen G (2003) Activation of STAT3 by IL-6 and IL-10 in primary human macrophages is differentially modulated by suppressor of cytokine signaling 3, J Immunol. 170, 3263–72. [DOI] [PubMed] [Google Scholar]
- 75.Pennell LM & Fish EN (2017) Interferon-beta regulates dendritic cell activation and migration in experimental autoimmune encephalomyelitis, Immunology. 152, 439–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Yen JH, Kong W & Ganea D (2010) IFN-beta inhibits dendritic cell migration through STAT-1-mediated transcriptional suppression of CCR7 and matrix metalloproteinase 9, J Immunol. 184, 3478–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Fujita H, Asahina A, Tada Y, Fujiwara H & Tamaki K (2005) Type I interferons inhibit maturation and activation of mouse Langerhans cells, J Invest Dermatol. 125, 126–33. [DOI] [PubMed] [Google Scholar]
- 78.Hauser MA & Legler DF (2016) Common and biased signaling pathways of the chemokine receptor CCR7 elicited by its ligands CCL19 and CCL21 in leukocytes, J Leukoc Biol. 99, 869–82. [DOI] [PubMed] [Google Scholar]
- 79.Yao H, Lan J, Li C, Shi H, Brosseau JP, Wang H, Lu H, Fang C, Zhang Y, Liang L, Zhou X, Wang C, Xue Y, Cui Y & Xu J (2019) Inhibiting PD-L1 palmitoylation enhances T-cell immune responses against tumours, Nat Biomed Eng. 3, 306–317. [DOI] [PubMed] [Google Scholar]
- 80.Horita H, Law A, Hong S & Middleton K (2017) Identifying Regulatory Posttranslational Modifications of PD-L1: A Focus on Monoubiquitinaton, Neoplasia. 19, 346–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Li CW, Lim SO, Xia W, Lee HH, Chan LC, Kuo CW, Khoo KH, Chang SS, Cha JH, Kim T, Hsu JL, Wu Y, Hsu JM, Yamaguchi H, Ding Q, Wang Y, Yao J, Lee CC, Wu HJ, Sahin AA, Allison JP, Yu D, Hortobagyi GN & Hung MC (2016) Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity, Nat Commun. 7, 12632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Beli P, Lukashchuk N, Wagner SA, Weinert BT, Olsen JV, Baskcomb L, Mann M, Jackson SP & Choudhary C (2012) Proteomic investigations reveal a role for RNA processing factor THRAP3 in the DNA damage response, Mol Cell. 46, 212–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Bennetzen MV, Larsen DH, Bunkenborg J, Bartek J, Lukas J & Andersen JS (2010) Site-specific phosphorylation dynamics of the nuclear proteome during the DNA damage response, Mol Cell Proteomics. 9, 1314–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Mertins P, Mani DR, Ruggles KV, Gillette MA, Clauser KR, Wang P, Wang X, Qiao JW, Cao S, Petralia F, Kawaler E, Mundt F, Krug K, Tu Z, Lei JT, Gatza ML, Wilkerson M, Perou CM, Yellapantula V, Huang KL, Lin C, McLellan MD, Yan P, Davies SR, Townsend RR, Skates SJ, Wang J, Zhang B, Kinsinger CR, Mesri M, Rodriguez H, Ding L, Paulovich AG, Fenyo D, Ellis MJ, Carr SA & Nci C (2016) Proteogenomics connects somatic mutations to signalling in breast cancer, Nature. 534, 55–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Mertins P, Yang F, Liu T, Mani DR, Petyuk VA, Gillette MA, Clauser KR, Qiao JW, Gritsenko MA, Moore RJ, Levine DA, Townsend R, Erdmann-Gilmore P, Snider JE, Davies SR, Ruggles KV, Fenyo D, Kitchens RT, Li S, Olvera N, Dao F, Rodriguez H, Chan DW, Liebler D, White F, Rodland KD, Mills GB, Smith RD, Paulovich AG, Ellis M & Carr SA (2014) Ischemia in tumors induces early and sustained phosphorylation changes in stress kinase pathways but does not affect global protein levels, Mol Cell Proteomics. 13, 1690–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Schweppe DK, Rigas JR & Gerber SA (2013) Quantitative phosphoproteomic profiling of human non-small cell lung cancer tumors, J Proteomics. 91, 286–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Shiromizu T, Adachi J, Watanabe S, Murakami T, Kuga T, Muraoka S & Tomonaga T (2013) Identification of missing proteins in the neXtProt database and unregistered phosphopeptides in the PhosphoSitePlus database as part of the Chromosome-centric Human Proteome Project, J Proteome Res. 12, 2414–21. [DOI] [PubMed] [Google Scholar]
- 88.Hansen JD, Du Pasquier L, Lefranc MP, Lopez V, Benmansour A & Boudinot P (2009) The B7 family of immunoregulatory receptors: a comparative and evolutionary perspective, Molecular immunology. 46, 457–72. [DOI] [PubMed] [Google Scholar]
- 89.Lin DY, Tanaka Y, Iwasaki M, Gittis AG, Su HP, Mikami B, Okazaki T, Honjo T, Minato N & Garboczi DN (2008) The PD-1/PD-L1 complex resembles the antigen-binding Fv domains of antibodies and T cell receptors, Proc Natl Acad Sci U S A. 105, 3011–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Dong H, Zhu G, Tamada K & Chen L (1999) B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion, Nat Med. 5, 1365–9. [DOI] [PubMed] [Google Scholar]
- 91.Chaudhri A, Xiao Y, Klee AN, Wang X, Zhu B & Freeman GJ (2018) PD-L1 Binds to B7-1 Only In Cis on the Same Cell Surface, Cancer Immunol Res. 6, 921–929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Kariv I, Truneh A & Sweet RW (1996) Analysis of the site of interaction of CD28 with its counter-receptors CD80 and CD86 and correlation with function, J Immunol. 157, 29–38. [PubMed] [Google Scholar]
- 93.Orabona C, Grohmann U, Belladonna ML, Fallarino F, Vacca C, Bianchi R, Bozza S, Volpi C, Salomon BL, Fioretti MC, Romani L & Puccetti P (2004) CD28 induces immunostimulatory signals in dendritic cells via CD80 and CD86, Nat Immunol. 5, 1134–42. [DOI] [PubMed] [Google Scholar]
- 94.Talay O, Shen CH, Chen L & Chen J (2009) B7-H1 (PD-L1) on T cells is required for T-cell-mediated conditioning of dendritic cell maturation, Proc Natl Acad Sci U S A. 106, 2741–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Fumio Matsumura RP, Sukhwinder Singh, Jürgen Scheller, Shigeko Yamashiro (2020) Investigation of fascin1, a marker of mature dendritic cells, reveals a New role for IL-6 signaling in chemotaxis, bioRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Sharma P & Allison JP (2015) The future of immune checkpoint therapy, Science. 348, 56–61. [DOI] [PubMed] [Google Scholar]
- 97.Blackburn SD, Shin H, Freeman GJ & Wherry EJ (2008) Selective expansion of a subset of exhausted CD8 T cells by alphaPD-L1 blockade, Proc Natl Acad Sci U S A. 105, 15016–21. [DOI] [PMC free article] [PubMed] [Google Scholar]