首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 46 毫秒
1.
Ship is an Src homology 2 domain containing inositol polyphosphate 5-phosphatase which has been implicated as an important signaling molecule in hematopoietic cells. In B cells, Ship becomes associated with Fcγ receptor IIB (FcγRIIB), a low affinity receptor for the Fc portion of immunoglobulin (Ig)G, and is rapidly tyrosine phosphorylated upon B cell antigen receptor (BCR)–FcγRIIB coligation. The function of Ship in lymphocytes was investigated in Ship−/− recombination-activating gene (Rag)−/− chimeric mice generated from gene-targeted Ship−/− embryonic stem cells. Ship−/−Rag−/− chimeras showed reduced numbers of B cells and an overall increase in basal serum Ig. Ship−/− splenic B cells displayed prolonged Ca2+ influx, increased proliferation in vitro, and enhanced mitogen-activated protein kinase (MAPK) activation in response to BCR–FcγRIIB coligation. These results demonstrate that Ship plays an essential role in FcγRIIB-mediated inhibition of BCR signaling, and that Ship is a crucial negative regulator of Ca2+ flux and MAPK activation.  相似文献   

2.
FcγRIIB is an inhibitory receptor expressed throughout B cell development. Diminished expression or function is associated with lupus in mice and humans, in particular through an effect on autoantibody production and plasma cell (PC) differentiation. Here, we analyzed the effect of B cell–intrinsic FcγRIIB expression on B cell activation and PC differentiation. Loss of FcγRIIB on B cells in Fcgr2b–conditional KO (Fcgr2b-cKO) mice led to a spontaneous increase in autoantibody titers. This increase was most striking for IgG3, suggestive of increased extrafollicular responses. Marginal zone (MZ) B cells had the highest expression of FcγRIIB in both mice and humans. This high expression of FcγRIIB was linked to increased MZ B cell activation, Erk phosphorylation, and calcium flux in the absence of FcγRIIB triggering. We observed a marked increase in IgG3+ PCs and B cells during extrafollicular PC responses in Fcgr2b-cKO mice. The increased IgG3 response following immunization of Fcgr2b-cKO mice was lost in MZ-deficient Notch2 Fcgr2b–double KO mice. Importantly, patients with systemic lupus erythematosus (SLE) had a decrease in FcγRIIB expression that was strongest in MZ B cells. Thus, we present a model in which high FcγRIIB expression in MZ B cells prevented their hyperactivation and ensuing autoimmunity.  相似文献   

3.
T cell receptors on CD4+ lymphocytes recognize antigen-derived peptides presented by major histocompatibility complex (MHC) class II molecules. A very limited set of peptides among those that may potentially bind MHC class II is actually presented to T lymphocytes. We here examine the role of two receptors mediating antigen internalization by antigen presenting cells, type IIb2 and type III receptors for IgG (FcγRIIb2 and FcγRIII, respectively), in the selection of peptides for presentation to T lymphocytes. B lymphoma cells expressing recombinant FcγRIIb2 or FcγRIII were used to assess the presentation of several epitopes from two different antigens. 4 out of the 11 epitopes tested were efficiently presented after antigen internalization through FcγRIIb2 and FcγRIII. In contrast, the 7 other epitopes were efficiently presented only when antigens were internalized through FcγRIII, but not through FcγRIIb2. The capacity to present these latter epitopes was transferred to a tail-less FcγRIIb2 by addition of the FcγRIII-associated γ chain cytoplasmic tail. Mutation of a single leucine residue at position 35 of the γ chain cytoplasmic tail resulted in the selective loss of presentation of these epitopes. Therefore, the nature of the receptor that mediates internalization determines the selection of epitopes presented to T lymphocytes within single protein antigens.Antigen receptors on CD4+ helper T lymphocytes recognize short peptides presented by class II molecules of MHC (1, 2). Antigenic peptides are generated by proteolytic degradation in the endocytic pathway, where they associate with MHC class II molecules. Only a very limited set of peptides among all the potential peptides is actually loaded onto MHC class II molecules in APCs (3). The mechanisms underlying the selection of peptides for MHC class II–restricted antigen presentation are yet unclear. However, we know that two independent complex processes of intracellular transport towards endosomes are crucial for MHC class II–restricted antigen presentation: the traffic of MHC class II molecules and the delivery of antigens (4, 5).MHC class II intracellular transport has been analyzed in detail. Newly synthesized MHC class II molecules reach endosomes, either directly from the trans-golgi network or after a short appearance at the plasma membrane, in association to the invariant (Ii)1 chain (5). Ii is then degraded and the class II–associated Ii chain peptide (CLIP) is replaced by an antigenic peptide under the control of HLA-DM (6). It has recently become clear that an alternative, Ii chain–independent pathway for MHC class II transport to endosomes also exists. Indeed, MHC class II molecules may reach the endocytic pathway from the cell surface by endocytosis (7), due to internalization signals present in the cytosolic domain of the MHC class II β chain (8). Newly synthesized and recycling MHC class II molecules may present different peptides (9). Accordingly, we have previously shown that different antigen receptors may also selectively target antigens for presentation by either of these MHC class II presentation pathways (10).In contrast, very little is known about the endocytic transport of antigen receptors. Physiologically, antigens are delivered to the endocytic pathway by different families of receptors, which strongly increase the efficiency of MHC class II–restricted antigen presentation (11). In B lymphocytes, surface Ig mediates both cell activation and the uptake of specific antigens (12), while the expression of a particular endocytosis-deficient receptor for the Fc portion of IgG (FcγRIIb1) prevents efficient presentation of irrelevant IgG-complexed antigens (13). Interestingly, the epitope specificity of surface Ig positively and negatively influences the presentation of various T cell epitopes (14, 15).In monocytes and dendritic cells, receptors for IgG (FcγRs), in addition to mannose receptors, mediate antigen internalization and strongly increase the efficiency of presentation to specific T cells (16). Two different FcγRs, type IIb2 and type III (FcγRIIb2 and FcγRIII) are expressed in dendritic cells. FcγRIIb2 is a monomeric receptor that, like FcγRIIb1, mediates the inhibition of cell activation when cocrosslinked to surface Ig (13). FcγIII is an heterotrimer consisting of an α chain and a dimer of γ chains, which couple the receptor to cytoplasmic effectors of signal transduction (17).To individually analyze the function of these two receptors, we expressed them by cDNA transfection into an FcγR-negative B cell lymphoma cell line (13, 18, 19). We have previously shown that the amino acid sequence, called immunoreceptor tyrosine kinase activation motif (ITAM), in the FcγIII-associated γ chain responsible for cell activation is also involved in receptor internalization (18, 19). In addition, mutation of either of the two tyrosine residues in the ITAM of the γ chain inhibits both cell activation and ligand internalization (19). Thus, in contrast to FcγRIIb2, which contains no ITAM, is not tyrosine phosphorylated, and does not induce cell activation, FcγRIII associates with and activates cytosolic tyrosine kinases after engagement by its ligand (18). In addition to this functional diversity of the two receptors, their expression is also selectively regulated, since TNF-α and IFN-γ increase the expression of FcγRIII and inhibit that of FcγRIIb2 in monocytes (20).The diversity in the signals required of FcγRIIb2 and FcγRIII internalization, as well as the differential regulation of their expression in APCs, suggest that the two receptors may have different antigen-presenting functions. We here examine the ability of FcγRIIb2 and FcγRIII to induce the presentation of various T cell epitopes from two different antigens, CI λ repressor and hen egg lysozyme (HEL). Internalization of antigen–antibody complexes through FcγRIII induced the efficient presentation of all the T cell epitopes tested, whereas FcγRIIb2 only induced the presentation of a few. Point mutation of leucine 35 to alanine (L35A) in the cytoplasmic tail of FcγRIII γ chain blocked signal transduction without affecting the internalization of immune complexes. This mutation also blocked the presentation of the epitopes that were only presented after internalization by FcγRIII. In contrast, the presentation of all the epitopes that were efficiently generated after internalization by FcγRIIb2 was not affected. Thus, the nature of the receptor that mediates antigen internalization influences the selection of the epitopes presented to T lymphocytes.  相似文献   

4.
Fc γ receptor (FcγR) coengagement can facilitate antibody-mediated receptor activation in target cells. In particular, agonistic antibodies that target tumor necrosis factor receptor (TNFR) family members have shown dependence on expression of the inhibitory FcγR, FcγRIIB. It remains unclear if engagement of FcγRIIB also extends to the activities of antibodies targeting immunoregulatory TNFRs expressed by T cells. We have explored the requirement for activating and inhibitory FcγRs for the antitumor effects of antibodies targeting the TNFR glucocorticoid-induced TNFR-related protein (GITR; TNFRSF18; CD357) expressed on activated and regulatory T cells (T reg cells). We found that although FcγRIIB was dispensable for the in vivo efficacy of anti-GITR antibodies, in contrast, activating FcγRs were essential. Surprisingly, the dependence on activating FcγRs extended to an antibody targeting the non-TNFR receptor CTLA-4 (CD152) that acts as a negative regulator of T cell immunity. We define a common mechanism that correlated with tumor efficacy, whereby antibodies that coengaged activating FcγRs expressed by tumor-associated leukocytes facilitated the selective elimination of intratumoral T cell populations, particularly T reg cells. These findings may have broad implications for antibody engineering efforts aimed at enhancing the therapeutic activity of immunomodulatory antibodies.Activating Fc γ receptors (FcγRs) stimulate immune cell effector mechanisms, such as antibody-dependent cell-mediated cytotoxicity (ADCC) and phagocytosis (ADCP), which combine to facilitate antibody-mediated tumor cell killing (Nimmerjahn and Ravetch, 2008; Hogarth and Pietersz, 2012). The importance of FcγR-mediated immune effector cell function has been demonstrated in preclinical efficacy studies for antibodies targeting a range of tumor cell–expressed receptors, including trastuzumab (HER2) and rituximab (CD20; Clynes et al., 2000; Nimmerjahn and Ravetch, 2012). The inhibitory FcγR, FcγRIIB, functions to modulate activating FcγR-mediated effector mechanisms in immune cells that coexpress both FcγR classes, such as macrophages and dendritic cells. FcγRIIB has recently been implicated in augmenting antibody-mediated receptor forward signaling through a mechanism of cross-linking in target cells expressing the TNF receptor (TNFR) family members TNFRSF10, TNFRSF10B (DR4 and DR5, respectively), and TNFRSF5 (CD40; Wilson et al., 2011; Li and Ravetch, 2012). It remains unclear what contribution FcγR biology has in the modality of antibody therapeutics that target other cell surface receptors. In particular, the emerging clinical benefit of agonistic antibodies targeting the T cell–APC interface raises the possibility that FcγR coengagement may contribute to their in vivo mechanism of action (Mellman et al., 2011).Preclinical studies in mice using agonistic antibodies targeted to glucocorticoid-induced TNFR-related protein (GITR)—a costimulatory TNFR expressed by regulatory and activated T cells—have shown compelling antitumor activity in syngeneic mouse tumor models (Turk et al., 2004; Ko et al., 2005). In vitro, stimulation of GITR with agonist antibodies can induce forward signaling into T cells, which promotes proliferation and cytokine production (Kanamaru et al., 2004; Ronchetti et al., 2007). In vivo, several mechanisms have been proposed to contribute to the antitumor activity of antibodies targeting GITR; however, the current paradigm stipulates that agonist properties of these antibodies promotes cytotoxic effector T cell generation, while dampening the immunosuppressive effects by FoxP3+ CD4+ T reg cells (Ronchetti et al., 2012; Schaer et al., 2012). The recent findings that antibodies targeted to TNFR family members require FcγRIIB interaction for their in vivo activities led us to explore a common mechanism for antibodies targeting TNFRs expressed by T cells, using GITR to test this paradigm.  相似文献   

5.
B cells from young lyn−/− mice are hyperresponsive to anti-IgM–induced proliferation, suggesting involvement of Lyn in negative regulation of B cell antigen receptor (BCR)-mediated signaling. Here we show that tyrosine phosphorylation of FcγRIIB and CD22 coreceptors, which are important for feedback suppression of BCR-induced signaling, was severely impaired in lyn−/− B cells upon their coligation with the BCR. Hypophosphorylation on tyrosine residues of these molecules resulted in failure of recruiting the tyrosine phosphatase SHP-1 and inositol phosphatase SHIP, SH2-containing potent inhibitors of BCR-induced B cell activation, to the coreceptors. Consequently, lyn−/− B cells exhibited defects in suppressing BCR-induced Ca2+ influx and proliferation. Thus, Lyn is critically important in tyrosine phosphorylation of the coreceptors, which is required for feedback suppression of B cell activation.  相似文献   

6.
7.
The ζ family includes ζ, η, and FcεRIγ (Fcγ). Dimers of the ζ family proteins function as signal transducing subunits of the T cell antigen receptor (TCR), the pre-TCR, and a subset of Fc receptors. In mice lacking ζ/η chains, T cell development is impaired, yet low numbers of CD4+ and CD8+ T cells develop. This finding suggests either that pre-TCR and TCR complexes lacking a ζ family dimer can promote T cell maturation, or that in the absence of ζ/η, Fcγ serves as a subunit in TCR complexes. To elucidate the role of ζ family dimers in T cell development, we generated mice lacking expression of all of these proteins and compared their phenotype to mice lacking only ζ/η or Fcγ. The data reveal that surface complexes that are expressed in the absence of ζ family dimers are capable of transducing signals required for α/β–T cell development. Strikingly, T cells generated in both ζ/η−/− and ζ/η−/−–Fcγ−/− mice exhibit a memory phenotype and elaborate interferon γ. Finally, examination of different T cell populations reveals that ζ/η and Fcγ have distinct expression patterns that correlate with their thymus dependency. A possible function for the differential expression of ζ family proteins may be to impart distinctive signaling properties to TCR complexes expressed on specific T cell populations.  相似文献   

8.
The high-affinity IgE receptor FcεRI is constitutively expressed in mast cells and basophils and is required for transmitting stimulatory signals upon engagement of IgE-bound allergens. FcεRI is also constitutively expressed in dendritic cells (DCs) and monocytes in humans; however, the specific functions of the FcεRI expressed by these cells are not completely understood. Here, we found that FcεRI expressed by human blood DC antigen 1–positive (BDCA1+) DCs and monocytes, but not basophils, traffics to endolysosomal compartments under steady-state conditions. Furthermore, IgE bound to FcεRI on BDCA1+ DCs was rapidly endocytosed, transported to the lysosomes, and degraded in vitro. IgE injected into mice expressing human FcεRIα (FCER1A-Tg mice) was endocytosed by conventional DCs and monocytes, and endocytosis was associated with rapid clearance of circulating IgE from these mice. Importantly, this rapid IgE clearance was dependent on monocytes or DCs but not basophils. These findings strongly suggest that constitutive internalization of human FcεRI by DCs and monocytes distinctively contributes to serum IgE clearance.  相似文献   

9.
Antibodies have important roles in controlling cellular immunity through interaction with activating or inhibitory Fcγ receptors (FcγRs). FcγR engagement can facilitate receptor cross-linking on target cells, or induce retrograde FcγR signals to stimulate or suppress antibody-dependent, cell-mediated depletion of antigen-bearing target cells. Recent studies uncover unexpectedly important roles for FcγRs in the anticancer action of antibodies designed to trigger tumor cell apoptosis or enhance antitumor immunity. Here, we outline a conceptual framework for understanding these findings and discuss their mechanistic and translational implications.The adaptive immune system is capable of recognizing an essentially limitless number of antigens via the combinatorial assembly of gene segments that encode antibody variable domains. This diversity has been exploited successfully in a growing number of therapeutic antibodies that bind to a wide range of clinically validated targets. Antibodies that recognize soluble antigens, such as the cytokines tumor necrosis factor (TNF), vascular endothelial growth factor, or interleukin-6, act as antagonists by blocking the interaction of a target ligand with its cognate receptor. In some cases, such antibodies may also augment clearance of the target antigen.In contrast, antibodies that bind to cell surface antigens, often transmembrane receptors such as HER2, EGFR, or DR5, may act as antagonists or agonists, respectively, to block or stimulate the action of the cognate target. Alternatively, antibodies may bind a cell surface target that lacks signaling function, such as the CD20 antigen, and act as an anchor for FcγR-based recruitment of immune-effector cells to kill the antigen-expressing target by antibody-dependent, cell-mediated cytotoxicity (ADCC). Therefore, antibodies that recognize cell surface receptors can be categorized by their function of either mediating target cell killing or modulating target receptor signal transduction. However, two new studies in this issue demonstrate that these activities are not mutually exclusive and that antibodies harboring both properties may be advantageous for cancer immunotherapy. Due to shared expression of cell surface antigens, such as CTLA-4 or glucocorticoid-induced TNFR-related protein (GITR) on protumorigenic regulatory T (T reg) cells and antitumorigenic effector T (T eff) cells, antibodies that target such receptors are capable of inducing antitumor immunity both by depleting T reg cells and by stimulating T eff cells. However, antibodies that conform to this dual mechanism of action have the risk of depleting T eff cells, which are the final mediators of tumor cell killing. Therefore, understanding the principles that govern antibody–FcγR interactions is crucial for designing effective antibody-based immunotherapies.

Antibody–FcγR interactions

FcγRs fall into two functional classes: activating and inhibitory (Nimmerjahn and Ravetch, 2006). The FcγR family comprises three activating (mouse FcγRI, FcγRIII, and FcγRIV; human FcγRI, FcγRIIA, and FcγRIIIA) and one inhibitory (FcγRIIB) receptor. Activating FcγRs associate with a common signaling chain (FcRγ), containing an immunoreceptor tyrosine-based activation motif (ITAM) that recruits Syk family kinases to stimulate effector function. In contrast, FcγRIIB contains an immunoreceptor tyrosine-based inhibitory motif (ITIM) that recruits specific phosphatases to oppose signaling by activating FcγRs. Innate-immune cells, such as macrophages, monocytes, dendritic cells, mast cells, and granulocytes, express both activating and inhibitory FcγRIIB (Amigorena et al., 1992; Nimmerjahn and Ravetch, 2008). IgG subtypes differ in FcγR affinity: human IgG1 and IgG3 have higher affinity for activating than inhibitory FcγR, as do mouse IgG2a and IgG2b (Dijstelbloem et al., 2001; Nimmerjahn and Ravetch, 2005, 2006).Antagonist antibodies may bind to a soluble ligand or a cell surface receptor to prevent signaling. Target inhibition per se typically does not require accessory FcγR-bearing cells; therefore, antagonist antibodies often act independently of FcγRs, and accordingly, IgG subtype. However, if the target is engaged at the cell surface and is sufficiently abundant, effector cells may be recruited via Fc–FcγR interactions to deplete the antigen-displaying cell, an outcome that can be desirable or undesirable depending on the context. Target cell depletion can be manipulated by selecting IgG subtypes that favor binding to activating or inhibitory FcγRs. Unwanted target cell depletion can be minimized by incorporating Fc mutations that decrease FcγR affinity (Presta et al., 2002; Carter, 2006; Lazar et al., 2006; Satoh et al., 2006; Jefferis, 2009). For example, asparagine 297, the site for N-linked glycosylation required for FcγR binding in the constant region, can be replaced by alanine. Further mutations to enhance or decrease specific FcγR interactions have also been reported. Alternatively, some antibody variants can be produced in Escherichia coli rather than mammalian cells to prevent Fc glycosylation. Fc effectorless antibodies have been demonstrated to be equally as potent at blocking ligand–receptor interactions as their wild-type counterparts.Recent work has revealed unexpectedly that agonist antibodies designed to stimulate the tumor necrosis factor receptor superfamily (TNFRSF) members DR4, DR5, or CD40 depend on FcγR interaction for robust agonist activity (Li and Ravetch, 2011; Wilson et al., 2011; Smith et al., 2012). As TNFRSF members usually require ligand-induced super-clustering for signal transmission, bivalent IgG molecules are unable to induce their efficient stimulation. In vitro activity can be enhanced by artificial cross-linking of the primary antibody, with secondary anti-Fc antibodies, or—perhaps more importantly—by providing contact with FcγR-bearing cells. Pretreating FcγR-expressing cells with actin polymerization inhibitors blocks this enhancement, suggesting that FcγR clustering is important for antibody-mediated stimulation of the target receptor (Wilson et al., 2011). Studies with mice deficient in specific FcγR subsets demonstrate that expression of the inhibitory FcγRIIB is sufficient—if not superior—for enabling in vivo efficacy of agonist antibodies targeting CD40, or the death receptors DR4 and DR5 (Li and Ravetch, 2011; Wilson et al., 2011). Reliance on FcγRIIB circumvents the potential for FcγR-mediated target cell depletion and is therefore advantageous for inducing signal transduction on target cells, such as CD40 signaling in dendritic cells. CD40 engagement on dendritic cells enhances their antigen presentation capabilities, thereby increasing T cell responses. Therefore, cross-linking CD40 via FcγRIIB on DCs enhances their T cell priming function, while minimizing depletion. Importantly, anti-CD40–induced adjuvant activity was unabated in mice deficient in activating FcγRs, yet was abrogated in FcγRIIB-deficient mice (Li and Ravetch, 2011). However, FcγR-dependent depletion of DR4- or DR5-expressing tumor cells could be useful to induce antitumor activity. These findings suggest that agonist antibodies targeting either proapoptotic or co-stimulatory TNFRSF members can rely on FcγRs as a dynamic cross-linking scaffold—a function that, at least in mice, may be supported more effectively by FcγRIIB.

Agonist GITR antibodies require activating FcγRs for antitumor efficacy

In this issue, Bulliard et al. report an apparent exception to the latter paradigm: agonist antibodies targeting the TNFRSF family member GITR require activating FcγR effector function to promote tumor regression in mice. GITR expression is induced on T eff cells upon T cell receptor (TCR) stimulation, and GITR cross-linking by GITR ligand or agonist antibodies co-stimulates TCR signaling (McHugh et al., 2002; Shimizu et al., 2002; Tone et al., 2003; Ronchetti et al., 2004; Stephens et al., 2004). In transplantable tumor models, anti-GITR treatment is hypothesized to induce tumor regression through T eff cell co-stimulation (Turk et al., 2004; Ko et al., 2005). Distinct from antibodies that target other TNFR superfamily members, anti-GITR activity was unaltered in Fcgr2b−/− mice. However, anti-GITR treatment was ineffective in knockout mice lacking the ITAM-containing FcγR chain required for signaling by all activating FcγRs. Hence, an alternative (or additional) mechanism of action—distinct from FcγRIIB-mediated GITR cross-linking to promote co-stimulation—may be critical for antitumor efficacy. Given that GITR-expressing CD8+ T cells and CD4+ T eff cells are needed for tumor cell killing, this mechanism may involve FcγR-dependent depletion of GITR-expressing T reg cells.T reg cells frequently express T cell activation markers induced by TCR signaling (Gavin et al., 2002; McHugh et al., 2002). In addition to their well-characterized role in maintaining peripheral tolerance to self-antigens, T reg cells have been demonstrated to suppress tumor immunity (Nishikawa and Sakaguchi, 2010; Josefowicz et al., 2012). T reg cells are highly enriched in tumors, both in mouse models and in various human cancers. Furthermore, in cancer patients, abundance of T reg cells within tumors is associated with poor prognosis, suggesting that these cells play an important role in suppressing antitumor immunity. Therefore, strategies to deplete intratumoral T reg cells may enhance the generation of tumor-directed T eff cell responses.GITR is weakly expressed on naive T eff cells, but is present on resting T reg cells and up-regulated on activated T eff cells. Bulliard et al. (2013) observed a significant reduction in tumor-associated, but not peripheral, T reg cells upon anti-GITR treatment of tumor-bearing mice. Hence, anti-GITR antibodies may enhance antitumor immunity by depleting GITR-positive T reg cells, in addition to co-stimulating antigen-experienced T eff cells (Coe et al., 2010). However, the shared expression of GITR on T reg and T eff cells makes both populations potentially susceptible to antibody-dependent, FcγR-mediated depletion. Indeed, diminished effector CD4+ and CD8+ T cell numbers were reported to accompany T reg cell depletion in tumor tissues. Therefore, elimination of intratumoral T reg cells, coupled with T eff cell co-stimulation and minimal T eff cell depletion, might provide an integrated mechanism of action for anti-GITR antibodies. The relative contribution of each of these mechanistic components remains to be clarified.

CTLA-4 blocking antibodies mediate T reg cell depletion

CTLA-4 is an inhibitory receptor that is induced on antigen-experienced T eff cells as a negative feedback regulator (Walunas et al., 1994; Krummel and Allison, 1995). Whereas CTLA-4 expression is induced on activated T eff cells, CTLA-4 is constitutively expressed by T reg cells, which require CTLA-4 function to aggregate preferentially around dendritic cells and inhibit their antigen-presenting activity (Onishi et al., 2008). As such, the multiorgan autoimmunity detected in Ctla4 germline knockout mice is phenocopied in T reg cell–specific Ctla4 conditional knockout mice (Tivol et al., 1995; Waterhouse et al., 1995; Chambers et al., 1997; Wing et al., 2008). Although anti–CTLA-4 therapy is one of the first clinically validated examples of effective cancer immunotherapy, its mechanism of action is poorly understood (Hodi et al., 2010). It is hypothesized that antagonist anti–CTLA-4 antibodies enhance tumor immunity by relieving inhibitory CTLA-4 signals on antigen-experienced T eff cells, as well as curtailing the suppressive function of T reg cells. Following the paradigm of antagonist antibodies, CTLA-4–blocking antibodies would be expected to exert their efficacy in the absence of Fc effector function.However, studies from Bulliard et al. (2013) and Simpson et al. in this issue provide compelling evidence that the activity of anti–CTLA-4 antibodies requires activating FcγR engagement to deplete T reg cells. Similar to anti-GITR therapy, anti–CTLA-4–mediated tumor regression was abrogated in FcγR-deficient mice, suggesting that retrograde signaling by activating FcγRs and consequent T reg cell depletion is important for antitumor activity. Accordingly, intratumoral T reg cell numbers were dramatically reduced in response to anti–CTLA-4 treatment. Importantly, this reduction was not associated with dedifferentiation or impaired generation of T reg cells, thereby supporting cellular depletion as the most plausible mechanism of efficacy.Because activated T eff cells express CTLA-4, they are also susceptible to antibody-mediated depletion. In this regard, Bulliard et al. (2013) demonstrated that both T reg and T eff cell numbers were reduced in response to anti–CTLA-4 treatment. In contrast, Simpson et al. (2013) observed that in an adoptive T cell transfer model, intratumoral T eff cells were not depleted by anti–CTLA-4 treatment, but rather increased in number. Differences in tumor model, antibody isotype or clone, or vaccine co-administration could potentially account for these discrepant results. Nevertheless, T reg cells may be more sensitive to antibody-mediated depletion because of their increased expression of CTLA-4 within tumor tissues. Consistent with this notion, tumor-infiltrating T reg cells expressed nearly fourfold higher CTLA-4 levels than did tumor-associated T eff cells in the Colon26 carcinoma model. Additionally, in the B16-BL6 melanoma model, CTLA-4 surface expression was slightly elevated in tumor-associated versus peripheral T reg cells or intratumoral T eff cells. It remains to be investigated whether this variation in receptor expression is detected in human intratumoral T cells and leads to differential depletion in cancer patients.

Conclusions

Important roles have emerged for FcγRs in mediating the antitumor efficacy of a new generation of therapeutic antibodies aiming to attack cancer directly, by triggering tumor cell apoptosis, or indirectly, by enhancing antitumor immunity (Fig. 1). First, FcγRs on tumor-infiltrating innate immune cells provide a cross-linking scaffold to enhance antibody-mediated activation of proapoptotic TNFRSF members such as DR4 and DR5 on cancer cells. Similarly, FcγR-mediated cross-linking supports activation of T eff cells via co-stimulatory TNFRSF members such as GITR. In some instances, this cross-linking function can be performed redundantly by activating or inhibitory FcγRs; however, mouse studies suggest that the inhibitory FcγRIIB may provide a more effective molecular scaffold, for reasons that have yet to be defined. Fc mutations that enhance affinity for FcγRs may serve to improve the efficacy of agonist antibodies targeting various TNFRSF members. In addition to cross-linking cell surface receptors to support forward signaling in target cells, FcγRs can mediate antibody-driven reverse signaling to activate FcγR-bearing cells and promote target cell depletion. For cancer immunotherapy, antibodies that fulfill both functions may be beneficial to deplete immunosuppressive T reg cells and stimulate T eff cells. The high prevalence of T reg cells in tumor tissues poses a significant barrier to generating a productive antitumor T cell response. Expression of antigens such as GITR or CTLA-4 on T reg cells affords the opportunity for enhanced antibody-based depletion of such immune-suppressive cells from the tumor microenvironment, thereby augmenting antitumor immunity. A potential caveat is that desirable T eff cells also might express the same antigens and hence be subject to similar depletion. Thus, maximal therapeutic efficacy likely necessitates a delicate balance between depleting T reg cells while sparing T eff cells. Defining the FcγR interactions responsible for these different outcomes should help optimize the effectiveness of antibodies to biologically relevant T cell antigens, including GITR, CTLA-4, and beyond.Open in a separate windowFigure 1.How Fcγ receptors enable anticancer efficacy of proapoptotic and immune-modulatory antibodies. Inhibitory (Inh.) or activating (Act.) FcγRs can provide a dynamic scaffold for cross-linking agonist antibodies targeting proapoptotic receptors such as DR4 or DR5 on the surface of cancer cells, thereby promoting direct tumor cell apoptosis (A). Similarly, FcγRs can support agonist antibody-mediated cross-linking of immune cell co-stimulatory receptors, e.g., CD40 on antigen-presenting cells (B), or GITR on T eff cells (C), to augment antitumor immunity. T eff cell responses can be enhanced further, through direct antibody-based antagonism of the inhibitory molecule CTLA-4 (D). Finally, activating FcγRs can promote ADCC-based depletion of T reg cells upon antibody binding to GITR or CTLA-4, thus alleviating T reg suppression and strengthening antitumor immunity (E).  相似文献   

10.
Stimulation of CD4+ helper T lymphocytes by antigen-presenting cells requires the degradation of exogenous antigens into antigenic peptides which associate with major histocompatibility complex (MHC) class II molecules in endosomal or lysosomal compartments. B lymphocytes mediate efficient antigen presentation first by capturing soluble antigens through clonally distributed antigen receptors (BCRs), composed of membrane immunoglobulin (Ig) associated with Ig-α/Ig-β heterodimers which, second, target antigens to MHC class II–containing compartments. We report that antigen internalization and antigen targeting through the BCR or its Ig-α–associated subunit to newly synthesized class II lead to the presentation of a large spectrum of T cell epitopes, including some cryptic T cell epitopes. To further characterize the intracellular mechanisms of BCR-mediated antigen presentation, we used two complementary experimental approaches: mutational analysis of the Ig-α cytoplasmic tail, and overexpression in B cells of dominant negative syk mutants. Thus, we found that the syk tyrosine kinase, an effector of the BCR signal transduction pathway, is involved in the presentation of peptide– MHC class II complexes through antigen targeting by BCR subunits.  相似文献   

11.
The effector activity of antibodies is dependent on engagement with Fcγ receptors (FcγRs) and activation of the associated intracellular signaling pathways. Preclinical evaluation of therapeutic humanized or chimeric mAbs to study the interactions of their Fc regions with FcγRs is hampered by substantial structural and functional FcγR diversity among species. In this report, we used mice expressing only human FcγRs to evaluate the contribution of FcγR-mediated pathways to the neutralizing activity of an anti-anthrax toxin chimeric mAb. We observed that the protective activity of this mAb was highly dependent upon FcγR engagement, with minimal protection against anthrax toxin observed in FcγR-deficient mice following mAb administration. We generated anti-anthrax toxin mAbs with specific Fc domain variants with selectively enhanced affinity for particular human FcγRs and assessed their activity in FcγR-humanized mice. We determined that Fc domain variants that were capable of selectively engaging activating FcγRs substantially enhanced the in vitro and in vivo activity of anthrax toxin-neutralizing antibodies. These findings indicate that the application of Fc domain engineering is a feasible strategy to enhance toxin-neutralizing activity and suggest that engineered antitoxin antibodies will have improved therapeutic efficacy.  相似文献   

12.
Phagocytosis of bacteria by macrophages and neutrophils is an essential component of host defense against infection. The mechanism whereby the interaction of opsonized particles with Fcγ receptors triggers the engulfment of opsonized particles remains incompletely understood, although activation of tyrosine kinases has been recognized as an early step. Recent studies in other systems have demonstrated that tyrosine kinases can in turn signal the activation of small GTPases of the ras superfamily. We therefore investigated the possible role of Rho in Fc receptor–mediated phagocytosis. To this end we microinjected J774 macrophages with C3 exotoxin from Clostridium botulinum, which ADP-ribosylates and inactivates Rho. C3 exotoxin induced the retraction of filopodia, the disappearance of focal complexes, and a global decrease in the F-actin content of J774 cells. In addition, these cells exhibited increased spreading and the formation of vacuolar structures. Importantly, inactivation of Rho resulted in the complete abrogation of phagocytosis. Inhibition of Fcγ receptor–mediated phagocytosis by C3 exotoxin was confirmed in COS cells, which become phagocytic upon transfection of the FcγRIIA receptor. Rho was found to be essential for the accumulation of phosphotyrosine and of F-actin around phagocytic cups and for Fcγ receptor–mediated Ca2+ signaling. The clustering of receptors in response to opsonin, an essential step in Fcγ-induced signaling, was the earliest event shown to be inhibited by C3 exotoxin. The effect of the toxin was specific, since clustering and internalization of transferrin receptors were unaffected by microinjection of C3. These data identify a role for small GTPases in Fcγ receptor–mediated phagocytosis by leukocytes.  相似文献   

13.
Receptors on macrophages for the Fc region of IgG (FcγR) mediate a number of responses important for host immunity. Signaling events necessary for these responses are likely initiated by the activation of Src-family and Syk-family tyrosine kinases after FcγR cross-linking. Macrophages derived from Syk-deficient (Syk) mice were defective in phagocytosis of particles bound by FcγRs, as well as in many FcγR-induced signaling events, including tyrosine phosphorylation of a number of cellular substrates and activation of MAP kinases. In contrast, Syk macrophages exhibited normal responses to another potent macrophage stimulus, lipopolysaccharide. Phagocytosis of latex beads and Escherichia coli bacteria was also not affected. Syk macrophages exhibited formation of polymerized actin structures opposing particles bound to the cells by FcγRs (actin cups), but failed to proceed to internalization. Interestingly, inhibitors of phosphatidylinositol 3-kinase also blocked FcγR-mediated phagocytosis at this stage. Thus, PI 3-kinase may participate in a Syk-dependent signaling pathway critical for FcγR-mediated phagocytosis. Macrophages derived from mice deficient for the three members of the Src-family of kinases expressed in these cells, Hck, Fgr, and Lyn, exhibited poor Syk activation upon FcγR engagement, accompanied by a delay in FcγR-mediated phagocytosis. These observations demonstrate that Syk is critical for FcγR-mediated phagocytosis, as well as for signal transduction in macrophages. Additionally, our findings provide evidence to support a model of sequential tyrosine kinase activation by FcγR's analogous to models of signaling by the B and T cell antigen receptors.  相似文献   

14.
Receptors for the Fc portion of immunoglobulin (Ig)G (FcγR) mediate phagocytosis of IgG-opsonized particles by a process that can be divided into four major steps: receptor–ligand binding, pseudopod extension, internalization, and lysosomal fusion. We have expressed single classes of FcγR in COS fibroblasts to examine the structural determinants necessary to complete the four steps of phagocytosis. Using phase contrast, fluorescence, confocal, and electron microscopy we have demonstrated that FcγR-expressing COS cells can phagocytose in a manner similar to that of professional phagocytes. We have further analyzed the capacity of the three classes of FcγR to phagocytose, placing special emphasis on the FcγRIA–γ chain complex, which allowed us to examine independently the roles of the ligand-binding unit (FcγRIA) and the signaling unit (γ chain). We found that receptor complexes containing a conserved tyrosine activation motif (ITAM), as found in the cytoplasmic domain of FcγRIIA and in the γ chain associated with FcγRIA and FcγRIIIA, readily internalized target particles. In contrast, FcγRIA alone, having no ITAM, was unable to internalize target particles efficiently, but did mediate pseudopod extension. Cotransfection of γ chain with FcγRIA restored the ability of the receptor to internalize target particles. A mutant FcγRIA in which the cytoplasmic domain had been deleted was also capable of mediating pseudopod extension, showing that neither the γ chain nor the cytoplasmic domain of FcγRIA were required for this step. Cytochalasin D, an inhibitor of actin polymerization, blocked particle internalization by all FcγR, but did not block pseudopod extension. Staining the FcγRIA COS cells for F-actin and for tyrosine phosphoproteins, we found that actin did not polymerize during FcγRIA-mediated pseudopod extension, nor were tyrosine kinases activated. Our data suggest that pseudopod extension and internalization are functionally distinct steps mediated through different pathways.Fc receptor for IgG (FcγR)-mediated phagocytosis is the process whereby IgG-coated particulate pathogens are internalized and moved to lysosomes for degradation. The mechanism requires fine coordination of the action of a variety of molecules including cell membrane receptors, signaling molecules, and cytoskeleton components. The receptors initiate the process by a zipper mechanism whereby sequential interaction of particle-bound IgG with phagocyte FcγR stimulates the spreading of pseudopods around the particle resulting in its eventual engulfment (1). Complete circumferential coating of the target particle by ligand is required, and FcγR additional to those involved in the initial binding of the particle are recruited during the process (2).Several characteristics of the intracellular mechanism of FcγR-mediated phagocytosis have been described. Engulfment of attached particles requires actin polymerization as demonstrated by the blockade of phagocytosis by inhibitors of actin polymerization such as cytochalasin D. Along with F-actin, tyrosine phosphoproteins accumulate beneath attached IgG-coated particles before FcγR-mediated internalization. Both F-actin accumulation and particle internalization can be blocked by tyrosine kinase inhibitors, indicating that the signal pathway requires tyrosine kinases (3). Indeed, several nonreceptor tyrosine kinases of the src and syk family have been coisolated with FcγR, and their activities have been shown to increase after FcγR clustering (46). Other signaling molecules such as protein kinase C and phosphoinositide-3 kinase have also been implicated in FcγR-mediated phagocytosis (7, 8).FcγR comprise a family of integral membrane glycoproteins with three standard components, i.e., an extracellular ligand-binding portion consisting of two or three Ig-like domains, a short hydrophobic transmembrane region, and a cytoplasmic tail. In humans, three classes (I, II, and III) of these receptors have been characterized, differing in fine structure and affinity for IgG, and within these classes a total of eight subclasses (A, B, C) exist, each encoded by a separate gene (9). Of the eight FcγR in humans, three have convincingly been shown to mediate phagocytosis (FcγRIA, FcγRIIA, and FcγRIIIA; reference 10). Two of these three associate noncovalently with a common subunit, γ chain. A common amino acid sequence is present in the γ chain associating with FcγRIA and FcγRIIIA and in the cytoplasmic tail of FcγRIIA. This conserved motif, designated immunoreceptor tyrosine activation motif (ITAM)1, links receptor clustering to the activation of tyrosine kinases (11– 13). Upon FcγR clustering, the tyrosines of the ITAM are phosphorylated (6, 14, 15) and, in turn, serve as docking sites for signaling proteins containing src homology 2 domains (16, 17).Analyzing the phagocytic mechanism of a single FcγR class has been difficult because monocytes and macrophages generally express multiple classes of FcγR. Therefore, structure–function studies of individual receptor classes have required expression of single FcγR classes into FcγR negative cells such as COS cell fibroblasts. This approach has allowed analysis of the phagocytic capacities of each receptor, the identification of receptor-associated molecules required for function (12, 1820), and of receptor structural sequences necessary for phagocytosis (2124).However, we have questioned two aspects of this model; first, whether phagocytosis by COS cells mimics the process as it occurs in a professional phagocyte, and second whether phase contrast and fluorescence microscopy alone are adequate indicators of bona fide phagocytosis. Therefore, before embarking intently upon the study of the COS cell model of phagocytosis, we critically scrutinized in these cells the four major steps of FcγR-mediated phagocytosis: receptor–ligand binding, pseudopod extension, internalization, and fusion of the phagosome with lysosomes, with four microscopic methods: phase contrast, fluorescence, confocal, and electron microscopy. Here we show that COS cells indeed complete these four steps in a manner morphologically similar to professional phagocytes, and we validate that phase contrast and fluorescence microscopy are reasonably accurate methods for estimating phagocytosis.We then focused on the signaling complex of FcγRIA with the γ chain to examine independently the effects of the ligand binding unit, FcγRIA, and the signaling unit, γ chain, in driving pseudopod extension and internalization. We found that although efficient particle internalization by the receptor complex requires the γ chain, pseudopod extension is mediated in the absence of γ chain and by a tailless FcγRIA. Moreover, pseudopod extension mediated by FcγRIA is not blocked by cytochalasin D, and F-actin does not polymerize nor do phosphotyrosine residues accumulate adjacent to pseudopod extensions. These data suggest that these two phases of phagocytosis, pseudopod extension and internalization, are functionally distinct steps mediated by different mechanisms.  相似文献   

15.
Migration of mature B lymphocytes within secondary lymphoid organs and recirculation between these sites are thought to allow B cells to obtain T cell help, to undergo somatic hypermutation, to differentiate into effector cells, and to home to sites of antibody production. The mechanisms that direct migration of B lymphocytes are unknown, but there is evidence that G protein–coupled receptors, and possibly chemokine receptors, may be involved. Stromal cell– derived factor (SDF)-1α is a CXC chemokine previously characterized as an efficacious chemoattractant for T lymphocytes and monocytes in peripheral blood. Here we show with purified tonsillar B cells that SDF-1α also attracts naive and memory, but not germinal center (GC) B lymphocytes. Furthermore, GC B cells could be converted to respond to SDF-1α by in vitro differentiation into memory B lymphocytes. Conversely, the migratory response in naive and memory B cells was significantly reduced after B cell receptor engagement and CD40 signaling. The receptor for SDF-1, CXC chemokine receptor 4 (CXCR4), was found to be expressed on responsive as well as unresponsive B cell subsets, but was more rapidly downregulated on responsive cells by ligand. Finally, messenger RNA for SDF-1 was detected by in situ hybridization in a layer of cells surrounding the GC. These findings show that responsiveness to the chemoattractant SDF-1α is regulated during B lymphocyte activation, and correlates with positioning of B lymphocytes within a secondary lymphoid organ.  相似文献   

16.
Sepsis survivors exhibit impaired responsiveness to antigen (Ag) challenge associated with increased mortality from infection. The contribution of follicular dendritic cells (FDCs) in the impaired humoral response in sepsis-surviving mice is investigated in this study. We demonstrated that mice subjected to sepsis from cecal ligation and puncture (CLP mice) have reduced NP-specific high-affinity class-switched Ig antibodies (Abs) compared with sham-operated control mice following immunization with the T cell–dependent Ag, NP-CGG. NP-specific germinal center (GC) B cells in CLP mice exhibited reduced TNF-α and AID mRNA expression compared with sham-operated mice. CLP mice showed a reduction in FDC clusters, a reduced binding of immune complexes on FDCs, and reduced mRNA expression of CR2, ICAM-1, VCAM-1, FcγRIIB, TNFR1, IKK2, and LTβR compared with sham-operated mice. Adoptive transfer studies showed that there was no B cell–intrinsic defect. In summary, our data suggest that the reduced Ag-specific Ab response in CLP mice is secondary to a disruption in FDC and GC B cell function.  相似文献   

17.
Sialylation of IgG Fc domain impairs complement-dependent cytotoxicity   总被引:1,自引:0,他引:1  
IgG molecules exert both pro- and antiinflammatory effector functions based on the composition of the fragment crystallizable (Fc) domain glycan. Sialylated IgG Fc domains have antiinflammatory properties that are attributed to their ability to increase the activation threshold of innate effector cells to immune complexes by stimulating the upregulation of the inhibitory Fcγ receptor IIB (FcγRIIB). Here, we report that IgG Fc sialylation of human monoclonal IgG1 molecules impairs their efficacy to induce complement-mediated cytotoxicity (CDC). Fc sialylation of a CD20-targeting antibody had no impact on antibody-dependent cellular cytotoxicity and did not change the affinity of the antibody for activating Fcγ receptors. In contrast, the presence of sialic acid abrogated the increased binding of C1q to Fc-galactosylated IgG1 and resulted in decreased levels of C3b deposition on the cell surface. Similar to monoclonal antibodies, sialic acid inhibited the increased C1q binding to galactosylated Fc fragments in human polyclonal IgG. In sera derived from patients with chronic inflammatory demyelinating polyneuropathy, an autoimmune disease of the peripheral nervous system in which humoral immune responses mediate tissue damage, induction of IgG Fc sialylation was associated with clinical disease remission. Thus, impairment of CDC represents an FcγR-independent mechanism by which Fc-sialylated glycovariants might limit proinflammatory IgG effector functions.  相似文献   

18.
The cytosolic SHP-1 and transmembrane CD45 protein tyrosine phosphatases (PTP) play critical roles in regulating signal transduction via the B cell antigen receptor (BCR). These PTPs differ, however, in their effects on BCR function. For example, BCR-mediated mitogenesis is essentially ablated in mice lacking CD45 (CD45), but is enhanced in SHP-1–deficient motheaten (me) and viable motheaten (mev) mice. To determine whether these PTPs act independently or coordinately in modulating the physiologic outcome of BCR engagement, we assessed B cell development and signaling in CD45-deficient mev (CD45/SHP-1) mice. Here we report that the CD45/SHP-1 cells undergo appropriate induction of protein kinase activity, mitogen-activated protein kinase activation, and proliferative responses after BCR aggregation. However, BCR-elicited increases in the tyrosine phosphorylation of several SHP-1–associated phosphoproteins, including CD19, were substantially enhanced in CD45/SHP-1, compared to wild-type and CD45 cells. In addition, we observed that the patterns of cell surface expression of μ, δ, and CD5, which distinguish the PTP-deficient from normal mice, are largely restored to normal levels in the double mutant animals. These findings indicate a critical role for the balance of SHP-1 and CD45 activities in determining the outcome of BCR stimulation and suggest that these PTPs act in a coordinate fashion to couple antigen receptor engagement to B cell activation and maturation.  相似文献   

19.
Mac-1 (αmβ2), a leukocyte adhesion receptor, has been shown in vitro to functionally interact with Fcγ receptors to facilitate immune complex (IC)–stimulated polymorphonuclear neutrophil (PMN) functions. To investigate the relevance of Mac-1–FcγR interactions in IC-mediated injury in vivo, we induced a model of Fc-dependent anti–glomerular basement membrane (GBM) nephritis in wild-type and Mac-1–deficient mice by the intravenous injection of anti-GBM antibody. The initial glomerular PMN accumulation was equivalent in Mac-1 null and wild-type mice, but thereafter increased in wild-type and decreased in mutant mice. The absence of Mac-1 interactions with obvious ligands, intercellular adhesion molecule 1 (ICAM-1), and C3 complement, is not responsible for the decrease in neutrophil accumulation in Mac-1– deficient mice since glomerular PMN accumulation in mice deficient in these ligands was comparable to those in wild-type mice. In vitro studies showed that spreading of Mac-1–null PMNs to IC-coated dishes was equivalent to that of wild-type PMNs at 5–12 min but was markedly reduced thereafter, and was associated with an inability of mutant neutrophils to redistribute filamentous actin. This suggests that in vivo, Mac-1 is not required for the initiation of Fc-mediated PMN recruitment but that Mac-1–FcγR interactions are required for filamentous actin reorganization leading to sustained PMN adhesion, and this represents the first demonstration of the relevance of Mac-1–FcγR interactions in vivo. PMN-dependent proteinuria, maximal in wild-type mice at 8 h, was absent in Mac-1 mutant mice at all time points. Complement C3–deficient mice also had significantly decreased proteinuria compared to wild-type mice. Since Mac-1 on PMNs is the principal ligand for ic3b, an absence of Mac-1 interaction with C3 probably contributed to the abrogation of proteinuria in Mac-1–null mice.  相似文献   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号