KEL RBC Transfusion Induces IgG Anti-KEL Antibodies Independent Of CD4 T Cells

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 41-41 ◽  
Author(s):  
Sean R. Stowell ◽  
Kathryn R. Girard-Pierce ◽  
Connie M Arthur ◽  
Nicole H. Smith ◽  
James C. Zimring ◽  
...  

Abstract Background While red blood cells (RBCs) transfusion can provide life saving therapy, patients who require chronic transfusion therapy may develop RBC alloantibodies that limit the availability of compatible RBCs for future transfusion and increase the risk of hemolytic transfusion reactions. However, not all patients generate alloantibodies following RBC exposure. Among factors that potentially influence RBC alloantibody formation, several studies suggest that a recipient’s MHC class II repertoire may predict an individual’s likelihood of responding to a particular RBC alloantigen. However, whether MHC class II antigens are required for efficient alloantibody formation following RBC transfusion remains unknown. As a result, we examined the potential role of MHC class II in the development of RBC alloantibodies following transfusion in a murine model of KEL alloimmunization. Methods RBCs transgenically expressing the human KEL antigen specifically under a β-globin promoter (KEL RBCs) were transfused into C57BL/6, C57BL/6 MHC class II knock out (KO) or KEL transgenic control recipients. Following transfusion, blood was harvested on days 3, 5, 7, 14, 21 and 28 following transfusion and serum was analyzed for IgM or IgG anti-KEL antibodies by indirect immunofluorescence using flow cytometry with KEL and control C57BL/6 RBCs as targets. To deplete CD4 T cells, mice were injected with anti-CD4 (clone GK1.5) 4 and 2 days prior to transfusion. As a control, additional C57BL/6 recipients were similarly injected with an isotype control. C57BL/6 recipients were also injected in parallel with GK1.5 or isotype control followed by splenocyte examination for CD4 T cell depletion using anti-CD3 and an anti-CD4 clone that recognizes a different CD4 epitope than GK1.5 (clone RM4-5). All experiments were completed at least three times with 3–5 recipients per group per experiment. Results Transfusion of KEL RBCs resulted in significant IgM anti-KEL antibody formation that peaked approximately 5 days following transfusion in both C57BL/6 and C57BL/6 MHC class II KO recipients. Similarly, IgG anti-KEL antibodies could also be detected in C57BL/6 or C57BL/6 MHC class II KO as early as 7 days following transfusion and continued to rise to similar peak levels within 14 to 21 days following KEL RBC transfusion. Injection of GK1.5, but not isotype control antibody, depleted CD4 T cells to less than 1% of their original level. Transfusion of KEL RBCs into C57BL/6, CD4 depleted C57BL/6 or isotype control treated C57BL/6 resulted in similar levels of IgM anti-KEL antibody that peaked approximately 5 days following transfusion. Likewise, transfusion of KEL RBCs induced similar levels of IgG anti-KEL antibodies within 7 days following transfusion that also peaked between 14 and 21 days in C57BL/6, CD4 depleted C57BL/6 or isotype control treated C57BL/6 recipients. (All the above differences achieved a p value of <0.05) Conclusions Despite the potential role of CD4 T cells in facilitating RBC alloantibody formation, these results suggest that significant IgG RBC alloantibody can occur independent of MHC class II or CD 4 T cells. Although it remains possible that CD4 T cells become activated following RBC alloantigen exposure, the lack of CD4 T cell requirement in this model suggests that some patients may be capable of mounting a clinically significant immune response following RBC transfusion in the absence of CD4 T cell help. As a result, MHC antigen presentation of unique RBC alloantigens may not be necessary for RBC alloimmunization to occur. Disclosures: Zimring: Immucor Inc.: Research Funding; Terumo: Research Funding; Haemonetics: Consultancy; Cerus: Honoraria.

Blood ◽  
2006 ◽  
Vol 108 (1) ◽  
pp. 270-277 ◽  
Author(s):  
Bruno Martin ◽  
Chantal Bécourt ◽  
Boris Bienvenu ◽  
Bruno Lucas

The role of self-recognition in the maintenance of the peripheral CD4+ T-cell pool has been extensively studied, but no clear answer has so far emerged. Indeed, in studies of the role of self-major histocompatibility complex (MHC) molecules in CD4+ T-cell survival, several parameters must be taken into account when interpreting the results: (1) in a lymphopenic environment, observations are biased by concomitant proliferation of T cells arising in MHC-expressing mice; (2) the peripheral T-cell compartment is qualitatively and quantitatively different in nonlymphopenic, normal, and MHC class II-deficient mice; and (3) in C57BL/6 Aβ-/- mice (traditionally considered MHC class II-deficient), the Aα chain and the Eβ chain associate to form a hybrid AαEβ MHC class II molecule. In light of these considerations, we revisited the role of interactions with MHC class II molecules in the survival of peripheral CD4+ T cells. We found that the answer to the question “is self-recognition required for CD4+ T cells to survive?” is not a simple yes or no. Indeed, although long-term survival of CD4+ T cells does not depend on self-recognition in lymphopenic mice, interactions with MHC class II molecules are required for maintaining the peripheral CD4+ T-cell pool in a nonlymphopenic environment. (Blood. 2006;108:270-277)


2005 ◽  
Vol 202 (8) ◽  
pp. 1109-1119 ◽  
Author(s):  
Nagendra R. Hegde ◽  
Claire Dunn ◽  
David M. Lewinsohn ◽  
Michael A. Jarvis ◽  
Jay A. Nelson ◽  
...  

Human cytomegalovirus (HCMV) infects endothelial, epithelial, and glial cells in vivo. These cells can express MHC class II proteins, but are unlikely to play important roles in priming host immunity. Instead, it seems that class II presentation of endogenous HCMV antigens in these cells allows recognition of virus infection. We characterized class II presentation of HCMV glycoprotein B (gB), a membrane protein that accumulates extensively in endosomes during virus assembly. Human CD4+ T cells specific for gB were both highly abundant in blood and cytolytic in vivo. gB-specific CD4+ T cell clones recognized gB that was expressed in glial, endothelial, and epithelial cells, but not exogenous gB that was fed to these cells. Glial cells efficiently presented extremely low levels of endogenous gB—expressed by adenovirus vectors or after HCMV infection—and stimulated CD4+ T cells better than DCs that were incubated with exogenous gB. Presentation of endogenous gB required sorting of gB to endosomal compartments and processing by acidic proteases. Although presentation of cellular proteins that traffic into endosomes is well known, our observations demonstrate for the first time that a viral protein sorted to endosomes is presented exceptionally well, and can promote CD4+ T cell recognition and killing of biologically important host cells.


Author(s):  
Sophia Schulte ◽  
Janna Heide ◽  
Christin Ackermann ◽  
Sven Peine ◽  
Michael Ramharter ◽  
...  

Abstract Relatively little is known about the ex vivo frequency and phenotype of the P. falciparum-specific CD4+ T cell response in humans. The exported protein 1 (EXP1) is expressed by plasmodia at both, the liver stage and blood stage, of infection making it a potential target for CD4+ and CD8+ effector T cells. Here, a fluorochrome-labelled HLA-DRB1*11:01-restriced MHC class II tetramer derived from the P. falciparum EXP1 (aa62-74) was established for ex vivo tetramer analysis and magnetic bead enrichment in ten patients with acute malaria. EXP1-specific CD4+ T cells were detectable in nine out of ten (90%) malaria patients expressing the HLA-DRB1*11 molecule with an average ex vivo frequency of 0.11% (0-0.22%) of total CD4+ T cells. The phenotype of EXP1-specific CD4+ T cells was further assessed using co-staining with activation (CD38, HLA-DR, CD26), differentiation (CD45RO, CCR7, KLRG1, CD127), senescence (CD57) and co-inhibitory (PD-1, TIGIT, LAG-3, TIM-3) markers as well as the ectonucleotidases CD39 and CD73. EXP1-specific tetramer+ CD4+ T cells had a distinct phenotype compared to bulk CD4+ T cells and displayed a highly activated effector memory phenotype with elevated levels of co-inhibitory receptors and activation markers: EXP1-specific CD4+ T cells universally expressed the co-inhibitory receptors PD-1 and TIGIT as well as the activation marker CD38 and showed elevated frequencies of CD39. These results demonstrate that MHC class II tetramer enrichment is a sensitive approach to investigate ex vivo antigen-specific CD4+ T cells in malaria patients that will aid further analysis of the role of CD4+ T cells during malaria.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3847-3847
Author(s):  
Patricia E. Zerra ◽  
Seema R. Patel ◽  
Connie M. Arthur ◽  
Kathryn R. Girard-Pierce ◽  
Ashley Bennett ◽  
...  

Abstract Background: While red blood cell (RBC) transfusion can be beneficial, exposure to allogeneic RBCs can result in the development of RBC alloantibodies that can make it difficult to obtain compatible RBCs for future transfusions. Aside from phenotype matching protocols, no strategy currently exists that is capable of preventing RBC alloimmunization following therapeutic transfusion. As RBC alloantigens represent diverse determinants capable of driving distinct immune pathways, common immunological nodes must be identified in order to successfully prevent RBC alloimmunization against a variety of different alloantigens. Recent results demonstrate that marginal zone (MZ) B cells mediate anti-KEL antibody formation in the complete absence of CD4 T cells. However, whether MZ B cells similarly regulate RBC alloantibody formation against other RBC alloantigens remains unknown. As a result, we examined the role of MZ B cells and CD4 T cells in the development of RBC alloantibodies following exposure to the HOD (hen egg lysozyme, ovalbumin and duffy) antigen. Methods: Each recipient was transfused with HOD or KEL RBCs following either MZ B cell or CD4 T cell depletion using a cocktail of MZ B cell (anti-CD11a and anti-CD49d) or anti-CD4 depleting antibody, 4 and 2 days prior to transfusion. Control groups received isotype control injections in parallel. MZ B cell deficient (CD19cre/+ X Notch2flx/flx) and CD4 T cell deficient (MHC class II knockout) recipients were also used to examine the role of MZ B cells and CD4 T cells, respectively. Serum collected on days 5 and 14 post-transfusion was evaluated for anti-HOD or anti-KEL antibodies by incubating HOD or KEL RBCs with serum, followed by detection of bound antibodies using anti-IgM and anti-IgG and subsequent flow cytometric analysis. Evaluation of antibody engagement and overall survival of HOD or KEL RBCs was accomplished by labeling RBCs with the lipophilic dye, DiI, prior to transfusion, followed by examination for bound antibody and RBC clearance on days 5 and 14 post-transfusion by flow cytometry. Results: Similar to the ability of MZ B cell depletion to reduce anti-KEL antibody formation following KEL RBC exposure, depletion of MZ B cells significantly reduced anti-HOD IgM and IgG antibodies following HOD RBC transfusion. In contrast, injection of recipients with isotype control antibodies in parallel failed to prevent alloantibody formation following HOD or KEL RBC transfusion. Similar results were obtained following HOD or KEL RBC transfusion into recipients genetically deficient in MZ B cells. In contrast, although MZ B cells were required for HOD and KEL RBC-alloantibody formation, manipulation of CD4 T cells differentially impacted the ability of each antigen to induce alloantibodies. While transfusion of HOD or KEL RBCs resulted in robust IgM alloantibodies in the absence of CD4 T cells, depletion or genetic elimination of CD4 T cells significantly inhibited anti-HOD IgG antibody formation, while failing to impact IgG anti-KEL antibody formation. Consistent with this, while manipulation of CD4 T cells protected HOD RBCs from antibody deposition and subsequent RBC clearance, this same approach failed to similarly protect KEL RBCs following transfusion. In contrast, depletion of MZ B cells not only prevented detectable alloantibody production, but also completely protected HOD or KEL RBCs from antibody deposition and subsequent RBC clearance. Conclusion: These results suggest that while MZ B cells mediate a robust IgM antibody response following either KEL or HOD antigen exposure, MZ B cells appear to possess the capacity to orchestrate unique downstream IgG responses through CD4 T cell dependent and independent pathways contingent on target alloantigen. As a result, while manipulation of CD4 T cells may prevent alloantibody formation against some antigens, targeting this immune population inadequately prevents RBC alloantibody formation against all RBC antigens. As chronic transfusion therapy exposes recipients to a wide variety of alloantigens, these results suggest that MZ B cells may represent a central initiating node that governs RBC alloimmunization against a variety of RBC alloantigens, and may therefore serve as a useful target in preventing alloantibody formation in chronically transfused individuals. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 23-23
Author(s):  
Muhammad Haroon Shaikh ◽  
Juan Gamboa Vargas ◽  
Josefina Peña Mosca ◽  
Duc Dung Le ◽  
Hermann Einsele ◽  
...  

Allogeneic T cell priming is considered as an essential event determining the outcome of allogeneic hematopoietic stem cell transplantation (allo-HCT), ideally triggering anti-leukemic responses (GvL effect) or, at worst, causing life-threatening acute graft-versus-host disease (aGvHD). During aGvHD initiation, alloreactive T cells are activated by host antigen presenting cells (APCs), rapidly expand and subsequently exert tissue damage. Recently, it was discovered that in absence of host hematopoietic APCs, aGvHD cannot be prevented, suggesting a crucial role of non-hematopoietic APCs for priming alloreactive T cells (Toubai et al., Blood 2012, Li et al., J Immunol. 2012, Koyama et al., Nat Med 2012). However, the exact location and identity of host non-hematopoietic APCs triggering alloreactive T cell responses remains controversial and needs to be proven in vivo. Fibroblastic reticular cells (FRCs) have shown to provide the crucial delta-like notch ligand to alloreactive T cells (Chung et al., JCI 2017) in aGvHD, therefore we investigated the role of FRCs MHC class II in aGvHD and their potential role as non-hematopoietic APCs in MHC class II dependent manner. In vitro cultured FRCs cell line as well as FRCs from lethally irradiated mice upregulate MHCII and co-stimulatory molecules. Moreover, FACS sorted FRCs (CD45-CD24-CD31-gp38+) were able to process DQ-OVA via MHC class II machinery, indicating that FRCs have the potential to activate CD4+ T cells. Employing allo-HCT mouse models in combination with flow cytometry and advanced microscopy techniques, we explored early alloreactive T cells activation initially in a myeloablatively conditioned MHC major mismatch allo-HCT setting (FVB/NàC57Bl/6). We generated MHCIIΔCcl19 mice with a Ccl19-intrinsic deletion of MHC class II on all Ccl19 expressing reticular lineage cells by crossing mice with floxed H2-Ab1 gene (H2-Ab1fl) with a mouse expressing Cre recombinase under the control of the Ccl19 promoter (Ccl19Cre). On day+3 after allo-HCT, CD4+ T cells activation (CD44 and CD25 expression) and proliferation (Ki67 expression and CFSE dilution) did not differ in the MHCIIΔCcl19 mice from H2-Ab1fl wildtype littermates. To further elucidate FRCs MHC class II in aGvHD milieu, we utilized iFABP-tOVA transgenic model in which OVA is expressed by intestinal epithelial cells as well as ectopically by FRCs of the lymphoid organs. OT-II cells transferred from RagΔ background mice failed to proliferate in the mLNs of lethally irradiated iFABP-tOVA, whereas excessive proliferation was observed in CD11c.DOG mice (where OVA is presented by CD11c-expressing cells). Taken together these results indicate that MHCII on FRCs does not play a role in direct antigen presentation and CD4+ T cell activation. Next, we asked whether MHCII on FRCs influences alloreactivity of CD4+ T cells in the symptomatic phase of aGvHD. Indeed, in MHCIIΔCcl19 mice, CD4+ T cells expressed higher levels of effector molecules: CD44 and CD127 as well as the proliferation marker Ki67 on day +30 of allo-HCT. Furthermore, the proportion of donor CD90.1+CD4+FoxP3+ regulatory T cells (Tregs) were reduced in MHCIIΔCcl19 mice as compared to H2-Ab1fl wild-type littermates. This led to overall poor survival of MHCIIΔCcl19 mice by day+60 after allo-HCT. At this time point in MHCIIΔCcl19 mice CD4+ T cells displayed higher levels of CD44, CD127 and Ki67 and down-regulated PD-1 and Lag3. To further elucidate the effect of FRCs MHC class II on CD4+FoxP3+ donor Tregs, we transplanted CD90.1+CD4+CD25hi Tregs, TCD BM from FVB mice along with naïve luc+ CD90.1+CD4+ T cells from FVB.L2G85 mice. Tregs protected against aGvHD in H2-Ab1fl littermate controls whereas Tregs could not protect MHCIIΔCcl19 recipients rendering them susceptible to aGvHD and to poor overall survival. Conclusively, these results indicate for the first time that MHC class II on FRCs assists to maintain donor Tregs in the SLOs after allo-HCT. Conclusively, we propose a model in which FRCs promote T cell alloreactivity by providing notch ligands (Chung et al., JCI 2017) in the initiation phase and mitigate aGvHD by maintenance of Tregs via MHC class II in the aGvHD-effector phase. Disclosures Einsele: Janssen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Amgen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Bristol-Myers Squibb: Consultancy, Honoraria, Research Funding, Speakers Bureau; Novartis: Honoraria, Speakers Bureau; Takeda: Consultancy, Honoraria, Speakers Bureau; Sanofi: Consultancy, Honoraria, Research Funding, Speakers Bureau; GlaxoSmithKline: Honoraria, Research Funding, Speakers Bureau.


1989 ◽  
Vol 170 (6) ◽  
pp. 2135-2140 ◽  
Author(s):  
J S Murray ◽  
J Madri ◽  
J Tite ◽  
S R Carding ◽  
K Bottomly

The present results demonstrate that CD4+ T cells activated in the primary in vivo response to antigen produce distinct patterns of cytokines depending upon the MHC class II haplotype of the responding mice. I-As mice were found to selectively activate IL-2/IFN-gamma-producing CD4+ T cells, whereas I-Ab mice exhibited selective activation of IL-4-producing CD4+ T cells in response to collagen IV. The effector response phenotype was found to correlate with the cytokine phenotype of CD4+ T cells activated in vivo; IL-2/IFN-gamma-producing cells giving rise to proliferative (cell-mediated) responses, IL-4-producing cells leading to secondary IgG (humoral) responses. Together the data support the notion that the outcome of a given immune response (e.g., protection vs. onset, tolerance vs. autoimmunity) may be determined in part by the type of CD4+ T cells initially activated by antigen. Moreover, the present experiments demonstrate for the first time that polymorphism in class II MHC can determine such selective activation of different cytokine-producing CD4+ T cell phenotypes.


Blood ◽  
2012 ◽  
Vol 119 (17) ◽  
pp. 4073-4082 ◽  
Author(s):  
Katharina N. Steinitz ◽  
Pauline M. van Helden ◽  
Brigitte Binder ◽  
David C. Wraith ◽  
Sabine Unterthurner ◽  
...  

Abstract Today it is generally accepted that B cells require cognate interactions with CD4+ T cells to develop high-affinity antibodies against proteins. CD4+ T cells recognize peptides (epitopes) presented by MHC class II molecules that are expressed on antigen-presenting cells. Structural features of both the MHC class II molecule and the peptide determine the specificity of CD4+ T cells that can bind to the MHC class II–peptide complex. We used a new humanized hemophilic mouse model to identify FVIII peptides presented by HLA-DRB1*1501. This model carries a knockout of all murine MHC class II molecules and expresses a chimeric murine-human MHC class II complex that contains the peptide-binding sites of the human HLA-DRB1*1501. When mice were treated with human FVIII, the proportion of mice that developed antibodies depended on the application route of FVIII and the activation state of the innate immune system. We identified 8 FVIII peptide regions that contained CD4+ T-cell epitopes presented by HLA-DRB1*1501 to CD4+ T cells during immune responses against FVIII. CD4+ T-cell responses after intravenous and subcutaneous application of FVIII involved the same immunodominant FVIII epitopes. Interestingly, most of the 8 peptide regions contained promiscuous epitopes that bound to several different HLA-DR proteins in in vitro binding assays.


1999 ◽  
Vol 190 (4) ◽  
pp. 555-566 ◽  
Author(s):  
Mark D. Denton ◽  
Christopher S. Geehan ◽  
Steve I. Alexander ◽  
Mohamed H. Sayegh ◽  
David M. Briscoe

Activated vascular endothelial cells (ECs) express major histocompatibility complex (MHC) class II molecules in vitro and in vivo in acute and chronic allograft rejection. However, human ECs may be limited in their ability to effectively activate CD4+ T cells, because they do not express members of the B7 family (CD80 and CD86) of costimulatory molecules. In this study, we show that ECs promote the full activation of CD4+ T cells via trans-costimulatory interactions. By reverse transcriptase polymerase chain reaction, Western blot, and FACS® analysis, we could not detect the expression of CD80 and CD86 on activated ECs and found minimal expression on purified CD4+ T cells. In contrast, both CD80 and CD86 were expressed in allogeneic CD4+ T cell–EC cocultures. Expression of CD86 peaked at early times between 12 and 24 h after coculture, whereas CD80 was not expressed until 72 h. Addition of anti-CD86 but not anti-CD80 monoclonal antibodies to cocultures inhibited IL-2 production and the proliferation of CD4+ T cells to allogeneic donor human umbilical vein ECs (HUVECs), as well as to skin and lung microvascular ECs. Furthermore, we found that interferon γ–activated ECs but not untreated ECs induced mRNA and cell surface expression of CD80 and CD86 on CD4+ T cells, and these T cells were functional to provide a trans-costimulatory signal to autologous CD4+ T cells. Blockade of MHC class II and lymphocyte function–associated antigen 3 but not other EC cell surface molecules on IFN-γ–activated ECs inhibited the induction of CD86 on CD4+ T cells. Transmigration of purified populations of monocytes across EC monolayers similarly resulted in the induction of functional CD86, but also induced the de novo expression of the cytokines interleukin (IL)-1α and IL-12. In addition, EC-modified monocytes supported enhanced proliferation of allogeneic and autologous CD4+ T cells. Taken together, these data define the ability of the endothelium to modify CD4+ T cells and monocytes for trans-costimulatory events. This unique function of the endothelium in alloimmune T cell activation has functional consequences for the direct and the indirect pathways of allorecognition.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A546-A546
Author(s):  
Margaux Saillard ◽  
Amelie Cachot ◽  
Georg Alexander Rockinger ◽  
Philippe Guillaume ◽  
Julien Schmidt ◽  
...  

BackgroundWhile cancer immunotherapy has mainly focused on exploiting CD8 T cells given their role in the direct elimination of tumor cells, increasing evidence highlights the crucial roles played by CD4 T cells in anti-tumor immunity. However, their very low frequency, the lack of robust algorithms to predict peptide binding to MHC class II molecules and the high polymorphism of MHC class II molecules render the study and use of circulating tumor antigen-specific CD4 T cells challenging. In this regard, the HLA-DRB3*02:02 gene encoding an HLA allele that is expressed by half of the Caucasian population, offers a way to identify CD4 T cell-defined tumor antigens with broad cancer patient coverage.MethodsHere, we aim to identify, isolate and functionally characterize ‘quasi-universal’ human tumor antigen-specific HLA-DRB3*02:02-restricted CD4 T cells in cancer patients. Using an algorithm we recently developed in house,1 tumor-associated antigenic peptides binding to this allele are identified. We have generated a large collection of HLA-DRB3*02:02-restricted CD4 T cell clones of different tumor-antigen specificities. We will perform in vitro co-cultures of CD4 T cell clones with tumor cells to measure cytokine secretion, their tumor cell killing and their phenotypic profile (PD-1, TIM3, TIGIT, 4-1BB, CD40L, LAG3, VISTA, OX40). We will sequence and clone the TCR of the most promising candidates for adoptive cell transfer therapy. Lastly, we will directly evaluate the presence of these cells ex-vivo and longitudinally monitor them in patients.ResultsN/AConclusionsTogether, these results should contribute valuable targets for coordinated CD4 and CD8 T cell-based immunotherapy of cancer.ReferenceRacle, J., et al., Robust prediction of HLA class II epitopes by deep motif deconvolution of immunopeptidomes. Nat Biotechnol 2019. 37(11): p. 1283–1286.


Sign in / Sign up

Export Citation Format

Share Document