Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Pharmaceutics ; 15(4)2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37111784

ABSTRACT

Calcineurin inhibitors have improved graft survival in solid-organ transplantation but their use is limited by toxicity, requiring a switch to another immunosuppressor in some cases. Belatacept is one option that has been shown to improve graft and patient survival despite being associated with a higher risk of acute cellular rejection. This risk of acute cellular rejection is correlated with the presence of belatacept-resistant T cells. We performed a transcriptomic analysis of in vitro-activated cells to identify pathways affected by belatacept in belatacept-sensitive cells (CD4+CD57-) but not in belatacept-resistant CD4+CD57+ T cells. mTOR was significantly downregulated in belatacept-sensitive but not belatacept-resistant T cells. The inhibition of mTOR strongly decreases the activation and cytotoxicity of CD4+CD57+ cells. In humans, the use of a combination of mTOR inhibitor and belatacept prevents graft rejection and decreases the expression of activation markers on CD4 and CD8 T cells. mTOR inhibition decreases the functioning of belatacept-resistant CD4+CD57+ T cells in vitro and in vivo. It could potentially be used in association with belatacept to prevent acute cellular rejection in cases of calcineurin intolerance.

2.
Front Med (Lausanne) ; 9: 949833, 2022.
Article in English | MEDLINE | ID: mdl-36072942

ABSTRACT

Background: Acute rejection rate is low after simultaneous liver-kidney transplantation (SLKT), leading some groups to minimize immunosuppressive (IS) regimens. However, the impact of preformed (pDSA) or de novo donor-specific antibodies (dnDSA) on the graft remains unclear. Methods: We performed a retrospective analysis of 102 consecutive SLKT patients to study the impact of anti-HLA antibodies. Results: Anti-HLA antibodies were detected in 75 recipients (class I 23.8%, both classes I and II 23.8%, and class II 14.3%). In total, 42.8% of the patients had pDSA and 21.7% developed dnDSA. Overall patient survival at 1-3 and 5 years, was respectively 88, 84, and 80%. Acute rejection occurred respectively in 3 (2.9%) liver and 6 kidney (5.9%) recipients. pDSA with titers over 10,000 mean fluorescence intensity (14.3%) was associated with lower patient survival (40 vs. 82%) but not with acute rejection. In a multivariable Cox regression analysis, the risk of death was associated with maleness, the highest titer of pDSA (p < 0.0007) or the sum of pDSA >10,000. Renal function did not differ between patients with class I pDSA (p = 0.631) and those with class II pDSA (p = 0.112) or between patients with and without a positive cross-match (p = 0.842). dnDSA were not associated with acute rejection, graft dysfunction or patient survival. IS minimization was not associated with rejection, graft dysfunction or death. Conclusion: In SLKT, high levels of pDSA >10,000 were associated with lower patient survival, but not rejection or graft survival. Minimization of maintenance immunosuppression regimen was not associated with a poorer outcome.

3.
Am J Transplant ; 22(2): 489-503, 2022 02.
Article in English | MEDLINE | ID: mdl-34431219

ABSTRACT

Belatacept was developed to replace calcineurin inhibitors in kidney transplantation. Its use is associated with better kidney transplant function, a lower incidence of anti-donor antibodies and higher graft survival. However, it is also associated with a higher risk of cellular rejection. We studied the activation and proliferation mechanisms of belatacept-resistant T lymphocytes (TLs), to identify new pathways for control. We performed a transcriptomic analysis on CD4+ CD57+ PD1- memory TLs, which are responsible for a higher incidence of graft rejection, after allogeneic stimulation with activated dendritic cells (aDCs) in the presence or absence of belatacept. After six hours of contact with aDCs, the (CD4+ CD57+ PD1- ) (CD4+ CD57+ PD1+ ) and (CD4+ CD57- ) lymphocytes had different transcriptional profiles with or without belatacept. In the CD4+ CD57+ PD1- population, the IFNα-dependent activation pathway was positively overrepresented, and IRF7 transcript levels were high. IRF7 was associated with IFNα/ß and IL-6 regulation. The inhibition of both these cytokines in a context of belatacept treatment inhibited the proliferation of CD4+ CD57+ PD1- T cells. Our results show that IRF7 is rapidly upregulated in belatacept-resistant CD4+ CD57+ PD1- TLs. The inhibition of type I IFN or IL-6 in association with belatacept treatment reduces the proliferation of belatacept-resistant TLs, paving the way for new treatments for use in organ transplantation.


Subject(s)
Immunosuppressive Agents , Kidney Transplantation , Abatacept/pharmacology , Cell Proliferation , Graft Rejection/etiology , Graft Survival , Immunosuppressive Agents/pharmacology , Kidney Transplantation/adverse effects
4.
Front Immunol ; 11: 875, 2020.
Article in English | MEDLINE | ID: mdl-32477353

ABSTRACT

Introduction: Focal and segmental glomerulosclerosis (FSGS) is a frequent form of glomerulonephritis that may be caused by a soluble permeability factor and regulated by the immune system. We previously described a soluble form of calcium/calmodulin-dependent serine/threonine kinase (CASK) acting as a permeability factor in patients with recurrent FSGS (rFSGS). Here, we aimed to identify the immune cells associated with CASK secretion in patients with rFSGS. Methods: FACS, western blotting and immunoprecipitation were performed to detect CASK in peripheral blood mononuclear cells, including CD3+, CD20+, and CD14+subsets, from patients with rFSGS, healthy donors, transplant patients and patients with nephrotic syndrome due to diabetes mellitus, and in KHM2 cells. Results: CASK was produced mostly by monocytes in patients with rFSGS but not by T or B lymphocytes. It was not detectein cells from control patients. CASK was also produced and secreted by M2 polarized macrophages and KMH2 cells, but not by M1 polarized macrophages. CASK secretion was not not inhibited by brefeldin A, suggesting an absence of classical secretion pathway involvement. Within cells, CASK was partly colocalized with ALIX, a molecule involved in exosome development, and these two molecules were coprecipitated from M2 macrophages. Moreover, exosomes derived from M2 macrophages induced podocyte cytoskeleton alterations and increased podocyte motility. Conclusion: These results suggest that the soluble permeability factor CASK is secreted by monocytes and M2 macrophages, via exosomes, to alter the glomerular filtration barrier in rFSGS.


Subject(s)
Glomerulosclerosis, Focal Segmental/immunology , Guanylate Kinases/immunology , Macrophages/metabolism , Adult , Aged , Animals , Biological Transport , Exosomes , Female , Glomerulosclerosis, Focal Segmental/blood , Guanylate Kinases/metabolism , Humans , Macrophages/immunology , Male , Middle Aged , Recurrence
5.
PLoS One ; 14(7): e0219353, 2019.
Article in English | MEDLINE | ID: mdl-31356645

ABSTRACT

INTRODUCTION: Focal and Segmental GlomeruloSclerosis (FSGS) can cause nephrotic syndrome with a risk of progression to end-stage renal disease. The idiopathic form has a high rate of recurrence after transplantation, suggesting the presence of a systemic circulating factor that causes glomerular permeability and can be removed by plasmapheresis or protein-A immunoadsorption. RESULTS: To identify this circulating factor, the eluate proteins bound on therapeutic immunoadsorption with protein-A columns were analyzed by comparative electrophoresis and mass spectrometry. A soluble form of calcium/calmodulin-dependent serine protein kinase (CASK) was identified. CASK was immunoprecipitated only in the sera of patients with recurrent FSGS after transplantation and not in control patients. Recombinant-CASK (rCASK) induced the reorganization of the actin cytoskeleton in immortalized podocytes, a redistribution of synaptopodin, ZO-1,vinculin and ENA. rCASK also induced alterations in the permeability of a monolayer of podocytes and increased the motility of pdodocytes in vitro. The extracellular domain of CD98, a transmembrane receptor expressed on renal epithelial cells, has been found to co-immunoprecipitated with rCASK. The invalidation of CD98 with siRNA avoided the structural changes of rCask treated cells suggesting its involvement in physiopathology of the disease. In mice, recombinant CASK induced proteinuria and foot process effacement in podocytes. CONCLUSION: Our results suggest that CASK can induce the recurrence of FSGS after renal transplantation.


Subject(s)
Glomerulosclerosis, Focal Segmental/blood , Guanylate Kinases/blood , Kidney Transplantation , Adult , Animals , Cells, Cultured , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Female , Fusion Regulatory Protein-1/metabolism , Glomerulosclerosis, Focal Segmental/complications , Humans , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Male , Membranes/metabolism , Membranes/ultrastructure , Mice , Middle Aged , Podocytes/metabolism , Podocytes/pathology , Podocytes/ultrastructure , Protein Binding , Proteinuria/complications , Recurrence
6.
Stem Cells Dev ; 28(1): 44-55, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30328799

ABSTRACT

Mesenchymal stem cells (MSCs) are powerful immunomodulators that regulate the diverse functions of immune cells involved in allogeneic reactions, such as T cells and natural killer (NK) cells, through cell-cell contact or secreted factors. Exosomes secreted by MSCs may be involved in their regulatory functions, providing new therapeutic tools. Here, we showed that fetal liver (FL) MSC-derived exosomes inhibit proliferation, activation, and cytotoxicity of NK cells. Exosomes bearing latency associated peptide (LAP), TGFß, and thrombospondin 1 (TSP1), a regulatory molecule for TGFß, induced downstream TGFß/Smad2/3 signaling in NK cells. The inhibition of TGFß, using a neutralizing anti-TGFß antibody, restored NK proliferation, differentiation, and cytotoxicity, demonstrating that FL-MSC-derived exosomes exert their inhibition on NK cell function via TGFß. These results suggest that FL-MSC-derived exosomes regulate NK cell functions through exosome-associated TGFß.


Subject(s)
Cell Communication , Exosomes/metabolism , Human Embryonic Stem Cells/metabolism , Killer Cells, Natural/immunology , Liver/cytology , Mesenchymal Stem Cells/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cells, Cultured , Human Embryonic Stem Cells/cytology , Humans , Liver/embryology , Mesenchymal Stem Cells/cytology , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
7.
Front Immunol ; 9: 787, 2018.
Article in English | MEDLINE | ID: mdl-29740433

ABSTRACT

B cells play a major role in the antibody-mediated rejection (AMR) of solid organ transplants, a major public health concern. The germinal center (GC) is involved in the generation of donor-specific antibody-producing plasma cells and memory B cells, which are often poorly controlled by current treatments. Myeloid cell leukemia-1 (Mcl-1), an antiapoptotic member of the B-cell lymphoma-2 family, is essential for maintenance of the GC reaction and B-cell differentiation. During chronic AMR (cAMR), tertiary lymphoid structures resembling GCs appear in the rejected organ, suggesting local lymphoid neogenesis. We report the infiltration of the kidneys with B cells expressing Mcl-1 in patients with cAMR. We modulated GC viability by impairing B-cell receptor signaling, by spleen tyrosine kinase (SYK) inhibition. SYK inhibition lowers viability and Mcl-1 protein levels in Burkitt's lymphoma cell lines. This downregulation of Mcl-1 is coordinated at the transcriptional level, possibly by signal transducer and activator of transcription 3 (STAT3), as shown by (1) the impaired translocation of STAT3 to the nucleus following SYK inhibition, and (2) the lower levels of Mcl-1 transcription upon STAT3 inhibition. Mcl-1 overproduction prevented cells from entering apoptosis following SYK inhibition. In vitro studies with primary tonsillar B cells confirmed that SYK inhibition impaired cell survival and decreased Mcl-1 protein levels. It also impaired B-cell activation and immunoglobulin G secretion by tonsillar B cells. These findings suggest that the SYK-Mcl-1 pathway could be targeted, to improve graft survival by manipulating the humoral immune response.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , Graft Rejection/immunology , Myeloid Cell Leukemia Sequence 1 Protein/immunology , Syk Kinase/immunology , Antibody Formation/immunology , Germinal Center/immunology , Humans , Immunity, Humoral/immunology , Lymphocyte Activation/immunology , Syk Kinase/antagonists & inhibitors
8.
Drugs ; 76(17): 1589-1600, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27785759

ABSTRACT

Long-term survival of solid allografts depends on both immunosuppressive efficacy and reducing the side effects associated with these therapies. Immunotherapies developed over the past 15 years to prevent organ rejection have greatly improved cardiovascular and renal function compared with classical therapies, such as calcineurin inhibitors and corticosteroids. Immunotherapies that target T cells through the co-stimulation blockade (CTLA-4-Ig) improve renal function and the survival of grafts and patients, but are associated with higher rates of T-cell-mediated acute rejection. Improvements to safe and efficacious therapeutic options could combine a co-stimulation blockade with a depleting immunotherapy. Herein, we describe the clinical outcomes and the likely causes of defects in the co-stimulation blockade, and comment on new therapeutic strategies to overcome these. Great progress has been made to optimize immunotherapy using the co-stimulation blockade, but the therapeutic combinations should be assessed further.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Calcineurin Inhibitors/therapeutic use , Graft Rejection/prevention & control , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/adverse effects , T-Lymphocytes/drug effects , Abatacept/therapeutic use , Graft Survival/drug effects , Humans , Immunotherapy/methods
9.
Cell Calcium ; 59(4): 198-207, 2016 04.
Article in English | MEDLINE | ID: mdl-27020659

ABSTRACT

Migration capacity is essential for dendritic cells (DCs) to present antigen to T cells for the induction of immune response. The DC migration is supposed to be a calcium-dependent process, while not fully understood. Here, we report a role of the KCa3.1/IK1/SK4 channels in the migration capacity of both immature (iDC) and mature (mDC) human CD14(+)-derived DCs. KCa3.1 channels were shown to control the membrane potential of human DC and the Ca(2+) entry, which is directly related to migration capacities. The expression of migration marker such as CCR5 and CCR7 was modified in both types of DCs by TRAM-34 (100nM). But, only the migration of iDC was decreased by use of both TRAM-34 and KCa3.1 siRNA. Confocal analyses showed a close localization of CCR5 with KCa3.1 in the steady state of iDC. Finally, the implication of KCa3.1 seems to be limited to the migration capacities as T cell activation of DCs appeared unchanged. Altogether, these results demonstrated that KCa3.1 channels have a pro-migratory effect on iDC migration. Our findings suggest that KCa3.1 in human iDC play a major role in their migration and constitute an attractive target for the cell therapy optimization.


Subject(s)
Calcium/metabolism , Cell Movement , Dendritic Cells/cytology , Dendritic Cells/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Cell Movement/drug effects , Dendritic Cells/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Pyrazoles/pharmacology
10.
J Immunol ; 192(12): 5660-70, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24829413

ABSTRACT

Human dendritic cells (hDCs) produce IL-2 and express IL-2R α-chain (CD25), but the role of IL-2 in DC functions is not well defined. A recent study suggested that the main function of CD25 on hDCs was to transpresent IL-2 to activate T lymphocytes. Our results demonstrate the expression of the three chains of the IL-2R on hDCs and that IL-2 induces STAT5 phosphorylation. Interestingly, use of inhibitors of p-STAT5 revealed that IL-2 increases LPS-induced IFN-γ through STAT5 phosphorylation. Finally, we report that IL-2 increases the ability of hDCs to activate helpless CD8(+) T cells, most likely because of IL-2-triggered IFN-γ synthesis, as we previously described. For the first time, to our knowledge, we disclose that IL-2 induces monocyte-derived hDC's functional maturation and activation through IL-2R binding. Interestingly, our study suggests a direct effect of anti-CD25 mAbs on hDCs that may contribute to their clinical efficacy.


Subject(s)
Dendritic Cells/immunology , Interferon-gamma/immunology , Interleukin-2/immunology , STAT5 Transcription Factor/immunology , Antibodies/pharmacology , Dendritic Cells/cytology , Female , Humans , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-2 Receptor alpha Subunit/immunology , Lipopolysaccharides/pharmacology , Male , Monocytes/cytology , Monocytes/immunology , Phosphorylation/drug effects , Phosphorylation/immunology
11.
Int Immunol ; 26(3): 173-81, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24222014

ABSTRACT

Regulatory T cells (Treg) play a crucial role in controlling immunity and transplant rejection. Two main groups of Treg have been described: antigen-induced Treg (iTreg) and natural Treg (nTreg). The ways to induce and the mechanisms of action of Treg subsets remained ill defined, particularly for their effects on CD8(+) T cells. CD8(+) T cells are major agents in the rejection of allografts; the aim of this study is to investigate the effects exerted on CD8(+) T cells by human CD4(+) iTreg induced by mycophenolic acid-treated dendritic cells. iTreg suppress the proliferation of CD8(+) T cells by allogeneic cell-cell interaction with mature dendritic cells and irrespectively of the TCR specificity of the CD8(+) T cells and cell-cell contact of iTreg with CD8(+) T cells. In our model, this suppression is independent of the action of IL-10 and TGF-ß1. iTreg were able to modify phenotype and inhibited IFN-γ and TNF-α secretion by CD8(+) T cells. Most interestingly, iTreg inhibit the synthesis of perforin and of granzymes A and B by CD8(+) T cells and impaired their cytotoxicity against allogeneic targets. In summary, our study showed the involvement of iTreg in the down-regulation of cytotoxic responses mediated by CD8(+) T cells in an allospecific context. Following studies that have shown the existence of a regulation control exerted by iTreg on CD4(+) T cells and dendritic cells, this work ultimately shows that this regulation can reach CD8(+) T-cell functions.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/drug effects , Mycophenolic Acid/pharmacology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , CD4 Antigens/metabolism , Cell Communication , Cell Differentiation , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Granzymes , Humans , Immunosuppression Therapy , Interferon-gamma/metabolism , Lymphocyte Activation , Perforin/metabolism , T-Cell Antigen Receptor Specificity/immunology , Transplantation Immunology , Tumor Necrosis Factor-alpha/metabolism
12.
Transpl Immunol ; 28(2-3): 120-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23507258

ABSTRACT

Rabbit antithymocyte globulin (rATG; Thymoglobulin(®)) is currently used to prevent acute rejection in kidney transplantation. The dose and regimen of rATG have not been optimized. Moreover, the impact of different treatment regimens on T-cell phenotype reconstitution remains unknown. We conducted a prospective randomized study of 17 renal transplant patients to determine the pharmacokinetics of total and active (bound to human cells) rATG and T-cell phenotype reconstitution after rATG administration. Patients received rATG at a total dose of 6mg/kg, administered either as 1.5mg/kg/day on days 0-3 (Group 1, n=8) or 3mg/kg on days 0 and 3 (Group 2, n=9). All patients received tacrolimus, mycophenolate mofetil and steroids. Blood samples were assayed for total rATG by enzyme linked immunosorbent assay and active rATG by flow cytometry. Maximum concentrations and terminal half-lives were similar between the two groups but at month 3 Group 1 had significantly lower values for total rATG (concentration was 6.2±1.1µg/mL versus 10.2±2.9µg/mL in Group 2, p=0.027) and total rATG dose-normalized AUC (374±83dayg/mL versus 508±149dayg/mL in Group 2, p=0.046). Time to sub-therapeutic levels (<1µg/mL) of active rATG was significantly shorter in Group 1 (18.75±6.9days versus 20±7.5days in Group 2, p<0.001). rATG induced significant depletion followed by slow reconstitution of CD3(+), CD4(+) and CD8(+) cells, with no marked differences between groups. B-cell count was unaffected, whereas CD3(-)CD56(+) NK-cell depletion was observed in both groups. rATG induced a significant decrease in the proportion of naïve CD4(+) T-cells, which plateaued after month 1 in Group 1 and after month 6 in Group 2. The proportion of central memory CD4(+) T-cells increased to a similar extent in both groups (Group 1: 38±18% at baseline, 74±23% at one year, p=0.009; Group 2: 32±14% at baseline, 65±14% at one year, p=0.001). In conclusion, our results suggest that the dosing regimen for rATG induction influences pharmacokinetic parameters without affecting the quality of immune reconstitution.


Subject(s)
Antilymphocyte Serum/pharmacology , Animals , Female , Graft Rejection/prevention & control , Humans , Immunity, Innate/drug effects , Immunologic Memory/drug effects , Kidney Transplantation , Male , Middle Aged , Rabbits , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
Blood ; 116(16): 3089-98, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20644110

ABSTRACT

The high frequency of allogeneic reactive CD8(+) T cells in human and their resistance to immunosuppression might be one of the reasons why successful tolerance-inducing strategies in rodents have failed in primates. Studies on the requirement for T-helper cells in priming CD8(+) T-cell responses have led to disparate findings. Recent studies have reported CD8(+)-mediated allograft rejection independently of T-helper cells; however, the mechanisms that govern the activation of these T cells are far from being elucidated. In this study, we demonstrated that lipopolysaccharide-treated dendritic cells (DCs) were able to induce proliferation and cytotoxic activity of allogeneic CD8(+) T cells independently of CD4(+) T cells, while adding mycophenolic acid (MPA) to LPS abolished this capacity and resulted in anergic CD8(+) T cells that secreted high levels of interleukin-4 (IL-4), IL-5, IL-10, and transforming growth factor-ß. Interestingly, we demonstrated that MPA inhibited the LPS-induced synthesis of tumor necrosis factor-α, IL-12, and interferon-γ (IFN-γ) in DCs. Importantly, we found that adding exogenous IFN-γ to MPA restored both the synthesis of cytokines and the ability to activate CD8(+) T cells. However, adding IL-12 or tumor necrosis factor-α had no effect. These results suggest that IFN-γ has an important role in licensing DCs to prime CD4-independent CD8 allogeneic T cells via an autocrine loop.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Interferon-gamma/immunology , Lymphocyte Activation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Coculture Techniques , Cytokines/immunology , Dendritic Cells/drug effects , Enzyme Inhibitors/pharmacology , Humans , Lipopolysaccharides/immunology , Mycophenolic Acid/pharmacology , Transforming Growth Factor beta/immunology , Tumor Necrosis Factor-alpha/immunology
14.
J Immunol ; 185(2): 1028-36, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20562265

ABSTRACT

Accumulating evidence suggests that CD4 help is needed at the memory stage to mount effective secondary CD8 T cell responses. In this paper, we report that memory CD4 T cells can provide efficient help to memory CD8 T cells after interaction of the two lymphocytes with distinct dendritic cells. Provision of help to CD8 T cells required direct cell-cell contact and involved both IL-2 and CD40 ligation, within a CD4-CD8 T cell synapse. Thus, following antigenic interaction with APCs, activated memory CD4 and CD8 T cells appear to separate from their respective APCs before meeting each other for help provision, regardless of their Ag specificity. CD4 help for memory CD8 T cells therefore appears to be conditioned primarily not by Ag specificity but by activation status.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Communication/immunology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Cytotoxicity, Immunologic/immunology , Dendritic Cells/cytology , Female , Flow Cytometry , Granzymes/metabolism , Immunologic Memory/immunology , Interleukin-2/immunology , Interleukin-2/metabolism , Lymphocyte Activation/immunology , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...