Ex Vivo Generation Of Functional Plasmacytoid and Myeloid Dendritic Cells Is Strongly Promoted By The Aryl Hydrocarbon Receptor Antagonist Stemregenin 1

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2025-2025
Author(s):  
Soley Thordardottir ◽  
Hangalapura Basav N. ◽  
Tim Hutten ◽  
Marta Cossu ◽  
Jan Spanholtz ◽  
...  

Abstract The prominent role of dendritic cells (DCs) in T cell activation is the rational for DC-based immunotherapy of cancer and infectious diseases. In cancer, DC therapy aims to induce tumor-specific effector T cell responses that can reduce or eliminate the tumor, and to develop immunological memory to control tumor relapse. So far, the vast majority of DC vaccination studies have been performed with DCs differentiated from monocytes (Mo-DCs) that are loaded with tumor-associated antigens (TAAs) or minor histocompatibility antigens (MiHA). This strategy has been reported to induce the expansion of antigen-specific CD4+ and/or CD8+ T cells in the majority of patients, however only a fraction of the patients develop clinical responses. Strategies to improve the potency of DC-based vaccines are to increase the stimulatory and migratory capacity of Mo-DCs, or to use alternative DC subtypes, such as naturally circulating plasmacytoid DCs (pDCs), BDCA1+ myeloid DCs (mDCs) or BDCA3+ mDCs. These DC subsets are potent inducers of antigen-specific T cell responses, and are therefore attractive cells to exploit for DC-based therapy. However, since their frequency in blood is very low, it is a challenge to obtain high enough numbers for immunotherapy. It would be advantageous if DCs, which are phenotypically and functionally similar to blood pDCs and mDCs, could be generated from CD34+ hematopoietic progenitor cells (HPCs). Interestingly, recent findings have indicated that the aryl hydrocarbon receptor (AhR) not only regulates toxic effects of environmental contaminants, but also plays a role in modulating hematopoiesis and the immune system. For instance, it has been reported that StemRegenin 1 (SR1), a small molecule inhibitor of AhR, promotes the ex vivo expansion of human CD34+ HPCs that are able to effectively engraft immunodeficient mice. Furthermore, differentiation of Langerhans cells and monocytes in vitro from HPCs can be inhibited by the addition of the AhR agonist VAF347. In light of these data, we investigated if we could generate DC subsets from CD34+ HPCs by supplementing SR1. Therefore, we cultured CD34+ HPCs in medium containing SCF, Flt3L, IL-6, TPO supplemented with 1 μM SR1 or DMSO as control. Interestingly, addition of SR1 explicitly promoted the emergence of pDCs (CD11c-HLA-DR+CD123hiBDCA2+BDCA4+ cells), BDCA1+ mDCs (Lin1-HLA-DR+BDCA1+BDCA3- cells) and BDCA3+ mDCs (Lin1-HLA-DR+BDCA1-BDCA3+ cells). After three weeks of culture, the frequency of these DC subsets was significantly higher in cultures with SR1 compared to control conditions; 2.9% vs. 0.04% for pDCs, 4.6% vs. 0.5% for BDCA1+ mDCs and 1.1% vs. 0.1% for BDCA3+ mDCs (n=3-5 donors). The average yield after three weeks of culture with SR1 starting from 105 CD34+ UCB cells was 3.8x106 pDCs, 5.3x106 BDCA1+ mDCs and 1.2x106 BDCA3+ mDCs (n=3-5 donors). Furthermore, SR1 also promoted the differentiation of DC subsets from CD34+ cells obtained from peripheral blood of G-CSF-mobilized donors. The average frequency of DCs in these SR1-cultures was 4.7%, 3.8% and 0.9% for pDCs, BDCA1+ and BDCA3+ mDCs, respectively (n=3 donors), which is comparable to the frequency obtained from UCB CD34+ cells. But the expansion potential of G-CSF-mobilized blood CD34+ HPCs was lower than that of UCB CD34+ cells, resulting in average DC yields of 0.6x106, 0.5x106 and 0.1x106 from 105 CD34+ cells (n=3). Flow cytometry analysis demonstrated that the SR1-induced pDCs and mDCs are phenotypically comparable to their naturally occurring counterpart in blood. Furthermore, the ex vivo-generated pDCs potently responded to stimulation with TLR7 and TLR9 ligands by secreting high amounts of IFN-α and upregulating CD83, CD80, CD86 and CCR7. The HPC-mDC subsets also upregulate CD80 and CD83 upon TLR3, TLR4 or TLR7/8 ligation. Finally, both the ex vivo-generated pDCs and mDCs induced potent allogeneic T cell responses and activated CD8+ effector T cells against hematopoietic-restricted MiHA. These findings demonstrate that our SR1 culture system not only allows detailed study of DC differentiation and molecular regulations in vitro, but it also offers the opportunity to evaluate the in vivo efficacy of cultured DC subsets upon vaccination into patients with cancer and viral infections. Disclosures: Spanholtz: Glycostem Therapeutics: Employment.

2020 ◽  
Vol 8 (2) ◽  
pp. e000691 ◽  
Author(s):  
Kelly-Anne Masterman ◽  
Oscar L Haigh ◽  
Kirsteen M Tullett ◽  
Ingrid M Leal-Rojas ◽  
Carina Walpole ◽  
...  

BackgroundDendritic cells (DCs) are crucial for the efficacy of cancer vaccines, but current vaccines do not harness the key cDC1 subtype required for effective CD8+ T-cell-mediated tumor immune responses. Vaccine immunogenicity could be enhanced by specific delivery of immunogenic tumor antigens to CD141+ DCs, the human cDC1 equivalent. CD141+ DCs exclusively express the C-type-lectin-like receptor CLEC9A, which is important for the regulation of CD8+ T cell responses. This study developed a new vaccine that harnesses a human anti-CLEC9A antibody to specifically deliver the immunogenic tumor antigen, NY-ESO-1 (New York esophageal squamous cell carcinoma 1), to human CD141+ DCs. The ability of the CLEC9A-NY-ESO-1 antibody to activate NY-ESO-1-specific naïve and memory CD8+ T cells was examined and compared with a vaccine comprised of a human DEC-205-NY-ESO-1 antibody that targets all human DCs.MethodsHuman anti-CLEC9A, anti-DEC-205 and isotype control IgG4 antibodies were genetically fused to NY-ESO-1 polypeptide. Cross-presentation to NY-ESO-1-epitope-specific CD8+ T cells and reactivity of T cell responses in patients with melanoma were assessed by interferon γ (IFNγ) production following incubation of CD141+ DCs and patient peripheral blood mononuclear cells with targeting antibodies. Humanized mice containing human DC subsets and a repertoire of naïve NY-ESO-1-specific CD8+ T cells were used to investigate naïve T cell priming. T cell effector function was measured by expression of IFNγ, MIP-1β, tumor necrosis factor and CD107a and by lysis of target tumor cells.ResultsCLEC9A-NY-ESO-1 antibodies (Abs) were effective at mediating delivery and cross-presentation of multiple NY-ESO-1 epitopes by CD141+ DCs for activation of NY-ESO-1-specific CD8+ T cells. When benchmarked to NY-ESO-1 conjugated to an untargeted control antibody or to anti-human DEC-205, CLEC9A-NY-ESO-1 was superior at ex vivo reactivation of NY-ESO-1-specific T cell responses in patients with melanoma. Moreover, CLEC9A-NY-ESO-1 induced priming of naïve NY-ESO-1-specific CD8+ T cells with polyclonal effector function and potent tumor killing capacity in vitro.ConclusionsThese data advocate human CLEC9A-NY-ESO-1 Ab as an attractive strategy for specific targeting of CD141+ DCs to enhance tumor immunogenicity in NY-ESO-1-expressing malignancies.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1354-1354
Author(s):  
Annkristin Heine ◽  
Tobias Holderried ◽  
Frank Grünebach ◽  
Silke Appel ◽  
Markus M. Weck ◽  
...  

Abstract Transfection of dendritic cells (DC) with in vitro transcribed RNA was shown to be a highly efficient method to generate antigen specific T cells, probably due to the induction of a polyclonal T cell response directed against multiple antigens presented on different HLA allels. However, the experimental evidence of this assumption remains to be demonstrated. To accomplish this, we used monocyte derived DC that were electroporated with RNA coding for the CMV pp65 antigen. The induction and expansion of antigen specific CD8+ and CD4+ T cells was assessed using a pannel of peptides derived from this antigen and presented on HLA-A2, -A1, -A11, -A24, -B35 and -B7 in IFN-g ELISPOT, 51Cr-release and proliferation assays. Autologous DC generated from CMV positive healthy donors were pulsed with peptides or transfected with pp65 RNA and utilized as stimulators. Autologous purified CD8+ and CD4+ lymphocytes were used as effector cells. By applying this approach we found that transfection of DC with pp65 RNA induces an expansion of polyclonal CD8+ mediated T cell responses that recognized peptide antigens presented on different HLA molecules. These in vitro generated cytotoxic T cells were able to efficiently lyse DC pulsed with pp65 derived peptides or transfected with the cognate RNA in an antigen specific manner after several in vitro restimulations. Furthermore, this experimental approach allowed the identification of the immunodominace of T cell epitopes presented upon RNA transfection. The HLA-2 directed responses were more pronounced as compared to the HLA-A1, -A11, -A24 or -B35 allels. In contrast, in 7 out of 7 HLA-A2 and HLA-B7 positive donors B7-peptides elicited a stronger T cell response than the A2-peptide, indicating the immunodominance of HLA-B7 epitopes. Interestingly, transfection of DC with pp65 RNA resulted in the induction of CD4+ antigen specific T cells that produced IFN-g and proliferated upon stimulation with transfected DC. In the next set of experiments we analyzed the possible induction of CMV specific T cells that recognize epitopes deduced from different antigens. Co-transfection of DC with a mixture of RNAs coding for the CMV pp65 and IE1 antigens elicited polyclonal T lymphocytes specific for peptides derived from both antigens. More importantly, polyclonal cytotoxic T cells could be elicited in peripheral blood of 2 out of 3 CMV negative donors demonstrating the efficiency of this approach. Our results demonstrate that DC transfected with RNA can elicit polyclonal T cell responses and have implications for the development of immunotherapeutic strategies to target viral or tumor associated antigens.


Blood ◽  
2005 ◽  
Vol 106 (1) ◽  
pp. 216-223 ◽  
Author(s):  
Elodie Segura ◽  
Carole Nicco ◽  
Bérangère Lombard ◽  
Philippe Véron ◽  
Graça Raposo ◽  
...  

Exosomes are secreted vesicles formed in late endocytic compartments. Immature dendritic cells (DCs) secrete exosomes, which transfer functional major histocompatibility complex (MHC)–peptide complexes to other DCs. Since immature and mature DCs induce different functional T-cell responses (ie, tolerance versus priming), we asked whether DC maturation also influenced the priming abilities of their exosomes. We show that exosomes secreted by lipopolysaccharide (LPS)–treated mature DCs are 50- to 100-fold more potent to induce antigen-specific T-cell activation in vitro than exosomes from immature DCs. In vitro, exosomes from mature DCs transfer to B lymphocytes the ability to prime naive T cells. In vivo, only mature exosomes trigger effector T-cell responses, leading to fast skin graft rejection. Proteomic and biochemical analyses revealed that mature exosomes are enriched in MHC class II, B7.2, intercellular adhesion molecule 1 (ICAM-1), and bear little milk-fat globule–epidermal growth factor–factor VIII (MFG-E8) as compared with immature exosomes. Functional analysis using DC-derived exosomes from knock-out mice showed that MHC class II and ICAM-1 are required for mature exosomes to prime naive T cells, whereas B7.2 and MFG-E8 are dispensable. Therefore, changes in protein composition and priming abilities of exosomes reflect the maturation signals received by DCs.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4821-4821
Author(s):  
Hakan Mellstedt ◽  
Parviz Kokhaei ◽  
Anders Osterborg ◽  
Aniruddha Choudhury

Abstract The slow indolent nature of B-CLL makes it eminently suitable for immune-based treatment strategies. Currently, there exist very few CLL-associated, defined antigen which can be used in vaccination approaches. The use of whole tumor preparations in conjunction with dendritic cells (DC) as cellular adjuvants presents an alternative method for generating therapeutic T-cell responses in CLL patients. We have examined various strategies of loading DC with whole tumor antigens; viz. tumor-DC hybrids, endocytosed apoptotic bodies, RNA or lysate from B-CLL cells. Dendritic cells were generated in vitro, using GM-CSF and IL-4 from immunomagnetically purified, CD14+ monocyte precursors obtained from the peripheral blood of B-CLL patients. The immature DC were loaded with whole tumor antigen using the above methods and matured using TNFα. The tumor antigen-loaded DC were compared for their ability to stimulate autologous T-cell responses. Among all the antigen-loading methods tested, DC that had endocytosed apoptotic bodies (Apo-DC) consistently generated the greatest numbers and magnitude of reactive T-cells as quantified in proliferation and ELISPOT assays. RT-PCR analysis for cytokine mRNA revealed that T-cells stimulated by Apo-DC resulted in an immune response that was almost entirely of the TH1 type as manifested by the production of mRNA for IFN-γ and TNFα. In contrast, other methods of loading antigen resulted in a mixed TH1-TH2 population with varying amounts of TH2 cytokines like IL-4 and IL-10. In a separate series of experiments we compared the T-cell stimulatory capacities of cryopreserved and thawed, with fresh antigen-loaded DC. The results of these studies are essential for the development of a protocol for clinical therapy of B-CLL patients using Apo-DC. Our results indicated that cryopreserved DC could be recovered with high viability. A comparison of functional abilities demonstrated that no significant differences in T-cell stimulatory capacity between cryopreserved and fresh Apo-DC could be noted over a wide variety of immunological assays. Cumulatively, our results suggest that Apo-DC may be a suitable vaccine candidate for immunotherapy of B-CLL patients.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Anna Rao ◽  
Otto Strauss ◽  
Efthymia Kokkinou ◽  
Mélanie Bruchard ◽  
Kumar P. Tripathi ◽  
...  

AbstractILCs and T helper cells have been shown to exert bi-directional regulation in mice. However, how crosstalk between ILCs and CD4+ T cells influences immune function in humans is unknown. Here we show that human intestinal ILCs co-localize with T cells in healthy and colorectal cancer tissue and display elevated HLA-DR expression in tumor and tumor-adjacent areas. Although mostly lacking co-stimulatory molecules ex vivo, intestinal and peripheral blood (PB) ILCs acquire antigen-presenting characteristics triggered by inflammasome-associated cytokines IL-1β and IL-18. IL-1β drives the expression of HLA-DR and co-stimulatory molecules on PB ILCs in an NF-κB-dependent manner, priming them as efficient inducers of cytomegalovirus-specific memory CD4+ T-cell responses. This effect is strongly inhibited by the anti-inflammatory cytokine TGF-β. Our results suggest that circulating and tissue-resident ILCs have the intrinsic capacity to respond to the immediate cytokine milieu and regulate local CD4+ T-cell responses, with potential implications for anti-tumor immunity and inflammation.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 781-781 ◽  
Author(s):  
Jacalyn Rosenblatt ◽  
Brett Glotzbecker ◽  
Heidi Mills ◽  
Whitney Keefe ◽  
Kerry Wellenstein ◽  
...  

Abstract Abstract 781 We have developed a promising cancer vaccine in which autologous tumor cells are fused with dendritic cells (DCs) resulting in the presentation of tumor antigens in the context of DC mediated costimulation. In animal models, vaccination with fusion cells results in eradication of established tumor, and in clinical trials, both immunologic and clinical responses have been observed. However, response to vaccination may be muted by inhibitory pathways that blunt activated T cell responses. The PD-1/PDL-1 pathway is an important element contributing to tumor mediated immune suppression. In infectious disease models, upregulation of T cell expression of PD-1 is associated with an exhausted phenotype facilitating the development of chronic viral infection. In contrast, PD-1 blockade results in the restoration of functionally active T cells and clearance of infection. The PD-1/PDL-1 pathway is also being evaluated a as a central mechanism by which tumors escape host immunity. CT-011, is a humanized anti PD-1 antibody that is currently evaluated in Phase II studies for the treatment of hematological malignancies and solid tumors. In this study, we evaluated expression of PD-1 on T cells derived from patients with advanced hematologic malignancies, and PDL-1 expression on primary myeloma cells, ex-vivo generated dendritic cells, and DC/tumor fusion cells. We evaluated the effect of PD-1 blockade with CT-011 on T cell response to DC/tumor fusion cell stimulation in vitro. Tumor cells were obtained from bone marrow aspirates of patients with multiple myeloma. Nonadherent peripheral blood mononuclear cells obtained from patients with multiple myeloma and normal volunteers were cultured in RPMI supplemented with 10U/ml IL-2, and expression of PD-1 on CD4+ T cells was assessed by flow cytometric analysis . DCs were generated from adherent mononuclear cells cultured with rhIL-4, GM-CSF and TNFα and fused with tumor cells by coculture in 50% solution of polyethylene glycol. T cells were stimulated by DC/tumor fusions in the presence or absence of 5ug/ml CT-011. We demonstrate that PD-1 expression is markedly upregulated on T cells in patients with advanced multiple myeloma. As compared to a control population of normal volunteers in which mean levels of PD-1 expression was 6% (n=7), mean expression in patients with multiple myeloma was 20% (n=9). These findings suggest that upregulation of PD-1 expression may play a central role in tumor mediated immune suppression. Mean expression of PDL-1 was 91% on dendritic cells generated from adherent peripheral blood mononuclear cells obtained from normal volunteers (n=6), and 66% on patient derived myeloma cells (n=3). In addition, PDL-1 expression is found in greater than 90% of DC/tumor fusions (n=2), which potentially provides an inhibitory signal dampening fusion mediated immunologic response. We examined the effect of PD-1 blockade on T cell response to DC/tumor fusions ex vivo with different anti PD-1 antibodies including CT-011. DC/tumor fusions were co-cultured with autologous T cells alone or with antibodies against PD-1 Enhanced fusion mediated stimulation of T cells was noted particularly with CT-011, resulting in a greater than 5 fold increase in T cell proliferation. Interferon gamma secretion by CD4+ T cells in response to stimulation by DC/myeloma fusion cells was increased from 4% to 11% in the presence of CT-011. In addition, IL-10 secretion by CD4+ cells following DC/myeloma fusion stimulation decreased from 6.5% to 3.5% following PD-1 blocakde . In summary, we have demonstrated that PD-1 expression is increased in T cells of patients with hematologic malignancy, and CT-011, a PD-1 blocking antibody, enhances activated T cell responses following stimulation with a DC/tumor fusion vaccine. A clinical trial in which patients with multiple myeloma are treated with DC/myeloma fusions in conjunction with CT-011 following autologous transplantation is planned. Disclosures: Schickler: CureTech, Ltd.: Employment. Rotem-Yehudar:CureTech, Ltd.: Employment.


2013 ◽  
Vol 87 (23) ◽  
pp. 12794-12804 ◽  
Author(s):  
Eddie A. James ◽  
Rebecca E. LaFond ◽  
Theresa J. Gates ◽  
Duy T. Mai ◽  
Uma Malhotra ◽  
...  

Yellow fever virus (YFV) can induce acute, life-threatening disease that is a significant health burden in areas where yellow fever is endemic, but it is preventable through vaccination. The live attenuated 17D YFV strain induces responses characterized by neutralizing antibodies and strong T cell responses. This vaccine provides an excellent model for studying human immunity. While several studies have characterized YFV-specific antibody and CD8+T cell responses, less is known about YFV-specific CD4+T cells. Here we characterize the epitope specificity, functional attributes, and dynamics of YFV-specific T cell responses in vaccinated subjects by investigating peripheral blood mononuclear cells by using HLA-DR tetramers. A total of 112 epitopes restricted by seven common HLA-DRB1 alleles were identified. Epitopes were present within all YFV proteins, but the capsid, envelope, NS2a, and NS3 proteins had the highest epitope density. Antibody blocking demonstrated that the majority of YFV-specific T cells were HLA-DR restricted. Therefore, CD4+T cell responses could be effectively characterized with HLA-DR tetramers.Ex vivotetramer analysis revealed that YFV-specific T cells persisted at frequencies ranging from 0 to 100 cells per million that are detectable years after vaccination. Longitudinal analysis indicated that YFV-specific CD4+T cells reached peak frequencies, often exceeding 250 cells per million, approximately 2 weeks after vaccination. As frequencies subsequently declined, YFV-specific cells regained CCR7 expression, indicating a shift from effector to central memory. Cells were typically CXCR3 positive, suggesting Th1 polarization, and produced gamma interferon and other cytokines after reactivationin vitro. Therefore, YFV elicits robust early effector CD4+T cell responses that contract, forming a detectable memory population.


Blood ◽  
2005 ◽  
Vol 106 (9) ◽  
pp. 3105-3113 ◽  
Author(s):  
Frank Neumann ◽  
Claudia Wagner ◽  
Klaus-Dieter Preuss ◽  
Boris Kubuschok ◽  
Claudia Schormann ◽  
...  

AbstractBecause of their frequent expression in a wide spectrum of malignant tumors but not in normal tissue except testis, cancer testis antigens are promising targets. However, except for HOM-TES-14/SCP1, their expression in malignant lymphomas is rare. SCP1 (synaptonemal complex protein 1) has been shown to elicit antibody responses in the autologous host, but no T-cell responses against HOM-TES-14/SCP1 have been reported. Using the SYFPEITHI algorithm, we selected peptides with a high binding affinity to major histocompatibility complex class 2 (MHC 2) molecules. The pentadecamer epitope p635-649 induced specific CD4+ T-cell responses that were shown to be restricted by HLA-DRB1*1401. The responses could be blocked by preincubation of T cells with anti-CD4 and antigen-presenting cells with anti–HLA-DR, respectively, proving the HLA-DR–restricted presentation of p635-649 and a CD4+ T-cell–mediated effector response. Responding CD4+ cells did not secrete interleukin-5 (IL-5), indicating that they belong to the TH1 subtype. The natural processing and presentation of p635-649 were demonstrated by pulsing autologous and allogeneic dendritic cells with a protein fragment covering p635-649. Thus, p635-649 is the first HOM-TES-14/SCP1–derived epitope to fulfill all prerequisites for use as a peptide vaccine in patients with HOM-TES-14/SCP1–expressing tumors, which is the case in two thirds of peripheral T-cell lymphomas.


2008 ◽  
Vol 82 (7) ◽  
pp. 3561-3573 ◽  
Author(s):  
Ellen R. Van Gulck ◽  
Guido Vanham ◽  
Leo Heyndrickx ◽  
Sandra Coppens ◽  
Katleen Vereecken ◽  
...  

ABSTRACT Developing an immunotherapy to keep human immunodeficiency virus type 1 (HIV-1) replication suppressed while discontinuing highly active antiretroviral therapy (HAART) is an important challenge. In the present work, we evaluated in vitro whether dendritic cells (DC) electroporated with gag mRNA can induce HIV-specific responses in T cells from chronically infected subjects. Monocyte-derived DC, from therapy-naïve and HAART-treated HIV-1-seropositive subjects, that were electroporated with consensus codon-optimized HxB2 gag mRNA efficiently expanded T cells, secreting gamma interferon (IFN-γ) and interleukin 2 (IL-2), as well as other cytokines and perforin, upon restimulation with a pool of overlapping Gag peptides. The functional expansion levels after 1 week of stimulation were comparable in T cells from HAART-treated and treatment-naïve patients and involved both CD4+ and CD8+ T cells, with evidence of bifunctionality in T cells. Epitope mapping of p24 showed that stimulated T cells had a broadened response toward previously nondescribed epitopes. DC, from HAART-treated subjects, that were electroporated with autologous proviral gag mRNA equally efficiently expanded HIV-specific T cells. Regulatory T cells did not prevent the induction of effector T cells in this system, whereas the blocking of PD-L1 slightly increased the induction of T-cell responses. This paper shows that DC, loaded with consensus or autologous gag mRNA, expand HIV-specific T-cell responses in vitro.


1989 ◽  
Vol 169 (4) ◽  
pp. 1255-1264 ◽  
Author(s):  
S E Macatonia ◽  
P M Taylor ◽  
S C Knight ◽  
B A Askonas

We used well-gassed hanging drop (20 microliters) cultures with high concentrations of purified T cells from normal BALB/c mice to examine whether dendritic cells (DC) can induce primary antiviral proliferative T cell responses and generate virus-specific CTL. We found that DC exposed to infectious influenza virus in vitro or in vivo in small numbers (0.1-1%) resulted in strong proliferation of responder T cells within 3 d, and this was strongly inhibited by antibodies to class II MHC molecules. In addition, in 5-d cultures, the influenza-treated DC generated CTL specifically able to lyse influenza-infected syngeneic target cells bearing MHC class I antigens. The most potent nucleoprotein (NP) epitope recognized by BALB/c CTL is peptide 147-158 (Arg156-) and influenza-infected DC in vitro stimulated CTL recognizing this peptide, thus mimicking the response in mice primed by intranasal influenza infection. We also induced T cell proliferation and virus-specific CTL in cultures of normal T cells by stimulating with DC pulsed with the natural NP sequence 147-158 or the potent peptide 147-158 (Arg156-). Small numbers of peritoneal exudate cells, after activation with Con A to produce class II MHC expression and after removal of DC with a specific mAb (33DI), did not lead to primary CTL generation but initiated secondary stimulation in vitro. Our results using the hanging drop culture method and DC as APC have implications for studying the T cell repertoire for viral components in humans without the necessity of previous immunization.


Sign in / Sign up

Export Citation Format

Share Document