scholarly journals CD8 T cells and dendritic cells: key players in the attenuated maternal immune response to influenza infection

2015 ◽  
Vol 107 ◽  
pp. 1-9 ◽  
Author(s):  
Rebecca L. Vanders ◽  
Vanessa E. Murphy ◽  
Peter G. Gibson ◽  
Philip M. Hansbro ◽  
Peter A.B. Wark
2021 ◽  
Author(s):  
◽  
John David Gibbins

<p>The immune system has the potential to selectively target and eliminate tumours cells. However, the induction of an immunosuppressive environment by factors released by tumours cells, or by the tumour stroma, in combination with difficulties in differentiating between healthy and malignant cells, contributes to inefficient or disabled anti-tumour immune responses. A variety of different immunotherapeutic approaches are being developed to tip the balance in favour of anti-tumour immunity. Many of these approaches are designed to stimulate improved activity of T cells with specificity for tumour-associated antigens.  This thesis explores how T cell-mediated responses are initiated and maintained in immunotherapy, with an emphasis on the role of antigen presentation by resident dendritic cells (DCs). An animal model was used in which a DC subset in the spleen that expresses the cell marker langerin could be selectively ablated during the course of therapy. As these DCs have been shown to be uniquely capable of acquiring circulating antigens and cellular debris, and have a heightened capacity for cross-priming CD8⁺ T cells, it was hypothesised that the function of these cells could play a significant role in determining the outcome of immunotherapies.  A model of adoptive T cell therapy was examined in mice challenged with an intravenously administered lymphoma that formed tumour foci in a variety of locations in the body. Treating established tumours by adoptively transferring in vitro activated effector CD8⁺ T cells significantly increased their symptom-free survival. The protection received by this therapy was dependent on a stimulus being provided by endogenous langerin⁺ CD8α⁺ DCs to the transferred T cells. In the absence of langerin⁺ CD8α⁺ DCs, the proportion and number of transferred anti-tumour CD8⁺ T cells was lower in the blood and spleen. However, no obvious differences in phenotype and function could be defined. Langerin⁺ CD8α⁺ DCs therefore contribute to the maintenance of an effective CD8⁺ T cell-based immunotherapy and the role of endogenous DCs should be taken into consideration during the design of immunotherapies.  To investigate the role of langerin⁺ CD8α⁺ DCs in initiating effector T cell responses, a novel whole-cell vaccine was developed for the treatment of acute myeloid leukaemia (AML). This vaccine exploited the stimulatory functions of invariant natural killer T cells, and was therefore administered intravenously to access the large invariant natural killer T cell compartment of the spleen. The vaccine completely protected mice from developing leukaemia when challenged with AML cells after vaccination, with CD4⁺ and CD8⁺ T cells mediating protection. The immune response generated by the vaccine was shown to be completely dependent on langerin⁺ CD8α⁺ DCs. In hosts with established tumours; however, the vaccine was ineffective. This may have been partially due to a reduced function of langerin⁺ CD8α⁺ DCs as their activation phenotype was significantly reduced in the presence of established AML; however, non-specific T cells could still be stimulated via these DCs. Reduced vaccine efficacy was associated with increased number and/or function of suppressor cells, including regulatory T cells and myeloid derived suppressor cells within the host. In addition, in leukemic hosts, the proportion of T cells in the spleen was reduced, and the function of AML-specific CD4⁺ T cells, but not CD8⁺ T cells, was impaired. Driving AML-bearing hosts into remission with chemotherapy prior to vaccination enabled the vaccine to protect the host from subsequent AML challenge. Langerin⁺ CD8α⁺ DCs are therefore responsible for initiating the vaccine-induced immune response in this model and their suppression may have contributed to the inefficacy of the vaccine in the presence of established tumours.</p>


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3228-3228
Author(s):  
Yukio Kondo ◽  
Kinya Ohata ◽  
Shinji Nakao

Abstract Aberrantly expressed self-antigens in leukemic cells are a kind of leukemia-associated-antigens (LAAs). Although such self-antigen-derived LAAs potentially induce high avidity CTLs in patients with leukemia, these CTLs usually do not persist in the patients because they undergo apoptosis upon encountering with leukemic cells. In allogeneic stem cell transplant (allo-SCT) recipients with leukemia, LAAs-specific high avidity CTL may be elicited from donor-derived naive T cells that are sensitized by residual leukemic cells. Cyclin-dependent kinase 2 (CDK2) is a cell cycle regulator protein that is aberrantly expressed by leukemia cells. We previously reported that two CDK2-derived peptides (CDK2 158-166, CDK2 178-186) avidly bound to HLA-A24 molecule to elicit each peptide-specific CTLs from HLA identical donor of a AML patient, and CDK2-specific CD8 + T cells preferentially killed the recipient AML cells which aberrantly expressed CDK2 protein (Blood.108 (11): a3173. 2006). When we assessed cryopreserved PBMCs obtained before and after allo-SCT from 16 HLA-A24+ patients (6 AML, 1 MDS, 1 CML, 2 ALL, 4 NHL and 2 RCC) using CDK2 158/A24 and CDK2 178/A24 pentamers, small populations (0.1–1.0%) of CDK2 158 and CDK2 178-specific CTL were detectable in 6 patients after SCT but not before SCT. All of the 6 patients had MRD at the time of SCT and achieved molecular CR after SCT. None of the 3 patients relapsed after SCT did not show CDK2 immunity. There is no relationship between the appearance of CDK2-specific CTLs and the development of GvHD (Figure). Leukemic myeloid dendritic cells (mDC) are known to present in vivo in patients with leukemia, and be able to trigger a protective antileukemic immune response by allogeneic T-cells. We hypothesized that residual circulating leukemic mDCs may sensitize donor-derived T cells by cross-presenting CDK2-peptides early after allo-SCT and elicit high avidity CTLs specific to leukemic cells. mDCs subset was identified with by three-color staining using mAbs against CD85k, CD33, CD14 and CD16. Circulating mDCs represented 5.4% of PBMCs in a patient with CML-CP at diagnosis. Leukemic mDCs from the patient were purified with CD1c mAb-conjugated maginetic beads and were assessed for their ability to stimulate allogeneic T cells to acquire specific cytotoxicity against CDK2-peptides. The purity of the mDCs as determined by flow cytometry was 92% of living isolated cells. Naive CD8 + T cells isolated from healthy individual were cultured with the leukemic mDCs for 12 days and subjected to pentamers staining. After the coculturing, the proportion of CDK2 158/A24 and CDK2 178/A24 pentamers + CD8 T cells increased 0.6% to 2.2% and 0.6% to 2.6%, respectively. These data suggest that CDK2-specific CTLs can be induced from donor-derived T cells due to in vivo sensitization of donor T cells by residual leukemic mDCs and this may be a mechanism responsible for the generation of CDK2-specific CTLs in allo-SCT recipients with MRD without vaccination of CDK2-peptides. Vaccination with CDK2-peptides after allo-SCT may be useful in both enhancing CDK2-specific immunity in patients with MRD and in generating CDK2 immunity in those without MRD. Figure: An immune response to CDK2 is associated with GvL and MRD at the time of allo-SCT, but not with GvHD. Figure:. An immune response to CDK2 is associated with GvL and MRD at the time of allo-SCT, but not with GvHD.


2021 ◽  
Author(s):  
◽  
John David Gibbins

<p>The immune system has the potential to selectively target and eliminate tumours cells. However, the induction of an immunosuppressive environment by factors released by tumours cells, or by the tumour stroma, in combination with difficulties in differentiating between healthy and malignant cells, contributes to inefficient or disabled anti-tumour immune responses. A variety of different immunotherapeutic approaches are being developed to tip the balance in favour of anti-tumour immunity. Many of these approaches are designed to stimulate improved activity of T cells with specificity for tumour-associated antigens.  This thesis explores how T cell-mediated responses are initiated and maintained in immunotherapy, with an emphasis on the role of antigen presentation by resident dendritic cells (DCs). An animal model was used in which a DC subset in the spleen that expresses the cell marker langerin could be selectively ablated during the course of therapy. As these DCs have been shown to be uniquely capable of acquiring circulating antigens and cellular debris, and have a heightened capacity for cross-priming CD8⁺ T cells, it was hypothesised that the function of these cells could play a significant role in determining the outcome of immunotherapies.  A model of adoptive T cell therapy was examined in mice challenged with an intravenously administered lymphoma that formed tumour foci in a variety of locations in the body. Treating established tumours by adoptively transferring in vitro activated effector CD8⁺ T cells significantly increased their symptom-free survival. The protection received by this therapy was dependent on a stimulus being provided by endogenous langerin⁺ CD8α⁺ DCs to the transferred T cells. In the absence of langerin⁺ CD8α⁺ DCs, the proportion and number of transferred anti-tumour CD8⁺ T cells was lower in the blood and spleen. However, no obvious differences in phenotype and function could be defined. Langerin⁺ CD8α⁺ DCs therefore contribute to the maintenance of an effective CD8⁺ T cell-based immunotherapy and the role of endogenous DCs should be taken into consideration during the design of immunotherapies.  To investigate the role of langerin⁺ CD8α⁺ DCs in initiating effector T cell responses, a novel whole-cell vaccine was developed for the treatment of acute myeloid leukaemia (AML). This vaccine exploited the stimulatory functions of invariant natural killer T cells, and was therefore administered intravenously to access the large invariant natural killer T cell compartment of the spleen. The vaccine completely protected mice from developing leukaemia when challenged with AML cells after vaccination, with CD4⁺ and CD8⁺ T cells mediating protection. The immune response generated by the vaccine was shown to be completely dependent on langerin⁺ CD8α⁺ DCs. In hosts with established tumours; however, the vaccine was ineffective. This may have been partially due to a reduced function of langerin⁺ CD8α⁺ DCs as their activation phenotype was significantly reduced in the presence of established AML; however, non-specific T cells could still be stimulated via these DCs. Reduced vaccine efficacy was associated with increased number and/or function of suppressor cells, including regulatory T cells and myeloid derived suppressor cells within the host. In addition, in leukemic hosts, the proportion of T cells in the spleen was reduced, and the function of AML-specific CD4⁺ T cells, but not CD8⁺ T cells, was impaired. Driving AML-bearing hosts into remission with chemotherapy prior to vaccination enabled the vaccine to protect the host from subsequent AML challenge. Langerin⁺ CD8α⁺ DCs are therefore responsible for initiating the vaccine-induced immune response in this model and their suppression may have contributed to the inefficacy of the vaccine in the presence of established tumours.</p>


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1909-1909
Author(s):  
Adham S Bear ◽  
Meghan M Turnis ◽  
Xiao-Tong Song ◽  
Russell Cruz ◽  
An Lu ◽  
...  

Abstract Abstract 1909 Introduction: Cancer vaccines have shown promise in small animal models of cancer, but have thus far been disappointing in clinical settings. Successful induction of a systemic and long-term anti-tumor immune response following vaccination is dependent on delivery of tumor-associated antigens to lymphoid tissues, in combination with the activation of professional antigen presenting cells (APCs). Here we describe a novel live T cell vaccine (TCV) that delivers antigenic peptides to secondary lymph nodes while simultaneously activating endogenous dendritic cells (DCs) through transgenic expression of CD40L or bacterial flagellin (fliC). Methods: To generate TCVs, murine splenocytes were isolated from wild-type type C57BL/6 mice. Following activation with anti-CD3/anti-CD28 microbeads, splenocytes were transduced with pRV2011-luciferase-IRES-Thy1.1, pRV2011-CD40L-IRES-Thy1.1 or pRV2011-fliC-IRES-Thy1.1 retrovirus. Analysis of TCV migration to lymphoid organs was performed by bioluminescence imaging for firefly luciferase. Following transduction with CD40L and fliC molecules, TCVs were measured for transduction efficiency (Thy1.1) and transgene expression using FACS analysis of CD40L or by Western blot, respectively. TCVs were subsequently pulsed with MHC class I-restricted epitopes for ovalbumin257-264 (SIINFEKL) or Trp2180-188 (SVYDFFVWL) peptides and injected intravenously at a dose of 1×107 TCVs per vaccination. To test the protective effects of TCVs, C57BL/6 mice were immunized at days 0 and 14 and then challenged with either 5×105 B16-OVA (for TCV-SIIN) or parental B16.F10 (for TCV-SVYD) melanoma tumor cells. To examine the ability of TCVs to eliminate established tumors, mice received B16-OVA or B16.F10 tumor cells followed by vaccination with TCVs on days 3, 9 and 15. Immunological studies were performed on a subset of mice (n=5 per group) to analyze induction of tumor-specific T cells using tetramer and IFN-g ELIspot assays. In vivo activation of lymph node DCs was performed by FACS analysis for CD11c+ DC co-expressing CD86 and I-A/I-E mouse MHC class II antibodies. Results: Following activation, TCVs were efficiently transduced with retrovirus (>85% CD40L) or expressed high levels of fliC. Bioluminescent imaging showed that luciferase-expressing TCVs rapidly migrated to lymphoid organs including the spleen and cervical and inguinal lymph nodes demonstrating the capacity of TCVs to co-localize with professional APCs. Importantly, irradiation (30 Gy) of TCVs completely abrogated migration and persistence highlighting the requirement for live TCVs. Next we examined whether TCV-CD40L or TCV-fliC could induce a protective immune response against B16 tumors. Administration of TCV-fliC-SIIN (OVA) and TCV-CD40L-SIIN primed peptide-specific CD8+ T cells, and led to decreased tumor growth and increased survival in mice subsequently challenged with B16-Ova (p<0.05). This response corresponded with a statistically significant (p<.05) increase in SIIN-specific CD8+ T cells as measured by tetramer FACS analysis and IFN-g ELIspot assays. Vaccination of mice with established tumors showed similar tumor suppression with both TCV designs (p<05). As OVA is a xenogenic antigen, we next examined whether TCVs pulsed with Trp2 peptide (SVYD) could induce similar protective effects. While vaccination with SVYD-pulsed T cells alone (no gene modification) did not inhibit tumor growth, expression of CD40L or fliC by TCV pulsed with Trp2 peptide suppressed B16.F10 tumor proliferation and increased survival in mice with pre-established tumors (p<.05). As found in the B16.OVA experiments, immunological protection correlated with a dramatic increase in SVYD-specific CD8+ T cells in the spleen, tumor draining lymph nodes and tumor. Conclusions: The efficient delivery of tumor-associated antigens to lymphoid tissues by TCVs overcomes a major limitation of alternative vaccine strategies. Vaccination with peptide-pulsed TCVs primes antigen-specific T cell responses with anti-tumor capability, and endogenous DC maturation leads to the inhibition of established B16-Ova and B16-F10 tumors. This illustrates the role of endogenous DC as mediators of the vaccine response and demonstrates the effectiveness of using TCVs to deliver antigen in the context of DC activating molecules. Disclosures: No relevant conflicts of interest to declare.


2012 ◽  
Vol 2012 ◽  
pp. 1-7 ◽  
Author(s):  
Simona Ronchetti ◽  
Giuseppe Nocentini ◽  
Maria Grazia Petrillo ◽  
Carlo Riccardi

As many members of the tumor necrosis factor receptor superfamily, glucocorticoid-induced TNFR-related gene (GITR) plays multiple roles mostly in the cells of immune system. CD8+T cells are key players in the immunity against viruses and tumors, and GITR has been demonstrated to be an essential molecule for these cells to mount an immune response. The aim of this paper is to focus on GITR function in CD8+cells, paying particular attention to numerous and recent studies that suggest its crucial role in mouse disease models.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A354-A354
Author(s):  
Andrea Ziblat ◽  
Brendan Horton ◽  
Emily Higgs ◽  
Ken Hatogai ◽  
Thomas Gajewski

BackgroundPD-1/PD-L1 blockade has shown clinical benefit across many cancer types. However, a large fraction of patients are resistant to immune checkpoint blockade therapy and others eventually relapse. Understanding the mechanisms involved in αPD1/PD-L1 immunotherapy efficacy may enable new strategies for improving clinical outcomes. Given that Batf3-lineage dendritic cells (DCs) are needed for spontaneous T cell priming in the tumor-draining lymph node and for recruitment of effector CD8+ T cells to the tumor, in the current work we examined whether Batf3+ DCs are also required during the effector phase of the anti-tumor immune response at the time of anti-PD-L1 administration for therapeutic efficacy.MethodsWe utilized the B16-SIY melanoma model, CD11c-DTR-GFP, and CD11c-DTR-GFP/Batf3 KO bone marrow chimeras to study the role Batf3+ DCs play during anti-PD-L1 immunotherapy. To focus on the effector phase of the immune response, we depleted CD11c+ cells with diphtheria toxin from day seven of tumor injection while simultaneously blocking new T cell entry with FTY720. As flow cytometry revealed high 4-1BBL expression on intratumoral Batf3-DCs, 4-1BB KO mice and anti-4-1BBL blocking antibodies were used. Tumor growth and phenotypic analysis of the tumor infiltrate were evaluated.ResultsStrikingly, we observed that CD11c+ cells, and specifically Batf3+ DCs, were required in the tumor prior to αPD-L1 treatment for immunotherapy efficacy. The normal intratumoral expansion of antigen (Ag)-specific CD8+ tumor-infiltrating T cells (TILs) and increased ratio between Ag-specific CD8+ TILs and regulatory T cells following anti-PD-L1 therapy was eliminated with Batf3+ DC depletion. Batf3+ DCs expressed high levels of 4-1BBL, and increased expression of 4-1BB on antigen-specific CD8+ TILs upon αPD-L1 treatment required Batf3+ DCs. Mechanistic experiments confirmed a requirement for 4-1BB expression on immune cells for αPD-L1 efficacy, and blocking antibodies against 4-1BBL eliminated anti-PD-L1 efficacy as well. Using appropriate bone marrow chimeras, agonistic 4-1BB antibodies were sufficient to bypass the need for CD11c+ DCs at the effector phase for tumor control. In human melanoma samples, co-localization of Batf3+ DCs and CD8+ T cells was observed in T cell-inflamed tumors, which correlated with anti-PD-1 efficacy in metastatic melanoma.ConclusionsOur results indicate that Batf3+ DCs are necessary during the effector phase of the anti-tumor immune response for anti-PD-L1 efficacy to occur, at least in part through 4-1BB/4-1BBL-mediated reinvigoration of Ag-specific CD8+ TILs.Ethics ApprovalThe study obtained ethics approval, IRB protocol 15-0837.


mSphere ◽  
2020 ◽  
Vol 5 (4) ◽  
Author(s):  
Roxane Tussiwand ◽  
Michael S. Behnke ◽  
Nicole M. Kretzer ◽  
Gary E. Grajales-Reyes ◽  
Theresa L. Murphy ◽  
...  

ABSTRACT Immunity to Toxoplasma gondii at early stages of infection in C57BL/6 mice depends on gamma interferon (IFN-γ) production by NK cells, while at later stages it is primarily mediated by CD8 T cells. We decided to explore the requirement for CD4 T cells during T. gondii infection in Batf3−/− mice, which lack CD8α+ dendritic cells (DCs) that are necessary for cross-presentation of cell-associated antigens to CD8 T cells. We show that in this immunodeficient background on a BALB/c background, CD4 T cells become important effector cells and are able to protect Batf3−/− mice from infection with the avirulent strain RHΔku80Δrop5. Independently of the initial NK cell activation, CD4 T cells in wild-type and Batf3−/− mice were the major source of IFN-γ. Importantly, memory CD4 T cells were sufficient to provide protective immunity following transfer into Batf3−/− mice and secondary challenge with the virulent RHΔku80 strain. Collectively, these results show that under situations where CD8 cell responses are impaired, CD4 T cells provide an important alternative immune response to T. gondii. IMPORTANCE Toxoplasma gondii is a widespread parasite of animals that causes zoonotic infections in humans. Although healthy individuals generally control the infection with only moderate symptoms, it causes serious illness in newborns and those with compromised immune systems such as HIV-infected AIDS patients. Because rodents are natural hosts for T. gondii, laboratory mice provide an excellent model for studying immune responses. Here, we used a combination of an attenuated mutant strain of the parasite that effectively vaccinates mice, with a defect in a transcriptional factor that impairs a critical subset of dendritic cells, to studying the immune response to infection. The findings reveal that in BALB/c mice, CD4 memory T cells play a dominant role in producing IFN-γ needed to control chronic infection. Hence, BALB/c mice may provide a more appropriate model for declining immunity seen in HIV-AIDS patients where loss of CD4 cells is associated with emergence of opportunistic infections.


Sign in / Sign up

Export Citation Format

Share Document