scholarly journals Expression of vFLIP in a Lentiviral Vaccine Vector Activates NF-κB, Matures Dendritic Cells, and Increases CD8+ T-Cell Responses

2008 ◽  
Vol 83 (4) ◽  
pp. 1555-1562 ◽  
Author(s):  
Helen M. Rowe ◽  
Luciene Lopes ◽  
Najmeeyah Brown ◽  
Sofia Efklidou ◽  
Timothy Smallie ◽  
...  

ABSTRACT Lentiviral vectors deliver antigens to dendritic cells (DCs) in vivo, but they do not trigger DC maturation. We therefore expressed a viral protein that constitutively activates NF-κB, vFLIP from Kaposi's sarcoma-associated herpesvirus (KSHV), in a lentivector to mature DCs. vFLIP activated NF-κB in mouse bone marrow-derived DCs in vitro and matured these DCs to a similar extent as lipopolysaccharide; costimulatory markers CD80, CD86, CD40, and ICAM-1 were upregulated and tumor necrosis factor alpha and interleukin-12 secreted. The vFLIP-expressing lentivector also matured DCs in vivo. When we coexpressed vFLIP in a lentivector with ovalbumin (Ova), we found an increased immune response to Ova; up to 10 times more Ova-specific CD8+ T cells secreting gamma interferon were detected in the spleens of vFLIP_Ova-immunized mice than in the spleens of mice immunized with GFP_Ova. Furthermore, this increased CD8+ T-cell response correlated with improved tumor-free survival in a tumor therapy model. A single immunization with vFLIP_Ova also reduced the parasite load when mice were challenged with OVA-Leishmania donovani. In conclusion, vFLIP from KSHV is a DC activator, maturing DCs in vitro and in vivo. This demonstrates that NF-κB activation is sufficient to induce many aspects of DC maturation and that expression of a constitutive NF-κB activator can improve the efficacy of a vaccine vector.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4489-4489
Author(s):  
Cirino Botta ◽  
Marco Rossi ◽  
Maria Rita Pitari ◽  
Annamaria Gullà ◽  
Teresa Del Giudice ◽  
...  

Dendritic cells (DCs) are potent antigen presenting cells that regulate the development of both innate and adaptive immune responses. According to their maturation status, DCs may ignite immune response or induce immune tolerance. Indeed, immature DCs (iDCs) present low levels of costimulatory molecules such as CD80 and CD86, and high levels of tolerogenic molecules such as B7H3. Upon exposure to maturation stimuli, DCs upregulate CD83 on their surface and gain the competence of stimulating T cell response. An efficient maturation is crucial for the generation of a specific cytotoxic T lymphocyte response, specially against cancer. However, recent reports have shown that Multiple Myeloma (MM) milieu can recruit DCs and reprogram them to sustain growth and survival of MM cells and protect them against immune response. Therapeutic approaches to restore DC functions rely on the identification of the pathways that are directly involved in induction of tolerance. Emerging evidence supports the role of microRNAs (miRNA) in the regulation of immune response and DC function. Among others, the miR-29 family seems to be involved in the modulation of NK activity, of Th1/Th2 phenotype switch and of DC differentiation from monocyte precursors. Besides, miR-29b targets and inhibits different and crucial immune-modulatory molecules such as B7H3, VEGF and IL-4. These findings suggest that miR-29 may play an important role in the multifaceted interplay between tumor cells and host’s immune system. To address this hypothesis, we generated iDCs from CD14+ monocytes of healthy donors and co-cultured them with: i) allogeneic (allo-) lymphocytes; ii) VEGF and IL-6 producing MM cells (RPMI8226 and U266); iii) allo-lymphocyte and MM cells. We found a consistent increase of miR-29b expression by RT-PCR during differentiation and maturation of DCs induced by allo-lymphocytes. However, when immature DCs were co-cultured with MM cells +/- allo-lymphocytes, a significant 3-fold reduction of miR-29b levels (p= 0.02) was observed (fig 1). This event occurred together with the absence of maturation markers, the persistence of high levels of B7H3 on the cell surface and with a raise in VEGF, IL-10, and IFN-gamma levels in the supernatant, confirming the MM-dependent impairment of the physiological DC maturation process. This latter concept was supported by the finding of increased number of CD4+CD25+Foxp3+T-regs in the DC/MM cell/allo-lymphocyte co-cultures as compared to the DC/allo-lymphocyte co-cultures (p= 0.05). To promote the recovery from the MM related immune-bias, we transiently transfected iDCs with miR-29b (29b-DCs) mimics or with a negative control (NC-DCs). We observed improved DC maturation (82.46% versus 39.89% of CD11c+/CD83+/CD86+ cells), reduced expression of B7H3 (33% reduction in MFI) and reduction of the T-reg number in 29b-DC/MM cell/allo-lymphocyte co-cultures as compared to NC-DC/MM cell/allo-lymphocyte co-culture. To investigate whether 29b-DCs were able to promote a specific CTL response against MM cells in vivo, we engrafted NOD/SCID γ chain-null mice with peripheral blood mononuclear cells (PBMCs) from HLA-A2+ healthy donors. DCs from the same donor were differentiated, transfected with either miR-29b or NC and then co-cultured with U266 for 48h. Mice were then vaccinated twice with either 29b-DCs or NC-DCs. Two weeks following the first injection, CD3+ human lymphocytes were recovered from mouse spleens (CD3 hu-splenocytes). We found an increased CD8/CD4 ratio in the CD3 hu-splenocytes collected from the 29b-DCs treated mouse as compared to control. To assess the capability of CD3+ hu-splenocytes to selectively kill U266 cells, we kept CD3 lymphocytes in culture in the presence of IL-15 for 48h. Then, we carried a cytotoxicity assay against U266 cell target. The highest specific lysis was attained with miR-29b DC primed CD3 hu-splenocytes (fig.2, p=0.03). Taken together, our data indicate that: a) miR-29b regulates DC differentiation/maturation and function; b) MM cells reduce the expression of miR-29b in DCs, thus contributing to the establishment of an immune-permissive microenvironment; c) replacement of miR-29b within DCs partially restores their differentiation and functions in vitro and their capability to induce antitumor specific T-cell response in vivo. On these findings, miR-29b mimics are attractive candidates to enhance immunotherapy approaches against MM. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2000 ◽  
Vol 96 (5) ◽  
pp. 1865-1872 ◽  
Author(s):  
Miriam Merad ◽  
Lawrence Fong ◽  
Jakob Bogenberger ◽  
Edgar G. Engleman

Bone marrow-derived dendritic cells (DC) represent a family of antigen-presenting cells (APC) with varying phenotypes. For example, in mice, CD8α+ and CD8α− DC are thought to represent cells of lymphoid and myeloid origin, respectively. Langerhans cells (LC) of the epidermis are typical myeloid DC; they do not express CD8α, but they do express high levels of myeloid antigens such as CD11b and FcγR. By contrast, thymic DC, which derive from a lymphoid-related progenitor, express CD8α but only low levels of myeloid antigens. CD8α+ DC are also found in the spleen and lymph nodes (LN), but the origin of these cells has not been determined. By activating and labeling CD8α− epidermal LC in vivo, it was found that these cells expressed CD8α on migration to the draining LN. Similarly, CD8α− LC generated in vitro from a CD8 wild-type mouse and injected into the skin of a CD8αKO mouse expressed CD8α when they reached the draining LN. The results also show that CD8α+ LC are potent APC. After migration from skin, they localized in the T-cell areas of LN, secreted high levels of interleukin-12, interferon-γ, and chemokine-attracting T cells, and they induced antigen-specific T-cell activation. These results demonstrate that myeloid DC in the periphery can express CD8α when they migrate to the draining LN. CD8α expression on these DC appears to reflect a state of activation, mobilization, or both, rather than lineage.


Blood ◽  
2005 ◽  
Vol 105 (8) ◽  
pp. 3206-3213 ◽  
Author(s):  
Jens Dannull ◽  
Smita Nair ◽  
Zhen Su ◽  
David Boczkowski ◽  
Christian DeBeck ◽  
...  

Abstract The objective of this study was to investigate whether the immunostimulatory properties of human monocyte-derived dendritic cells (DCs) could be enhanced by triggering OX40/OX40L signaling. Since monocyte-derived DCs possess only low-cell surface levels of OX40L in the absence of CD40 signaling, OX40L was expressed by transfection of DCs with the corresponding mRNA. We show that OX40L mRNA transfection effectively enhanced the immunostimulatory function of DCs at multiple levels: OX40L mRNA transfection augmented allogeneic and HLA class II epitope-specific CD4+ T-cell responses, improved the stimulation of antigen-specific cytotoxic T lymphocytes (CTLs) in vitro without interfering with the prostaglandin E2 (PGE2)–mediated migratory function of the DCs, and facilitated interleukin 12 p70 (IL-12p70)–independent T helper type 1 (Th1) polarization of naive CD4+ T-helper cells. Furthermore, vaccination of tumor-bearing mice using OX40L mRNA–cotransfected DCs resulted in significant enhancement of therapeutic antitumor immunity due to in vivo priming of Th1-type T-cell responses. Our data suggest that transfection of DCs with OX40L mRNA may represent a promising strategy that could be applied in clinical immunotherapy protocols, while circumventing the current unavailability of reagents facilitating OX40 ligation.


Blood ◽  
2002 ◽  
Vol 100 (2) ◽  
pp. 569-577 ◽  
Author(s):  
Yong Wang ◽  
Yanyun Zhang ◽  
Hiroyuki Yoneyama ◽  
Nobuyuki Onai ◽  
Taku Sato ◽  
...  

Abstract CD8α+ dendritic cells (DCs) represent a functionally distinct DC subset in vivo, which plays a critical role in initiating various cellular immune responses. However, the committed precursor of CD8α+ DCs remains to be identified. We reported here that murine splenic CD8α+CD11c− lineage phenotype (Lin)− cells could differentiate into CD8α+DCs in vivo after intravenous transplantation. Immunohistochemistry staining showed that donor-derived DCs mainly located in T-cell areas of the spleen. Functionally, these CD8α+CD11c−Lin− cell–derived DCs were capable of stimulating allogenic T-cell response, as well as secreting bioactive interleukin 12 p70 and interferon γ. Freshly isolated CD8α+CD11c−Lin− cells expressed CC chemokine receptor (CCR)2, CCR5, and CCR7 messenger RNA, whereas CD8α+ DCs derived from CD8α+CD11c−Lin− cells further obtained the expression of CCR6 and macrophage-derived chemokine. Flow cytometry analysis showed that CD8α+CD11c−Lin− cells were identified in bone marrow and lymph nodes. Moreover, transplanted splenic CD8α+CD11c−Lin− cells could also home to thymus and lymph nodes and were capable of developing into CD8α+ DCs in these locations. However, CD8α+CD11c−Lin−cells failed to differentiate into CD8α− DCs, T cells, natural killer cells, or other myeloid lineage cells in irradiated chimeras. Taken together, all these findings suggest that CD8α+CD11c−Lin− cells are a committed precursor of CD8α+ DCs.


Antioxidants ◽  
2020 ◽  
Vol 9 (10) ◽  
pp. 1021
Author(s):  
Ho-Jae Lee ◽  
Joon Seok Park ◽  
Hyun Jung Yoo ◽  
Hae Min Lee ◽  
Byung Cheon Lee ◽  
...  

Immune activation associates with the intracellular generation of reactive oxygen species (ROS). To elicit effective immune responses, ROS levels must be balanced. Emerging evidence shows that ROS-mediated signal transduction can be regulated by selenoproteins such as methionine sulfoxide reductase B1 (MsrB1). However, how the selenoprotein shapes immunity remains poorly understood. Here, we demonstrated that MsrB1 plays a crucial role in the ability of dendritic cells (DCs) to provide the antigen presentation and costimulation that are needed for cluster of differentiation antigen four (CD4) T-cell priming in mice. We found that MsrB1 regulated signal transducer and activator of transcription-6 (STAT6) phosphorylation in DCs. Moreover, both in vitro and in vivo, MsrB1 potentiated the lipopolysaccharide (LPS)-induced Interleukin-12 (IL-12) production by DCs and drove T-helper 1 (Th1) differentiation after immunization. We propose that MsrB1 activates the STAT6 pathway in DCs, thereby inducing the DC maturation and IL-12 production that promotes Th1 differentiation. Additionally, we showed that MsrB1 promoted follicular helper T-cell (Tfh) differentiation when mice were immunized with sheep red blood cells. This study unveils as yet unappreciated roles of the MsrB1 selenoprotein in the innate control of adaptive immunity. Targeting MsrB1 may have therapeutic potential in terms of controlling immune reactions.


Blood ◽  
2000 ◽  
Vol 96 (5) ◽  
pp. 1865-1872 ◽  
Author(s):  
Miriam Merad ◽  
Lawrence Fong ◽  
Jakob Bogenberger ◽  
Edgar G. Engleman

Abstract Bone marrow-derived dendritic cells (DC) represent a family of antigen-presenting cells (APC) with varying phenotypes. For example, in mice, CD8α+ and CD8α− DC are thought to represent cells of lymphoid and myeloid origin, respectively. Langerhans cells (LC) of the epidermis are typical myeloid DC; they do not express CD8α, but they do express high levels of myeloid antigens such as CD11b and FcγR. By contrast, thymic DC, which derive from a lymphoid-related progenitor, express CD8α but only low levels of myeloid antigens. CD8α+ DC are also found in the spleen and lymph nodes (LN), but the origin of these cells has not been determined. By activating and labeling CD8α− epidermal LC in vivo, it was found that these cells expressed CD8α on migration to the draining LN. Similarly, CD8α− LC generated in vitro from a CD8 wild-type mouse and injected into the skin of a CD8αKO mouse expressed CD8α when they reached the draining LN. The results also show that CD8α+ LC are potent APC. After migration from skin, they localized in the T-cell areas of LN, secreted high levels of interleukin-12, interferon-γ, and chemokine-attracting T cells, and they induced antigen-specific T-cell activation. These results demonstrate that myeloid DC in the periphery can express CD8α when they migrate to the draining LN. CD8α expression on these DC appears to reflect a state of activation, mobilization, or both, rather than lineage.


2020 ◽  
Author(s):  
Yajing Liu ◽  
Lintong Yao ◽  
Yun Zhang ◽  
Wenhui shen ◽  
Chunxia Chen ◽  
...  

Abstract BackgroundVaccination is a promising anticancer strategy, but the limited delivery routes and short retention of antigens and immunomodulatory agents are problems that need to be solved in vaccine design. Because silicon nanoparticles have a tunable pore size and high loading capacity, they have been used in a variety of drug delivery systems, but their roles in tumor vaccine and tumor immunotherapy need to be examined.MethodsCD40 mAb was attached to mesoporous silica nanoparticles (MSNs) through covalent conjunction, and MSN-CD40/OVA/CpG nanoparticles were examined by Fourier transform-infrared spectroscopy, transmission electron microscopy and nanoparticle analyzer. In vitro functions of nanoparticles were detected by cytotoxicity, cellular uptake, DC maturation, cross-presentation and T cell priming. In vivo functions were monitored by tumor elimination, DC maturation, cross-presentation and T cell activity.ResultsWe encapsulated anti-CD40 monoclonal antibodies, ovalbumin (OVA) antigen, and a toll-like receptor-9 agonist (CpG) in mesoporous silica nanoparticles (MSNs). The resulting MSN-CD40/OVA/CpG nanoparticles were efficiently phagocytized by splenocytes and bone marrow-derived dendritic cells (BMDC). The MSN-CD40/OVA/CpG nanoparticles induced the BMDC to express the costimulatory molecules CD80 and CD86, and release tumor necrosis factor-α. We found that MSN-CD40/OVA/CpG nanoparticles correctly enhanced antigen cross-priming, and stimulated T cell proliferation and interferon γ (IFNγ) production in vitro. In vivo, the MSN-CD40/OVA/CpG nanoparticles strongly increased intracellular IFNγ secretion and its release from OVA257–264 peptide-specific splenocytes into the cell supernatant, induced dendritic cell expression of major histocompatibility complex-II, and stimulated lymphocyte CD80 and CD86 expression. The MSN-CD40/OVA/CpG nanoparticles also inhibited tumor growth, enhanced tumor infiltration of CD8+ and CD4+ T cells, and stimulated IFNγ secretion from splenocytes. In conclusion, we believe these MSN-CD40/OVA/CpG nanoparticles are a promising strategy for improving antigen cross-presentation, cytotoxic T lymphocyte immune activity, and anti-tumor immunotherapy.


2009 ◽  
Vol 27 (3) ◽  
pp. 736-742 ◽  
Author(s):  
Yu-gang Wu ◽  
Guang-zhou Wu ◽  
Liang Wang ◽  
Yan-Yun Zhang ◽  
Zhong Li ◽  
...  

2006 ◽  
Vol 80 (4) ◽  
pp. 1826-1836 ◽  
Author(s):  
Allison T. Thiele ◽  
Tina L. Sumpter ◽  
Joanna A. Walker ◽  
Qi Xu ◽  
Cheong-Hee Chang ◽  
...  

ABSTRACT Adenovirus (Ad) infection has been identified as predisposing hosts to the development of pulmonary disease through unknown mechanisms. Lung dendritic cells (DCs) are vital for initiating pulmonary immune responses; however, the effects of Ad infection on primary lung DC have not been studied. In contrast to the effects on bone marrow- and monocyte-derived DCs, the current study shows that Ad infection of murine BALB/c lung DCs in vitro and in vivo suppresses DC-induced T-cell proliferation. The effect of Ad on DCs was not due to a downregulation of major histocompatibility complex or costimulatory molecules. Analysis of the production of interleukin-12 (IL-12), alpha interferon (IFN-α), and IFN-γ by the Ad-infected DCs shows no significant differences over noninfected control lung DCs. Ad-induced suppression was not due to a deficiency of IL-2 or other DC-secreted factors and was dependent on viral protein synthesis, as UV irradiation of Ad abrogated the suppressive effect. Results suggest that Ad-infected DCs induce T cells to be nonresponsive to IL-2 during primary coculture, as the addition of IL-2 in secondary cultures recovered T-cell proliferation. In vivo studies supported in vitro results showing that Ad infection resulted in lung T cells with decreased proliferative ability. This study demonstrates that Ad infection induces local immunoincompetence by altering DC-T-cell interactions.


Blood ◽  
2006 ◽  
Vol 108 (4) ◽  
pp. 1189-1197 ◽  
Author(s):  
Hua Tang ◽  
Zhenhong Guo ◽  
Minghui Zhang ◽  
Jianli Wang ◽  
Guoyou Chen ◽  
...  

Abstract Regulatory dendritic cells (DCs) have been reported recently, but their origin is poorly understood. Our previous study demonstrated that splenic stroma can drive mature DCs to proliferate and differentiate into regulatory DCs, and their natural counterpart with similar regulatory function in normal spleens has been identified. Considering that the spleen microenvironment supports hematopoiesis and that hematopoietic stem cells (HSCs) are found in spleens of adult mice, we wondered whether splenic microenvironment could differentiate HSCs into regulatory DCs. In this report, we demonstrate that endothelial splenic stroma induce HSCs to differentiate into a distinct regulatory DC subset with high expression of CD11b but low expression of Ia. CD11bhiIalo DCs secreting high levels of TGF-β, IL-10, and NO can suppress T-cell proliferation both in vitro and in vivo. Furthermore, CD11bhiIalo DCs have the ability to potently suppress allo-DTH in vivo, indicating their preventive or therapeutic perspectives for some immunologic disorders. The inhibitory function of CD11bhiIalo DCs is mediated through NO but not through induction of regulatory T (Treg) cells or T-cell anergy. IL-10, which is secreted by endothelial splenic stroma, plays a critical role in the differentiation of the regulatory CD11bhiIalo DCs from HSCs. These results suggest that splenic microenvironment may physiologically induce regulatory DC differentiation in situ.


Sign in / Sign up

Export Citation Format

Share Document