Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
1.
Ann N Y Acad Sci ; 1445(1): 62-73, 2019 06.
Article in English | MEDLINE | ID: mdl-30945313

ABSTRACT

One successful class of cancer immunotherapies, immune checkpoint inhibitory antibodies, disrupts key pathways that regulate immune checkpoints, such as cytotoxic T lymphocyte-associated antigen-4 (CTLA-4). These agents unleash the potency of antigen-experienced T cells that have already been induced as a consequence of the existing tumor. But only 20% of cancers naturally induce T cells. For most cancers, vaccines are require to induce and mobilize T effector cells (Teffs ) to traffick into tumors. We evaluated the effects of anti-CTLA-4 given in combination with an antigen-specific dendritic cell vaccine on intratumoral Teffs in a murine pancreatic cancer model. The dendritic cell-targeted tumor antigen plus anti-CTLA-4 significantly increased the number of vaccine-induced CD4+ Teffs within the tumor. This increase was accompanied by a reduction in the size of the peripheral CD4+ Teff pool. We also found that IL-3 production by activated CD4+ T cells was significantly increased with this combination. Importantly, the CD4+ Teff response was attenuated in Il3-/- mice, suggesting mediation of the effect by IL-3. Finally, the induced T cell infiltration was associated with activation of the tumor endothelium by T cell-derived IL-3. Our findings collectively provide a new insight into the mechanism driving Teff infiltration and vascular activation in a murine pancreatic cancer model, specifically identifying a new role for IL-3 in the anticancer immune response.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/antagonists & inhibitors , Cancer Vaccines/immunology , Interleukin-3/immunology , Pancreatic Neoplasms/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Disease Models, Animal , Interleukin-3/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Neoplasms/immunology
2.
Immun Inflamm Dis ; 7(2): 55-67, 2019 06.
Article in English | MEDLINE | ID: mdl-28474788

ABSTRACT

INTRODUCTION: Targeting antigens to dendritic cells (DCs) in vivo via a DC-restricted endocytic receptor, DEC205, has been validated to enhance immunity in several vaccine platforms. Particularly atttractive is selected delivery of proteins to DCs in vivo because it enables proteins to be more immunogenic and provides a cheaper and effective way for repeated immunizations. METHODS: In this study, we tested the efficacy of a single chain antibody to DEC205 (scDEC) to deliver protein antigens selectively to DCs in vivo and to induce protective immunity. RESULTS: In comparison to soluble Ovalbumin (OVA) antigen, when recombinant scDEC:OVA protein was injected subcutaneously (s.c.) into mice, the OVA protein was selectively presented by DCs to both TCR transgenic CD8+ and CD4+ T cells approximately 500 and 100 times more efficient than soluble OVA, respectively, and could persist for seven days following s.c. injection of the scDEC205:OVA. Similarly selective targeting of HIV Gag P24 to DCs in vivo using scDEC-Gag protein plus polyICLC vaccine resulted in strong, long lasting, polyfuntional CD4+ T cells in mice which were protective against airway challenge by a recombinant vaccinia-gag virus. CONCLUSION: Thus targeting protein antigens to DCs using scDEC can be used either alone or in combination with other strategies for effective immunization.


Subject(s)
AIDS Vaccines/immunology , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Dendritic Cells/immunology , Single-Chain Antibodies/immunology , gag Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , Adjuvants, Immunologic , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CHO Cells , Cell Line , Cricetulus , Dendritic Cells/metabolism , Disease Models, Animal , Female , HIV/immunology , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/prevention & control , Humans , Immunization , Immunogenicity, Vaccine/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Single-Chain Antibodies/pharmacology , T-Cell Antigen Receptor Specificity/immunology
4.
PLoS One ; 11(1): e0146412, 2016.
Article in English | MEDLINE | ID: mdl-26745371

ABSTRACT

Cytokines secreted from dendritic cells (DCs) play an important role in the regulation of T helper (Th) cell differentiation and activation into effector cells. Therefore, controlling cytokine secretion from DCs may potentially regulate Th differentiation/activation. DCs also induce de-novo generation of regulatory T cells (Treg) that modulate the immune response. In the current study we used the mixed leukocyte reaction (MLR) to investigate the effect of allospecific Treg on IL-12, TNFα and IL-6 secretion by DCs. Treg cells were found to markedly down-regulate IL-12 secretion from DCs following stimulation with TLR7/8 agonist. This down-regulation of IL-12 was neither due to a direct suppression of its production by the DCs nor a result of marked DC death. We found that IL-12 was rather actively consumed by Treg cells. IL-12 consumption was mediated by a subpopulation of IL-12Rß2-expressing Treg cells and was dependent on MHC class-II expressed on dendritic cells. Furthermore, IL-12 consumption by Tregs increased their suppressive effect on T cell proliferation and Th1 activation. These results provide a new pathway of Th1 response regulation where IL-12 secreted by DCs is consumed by a sub-population of IL-12Rß2-expressing Treg cells. Consumption of IL-12 by Tregs not only reduces the availability of IL-12 to Th effector cells but also enhances the Treg immunosuppressive effect. This DC-induced IL-12Rß2-expressing Treg subpopulation may have a therapeutic advantage in suppressing Th1 mediated autoimmunity.


Subject(s)
Cell Lineage/immunology , Dendritic Cells/cytology , Interleukin-12/genetics , Receptors, Interleukin-12/genetics , T-Lymphocytes, Regulatory/cytology , Th1 Cells/cytology , Animals , Biological Transport , Cell Differentiation , Cell Lineage/genetics , Cell Proliferation , Dendritic Cells/immunology , Female , Gene Expression Regulation , Immunophenotyping , Interleukin-12/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lymphocyte Culture Test, Mixed , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Receptors, Interleukin-12/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th1-Th2 Balance , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
5.
PLoS One ; 10(4): e0124828, 2015.
Article in English | MEDLINE | ID: mdl-25915045

ABSTRACT

Airways infection with Mycobacterium tuberculosis (Mtb) is contained mostly by T cell responses, however, Mtb has developed evasion mechanisms which affect antigen presenting cell (APC) maturation/recruitment delaying the onset of Ag-specific T cell responses. Hypothetically, bypassing the natural infection routes by delivering antigens directly to APCs may overcome the pathogen's naturally evolved evasion mechanisms, thus facilitating the induction of protective immune responses. We generated a murine monoclonal fusion antibody (α-DEC-ESAT) to deliver Early Secretory Antigen Target (ESAT)-6 directly to DEC205+ APCs and to assess its in vivo effects on protection associated responses (IFN-γ production, in vivo CTL killing, and pulmonary mycobacterial load). Treatment with α-DEC-ESAT alone induced ESAT-6-specific IFN-γ producing CD4+ T cells and prime-boost immunization prior to Mtb infection resulted in early influx (d14 post-infection) and increased IFN-γ+ production by specific T cells in the lungs, compared to scarce IFN-γ production in control mice. In vivo CTL killing was quantified in relevant tissues upon transferring target cells loaded with mycobacterial antigens. During infection, α-DEC-ESAT-treated mice showed increased target cell killing in the lungs, where histology revealed cellular infiltrate and considerably reduced bacterial burden. Targeting the mycobacterial antigen ESAT-6 to DEC205+ APCs before infection expands specific T cell clones responsible for early T cell responses (IFN-γ production and CTL activity) and substantially reduces lung bacterial burden. Delivering mycobacterial antigens directly to APCs provides a unique approach to study in vivo the role of APCs and specific T cell responses to assess their potential anti-mycobacterial functions.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, CD/genetics , Antigens, CD/metabolism , Bacterial Load , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Line , Cytotoxicity, Immunologic , Disease Models, Animal , Flow Cytometry , Immunization , Interferon-gamma/biosynthesis , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Male , Mice , Minor Histocompatibility Antigens , Mycobacterium tuberculosis/pathogenicity , Peptides/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/pathology
6.
EMBO Mol Med ; 6(9): 1191-204, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25085878

ABSTRACT

Skin dendritic cells (DCs) control the immunogenicity of cutaneously administered vaccines. Antigens targeted to DCs via the C-type lectin Langerin/CD207 are cross-presented to CD8(+) T cells in vivo. We investigated the relative roles of Langerhans cells (LCs) and Langerin(+) dermal DCs (dDCs) in different vaccination settings. Poly(I:C) and anti-CD40 agonist antibody promoted cytotoxic responses upon intradermal immunization with ovalbumin (OVA)-coupled anti-Langerin antibodies (Langerin/OVA). This correlated with CD70 upregulation in Langerin(+) dDCs, but not LCs. In chimeric mice where Langerin targeting was restricted to dDCs, CD8(+) T-cell memory was enhanced. Conversely, providing Langerin/OVA exclusively to LCs failed to prime cytotoxicity, despite initial antigen cross-presentation to CD8(+) T cells. Langerin/OVA combined with imiquimod could not prime CD8(+) T cells and resulted in poor cytotoxicity in subsequent responses. This tolerance induction required targeting and maturation of LCs. Altogether, Langerin(+) dDCs prime long-lasting cytotoxic responses, while cross-presentation by LCs negatively influences CD8(+) T-cell priming. Moreover, this highlights that DCs exposed to TLR agonists can still induce tolerance and supports the existence of qualitatively different DC maturation programs.


Subject(s)
Antigens, Surface/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Langerhans Cells/immunology , Lectins, C-Type/immunology , Mannose-Binding Lectins/immunology , Animals , Antigen Presentation , Cross-Priming , Immunologic Memory , Mice , Mice, Inbred C57BL , Models, Immunological
7.
J Exp Med ; 211(9): 1875-91, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25135299

ABSTRACT

DCs are critical for initiating immunity. The current paradigm in vaccine biology is that DCs migrating from peripheral tissue and classical lymphoid-resident DCs (cDCs) cooperate in the draining LNs to initiate priming and proliferation of T cells. Here, we observe subcutaneous immunity is Fms-like tyrosine kinase 3 ligand (Flt3L) dependent. Flt3L is rapidly secreted after immunization; Flt3 deletion reduces T cell responses by 50%. Flt3L enhances global T cell and humoral immunity as well as both the numbers and antigen capture capacity of migratory DCs (migDCs) and LN-resident cDCs. Surprisingly, however, we find immunity is controlled by cDCs and actively tempered in vivo by migDCs. Deletion of Langerin(+) DC or blockade of DC migration improves immunity. Consistent with an immune-regulatory role, transcriptomic analyses reveals different skin migDC subsets in both mouse and human cluster together, and share immune-suppressing gene expression and regulatory pathways. These data reveal that protective immunity to protein vaccines is controlled by Flt3L-dependent, LN-resident cDCs.


Subject(s)
Dendritic Cells/immunology , Membrane Proteins/immunology , Vaccines/immunology , Animals , Antigen Presentation , Antigens, Surface/genetics , Antigens, Surface/immunology , Dendritic Cells/classification , Female , Gene Expression , Humans , Immunity, Humoral/genetics , Injections, Intradermal , Injections, Subcutaneous , Interferon-gamma/biosynthesis , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Ligands , Male , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/immunology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/immunology , Proteins/immunology , T-Lymphocyte Subsets/immunology , Transcription Factors/immunology , Vaccines/administration & dosage
8.
PLoS Biol ; 12(1): e1001759, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24409099

ABSTRACT

Type I interferons (IFNs) play an important role in direct antiviral defense as well as linking the innate and adaptive immune responses. On dendritic cells (DCs), IFNs facilitate their activation and contribute to CD8(+) and CD4(+) T cell priming. However, the precise molecular mechanism by which IFNs regulate maturation and immunogenicity of DCs in vivo has not been studied in depth. Here we show that, after in vivo stimulation with the TLR ligand poly IC, IFNs dominate transcriptional changes in DCs. In contrast to direct TLR3/mda5 signaling, IFNs are required for upregulation of all pathways associated with DC immunogenicity. In addition, metabolic pathways, particularly the switch from oxidative phosphorylation to glycolysis, are also regulated by IFNs and required for DC maturation. These data provide evidence for a metabolic reprogramming concomitant with DC maturation and offer a novel mechanism by which IFNs modulate DC maturation.


Subject(s)
Dendritic Cells/immunology , Gene Expression Regulation/immunology , Glycolysis/drug effects , Interferon-alpha/genetics , Oxidative Phosphorylation/drug effects , Poly I-C/pharmacology , Adaptive Immunity , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Immunity, Innate , Injections, Intraperitoneal , Interferon-Induced Helicase, IFIH1 , Interferon-alpha/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Transcription, Genetic
9.
J Invest Dermatol ; 134(5): 1265-1275, 2014 May.
Article in English | MEDLINE | ID: mdl-24288007

ABSTRACT

Skin-derived dendritic cells (DCs) are potent antigen-presenting cells with critical roles in both adaptive immunity and tolerance to self. Skin DCs carry antigens and constitutively migrate to the skin-draining lymph nodes (LNs). In mice, Langerin-CD11b- dermal DCs are a low-frequency, heterogeneous, migratory DC subset that traffics to LNs (Langerin-CD11b- migDCs). Here, we build on the observation that Langerin-CD11b- migDCs are Fms-like tyrosine kinase 3 ligand (Flt3L) dependent and strongly Flt3L responsive, which may relate them to classical DCs. Examination of DC capture of FITC from painted skin, DC isolation from skin explant culture, and from the skin of CCR7 knockout mice, which accumulate migDCs, demonstrate these cells are cutaneous residents. Langerin-CD11b- Flt3L-responsive DCs are largely CD24(+) and CX3CR1(low) and can be depleted from Zbtb46-DTR mice, suggesting classical DC lineage. Langerin-CD11b- migDCs present antigen with equal efficiency to other DC subsets ex vivo, including classical CD8α cDCs and Langerin+CD103+ dermal DCs. Finally, transcriptome analysis suggests a close relationship with other skin DCs, and a lineage relationship with other classical DCs. This work demonstrates that Langerin- CD11b- dermal DCs, a previously overlooked cell subset, may be an important contributor to the cutaneous immune environment.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/immunology , Membrane Proteins/immunology , Skin/cytology , Skin/immunology , Adaptive Immunity/immunology , Animals , Antigens, Surface/immunology , Antigens, Surface/metabolism , CD11b Antigen/immunology , CD11b Antigen/metabolism , CX3C Chemokine Receptor 1 , Cell Movement/immunology , Female , Immune Tolerance/immunology , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Male , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR7/genetics , Receptors, Chemokine/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
10.
J Exp Med ; 210(9): 1871-88, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-23960190

ABSTRACT

Developing efficacious vaccines against enteric diseases is a global challenge that requires a better understanding of cellular recruitment dynamics at the mucosal surfaces. The current paradigm of T cell homing to the gastrointestinal (GI) tract involves the induction of α4ß7 and CCR9 by Peyer's patch and mesenteric lymph node (MLN) dendritic cells (DCs) in a retinoic acid-dependent manner. This paradigm, however, cannot be reconciled with reports of GI T cell responses after intranasal (i.n.) delivery of antigens that do not directly target the GI lymphoid tissue. To explore alternative pathways of cellular migration, we have investigated the ability of DCs from mucosal and nonmucosal tissues to recruit lymphocytes to the GI tract. Unexpectedly, we found that lung DCs, like CD103(+) MLN DCs, up-regulate the gut-homing integrin α4ß7 in vitro and in vivo, and induce T cell migration to the GI tract in vivo. Consistent with a role for this pathway in generating mucosal immune responses, lung DC targeting by i.n. immunization induced protective immunity against enteric challenge with a highly pathogenic strain of Salmonella. The present report demonstrates novel functional evidence of mucosal cross talk mediated by DCs, which has the potential to inform the design of novel vaccines against mucosal pathogens.


Subject(s)
Cell Movement/immunology , Dendritic Cells/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Immunity, Mucosal/immunology , Lung/pathology , T-Lymphocytes/immunology , Administration, Intranasal , Adoptive Transfer , Animals , Antigens, CD/metabolism , Antigens, Surface/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Movement/drug effects , Dendritic Cells/drug effects , Dendritic Cells/pathology , Fingolimod Hydrochloride , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Immunity, Mucosal/drug effects , Immunization , Integrins/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Propylene Glycols/pharmacology , Receptors, CCR/metabolism , Salmonella/drug effects , Salmonella/immunology , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/pathology , Salmonella Infections, Animal/prevention & control , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology
11.
PLoS One ; 8(6): e67453, 2013.
Article in English | MEDLINE | ID: mdl-23840706

ABSTRACT

Efficient vaccination against the parasite Leishmania major, the causative agent of human cutaneous leishmaniasis, requires development of type 1 T-helper (Th1) CD4(+) T cell immunity. Because of their unique capacity to initiate and modulate immune responses, dendritic cells (DCs) are attractive targets for development of novel vaccines. In this study, for the first time, we investigated the capacity of a DC-targeted vaccine to induce protective responses against L. major. To this end, we genetically engineered the N-terminal portion of the stress-inducible 1 protein of L. major (LmSTI1a) into anti-DEC205/CD205 (DEC) monoclonal antibody (mAb) and thereby delivered the conjugated protein to DEC(+) DCs in situ in the intact animal. Delivery of LmSTI1a to adjuvant-matured DCs increased the frequency of antigen-specific CD4(+) T cells producing IFN-γ(+), IL-2(+), and TNF-α(+) in two different strains of mice (C57BL/6 and Balb/c), while such responses were not observed with the same doses of a control Ig-LmSTI1a mAb without receptor affinity or with non-targeted LmSTI1a protein. Using a peptide library for LmSTI1a, we identified at least two distinct CD4(+) T cell mimetopes in each MHC class II haplotype, consistent with the induction of broad immunity. When we compared T cell immune responses generated after targeting DCs with LmSTI1a or other L. major antigens, including LACK (Leishmania receptor for activated C kinase) and LeIF (Leishmania eukaryotic ribosomal elongation and initiation factor 4a), we found that LmSTI1a was superior for generation of IFN-γ-producing CD4(+) T cells, which correlated with higher protection of susceptible Balb/c mice to a challenge with L. major. For the first time, this study demonstrates the potential of a DC-targeted vaccine as a novel approach for cutaneous leishmaniasis, an increasing public health concern that has no currently available effective treatment.


Subject(s)
Antigens, Protozoan/immunology , Dendritic Cells/immunology , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Protozoan Proteins/immunology , Th1 Cells/immunology , Animals , Cell Proliferation , Dendritic Cells/parasitology , Female , Immunization , Interferon-gamma/metabolism , Leishmaniasis, Cutaneous/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism
12.
Blood ; 121(25): 5034-44, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23482932

ABSTRACT

Functional differences between human dendritic cell (DC) subsets and the potential benefits of targeting them with vaccines remain poorly defined. Here we describe that mice with reconstituted human immune system components (huNSG mice) develop all human conventional and plasmacytoid DC compartments in lymphoid organs. Testing different Toll-like receptor agonists for DC maturation in vivo, we found that IL-12p70 and interferon (IFN)-α production correlated with the maturation of CD141+ (BDCA3+) conventional DCs in huNSG mice. Furthermore, depletion of CD141+ DCs before stimulation significantly reduced IFN-α levels in vivo. This DC subset produced similar total amounts but different subtypes of IFN-α in response to synthetic double-stranded RNA compared with plasmacytoid DCs in response to a single-stranded RNA equivalent. Moreover, synthetic double-stranded RNA as adjuvant and antigen targeting to the endocytic receptor DEC-205, a combination that focuses antigen presentation for T-cell priming on CD141+ DCs, stimulated antigen-specific human CD4+ T-cell responses. Thus, the human CD141+ DC subset is a prominent source of IFN-α and interleukin-12 production and should be further evaluated for vaccine development.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/immunology , Interferon-alpha/biosynthesis , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , RNA, Double-Stranded/immunology , Receptors, Cell Surface/immunology , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/cytology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interferon-alpha/immunology , Mice , Minor Histocompatibility Antigens
13.
Vaccine ; 31(19): 2366-71, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23499580

ABSTRACT

Dengue virus (DENV) is the causal agent of severe disease and, in some cases, mortality in humans, but no licensed vaccines against dengue are available. An effective vaccine against dengue requires long-term humoral and cellular immune responses. Several viral proteins have been the subjects of intense research, especially the envelope (E) protein, aimed at developing a vaccine. Domain III of the envelope protein (EDIII) has been identified as a potential candidate because it is involved in binding to host cell receptors and contains epitopes that elicit virus neutralizing antibodies. However, this domain is not sufficiently antigenic when is expressed and administered as antigen to elicit a strong immune response. One alternative to enhance immunogenicity is to target the antigen to dendritic cells to induce T-cells for broad antibody responses. In this work, a single chain antibody fragment (scFv) raised against the DEC-205 receptor fused with the EDIII was successfully expressed in Nicotiana benthamiana. The recombinant protein was expressed and purified from the plant and evaluated in BALB/c mice to test its immunogenicity and ability to induce neutralizing antibodies against DENV. The mice immunized with the recombinant protein produced specific and strong humoral immune responses to DENV. Only two immunizations were required to generate a memory response to DENV without the presence of adjuvants. Also, recognition of the recombinant protein with sera from DENV-infected patients was observed. These findings suggest that this strategy has potential for development of an effective vaccine against DENV.


Subject(s)
Antigens, CD/immunology , Dengue Vaccines/immunology , Dengue/prevention & control , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , Recombinant Fusion Proteins/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Antibody Formation , Dengue/immunology , Dengue Virus/immunology , Dengue Virus/metabolism , Epitopes/immunology , Epitopes/metabolism , Humans , Mice , Mice, Inbred BALB C , Minor Histocompatibility Antigens , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism
14.
Nat Chem Biol ; 9(4): 250-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23416331

ABSTRACT

Targeted delivery of antigens to dendritic cells (DCs) is a promising vaccination strategy. However, to ensure immunity, the approach depends on coadministration of an adjuvant. Here we ask whether targeting of both adjuvant and antigen to DCs is sufficient to induce immunity. Using a protein ligation method, we develop a general approach for linking the immune stimulant, poly dA:dT (pdA:dT), to a monoclonal antibody (mAb) specific for DEC205 (DEC). We show that DEC-specific mAbs deliver pdA:dT to DCs for the efficient production of type I interferon in human monocyte-derived DCs and in mice. Notably, adaptive T-cell immunity is elicited when mAbs specific for DEC-pdA:dT are used as the activation stimuli and are administered together with a DC-targeted antigen. Collectively, our studies indicate that DCs can integrate innate and adaptive immunity in vivo and suggest that dual delivery of antigen and adjuvant to DCs might be an efficient approach to vaccine development.


Subject(s)
Adaptive Immunity/drug effects , Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Antigens/immunology , Dendritic Cells/drug effects , Immunity, Innate/drug effects , Immunoconjugates/immunology , Lectins, C-Type/immunology , Poly dA-dT/immunology , Receptors, Cell Surface/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antigens/administration & dosage , Antigens/chemistry , Antigens, CD/administration & dosage , Antigens, CD/chemistry , Dendritic Cells/immunology , Drug Delivery Systems , Genetic Vectors , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Interferon Type I/biosynthesis , Interferon Type I/immunology , Lectins, C-Type/administration & dosage , Lectins, C-Type/chemistry , Mice , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Plasmids , Poly dA-dT/administration & dosage , Poly dA-dT/chemistry , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/chemistry
15.
J Clin Invest ; 123(2): 844-54, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23298832

ABSTRACT

Harnessing DCs for immunotherapies in vivo requires the elucidation of the physiological role of distinct DC populations. Migratory DCs traffic from peripheral tissues to draining lymph nodes charged with tissue self antigens. We hypothesized that these DC populations have a specialized role in the maintenance of peripheral tolerance, specifically, to generate suppressive Foxp3+ Tregs. To examine the differential capacity of migratory DCs versus blood-derived lymphoid-resident DCs for Treg generation in vivo, we targeted a self antigen, myelin oligodendrocyte glycoprotein, using antibodies against cell surface receptors differentially expressed in these DC populations. Using this approach together with mouse models that lack specific DC populations, we found that migratory DCs have a superior ability to generate Tregs in vivo, which in turn drastically improve the outcome of experimental autoimmune encephalomyelitis. These results provide a rationale for the development of novel therapies targeting migratory DCs for the treatment of autoimmune diseases.


Subject(s)
Dendritic Cells/immunology , Peripheral Tolerance/immunology , Animals , Antigens, CD/immunology , Antigens, Surface/immunology , Autoantigens , Cell Movement/immunology , Dendritic Cells/classification , Dendritic Cells/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Forkhead Transcription Factors/metabolism , Langerhans Cells/immunology , Langerhans Cells/physiology , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Mannose-Binding Lectins/antagonists & inhibitors , Mannose-Binding Lectins/immunology , Mice , Mice, Knockout , Minor Histocompatibility Antigens , Myelin-Oligodendrocyte Glycoprotein/immunology , Peripheral Tolerance/physiology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/immunology , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , T-Lymphocytes, Regulatory/immunology
16.
Vaccine ; 30(45): 6359-67, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22947140

ABSTRACT

It is of great interest to develop a pneumonic plague vaccine that would induce combined humoral and cellular immunity in the lung. Here we investigate a novel approach based on targeting of dendritic cells using the DEC-205/CD205 receptor (DEC) via the intranasal route as way to improve mucosal cellular immunity to the vaccine. Intranasal administration of Yersinia pestis LcrV (V) protein fused to anti-DEC antibody together with poly IC as an adjuvant induced high frequencies of IFN-γ secreting CD4(+) T cells in the airway and lung as well as pulmonary IgG and IgA antibodies. Anti-DEC:LcrV was more efficient to induce IFN-γ/TNF-α/IL-2 secreting polyfunctional CD4(+) T cells when compared to non-targeted soluble protein vaccine. In addition, the intranasal route of immunization with anti-DEC:LcrV was associated with improved survival upon pulmonary challenge with the virulent CO92 Y. pestis. Taken together, these data indicate that targeting dendritic cells via the mucosal route is a potential new avenue for the development of a mucosal vaccine against pneumonic plague.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/immunology , Immunity, Mucosal , Lectins, C-Type/immunology , Lung/immunology , Plague/prevention & control , Receptors, Cell Surface/immunology , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Immunity, Cellular , Immunity, Humoral , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Plague Vaccine/immunology , Poly I-C/immunology , Poly I-C/pharmacology , Pore Forming Cytotoxic Proteins/immunology , Yersinia pestis/pathogenicity
17.
PLoS One ; 7(7): e41897, 2012.
Article in English | MEDLINE | ID: mdl-22860026

ABSTRACT

Mass Spectrometry (MS) is becoming a preferred method to identify class I and class II peptides presented on major histocompability complexes (MHC) on antigen presenting cells (APC). We describe a combined computational and MS approach to identify exogenous MHC II peptides presented on mouse spleen dendritic cells (DCs). This approach enables rapid, effective screening of a large number of possible peptides by a computer-assisted strategy that utilizes the extraordinary human ability for pattern recognition. To test the efficacy of the approach, a mixture of epitope peptide mimics (mimetopes) from HIV gag p24 sequence were added exogenously to Fms-like tyrosine kinase 3 ligand (Flt3L)-mobilized splenic DCs. We identified the exogenously added peptide, VDRFYKTLRAEQASQ, and a second peptide, DRFYKLTRAEQASQ, derived from the original exogenously added 15-mer peptide. Furthermore, we demonstrated that our strategy works efficiently with HIV gag p24 protein when delivered, as vaccine protein, to Flt3L expanded mouse splenic DCs in vitro through the DEC-205 receptor. We found that the same MHC II-bound HIV gag p24 peptides, VDRFYKTLRAEQASQ and DRFYKLTRAEQASQ, were naturally processed from anti-DEC-205 HIV gag p24 protein and presented on DCs. The two identified VDRFYKTLRAEQASQ and DRFYKLTRAEQASQ MHC II-bound HIV gag p24 peptides elicited CD4(+) T-cell mediated responses in vitro. Their presentation by DCs to antigen-specific T cells was inhibited by chloroquine (CQ), indicating that optimal presentation of these exogenously added peptides required uptake and vesicular trafficking in mature DCs. These results support the application of our strategy to identify and characterize peptide epitopes derived from vaccine proteins processed by DCs and thus has the potential to greatly accelerate DC-based vaccine development.


Subject(s)
Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Epitopes, T-Lymphocyte/immunology , HIV Core Protein p24/immunology , Amino Acid Sequence , Animals , Antigen Presentation , Antigens, Viral/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/metabolism , HIV Core Protein p24/metabolism , Histocompatibility Antigens Class II/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Peptide Fragments/immunology , Peptide Fragments/metabolism
18.
J Immunol Methods ; 384(1-2): 184-90, 2012 Oct 31.
Article in English | MEDLINE | ID: mdl-22841832

ABSTRACT

DEC205/CD205, an endocytic receptor of C-type multilectin, is expressed highly in dendritic cells (DCs). DEC205 was shown to efficiently deliver vaccine antigens in surrogate ligands to the antigen processing and presentation machinery of DCs, which resulted in the development of DC-targeted vaccines employing anti-DC monoclonal antibodies (mAbs). During our studies to characterize a variety of anti-DC mAbs including anti-DEC205 by flow cytometric analysis, we discovered that a secondary anti-immunoglobulin antibody conjugated with PE-Cy5.5 bound strongly to the cells expressing mouse DEC205 (mDEC205) without incubation of a primary anti-mDEC205 mAb. In the present study we demonstrate that various antibodies and streptavidin conjugated with PE-Cy5.5 bind to the mDEC205-expressing cells including CHO, KIT6, and HEK293 cells. The interaction between the PE-Cy5.5 conjugates and the cells expressing mDEC205 appears distinctive, since none of the PE-Cy5.5 conjugates bind to the cells that express human DEC205 on surface. Besides, only PE-Cy5.5 conjugates bind strongly to mDEC205-expressing cells; PerCP-Cy5.5, APC-Cy5.5, and Cy5.5 conjugates bind weakly; PE, PE-Cy5, Cy5, FITC, or Alexa488 conjugates do not bind to mDEC205-expressing cells. Therefore the use of PE-Cy5.5 conjugates, widely utilized in multicolor flow cytometry, requires precaution against nonspecific binding to mDEC205-positive cells.


Subject(s)
Antigens, CD/immunology , Immunoconjugates/immunology , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Binding, Competitive/immunology , CHO Cells , Carbocyanines/chemistry , Cricetinae , Cricetulus , Dendritic Cells/immunology , Flow Cytometry , Gene Expression , HEK293 Cells , Humans , Hybridomas , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Protein Binding/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Species Specificity , Streptavidin/chemistry
19.
J Exp Med ; 209(5): 1011-28, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22547652

ABSTRACT

Delta-like ligand 4 (Dll4)-Notch signaling is essential for T cell development and alternative thymic lineage decisions. How Dll4-Notch signaling affects pro-T cell fate and thymic dendritic cell (tDC) development is unknown. We found that Dll4 pharmacological blockade induces accumulation of tDCs and CD4(+)CD25(+)FoxP3(+) regulatory T cells (T(reg) cells) in the thymic cortex. Both genetic inactivation models and anti-Dll4 antibody (Ab) treatment promote de novo natural T(reg) cell expansion by a DC-dependent mechanism that requires major histocompatibility complex II expression on DCs. Anti-Dll4 treatment converts CD4(-)CD8(-)c-kit(+)CD44(+)CD25(-) (DN1) T cell progenitors to immature DCs that induce ex vivo differentiation of naive CD4(+) T cells into T(reg) cells. Induction of these tolerogenic DN1-derived tDCs and the ensuing expansion of T(reg) cells are Fms-like tyrosine kinase 3 (Flt3) independent, occur in the context of transcriptional up-regulation of PU.1, Irf-4, Irf-8, and CSF-1, genes critical for DC differentiation, and are abrogated in thymectomized mice. Anti-Dll4 treatment fully prevents type 1 diabetes (T1D) via a T(reg) cell-mediated mechanism and inhibits CD8(+) T cell pancreatic islet infiltration. Furthermore, a single injection of anti-Dll4 Ab reverses established T1D. Disease remission and recurrence are correlated with increased T(reg) cell numbers in the pancreas-draining lymph nodes. These results identify Dll4-Notch as a novel Flt3-alternative pathway important for regulating tDC-mediated T(reg) cell homeostasis and autoimmunity.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/physiology , Diabetes Mellitus, Type 1/prevention & control , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/metabolism , Animals , Antibodies/pharmacology , Blotting, Western , Calcium-Binding Proteins , DNA Primers/genetics , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Genes, MHC Class II/immunology , Intracellular Signaling Peptides and Proteins/immunology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Pancreas/pathology , Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Thymus Gland/immunology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
20.
Breast Cancer Res ; 14(2): R39, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-22397502

ABSTRACT

INTRODUCTION: Given their relative simplicity of manufacture and ability to be injected repeatedly, vaccines in a protein format are attractive for breast and other cancers. However, soluble human epidermal growth factor receptor (HER2)/neu protein as a vaccine has not been immunogenic. When protein is directly targeted to antigen uptake receptors, such as DEC205 (DEC), efficient processing and presentation of antigen take place. The aim of this study was to determine the immunogenicity of a HER2 protein vaccine that directly targets to DEC+ dendritic cells (DCs) in a mouse breast cancer model. METHODS: We genetically engineered the HER2 extracellular domain into a monoclonal antibody specific for DEC (DEC-HER2). Mice of various genetic backgrounds were immunized with DEC-HER2 in combination with DC maturation stimuli (poly IC ± CD40 Ab). Vaccine-induced T cell immunity was determined by analyzing the ability of CD4+/CD8+ T cell to produce interferon (IFN)-gamma and proliferate upon antigen rechallenge. Sera were assessed for the presence of antigen specific antibody (Ab). For vaccine efficacy, FVB/N mice were immunized with DEC-HER2 in combination with poly IC and protection against neu-expressing mammary tumors was assessed. Protection mechanisms and tumor-specific T cell responses were also evaluated. RESULTS: We demonstrate that DEC-HER2 fusion mAb, but not Ctrl Ig-HER2, elicits strong, broad and multifunctional CD4+ T cell immunity, CD8+ T cell responses, and humoral immunity specific for HER2 antigen. Cross-reactivity to rat neu protein was also observed. Importantly, mice xeno-primed with DEC-HER2 were protected from a neu-expressing mammary tumor challenge. Both CD4+ and CD8+ T cells mediated the tumor protection. Robust anti-tumor T cell immunity was detected in tumor protected mice. CONCLUSIONS: Immunization of mice with HER2 protein vaccine targeting DEC+ DCs in vivo induced high levels of T- and B-cell immunity. Non-targeted HER2 protein was poorly immunogenic for CD4+ and CD8+ T cells. This vaccination approach provided long-term survival benefit for mice challenged with neu-expressing tumor following as little as 2.7 µg of HER2 protein incorporated in the vaccine. Vaccine-induced CD4+ and CD8+ T cells were both essential for tumor protection. This immunization strategy demonstrates great potential towards the development of vaccines for breast cancer patients.


Subject(s)
Breast Neoplasms/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Receptor, ErbB-2/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/genetics , Cell Line, Tumor , Female , Humans , Immunity, Humoral , Interferon-gamma/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred Strains , Mice, Transgenic , Poly I-C/immunology , Poly I-C/pharmacology , Protein Structure, Tertiary/genetics , Receptor, ErbB-2/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL