Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Oncoimmunology ; 9(1): 1758606, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32923117

ABSTRACT

Antitumor immunity is mediated by Th1 CD4+ and CD8+ T lymphocytes, which induce tumor-specific cytolysis, whereas Th17 CD4+ T cells have been described to promote tumor growth. Here, we explored the influence of IL-17 on the ability of therapeutic vaccines to induce the rejection of tumors in mice using several adjuvants known to elicit either Th1 or Th17-type immunity. Immunization of mice with Th1-adjuvanted vaccine induced high levels of IFN-γ-producing T cells, whereas injection with Th17-promoting adjuvants triggered the stimulation of both IL-17 and IFN-γ-producing T cells. However, despite their capacity to induce strong Th1 responses, these Th17-promoting adjuvants failed to induce the eradication of tumors. In addition, the systemic administration of IL-17A strongly decreases the therapeutic effect of Th1-adjuvanted vaccines in two different tumor models. This suppressive effect correlated with the capacity of systemically delivered IL-17A to inhibit the induction of CD8+ T-cell responses. The suppressive effect of IL-17A on the induction of CD8+ T-cell responses was abolished in mice depleted of neutrophils, clearly demonstrating the role played by these cells in the inhibitory effect of IL-17A in the induction of antitumor responses. These results demonstrate that even though strong Th1-type responses favor tumor control, the simultaneous activation of Th17 cells may redirect or curtail tumor-specific immunity through a mechanism involving neutrophils. This study establishes that IL-17 plays a detrimental role in the development of an effective antitumor T cell response and thus could strongly affect the efficiency of immunotherapy through the inhibition of CTL responses.


Subject(s)
CD8-Positive T-Lymphocytes , Cancer Vaccines , Interleukin-17 , Neoplasms , Adjuvants, Immunologic , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/pharmacology , Female , Interleukin-17/pharmacology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Th1 Cells/immunology
2.
Nat Commun ; 9(1): 2241, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29884826

ABSTRACT

Cross-presentation allows exogenous antigen presentation in association with major histocompatibility complex class I molecules, a process crucial for the priming of CD8+ T-cell responses against viruses and tumors. By contrast to conventional dendritic cells (cDC), which cross-present antigens in the steady state, plasmacytoid dendritic cells (pDC) acquire this ability only after stimulation by Toll-like receptor (TLR) ligands. The intracellular pathways accounting for this functional difference are still unknown. Here we show that the induction of cross-presentation by pDCs is regulated by mitochondria through a reactive oxygen species (ROS)-dependent mechanism, involving pH alkalization and antigen protection. The reduction of mitochondrial ROS production dramatically decreases the cross-presentation capacity of pDCs, leading to a strong reduction of their capacity to trigger CD8+ T-cell responses. Our results demonstrate the importance of mitochondrial metabolism in pDC biology, particularly for the induction of adaptive immune responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Mitochondria/immunology , Reactive Oxygen Species/immunology , Animals , Antigen Presentation/immunology , Antigens/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cross-Priming/immunology , Dendritic Cells/metabolism , Histocompatibility Antigens Class I/immunology , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
3.
Cancer Res ; 78(11): 3014-3026, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29523540

ABSTRACT

A growing number of observations has suggested that plasmacytoid dendritic cells (pDC) play a critical role in tumor biology. In patients, infiltration of tumors by pDCs generally correlates with a poor prognosis, suggesting that pDCs may play an important role in the host-tumor relationship. Here, we analyze the influence of pDCs in solid tumor development using two different tumor models: TC-1 and B16-OVA. Phenotypic and functional gene profiling analysis of tumor-associated pDCs showed that the tumor microenvironment affected their activation status and ability to produce cytokines and chemokines. In addition, tumor cells secreted factors that inhibit the ability of pDCs to produce type I IFN. Among the various cytokines and chemokines produced by the tumor cells, we demonstrate that TGFß is the main factor responsible for this inhibition. Using a mouse model deficient for pDCs, we also show that pDCs promote TC-1 tumor growth and that natural killer (NK) cells and regulatory T cells are involved in the protumoral effect of pDCs. Overall, our results evidence the cross-talk among pDCs, NK, and regulatory T cells in the promotion of tumor growth and their role in the development of antitumor immune responses.Significance: These findings highlight the importance of pDCs in the cross-talk between tumor cells and the immune system. Cancer Res; 78(11); 3014-26. ©2018 AACR.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate/immunology , Signal Transduction/immunology , Transforming Growth Factor beta/immunology , Animals , Chemokines/immunology , Disease Models, Animal , Female , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment
4.
Oncoimmunology ; 7(3): e1404213, 2018.
Article in English | MEDLINE | ID: mdl-29399403

ABSTRACT

Regulatory T-cells (Tregs) are crucial for the maintenance of immune tolerance and homeostasis as well as for preventing autoimmune diseases, but their impact on the survival of cancer patients remains controversial. In the TC-1 mouse model of human papillomavirus (HPV)-related carcinoma, we have previously demonstrated that the therapeutic efficacy of the CyaA-E7-vaccine, targeting the HPV-E7 antigen, progressively declines with tumor growth, in correlation with increased intratumoral recruitment of Tregs. In the present study, we demonstrated that these TC-1 tumor-infiltrating Tregs were highly activated, with increased expression of immunosuppressive molecules. Both intratumoral effector CD4+ T-cells (Teffs) and Tregs expressed high levels of PD-1, but anti-PD-1 antibody treatment did not impact the growth of the TC-1 tumor nor restore the therapeutic effect of the CyaA-E7 vaccine. To analyze the mechanisms by which Tregs are recruited to the tumor site, we used MHC-II KO mice with drastically reduced numbers of CD4+ effector T-cells. We demonstrated that these mice still had significant numbers of Tregs in their lymphoid organs which were recruited to the tumor. In MHC-II KO mice, the growth of the TC-1 tumor was delayed in correlation with a strong increase in the intratumoral recruitment of CD8+ T-cells. In addition, in mice that spontaneously rejected their tumors, the infiltration of E7-specific CD8+ T-cells was significantly higher than in MHC-II KO mice with a growing tumor. These results demonstrate that tumor-specific CD8+ T-cells can be efficiently activated and recruited in the absence of MHC class II molecules and of CD4+ T-cell help.

5.
PLoS Pathog ; 12(7): e1005770, 2016 07.
Article in English | MEDLINE | ID: mdl-27467705

ABSTRACT

Mycobacterium tuberculosis (Mtb), possesses at least three type VII secretion systems, ESX-1, -3 and -5 that are actively involved in pathogenesis and host-pathogen interaction. We recently showed that an attenuated Mtb vaccine candidate (Mtb Δppe25-pe19), which lacks the characteristic ESX-5-associated pe/ppe genes, but harbors all other components of the ESX-5 system, induces CD4+ T-cell immune responses against non-esx-5-associated PE/PPE protein homologs. These T cells strongly cross-recognize the missing esx-5-associated PE/PPE proteins. Here, we characterized the fine composition of the functional cross-reactive Th1 effector subsets specific to the shared PE/PPE epitopes in mice immunized with the Mtb Δppe25-pe19 vaccine candidate. We provide evidence that the Mtb Δppe25-pe19 strain, despite its significant attenuation, is comparable to the WT Mtb strain with regard to: (i) its antigenic repertoire related to the different ESX systems, (ii) the induced Th1 effector subset composition, (iii) the differentiation status of the Th1 cells induced, and (iv) its particular features at stimulating the innate immune response. Indeed, we found significant contribution of PE/PPE-specific Th1 effector cells in the protective immunity against pulmonary Mtb infection. These results offer detailed insights into the immune mechanisms underlying the remarkable protective efficacy of the live attenuated Mtb Δppe25-pe19 vaccine candidate, as well as the specific potential of PE/PPE proteins as protective immunogens.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cross Reactions , Disease Models, Animal , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Th1 Cells
6.
Cancer Res ; 75(16): 3279-91, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26122844

ABSTRACT

The metabolic sensor mTOR broadly regulates cell growth and division in cancer cells, leading to a significant focus on studies of rapamycin and its analogues as candidate anticancer drugs. However, mTOR inhibitors have failed to produce useful clinical efficacy, potentially because mTOR is also critical in T cells implicated in immunosurveillance. Indeed, recent studies using rapamycin have demonstrated the important role of mTOR in differentiation and induction of the CD8+ memory in T-cell responses associated with antitumor properties. In this study, we demonstrate that rapamycin harms antitumor immune responses mediated by T cells in the setting of cancer vaccine therapy. Specifically, we analyzed how rapamycin affects the antitumor efficacy of a human papilloma virus E7 peptide vaccine (CyaA-E7) capable of eradicating tumors in the TC-1 mouse model of cervical cancer. In animals vaccinated with CyaA-E7, rapamycin administration completely abolished recruitment of CD8+ T cells into TC-1 tumors along with the ability of the vaccine to reduce infiltration of T regulatory cells and myeloid-derived suppressor cells. Moreover, rapamycin completely abolished vaccine-induced cytotoxic T-cell responses and therapeutic activity. Taken together, our results demonstrate the powerful effects of mTOR inhibition in abolishing T-cell-mediated antitumor immune responses essential for the therapeutic efficacy of cancer vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Neoplasms/immunology , Sirolimus/immunology , Animals , Antibiotics, Antineoplastic/immunology , Antibiotics, Antineoplastic/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Female , Flow Cytometry , Humans , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/metabolism , Neoplasms/prevention & control , Papillomavirus E7 Proteins/immunology , Sirolimus/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects , Tumor Burden/immunology
7.
J Immunol ; 193(4): 1787-98, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25024388

ABSTRACT

Deciphering the mechanisms that allow the induction of strong immune responses is crucial to developing efficient vaccines against infectious diseases and cancer. Based on the discovery that the adenylate cyclase from Bordetella pertussis binds to the CD11b/CD18 integrin, we developed a highly efficient detoxified adenylate cyclase-based vector (CyaA) capable of delivering a large variety of Ags to the APC. This vector allows the induction of protective and therapeutic immunity against viral and tumoral challenges as well as against transplanted tumors in the absence of any added adjuvant. Two therapeutic vaccine candidates against human papilloma viruses and melanoma have been developed recently, based on the CyaA vector, and are currently in clinical trials. We took advantage of one of these highly purified vaccines, produced under good manufacturing practice-like conditions, to decipher the mechanisms by which CyaA induces immune responses. In this study, we demonstrate that CyaA binds both human and mouse CD11b(+) dendritic cells (DCs) and induces their maturation, as shown by the upregulation of costimulatory and MHC molecules and the production of proinflammatory cytokines. Importantly, we show that DCs sense CyaA through the TLR4/Toll/IL-1R domain-containing adapter-inducing IFN-ß pathway, independent of the presence of LPS. These findings show that CyaA possesses the intrinsic ability to not only target DCs but also to activate them, leading to the induction of strong immune responses. Overall, this study demonstrates that Ag delivery to CD11b(+) DCs in association with TLR4/Toll/IL-1R domain-containing adapter-inducing IFN-ß activation is an efficient strategy to promote strong specific CD8(+) T cell responses.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Adenylate Cyclase Toxin/immunology , CD11b Antigen/immunology , Dendritic Cells/immunology , Animals , B7-1 Antigen/biosynthesis , B7-2 Antigen/biosynthesis , Bordetella pertussis/immunology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/cytology , Female , Interferon-beta/immunology , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Receptors, Interleukin-1/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tyrosine/genetics
8.
PLoS Pathog ; 8(4): e1002580, 2012.
Article in English | MEDLINE | ID: mdl-22496638

ABSTRACT

Bordetella adenylate cyclase toxin-hemolysin (CyaA) penetrates the cytoplasmic membrane of phagocytes and employs two distinct conformers to exert its multiple activities. One conformer forms cation-selective pores that permeabilize phagocyte membrane for efflux of cytosolic potassium. The other conformer conducts extracellular calcium ions across cytoplasmic membrane of cells, relocates into lipid rafts, translocates the adenylate cyclase enzyme (AC) domain into cells and converts cytosolic ATP to cAMP. We show that the calcium-conducting activity of CyaA controls the path and kinetics of endocytic removal of toxin pores from phagocyte membrane. The enzymatically inactive but calcium-conducting CyaA-AC⁻ toxoid was endocytosed via a clathrin-dependent pathway. In contrast, a doubly mutated (E570K+E581P) toxoid, unable to conduct Ca²âº into cells, was rapidly internalized by membrane macropinocytosis, unless rescued by Ca²âº influx promoted in trans by ionomycin or intact toxoid. Moreover, a fully pore-forming CyaA-ΔAC hemolysin failed to permeabilize phagocytes, unless endocytic removal of its pores from cell membrane was decelerated through Ca²âº influx promoted by molecules locked in a Ca²âº-conducting conformation by the 3D1 antibody. Inhibition of endocytosis also enabled the native B. pertussis-produced CyaA to induce lysis of J774A.1 macrophages at concentrations starting from 100 ng/ml. Hence, by mediating calcium influx into cells, the translocating conformer of CyaA controls the removal of bystander toxin pores from phagocyte membrane. This triggers a positive feedback loop of exacerbated cell permeabilization, where the efflux of cellular potassium yields further decreased toxin pore removal from cell membrane and this further enhances cell permeabilization and potassium efflux.


Subject(s)
Adenylate Cyclase Toxin/pharmacology , Cell Membrane Permeability/drug effects , Macrophages/metabolism , Membrane Microdomains/metabolism , Potassium/metabolism , Animals , Cell Line , Clathrin/metabolism , Endocytosis/drug effects , Ion Transport/drug effects , Macrophages/cytology , Mice
9.
Vaccine ; 30(18): 2848-58, 2012 Apr 16.
Article in English | MEDLINE | ID: mdl-22387222

ABSTRACT

The complement system and Toll-like receptors (TLR) are key innate defense systems which might interact synergistically on dendritic cells (DC) to reinforce adaptive immunity. In a previous work, we found that the extra domain A from fibronectin EDA (an endogenous ligand for TLR4) can favour antigen delivery to DC and induce their maturation. Given the potential of anaphylatoxins to cause inflammation and activation of myeloid cells, we hypothesized that a fusion protein between EDA, and anaphylatoxins C3a, C4a or C5a together with an antigen might improve the immunogenicity of the antigen. Naked DNA immunization with a construct expressing the fusion protein between C5a, EDA and the cytotoxic T cell epitope SIINFEKL from ovalbumin, induced strong antigen specific T cell responses. The purified recombinant fusion protein EDA-SIINFEKL-C5a induced activation of dendritic cells, the production of proinflammatory cytokines/chemokines and stimulated antigen presenting cell migration and NK cell activation. As compared to EDA-SIINFEKL, the fusion protein EDA-SIINFEKL-C5a did not induce the production of the immunosuppressive molecules IL-10, CCL17, CCL1, CXCL12 or XCL1 by DC. Moreover, EDA-SIINFEKL-C5a induced strong specific T cell responses in vivo and protected mice against E.G7-OVA tumor growth more efficiently than EDA-SIINFEKL or SIINFEKL-C5a recombinant proteins. Our results suggest that fusion proteins containing EDA, the anaphylatoxin C5a and the antigen may serve as a suitable strategy for the development of anti-tumor or anti-viral vaccines.


Subject(s)
Anaphylatoxins/immunology , Complement C5a/immunology , Ectodysplasins/immunology , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 4/agonists , Anaphylatoxins/genetics , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Complement C5a/genetics , Cytokines/metabolism , Ectodysplasins/genetics , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/prevention & control , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
10.
Vaccine ; 28(44): 7146-54, 2010 Oct 18.
Article in English | MEDLINE | ID: mdl-20817012

ABSTRACT

Staphylococcus epidermidis releases a complex of at least four peptides, termed phenol-soluble modulins (PSM), which stimulate macrophages to produce proinflammatory cytokines via activation of TLR2 signalling pathway. We demonstrated that covalent linkage of PSM peptides to an antigen facilitate its capture by dendritic cells and, in combination with different TLR ligands, can favour the in vivo induction of strong and persistent antigen-specific immune responses. Treatment of mice grafted with HPV16-E7-expressing tumor cells (TC-1) with poly(I:C) and a peptide containing αMod linked to the H-2D(b)-restricted cytotoxic T-cell epitope E7(49-57) from HPV16-E7 protein allowed complete tumor regression in 100% of the animals. Surprisingly, this immunomodulatory property of modulin-derived peptides was TLR2 independent and partially dependent upon the EGF-receptor signalling pathway. Our results suggest that alpha or gamma modulin peptides may serve as a suitable antigen carrier for the development of anti-tumoral or anti-viral vaccines.


Subject(s)
Antigens, Neoplasm/immunology , Bacterial Toxins/immunology , Neoplasms, Experimental/drug therapy , Papillomavirus E7 Proteins/immunology , Animals , Antigen Presentation , Antigens, Neoplasm/administration & dosage , Bacterial Toxins/administration & dosage , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , ErbB Receptors/metabolism , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/prevention & control , Papillomavirus E7 Proteins/administration & dosage , Poly I-C/immunology , Staphylococcus epidermidis , Toll-Like Receptor 2/immunology
11.
Vaccine ; 28(42): 6930-41, 2010 Oct 04.
Article in English | MEDLINE | ID: mdl-20728521

ABSTRACT

HIV-Tat based vaccines have been proposed as an attractive option to prevent or treat AIDS. A vaccine to induce optimal anti-Tat neutralizing antibody responses was designed by inserting this protein, or its dominant B-cell epitope, into the CyaA vector, which targets dendritic cells (DC). Tat was inserted into various sites of CyaA, including regions that do not translocate into the cytosol of the targeted DC. The presentation of the Tat CD4(+) T-cell epitope delivered by the CyaA-Tat proteins was observed with a recombinant CyaA in which the entire AC domain was replaced by the entire Tat protein (Tat-Δ373 CyaA) but was abolished with large deletions of the N-terminal region. Moreover, CyaA carrying multiple copies of the dominant Tat: 1-21 B-cell epitope were shown to induce high titers of anti-Tat antibodies, even after a single immunization, that persisted up to 10 weeks post-immunization.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Neutralizing/blood , Dendritic Cells/immunology , Epitopes, B-Lymphocyte/immunology , tat Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/genetics , Adenylate Cyclase Toxin/immunology , Animals , Antigen Presentation , CHO Cells , CpG Islands , Cricetinae , Cricetulus , Cytokines/immunology , Epitopes, T-Lymphocyte/immunology , Female , HIV Antibodies/blood , Immunity, Cellular , Mice , Mice, Inbred BALB C , Neutralization Tests , Poly I-C/immunology , Recombinant Proteins/immunology , tat Gene Products, Human Immunodeficiency Virus/genetics
12.
Mol Microbiol ; 75(6): 1550-62, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20199594

ABSTRACT

The adenylate cyclase toxin-haemolysin of Bordetella (CyaA) targets CD11b(+) myeloid phagocytes and translocates across their cytoplasmic membrane an adenylate cyclase (AC) enzyme that catalyses conversion of cytosolic ATP into cAMP. In parallel, CyaA acts as a cytolysin forming cation-selective pores, which permeabilize cell membrane and eventually provoke cell lysis. Using cytolytic activity, potassium efflux and patch-clamp assays, we show that a combination of substitutions within the pore-forming (E570Q) and acylation-bearing domain (K860R) ablates selectively the cell-permeabilizing activity of CyaA. At the same time, however, the capacity of such mutant CyaA to translocate the AC domain across cytoplasmic membrane into cytosol of macrophage cells and to elevate cellular cAMP concentrations remained intact. Moreover, the combination of E570Q+K860R substitutions suppressed the residual cytolytic activity of the enzymatically inactive CyaA/OVA/AC(-) toxoid on CD11b-expressing monocytes, while leaving unaffected the capacity of the mutant toxoid to deliver in vitro a reporter CD8(+) T cell epitope from ovalbumin (OVA) to the cytosolic pathway of dendritic cells for MHC class I-restricted presentation and induce in vivo an OVA-specific cytotoxic T cell response. CyaA, hence, employs a mechanism of AC enzyme domain translocation across cellular membrane that avoids passage across the cytolytic pore formed by toxin oligomers.


Subject(s)
Adenylate Cyclase Toxin/metabolism , Cell Membrane/metabolism , Adenylate Cyclase Toxin/genetics , Amino Acid Substitution , Animals , Cells, Cultured , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , Macrophages/immunology , Mice , Models, Biological , Monocytes/immunology , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Ovalbumin/immunology , Protein Transport , T-Lymphocytes, Cytotoxic/immunology
13.
Vaccine ; 24(17): 3490-9, 2006 Apr 24.
Article in English | MEDLINE | ID: mdl-16524647

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) Tat is a key protein playing a major role in the infectivity of the virus. Thus, HIV-Tat based vaccines have been proposed as an attractive option to treat AIDS. Recently, we have shown that the recombinant detoxified adenylate cyclase (CyaA) from Bordetella pertussis carrying HIV-Tat (CyaA-E5-Tat), targets dendritic cells (DCs) and induces specific Th1 polarized and neutralizing antibody responses in mice. To further explore the potentialities of this prototype vaccine for human use, we analyzed the CyaA-E5-Tat induced antibody responses in non-human primates and established the biological characteristics of these antibodies. African Green Monkeys (AGM) were immunized with CyaA-E5-Tat in the presence or in the absence of alum adjuvant. First, we showed that the anti-CyaA antibodies induced by such immunization does not interfere with the binding of CyaA-E5-Tat to its receptor at the DC surface, the alphaMbeta2 integrin. Monkeys immunized with CyaA-E5-Tat, with or without alum, produced anti-Tat antibodies that mainly recognized the N-terminal domain of the Tat protein. Importantly, all sera obtained after three immunizations displayed the capacity to bind to Tat and neutralize its transactivating function in vitro. Finally, in the absence of alum, CyaA-E5-Tat, induced Th1 Tat specific T cell responses. These findings reveal that CyaA-E5-Tat is efficiently delivered in non-human primates and had a significant impact on the generation of neutralizing anti-Tat antibodies. These observations are, thus, encouraging for the use of the CyaA vector in human and also suggest that CyaA-E5-Tat might be a useful tool to decipher the biological characteristic of such antibodies.


Subject(s)
Adenylyl Cyclases/administration & dosage , Dendritic Cells/immunology , Gene Products, tat/immunology , HIV Antibodies/biosynthesis , HIV-1/immunology , Th1 Cells/immunology , AIDS Vaccines/immunology , Animals , CD11b Antigen/metabolism , Cell Polarity , Chlorocebus aethiops , Dendritic Cells/metabolism , Female , Humans , Interferon-gamma/biosynthesis , Male , tat Gene Products, Human Immunodeficiency Virus
14.
J Virol ; 79(15): 9872-84, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16014948

ABSTRACT

HIV-Tat, a conserved protein playing a key role in the early life cycle of the human immunodeficiency virus (HIV) has been proposed as a potential AIDS vaccine. An HIV-Tat-based vaccine should elicit a broad, long-lasting, and neutralizing immune response. We have previously demonstrated that the adenylate cyclase (CyaA) from Bordetella pertussis targets dendritic cells and delivers CD8(+) and CD4(+) T-cell epitopes into the major histocompatibility complex class I and class II presentation pathways. We have also showed that CyaA induced specific and protective cytotoxic T cell responses in vivo. Here, we designed a prototype vaccine based on the HIV type 1 Tat delivered by CyaA (CyaA-E5-Tat) and tested its capacity to induce HIV-Tat-specific cellular as well as antibody responses. We showed that immunization of mice by CyaA-E5-Tat in the absence of adjuvant elicited strong and long-lasting neutralizing anti-Tat antibody responses more efficient than those obtained after immunization with Tat toxoid in aluminum hydroxide adjuvant. Analyses of the anti-Tat immunoglobulin G isotypes and the cytokine pattern showed that CyaA-E5-Tat induced a Th1-polarized immune response in contrast to the Th2-polarized immune responses obtained with the Tat toxoid. In addition, our data demonstrated that HIV-Tat-specific gamma interferon-producing CD8(+) T cells were generated after vaccination with CyaA-E5-Tat in a CD4(+) T-cell-independent manner. Based on these findings, CyaA-E5-Tat represents an attractive vaccine candidate for both preventive and therapeutic vaccination involving CyaA as an efficient nonreplicative vector for protein delivery.


Subject(s)
AIDS Vaccines/immunology , Adenylate Cyclase Toxin/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Products, tat/immunology , HIV Antibodies/blood , HIV Infections/immunology , Immunization , Adenylate Cyclase Toxin/metabolism , Animals , Drug Evaluation, Preclinical , Female , HIV Infections/blood , Interferon-gamma/biosynthesis , Lymphocyte Count , Mice , Mice, Inbred BALB C , Neutralization Tests , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Spleen/immunology , T-Cell Antigen Receptor Specificity , Th1 Cells/immunology , Vaccines, Synthetic/immunology , tat Gene Products, Human Immunodeficiency Virus
15.
Vaccine ; 23(5): 604-14, 2004 Dec 16.
Article in English | MEDLINE | ID: mdl-15542180

ABSTRACT

The adenylate cyclase (CyaA) produced by Bordetella pertussis is able to deliver CD8+ and CD4+ T-cell epitopes genetically grafted within the catalytic domain of the molecule into antigen presenting cells in vivo. We develop now a new approach in which peptides containing CD8+ epitopes are chemically linked to CyaA. We show that CTL responses were induced in mice immunized with CyaA bearing these CD8+ epitopes. Moreover, we demonstrate that the OVA257-264 CD8+ epitope chemically grafted to CyaA is presented to CD8+ T cells by a mechanism requiring (1) proteasome processing, (2) TAP and (3) neosynthesis of MHC class I molecules. Thus, this novel strategy represents a very versatile system as a single CyaA carrier protein could be easily and rapidly coupled to any desired synthetic peptide.


Subject(s)
Adenylate Cyclase Toxin/immunology , Bacterial Vaccines/immunology , Bordetella pertussis/immunology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , T-Lymphocytes, Cytotoxic/immunology , Adenylate Cyclase Toxin/metabolism , Animals , Antigen Presentation/drug effects , Antigens, Ly/metabolism , Bacterial Vaccines/chemistry , Bacterial Vaccines/therapeutic use , Bordetella pertussis/enzymology , Brefeldin A/pharmacology , CD8 Antigens/immunology , Catalytic Domain , Dendritic Cells/metabolism , Epitopes, T-Lymphocyte/genetics , Female , Golgi Apparatus/metabolism , Histocompatibility Antigens Class I/biosynthesis , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL
16.
Int Immunol ; 15(10): 1161-71, 2003 Oct.
Article in English | MEDLINE | ID: mdl-13679386

ABSTRACT

A number of studies in animal models and humans have shown that both humoral andcell-mediated immune responses play an important role in the control of viral infection and tumor development. In most cases, vaccination with non-vectorized peptides or proteins induces low antibody responses and fails to elicit specific cytotoxic T lymphocytes (CTL). In order to make vaccination more efficient, we chemically coupled the non-toxic B subunit of Shiga toxin (STxB) to a full-size antigenic model protein, ovalbumin (OVA), yielding STxB-OVA. We found that STxB-OVA delivers OVA-derived peptides into both the MHC class I- and II-restricted presentation pathways in mouse dendritic cells. Accordingly, the study of STxB trafficking in these cells revealed that, after internalization, a fraction of STxB followed the retrograde transport pathway to the endoplasmic reticulum, while another fraction was targeted to late endosomes/lysosomes. Vaccination of mice with STxB-OVA primed a specific anti-OVA CTL response without the use of adjuvants. Splenocytes and, particularly, CD4(+) T cells from mice immunized with STxB-OVA also produced higher amounts of the T(h)1 cytokine IFN-gamma and IgG2a-type antibodies than mice immunized with non-vectorized ovalbumin. In conclusion, this study identifies a unique non-live vaccine delivery system for polyepitopic antigens that elicits antigen-specific CTL, a humoral immune response and a T(h)1-type polarization without the use of adjuvant.


Subject(s)
Antibody Formation , Ovalbumin/immunology , Shiga Toxins/genetics , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Animals , Antigens/immunology , Cysteine Endopeptidases/physiology , Female , Genes, MHC Class I/genetics , Genes, MHC Class II/genetics , Genes, MHC Class II/immunology , Mice , Multienzyme Complexes/physiology , Ovalbumin/genetics , Ovalbumin/metabolism , Proteasome Endopeptidase Complex , Protein Processing, Post-Translational , Receptors, Cell Surface/physiology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Shiga Toxin/immunology , Shiga Toxin/metabolism , Signal Transduction
17.
Eur J Immunol ; 32(11): 3071-81, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12385027

ABSTRACT

The precise contribution of mouse dendritic cells (DC) CD8 alpha +CD11blow and CD8 alpha -CD11bhigh subsets to CTL priming is not fully defined. Here we show that CyaA, the adenylate cyclase toxin of Bordetella pertussis, an invasive bacterial toxin that binds cells through CD11b/CD18 can be exploited for the targeted delivery of an exogenous peptide to the CD8 alpha -CD11bhigh subset in vivo. Antigen (Ag) genetically inserted in the N-terminal domain of mutant CyaA devoid of catalytic activity, are targeted to CD8 alpha -CD11bhigh DC by the CD11b/CD18-dependent binding of CyaA to the cell surface. Ag is then presented by MHC class I molecules of CD8 alpha -CD11bhigh DC after a TAP-dependent, cytosolic processing. As a result, CTL are primed after a single injection, bypassing requirement for adjuvant, CD4+ T cell help and CD40 signaling. Beside the interest of the CyaA vector for vaccine development, these results show that Ag presentation focused on CD8 alpha -CD11bhigh DC in vivo is sufficient for eliciting a vigorous CTL response and that CD11b/CD18 could be a suitable surface molecule for targeting Ag to DC.


Subject(s)
Adenylate Cyclase Toxin/metabolism , Antigen Presentation , Dendritic Cells/metabolism , Toxoids/metabolism , Adenylate Cyclase Toxin/immunology , Animals , CD11b Antigen/analysis , CD11c Antigen/analysis , CD8 Antigens/analysis , Cell Line , Cytosol/metabolism , Female , Immunization , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Recombinant Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology
18.
Vaccine ; 20(19-20): 2463-73, 2002 Jun 07.
Article in English | MEDLINE | ID: mdl-12057601

ABSTRACT

Epitope-based vaccination strategies designed to induce strong tumor-specific CD8(+) T cell responses are being widely considered for cancer immunotherapy. Here, two recombinant tubular structures, NS1-Mela 1 and NS1-Mela 2, carrying, respectively two HLA-A2 epitopes derived from human melanoma antigens were constructed and their capability to induce CTL responses in vivo were studied in HLA-A2 transgenic mice. Strong CTL responses specific for GnT-V/NA 17-A and gp100 (154-162) epitopes were generated in HLA-A2 transgenic mice immunized by the construct NS1-Mela l carrying these two epitopes. The second construct NS1-Mela 2 carrying both Tyrosinase (369-377Da) and Melan-A/Mart-1 (27-35) epitopes induced a weak Tyrosinase-specific CTL response in mice but failed to induce specific CTL responses against the Melan-A/Mart-1 (27-35) epitope in the tested mice. Thus, recombinant tubular structures containing multiple tumoral epitopes may lead to new strategies for the induction of strong tumor-specific CTL responses in cancer patients.


Subject(s)
Epitopes/immunology , HLA-A2 Antigen/immunology , Melanoma/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Baculoviridae/genetics , Base Sequence , Blotting, Western , Cancer Vaccines/immunology , DNA Primers , Enzyme-Linked Immunosorbent Assay , Humans , Interferon-gamma/biosynthesis , Mice , Mice, Knockout , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...