ABSTRACT
Paracoccidioidomycosis (PCM) is a systemic mycosis caused by Paracoccidioides brasiliensis, a thermally dimorphic fungus, which is the most frequent endemic systemic mycosis in many Latin American countries, where ~10 million people are believed to be infected. In Brazil, it is ranked as the tenth most common cause of death among chronic infectious diseases. Hence, vaccines are in development to combat this insidious pathogen. It is likely that effective vaccines will need to elicit strong T cell-mediated immune responses composed of IFNγ secreting CD4+ helper and CD8+ cytolytic T lymphocytes. To induce such responses, it would be valuable to harness the dendritic cell (DC) system of antigen-presenting cells. To assess the potential of targeting P10, which is a peptide derived from gp43 secreted by the fungus, directly to DCs, we cloned the P10 sequence in fusion with a monoclonal antibody to the DEC205 receptor, an endocytic receptor that is abundant on DCs in lymphoid tissues. We verified that a single injection of the αDEC/P10 antibody caused DCs to produce a large amount of IFNγ. Administration of the chimeric antibody to mice resulted in a significant increase in the levels of IFN-γ and IL-4 in lung tissue relative to control animals. In therapeutic assays, mice pretreated with αDEC/P10 had significantly lower fungal burdens compared to control infected mice, and the architecture of the pulmonary tissues of αDEC/P10 chimera-treated mice was largely normal. Altogether, the results obtained so far indicate that targeting P10 through a αDEC/P10 chimeric antibody in the presence of polyriboinosinic: polyribocytidylic acid is a promising strategy in vaccination and therapeutic protocols to combat PCM.
ABSTRACT
Dendritic cell (DC) targeting by DEC205+ cells effectively promotes the internalization of antigens that may trigger a specific immune response. In this study, we evaluated the ability of a recombinant antibody, anti-DEC205 (rAb ZH9F7), to trigger cellular endocytosis in subpopulations of DCs and targeted cells after intradermal injection and subsequent migration toward lymph nodes. Furthermore, the cellular immune response was evaluated in pigs after intradermal application of the antigenized rAb ZH9F7 combined with porcine circovirus type 2 cap antigen (rAb ZH9F7-Cap). We demonstrated that rAb ZH9F7 recognized conventional type 1 and 2 DCs from the blood and skin and monocytes. It promoted receptor-mediated endocytosis and migration of cDCs and moDCs toward regional lymph nodes. Intradermal application of rAb ZH9F7-Cap induced a higher frequency of IFN-γ-secreting CD4+CD8+ T lymphocytes and antibodies against Cap protein than that in the control group. In conclusion, the rAb ZH9F7-Cap system promoted the target of skin cDC1 and cDC2, provoking migration to the regional lymph nodes and inducing a Th1 response, as evidenced by the proliferation of double-positive CD4+CD8+ T cells, which correlates with an enhanced ability to target the cDC1 subset both in vitro and in vivo.
ABSTRACT
The generation of successful anticancer vaccines relies on the ability to induce efficient and long-lasting immune responses to tumor antigens. In this scenario, dendritic cells (DCs) are essential cellular components in the generation of antitumor immune responses. Thus, delivery of tumor antigens to specific DC populations represents a promising approach to enhance the efficiency of antitumor immunotherapies. In the present study, we employed antibody-antigen conjugates targeting a specific DC C-type lectin receptor. For that purpose, we genetically fused the anti-DEC205 monoclonal antibody to the type 16 human papillomavirus (HPV-16) E7 oncoprotein to create a therapeutic vaccine to treat HPV-associated tumors in syngeneic mouse tumor models. The therapeutic efficacy of the αDEC205-E7 mAb was investigated in three distinct anatomical tumor models (subcutaneous, lingual and intravaginal). The immunization regimen comprised two doses of the αDEC205-E7 mAb coadministered with a DC maturation stimulus (Polyinosinic:polycytidylic acid, poly (I:C)) as an adjuvant. The combined immunotherapy produced robust antitumor effects on both the subcutaneous and orthotopic tumor models, stimulating rapid tumor regression and long-term survival. These outcomes were related to the activation of tumor antigen-specific CD8+ T cells in both systemic compartments and lymphoid tissues. The αDEC205-E7 antibody plus poly (I:C) administration induced long-lasting immunity and controlled tumor relapses. Our results highlight that the delivery of HPV tumor antigens to DCs, particularly via the DEC205 surface receptor, is a promising therapeutic approach, providing new opportunities for the development of alternative immunotherapies for patients with HPV-associated tumors at different anatomical sites.
Subject(s)
Antigens, CD/immunology , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Lectins, C-Type/immunology , Minor Histocompatibility Antigens/immunology , Neoplasms, Experimental/prevention & control , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/prevention & control , Receptors, Cell Surface/immunology , Adjuvants, Immunologic , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Female , Humans , Immunologic Memory , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/virology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Poly I-C/administration & dosageABSTRACT
Rotavirus is the most common cause of severe diarrhea in infants and children worldwide and is responsible for about 215,000 deaths annually. Over 85% of these deaths originate in low-income/developing countries in Asia and Africa. Therefore, it is necessary to explore the development of vaccines that avoid the use of "living" viruses and furthermore, vaccines that have viral antigens capable of generating powerful heterotypic responses. Our strategy is based on the expression of the fusion of the anti-DEC205 single-chain variable fragment (scFv) coupled by an OLLAS tag to a viral protein (VP6) of Rotavirus in Nicotiana plants. It was possible to express transiently in N. benthamiana and N. sylvestris a recombinant protein consisting of the single chain variable fragment linked by an OLLAS tag to the VP6 protein. The presence of the recombinant protein, which had a molecular weight of approximately 75 kDa, was confirmed by immunodetection, in both plant species and in both cellular compartments (cytoplasm and apoplast) where it was expressed. In addition, the recombinant protein was modeled, and it was observed that some epitopes of interest are exposed on the surface, which could favor their immunogenic response.
Subject(s)
Antigens, Viral/genetics , Capsid Proteins/genetics , Nicotiana/growth & development , Rotavirus/metabolism , Single-Chain Antibodies/genetics , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antigens, Viral/chemistry , Antigens, Viral/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Models, Molecular , Molecular Weight , Protein Engineering , Protein Structure, Secondary , Recombinant Proteins/metabolism , Rotavirus/genetics , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism , Nicotiana/genetics , Nicotiana/metabolismABSTRACT
HPV E5 is an oncoprotein mainly expressed in premalignant lesions, which makes it an important target for a vaccine to prevent or cure cervical cancer (CC). In this study, we evaluated whether E5 targeted to DEC-205, present in dendritic cells (DCs), could induce a therapeutic protection against HPV16-induced tumor cells in a mouse model. The HPV-16 E5 (16E5) protein was cross-linked to a monoclonal antibody (mAb) specific to mouse DEC-205 (anti-DEC-205:16E5) or to an isotype control mAb (isotype:16E5). Rotavirus VP6 was cross-linked to the mouse anti-DEC-205 mAb (anti-DEC-205:VP6) as a non-specific antigen control. BALB/c mice were inoculated subcutaneously (s.c.) with the 16E5-expressing BMK-16/myc tumor cells, and 7 and 14 days later the mice were immunized s.c. with the conjugates, free 16E5 or PBS in the presence of adjuvant. Tumor growth was monitored to evaluate protection. A strong protective immune response against the tumor cells was induced when the mice were inoculated with the anti-DEC-205:16E5 conjugate, since 70% of the mice controlled the tumor growth and survived, whereas the remaining 30% developed tumors and died by day 72. In contrast, 100% of the mice in the control groups died by day 30. The anti-DEC-205:16E5 conjugate was found to induce 16E5-specific memory T cells, with a Th1/Th17 profile. Both CD4+ and CD8+ T cells contributed to the observed protection. Finally, treating mice that had developed tumors with an anti-PD-1 mAb, delayed the tumor growth for more than 20 days. These results show that targeting 16E5 to DEC-205, alone or combined with an immune checkpoint blockade, could be a promising protocol for the treatment of the early stages of HPV-associated cancer.
Subject(s)
Dendritic Cells/immunology , Human papillomavirus 16/immunology , Neoplasms/etiology , Neoplasms/therapy , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/complications , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Biomarkers, Tumor , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Immunization , Immunologic Memory , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Neoplasms/diagnosis , Papillomavirus Infections/virology , T-Lymphocytes/immunology , T-Lymphocytes/metabolismABSTRACT
Recombinant hybrid antibodies are commonly used in antigen-targeting assays to reduce the immunogenic potential associated with using classic mouse antibodies in other species. The DEC205 receptor has become an attractive target due to its effectiveness in activating the immune response and is considered a promising vaccination target. The aim of this study was to produce a fully chimeric mouse x pig anti-porcine DEC205 recombinant antibody (rAb). Based on a mouse anti-porcine DEC205 monoclonal antibody (mAb), we designed and expressed a chimeric mouse x pig rAb using the Expi293f system. The resulting rAb maintained the recognition capacity of the native mouse mAb toward the porcine DEC205 receptor, as evidenced by western blot analysis. By using flow cytometry, we evaluated the ability of the rAb to recognize DEC205+ dendritic cells. In conclusion, the chimeric mouse x pig anti-DEC205 rAb can be used in antigen-targeting assays as a vaccination strategy in pigs.
Subject(s)
Antibodies, Monoclonal/biosynthesis , Antigens, CD/immunology , Lectins, C-Type/immunology , Minor Histocompatibility Antigens/immunology , Receptors, Cell Surface/immunology , Animals , Antibodies, Monoclonal/immunology , Mice , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , SwineABSTRACT
Targeting dendritic cells (DCs) by means of monoclonal antibodies (mAbs) capable of binding their surface receptors (DEC205 and DCIR2) has previously been shown to enhance the immunogenicity of genetically fused antigens. This approach has been repeatedly demonstrated to enhance the induced immune responses to passenger antigens and thus represents a promising therapeutic and/or prophylactic strategy against different infectious diseases. Additionally, under experimental conditions, chimeric αDEC205 or αDCIR2 mAbs are usually administered via an intraperitoneal (i.p.) route, which is not reproducible in clinical settings. In this study, we characterized the delivery of chimeric αDEC205 or αDCIR2 mAbs via an intradermal (i.d.) route, compared the elicited humoral immune responses, and evaluated the safety of this potential immunization strategy under preclinical conditions. As a model antigen, we used type 2 dengue virus (DENV2) nonstructural protein 1 (NS1). The results show that the administration of chimeric DC-targeting mAbs via the i.d. route induced humoral immune responses to the passenger antigen equivalent or superior to those elicited by i.p. immunization with no toxic effects to the animals. Collectively, these results clearly indicate that i.d. administration of DC-targeting chimeric mAbs presents promising approaches for the development of subunit vaccines, particularly against DENV and other flaviviruses.
ABSTRACT
Conventional dendritic cells (cDCs) are specialized in antigen presentation. In the mouse spleen, cDCs are classified in cDC1s and cDC2s, and express DEC205 and DCIR2 endocytic receptors, respectively. Monoclonal antibodies (mAbs) αDEC205 (αDEC) and αDCIR2 have been fused to different antigens to deliver them to cDC1s or cDC2s. We immunized mice with αDEC and αDCIR2 fused to an antigen using Poly(I:C) as adjuvant. The initial immune response was analyzed from days 3 to 6 after the immunization. We also studied the influence of a booster dose. Our results showed that antigen targeting to cDC1s promoted a pro-inflammatory TH 1 cell response. Antigen targeting to cDC2s induced TFH cells, GCs, and plasma cell differentiation. After boost, antigen targeting to cDC1s improved the TH 1 cell response and induced TH 1-like TFH cells that led to an increase in specific antibody titers and IgG class switch. Additionally, a population of regulatory T cells was also observed. Antigen targeting to cDC2s did not improve the specific antibody response after boost. Our results add new information on the immune response induced after the administration of a booster dose with αDEC and αDCIR2 fusion mAbs. These results may be useful for vaccine design using recombinant mAbs.
Subject(s)
Dendritic Cells/immunology , Receptors, Cell Surface/immunology , T Follicular Helper Cells/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Formation/immunology , Antigen Presentation/immunology , Female , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/immunologyABSTRACT
Conventional dendritic cells (cDCs) cannot be infected by porcine reproductive and respiratory syndrome virus (PRRSV) but respond to infection via cytokine production, indicating a possible role in initiation/regulation of the immune response against PRRSV. In this work, we evaluated the responses of splenic and blood cDCs, with DEC205+CADM1+CD172a+/- phenotype, as well as those of CD163+ cells against PRRSV and porcine epidemic diarrhea virus (PEDV). Both populations were incubated in the presence of PRRSV or PEDV with and without naïve CD3+ T cells, and cytokine responses were evaluated by qPCR and ELISA. Our results showed that cDCs, but not CD163+ cells, produced IL-12 in response to PRRSV. PEDV did not induce IL-12 production. Cocultures of cDCs and autologous naïve CD3+ cells resulted in decreased IL-12 production and low expression of IFN-γ transcripts in response to PRRSV. Interestingly, cDCs increased the proliferation of naïve T cells in the presence of PRRSV compared with that achieved with monocytes and peripheral blood mononuclear cells (PBMCs). Cocultures of CD163+ cells induced IL-10 and IL-4 expression in the presence of PRRSV and PEDV, respectively. In conclusion, cDCs can selectively produce IL-12 in response to PRRSV but poorly participate in the activation of naïve T cells.
Subject(s)
Coronavirus Infections/veterinary , Dendritic Cells/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , T-Lymphocytes , Animals , Antigens, CD/blood , Antigens, Differentiation, Myelomonocytic/blood , Cell Adhesion Molecule-1/blood , Coronavirus Infections/immunology , Cytokines/blood , Dendritic Cells/virology , Interleukin-10/blood , Interleukin-12/blood , Interleukin-4/blood , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Monocytes/immunology , Monocytes/virology , Porcine epidemic diarrhea virus , Porcine respiratory and reproductive syndrome virus , Primary Cell Culture , Receptors, Cell Surface/blood , Spleen/cytology , Spleen/immunology , Spleen/virology , Swine , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , T-Lymphocytes/immunology , T-Lymphocytes/virologyABSTRACT
Activation of the immune system using antigen targeting to the dendritic cell receptor DEC205 presents great potential in the field of vaccination. The objective of this work was to evaluate the immunogenicity and protectiveness of a recombinant mouse x pig chimeric antibody fused with peptides of structural and nonstructural proteins of porcine respiratory and reproductive syndrome virus (PRRSV) directed to DEC205+ cells. Priming and booster immunizations were performed three weeks apart and administered intradermally in the neck area. All pigs were challenged with PRRSV two weeks after the booster immunization. Immunogenicity was evaluated by assessing the presence of antibodies anti-PRRSV, the response of IFN-γ-producing CD4+ cells, and the proliferation of cells. Protection was determined by assessing the viral load in the blood, lungs, and tonsils using qRT-PCR. The results showed that the vaccine exhibited immunogenicity but conferred limited protection. The vaccine group had a lower viral load in the tonsils and a significantly higher production of antibodies anti-PRRSV than the control group (p < 0.05); the vaccine group also produced more CD4+IFN-γ+ cells in response to peptides from the M and Nsp2 proteins. In conclusion, this antigenized recombinant mouse x pig chimeric antibody had immunogenic properties that could be enhanced to improve the level of protection and vaccine efficiency.
ABSTRACT
Cellular immune responses are implicated in resistance to HIV and have been considered for the development of an effective vaccine. Despite their safety profile, subunit vaccines need to be delivered combined with an adjuvant. In the last years, in vivo antigen targeting to dendritic cells (DCs) using chimeric monoclonal antibodies (mAb) against the DC endocytic receptor DEC205/CD205 was shown to support long-term T cell immunity. Here, we evaluated the ability of different adjuvants to modulate specific cellular immune response when eight CD4+ HIV-derived epitopes (HIVBr8) were targeted to DEC205+ DCs in vivo. Immunization with two doses of αDECHIVBr8 mAb along with poly(I:C) induced Th1 cytokine production and higher frequency of HIV-specific polyfunctional and long-lived T cells than MPL or CpG ODN-assisted immunization. Although each adjuvant elicited responses against the 8 epitopes present in the vaccine, the magnitude of the T cell response was higher in the presence of poly(I:C). Moreover, poly(I:C) up regulated the expression of costimulatory molecules in both cDC1 and cDC2 DCs subsets. In summary, the use of poly(I:C) in a vaccine formulation that targets multiple epitopes to the DEC205 receptor improved the potency and the quality of HIV-specific responses when compared to other vaccine-adjuvant formulations. This study highlights the importance of the rational selection of antigen/adjuvant combination to potentiate the desired immune responses.
Subject(s)
AIDS Vaccines/immunology , Dendritic Cells/immunology , Epitopes/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV/immunology , Poly I-C , T-Lymphocytes/immunology , Adjuvants, Immunologic , Animals , Antigens, CD/metabolism , Biomarkers , Cytokines/metabolism , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Immunity, Cellular , Immunophenotyping , Lymphocyte Activation , Mice , Poly I-C/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolismABSTRACT
The ectodomain of the influenza A virus (IAV) M2 protein (M2e) is highly conserved, and it represents a promising candidate for the development of an "universal vaccine". However, the low immunogenicity associated to M2e in a natural infection or in response to seasonal vaccines has led to explore new approaches to enhance it. In recent years, it has become clear that targeting antigens to dendritic cells (DC) is an efficient way to enhance immune responses against pathogens. In this work, the M2e peptide was chemically cross-linked to a monoclonal antibody (mAb) specific for DEC-205 (α-DEC-205:M2e), present on DC. BALB/c mice were inoculated subcutaneously (s.c.) three times with the conjugate equivalent to 1⯵g of M2e, in the presence of polyinosinic-polycytidylic acid (poly I:C) as adjuvant. As controls, other groups of mice were inoculated under the same conditions with M2e cross-linked to an isotype control mAb (isotype:M2e), 5⯵g of free M2e peptide, ovalbumin (OVA) cross-linked to the α-DEC-205 mAb (α-DEC-205:OVA) or poly I:C alone. Immunization with α-DEC-205-M2e induced high levels of serum antibodies (Abs) compared to isotype:M2e or to free M2e peptide, and in all cases IgG1 was predominant over IgG2a Abs. Furthermore, immunization with the α-DEC-205:M2e conjugate did not prevent morbidity, but it induced up to 76% protection against a heterosubtypic IAV lethal challenge. Contrasting with the 20 to 40% protection induced by isotype:M2e or by free M2e peptide. The protection induced by α-DEC-205:M2e conjugate was dependent on non-neutralizing serum Abs and independent of effector CD4+ T cells. These results show that targeting M2e to DEC-205 is a very effective alternative to induce strong heterosubtypic protection against an IAV infection.
Subject(s)
Antibodies, Viral/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Lectins, C-Type/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Adjuvants, Immunologic , Animals , Antibody Specificity/immunology , Antigens, CD , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Female , Humans , Immunogenicity, Vaccine , Influenza, Human/metabolism , Mice , Minor Histocompatibility Antigens , VaccinationABSTRACT
Porcine reproductive and respiratory syndrome virus (PRRSV) infects monocyte-derived DCs, and previous reports have shown that PRRSV does not infect conventional DCs (cDCs) in vitro, but the effects on cDCs from lymphoid tissues are unknown. This study analyzed the response and susceptibility of tonsil DEC205+cDCs from infected pigs. We confirmed the phenotype and lineage of bona fide tonsil cDCs with the mRNA expression of FLT3+ and the phenotype MHCII+CADM1highDEC205+ (DEC205+cDCs). These cells were not infected by PRRSV, whereas CD163+ tonsil cells were infected. The numbers of tonsil cDCs and CD163+ cells were not affected by PRRSV, in contrast to the reduction in alveolar macrophage numbers. DEC205+cDCs exhibited an increase in the expression of IL-12 at 5 days postinfection, suggesting a proinflammatory response by these cells to the virus. In summary, this study confirms that, in vitro and in vivo, cDCs are not susceptible to PRRSV but can respond against it.
Subject(s)
Dendritic Cells/virology , Palatine Tonsil/cytology , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/physiology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Adhesion Molecule-1/genetics , Cell Adhesion Molecule-1/metabolism , Gene Expression Regulation/immunology , Interleukin-12/genetics , Interleukin-12/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Major Histocompatibility Complex , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Porcine Reproductive and Respiratory Syndrome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Swine , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolismABSTRACT
CD4+ T cells are major players in the immune response against several diseases; including AIDS, leishmaniasis, tuberculosis, influenza and cancer. Their activation has been successfully achieved by administering antigen coupled with antibodies, against DC-specific receptors in combination with adjuvants. Unfortunately, most of the adjuvants used so far in experimental models are unsuitable for human use. Therefore, human DC-targeted vaccination awaits the description of potent, yet nontoxic adjuvants. The nontoxic cholera B subunit (CTB) can be safely used in humans and it has the potential to activate CD4+ T cell responses. However, it remains unclear whether CTB can promote DC activation and can act as an adjuvant for DC-targeted antigens. Here, we evaluated the CTB's capacity to activate DCs and CD4+ T cell responses, and to generate long-lasting protective immunity. Intradermal (i.d.) administration of CTB promoted late and prolonged activation and accumulation of skin and lymphoid-resident DCs. When CTB was co-administered with anti-DEC205-OVA, it promoted CD4+ T cell expansion, differentiation, and infiltration to peripheral nonlymphoid tissues, i.e., the skin, lungs and intestine. Indeed, CTB promoted a polyfunctional CD4+ T cell response, including the priming of Th1 and Th17 cells, as well as resident memory T (RM) cell differentiation in peripheral nonlymphoid tissues. It is worth noting that CTB together with a DC-targeted antigen promoted local and systemic protection against experimental melanoma and murine rotavirus. We conclude that CTB administered i.d. can be used as an adjuvant to DC-targeted antigens for the induction of broad CD4+ T cell responses as well as for promoting long-lasting protective immunity.
Subject(s)
Adjuvants, Immunologic/administration & dosage , Cholera Toxin/administration & dosage , Dendritic Cells/immunology , Lectins, C-Type/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Vaccination/methods , Animals , Antigens, CD/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Humans , Injections, Intradermal , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , Male , Melanoma/immunology , Melanoma/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens/immunology , Receptors, Cell Surface/immunology , Rotavirus/immunology , Rotavirus Infections/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/virology , Th1 Cells/immunology , Th17 Cells/immunology , Treatment OutcomeABSTRACT
Conventional dendritic cells (cDCs) are divided into the following different subtypes: cDC1, which promotes a Th1 response, and cDC2, which stimulates a Th2 and Th17 response. These cells have not been characterized in porcine lymphoid tissues. DEC205 is a receptor that increases antigen presentation and allows DCs to cross-present antigens. The objectives of this work were to characterize cDCs subsets in the tonsil, submaxillary and mesenteric lymph nodes and spleen lymphoid tissues and to determine their expression of DEC205 by flow cytometry. The cDC1 (MHCIIhighCADM1highCD172a-/low) and cDC2 (MHCIIhighCADM1highCD172a+) phenotypes were confirmed by the expression of characteristic cDC1 and cDC2 transcripts (FLT3, XCR1 and FCER1α). Among all lymphoid tissues, the spleen had the highest frequency of total cDCs. The cDC1:cDC2 ratio showed that all lymph tissues had higher levels of cDC1 than levels of cDC2. DEC205+ cDCs were found in all analyzed tissues, albeit with different frequencies. Our research will facilitate the study on the function of these cells and the investigation of the strategies for DEC205 targeting and functional studies.
Subject(s)
Dendritic Cells/cytology , Lymph Nodes/cytology , Palatine Tonsil/cytology , Spleen/cytology , Swine/immunology , Animals , Dendritic Cells/immunology , Lymph Nodes/immunology , Palatine Tonsil/immunology , Spleen/immunologyABSTRACT
Rotavirus (RV) is the primary etiologic agent of severe gastroenteritis in human infants. Although two attenuated RV-based vaccines have been licensed to be applied worldwide, they are not so effective in low-income countries, and the induced protection mechanisms have not been clearly established. Thus, it is important to develop new generation vaccines that induce long lasting heterotypic immunity. VP6 constitutes the middle layer protein of the RV virion. It is the most conserved protein and it is the target of protective T-cells; therefore, it is a potential candidate antigen for a new generation vaccine against the RV infection. We determined whether targeting the DEC-205 present in dendritic cells (DCs) with RV VP6 could induce protection at the intestinal level. VP6 was cross-linked to a monoclonal antibody (mAb) against murine DEC-205 (αDEC-205:VP6), and BALB/c mice were inoculated subcutaneously (s.c.) twice with the conjugated containing 1.5 µg of VP6 in the presence of polyinosinic-polycytidylic acid (Poly I:C) as adjuvant. As controls and following the same protocol, mice were immunized with ovalbumin (OVA) cross-linked to the mAb anti-DEC-205 (αDEC-205:OVA), VP6 cross-linked to a control isotype mAb (Isotype:VP6), 3 µg of VP6 alone, Poly I:C or PBS. Two weeks after the last inoculation, mice were orally challenged with a murine RV. Mice immunized with α-DEC-205:VP6 and VP6 alone presented similar levels of serum Abs to VP6 previous to the virus challenge. However, after the virus challenge, only α-DEC-205:VP6 induced up to a 45% IgA-independent protection. Memory T-helper (Th) cells from the spleen and the mesenteric lymph node (MLN) showed a Th1-type response upon antigen stimulation in vitro. These results show that when VP6 is administered parenterally targeting DEC-205, it can induce protection at the intestinal level at a very low dose, and this protection may be Th1-type cell dependent.
Subject(s)
Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Rotavirus Infections/prevention & control , Rotavirus Vaccines/administration & dosage , Rotavirus Vaccines/immunology , Rotavirus/immunology , Adjuvants, Immunologic , Animals , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Dendritic Cells/immunology , Escherichia coli Proteins/immunology , Female , Humans , Immunologic Memory , Mice, Inbred BALB C , Minor Histocompatibility Antigens , Poly I-C/administration & dosage , Poly I-C/immunology , Rotavirus/pathogenicity , Rotavirus Infections/immunology , Rotavirus Infections/virology , Th1 Cells/immunology , Virus SheddingABSTRACT
A paracoccidioidomicose (PCM) é a micose sistêmica mais frequente no Brasil. Na última década, foi demonstrado que é possível enviar antígenos diretamente para as células dendríticas utilizando o anticorpo αDEC205 e na presença de um estímulo de maturação, o resultado é a indução de uma resposta imunológica. Verificamos que o anticorpo αDEC fusionado ao peptídeo P10 induziu uma resposta por células produtoras de IFN-γ após uma única dose em relação à administração de P10, mesmo tendo sido administrado em uma concentração menor. Entretanto, essa resposta não se manteve após segunda dose do anticorpo. Após desafio dos animais com P. brasiliensis, imunizados com duas doses do anticorpo quimérico, detectamos níveis de IFN-γ e IL-4 no tecido pulmonar estatisticamente maiores no grupo αDEC/P10 e ISO/P10 em relação à administração de P10, todos em presença de Poly I:C. Em ensaios de terapia, verificamos no pulmão de camundongos tratados com o anticorpo quimérico, principal órgão envolvido em modelo animal de PCM, baixa concentração de IFN-γ e IL-10 em relação aos controles. Em adição, ficou evidente que nos animais tratados com o anticorpo αDEC/P10 o tecido pulmonar está compatível com o tecido de animais não infectados, enquanto que na ausência de tratamento adequado encontramos aglomerados de leveduras e um tecido com aumento no infiltrado celular. Esses achados indicam uma boa evolução clínica em animais tratados e indicam que o direcionamento do P10 através do anticorpo quimérico αDEC/P10, na presença de Poly I:C, é uma estratégia promissora para terapia contra P. brasiliensis
Paracoccidioidomycosis (PCM) is the most common systemic mycosis in Brazil. In the last decade, it was demonstrated that antigens can be directly target to the dendritic cells using the antibody αDEC205 in the presence of a maturation stimulus, resulting in the induction of a strong immune response. We found that αDEC205 antibody fused to peptide P10 induced great response by IFN-γ producing cells after a single dose in relation to the administration of P10, although it has been administered in a lower concentration. However, this response was not maintained after second dose of antibody. Animals challenge with P. brasiliensis, after immunization with two doses of the chimeric antibody, produced high levels IFN-γ and IL-4 in lung tissue significantly higher in αDEC/P10 group in relation to the administration of P10, all in the presence of Poly I:C. In therapy assays, we found in the lungs of mice treated with the chimeric antibody, the main organ involved in an animal model of PCM, low concentration of IFN-γ and IL-10 compared to controls. In addition, it became evident that animals treated with αDEC/P10 antibody have a lung tissue much closer to that of non-infected tissue, while in the absence of suitable treatment we find clusters of yeasts and tissue filled with cellular infiltrates. Altogether, these findings show a clinical improvement in treated animals and indicate that targeting of P10 through the chimeric antibody αDEC/P10 in the presence of Poly I:C, is a promising strategy for therapy against P. brasiliensis