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










Publication year range
1.
Front Immunol ; 14: 1278184, 2023.
Article in English | MEDLINE | ID: mdl-37954612

ABSTRACT

Oral administration of antigen induces regulatory T cells (Treg) that can not only control local immune responses in the small intestine, but also traffic to the central immune system to deliver systemic suppression. Employing murine models of the inherited bleeding disorder hemophilia, we find that oral antigen administration induces three CD4+ Treg subsets, namely FoxP3+LAP-, FoxP3+LAP+, and FoxP3-LAP+. These T cells act in concert to suppress systemic antibody production induced by therapeutic protein administration. Whilst both FoxP3+LAP+ and FoxP3-LAP+ CD4+ T cells express membrane-bound TGF-ß (latency associated peptide, LAP), phenotypic, functional, and single cell transcriptomic analyses reveal distinct characteristics in the two subsets. As judged by an increase in IL-2Rα and TCR signaling, elevated expression of co-inhibitory receptor molecules and upregulation of the TGFß and IL-10 signaling pathways, FoxP3+LAP+ cells are an activated form of FoxP3+LAP- Treg. Whereas FoxP3-LAP+ cells express low levels of genes involved in TCR signaling or co-stimulation, engagement of the AP-1 complex members Jun/Fos and Atf3 is most prominent, consistent with potent IL-10 production. Single cell transcriptomic analysis further reveals that engagement of the Jun/Fos transcription factors is requisite for mediating TGFß expression. This can occur via an Il2ra dependent or independent process in FoxP3+LAP+ or FoxP3-LAP+ cells respectively. Surprisingly, both FoxP3+LAP+ and FoxP3-LAP+ cells potently suppress and induce FoxP3 expression in CD4+ conventional T cells. In this process, FoxP3-LAP+ cells may themselves convert to FoxP3+ Treg. We conclude that orally induced suppression is dependent on multiple regulatory cell types with complementary and interconnected roles.


Subject(s)
Interleukin-10 , T-Lymphocytes, Regulatory , Mice , Animals , Interleukin-10/metabolism , Transforming Growth Factor beta/metabolism , Forkhead Transcription Factors/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
2.
Int J Mol Sci ; 24(13)2023 Jun 21.
Article in English | MEDLINE | ID: mdl-37445595

ABSTRACT

There is evidence that IL-22 and IL-17 participate in the pathogenesis of allergic asthma. To investigate the role of IL-22, we used IL-22 deficient mice (IL-22 KO) sensitized and challenged with ovalbumin (OVA) and compared with wild type (WT) animals exposed to OVA. IL-22 KO animals exposed to OVA showed a decreased number and frequency of eosinophils, IL-5 and IL-13 in the airways, reduced mucus production and pulmonary inflammation. In addition, IL-22 KO animals exhibited a decreased percentage and number of lung CD11c+CD11b+ cells and increased apoptosis of eosinophils. Th17 cell transfer generated from IL-22 KO to animals previously sensitized and challenged with OVA caused a reduction in eosinophil frequency and number in the airways compared to animals transferred with Th17 cells generated from WT mice. Therefore, IL-22 is deleterious with concomitant secretion of IL-17. Our findings show a pro-inflammatory role for IL-22, confirmed in a model of allergen-free and allergen-specific immunotherapy. Moreover, during the comorbidity asthma and pneumonia that induces neutrophil inflammation, IL-22 was not detrimental. Our results show that targeting IL-22 would negatively affect the survival of eosinophils, reduce the expansion or migration of CD11c+CD11b+ cells, and negatively regulate allergic asthma.


Subject(s)
Asthma , Pneumonia , Mice , Animals , Interleukin-17/genetics , Asthma/pathology , Lung/pathology , Eosinophils , Pneumonia/pathology , Allergens , Comorbidity , Ovalbumin , Disease Models, Animal , Bronchoalveolar Lavage Fluid , Mice, Inbred BALB C
3.
Blood ; 142(3): 290-305, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37192286

ABSTRACT

Despite >80 years of clinical experience with coagulation factor VIII (FVIII) inhibitors, surprisingly little is known about the in vivo mechanism of this most serious complication of replacement therapy for hemophilia A. These neutralizing antidrug alloantibodies arise in ∼30% of patients. Inhibitor formation is T-cell dependent, but events leading up to helper T-cell activation have been elusive because of, in part, the complex anatomy and cellular makeup of the spleen. Here, we show that FVIII antigen presentation to CD4+ T cells critically depends on a select set of several anatomically distinct antigen-presenting cells, whereby marginal zone B cells and marginal zone and marginal metallophilic macrophages but not red pulp macrophages (RPMFs) participate in shuttling FVIII to the white pulp in which conventional dendritic cells (DCs) prime helper T cells, which then differentiate into follicular helper T (Tfh) cells. Toll-like receptor 9 stimulation accelerated Tfh cell responses and germinal center and inhibitor formation, whereas systemic administration of FVIII alone in hemophilia A mice increased frequencies of monocyte-derived and plasmacytoid DCs. Moreover, FVIII enhanced T-cell proliferation to another protein antigen (ovalbumin), and inflammatory signaling-deficient mice were less likely to develop inhibitors, indicating that FVIII may have intrinsic immunostimulatory properties. Ovalbumin, which, unlike FVIII, is absorbed into the RPMF compartment, fails to elicit T-cell proliferative and antibody responses when administered at the same dose as FVIII. Altogether, we propose that an antigen trafficking pattern that results in efficient in vivo delivery to DCs and inflammatory signaling, shape the immunogenicity of FVIII.


Subject(s)
CD4-Positive T-Lymphocytes , Factor VIII , Hemophilia A , Hemostatics , Animals , Mice , Dendritic Cells/metabolism , Factor VIII/immunology , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemostatics/immunology , Hemostatics/therapeutic use , Ovalbumin/immunology
4.
Cell Immunol ; 385: 104675, 2023 03.
Article in English | MEDLINE | ID: mdl-36746071

ABSTRACT

Active tolerance to ingested dietary antigens forms the basis for oral immunotherapy to food allergens or autoimmune self-antigens. Alternatively, oral administration of anti-CD3 monoclonal antibody can be effective in modulating systemic immune responses without T cell depletion. Here we assessed the efficacy of full length and the F(ab')2 fragment of oral anti-CD3 to prevent anti-drug antibody (ADA) formation to clotting factor VIII (FVIII) protein replacement therapy in hemophilia A mice. A short course of low dose oral anti-CD3 F(ab')2 reduced the production of neutralizing ADAs, and suppression was significantly enhanced when oral anti-CD3 was timed concurrently with FVIII administration. Tolerance was accompanied by the early induction of FoxP3+LAP-, FoxP3+LAP+, and FoxP3-LAP+ populations of CD4+ T cells in the spleen and mesenteric lymph nodes. FoxP3+LAP+ Tregs expressing CD69, CTLA-4, and PD1 persisted in spleens of treated mice, but did not produce IL-10. Finally, we attempted to combine the anti-CD3 approach with oral intake of FVIII antigen (using our previously established method of using lettuce plant cells transgenic for FVIII antigen fused to cholera toxin B (CTB) subunit, which suppresses ADAs in part through induction of IL-10 producing FoxP3-LAP+ Treg). However, combining these two approaches failed to improve suppression of ADAs. We conclude that oral anti-CD3 treatment is a promising approach to prevention of ADA formation in systemic protein replacement therapy, albeit via mechanisms distinct from and not synergistic with oral intake of bioencapsulated antigen.


Subject(s)
Hemophilia A , Mice , Animals , Hemophilia A/drug therapy , Factor VIII , Interleukin-10/metabolism , Antibody Formation , Antibodies, Monoclonal , Forkhead Transcription Factors/metabolism , Immune Tolerance , T-Lymphocytes, Regulatory
5.
Res Pract Thromb Haemost ; 7(8): 102248, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38193070

ABSTRACT

Background: Factor IX inhibitor formation is the most serious complication of replacement therapy for the bleeding disorder hemophilia B, exacerbated by severe allergic reactions occurring in up to 60% of patients with inhibitors. Low success rates of immune tolerance induction therapy in hemophilia B necessitate the search for novel immune tolerance therapies. Skin-associated lymphoid tissues have been successfully targeted in allergen-specific immunotherapy. Objectives: We aimed to develop a prophylactic immune tolerance protocol based on intradermal administration of FIX that would prevent inhibitor formation and/or anaphylaxis in response to replacement therapy. Methods: We measured FIX inhibitor, anti-FIX immunoglobulin G1, and immunoglobulin E titers using the Bethesda assay and enzyme-linked immunosorbent assay after 4 weeks of twice-weekly intradermal FIX or FIX-Fc administration followed by 5 to 6 weeks of weekly systemic FIX injections in C3H/HeJ hemophilia B mice. We also measured skin antigen-presenting, follicular helper T, and germinal center B cell frequencies in skin-draining lymph nodes after a single or repeat intradermal FIX administration. Results: Intradermal administration enhanced FIX inhibitor formation in response to systemic administration. We further found that intradermal administration alone triggers inhibitor formation, even at a low dose of 0.4 IU/kg, which is 100-fold lower than the intravenous dose of 40 IU/kg typically required to induce inhibitor development in hemophilia B mice. Also, intradermal administration triggered germinal center formation in skin-draining lymph nodes and sensitized mice to systemic administration. Factor IX-Fc fusion protein did not modulate inhibitor formation. Conclusion: Intradermal FIX administration is highly immunogenic, suggesting that the skin compartment is not amenable to immune tolerance induction or therapeutic delivery of clotting factors.

6.
Mol Ther ; 30(12): 3552-3569, 2022 12 07.
Article in English | MEDLINE | ID: mdl-35821634

ABSTRACT

Hepatic adeno-associated viral (AAV) gene transfer has the potential to cure the X-linked bleeding disorder hemophilia A. However, declining therapeutic coagulation factor VIII (FVIII) expression has plagued clinical trials. To assess the mechanistic underpinnings of this loss of FVIII expression, we developed a hemophilia A mouse model that shares key features observed in clinical trials. Following liver-directed AAV8 gene transfer in the presence of rapamycin, initial FVIII protein expression declines over time in the absence of antibody formation. Surprisingly, loss of FVIII protein production occurs despite persistence of transgene and mRNA, suggesting a translational shutdown rather than a loss of transduced hepatocytes. Some of the animals develop ER stress, which may be linked to hepatic inflammatory cytokine expression. FVIII protein expression is preserved by interleukin-15/interleukin-15 receptor blockade, which suppresses CD8+ T and natural killer cell responses. Interestingly, mice with initial FVIII levels >100% of normal had diminishing expression while still under immune suppression. Taken together, our findings of interanimal variability of the response, and the ability of the immune system to shut down transgene expression without utilizing cytolytic or antibody-mediated mechanisms, illustrate the challenges associated with FVIII gene transfer. Our protocols based upon cytokine blockade should help to maintain efficient FVIII expression.


Subject(s)
Factor VIII , Interleukin-15 , Mice , Animals , Factor VIII/genetics , Interleukin-15/genetics , Sirolimus/pharmacology
7.
Front Immunol ; 12: 672449, 2021.
Article in English | MEDLINE | ID: mdl-34135899

ABSTRACT

Adeno associated viral (AAV) vectors have emerged as a preferred platform for in vivo gene replacement therapy and represent one of the most promising strategies to treat monogenetic disorders such as hemophilia. However, immune responses to gene transfer have hampered human gene therapy in clinical trials. Over the past decade, it has become clear that innate immune recognition provides signals for the induction of antigen-specific responses against vector or transgene product. In particular, TLR9 recognition of the vector's DNA genome in plasmacytoid dendritic cells (pDCs) has been identified as a key factor. Data from clinical trials and pre-clinical studies implement CpG motifs in the vector genome as drivers of immune responses, especially of CD8+ T cell activation. Here, we demonstrate that cross-priming of AAV capsid-specific CD8+ T cells depends on XCR1+ dendritic cells (which are likely the main cross-presenting cell that cooperates with pDCs to activate CD8+ T cells) and can be minimized by the elimination of CpG motifs in the vector genome. Further, a CpG-depleted vector expressing human coagulation factor IX showed markedly reduced (albeit not entirely eliminated) CD8+ T cell infiltration upon intramuscular gene transfer in hemophilia B mice when compared to conventional CpG+ vector (comprised of native sequences), resulting in better preservation of transduced muscle fibers. Therefore, this deimmunization strategy is helpful in reducing the potential for CD8+ T cell responses to capsid or transgene product. However, CpG depletion had minimal effects on antibody responses against capsid or transgene product, which appear to be largely independent of CpG motifs.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dependovirus/immunology , Genetic Therapy/methods , Genetic Vectors/immunology , Oligodeoxyribonucleotides/immunology , Animals , Humans , Mice , Mice, Inbred C57BL
8.
Plant Biotechnol J ; 19(10): 1952-1966, 2021 10.
Article in English | MEDLINE | ID: mdl-33949086

ABSTRACT

Anti-drug antibody (ADA) formation is a major complication in treatment of the X-linked bleeding disorder haemophilia B (deficiency in coagulation factor IX, FIX). Current clinical immune tolerance protocols are often not effective due to complications such as anaphylactic reactions against FIX. Plant-based oral tolerance induction may address this problem, as illustrated by the recent first regulatory approval of orally delivered plant cells to treat peanut allergy. Our previous studies showed that oral delivery of plant cells expressing FIX fused to the transmucosal carrier CTB (cholera toxin subunit B) in chloroplasts suppressed ADA in animals with haemophilia B. We report here creation of the first lettuce transplastomic lines expressing a coagulation factor, in the absence of antibiotic resistance gene. Stable integration of the CTB-FIX gene and homoplasmy (transformation of ˜10 000 copies in each cell) were maintained in both T1 and T2 generation marker-free plants. CTB-FIX expression in lyophilized leaves of T1 and T2 marker-free plants was 1.0-1.5 mg/g dry weight, confirming that the marker excision did not affect antigen levels. Oral administration of CTB-FIX to Sprague Dawley rats at 0.25, 1 or 2.5 mg/kg did not produce overt adverse effects or toxicity. The no-observed-adverse-effect level (NOAEL) is at least 2.5 mg/kg for a single oral administration in rats. Oral administration of CTB-FIX at 0.3 or 1.47 mg/kg either mixed in food or as an oral suspension to Beagle dogs did not produce any observable toxicity. These toxicology studies should facilitate filing of regulatory approval documents and evaluation in haemophilia B patients.


Subject(s)
Hemophilia B , Administration, Oral , Animals , Chloroplasts , Cholera Toxin , Dogs , Factor IX/genetics , Hemophilia B/drug therapy , Humans , Rats , Rats, Sprague-Dawley
9.
Cell Immunol ; 359: 104251, 2021 01.
Article in English | MEDLINE | ID: mdl-33248367

ABSTRACT

Oral antigen administration to induce regulatory T cells (Treg) takes advantage of regulatory mechanisms that the gastrointestinal tract utilizes to promote unresponsiveness against food antigens or commensal microorganisms. Recently, antigen-based oral immunotherapies (OITs) have shown efficacy as treatment for food allergy and autoimmune diseases. Similarly, OITs appear to prevent anti-drug antibody responses in replacement therapy for genetic diseases. Intestinal epithelial cells and microbiota possibly condition dendritic cells (DC) toward a tolerogenic phenotype that induces Treg via expression of several mediators, e.g. IL-10, transforming growth factor-ß, retinoic acid. Several factors, such as metabolites derived from microbiota or diet, impact the stability and expansion of these induced Treg, which include, but are not limited to, FoxP3+ Treg, LAP+ Treg, and/or Tr1 cells. Here, we review various orally induced Treg, their plasticity and cooperation between the Treg subsets, as well as underlying mechanisms controlling their induction and role in oral tolerance.


Subject(s)
Immune Tolerance/immunology , Immunotherapy/methods , T-Lymphocytes, Regulatory/immunology , Administration, Oral , Allergens/immunology , Animals , Dendritic Cells/immunology , Food Hypersensitivity/immunology , Forkhead Transcription Factors/metabolism , Humans , Immunologic Factors , Intestinal Mucosa/immunology , Intestines/immunology , Peptides/metabolism , Protein Precursors/metabolism , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism
10.
Front Immunol ; 11: 844, 2020.
Article in English | MEDLINE | ID: mdl-32508814

ABSTRACT

Fusion proteins, which consist of factor VIII or factor IX and the transmucosal carrier cholera toxin subunit B, expressed in chloroplasts and bioencapsulated within plant cells, initiate tolerogenic immune responses in the intestine when administered orally. This approach induces regulatory T cells (Treg), which suppress inhibitory antibody formation directed at hemophilia proteins induced by intravenous replacement therapy in hemophilia A and B mice. Further analyses of Treg CD4+ lymphocyte sub-populations in hemophilia B mice reveal a marked increase in the frequency of CD4+CD25-FoxP3-LAP+ T cells (but not of CD4+CD25+FoxP3+ T cells) in the lamina propria of the small but not large intestine. The adoptive transfer of very small numbers of CD4+CD25-LAP+ Treg isolated from the spleen of tolerized mice was superior in suppression of antibodies directed against FIX when compared to CD4+CD25+ T cells. Thus, tolerance induction by oral delivery of antigens bioencapsulated in plant cells occurs via the unique immune system of the small intestine, and suppression of antibody formation is primarily carried out by induced latency-associated peptide (LAP) expressing Treg that likely migrate to the spleen. Tolerogenic antigen presentation in the small intestine requires partial enzymatic degradation of plant cell wall by commensal bacteria in order to release the antigen. Microbiome analysis of hemophilia B mice showed marked differences between small and large intestine. Remarkably, bacterial species known to produce a broad spectrum of enzymes involved in degradation of plant cell wall components were found in the small intestine, in particular in the duodenum. These were highly distinct from populations of cell wall degrading bacteria found in the large intestine. Therefore, FIX antigen presentation and Treg induction by the immune system of the small intestine relies on activity of a distinct microbiome that can potentially be augmented to further enhance this approach.


Subject(s)
Cholera Toxin/immunology , Factor IX/immunology , Gastrointestinal Microbiome/immunology , Hemophilia B/immunology , Hemophilia B/microbiology , Immune Tolerance/immunology , Intestine, Small/immunology , Intestine, Small/microbiology , Plant Cells/metabolism , Administration, Oral , Adoptive Transfer/methods , Animals , Antigen Presentation , Antigens/genetics , Antigens/immunology , Cholera Toxin/genetics , Disease Models, Animal , Factor IX/genetics , Gene Deletion , Genome, Chloroplast , Lactuca/genetics , Male , Mice , Mice, Transgenic , Plants, Genetically Modified , T-Lymphocytes, Regulatory/immunology
11.
J Ethnopharmacol ; 252: 112496, 2020 Apr 24.
Article in English | MEDLINE | ID: mdl-31870795

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Propolis is a natural product produced by honeybees used as a medicine at least to 300 BC. In the last decades, several studies showed biological and pharmacological properties of propolis, witch scientifically explains the empirical use for centuries. The anti-inflammatory activity of propolis with the purpose to reduce Th2 inflammation has been evaluated in allergic asthma. However, it remains to be determined how propolis negatively regulates the immune response after allergen re-exposure. AIM OF THE STUDY: We hypothesized that the anti-inflammatory activity of propolis is dependent on the induction of myeloid derived suppressor cells (MDSC) and regulatory T cells. MATERIALS AND METHODS: To assess this hypothesis, we used an ovalbumin-induced asthma model to evaluate the effect of EPP-AF® dry extract from Brazilian green propolis. RESULTS: Propolis treatment decreased pulmonary inflammation and mucus production as well as eosinophils and IL-5 in the broncoalveolar lavage. Propolis enhanced also in vitro differentiation and in vivo frequency of lung MDSC and CD4+Foxp3+ regulatory T cells. CONCLUSIONS: Together these results confirm the immunomodulatory potential of propolis during sensitization and challenge with allergen. In addition, the collecting findings show, for the first time, that propolis increases the frequency of MDSC and CD4+Foxp3+ regulatory T cells in the lungs, and suggest that it could be use as target for development of new immunotherapy or adjuvant immunotherapy for asthma.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Immunologic Factors/therapeutic use , Myeloid-Derived Suppressor Cells/drug effects , Propolis/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Allergens , Animals , Anti-Inflammatory Agents/pharmacology , Asthma/chemically induced , Asthma/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Differentiation/drug effects , Female , Immunologic Factors/pharmacology , Immunotherapy , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-5/immunology , Lung/drug effects , Lung/immunology , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Ovalbumin , Propolis/pharmacology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/drug effects , Th2 Cells/immunology
12.
Cell Death Dis ; 10(1): 11, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30584243

ABSTRACT

Chronic pulmonary inflammation marked predominantly by CD4+IFN-γ+ cells is the hallmark of tuberculosis pathogenesis in immunocompetent adults, who are substantially affected by this disease. Moreover, CD4+Foxp3+ cell-mediated suppression contributes to infection susceptibility. We addressed the role of CD4+Foxp3+ cells in tuberculosis pathogenesis, because this aspect has not been addressed during chronic infection. We targeted CCR4, which induces the influx of CD4+Foxp3+ cells into the lungs. CCR4-/- mice exhibited a lower frequency of CD4+Foxp3+ cells at 15, 30, and 70 days of infection than their wild-type counterparts. However, only at 70 days of infection was an exacerbated IFN-γ-mediated immune response associated with apparent tuberculosis pathogenesis and susceptibility. In addition, CCR4-/- mice exhibited a decrease in the suppressor function of CD4+Foxp3+ cells. Adoptive transfer of Foxp3+ cells into infected CCR4-/- mice restored pulmonary inflammation and bacterial load to levels observed in wild-type mice. Our findings suggest that CD4+Foxp3+ cells play a time-dependent role in tuberculosis and highlight that CCR4 plays a critical role in the balance of IFN-γ-mediated inflammation by regulating the influx and function of CD4+Foxp3+ cells. Our findings are translationally relevant, as CD4+Foxp3+ cells or CCR4 could be a target for immunotherapy, considering the heterogeneity of tuberculosis in immunocompetent adults.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Mycobacterium tuberculosis/immunology , Receptors, CCR4/immunology , Tuberculosis, Pulmonary/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Interferon-gamma/genetics , Mice , Mice, Knockout , Receptors, CCR4/genetics , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/pathology
13.
Immunology ; 148(1): 102-13, 2016 May.
Article in English | MEDLINE | ID: mdl-26840507

ABSTRACT

M1 macrophages are more effective in the induction of the inflammatory response and clearance of Mycobacterium tuberculosis than M2 macrophages. Infected C57BL/6 mice generate a stronger cellular immune response compared with BALB/c mice. We hypothesized that infected C57BL/6 mice would exhibit a higher frequency and function of M1 macrophages than infected BALB/c mice. Our findings show a higher ratio of macrophages to M2 macrophages in the lungs of chronically infected C57BL/6 mice compared with BALB/c mice. However, there was no difference in the functional ability of M1 and M2 macrophages for the two strains in vitro. In vivo, a deleterious role for M2 macrophages was confirmed by M2 cell transfer, which rendered the infected C57BL/6, but not the BALB/c mice, more susceptible and resulted in mild lung inflammation compared with C57BL/6 mice that did not undergo cell transfer. M1 cell transfer induced a higher inflammatory response, although not protective, in infected BALB/c mice compared with their counterparts that did not undergo cell transfer. These findings demonstrate that an inflammation mediated by M1 macrophages may not induce bacterial tolerance because protection depends on the host genetic background, which drives the magnitude of the inflammatory response against M. tuberculosis in the pulmonary microenvironment. The contribution of our findings is that although M1 macrophage is an effector leucocyte with microbicidal machinery, its dominant role depends on the balance of M1 and M2 subsets, which is driven by the host genetic background.


Subject(s)
Lung/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Animals , Disease Susceptibility , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle, Smooth, Vascular/enzymology , Nitric Oxide Synthase Type II/analysis , Species Specificity
14.
Immunology ; 147(2): 240-50, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26595158

ABSTRACT

The voltage-gated potassium channel Kv1.3 is a novel target for immunomodulation of autoreactive effector memory T cells, which play a major role in the pathogenesis of autoimmune diseases. In this study, the Ts6 and Ts15 toxins isolated from Tityus serrulatus (Ts) were investigated for their immunosuppressant roles on CD4(+) cell subsets: naive, effector (TEF ), central memory (TCM) and effector memory (TEM). The electrophysiological assays confirmed that both toxins were able to block Kv1.3 channels. Interestingly, an extended Kv channel screening shows that Ts15 blocks Kv2.1 channels. Ts6 and Ts15 significantly inhibit the proliferation of TEM cells and interferon-γ production; however, Ts15 also inhibits other CD4(+) cell subsets (naive, TEF and TCM). Based on the Ts15 inhibitory effect of proliferation of all CD4(+) cell subsets, and based on its blocking effect on Kv2.1, we investigated the Kv2.1 expression in T cells. The assays showed that CD4(+) and CD8(+) cells express the Kv2.1 channels mainly extracellularly with TCM cells expressing the highest number of Kv2.1 channels. We also provide in vivo experimental evidence to the protective effect of Ts6 and Ts15 on delayed-type hypersensitivity reaction. Altogether, this study presents the immunosuppressive behaviour of Ts6 and Ts15 toxins, indicating that these toxins could be promising candidates for autoimmune disease therapy. Moreover, this is the first report illustrating the involvement of a novel K(+) channel subtype, Kv2.1, and its distribution in T-cell subsets.


Subject(s)
Immunosuppressive Agents/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Scorpion Venoms/pharmacology , T-Lymphocytes/drug effects , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/metabolism , Hypersensitivity, Delayed/prevention & control , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/metabolism , Lymphocyte Activation/drug effects , Male , Membrane Potentials , Mice, Inbred BALB C , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Serum Albumin, Bovine , Shab Potassium Channels/antagonists & inhibitors , Shab Potassium Channels/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenopus laevis
15.
Immunology ; 144(4): 574-86, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25322675

ABSTRACT

The magnitude of the cellular adaptive immune response is critical for the control of Mycobacterium tuberculosis infection in the chronic phase. In addition, the genetic background is equally important for resistance or susceptibility to tuberculosis. In this study, we addressed whether lung populations of dendritic cells, obtained from genetically different hosts, would play a role in the magnitude and function of CD4(+) populations generated after M. tuberculosis infection. Thirty days post-infection, C57BL/6 mice, which generate a stronger interferon-γ (IFN-γ)-mediated immune response than BALB/c mice, exhibited a higher number and frequency of lung CD11c(+)  CD11b(-)  CD103(+) cells compared with BALB/c mice, which exhibited a high frequency of lung CD11c(+)  CD11b(+)  CD103(-) cells. CD11c(+)  CD11b(-)  CD103(+) cells, purified from lungs of infected C57BL/6 mice, but not from infected BALB/c mice, induced a higher frequency of IFN-γ-producing or interleukin-17 (IL-17)-producing CD4(+) cells. Moreover, CD4(+) cells also arrive at the lung of C57BL/6 mice faster than in BALB/c mice. This pattern of immune response seems to be associated with higher gene expression for CCL4, CCL19, CCL20 and CCR5 in the lungs of infected C57BL/6 mice compared with infected BALB/c mice. The results described here show that the magnitude of IFN-γ-producing or IL-17-producing CD4(+) cells is dependent on CD11c(+)  CD11b(-)  CD103(+) cells, and this pattern of immune response is directly associated with the host genetic background. Therefore, differences in the genetic background contribute to the identification of immunological biomarkers that can be used to design human assays to predict progression of M. tuberculosis infection.


Subject(s)
Antigens, CD/immunology , CD11c Antigen/immunology , Integrin alpha Chains/immunology , Interferon-gamma/immunology , Interleukin-17/immunology , Lung/immunology , Mycobacterium tuberculosis/immunology , Th17 Cells/immunology , Tuberculosis, Pulmonary/immunology , Animals , Antigens, CD/metabolism , CD11b Antigen/immunology , CD11b Antigen/metabolism , CD11c Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Disease Models, Animal , Female , Genotype , Immunity, Cellular , Integrin alpha Chains/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Lung/metabolism , Lung/microbiology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mycobacterium tuberculosis/pathogenicity , Phenotype , Signal Transduction , Species Specificity , Th17 Cells/metabolism , Th17 Cells/microbiology , Time Factors , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/microbiology
16.
Toxicon ; 76: 1-10, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23998940

ABSTRACT

In various types of snake venom, the major toxic components are proteinases and members of the phospholipase A2 family, although other enzymes also contribute to the toxicity. In this study, we evaluated the proteolytic, phospholipase, and L-Amino acid oxidase activities in the venom of five Bothrops species-Bothrops jararaca, Bothrops jararacussu, Bothrops moojeni, Bothrops neuwiedi, and Bothrops alternatus-all of which are used in the production of commercial antivenom, prepared in horses. The enzymatic activities of each species' venom were classified as high, moderate, or low. B. moojeni venom demonstrated the highest enzymatic activity profile, followed by the venom of B. neuwiedi, B. jararacussu, B. jararaca, and B. alternatus. To our knowledge, this is the first study to compare all of these enzymes from multiple species, which is significant in view of the activity of L-amino acid oxidase across Bothrops species.


Subject(s)
Bothrops , Crotalid Venoms/enzymology , Animals , Brazil , Cattle , Crotalid Venoms/chemistry , L-Amino Acid Oxidase/chemistry , Peptide Hydrolases/chemistry , Phospholipases/chemistry , Proteolysis , Sheep , Species Specificity
17.
J Immunotoxicol ; 10(2): 160-8, 2013.
Article in English | MEDLINE | ID: mdl-22954026

ABSTRACT

Approximately 20,000 snakebites are registered each year in Brazil. The classical treatment for venomous snakebite involves the administration of sera obtained from immunized horses. Moreover, the production and care of horses is costly, and the use of heterologous sera can cause hypersensitivity reactions. The production of human antibody fragments by phage display technology is seen as a means of overcoming some of these disadvantages. The studies here attempted to test human monoclonal antibodies specific to Bothrops jararacussu against other Bothrops sp. venoms, using the Griffin.1 library of human single-chain fragment-variable (scFv) phage antibodies. Using the Griffin.1 phage antibody library, this laboratory previously produced scFvs capable of inhibiting the phospholipase and myotoxic activities of Bothrops jararacussu venom. The structural and functional similarities of the various forms of phospholipase A2 (PLA2) in Bothrops venom served as the basis for the present study wherein the effectiveness of those same scFvs were evaluated against B. jararaca, B. neuwiedi, and B. moojeni venoms. Each clone was found to recognize all three Bothrops venoms, and purified scFvs partially inhibited their in vitro phospholipase activity. In vivo assays demonstrated that the scFv clone P2B7 reduced myotoxicity and increased the survival of animals that received the test venoms. The results here indicate that the scFv P2B7 is a candidate for inclusion in a mixture of specific antibodies to produce a human anti-bothropic sera. This data demonstrates that the human scFv P2B7 represents an alternative therapeutic approach to heterologous anti-bothropic sera available today.


Subject(s)
Antibodies, Monoclonal/immunology , Antivenins/immunology , Bothrops/immunology , Crotalid Venoms/immunology , Single-Chain Antibodies/immunology , Animals , Antibodies, Monoclonal/chemistry , Antivenins/chemistry , Cell Surface Display Techniques , Creatine Kinase/blood , Crotalid Venoms/chemistry , Female , Hemolytic Plaque Technique , Humans , Mice , Phospholipases A2/chemistry , Species Specificity , Structural Homology, Protein
18.
J Immunotoxicol ; 9(2): 173-83, 2012.
Article in English | MEDLINE | ID: mdl-22424317

ABSTRACT

In Brazil, the species Tityus serrulatus is responsible for the most severe cases of scorpion envenomation. There is currently a need for new scorpion anti-venoms that are more effective and less harmful. This study attempted to produce human monoclonal antibodies capable of inhibiting the activity of T. serrulatus venom (TsV), using the Griffin.1 library of human single-chain fragment-variable (scFv) phage antibodies. Four rounds of phage antibody selection were performed, and the round with the highest phage antibody titer was chosen for the production of monoclonal phage antibodies and for further analysis. The scFv 2A, designated serrumab, was selected for the production and purification of soluble antibody fragments. In a murine peritoneal macrophage cell line (J774.1), in vitro assays of the cytokines interleukin (IL)-6, tumor necrosis factor (TNF)-α, and IL-10 were performed. In male BALB/c mice, in vivo assays of plasma urea, creatinine, aspartate transaminase, and glucose were performed, as well as of neutrophil recruitment and leukocyte counts. It was found that serrumab inhibited the TsV-induced increases in the production of IL-6, TNFα, and IL-10 in J774.1 cells. The in vivo inhibition assay showed that serrumab also prevented TsV-induced increases in the plasma levels of urea, creatinine, aspartate transaminase, and glucose, as well as preventing the TsV-induced increase in neutrophil recruitment. The results indicate that the human monoclonal antibody serrumab is a candidate for inclusion in a mixture of specific antibodies to the various toxins present in TsV. Therefore, serrumab shows promise for use in the production of new anti-venom.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antivenins/immunology , Insect Proteins/immunology , Scorpion Venoms/immunology , Scorpions/physiology , Single-Chain Antibodies/immunology , Animals , Antibodies, Monoclonal, Humanized/biosynthesis , Antibodies, Monoclonal, Humanized/therapeutic use , Antivenins/biosynthesis , Antivenins/therapeutic use , Cell Survival/drug effects , Cytokines/metabolism , Insect Proteins/antagonists & inhibitors , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred BALB C , Neutralization Tests , Peptide Library , Scorpion Venoms/antagonists & inhibitors , Scorpion Venoms/toxicity , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/therapeutic use
19.
Basic Clin Pharmacol Toxicol ; 110(3): 290-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22017759

ABSTRACT

The hybrid created from the crossbreeding of European and African bees, known as the Africanised bee, has provided numerous advantages for current beekeeping. However, this new species exhibits undesirable behaviours, such as colony defence instinct and a propensity to attack en masse, which can result in serious accidents. To date, there is no effective treatment for cases of Africanised bee envenomation. One promising technique for developing an efficient antivenom is the use of phage display technology, which enables the production of human antibodies, thus avoiding the complications of serum therapy, such as anaphylaxis and serum sickness. The aim of this study was to produce human monoclonal single-chain Fv (scFv) antibody fragments capable of inhibiting the toxic effects of Africanised bee venom. We conducted four rounds of selection of antibodies against the venom and three rounds of selection of antibodies against purified melittin. Three clones were selected and tested by enzyme-linked immunosorbent assay to verify their specificity for melittin and phospholipase A2. Two clones (C5 and C12) were specific for melittin, and one (A7) was specific for phospholipase A2. In a kinetic haemolytic assay, these clones were evaluated individually and in pairs. The A7-C12 combination had the best synergistic effect and was chosen to be used in the assays of myotoxicity inhibition and lethality. The A7-C12 combination inhibited the in vivo myotoxic effect of the venom and increased the survival of treated animals.


Subject(s)
Antivenins/immunology , Bee Venoms/toxicity , Melitten/immunology , Phospholipases A2/immunology , Animals , Antibodies, Monoclonal/immunology , Bee Venoms/immunology , Bees , Enzyme-Linked Immunosorbent Assay , Female , Humans , Insect Bites and Stings/immunology , Insect Bites and Stings/therapy , Mice , Single-Chain Antibodies/immunology , Survival
20.
Braz. j. pharm. sci ; 47(1): 31-38, Jan.-Mar. 2011. ilus, graf, tab
Article in English | LILACS | ID: lil-586522

ABSTRACT

Monoclonal antibodies represent the fastest growing class of biopharmaceutical products and have a host of applications in medical research, diagnosis, therapy, and basic science. The production of recombinant monoclonal antibodies has revolutionized the generation of immunoglobulins, and their use represents a strategic breakthrough, affecting the global pharmaceutical market for therapeutic proteins. In the present work, a review of scFv, and the number of related patents, has been carried out. The results show that several countries have scFv patents, most notably the United States, China and United Kingdom. The target of these scFv antibodies was also assessed and the results demonstrate that most are directed toward cancer therapy.


Anticorpos monoclonais representam a classe de maior crescimento em produtos de biofármacos e possuem várias aplicações em pesquisa médica, diagnóstico, terapias e ciência básica. A produção de anticorpos monoclonais recombinantes revolucionou a geração de imunoglobulinas e sua utilização implica em avanço estratégico, afetando o mercado farmacêutico global de proteínas terapêuticas. No presente trabalho, uma revisão sobre scFv e a relação do seu número de patentes foi analisada. Os resultados mostram que vários países apresentam patentes de scFv com destaque para os Estados Unidos, China e Reino Unido. Os alvos desses anticorpos também foram avaliados e as análises revelaram que a maioria é destinado a terapias contra o câncer.


Subject(s)
Antigens, Differentiation , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Biopharmaceutics/trends , Drug Synergism , Intellectual Property of Pharmaceutic Products and Process , Colony-Stimulating Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...