Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Nitric Oxide ; 148: 1-12, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38636582

ABSTRACT

Epidemiological studies show a strong correlation between diabetes and the increased risk of developing different cancers, including melanoma. In the present study, we investigated the impact of a streptozotocin (STZ)-induced hyperglycemic environment on B16F10-Nex2 murine melanoma development. Hyperglycemic male C57Bl/6 mice showed increased subcutaneous tumor development, partially inhibited by metformin. Tumors showed increased infiltrating macrophages, and augmented IL-10 and nitric oxide (NO) concentrations. In vivo neutralization of IL-10, NO synthase inhibition, and depletion of macrophages reduced tumor development. STZ-treated TLR4 KO animals showed delayed tumor development; the transfer of hyperglycemic C57Bl/6 macrophages to TLR4 KO reversed this effect. Increased concentrations of IL-10 present in tumor homogenates of hyperglycemic mice induced a higher number of pre-angiogenic structures in vitro, and B16F10-Nex2 cells incubated with different glucose concentrations in vitro produced increased levels of IL-10. In summary, our findings show that a hyperglycemic environment stimulates murine melanoma B16F10-Nex2 primary tumor growth, and this effect is dependent on tumor cell stimulation, increased numbers of macrophages, and augmented IL-10 and NO concentrations. These findings show the involvement of tumor cells and other components of the tumor microenvironment in the development of subcutaneous melanoma under hyperglycemic conditions, defining novel targets for melanoma control in diabetic patients.


Subject(s)
Hyperglycemia , Interleukin-10 , Macrophages , Melanoma, Experimental , Mice, Inbred C57BL , Nitric Oxide , Animals , Interleukin-10/metabolism , Nitric Oxide/metabolism , Male , Hyperglycemia/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Macrophages/metabolism , Macrophages/drug effects , Mice , Mice, Knockout , Cell Line, Tumor
2.
Biosci. rep ; 41(2): 1-11, Feb. 2021. ilus, graf, tab
Article in English | CONASS, Sec. Est. Saúde SP, SESSP-IDPCPROD, Sec. Est. Saúde SP | ID: biblio-1147459

ABSTRACT

Introduction: Almost 20% of patients with acute myocardial infarction (MI) develop heart failure, even when early reperfused [1]. Left ventricular remodeling seems related to the size of myocardial infarction and timely reperfusion, as well as to the inflammatory responses and residual ischemia [2]. Experimental studies suggested that B lymphocytes may influence the myocardial infarcted mass [3], although there are few data about the role of these cells in humans. Furthermore, a possible atheroprotective role for B1 lymphocytes has been proposed based on the production of interleukin 10 (IL-10) and natural antibodies, which may switch the proinflammatory response to more appropriate healing, promoting cell recovery and the clearance of apoptotic cellular debris [4]. On the other hand, classic B lymphocytes or B2 cells are linked to progression of atherosclerosis, possibly by their interaction with CD4+ T lymphocytes [4]. In 2011, Griffin and colleagues proposed CD19+CD20+CD43+CD70- lymphocyte cells as the human B1 phenotype, and these cells spontaneously produced IgM and IL-10 [5]. However, according to the presence or absence of the CD11b on the surface of these cells, the capacity of IgM production and activation Stem cells in blood marrow differentiate in T or B lymphocyte, according to the presence of CD3 or CD19, respectively. Lymphocyte final maturation takes place in thymus for T cells; or in spleen and lymphatic tissue for classic B cells. B1 lymphocytes are well described in experimental studies. These cells are notorious for their capacity of spontaneous production of IgM and according to the presence of the CD11b, two distinct subtypes are recognized: CD11b- B1 lymphocytes, producing IgM, and CD11b+ B1 lymphocytes, related to the expansion of CD4+ T lymphocytes.


Subject(s)
B-Lymphocytes , Magnetic Resonance Spectroscopy , Cytokines , Myocardial Infarction
3.
Mol Immunol ; 131: 144-154, 2021 03.
Article in English | MEDLINE | ID: mdl-33422341

ABSTRACT

Sticholysins (Sts) I and II (StI and StII) are pore-forming proteins (PFPs), purified from the Caribbean Sea anemone Stichodactyla helianthus. StII encapsulated into liposomes induces a robust antigen-specific cytotoxic CD8+ T lymphocytes (CTL) response and in its free form the maturation of bone marrow-derived dendritic cells (BM-DCs). It is probable that the latter is partially supporting in part the immunomodulatory effect on the CTL response induced by StII-containing liposomes. In the present work, we demonstrate that the StII's ability of inducing maturation of BM-DCs is also shared by StI, an isoform of StII. Using heat-denatured Sts we observed a significant reduction in the up-regulation of maturation markers indicating that both PFP's ability to promote maturation of BM-DCs is dependent on their conformational characteristics. StII-mediated DC maturation was abrogated in BM-DCs from toll-like receptor (TLR) 4 and myeloid differentiation primary response gene 88 (MyD88)-knockout mice but not in cells from TLR2-knockout mice. Furthermore, the antigen-specific CTL response induced by StII-containing liposomes was reduced in TLR4-knockout mice. These results indicate that StII, and probably by extension StI, has the ability to induce maturation of DCs through a TLR4/MyD88-dependent pathway, and that this activation contributes to the CTL response generated by StII-containing liposomes.


Subject(s)
Cnidarian Venoms/metabolism , Dendritic Cells/metabolism , Toll-Like Receptor 4/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Organic Chemicals/metabolism , Signal Transduction/physiology
4.
Biosci Rep ; 41(2)2021 02 26.
Article in English | MEDLINE | ID: mdl-33495783

ABSTRACT

Despite early reperfusion, patients with ST segment elevation myocardial infarction (STEMI) may present large myocardial necrosis and significant impairment of ventricular function. The present study aimed to evaluate the role of subtypes of B lymphocytes and related cytokines in the infarcted mass and left ventricular ejection fraction obtained by cardiac magnetic resonance imaging performed after 30 days of STEMI. This prospective study included 120 subjects with STEMI submitted to pharmacoinvasive strategy. Blood samples were collected in subjects in the first (D1) and 30th (D30) days post STEMI. The amount of CD11b+ B1 lymphocytes (cells/ml) at D1 were related to the infarcted mass (rho = 0.43; P=0.033), measured by cardiac MRI at D30. These B1 cells were associated with CD4+ T lymphocytes at D1 and D30, while B2 classic lymphocytes at day 30 were related to left ventricular ejection fraction (LVEF). Higher titers of circulating IL-4 and IL-10 were observed at D30 versus D1 (P=0.013 and P<0.001, respectively). Titers of IL-6 at D1 were associated with infarcted mass (rho = 0.41, P<0.001) and inversely related to LVEF (rho = -0.38, P<0.001). After multiple linear regression analysis, high-sensitivity troponin T and IL-6 collected at day 1 were independent predictors of infarcted mass and, at day 30, only HDL-C. Regarding LVEF, high-sensitivity troponin T and high-sensitivity C-reactive protein were independent predictors at day 1, and B2 classic lymphocytes, at day 30. In subjects with STEMI, despite early reperfusion, the amount of infarcted mass and ventricular performance were related to inflammatory responses triggered by circulating B lymphocytes.


Subject(s)
B-Lymphocytes/immunology , Myocardial Infarction/immunology , Adult , Antigens, CD/immunology , Female , Humans , Interleukin-10/blood , Interleukin-4/blood , Magnetic Resonance Imaging , Male , Myocardial Infarction/diagnostic imaging , Sensitivity and Specificity , Troponin T/blood
5.
Front Immunol ; 11: 345, 2020.
Article in English | MEDLINE | ID: mdl-32194563

ABSTRACT

The palladacycle complex DPPE 1.2 was previously shown to inhibit Leishmania (Leishmania) amazonensis infection in vitro and in vivo. The present study aimed to evaluate the effect of DPPE 1.2 associated with a recombinant cysteine proteinase, rLdccys1, and the adjuvant Propionibacterium acnes on L. (L.) amazonensis infection in two mouse strains, BALB/c, and C57BL/6. Treatment with this association potentiated the leishmanicidal effect of DPPE 1.2 resulting in a reduction of parasite load in both strains of mice which was higher compared to that found in groups treated with either DPPE 1.2 alone or associated with P. acnes or rLdccys1. The reduction of parasite load in both mice strains was followed by immunomodulation mediated by an increase of memory CD4+ and CD8+ T lymphocytes, IFN-γ levels and reduction of active TGF-ß in treated animals. No infection relapse was observed 1 month after the end of treatment in mice which received DPPE 1.2 associated with rLdccys1 or rLdccys1 plus P. acnes in comparison to that exhibited by animals treated with DPPE 1.2 alone. Evaluation of serum levels of AST, ALT, urea, and creatinine showed no alterations among treated groups, indicating that this treatment schedule did not induce hepato or nephrotoxicity. These data indicate the potential use of this association as a therapeutic alternative for cutaneous leishmaniasis caused by L. (L) amazonensis.


Subject(s)
Antiprotozoal Agents/therapeutic use , Cysteine Endopeptidases/therapeutic use , Immunotherapy/methods , Leishmaniasis, Cutaneous/drug therapy , Propionibacterium acnes , Protozoan Proteins/therapeutic use , Animals , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/toxicity , Combined Modality Therapy , Cysteine Endopeptidases/administration & dosage , Cysteine Endopeptidases/immunology , Cysteine Endopeptidases/toxicity , Drug Evaluation, Preclinical , Female , Immunologic Memory , Interferon-gamma/metabolism , Leishmania mexicana , Leishmaniasis, Cutaneous/immunology , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protozoan Proteins/administration & dosage , Protozoan Proteins/immunology , Protozoan Proteins/toxicity , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/metabolism
6.
PLoS One ; 13(10): e0205148, 2018.
Article in English | MEDLINE | ID: mdl-30300366

ABSTRACT

Hybrid vaccines have been investigated in clinical and experimental studies once expresses total antigens of a tumor cell combined with the ability of a dendritic cell (DC) to stimulate immune responses. However, the response triggered by these vaccines is often weak, requiring the use of adjuvants to increase vaccine immunogenicity. Killed Propionibacterium acnes (P. acnes) exerts immunomodulatory effects by increasing the phagocytic and tumoricidal activities of macrophages, promoting DC maturation, inducing pro-inflammatory cytokines production and increasing the humoral response to different antigens. Here, we evaluated the effect of P. acnes on a specific antitumor immune response elicited by a hybrid vaccine in a mouse melanoma model. Hybrid vaccine associated with P. acnes increased the absolute number of memory T cells, the IFN-γ secretion by these cells and the IgG-specific titers to B16F10 antigens, polarizing the immune response to a T helper 1 pattern. Furthermore, the addition of P. acnes to a hybrid vaccine increased the cytotoxic activity of splenocytes toward B16F10 in vitro and avoided late tumor progression in a pulmonary colonization model. These results revealed the adjuvant effect of a killed P. acnes suspension, as it improved specific humoral and cellular immune responses elicited by DC-tumor cell hybrid vaccines.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines/immunology , Dendritic Cells/immunology , Immunogenicity, Vaccine , Melanoma, Experimental/immunology , Propionibacterium acnes/immunology , Animals , Antigens, Neoplasm/immunology , Cells, Cultured , Chemokine CCL1/immunology , Disease Progression , Female , Immunoglobulin G/metabolism , Interferon-gamma/metabolism , Lymph Nodes/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice, Inbred C57BL , Spleen/immunology , Tumor Burden , Vaccines, Inactivated
7.
Front Microbiol ; 9: 1427, 2018.
Article in English | MEDLINE | ID: mdl-30018604

ABSTRACT

The present study focused on the activity of the palladacycle complex DPPE 1.1 on Leishmania (Leishmania) amazonensis. Promastigotes of L. (L.) amazonensis were destroyed in vitro by nanomolar concentrations of DPPE 1.1, whereas intracellular amastigotes were killed at drug concentrations fivefold less toxic than those harmful to macrophages. L. (L.) amazonensis-infected BALB/c mice were treated by intralesional injection of DPPE 1.1. Animals treated with 3.5 and 7.0 mg/kg of DPPE 1.1 showed a significant decrease of foot lesion sizes and a parasite load reduction of 93 and 99%, respectively, when compared to untreated controls. Furthermore, DPPE 1.1 was non-toxic to treated animals. The cathepsin B activity of L. (L.) amazonensis amastigotes was inhibited by DPPE 1.1 as demonstrated spectrofluorometrically by use of a specific fluorogenic substrate. Analysis of T-cells populations in mice treated with DPPE 1.1 and untreated controls was performed by fluorescence-activated cell sorter (FACS). IFN-γ was measured in supernatants of lymphocytes from popliteal and inguinal lymph nodes isolated from treated and untreated mice and stimulated with L. (L.) amazonensis amastigotes extract and active TGF-ß was evaluated in supernatants of foot lesions; both dosages were carried out by means of a double-sandwich ELISA assay. A significant increase of TCD4+ and TCD8+ lymphocytes and IFN-γ secretion was displayed in mice treated with DPPE 1.1 compared to untreated animals, whereas a significant reduction of active TGF-ß was observed in treated mice. These findings open perspectives for further investment in DPPE 1.1 as an alternative option for the chemotherapy of cutaneous leishmaniasis.

8.
J Immunol ; 198(7): 2772-2784, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28258198

ABSTRACT

Vaccine strategies to enhance CD8+ CTL responses remain a current challenge because they should overcome the plasmatic and endosomal membranes for favoring exogenous Ag access to the cytosol of APCs. As a way to avoid this hurdle, sticholysin (St) II, a pore-forming protein from the Caribbean Sea anemone Stichodactyla helianthus, was encapsulated with OVA into liposomes (Lp/OVA/StII) to assess their efficacy to induce a CTL response. OVA-specific CD8+ T cells transferred to mice immunized with Lp/OVA/StII experienced a greater expansion than when the recipients were injected with the vesicles without St, mostly exhibiting a memory phenotype. Consequently, Lp/OVA/StII induced a more potent effector function, as shown by CTLs, in vivo assays. Furthermore, treatment of E.G7-OVA tumor-bearing mice with Lp/OVA/StII significantly reduced tumor growth being more noticeable in the preventive assay. The contribution of CD4+ and CD8+ T cells to CTL and antitumor activity, respectively, was elucidated. Interestingly, the irreversibly inactive variant of the StI mutant StI W111C, encapsulated with OVA into Lp, elicited a similar OVA-specific CTL response to that observed with Lp/OVA/StII or vesicles encapsulating recombinant StI or the reversibly inactive StI W111C dimer. These findings suggest the relative independence between StII pore-forming activity and its immunomodulatory properties. In addition, StII-induced in vitro maturation of dendritic cells might be supporting these properties. These results are the first evidence, to our knowledge, that StII, a pore-forming protein from a marine eukaryotic organism, encapsulated into Lp functions as an adjuvant to induce a robust specific CTL response.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/immunology , Cnidarian Venoms/administration & dosage , Neoplasms, Experimental/pathology , T-Lymphocytes, Cytotoxic/drug effects , Animals , Cnidarian Venoms/immunology , Female , Flow Cytometry , Liposomes/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology
9.
Front Microbiol ; 8: 333, 2017.
Article in English | MEDLINE | ID: mdl-28321209

ABSTRACT

Palladacycle complex DPPE 1.2 was previously reported to inhibit the in vitro and in vivo infection by Leishmania (Leishmania) amazonensis. The aim of the present study was to compare the effect of DPPE 1.2, in association with heat-killed Propionibacterium acnes, on L. (L.) amazonensis infection in two mouse strains, BALB/c and C57BL/6, and to evaluate the immune responses of the treated animals. Foot lesions of L. (L.) amazonensis-infected mice were injected with DPPE 1.2 alone, or associated with P. acnes as an adjuvant. Analysis of T-cell populations in the treated mice and in untreated controls was performed by FACS. Detection of IFN-γ-secreting lymphocytes was carried out by an ELISPOT assay and active TGF-ß was measured by means of a double-sandwich ELISA test. The treatment with DPPE 1.2 resulted in a significant reduction of foot lesion sizes and parasite burdens in both mouse strains, and the lowest parasite burden was found in mice treated with DPPE 1.2 plus P. acnes. Mice treated with DPPE 1.2 alone displayed a significant increase of TCD4+ and TCD8+ lymphocytes and IFN-γ secretion which were significantly higher in animals treated with DPPE 1.2 plus P. acnes. A significant reduction of active TGF-ß was observed in mice treated with DPPE 1.2 alone or associated with P. acnes. Moreover, DPPE 1.2 associated to P. acnes was non-toxic to treated animals. The destruction of L. (L.) amazonensis by DPPE 1.2 was followed by host inflammatory responses which were exacerbated when the palladacycle complex was associated with P. acnes.

10.
Front Immunol ; 7: 138, 2016.
Article in English | MEDLINE | ID: mdl-27148259

ABSTRACT

The role of B cells as antigen-presenting cells (APCs) has been extensively studied, mainly in relation to the activation of memory T cells. Considering the B cell subtypes, the role of B-1 cells as APCs is beginning to be explored. Initially, it was described that B-1 cells are activated preferentially by T-independent antigens. However, some reports demonstrated that these cells are also involved in a T-dependent response. The aim of this review is to summarize information about the ability of B-1 cells to play a role as APCs and to briefly discuss the role of the BCR and toll-like receptor signals in this process. Furthermore, some characteristics of B-1 cells, such as natural IgM production and phagocytic ability, could interfere in the participation of these cells in the onset of an adaptive response.

11.
Immunobiology ; 217(9): 831-41, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22257707

ABSTRACT

The pathogenesis of focal segmental glomerulosclerosis (FSGS) appears to be associated with type-2 cytokines and podocyte dysfunction. In this study, we tested the hypothesis that immunization with the polysaccharide fraction of Propionibacterium acnes (PS), a pro-Th1 agonist, may subvert the type-2 profile and protect podocytes from adriamycin-induced glomerulosclerosis. Adriamycin injection resulted in albuminuria and increased serum creatinine in association with loss of glomerular podocin and podoplanin expression, which is consistent with podocyte dysfunction. Renal tissue analysis revealed the expression of transcripts for GATA3 and fibrogenic-related proteins, such as TGF-ß, tissue inhibitor of metalloproteinase-1 (TIMP-1) and metalloproteinase 9 (MMP9). In association with the expression of fibrogenic transcripts, we observed peri-glomerular expression of α-smooth muscle actin (α-SMA), indicating epithelial-to-mesenchymal transition, and increased expression of proliferating cell nuclear antigen (PCNA) in tubular cells, suggesting intense proliferative activity. Previous immunization with PS inhibited albuminuria and serum creatinine in association with the preservation of podocyte proteins and inhibition of fibrogenic transcripts and the expression of α-SMA and PCNA proteins. Tissue analysis also revealed that PS treatment induced expression of mRNA for GD3 synthase, which is a glycosiltransferase related to the synthesis of GD3, a ganglioside associated with podocyte physiology. In addition, PS treatment inhibited the influx of inflammatory CD8(pos) and CD11b(pos) cells to kidney tissue. Finally, PS treatment on day 4 post-ADM, a period when proteinuria was already established, was able to improve renal function. Thus, we demonstrate that the PS fraction of P. acnes can inhibit FSGS pathogenesis, suggesting that immunomodulation can represent an alternative approach for disease management.


Subject(s)
Glomerulosclerosis, Focal Segmental/etiology , Polysaccharides, Bacterial/pharmacology , Propionibacterium acnes/chemistry , Protective Agents/pharmacology , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Doxorubicin/adverse effects , Epithelial-Mesenchymal Transition/drug effects , Fibrosis/genetics , Gene Expression Regulation/drug effects , Glomerulosclerosis, Focal Segmental/drug therapy , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred BALB C , Polysaccharides, Bacterial/administration & dosage , Polysaccharides, Bacterial/isolation & purification , Protective Agents/administration & dosage , Protective Agents/isolation & purification , Proteinuria/drug therapy , Renal Insufficiency/chemically induced , Renal Insufficiency/drug therapy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...