Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biomolecules ; 11(11)2021 11 04.
Article in English | MEDLINE | ID: mdl-34827634

ABSTRACT

Yersinia enterocolitica (Ye) inserts outer proteins (Yops) into cytoplasm to infect host cells. However, in spite of considerable progress, the mechanisms implicated in this process, including the association of Yops with host proteins, remain unclear. Here, we evaluated the functional role of Galectin-1 (Gal1), an endogenous ß-galactoside-binding protein, in modulating Yop interactions with host cells. Our results showed that Gal1 binds to Yops in a carbohydrate-dependent manner. Interestingly, Gal1 binding to Yops protects these virulence factors from trypsin digestion. Given that early control of Ye infection involves activation of macrophages, we evaluated the role of Gal1 and YopP in the modulation of macrophage function. Although Gal1 and YopP did not influence production of superoxide anion and/or TNF by Ye-infected macrophages, they coordinately inhibited nitric oxide (NO) production. Notably, recombinant Gal1 (rGal1) did not rescue NO increase observed in Lgals1-/- macrophages infected with the YopP mutant Ye ∆yopP. Whereas NO induced apoptosis in macrophages, no significant differences in cell death were detected between Gal1-deficient macrophages infected with Ye ∆yopP, and WT macrophages infected with Ye wt. Strikingly, increased NO production was found in WT macrophages treated with MAPK inhibitors and infected with Ye wt. Finally, rGal1 administration did not reverse the protective effect in Peyer Patches (PPs) of Lgals1-/- mice infected with Ye ∆yopP. Our study reveals a cooperative role of YopP and endogenous Gal1 during Ye infection.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Galectin 1/metabolism , Immunity , Nitric Oxide/biosynthesis , Yersinia enterocolitica/immunology , Amino Acid Sequence , Animals , Bacterial Outer Membrane Proteins/chemistry , Mice, Inbred C57BL , Mice, Knockout , Peptide Hydrolases/metabolism , Protein Binding , Proteolysis , Proteomics , Virulence Factors/metabolism , Yersinia enterocolitica/pathogenicity
2.
Cancers (Basel) ; 13(5)2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33804419

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is a heterogeneous group of tumors usually diagnosed at an advanced stage and characterized by a poor prognosis. The main risk factors associated with its development include tobacco and alcohol consumption and Human Papillomavirus (HPV) infections. The immune system has a significant role in the oncogenesis and evolution of this cancer type. Notably, the immunosuppressive tumor microenvironment triggers immune escape through several mechanisms. The improved understanding of the antitumor immune response in solid tumors and the role of the immune checkpoint molecules and other immune regulators have led to the development of novel therapeutic strategies that revolutionized the clinical management of HNSCC. However, the limited overall response rate to immunotherapy urges identifying predictive biomarkers of response and resistance to treatment. Here, we review the role of the immune system and immune checkpoint pathways in HNSCC, the most relevant clinical findings linked to immunotherapeutic strategies and predictive biomarkers of response and future treatment perspectives.

3.
Front Pharmacol ; 11: 901, 2020.
Article in English | MEDLINE | ID: mdl-32625094

ABSTRACT

During the resolution of acute inflammation, macrophages undergo reprogramming from pro-inflammatory, to anti-inflammatory/reparative, and eventually to pro-resolving macrophages. Galectin-1 (Gal-1) is a bona fide pro-resolving lectin while interferon ß (IFN-ß) was recently shown to facilitate macrophage reprogramming and resolution of inflammation. In this study, we found Gal-1null mice exhibit a hyperinflammatory phenotype during the resolution of zymosan A-induced peritonitis but not during the early inflammatory response. This phenotype was characterized by reduced macrophage numbers, increased secretion of pro-inflammatory cytokines, such as interleukin-12 (IL-12), and reduced secretion of anti-inflammatory cytokines, such as interleukin-10 (IL-10). In addition, we found a delayed expression of the pro-resolving enzyme 12/15-lipoxygenase in macrophages and heightened levels of the inflammatory protease proteinase-3 (PR3) in peritoneal fluids from Gal-1null mice. Moreover, we observed sex-dependent differences in the inflammatory profile of Gal-1null mice. Notably, we found that IFN-ß levels were reduced in resolution-phase exudates from Gal-1null mice. Administration of IFN-ß in vivo or ex vivo treatment was able to rescue, at least in part, the hyperinflammatory profile of Gal-1null mice. In particular, IFN-ß recovered a subset of F4/80+GR-1+ macrophages, restored IL-12 and IL-10 secretion from macrophages to WT values and diminished abnormal peritoneal PR3 levels in Gal-1null mice. In conclusion, our results revealed a new Gal-1-IFN-ß axis that facilitates the resolution of inflammation and might restrain uncontrolled inflammatory disorders.

4.
Cell Physiol Biochem ; 52(2): 354-367, 2019.
Article in English | MEDLINE | ID: mdl-30816679

ABSTRACT

BACKGROUND/AIMS: Although a cross-talk between immune and endocrine systems has been well established, the precise pathways by which these signals co-regulate pro- and antiinflammatory responses on antigen-presenting cells remain poorly understood. In this work we investigated the mechanisms by which triiodothyronine (T3) controls T cell activity via dendritic cell (DC) modulation. METHODS: DCs from wild-type (WT) and IL-6-deficient mice were pulsed with T3. Cytokine production and programmed death protein ligands (PD-L) 1 and 2 expression were assayed by flow cytometry and ELISA. Interferon-regulatory factor-4 (IRF4) expression was evaluated by RT-qPCR and flow cytometry. The ability of DCs to stimulate allogenic splenocytes was assessed in a mixed lymphocyte reaction and the different profile markers were analyzed by flow cytometry and ELISA. For in vivo experiments, DCs treated with ovalbumin and T3 were injected into OTII mice. Proliferation, cytokine production, frequency of FoxP3+ regulatory T (Treg) cells and PD-1+ cells were determined by MTT assay, ELISA and flow cytometry, respectively. RESULTS: T3 endows DCs with pro-inflammatory potential capable of generating IL-17-dominant responses and down-modulating expression of PD-L1 and 2. T3-stimulated WT-DCs increased the proportion of IL-17-producing splenocytes, an effect which was eliminated when splenocytes were incubated with T3-treated DCs derived from IL-6-deficient mice. Enhanced IL-17 expression was recorded in both, CD4- and CD4+ populations and involved the IRF-4 pathway. Particularly, γδ-T cells but not natural killer (NK), NKT, B lymphocytes nor CD8+ T cells were the major source of IL-17-production from CD4- cells. Moreover, T3-conditioned DCs promoted a decrease of the FoxP3+ Treg population. Furthermore, T3 down-modulated PD-1 expression on CD4- cells thereby limiting inhibitory signals driven by this co-inhibitory pathway. Thus, T3 acts at the DC level to drive proinflammatory responses in vitro. Accordingly, we found that T3 induces IL-17 and IFNγ-dominant antigen-specific responses in vivo. CONCLUSION: These results emphasize the relevance of T3 as an additional immune-endocrine checkpoint and a novel therapeutic target to modulate IL-17-mediated pro-inflammatory responses.


Subject(s)
Dendritic Cells/immunology , Interleukin-17/immunology , Signal Transduction/drug effects , Triiodothyronine/pharmacology , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Dendritic Cells/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interleukin-17/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Mice , Mice, Knockout , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Signal Transduction/immunology
5.
BMC Immunol ; 18(1): 8, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28166724

ABSTRACT

BACKGROUND: Hyperactive secretion and pathogenic effects of interleukin (IL)-17 and IgA have been detected in different arthropathies. Recent evidence has revealed that TH17 cytokines regulate mucosal IgA secretion. However, it is unknown whether and how IL-17 mediates synovial IgA production. Here we aim to investigate the connection of synovial IL-17 with IgA production in the joint. In this study we included synovial fluids (SF) from patients with rheumatoid arthritis (RA; n = 66), spondyloarthritis (SpA; n = 18) and osteoarthritis (OA; n = 36). The levels of IL-17, IL-6, transforming growth factor (TGF)-ß1, B-cell-activating factor of the TNF family (BAFF) and anti-lipopolyssacharide (LPS) immunoglobulin (Ig)A were investigated by enzyme-linked immunosorbent assay (ELISA). Total IgA was measured by radial immunodiffusion assay. Synovial fluid-derived mononuclear cells (SFMC) were stimulated with bacterial antigens or SF-conditioned media, and cytokines and IgA were analyzed in the supernatants. RESULTS: IL-17, IL-6 and TGF-ß1 were increased in SF from both RA and SpA compared with OA patients. Concentration of IL-17 correlated with the disease activity score (DAS)-28, IL-6 and anti-LPS IgA levels. Bacterial-stimulated SFMCs from RA and SpA patients secreted higher IL-17 than vehicle-stimulated SFMCs. Conditioned media with SF containing IL-17 induced anti-LPS IgA production by SFMCs which was independent of IL-6 activity. Concentrations of synovial TGF-ß1 and BAFF correlated with anti-LPS and total IgA levels, respectively. Blockade of IL-17 decreased the production of TGF-ß1 and anti-LPS IgA by SF-stimulated SFMCs. CONCLUSIONS: This study reports a connection between IL-17 and IgA secretion in the joint. In addition, it demonstrates that enterobacterial antigens trigger synovial IL-17 production, and that TGF-ß1 and BAFF may mediate the effect of IL-17 on IgA production. This circuit may contribute to the pathogenesis of inflammatory joint diseases.


Subject(s)
Enterobacteriaceae Infections/immunology , Inflammation/immunology , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Joints/immunology , Spondylitis, Ankylosing/immunology , Th17 Cells/immunology , Adolescent , Adult , Aged , Aged, 80 and over , B-Cell Activating Factor/metabolism , Child , Female , Humans , Immunoglobulin A/metabolism , Interleukin-17/immunology , Intestinal Mucosa/microbiology , Male , Middle Aged , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism , Young Adult
6.
PLoS One ; 9(4): e96402, 2014.
Article in English | MEDLINE | ID: mdl-24788652

ABSTRACT

Platelets contribute to vessel formation through the release of angiogenesis-modulating factors stored in their α-granules. Galectins, a family of lectins that bind ß-galactoside residues, are up-regulated in inflammatory and cancerous tissues, trigger platelet activation and mediate vascularization processes. Here we aimed to elucidate whether the release of platelet-derived proangiogenic molecules could represent an alternative mechanism through which galectins promote neovascularization. We show that different members of the galectin family can selectively regulate the release of angiogenic molecules by human platelets. Whereas Galectin (Gal)-1, -3, and -8 triggered vascular endothelial growth factor (VEGF) release, only Gal-8 induced endostatin secretion. Release of VEGF induced by Gal-8 was partially prevented by COX-1, PKC, p38 and Src kinases inhibitors, whereas Gal-1-induced VEGF secretion was inhibited by PKC and ERK blockade, and Gal-3 triggered VEGF release selectively through a PKC-dependent pathway. Regarding endostatin, Gal-8 failed to stimulate its release in the presence of PKC, Src and ERK inhibitors, whereas aspirin or p38 inhibitor had no effect on endostatin release. Despite VEGF or endostatin secretion, platelet releasates generated by stimulation with each galectin stimulated angiogenic responses in vitro including endothelial cell proliferation and tubulogenesis. The platelet angiogenic activity was independent of VEGF and was attributed to the concerted action of other proangiogenic molecules distinctly released by each galectin. Thus, secretion of platelet-derived angiogenic molecules may represent an alternative mechanism by which galectins promote angiogenic responses and its selective blockade may lead to the development of therapeutic strategies for angiogenesis-related diseases.


Subject(s)
Blood Platelets/metabolism , Galectin 1/metabolism , Galectin 3/metabolism , Galectins/metabolism , Neovascularization, Physiologic , Cell Line , Endostatins/metabolism , Humans , Vascular Endothelial Growth Factor A/metabolism
7.
Cancer Immunol Immunother ; 62(12): 1781-95, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24114144

ABSTRACT

The progesterone analog medroxyprogesterone acetate (MPA) is widely used as a hormone replacement therapy in postmenopausal women and as contraceptive. However, prolonged administration of MPA is associated with increased incidence of breast cancer through ill-defined mechanisms. Here, we explored whether exposure to MPA during mammary tumor growth affects myeloid-derived suppressor cells (MDSCs; CD11b(+)Gr-1(+), mostly CD11b(+)Ly6G(+)Ly6C(int) and CD11b(+)Ly6G(-)Ly6C(high) cells) and natural killer (NK) cells, potentially restraining tumor immunosurveillance. We used the highly metastatic 4T1 breast tumor (which does not express the classical progesterone receptor and expands MDSCs) to challenge BALB/c mice in the absence or in the presence of MPA. We observed that MPA promoted the accumulation of NK cells in spleens of tumor-bearing mice, but with reduced degranulation ability and in vivo cytotoxic activity. Simultaneously, MPA induced a preferential expansion of CD11b(+)Ly6G(+)Ly6C(int) cells in spleen and bone marrow of 4T1 tumor-bearing mice. In vitro, MPA promoted nuclear mobilization of the glucocorticoid receptor (GR) in 4T1 cells and endowed these cells with the ability to promote a preferential differentiation of bone marrow cells into CD11b(+)Ly6G(+)Ly6C(int) cells that displayed suppressive activity on NK cell degranulation. Sorted CD11b(+)Gr-1(+) cells from MPA-treated tumor-bearing mice exhibited higher suppressive activity on NK cell degranulation than CD11b(+)Gr-1(+) cells from vehicle-treated tumor-bearing mice. Thus, MPA, acting through the GR, endows tumor cells with an enhanced capacity to expand CD11b(+)Ly6G(+)Ly6C(int) cells that subsequently display a stronger suppression of NK cell-mediated anti-tumor immunity. Our results describe an alternative mechanism by which MPA may affect immunosurveillance and have potential implication in breast cancer incidence.


Subject(s)
Antigens, Ly/immunology , Breast Neoplasms/immunology , CD11b Antigen/immunology , Killer Cells, Natural/immunology , Medroxyprogesterone Acetate/pharmacology , Myeloid Cells/immunology , Animals , Antigens, Ly/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , CD11b Antigen/metabolism , Cell Differentiation , Cell Proliferation , Cytotoxicity, Immunologic , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Receptors, Glucocorticoid/metabolism , STAT3 Transcription Factor/metabolism , Tumor Cells, Cultured
8.
IUBMB Life ; 63(7): 521-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21698756

ABSTRACT

Platelet activation at sites of vascular injury leads to the formation of a hemostatic plug. Activation of platelets is therefore crucial for normal hemostasis. However, uncontrolled platelet activation may also lead to the formation of occlusive thrombi that can cause ischemic events. Platelets can be activated by soluble molecules including thrombin, TXA2 , adenosine diphosphate (ADP), and serotonin or by adhesive extracellular matrix (ECM) proteins such as von Willebrand factor and collagen. In this article, we review recent advances on the role of galectins in platelet physiology. By acting in either soluble or immobilized form, these glycan-binding proteins trigger platelet activation through modulation of discrete signaling pathways. We also offer new hypotheses and some speculations about the role of platelet-galectin interactions not only in hemostasis and thrombosis but also in inflammation and related diseases such as atherosclerosis and cancer.


Subject(s)
Blood Platelets/physiology , Galectins/metabolism , Platelet Activation/physiology , Signal Transduction/physiology , Animals , Blood Coagulation/physiology , Hemostasis/physiology , Humans , Neoplasms/metabolism , Neoplasms/pathology , Thrombosis/physiopathology
9.
Am J Pathol ; 170(2): 546-56, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17255323

ABSTRACT

During acute infection with Trypanosoma cruzi, the causative agent of Chagas' disease, the thymus undergoes intense atrophy followed by a premature escape of CD4+CD8+ immature cortical thymocytes. Here we report a pivotal role for the endogenous lectin galectin-3 in accelerating death of thymocytes and migration of these cells away from the thymus after T. cruzi infection. We observed a pronounced increase in galectin-3 expression that paralleled the extensive depletion of CD4+CD8+ immature thymocytes after infection. In vitro, recombinant galectin-3 induced increased levels of death in cortical immature thymocytes. Consistent with the role of galectin-3 in promoting cell death, thymuses from gal-3-/- mice did not show cortical thymocyte depletion after parasite infection in vivo. In addition, galectin-3 accelerated laminin-driven CD4+CD8+ thymocyte migration in vitro and in vivo induced exportation of CD4+CD8+ cells from the thymus to the peripheral compartment. Our findings provide evidence of a novel role for galectin-3 in the regulation of thymus physiology and identify a potential mechanism based on protein-glycan interactions in thymic atrophy associated with acute T. cruzi infection.


Subject(s)
CD4 Antigens/metabolism , CD8 Antigens/metabolism , Chagas Disease/metabolism , Galectin 3/metabolism , Thymus Gland/metabolism , Trypanosoma cruzi , Animals , Atrophy/genetics , Atrophy/metabolism , Atrophy/pathology , Cell Death/genetics , Cell Differentiation/genetics , Chagas Disease/genetics , Galectin 3/deficiency , Gene Expression Regulation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Thymus Gland/parasitology , Thymus Gland/pathology
10.
Medicina (B.Aires) ; 61(1): 85-92, 2001. ilus, tab
Article in Spanish | LILACS | ID: lil-286387

ABSTRACT

Las galectinas constituyen una familia de proteínas extremadamente conservada a través de la evolución. En función de su propiedad de decifrar glicocódigos específicos, estas proteínas han sido involucradas en un amplio espectro de eventos biológicos. Recientes avances han demostrados que estas proteínas han juegan un rol fundamental en procesos relacionados a la regulación de la respuesta inmune, tales como adhesión linfocitaria, crecimiento celular, producción de citoquinas y regulación de la muerte celular progamada. En el presente artículo se analizan las implicancias de estas familias de proteínas en desórdenes autoinmunes, inflamación aguda y crónica, transtornos alérgicos, infecciones y enfermedades neoplásicas. La utilización de estas proteínas endógenas y sus antagonistas en el diagnóstico y tratamiento de estas patologías abre un nuevo horizonte en el campo de la inmunopatología molecular.


Subject(s)
Humans , Hemagglutinins/immunology , Adjuvants, Immunologic/physiology , Hemagglutinins/physiology
11.
Medicina [B.Aires] ; 61(1): 85-92, 2001. ilus, tab
Article in Spanish | BINACIS | ID: bin-10508

ABSTRACT

Las galectinas constituyen una familia de proteínas extremadamente conservada a través de la evolución. En función de su propiedad de decifrar glicocódigos específicos, estas proteínas han sido involucradas en un amplio espectro de eventos biológicos. Recientes avances han demostrados que estas proteínas han juegan un rol fundamental en procesos relacionados a la regulación de la respuesta inmune, tales como adhesión linfocitaria, crecimiento celular, producción de citoquinas y regulación de la muerte celular progamada. En el presente artículo se analizan las implicancias de estas familias de proteínas en desórdenes autoinmunes, inflamación aguda y crónica, transtornos alérgicos, infecciones y enfermedades neoplásicas. La utilización de estas proteínas endógenas y sus antagonistas en el diagnóstico y tratamiento de estas patologías abre un nuevo horizonte en el campo de la inmunopatología molecular. (AU)


Subject(s)
Humans , Hemagglutinins/immunology , Hemagglutinins/physiology , Adjuvants, Immunologic/physiology
13.
Mem. Inst. Oswaldo Cruz ; 95(supl.1): 225-33, 2000. tab
Article in English | LILACS | ID: lil-274885

ABSTRACT

Rheumatoid arthritis (RA) is characterized by chronic inflammation of the synovial joints resulting from hyperplasia of synovial fibroblasts and infiltration of lymphocytes, macrophages and plasma cells, all of which manifest signs of activation. All these cells proliferate abnormally, invade bone and cartilage, produce an elevated amount of pro-inflammatory cytokines, metalloproteinases and trigger osteoclast formation and activation. Some of the pathophysiological consequences of the disease may be explained by the inadequate apoptosis, which may promote the survival of autoreactive T cells, macrophages or synovial fibroblasts. Although RA does not result from single genetic mutations, elucidation of the molecular mechanisms implicated in joint destruction has revealed novel targets for gene therapy. Gene transfer strategies include inhibition of pro-inflammatory cytokines, blockade of cartilage-degrading metalloproteinases, inhibition of synovial cell activation and manipulation of the Th1-Th2 cytokine balance. Recent findings have iluminated the idea that induction of apoptosis in the rheumatoid joint can be also used to gain therapeutic advantage in the disease. In the present review we will discuss different strategies used for gene transfer in RA and chronic inflammation. Particularly, we will high-light the importance of programmed cell death as a novel target for gene therapy using endogenous biological mediators, such as galectin-1, a beta-galactoside-binding protein that induces apoptosis of activated T cells and immature thymocytes


Subject(s)
Humans , Apoptosis/immunology , Arthritis, Rheumatoid/therapy , Biomarkers , Cytokines/immunology , Genetic Therapy , Hemagglutinins , Arthritis, Rheumatoid/immunology , Gene Transfer Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...