Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomed Pharmacother ; 172: 116283, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38377735

RESUMEN

BACKGROUND: Galectins (Gal's) are a family of carbohydrate-binding proteins that are known to support the tumour microenvironment through their immunosuppressive activity and ability to promote metastasis. As such they are attractive therapeutic targets, but little is known about the cellular expression pattern of galectins within the tumour and its neighbouring stromal microenvironment. Here we investigated the cellular expression pattern of Gals within pancreatic ductal adenocarcinoma (PDAC). METHODS: Galectin gene and protein expression were analysed by scRNAseq (n=4) and immunofluorescence imaging (n=19) in fibroblasts and epithelial cells of pancreatic biopsies from PDAC patients. Galectin surface expression was also assessed on tumour adjacent normal fibroblasts and cancer associated primary fibroblasts from PDAC biopsies using flow cytometry. RESULTS: scRNAseq revealed higher Gal-1 expression in fibroblasts and higher Gal-3 and -4 expression in epithelial cells. Both podoplanin (PDPN+, stromal/fibroblast) cells and EpCAM+ epithelial cells expressed Gal-1 protein, with highest expression seen in the stromal compartment. By contrast, significantly more Gal-3 and -4 protein was expressed in ductal cells expressing either EpCAM or PDPN, when compared to the stroma. Ductal Gal-4 cellular expression negatively correlated with ductal Gal-1, but not Gal-3 expression. Higher ductal cellular expression of Gal-1 correlated with smaller tumour size and better patient survival. CONCLUSIONS: In summary, the intricate interplay and cell-specific expression patterns of galectins within the PDAC tissue, particularly the inverse correlation between Gal-1 and Gal-4 in ducts and its significant association with patient survival, highlights the complex molecular landscape underlying PDAC and provides valuable insights for future therapeutic interventions.


Asunto(s)
Benzamidas , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Tirosina/análogos & derivados , Humanos , Molécula de Adhesión Celular Epitelial , Neoplasias Pancreáticas/genética , Carcinoma Ductal Pancreático/genética , Factores de Transcripción , Galectinas/genética , Microambiente Tumoral
2.
Nat Metab ; 5(11): 1969-1985, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37884694

RESUMEN

T cell activation is associated with a profound and rapid metabolic response to meet increased energy demands for cell division, differentiation and development of effector function. Glucose uptake and engagement of the glycolytic pathway are major checkpoints for this event. Here we show that the low-affinity, concentration-dependent glucose transporter 2 (Glut2) regulates the development of CD8+ T cell effector responses in mice by promoting glucose uptake, glycolysis and glucose storage. Expression of Glut2 is modulated by environmental factors including glucose and oxygen availability and extracellular acidification. Glut2 is highly expressed by circulating, recently primed T cells, allowing efficient glucose uptake and storage. In glucose-deprived inflammatory environments, Glut2 becomes downregulated, thus preventing passive loss of intracellular glucose. Mechanistically, Glut2 expression is regulated by a combination of molecular interactions involving hypoxia-inducible factor-1 alpha, galectin-9 and stomatin. Finally, we show that human T cells also rely on this glucose transporter, thus providing a potential target for therapeutic immunomodulation.


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa , Glucosa , Ratones , Humanos , Animales , Glucosa/metabolismo , Transporte Biológico/fisiología , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Diferenciación Celular , Linfocitos T CD8-positivos/metabolismo
3.
Front Immunol ; 14: 1226196, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37622117

RESUMEN

Background: The NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome has been described in both immune cells and platelets, but its role in the megakaryocyte (MK) lineage remains elusive. Objective: The aim of this study was to explore the role of NLRP3 inflammasome in megakaryocytes and platelets. Methods: We generated Nlrp3 A350V/+/Gp1ba-CreKI/+ mice carrying a mutation genetically similar to the one observed in human Muckle-Wells syndrome, which leads to hyperactivity of NLRP3 specifically in MK and platelets. Results: Platelets from the mutant mice expressed elevated levels of both precursor and active form of caspase-1, suggesting hyperactivity of NLRP3 inflammasome. Nlrp3 A350V/+/Gp1ba-CreKI/+ mice developed normally and had normal platelet counts. Expression of major platelet receptors, platelet aggregation, platelet deposition on collagen under shear, and deep vein thrombosis were unchanged. Nlrp3 A350V/+/Gp1ba-CreKI/+ mice had mild anemia, reduced Ter119+ cells in the bone marrow, and splenomegaly. A mild increase in MK TGF-ß1 might be involved in the anemic phenotype. Intraperitoneal injection of zymosan in Nlrp3 A350V/+/Gp1ba-CreKI/+ mice induced increased neutrophil egression and elevated levels of a set of proinflammatory cytokines, alongside IL-10 and G-CSF, in the peritoneal fluid as compared with control animals. Conclusion: MK/platelet NLRP3 inflammasome promotes the acute inflammatory response and its hyperactivation in mice leads to mild anemia and increased extramedullary erythropoiesis.


Asunto(s)
Anemia , Megacariocitos , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Ratones , Inflamasomas , Ratones Endogámicos NOD , Proteína con Dominio Pirina 3 de la Familia NLR/genética
4.
J Leukoc Biol ; 114(4): 301-314, 2023 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-37309034

RESUMEN

Aging is associated with exacerbated systemic inflammation (inflammaging) and the progressive loss of immune system function (immunosenescence). Leukocyte migration is necessary for effective immunity; however, dysregulated trafficking of leukocytes into tissue contributes to inflammaging and the development of age-related inflammatory diseases. Aging modulates leukocyte trafficking under inflammatory conditions; however, whether aging modulates leukocyte trafficking under homeostatic conditions remains to be elucidated. Although immune responses are evidently sexually dimorphic, limited studies have investigated the effect of sex on age-related changes to leukocyte trafficking processes. Here, we investigated age-related and sex-specific changes to the leukocyte populations within the peritoneal cavity of young (3-mo), middle-aged (18-mo) and old (21-mo) male and female wild-type mice in the steady state. We found an age-related increase in the number of leukocytes within the peritoneal cavity of female mice, predominantly B cells, which may reflect increased trafficking through this tissue with age. This was accompanied by an increased inflammatory environment within the aged cavity, including increased levels of chemoattractants, including B cell chemoattractants CXCL13 and CCL21, soluble adhesion molecules, and proinflammatory cytokines, which was more pronounced in aged female mice. Intravital microscopy techniques revealed altered vascular structure and increased vascular permeability within the peritoneal membrane of aged female mice, which may support increased leukocyte trafficking to the cavity with age. Together, these data indicate that aging affects homeostatic leukocyte trafficking processes in a sex-specific fashion.


Asunto(s)
Leucocitos , Cavidad Peritoneal , Masculino , Femenino , Animales , Ratones , Inflamación , Peritoneo , Factores Quimiotácticos
5.
Clin Exp Immunol ; 212(1): 1-10, 2023 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-36891817

RESUMEN

Dysregulation of leukocyte trafficking, lipid metabolism, and other metabolic processes are the hallmarks that underpin and drive pathology in obesity. Current clinical management targets alternations in lifestyle choices (e.g. exercise, weight loss) to limit the impact of the disease. Crucially, re-gaining control over the pathogenic cellular and molecular processes may offer an alternative, complementary strategy for obese patients. Here we investigate the impact of the immunopeptide, PEPITEM, on pancreas homeostasis and leukocyte trafficking in mice on high-fed obesogenic diet (HFD). Both prophylactic and therapeutic treatment with PEPITEM alleviated the effects of HFD on the pancreas, reducing pancreatic beta cell size. Moreover, PEPITEM treatment also limited T-cell trafficking (CD4+ T-cells and KLRG1+ CD3+ T-cells) to obese visceral, but not subcutaneous, adipose tissue. Similarly, PEPITEM treatment reduced macrophage numbers within the peritoneal cavity of mice on HFD diet at both 6 and 12 weeks. By contrast, PEPITEM therapy elevated numbers of T and B cells were observed in the secondary lymphoid tissues (e.g. spleen and inguinal lymph node) when compared to the untreated HFD controls. Collectively our data highlights the potential for PEPITEM as a novel therapy to combat the systemic low-grade inflammation experienced in obesity and minimize the impact of obesity on pancreatic homeostasis. Thus, offering an alternative strategy to reduce the risk of developing obesity-related co-morbidities, such as type 2 diabetes mellitus, in individuals at high risk and struggling to control their weight through lifestyle modifications.


Asunto(s)
Diabetes Mellitus Tipo 2 , Ratones , Animales , Diabetes Mellitus Tipo 2/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/patología , Inflamación/patología , Dieta , Linfocitos T CD4-Positivos/metabolismo , Ratones Endogámicos C57BL , Tejido Adiposo
6.
Atherosclerosis ; 363: 57-68, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36459823

RESUMEN

BACKGROUND AND AIMS: Atherosclerosis is widely accepted to be an inflammatory disease driven by lipid accumulation and leukocyte recruitment. More recently, galectins, a family of ß-galactoside binding proteins, have been shown to play a role in leukocyte recruitment among other immunomodulatory functions. Galectin (Gal) -9, a tandem repeat type galectin expressed by the endothelium in inflammatory environments, has been proposed to promote leukocyte recruitment. However, the role of Gal-9 in the context of monocyte recruitment remains elusive. METHODS AND RESULTS: Here, we characterise the immunomodulatory role of Gal-9 in context of atherosclerosis. We show that ApoE-/-Gal-9-/- mice have a significantly reduced aortic plaque burden compared to their ApoE-/- littermate controls after 12 weeks of high fat diet. RNA sequencing data from two independent studies reveal Lgals9 expression in leukocyte clusters isolated from murine atherosclerotic plaques. Additionally, soluble Gal-9 protein induces monocyte activation and a pro-inflammatory phenotype in macrophages. Furthermore, we show that immobilised recombinant Gal-9 acts as capture and adhesion molecule for CD14+ monocytes in a ß2-integrin and glycan dependent manner, while adhesion of monocytes to stimulated endothelium is reduced when Gal-9 is knocked down. Gal-9 also facilitates enhanced recruitment of leukocytes from peripheral arterial disease (PAD) patients compared to healthy young and aged controls. We further characterise the endothelium as source of circulating Gal-9, which is increased in plasma of PAD patients compared to healthy controls. CONCLUSIONS: These results highlight a pathological role for Gal-9 as promoter of monocyte recruitment and atherosclerotic plaque progression, making it a novel target in the prevention of plaque formation and progression.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Ratones , Animales , Ratones Endogámicos C57BL , Células Cultivadas , Aterosclerosis/patología , Placa Aterosclerótica/metabolismo , Monocitos/metabolismo
7.
Blood ; 140(24): 2626-2643, 2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36026606

RESUMEN

S100A8/A9, also known as "calprotectin" or "MRP8/14," is an alarmin primarily secreted by activated myeloid cells with antimicrobial, proinflammatory, and prothrombotic properties. Increased plasma levels of S100A8/A9 in thrombo-inflammatory diseases are associated with thrombotic complications. We assessed the presence of S100A8/A9 in the plasma and lung autopsies from patients with COVID-19 and investigated the molecular mechanism by which S100A8/A9 affects platelet function and thrombosis. S100A8/A9 plasma levels were increased in patients with COVID-19 and sustained high levels during hospitalization correlated with poor outcomes. Heterodimeric S100A8/A9 was mainly detected in neutrophils and deposited on the vessel wall in COVID-19 lung autopsies. Immobilization of S100A8/A9 with collagen accelerated the formation of a fibrin-rich network after perfusion of recalcified blood at venous shear. In vitro, platelets adhered and partially spread on S100A8/A9, leading to the formation of distinct populations of either P-selectin or phosphatidylserine (PS)-positive platelets. By using washed platelets, soluble S100A8/A9 induced PS exposure but failed to induce platelet aggregation, despite GPIIb/IIIa activation and alpha-granule secretion. We identified GPIbα as the receptor for S100A8/A9 on platelets inducing the formation of procoagulant platelets with a supporting role for CD36. The effect of S100A8/A9 on platelets was abolished by recombinant GPIbα ectodomain, platelets from a patient with Bernard-Soulier syndrome with GPIb-IX-V deficiency, and platelets from mice deficient in the extracellular domain of GPIbα. We identified the S100A8/A9-GPIbα axis as a novel targetable prothrombotic pathway inducing procoagulant platelets and fibrin formation, in particular in diseases associated with high levels of S100A8/A9, such as COVID-19.


Asunto(s)
Plaquetas , COVID-19 , Calgranulina A , Calgranulina B , Complejo GPIb-IX de Glicoproteína Plaquetaria , Animales , Ratones , Plaquetas/metabolismo , Calgranulina A/metabolismo , COVID-19/metabolismo , Fibrina/metabolismo , Fosfatidilserinas/metabolismo , Agregación Plaquetaria , Humanos , Calgranulina B/metabolismo , Autopsia , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo
8.
Front Cardiovasc Med ; 9: 883179, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35833186

RESUMEN

Coronary artery disease is among the primary causes of death worldwide. While synthetic grafts allow replacement of diseased tissue, mismatched mechanical properties between graft and native tissue remains a major cause of graft failure. Multi-layered grafts could overcome these mechanical incompatibilities by mimicking the structural heterogeneity of the artery wall. However, the layer-specific biomechanics of synthetic grafts under physiological conditions and their impact on endothelial function is often overlooked and/or poorly understood. In this study, the transmural biomechanics of four synthetic graft designs were simulated under physiological pressure, relative to the coronary artery wall, using finite element analysis. Using poly(vinyl alcohol) (PVA)/gelatin cryogel as the representative biomaterial, the following conclusions are drawn: (I) the maximum circumferential stress occurs at the luminal surface of both the grafts and the artery; (II) circumferential stress varies discontinuously across the media and adventitia, and is influenced by the stiffness of the adventitia; (III) unlike native tissue, PVA/gelatin does not exhibit strain stiffening below diastolic pressure; and (IV) for both PVA/gelatin and native tissue, the magnitude of stress and strain distribution is heavily dependent on the constitutive models used to model material hyperelasticity. While these results build on the current literature surrounding PVA-based arterial grafts, the proposed method has exciting potential toward the wider design of multi-layer scaffolds. Such finite element analyses could help guide the future validation of multi-layered grafts for the treatment of coronary artery disease.

9.
J Thromb Haemost ; 20(4): 936-950, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34936188

RESUMEN

BACKGROUND: Platelets are multifunctional cellular mediators in many physiological and pathophysiological processes such as thrombosis, angiogenesis, and inflammation. Several members of galectins, a family of carbohydrate-binding proteins with a broad range of immunomodulatory actions, have been reported to activate platelets. OBJECTIVE: In this study, we investigated the role of galectin-9 (Gal-9) as a novel ligand for platelet glycoprotein VI (GPVI) and C-type lectin-like receptor 2 (CLEC-2). METHODS: Platelet spreading, aggregation, and P-selectin expression in response to Gal-9 were measured in washed platelet suspensions via static adhesion assay, light transmission aggregometry, and flow cytometry, respectively. Solid-phase binding assay and protein phosphorylation studies were utilized to validate the interaction between Gal-9 and GPVI, and immunoprecipitation for detecting CLEC-2 phosphorylation. Wild-type (WT), GPVI-knockout (Gp6-/- ), and GPVI and CLEC-2-double knockout (Gp6-/- /Gp1ba-Cre-Clec1bfl/fl ) mice were used. RESULTS: We have shown that recombinant Gal-9 stimulates aggregation in human and mouse washed platelets dose-dependently. Platelets from both species adhere and spread on immobilized Gal-9 and express P-selectin. Gal-9 competitively inhibited the binding of human recombinant D1 and D2 domains of GPVI to collagen. Gal-9 stimulated tyrosine phosphorylation of CLEC-2 and proteins known to lie downstream of GPVI and CLEC-2 including spleen tyrosine kinase and linker of activated T cells in human platelets. GPVI-deficient murine platelets exhibited significantly impaired aggregation in response to Gal-9, which was further abrogated in GPVI and CLEC-2-double-deficient platelets. CONCLUSIONS: We have identified Gal-9 as a novel platelet agonist that induces activation through interaction with GPVI and CLEC-2.


Asunto(s)
Lectinas Tipo C , Selectina-P , Animales , Plaquetas/metabolismo , Proteínas Portadoras/metabolismo , Galectinas/metabolismo , Humanos , Lectinas Tipo C/metabolismo , Ratones , Selectina-P/metabolismo , Activación Plaquetaria , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/metabolismo
10.
FASEB J ; 36(1): e22065, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34847625

RESUMEN

Neutrophil trafficking is a key component of the inflammatory response. Here, we have investigated the role of the immunomodulatory lectin Galectin-9 (Gal-9) on neutrophil recruitment. Our data indicate that Gal-9 is upregulated in the inflamed vasculature of RA synovial biopsies and report the release of Gal-9 into the extracellular environment following endothelial cell activation. siRNA knockdown of endothelial Gal-9 resulted in reduced neutrophil adhesion and neutrophil recruitment was significantly reduced in Gal-9 knockout mice in a model of zymosan-induced peritonitis. We also provide evidence for Gal-9 binding sites on human neutrophils; Gal-9 binding induced neutrophil activation (increased expression of ß2 integrins and reduced expression of CD62L). Intra-vital microscopy confirmed a pro-recruitment role for Gal-9, with increased numbers of transmigrated neutrophils following Gal-9 administration. We studied the role of both soluble and immobilized Gal-9 on human neutrophil recruitment. Soluble Gal-9 significantly strengthened the interaction between neutrophils and the endothelium and inhibited neutrophil crawling on ICAM-1. When immobilized, Gal-9 functioned as an adhesion molecule and captured neutrophils from the flow. Neutrophils adherent to Gal-9 exhibited a spread/activated phenotype that was inhibited by CD18 and CD44 neutralizing antibodies, suggesting a role for these molecules in the pro-adhesive effects of Gal-9. Our data indicate that Gal-9 is expressed and released by the activated endothelium and functions both in soluble form and when immobilized as a neutrophil adhesion molecule. This study paves the way for further investigation of the role of Gal-9 in leukocyte recruitment in different inflammatory settings.


Asunto(s)
Antígenos CD18/metabolismo , Galectinas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Receptores de Hialuranos/metabolismo , Neutrófilos/metabolismo , Migración Transendotelial y Transepitelial , Animales , Adhesión Celular , Humanos , Ratones
11.
Front Immunol ; 12: 687711, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34140956

RESUMEN

Leukocyte recruitment to the site of injury is a crucial event in the regulation of an inflammatory response. Tight regulation of interactions between the endothelium and circulating leukocytes is necessary to ensure a protective response to injury does not result in inflammatory disease. Rising interest in the broad immunoregulatory roles displayed by members of the glycan-binding galectin family suggests that these proteins could be an attractive target for therapeutic intervention, since their expression is significantly altered in disease. The focus of this review is to summarize current knowledge on the role of galectins in leukocyte trafficking during inflammation and the clinical approaches being taken to target these interactions for treatment of inflammatory disease.


Asunto(s)
Adhesión Celular , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Galectinas/metabolismo , Inflamación/metabolismo , Rodamiento de Leucocito , Leucocitos/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Adhesión Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Galectinas/antagonistas & inhibidores , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Rodamiento de Leucocito/efectos de los fármacos , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Transducción de Señal
12.
Front Cell Dev Biol ; 9: 624082, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33614653

RESUMEN

Leukocyte recruitment is a highly controlled cascade of interactions between proteins expressed by the endothelium and circulating leukocytes. The involvement of glycans and glycan-binding proteins in the leukocyte recruitment cascade has been well-characterised. However, our understanding of these interactions and their regulation has expanded substantially in recent years to include novel lectins and regulatory pathways. In this review, we discuss the role of glycans and glycan-binding proteins, mediating the interactions between endothelium and leukocytes both directly and indirectly. We also highlight recent findings of key enzymes involved in glycosylation which affect leukocyte recruitment. Finally, we investigate the potential of glycans and glycan binding proteins as therapeutic targets to modulate leukocyte recruitment and transmigration in inflammation.

13.
Biomed Pharmacother ; 130: 110595, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32771893

RESUMEN

Macrophages are key cells in both acute and chronic inflammatory settings. Their activation and function highly depends on the cytokines, chemokines and adhesion molecules that direct monocytes to infiltrate tissues, differentiate into macrophages, and finally lead to the clearance of such inflammatory signals. Galectins, ß-galactoside-binding lectins, are differentially expressed by various immune cells, and some members of this family have been identified as regulators of leukocyte recruitment and activation. Galectin-1 (Gal-1) and galectin-9 (Gal-9) expression has been described in immune cells, but the specific molecular mechanisms by which they modulate the inflammatory response in macrophages/monocytes are not completely understood. In this study we sought to comprehensively characterise the expression profile of endogenous Gal-1 and Gal-9 in different murine and human monocyte/macrophage populations in response to different inflammatory stimuli. All subsets of murine and human macrophages expressed significant levels of Gal-1 and -9. Interestingly, murine bone marrow derived macrophages stimulated with M2 (pro-resolution) polarising agents preferentially upregulated Gal-1, while Gal-9 expression was upregulated by M1/pro-inflammatory stimulation. However, we observed differing results in human monocyte derived macrophages. Collectively, our findings report a differential expression pattern of endogenous Gal-1 and -9 in macrophage and monocyte subsets in response to a range of inflammatory stimuli. Future studies will endeavour to elucidate whether the galectins make attractive therapeutic targets or agents for regulating the inflammatory response.


Asunto(s)
Galectina 1/biosíntesis , Galectinas/biosíntesis , Inflamación/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Adulto , Anciano , Animales , Células de la Médula Ósea/metabolismo , Células Cultivadas , Femenino , Humanos , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Adulto Joven
14.
Pharmacol Res ; 157: 104807, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32330552

RESUMEN

Many years have elapsed since the discovery of anti-inflammatories as effective therapeutics for the treatment of inflammatory-related diseases, but we are still uncovering their various mechanisms of action. Recent biochemical and pharmacological studies have shown that in different tissues and cell types lipid mediators from thearachidonic acid cascade, play a crucial role in the initiation and resolution of inflammation by shifting from pro-inflammatory prostaglandin (PG)E2 to anti-inflammatory PGD2 and PGJ2. Considering that until now very little is known about the biological effects evoked by microsomal prostaglandin E synthase-1 (mPGES-1) and contextually by peroxisome proliferator-activated receptor γ (PPARγ) modulation (key enzymes involved in PGE2 and PGD2/PGJ2metabolism), in this opinion paper we sought to define the coordinate functional regulation between these two enzymes at the "crossroads of phlogistic pathway" involved in the induction and resolution of inflammation.


Asunto(s)
Mediadores de Inflamación/metabolismo , Inflamación/enzimología , PPAR gamma/metabolismo , Prostaglandina-E Sintasas/metabolismo , Transducción de Señal , Animales , Antiinflamatorios/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Transducción de Señal/efectos de los fármacos
15.
Front Pharmacol ; 11: 274, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32265698

RESUMEN

Galectin-1 (Gal-1) exerts immune-regulatory and anti-inflammatory actions in animal models of acute and chronic inflammation. Its release into the extracellular milieu often correlates with the peak of inflammation suggesting that it may serve a pro-resolving function. Gal-1 is reported to inhibit neutrophil recruitment and induce surface exposure of phosphatidylserine (PS), an "eat me" signal on the surface of neutrophils, yet its role in resolution remains to be fully elucidated. We hypothesized that the anti-inflammatory and pro-resolving properties of Gal-1 are mediated through its ability to inhibit neutrophil recruitment and potentiate neutrophil clearance. To investigate this, a murine model of self-resolving inflammation was utilized to uncover the role of both the endogenous and exogenous protein using Gal-1 null mice and recombinant protein, respectively. We found that peritoneal macrophages express increased Gal-1 during the resolution phase and enhanced neutrophil recruitment occurs in the early phases of zymosan peritonitis in Gal-1 null mice compared to their wild-type (WT) counterparts. Administration of recombinant Gal-1 following the peak of inflammation led to reduced neutrophil numbers at 24 and 48 h, shortening the resolution interval from 39 to 14 h. Gal-1 treatment also enhanced neutrophil apoptosis, indicating a pro-resolving action. Together these results indicate an important role for Gal-1 in the timely resolution of acute inflammation.

16.
Haematologica ; 105(5): 1248-1261, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31467123

RESUMEN

Interactions between platelets, leukocytes and the vessel wall provide alternative pathological routes of thrombo-inflammatory leukocyte recruitment. We found that when platelets were activated by a range of agonists in whole blood, they shed platelet-derived extracellular vesicles which rapidly and preferentially bound to blood monocytes compared to other leukocytes. Platelet-derived extracellular vesicle binding to monocytes was initiated by P-selectin-dependent adhesion and was stabilised by binding of phosphatidylserine. These interactions resulted in the progressive transfer of the platelet adhesion receptor GPIbα to monocytes. GPIbα+-monocytes tethered and rolled on immobilised von Willebrand Factor or were recruited and activated on endothelial cells treated with TGF-ß1 to induce the expression of von Willebrand Factor. In both models monocyte adhesion was ablated by a function-blocking antibody against GPIbα. Monocytes could also bind platelet-derived extracellular vesicle in mouse blood in vitro and in vivo Intratracheal instillations of diesel nanoparticles, to model chronic pulmonary inflammation, induced accumulation of GPIbα on circulating monocytes. In intravital experiments, GPIbα+-monocytes adhered to the microcirculation of the TGF-ß1-stimulated cremaster muscle, while in the ApoE-/- model of atherosclerosis, GPIbα+-monocytes adhered to the carotid arteries. In trauma patients, monocytes bore platelet markers within 1 hour of injury, the levels of which correlated with severity of trauma and resulted in monocyte clearance from the circulation. Thus, we have defined a novel thrombo-inflammatory pathway in which platelet-derived extracellular vesicles transfer a platelet adhesion receptor to monocytes, allowing their recruitment in large and small blood vessels, and which is likely to be pathogenic.


Asunto(s)
Plaquetas , Vesículas Extracelulares , Animales , Células Endoteliales , Humanos , Inflamación , Ratones , Monocitos , Complejo GPIb-IX de Glicoproteína Plaquetaria
17.
Biomed Pharmacother ; 123: 109788, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31865142

RESUMEN

Antimicrobial peptides (AMPs) are an ancient group of defense molecules distributed in nature being found in mammals, birds, amphibians, insects, plants, and microorganisms. They display antimicrobial as well as immunomodulatory properties. The aim of this study was to investigate, for the first time, the anti-inflammatory activities of two synthetic temporin-L analogues (here named peptide 1 and 2) by an in vivo model of inflammation caused by intraperitoneal sub-lethal dose of zymosan. Our results show that peptide 1 and 2 exert anti-inflammatory activity in vivo in response to zymosan-induce peritonitis. Simultaneous administration of 10 mg/kg of both temporins, with a sub-lethal dose of zymosan (500 mg/kg), significantly rescued mice from the classical hallmarks of inflammation, including leukocyte infiltration and synthesis of inflammatory mediators including IL-6, TNF-α and MCP-1. More importantly, flow cytometry analysis highlighted a selective modulation of infiltrating inflammatory monocytes (defined as B220-/GR1hi-F480hi/CD115+) after peptide 2 treatment. Our results and presented models offer the possibility to test, in a preclinical setting, the potential of temporin analogues as anti-inflammatory agents.


Asunto(s)
Antiinflamatorios/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Inflamación/tratamiento farmacológico , Peritonitis/tratamiento farmacológico , Enfermedad Aguda , Animales , Antiinflamatorios/química , Péptidos Catiónicos Antimicrobianos/química , Línea Celular , Modelos Animales de Enfermedad , Inflamación/patología , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Monocitos/efectos de los fármacos , Monocitos/patología , Peritonitis/patología , Zimosan
18.
Data Brief ; 25: 104381, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31489355

RESUMEN

The data supplied in this work are related to the research article entitled "IL-17A neutralizing antibody regulates monosodium urate crystal-induced gouty inflammation" [1]. This data article presents the results of the gating strategy applied to identify Treg population in peripheral blood of mice injected with MSU crystals and MSU crystals + interleukin-17 antibody (IL-17Ab). Lastly, this article provides in-depth immunophenotyping data relating to all specific and isotype control antibodies used in the phenotypical characterization of circulating Treg (defined as CD4+CD25+Foxp3+), Th17 (defined as CD4+IL-17+) cells and joint-infiltrated (in situ) inflammatory monocytes (defined as B220-GR1highF480highCD115+).

19.
Pharmacol Res ; 147: 104351, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31315067

RESUMEN

Gout is a paradigm of acute, self-limiting inflammation caused by the deposition of monosodium urate (MSU) crystals within intra-and/or peri-articular areas, leading to excruciating pain, joint swelling and stiffness. The infiltration of leukocytes drives the inflammatory response and remains an attractive target for therapeutic intervention. In this context, emerging evidence supports the view that systemic differentiation of Th17 cells and their in situ infiltration as one of the potential mechanisms by which these cells, and their main product IL-17, causes damage to target tissues. To test if IL-17 was having a detrimental role in gouty onset and progression we targeted this cytokine, using a neutralizing antibody strategy, in an experimental model of gout. Joint inflammation was induced in CD-1 mice by the intra-articular (i.a.) administration of MSU crystals (200 µg/20 µl). Animals from IL-17Ab-treated groups received 1, 3 and 10 µg (i.a.) in 20 µl of neutralizing antibody after MSU crystals administration. Thereafter, joints were scored macroscopically, and knee joint oedema determined with a caliper. Histological analysis, myeloperoxidase assay and western blots analysis for COX-2/mPGEs-1/IL-17R pathway were conducted at 18 h (peak of inflammation) to evaluate leukocytes infiltration and activation, followed by the analysis, in situ, of pro/anti-inflammatory cytokines and chemokines. Flow cytometry was also used to evaluate the modulation of infiltrated inflammatory monocytes and systemic Th17 and Treg profile. Treatment with IL-17Ab revealed a dose-dependent reduction of joint inflammation scores with maximal inhibition at 10 µg. The neutralizing antibody was also able to significantly reduce leukocytes infiltration and MPO activity as well the expression of JE, IL-1α, IL-1ß, IL-16, IL-17, C5a, BLC and, with a less extent IP-10, Rantes, KC, TIMP-1, SDF-1 and metalloproteinases in inflamed tissues. Biochemical analysis also revealed that IL-17Ab treatment modulated COX-2/mPGEs-1 pathway (and related PGE2 production) without interfering with IL-17R expression. Furthermore, flow cytometry analysis highlighted a selective modulation of infiltrating inflammatory monocytes (B220-/GR1hi-F480hi/CD115+) and circulating Th17, but not Treg, cells after IL-17Ab treatment. Collectively the results of this study report for the first time, that i.a. injection of MSU crystals stimulates in vivo production of Th17 cells and Th17-related inflammatory cyto-chemokines. In addition, we have demonstrated that the administration of a neutralizing antibody against IL-17 attenuates joint symptoms, swelling and leukocytes infiltration to the inflamed tissue, possibly providing a new strategy for the treatment of gouty inflammation and/or arthritis.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Gota/inmunología , Interleucina-17/inmunología , Ácido Úrico , Animales , Edema/inmunología , Edema/patología , Gota/patología , Inflamación/inmunología , Inflamación/patología , Inyecciones Intraarticulares , Articulación de la Rodilla/inmunología , Articulación de la Rodilla/patología , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...