Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Intervalo de año de publicación
1.
Angiogenesis ; 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38878257

RESUMEN

Snake venoms are intricate mixtures of enzymes and bioactive factors that induce a range of detrimental effects in afflicted hosts. Certain Viperids, including Bothrops jararacussu, harbor C-type lectins (CTLs) known for their modulation of a variety of host cellular responses. In this study, we isolated and purified BjcuL, a CTL from B. jararacussu venom and investigated its impact on endothelial cell behavior, contrasting it with human galectin-1 (Gal-1), a prototype member of the galectin family with shared ß-galactoside-binding activity. We found that BjcuL binds to human dermal microvascular endothelial cells (HMECs) in a concentration- and carbohydrate-dependent fashion and reprograms the function of these cells, favoring a pro-inflammatory and pro-coagulant endothelial phenotype. In light of the quest for universal antagonists capable of mitigating the harmful consequences of snake venoms, BjcuL emerges as a promising target to be blocked in order to regulate pathological endothelial cell responses.

2.
PLoS Pathog ; 16(12): e1009176, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33347509

RESUMEN

Multiple sclerosis (MS) is a highly disabling neurodegenerative autoimmune condition in which an unbalanced immune response plays a critical role. Although the mechanisms remain poorly defined, helminth infections are known to modulate the severity and progression of chronic inflammatory diseases. The tyrosine kinase receptors TYRO3, AXL, and MERTK (TAM) have been described as inhibitors of the immune response in various inflammatory settings. We show here that patients with concurrent natural helminth infections and MS condition (HIMS) had an increased expression of the negative regulatory TAM receptors in antigen-presenting cells and their agonist GAS6 in circulating CD11bhigh and CD4+ T cells compared to patients with only MS. The Th17 subset was reduced in patients with HIMS with a subsequent downregulation of its pathogenic genetic program. Moreover, these CD4+ T cells promoted lower levels of the co-stimulatory molecules CD80, CD86, and CD40 on dendritic cells compared with CD4+ T cells from patients with MS, an effect that was GAS6-dependent. IL-10+ cells from patients with HIMS showed higher GAS6 expression levels than Th17 cells, and inhibition of phosphatidylserine/GAS6 binding led to an expansion of Th17 effector genes. The addition of GAS6 on activated CD4+ T cells from patients with MS restrains the Th17 gene expression signature. This cohort of patients with HIMS unravels a promising regulatory mechanism to dampen the Th17 inflammatory response in autoimmunity.


Asunto(s)
Helmintiasis/complicaciones , Helmintiasis/inmunología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/parasitología , Células Th17/inmunología , Adulto , Animales , Femenino , Humanos , Masculino
3.
Int J Mol Sci ; 23(18)2022 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-36142365

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been responsible for the severe pandemic of acute respiratory disease, coronavirus disease 2019 (COVID-19), experienced in the 21st century. The clinical manifestations range from mild symptoms to abnormal blood coagulation and severe respiratory failure. In severe cases, COVID-19 manifests as a thromboinflammatory disease. Damage to the vascular compartment caused by SARS-CoV-2 has been linked to thrombosis, triggered by an enhanced immune response. The molecular mechanisms underlying endothelial activation have not been fully elucidated. We aimed to identify the proteins correlated to the molecular response of human umbilical vein endothelial cells (HUVECs) after exposure to SARS-CoV-2, which might help to unravel the molecular mechanisms of endothelium activation in COVID-19. In this direction, we exposed HUVECs to SARS-CoV-2 and analyzed the expression of specific cellular receptors, and changes in the proteome of HUVECs at different time points. We identified that HUVECs exhibit non-productive infection without cytopathic effects, in addition to the lack of expression of specific cell receptors known to be essential for SARS-CoV-2 entry into cells. We highlighted the enrichment of the protein SUMOylation pathway and the increase in SUMO2, which was confirmed by orthogonal assays. In conclusion, proteomic analysis revealed that the exposure to SARS-CoV-2 induced oxidative stress and changes in protein abundance and pathways enrichment that resembled endothelial dysfunction.


Asunto(s)
Fenómenos Biológicos , COVID-19 , Células Endoteliales , Humanos , Proteoma , Proteómica , SARS-CoV-2
4.
Platelets ; 32(3): 305-313, 2021 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-31984825

RESUMEN

Platelets have a well-recognized role in hemostasis and thrombosis, and they are important amplifiers of inflammation and innate immune responses. The formation of DNA extracellular traps (ETs) is a complex cellular mechanism, which occurs in response to microbial infections and sterile inflammation, and results in the release of DNA complexed with histones and various granular proteins. ETs were first discovered in neutrophils (NETs); however, it is now accepted that other leukocytes, including eosinophils (EETs) and monocytes/macrophages (MoETs/METs), can also generate them. Moreover, several types of ETs have been described.Increasing evidence has demonstrated that platelets modulate the formation of ETs. This review summarizes recent findings about the physiopathological role of platelets in the formation of ETs during infection and future perspectives in the field.


Asunto(s)
Plaquetas/metabolismo , Trampas Extracelulares/metabolismo , Infecciones/sangre , Humanos
5.
Cell Microbiol ; 21(2): e12990, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30537301

RESUMEN

Leptospirosis is a global zoonosis caused by pathogenic Leptospira. Neutrophils are key cells against bacterial pathogens but can also contribute to tissue damage. Because the information regarding the role of human neutrophils in leptospirosis is scant, we comparatively analysed the human neutrophil's response to saprophytic Leptospira biflexa serovar Patoc (Patoc) and the pathogenic Leptospira interrogans serovar Copenhageni (LIC). Both species triggered neutrophil responses involved in migration, including the upregulation of CD11b expression, adhesion to collagen, and the release of IL-8. In addition, both species increased levels of pro-inflammatory IL-1ß and IL-6 associated with the inflammasome and NFκB pathway activation and delayed neutrophil apoptosis. LIC was observed on the neutrophil surface and not phagocytized. In contrast, Patoc generated intracellular ROS associated with its uptake. Neutrophils express the TYRO3, AXL, and MER receptor protein tyrosine kinases (TAM), but only LIC selectively increased the level of AXL. TLR2 but not TLR4-blocking antibodies abrogated the IL-8 secretion triggered by both Leptospira species. In summary, we demonstrate that Leptospira species trigger a robust neutrophil activation and pro-inflammatory response. These findings may be useful to find new diagnostic markers and therapeutic strategies against leptospirosis.


Asunto(s)
Leptospira/inmunología , Leptospirosis/inmunología , Leptospirosis/patología , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Antígeno CD11b/metabolismo , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Leptospira interrogans/inmunología , Leptospirosis/microbiología
6.
Arterioscler Thromb Vasc Biol ; 39(10): e219-e232, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31434496

RESUMEN

OBJECTIVE: Ceramide 1-phosphate (C1P) is a bioactive sphingolipid highly augmented in damaged tissues. Because of its abilities to stimulate migration of murine bone marrow-derived progenitor cells, it has been suggested that C1P might be involved in tissue regeneration. In the present study, we aimed to investigate whether C1P regulates survival and angiogenic activity of human progenitor cells with great therapeutic potential in regenerative medicine such as endothelial colony-orming cells (ECFCs). Approach and Results: C1P protected ECFC from TNFα (tumor necrosis factor-α)-induced and monosodium urate crystal-induced death and acted as a potent chemoattractant factor through the activation of ERK1/2 (extracellular signal-regulated kinases 1 and 2) and AKT pathways. C1P treatment enhanced ECFC adhesion to collagen type I, an effect that was prevented by ß1 integrin blockade, and to mature endothelial cells, which was mediated by the E-selectin/CD44 axis. ECFC proliferation and cord-like structure formation were also increased by C1P, as well as vascularization of gel plug implants loaded or not with ECFC. In a murine model of hindlimb ischemia, local administration of C1P alone promoted blood perfusion and reduced necrosis in the ischemic muscle. Additionally, the beneficial effects of ECFC infusion after ischemia were amplified by C1P pretreatment, resulting in a further and significant enhancement of leg reperfusion and muscle repair. CONCLUSIONS: Our findings suggest that C1P may have therapeutic relevance in ischemic disorders, improving tissue repair by itself, or priming ECFC angiogenic responses such as chemotaxis, adhesion, proliferation, and tubule formation, which result in a better outcome of ECFC-based therapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Ceramidas/farmacología , Células Progenitoras Endoteliales/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Regeneración/efectos de los fármacos , Animales , Diferenciación Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/efectos de los fármacos , Humanos , Isquemia/tratamiento farmacológico , Isquemia/metabolismo , Ratones , Morfogénesis/efectos de los fármacos , Sensibilidad y Especificidad
7.
J Mol Cell Cardiol ; 125: 149-161, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30393107

RESUMEN

Coxsackievirus B3 (CVB3) is a globally prevalent enterovirus of the Picornaviridae family that is frequently associated with viral myocarditis (VM). Neutrophils, as first responders, may be key cells in determining viral disease outcomes; however, neutrophils have been poorly studied with respect to viral infection. Although neutrophils have been ascribed a relevant role in early cardiac inflammation, their precise role in CVB3 infection has not yet been evaluated. In this study, we aimed to determine if the interaction between human neutrophils and CVB3 could lead to viral replication and/or modulation of neutrophil survival and biological functions, and whether neutrophil depletion in a murine model has a beneficial or harmful effect on CVB3 infection. Our results show that CVB3 interacted with but did not replicate in human neutrophils. Neutrophils recognized CVB3 mainly through endosomal TLR-8, and infection triggered NFκB activation. Virus internalization resulted in increased cell survival, up-regulation of CD11b, enhanced adhesion to fibrinogen and fibronectin, and the secretion of IL-6, IL-1ß, TNF-α, and IL-8. Supernatants from infected neutrophils exerted chemotactic activity partly mediated by IL-8. The infected neutrophils released myeloperoxidase and triggered neutrophil extracellular trap formation in the presence of TNF-α. In mice infected with CVB3, viral RNA was detected in neutrophils as well as in mononuclear cells. After neutrophil depletion, mice showed reduced VM reflected by a reduction in viral titers, cell exudates, and CCL-2 mRNA levels, as well as the abrogation of reactive cardiomyocyte hypertrophy. Our results indicate that neutrophils have relevant direct and indirect roles in the pathogenesis of CVB3-induced VM.


Asunto(s)
Infecciones por Coxsackievirus/metabolismo , Miocarditis/metabolismo , Miocarditis/virología , Neutrófilos/metabolismo , Animales , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/virología , Enterovirus Humano B/inmunología , Enterovirus Humano B/patogenicidad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Interleucina-1beta/metabolismo , Miocarditis/inmunología , Miocardio/inmunología , Miocardio/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/metabolismo , Replicación Viral/fisiología
8.
Clin Immunol ; 183: 254-262, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28863968

RESUMEN

NETosis is a host defense mechanism associated with inflammation and tissue damage. Experimental models show that platelets and von Willebrand factor (VWF) are key elements for intravascular NETosis. We determined NETosis in septic and burn patients at 1 and 4days post-admission (dpa). Nucleosomes were elevated in patients. In septics, they correlated with Human Neutrophil Elastase (HNE)-DNA complexes and SOFA score at 1dpa, and were associated with mortality. Patient's neutrophils had spontaneous NETosis and were unresponsive to stimulation. Although platelet P-selectin and TNF-α were increased in both groups, higher platelet TLR-4 expression, VWF levels and IL-6 were found in septics at 1dpa. Neither platelet activation markers nor cytokines correlated with nucleosomes or HNE-DNA. Nucleosomes could be indicators of organ damage and predictors of mortality in septic but not in burn patients. Platelet activation, VWF and cytokines do not appear to be key mediators of NETosis in these patient groups.


Asunto(s)
Quemaduras/metabolismo , Trampas Extracelulares/fisiología , Nucleosomas/metabolismo , Sepsis/metabolismo , Adulto , Anciano , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación , Modelos Lineales , Masculino , Persona de Mediana Edad , Activación Plaquetaria , Factor de von Willebrand
9.
Blood ; 126(2): 242-6, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-25979951

RESUMEN

Neutrophil extracellular traps (NETs) can be released in the vasculature. In addition to trapping microbes, they promote inflammatory and thrombotic diseases. Considering that P-selectin induces prothrombotic and proinflammatory signaling, we studied the role of this selectin in NET formation. NET formation (NETosis) was induced by thrombin-activated platelets rosetting with neutrophils and was inhibited by anti-P-selectin aptamer or anti-P-selectin glycoprotein ligand-1 (PSGL-1) inhibitory antibody but was not induced by platelets from P-selectin(-/-) mice. Moreover, NETosis was also promoted by P-selectin-immunoglobulin fusion protein but not by control immunoglobulin. We isolated neutrophils from mice engineered to overproduce soluble P-selectin (P-selectin(ΔCT/ΔCT) mice). Although the levels of circulating DNA and nucleosomes (indicative of spontaneous NETosis) were normal in these mice, basal neutrophil histone citrullination and presence of P-selectin on circulating neutrophils were elevated. NET formation after stimulation with platelet activating factor, ionomycin, or phorbol 12-myristate 13-acetate was significantly enhanced, indicating that the P-selectin(ΔCT/ΔCT) neutrophils were primed for NETosis. In summary, P-selectin, cellular or soluble, through binding to PSGL-1, promotes NETosis, suggesting that this pathway is a potential therapeutic target for NET-related diseases.


Asunto(s)
Trampas Extracelulares/genética , Selectina-P/fisiología , Trombosis/genética , Vasculitis/genética , Animales , Plaquetas/fisiología , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Selectina-P/genética , Selectina-P/farmacología , Activación Plaquetaria/genética , Proteínas Recombinantes de Fusión/farmacología , Trombosis/patología , Vasculitis/patología
10.
Mediators Inflamm ; 2017: 2810606, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28852268

RESUMEN

M strain, the most prevalent multidrug-resistant strain of Mycobacterium tuberculosis (Mtb) in Argentina, has mounted mechanisms to evade innate immune response. The role of human bronchial epithelium in Mtb infection remains unknown as well as its crosstalk with neutrophils (PMN). In this work, we evaluate whether M and H37Rv strains invade and replicate within bronchial epithelial cell line Calu-6 and how conditioned media (CM) derived from infected cells alter PMN responses. We demonstrated that M infects and survives within Calu-6 without promoting death. CM from M-infected Calu-6 (M-CM) did not attract PMN in correlation with its low IL-8 content compared to H37Rv-CM. Also, PMN activation and ROS production in response to irradiated H37Rv were impaired after treatment with M-CM due to the lack of TNF-α. Interestingly, M-CM increased H37Rv replication in PMN which would allow the spreading of mycobacteria upon PMN death and sustain IL-8 release. Thus, our results indicate that even at low invasion/replication rate within Calu-6, M induces the secretion of factors altering the crosstalk between these nonphagocytic cells and PMN, representing an evasion mechanism developed by M strain to persist in the host. These data provide new insights on the role of bronchial epithelium upon M infection.


Asunto(s)
Interleucina-8/metabolismo , Mycobacterium tuberculosis/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Quimiocinas/metabolismo , Quimiotaxis/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Citometría de Flujo , Humanos , Inmunidad Innata/efectos de los fármacos , Fagocitosis/efectos de los fármacos
11.
J Virol ; 89(14): 7409-13, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25926646

RESUMEN

The arenavirus Junin virus (JUNV) is the etiologic agent of Argentine hemorrhagic fever. We characterized the JUNV infection of human peripheral blood-derived plasmacytoid dendritic cells (hpDC), demonstrating that hpDC are susceptible to infection with the C#1 strain (attenuated) and even more susceptible to infection with the P (virulent) JUNV strain. However, hpDC elicited different responses in terms of viability, activation, maturation, and cytokine expression after infection with both JUNV strains.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , Virus Junin/inmunología , Diferenciación Celular , Supervivencia Celular , Citocinas/biosíntesis , Humanos , Virus Junin/patogenicidad
12.
J Mol Cell Cardiol ; 85: 58-70, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26002282

RESUMEN

Macrophage influx and galectin 3 production have been suggested as major players driving acute inflammation and chronic fibrosis in many diseases. However, their involvement in the pathogenesis of viral myocarditis and subsequent cardiomyopathy are unknown. Our aim was to characterise the role of macrophages and galectin 3 on survival, clinical course, viral burden, acute pathology, and chronic fibrosis in coxsackievirus B3 (CVB3)-induced myocarditis. Our results showed that C3H/HeJ mice infected with CVB3 and depleted of macrophages by liposome-encapsulated clodronate treatment compared with infected untreated mice presented higher viral titres but reduced acute myocarditis and chronic fibrosis, compared with untreated infected mice. Increased galectin 3 transcriptional and translational expression levels correlated with CVB3 infection in macrophages and in non-depleted mice. Disruption of the galectin 3 gene did not affect viral titres but reduced acute myocarditis and chronic fibrosis compared with C57BL/6J wild-type mice. Similar results were observed after pharmacological inhibition of galectin 3 with N-acetyl-d-lactosamine in C3H/HeJ mice. Our results showed a critical role of macrophages and their galectin 3 in controlling acute viral-induced cardiac injury and the subsequent fibrosis. Moreover, the fact that pharmacological inhibition of galectin 3 induced similar results to macrophage depletion regarding the degree of acute cardiac inflammation and chronic fibrosis opens up the possibility of new pharmacological strategies for viral myocarditis.


Asunto(s)
Infecciones por Coxsackievirus/complicaciones , Galectina 3/fisiología , Macrófagos/inmunología , Miocarditis/inmunología , Animales , Línea Celular , Enterovirus , Fibrosis , Masculino , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Miocarditis/metabolismo , Miocarditis/virología
13.
BMC Immunol ; 16: 26, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25913718

RESUMEN

Blood platelets are first aimed at ensuring primary hemostasis. Beyond this role, they have been acknowledged as having functions in the maintenance of the vascular arborescence and, more recently, as being also innate immune cells, devoted notably to the detection of danger signals, of which infectious ones. Platelets express pathogen recognition receptors that can sense bacterial and viral moieties. Besides, several molecules that bind epithelial or sub-endothelial molecules and, so forth, are involved in hemostasis, happen to be able to ligate viral determinants, making platelets capable of either binding viruses or even to be infected by some of them. Further, as platelets express both Fc-receptors for Ig and complement receptors, they also bind occasionally virus-Ig or virus-Ig-complement immune complexes. Interplays of viruses with platelets are very complex and viral infections often interfere with platelet number and functions. Through a few instances of viral infections, the present review aims at presenting some of the most important interactions from pathophysiological and clinical points of view, which are observed between human viruses and platelets.


Asunto(s)
Plaquetas/inmunología , Virosis/inmunología , Virus/inmunología , Animales , Plaquetas/virología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Acoplamiento Viral
14.
Glycobiology ; 24(12): 1252-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24928621

RESUMEN

Under normal conditions, platelets do not interact with blood vessel walls; however, upon activation, platelets firmly attach to endothelial cells. Communication between platelets and endothelial cells during the normal or activated state takes place at multiple levels. Cross-talk may occur over a distance via transient interactions or through receptor-mediated cell-cell adhesion. Platelets may release or transfer substances that affect endothelial cell function and vice versa. Excessive dialogue between platelets and the endothelium exists in several disease states as a causative factor and/or as a consequence of the disease process. Glycans are covalent assemblies of sugars that exist in either free form or in covalent complexes with proteins or lipids. Among other functions, glycans confer stability to the proteins to which they are attached, play key roles in signal transduction and control cell development and differentiation. Glycans not only influence the structure and function of hemostatic molecules but are also increasingly recognized as key molecules regulating platelet-endothelial interactions. The present review outlines the current knowledge regarding glycan-mediated interactions between platelets and endothelial cells and their role in physiopathological processes.


Asunto(s)
Plaquetas/citología , Plaquetas/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Polisacáridos/metabolismo , Comunicación Celular , Glicómica , Humanos
15.
Glycobiology ; 24(10): 966-73, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24957054

RESUMEN

Galectins (Gals), a family of mammalian lectins, play diverse roles under physiological and pathological conditions. Here, we analyzed the tandem-repeat Gal-8 synthesis, secretion and effects on the endothelium physiology. Gal-8M and Gal-8L isoforms were secreted under basal conditions by human microvascular endothelial cells (HMEC-1). However, expression and secretion of the Gal-8M isoform, but not Gal-8L, were increased in response to bacterial lipopolysaccharide (LPS) stimulus and returned to control values after LPS removal. Similarly, cell surface Gal-8 exposure was increased after stimulation with LPS. To evaluate Gal-8 effects on the endothelium physiology, HMEC-1 cells were incubated in the presence of recombinant Gal-8M. Pretreated HMEC-1 cells became proadhesive to human normal platelets, indicating that Gal-8 actually activates endothelial cells. This effect was specific for lectin activity as it was prevented by the simultaneous addition of lactose, but not by sucrose. Endothelial cells also increased their exposition of von Willebrand factor after Gal-8 treatment, which constitutes another feature of cell activation that could be, in turn, responsible for the observed platelet adhesion. Several pro-inflammatory molecules were abundantly produced by Gal-8 stimulated endothelial cells: CXCL1 (GRO-α), GM-CSF, IL-6 and CCL5 (RANTES), and in a lower degree CCL2 (MCP-1), CXCL3 (GRO-γ) and CXCL8 (IL-8). In agreement, Gal-8M induced nuclear factor kappa B phosphorylation. Altogether, these results not only confirm the pro-inflammatory role we have already proposed for Gal-8 in other cellular systems but also suggest that this lectin is orchestrating the interaction between leukocytes, platelets and endothelial cells.


Asunto(s)
Endotelio/metabolismo , Galectinas/metabolismo , Interleucina-8/metabolismo , Galectinas/genética , Humanos , Interleucina-8/genética , Lipopolisacáridos/química , Secuencias Repetidas en Tándem/genética
16.
Angiogenesis ; 17(4): 867-79, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24854678

RESUMEN

OBJECTIVE: Acidosis is present in several pathological conditions where vasculogenesis takes place including ischemia, tumor growth and wound healing. We have previously demonstrated that acidosis induces human CD34+ cell apoptosis. Considering that endothelial colony-forming cells (ECFC) are a subpopulation of CD34+ cells and key players in vasculogenesis, in the present study we investigated the effect of acidosis on the survival and functionality of ECFC. APPROACH AND RESULTS: Endothelial colony-forming cells obtained by differentiation of human cord blood CD34+ cells in endothelial growth medium-2 for 14-21 days were exposed at pH 7.4, 7.0 or 6.6. We found that acidosis failed to induce ECFC apoptosis and, although an early reduction in proliferation, chemotaxis, wound healing and capillary-like tubule formation was observed, once the medium pH was restored to 7.4, ECFC proliferation and tubulogenesis were augmented. Stromal cell derived factor-1 (SDF1)-driven migration and chemokine receptor type 4 surface expression were also increased. The maximal proangiogenic effect exerted by acidic preconditioning was observed after 6 h at pH 6.6. Furthermore, preconditioned ECFC showed an increased ability to promote tissue revascularization in a murine model of hind limb ischemia. Immunoblotting assays showed that acidosis activated AKT and ERK1/2 and inhibited p38 pathways. Proliferation rises triggered by acidic preconditioning were no longer observed after AKT or ERK1/2 inhibition, whereas p38 suppression not only mimicked but also potentiated the effect of acidosis on ECFC tubule formation abilities. CONCLUSIONS: These results demonstrate that acidic preconditioning greatly increases ECFC-mediated angiogenesis in vitro including ECFC proliferation, tubulogenesis and SDF1-driven chemotaxis and is a positive regulator of microvessel formation in vivo.


Asunto(s)
Ácidos/química , Medios de Cultivo/química , Células Endoteliales/citología , Células Madre/citología , Animales , Antígenos CD34/metabolismo , Apoptosis , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Quimiotaxis , Humanos , Concentración de Iones de Hidrógeno , Isquemia/patología , Masculino , Ratones , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Cicatrización de Heridas
17.
J Pharmacol Exp Ther ; 345(3): 430-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23536315

RESUMEN

The formation of neutrophil extracellular traps (NETs) is a newly described phenomenon that increases the bacteria-killing ability and the inflammatory response of neutrophils. Because NET generation occurs in an inflammatory microenvironment, we examined its regulation by anti-inflammatory drugs. Treatment of neutrophils with dexamethasone had no effect, but acetylsalicylic acid (ASA) treatment prevented NET formation. NETosis was also abrogated by the presence of BAY 11-7082 [(E)-3-[4-methylphenylsulfonyl]-2-propenenitrile] and Ro 106-9920 [6-(phenylsulfinyl)tetrazolo[1,5-b]pyridazine], two structurally unrelated nuclear factor-κB (NF-κB) inhibitors. The decrease in NET formation mediated by ASA, BAY-11-7082, and Ro 106-9920 was correlated with a significant reduction in the phosphorylation of NF-κB p65 subunit, indicating that the activation of this transcription factor is a relevant signaling pathway involved in the generation of DNA traps. The inhibitory effect of these drugs was also observed when NET generation was induced under acidic or hyperthermic conditions, two stress signals of the inflammatory microenvironment. In a mouse peritonitis model, while pretreatment of animals with ASA or BAY 11-7082 resulted in a marked suppression of NET formation along with increased bacteremia, dexamethasone had no effect. Our results show that NETs have an important role in the local control of infection and that ASA and NF-κB blockade could be useful therapies to avoid undesired effect of persistent neutrophil activation.


Asunto(s)
Antiinflamatorios/farmacología , Espacio Extracelular/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Acidosis/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Apoptosis/efectos de los fármacos , Aspirina/farmacología , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/prevención & control , Western Blotting , Permeabilidad Capilar/efectos de los fármacos , ADN/biosíntesis , ADN/genética , Dexametasona/farmacología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Endogámicos BALB C , Microscopía Fluorescente , FN-kappa B/antagonistas & inhibidores , Nitrilos/farmacología , Lavado Peritoneal , Sulfonas/farmacología
18.
Biol Chem ; 394(7): 857-63, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23509216

RESUMEN

Platelet activation at sites of vascular injury leads to the formation of a hemostatic plug and is crucial for hemostasis. However, uncontrolled platelet activation may lead to the formation of occlusive thrombi. Several soluble or matricellular proteins can activate platelets. In this article, we review recent advances in knowledge of the role of galectins in platelet physiology. In soluble or immobilized form, these endogenous glycan-binding proteins trigger platelet activation through the modulation of discrete signaling pathways. We discuss the role of platelet-galectin interactions not only in hemostasis, but also in chronic inflammation, atherosclerosis and cancer.


Asunto(s)
Plaquetas/fisiología , Galectinas/sangre , Hemostasis/fisiología , Activación Plaquetaria/fisiología , Adhesividad Plaquetaria/fisiología , Animales , Humanos
19.
Microb Pathog ; 56: 21-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23092690

RESUMEN

We report the study of a predicted outer-membrane leptospiral protein encoded by the gene lic11207. This protein is conserved in several pathogenic leptospiral strains but is absent in the saprophyte Leptospira biflexa. This putative outer-membrane protein has a domain of unknown function (DUF) 1565 found in several phylogenetically diverse bacteria and in the archaeon Methanosarcina acetivorans. The gene was cloned and expressed in Escherichia coli BL21 (SI) strain using the expression vector pDEST17. The 34 kDa recombinant protein was tagged with N-terminal hexahistidine and purified by metal-charged chromatography. The purified protein was used to assess: reactivity with human convalescent sera; in vivo expression; ability to activate endothelial cells (EC); and ability to modulate the apoptosis of polymorphonuclear cells (PMNs). The LIC11207 coding sequence was identified in vivo in the hamster renal tubules during experimental infection with Leptospira interrogans. The rLIC11207 showed significant antigenicity against human convalescent sera when compared with sera from healthy donors. The recombinant protein did not alter the surface expression of E-selectin or intercellular adhesion molecule 1 (ICAM-1) in EC and failed to induce the release of von Willebrand factor (vWF). Interestingly, rLIC11207 delayed apoptosis of PMNs suggesting a possible role of this protein during the infection.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Apoptosis , Proteínas de la Membrana Bacteriana Externa/metabolismo , Leptospira interrogans/patogenicidad , Neutrófilos/efectos de los fármacos , Factores de Virulencia/metabolismo , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Cricetinae , Modelos Animales de Enfermedad , Células Endoteliales/microbiología , Perfilación de la Expresión Génica , Humanos , Túbulos Renales/patología , Leptospira interrogans/inmunología , Leptospirosis/microbiología , Leptospirosis/patología , Neutrófilos/microbiología , Factores de Virulencia/genética , Factores de Virulencia/inmunología
20.
FASEB J ; 26(7): 2788-98, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22456341

RESUMEN

Understanding noncanonical mechanisms of platelet activation represents an important challenge for the identification of novel therapeutic targets in bleeding disorders, thrombosis, and cancer. We previously reported that galectin-1 (Gal-1), a ß-galactoside-binding protein, triggers platelet activation in vitro. Here we investigated the molecular mechanisms underlying this function and the physiological relevance of endogenous Gal-1 in hemostasis. Mass spectrometry analysis, as well as studies using blocking antibodies against the anti-α(IIb) subunit ofα(IIb)ß(3) integrin or platelets from patients with Glanzmann's thrombasthenia syndrome (α(IIb)ß(3) deficiency), identified this integrin as a functional Gal-1 receptor in platelets. Binding of Gal-1 to platelets triggered the phosphorylation of ß(3)-integrin, Syk, MAPKs, PI3K, PLCγ2, thromboxane (TXA(2)) release, and Ca(2+) mobilization. Not only soluble but also immobilized Gal-1 promoted platelet activation. Gal-1-deficient (Lgals1(-/-)) mice showed increased bleeding time (P<0.0002, knockout vs. wild type), which was not associated with an abnormal platelet count. Lgals1(-/-) platelets exhibited normal aggregation to PAR4, ADP, arachidonic acid, or collagen but abnormal ATP release at low collagen concentrations. Impaired spreading on fibrinogen and clot retraction with normal levels of α(IIb)ß(3) was also observed in Lgals1(-/-) platelets, indicating a failure in the "outside-in" signaling through this integrin. This study identifies a noncanonical mechanism, based on galectin-integrin interactions, for regulating platelet activation.


Asunto(s)
Galectina 1/sangre , Hemostasis/fisiología , Activación Plaquetaria/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Animales , Tiempo de Sangría , Galectina 1/deficiencia , Galectina 1/genética , Humanos , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Activación Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/fisiología , Unión Proteica , Transducción de Señal , Trombastenia/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA