Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunology ; 171(2): 181-197, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37885279

RESUMEN

Haemolytic disorders, such as sickle cell disease, are accompanied by the release of high amounts of labile heme into the intravascular compartment resulting in the induction of proinflammatory and prothrombotic complications in affected patients. In addition to the relevance of heme-regulated proteins from the complement and blood coagulation systems, activation of the TLR4 signalling pathway by heme was ascribed a crucial role in the progression of these pathological processes. Heme binding to the TLR4-MD2 complex has been proposed recently, however, essential mechanistic information of the processes at the molecular level, such as heme-binding kinetics, the heme-binding capacity and the respective heme-binding sites (HBMs) is still missing. We report the interaction of TLR4, MD2 and the TLR4-MD2 complex with heme and the consequences thereof by employing biochemical, spectroscopic, bioinformatic and physiologically relevant approaches. Heme binding occurs transiently through interaction with up to four HBMs in TLR4, two HBMs in MD2 and at least four HBMs in their complex. Functional studies highlight that mutations of individual HBMs in TLR4 preserve full receptor activation by heme, suggesting that heme interacts with TLR4 through different binding sites independently of MD2. Furthermore, we confirm and extend the major role of TLR4 for heme-mediated cytokine responses in human immune cells.


Asunto(s)
Transducción de Señal , Receptor Toll-Like 4 , Humanos , Receptor Toll-Like 4/metabolismo , Sitios de Unión , Citocinas/metabolismo , Antígeno 96 de los Linfocitos/metabolismo , Lipopolisacáridos
2.
Eur J Immunol ; 53(6): e2250016, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37061852

RESUMEN

Macrophage migration inhibitory factor (MIF) is present in high amounts in the BALF and serum of asthmatic patients, contributing to the pathogenesis of experimental asthma induced by OVA in mice. Whether MIF contributes to the physiopathology on a more complex and relevant asthma model has not been characterized. Mif-deficient (Mif-/- ) or WT mice treated with anti-MIF antibody were challenged multiple times using house dust mite (HDM) extract by the intranasal route. HDM-challenged Mif-/- mice presented decreased airway hyperresponsiveness, lung infiltration of eosinophils, mucus hypersecretion, and subepithelial fibrosis compared to HDM-challenged WT mice. Amounts of IL-4, IL-5, and IL-13 were decreased in the lungs of Mif-/- mice upon HDM challenges, but the increase of CCL11 was preserved, compared to HDM-challenged WT mice. We also observed increased numbers of group 2 innate lymphoid cells and Th2 cells in the BALF and mediastinal LNs (mLN)-induced challenged by HDM of WT mice, but not in HDM-challenged Mif-/- mice. Anti-MIF treatment abrogated the airway infiltration of eosinophils, mucus hypersecretion, and subepithelial fibrosis in the lungs of HDM-challenged mice. In conclusion, MIF ablation prevents the pathologic hallmarks of asthma in HDM-challenged mice, reinforcing the promising target of MIF for asthma therapy.


Asunto(s)
Asma , Factores Inhibidores de la Migración de Macrófagos , Animales , Ratones , Pyroglyphidae , Factores Inhibidores de la Migración de Macrófagos/genética , Inmunidad Innata , Linfocitos/patología , Pulmón , Inflamación/patología , Fibrosis
3.
Mol Microbiol ; 117(2): 293-306, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34783412

RESUMEN

Salmonellosis is a public health problem caused by Salmonella sp., a highly adapted facultative intracellular pathogen. After internalization, Salmonella sp. Manipulates several host processes, mainly through the activation of the type III secretion system (T3SS), including modification of host lipid metabolism and lipid droplet (LD) accumulation. LDs are dynamic and complex lipid-rich organelles involved in several cellular processes. The present study investigated the mechanism involved in LD biogenesis in Salmonella-infected macrophages and its role in bacterial pathogenicity. Here, we reported that S. Typhimurium induced a rapid time-dependent increase of LD formation in macrophages. The LD biogenesis was demonstrated to depend on Salmonella's viability and SPI1-related T3SS activity, with the participation of Toll-Like Receptor (TLR) signaling. We also observed that LD accumulation occurs through TLR2-dependent signaling and is counter-regulated by TLR4. Last, the pharmacologic modulation of LD formation by inhibiting diacylglycerol O-acyltransferase 1 (DGAT1) and cytosolic phospholipase A2 (cPLA2) significantly reduced the intracellular bacterial proliferation and impaired the prostaglandin E2 (PGE2 ) synthesis. Collectively, our data suggest the role of LDs on S. typhimurium intracellular survival and replication in macrophages. This data set provides new perspectives for future investigations about LDs in host-pathogen interaction.


Asunto(s)
Gotas Lipídicas , Infecciones por Salmonella , Humanos , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/fisiología , Macrófagos/microbiología , Sistemas de Secreción Tipo III/metabolismo
4.
PLoS Pathog ; 16(7): e1008599, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32692767

RESUMEN

Heme oxygenase (HO-1) mediates the enzymatic cleavage of heme, a molecule with proinflammatory and prooxidant properties. HO-1 activity deeply impacts host capacity to tolerate infection through reduction of tissue damage or affecting resistance, the ability of the host to control pathogen loads. In this Review, we will discuss the contribution of HO-1 in different and complex protozoan infections, such as malaria, leishmaniasis, Chagas disease, and toxoplasmosis. The complexity of these infections and the pleiotropic effects of HO-1 constitute an interesting area of study and an opportunity for drug development.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Infecciones por Protozoos/enzimología , Animales , Humanos , Tolerancia Inmunológica/fisiología
5.
PLoS Pathog ; 16(8): e1008230, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32797076

RESUMEN

Neutrophil extracellular traps (NETs) evolved as a unique effector mechanism contributing to resistance against infection that can also promote tissue damage in inflammatory conditions. Malaria infection can trigger NET release, but the mechanisms and consequences of NET formation in this context remain poorly characterized. Here we show that patients suffering from severe malaria had increased amounts of circulating DNA and increased neutrophil elastase (NE) levels in plasma. We used cultured erythrocytes and isolated human neutrophils to show that Plasmodium-infected red blood cells release macrophage migration inhibitory factor (MIF), which in turn caused NET formation by neutrophils in a mechanism dependent on the C-X-C chemokine receptor type 4 (CXCR4). NET production was dependent on histone citrullination by peptidyl arginine deiminase-4 (PAD4) and independent of reactive oxygen species (ROS), myeloperoxidase (MPO) or NE. In vitro, NETs functioned to restrain parasite dissemination in a mechanism dependent on MPO and NE activities. Finally, C57/B6 mice infected with P. berghei ANKA, a well-established model of cerebral malaria, presented high amounts of circulating DNA, while treatment with DNAse increased parasitemia and accelerated mortality, indicating a role for NETs in resistance against Plasmodium infection.


Asunto(s)
Eritrocitos/inmunología , Trampas Extracelulares/inmunología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Malaria/inmunología , Neutrófilos/inmunología , Plasmodium/inmunología , Receptores CXCR4/metabolismo , Animales , Eritrocitos/metabolismo , Eritrocitos/parasitología , Trampas Extracelulares/metabolismo , Trampas Extracelulares/parasitología , Humanos , Malaria/metabolismo , Malaria/parasitología , Malaria/patología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Neutrófilos/parasitología , Parasitemia/inmunología , Parasitemia/metabolismo , Parasitemia/parasitología , Parasitemia/patología
6.
J Immunol ; 205(10): 2795-2805, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33037139

RESUMEN

Hemolysis causes an increase of intravascular heme, oxidative damage, and inflammation in which macrophages play a critical role. In these cells, heme can act as a prototypical damage-associated molecular pattern, inducing TLR4-dependent cytokine production through the MyD88 pathway, independently of TRIF. Heme promotes reactive oxygen species (ROS) generation independently of TLR4. ROS and TNF production contribute to heme-induced necroptosis and inflammasome activation; however, the role of ROS in proinflammatory signaling and cytokine production remains unknown. In this study, we demonstrate that heme activates at least three signaling pathways that contribute to a robust MAPK phosphorylation and cytokine expression in mouse macrophages. Although heme did not induce a detectable Myddosome formation, the TLR4/MyD88 axis was important for phosphorylation of p38 and secretion of cytokines. ROS generation and spleen tyrosine kinase (Syk) activation induced by heme were critical for most proinflammatory signaling pathways, as the antioxidant N-acetyl-l-cysteine and a Syk inhibitor differentially blocked heme-induced ROS, MAPK phosphorylation, and cytokine production in macrophages. Early generated mitochondrial ROS induced by heme was Syk dependent, selectively promoted the phosphorylation of ERK1/2 without affecting JNK or p38, and contributed to CXCL1 and TNF production. Finally, lethality caused by sterile hemolysis in mice required TLR4, TNFR1, and mitochondrial ROS, supporting the rationale to target these pathways to mitigate tissue damage of hemolytic disorders.


Asunto(s)
Hemo/metabolismo , Hemólisis/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/inmunología , Animales , Quimiocina CXCL1/metabolismo , Modelos Animales de Enfermedad , Humanos , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Noqueados , Mitocondrias/inmunología , Mitocondrias/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Quinasa Syk/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
PLoS Pathog ; 14(4): e1006928, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29672619

RESUMEN

The activation of macrophage respiratory burst in response to infection with Trypanosoma cruzi inflicts oxidative damage to the host's tissues. For decades, the role of reactive oxygen species (ROS) in the elimination of T. cruzi was taken for granted, but recent evidence suggests parasite growth is stimulated in oxidative environments. It is still a matter of debate whether indeed oxidative environments provide ideal conditions (e.g., iron availability in macrophages) for T. cruzi growth and whether indeed ROS signals directly to stimulate growth. Nitric oxide (NO) and ROS combine to form peroxynitrite, participating in the killing of phagocytosed parasites by activated macrophages. In response to infection, mitochondrial ROS are produced by cardiomyocytes. They contribute to oxidative damage that persists at the chronic stage of infection and is involved in functional impairment of the heart. In this review, we discuss how oxidative stress helps parasite growth during the acute stage and how it participates in the development of cardiomyopathy at the chronic stage.


Asunto(s)
Enfermedad de Chagas/complicaciones , Cardiopatías/etiología , Macrófagos/microbiología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Trypanosoma cruzi/patogenicidad , Animales , Enfermedad de Chagas/microbiología , Humanos
8.
Proc Natl Acad Sci U S A ; 113(47): E7474-E7482, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27821769

RESUMEN

Hemolytic diseases include a variety of conditions with diverse etiologies in which red blood cells are destroyed and large amounts of hemeproteins are released. Heme has been described as a potent proinflammatory molecule that is able to induce multiple innate immune responses, such as those triggered by TLR4 and the NLRP3 inflammasome, as well as necroptosis in macrophages. The mechanisms by which eukaryotic cells respond to the toxic effects induced by heme to maintain homeostasis are not fully understood, however. Here we describe a previously uncharacterized cellular response induced by heme: the formation of p62/SQTM1 aggregates containing ubiquitinated proteins in structures known as aggresome-like induced structures (ALIS). This action is part of a response driven by the transcription factor NRF2 to the excessive generation of reactive oxygen species induced by heme that results in the expression of genes involved in antioxidant responses, including p62/SQTM1. Furthermore, we show that heme degradation by HO-1 is required for ALIS formation, and that the free iron released on heme degradation is necessary and sufficient to induce ALIS. Moreover, ferritin, a key protein in iron metabolism, prevents excessive ALIS formation. Finally, in vivo, hemolysis promotes an increase in ALIS formation in target tissues. Our data unravel a poorly understood aspect of the cellular responses induced by heme that can be explored to better understand the effects of free heme and free iron during hemolytic diseases such as sickle cell disease, dengue fever, malaria, and sepsis.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Hemo/metabolismo , Hierro/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Sequestosoma-1/metabolismo , Animales , Ferritinas/metabolismo , Células HEK293 , Hemo/química , Humanos , Ratones , Estrés Oxidativo , Agregado de Proteínas , Proteolisis , Células RAW 264.7 , Proteína Sequestosoma-1/química , Ubiquitinación , Regulación hacia Arriba
9.
PLoS Pathog ; 12(10): e1005947, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27788262

RESUMEN

Chronic chagasic cardiomyopathy (CCC) develops years after acute infection by Trypanosoma cruzi and does not improve after trypanocidal therapy, despite reduction of parasite burden. During disease, the heart undergoes oxidative stress, a potential causative factor for arrhythmias and contractile dysfunction. Here we tested whether antioxidants/ cardioprotective drugs could improve cardiac function in established Chagas heart disease. We chose a model that resembles B1-B2 stage of human CCC, treated mice with resveratrol and performed electrocardiography and echocardiography studies. Resveratrol reduced the prolonged PR and QTc intervals, increased heart rates and reversed sinus arrhythmia, atrial and atrioventricular conduction disorders; restored a normal left ventricular ejection fraction, improved stroke volume and cardiac output. Resveratrol activated the AMPK-pathway and reduced both ROS production and heart parasite burden, without interfering with vascularization or myocarditis intensity. Resveratrol was even capable of improving heart function of infected mice when treatment was started late after infection, while trypanocidal drug benznidazole failed. We attempted to mimic resveratrol's actions using metformin (AMPK-activator) or tempol (SOD-mimetic). Metformin and tempol mimicked the beneficial effects of resveratrol on heart function and decreased lipid peroxidation, but did not alter parasite burden. These results indicate that AMPK activation and ROS neutralization are key strategies to induce tolerance to Chagas heart disease. Despite all tissue damage observed in established Chagas heart disease, we found that a physiological dysfunction can still be reversed by treatment with resveratrol, metformin and tempol, resulting in improved heart function and representing a starting point to develop innovative therapies in CCC.


Asunto(s)
Antioxidantes/farmacología , Cardiomiopatía Chagásica/patología , Estilbenos/farmacología , Animales , Óxidos N-Cíclicos/farmacología , Modelos Animales de Enfermedad , Femenino , Masculino , Metformina/farmacología , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , Resveratrol , Marcadores de Spin
10.
J Immunol ; 196(12): 5056-63, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27183605

RESUMEN

Leishmaniasis is an important parasitic disease found in the tropics and subtropics. Cutaneous and visceral leishmaniasis affect an estimated 1.5 million people worldwide. Despite its human health relevance, relatively little is known about the cell death pathways that control Leishmania replication in the host. Necroptosis is a recently identified form of cell death with potent antiviral effects. Receptor interacting protein kinase 1 (RIPK1) is a critical kinase that mediates necroptosis downstream of death receptors and TLRs. Heme, a product of hemoglobin catabolism during certain intracellular pathogen infections, is also a potent inducer of macrophage necroptosis. We found that human visceral leishmaniasis patients exhibit elevated serum levels of heme. Therefore, we examined the impact of heme and necroptosis on Leishmania replication. Indeed, heme potently inhibited Leishmania replication in bone marrow-derived macrophages. Moreover, we found that inhibition of RIPK1 kinase activity also enhanced parasite replication in the absence of heme. We further found that the mitochondrial phosphatase phosphoglycerate mutase family member 5 (PGAM5), a putative downstream effector of RIPK1, was also required for inhibition of Leishmania replication. In mouse infection, both PGAM5 and RIPK1 kinase activity are required for IL-1ß expression in response to Leishmania However, PGAM5, but not RIPK1 kinase activity, was directly responsible for Leishmania-induced IL-1ß secretion and NO production in bone marrow-derived macrophages. Collectively, these results revealed that RIPK1 and PGAM5 function independently to exert optimal control of Leishmania replication in the host.


Asunto(s)
Interacciones Huésped-Parásitos , Leishmania/crecimiento & desarrollo , Leishmania/inmunología , Leishmaniasis/parasitología , Fosfoproteínas Fosfatasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Muerte Celular , Hemo/análisis , Hemo/farmacología , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Leishmania/efectos de los fármacos , Leishmaniasis/sangre , Leishmaniasis/inmunología , Leishmaniasis/microbiología , Leishmaniasis Visceral/sangre , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/fisiología , Ratones , Óxido Nítrico/biosíntesis , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores
11.
Mediators Inflamm ; 2017: 2086840, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28894350

RESUMEN

TLRs recognize a broad spectrum of microorganism molecules, triggering a variety of cellular responses. Among them, phagocytosis is a critical process for host defense. Leukotrienes (LTs), lipid mediators produced from 5-lipoxygenase (5-LO) enzyme, increase FcγR-mediated phagocytosis. Here, we evaluated the participation of TLR2, TLR3, TLR4, and TLR9 in FcγR-mediated phagocytosis and whether this process is modulated by LTs. Rat alveolar macrophages (AMs), murine bone marrow-derived macrophages (BMDMs), and peritoneal macrophages (PMs) treated with TLR2, TLR3, and TLR4 agonists, but not TLR9, enhanced IgG-opsonized sheep red blood cell (IgG-sRBC) phagocytosis. Pretreatment of AMs or BMDMs with drugs that block LT synthesis impaired the phagocytosis promoted by TLR ligands, and TLR potentiation was also abrogated in PMs and BMDMs from 5-LO-/- mice. LTB4 production induced by IgG engagement was amplified by TLR ligands, while cys-LTs were amplified by activation of TLR2 and TLR4, but not by TLR3. We also noted higher ERK1/2 phosphorylation in IgG-RBC-challenged cells when preincubated with TLR agonists. Furthermore, ERK1/2 inhibition by PD98059 reduced the phagocytic activity evoked by TLR agonists. Together, these data indicate that TLR2, TLR3, and TLR4 ligands, but not TLR9, amplify IgG-mediated phagocytosis by a mechanism which requires LT production and ERK-1/2 pathway activation.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Animales , Araquidonato 5-Lipooxigenasa/genética , Flavonoides/farmacología , Immunoblotting , Leucotrienos/metabolismo , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Noqueados , Fagocitosis/efectos de los fármacos , Fagocitosis/genética , Fagocitosis/fisiología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Ratas , Ratas Wistar , Ovinos , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
12.
Proc Natl Acad Sci U S A ; 111(39): E4110-8, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25225402

RESUMEN

The increase of extracellular heme is a hallmark of hemolysis or extensive cell damage. Heme has prooxidant, cytotoxic, and inflammatory effects, playing a central role in the pathogenesis of malaria, sepsis, and sickle cell disease. However, the mechanisms by which heme is sensed by innate immune cells contributing to these diseases are not fully characterized. We found that heme, but not porphyrins without iron, activated LPS-primed macrophages promoting the processing of IL-1ß dependent on nucleotide-binding domain and leucine rich repeat containing family, pyrin domain containing 3 (NLRP3). The activation of NLRP3 by heme required spleen tyrosine kinase, NADPH oxidase-2, mitochondrial reactive oxygen species, and K(+) efflux, whereas it was independent of heme internalization, lysosomal damage, ATP release, the purinergic receptor P2X7, and cell death. Importantly, our results indicated the participation of macrophages, NLRP3 inflammasome components, and IL-1R in the lethality caused by sterile hemolysis. Thus, understanding the molecular pathways affected by heme in innate immune cells might prove useful to identify new therapeutic targets for diseases that have heme release.


Asunto(s)
Hemo/metabolismo , Hemólisis/fisiología , Inflamasomas/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/deficiencia , Caspasa 1/genética , Caspasa 1/metabolismo , Hemo/química , Hemo/inmunología , Hemólisis/inmunología , Humanos , Inflamasomas/inmunología , Interleucina-1beta/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Potasio/metabolismo , Protoporfirinas/química , Protoporfirinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
13.
Blood ; 122(20): 3405-14, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24009231

RESUMEN

Dengue is the most frequent hemorrhagic viral disease and re-emergent infection in the world. Although thrombocytopenia is characteristically observed in mild and severe forms of dengue, the role of platelet activation in dengue pathogenesis has not been fully elucidated. We hypothesize that platelets have major roles in inflammatory amplification and increased vascular permeability during severe forms of dengue. Here we investigate interleukin (IL)-1ß synthesis, processing, and secretion in platelets during dengue virus (DV) infection and potential contribution of these events to endothelial permeability during infection. We observed increased expression of IL-1ß in platelets and platelet-derived microparticles from patients with dengue or after platelet exposure to DV in vitro. We demonstrated that DV infection leads to assembly of nucleotide-binding domain leucine rich repeat containing protein (NLRP3) inflammasomes, activation of caspase-1, and caspase-1-dependent IL-1ß secretion. Our findings also indicate that platelet-derived IL-1ß is chiefly released in microparticles through mechanisms dependent on mitochondrial reactive oxygen species-triggered NLRP3 inflammasomes. Inflammasome activation and platelet shedding of IL-1ß-rich microparticles correlated with signs of increased vascular permeability. Moreover, microparticles from DV-stimulated platelets induced enhanced permeability in vitro in an IL-1-dependent manner. Our findings provide new evidence that platelets contribute to increased vascular permeability in DV infection by inflammasome-dependent release of IL-1ß.


Asunto(s)
Plaquetas/metabolismo , Permeabilidad Capilar/fisiología , Proteínas Portadoras/fisiología , Dengue/fisiopatología , Endotelio Vascular/fisiopatología , Inflamasomas/fisiología , Interleucina-1beta/metabolismo , Adulto , Plaquetas/efectos de los fármacos , Plaquetas/ultraestructura , Caspasa 1/fisiología , Micropartículas Derivadas de Células/metabolismo , Dengue/sangre , Femenino , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/farmacología , Indoles/farmacología , Interleucina-1beta/biosíntesis , Masculino , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Compuestos Organofosforados/farmacología , Piperidinas/farmacología , Activación Plaquetaria , Especies Reactivas de Oxígeno/metabolismo , Clorometilcetona de Tosilfenilalanila/análogos & derivados , Clorometilcetona de Tosilfenilalanila/farmacología , Regulación hacia Arriba/efectos de los fármacos
14.
Blood ; 119(10): 2368-75, 2012 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-22262768

RESUMEN

Diseases that cause hemolysis or myonecrosis lead to the leakage of large amounts of heme proteins. Free heme has proinflammatory and cytotoxic effects. Heme induces TLR4-dependent production of tumor necrosis factor (TNF), whereas heme cytotoxicity has been attributed to its ability to intercalate into cell membranes and cause oxidative stress. We show that heme caused early macrophage death characterized by the loss of plasma membrane integrity and morphologic features resembling necrosis. Heme-induced cell death required TNFR1 and TLR4/MyD88-dependent TNF production. Addition of TNF to Tlr4(-/-) or to Myd88(-/-) macrophages restored heme-induced cell death. The use of necrostatin-1, a selective inhibitor of receptor-interacting protein 1 (RIP1, also known as RIPK1), or cells deficient in Rip1 or Rip3 revealed a critical role for RIP proteins in heme-induced cell death. Serum, antioxidants, iron chelation, or inhibition of c-Jun N-terminal kinase (JNK) ameliorated heme-induced oxidative burst and blocked macrophage cell death. Macrophages from heme oxygenase-1 deficient mice (Hmox1(-/-)) had increased oxidative stress and were more sensitive to heme. Taken together, these results revealed that heme induces macrophage necrosis through 2 synergistic mechanisms: TLR4/Myd88-dependent expression of TNF and TLR4-independent generation of ROS.


Asunto(s)
Hemo/farmacología , Macrófagos/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Imidazoles/farmacología , Indoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Células 3T3 NIH , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factores de Necrosis Tumoral/farmacología
15.
J Immunol ; 188(9): 4460-7, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22461696

RESUMEN

Visceral leishmaniasis (VL) remains a major public health problem worldwide. This disease is highly associated with chronic inflammation and a lack of the cellular immune responses against Leishmania. It is important to identify major factors driving the successful establishment of the Leishmania infection to develop better tools for the disease control. Heme oxygenase-1 (HO-1) is a key enzyme triggered by cellular stress, and its role in VL has not been investigated. In this study, we evaluated the role of HO-1 in the infection by Leishmania infantum chagasi, the causative agent of VL cases in Brazil. We found that L. chagasi infection or lipophosphoglycan isolated from promastigotes triggered HO-1 production by murine macrophages. Interestingly, cobalt protoporphyrin IX, an HO-1 inductor, increased the parasite burden in both mouse and human-derived macrophages. Upon L. chagasi infection, macrophages from Hmox1 knockout mice presented significantly lower parasite loads when compared with those from wild-type mice. Furthermore, upregulation of HO-1 by cobalt protoporphyrin IX diminished the production of TNF-α and reactive oxygen species by infected murine macrophages and increased Cu/Zn superoxide dismutase expression in human monocytes. Finally, patients with VL presented higher systemic concentrations of HO-1 than healthy individuals, and this increase of HO-1 was reduced after antileishmanial treatment, suggesting that HO-1 is associated with disease susceptibility. Our data argue that HO-1 has a critical role in the L. chagasi infection and is strongly associated with the inflammatory imbalance during VL. Manipulation of HO-1 pathways during VL could serve as an adjunctive therapeutic approach.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/inmunología , Hemo-Oxigenasa 1/inmunología , Leishmania/inmunología , Leishmaniasis Visceral/inmunología , Macrófagos Peritoneales/inmunología , Proteínas de la Membrana/inmunología , Animales , Brasil , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Glicoesfingolípidos/inmunología , Glicoesfingolípidos/metabolismo , Glicoesfingolípidos/farmacología , Hemo-Oxigenasa 1/biosíntesis , Hemo-Oxigenasa 1/genética , Humanos , Leishmania/metabolismo , Leishmaniasis Visceral/enzimología , Leishmaniasis Visceral/genética , Leishmaniasis Visceral/patología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/parasitología , Macrófagos Peritoneales/patología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , Fármacos Fotosensibilizantes/farmacología , Protoporfirinas/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
16.
Fungal Genet Biol ; 60: 64-73, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23608320

RESUMEN

The principal capsular component of Cryptococcus neoformans, glucuronoxylomannan (GXM), interacts with surface glycans, including chitin-like oligomers. Although the role of GXM in cryptococcal infection has been well explored, there is no information on how chitooligomers affect fungal pathogenesis. In this study, surface chitooligomers of C. neoformans were blocked through the use of the wheat germ lectin (WGA) and the effects on animal pathogenesis, interaction with host cells, fungal growth and capsule formation were analyzed. Treatment of C. neoformans cells with WGA followed by infection of mice delayed mortality relative to animals infected with untreated fungal cells. This observation was associated with reduced brain colonization by lectin-treated cryptococci. Blocking chitooligomers also rendered yeast cells less efficient in their ability to associate with phagocytes. WGA did not affect fungal viability, but inhibited GXM release to the extracellular space and capsule formation. In WGA-treated yeast cells, genes that are involved in capsule formation and GXM traffic had their transcription levels decreased in comparison with untreated cells. Our results suggest that cellular pathways required for capsule formation and pathogenic mechanisms are affected by blocking chitin-derived structures at the cell surface of C. neoformans. Targeting chitooligomers with specific ligands may reveal new therapeutic alternatives to control cryptococcosis.


Asunto(s)
Cryptococcus neoformans/patogenicidad , Cápsulas Fúngicas/metabolismo , Fagocitosis/efectos de los fármacos , Polisacáridos/metabolismo , Aglutininas del Germen de Trigo/farmacología , Animales , Encéfalo/microbiología , Quitina/metabolismo , Criptococosis/tratamiento farmacológico , Criptococosis/patología , Cryptococcus neoformans/crecimiento & desarrollo , Cryptococcus neoformans/metabolismo , Cápsulas Fúngicas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Aglutininas del Germen de Trigo/metabolismo
17.
J Immunol ; 186(11): 6562-7, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21536805

RESUMEN

High concentrations of free heme found during hemolytic events or cell damage leads to inflammation, characterized by neutrophil recruitment and production of reactive oxygen species, through mechanisms not yet elucidated. In this study, we provide evidence that heme-induced neutrophilic inflammation depends on endogenous activity of the macrophage-derived lipid mediator leukotriene B(4) (LTB(4)). In vivo, heme-induced neutrophil recruitment into the peritoneal cavity of mice was attenuated by pretreatment with 5-lipoxygenase (5-LO) inhibitors and leukotriene B(4) receptor 1 (BLT1) receptor antagonists as well as in 5-LO knockout (5-LO(-/-)) mice. Heme administration in vivo increased peritoneal levels of LTB(4) prior to and during neutrophil recruitment. Evidence that LTB(4) was synthesized by resident macrophages, but not mast cells, included the following: 1) immuno-localization of heme-induced LTB(4) was compartmentalized exclusively within lipid bodies of resident macrophages; 2) an increase in the macrophage population enhanced heme-induced neutrophil migration; 3) depletion of resident mast cells did not affect heme-induced LTB(4) production or neutrophil influx; 4) increased levels of LTB(4) were found in heme-stimulated peritoneal cavities displaying increased macrophage numbers; and 5) in vitro, heme was able to activate directly macrophages to synthesize LTB(4). Our findings uncover a crucial role of LTB(4) in neutrophil migration induced by heme and suggest that beneficial therapeutic outcomes could be achieved by targeting the 5-LO pathway in the treatment of inflammation associated with hemolytic processes.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Hemo/farmacología , Leucotrieno B4/metabolismo , Neutrófilos/efectos de los fármacos , Animales , Araquidonato 5-Lipooxigenasa/genética , Araquidonato 5-Lipooxigenasa/metabolismo , Células Cultivadas , Femenino , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/citología , Neutrófilos/metabolismo , Receptores de Leucotrieno B4/metabolismo , Tioglicolatos/farmacología , p-Metoxi-N-metilfenetilamina/farmacología
18.
iScience ; 26(7): 107219, 2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37529320

RESUMEN

The vast spectrum of clinical features of COVID-19 keeps challenging scientists and clinicians. Low resistance to infection might result in long-term viral persistence, but the underlying mechanisms remain unclear. Here, we studied the immune response of immunocompetent COVID-19 patients with prolonged SARS-CoV-2 infection by immunophenotyping, cytokine and serological analysis. Despite viral loads and symptoms comparable to regular mildly symptomatic patients, long-term carriers displayed weaker systemic IFN-I responses and fewer circulating pDCs and NK cells at disease onset. Type 1 cytokines remained low, while type-3 cytokines were in turn enhanced. Of interest, we observed no defects in antigen-specific cytotoxic T cell responses, and circulating antibodies displayed higher affinity against different variants of SARS-CoV-2 Spike protein in these patients. The identification of distinct immune responses in long-term carriers adds up to our understanding of essential host protective mechanisms to ensure tissue damage control despite prolonged viral infection.

19.
Microvasc Res ; 83(2): 185-93, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22036674

RESUMEN

Experiments were designed to determine if the vasodilatory peptides maxadilan and pituitary adenylate cyclase-activating peptide (PACAP-38) may cause plasma leakage through activation of leukocytes and to what extent these effects could be due to PAC1 and CXCR1/2 receptor stimulation. Intravital microscopy of hamster cheek pouches utilizing FITC-dextran and rhodamine, respectively, as plasma and leukocyte markers was used to measure arteriolar diameter, plasma leakage and leukocyte accumulation in a selected area (5mm(2)) representative of the hamster cheek pouch microcirculation. Our studies showed that the sand fly vasodilator maxadilan and PACAP-38 induced arteriolar dilation, leukocyte accumulation and plasma leakage in postcapillary venules. The recombinant mutant of maxadilan M65 and an antagonist of CXCR1/2 receptors, reparixin, and an inhibitor of CD11b/CD18 up-regulation, ropivacaine, inhibited all these effects as induced by maxadilan. Dextran sulfate, a complement inhibitor with heparin-like anti-inflammatory effects, inhibited plasma leakage and leukocyte accumulation but not arteriolar dilation as induced by maxadilan and PACAP-38. In vitro studies with isolated human neutrophils showed that maxadilan is a potent stimulator of neutrophil migration comparable with fMLP and leukotriene B(4) and that M65 and reparixin inhibited such migration. The data suggest that leukocyte accumulation and plasma leakage induced by maxadilan involves a mechanism related to PAC1- and CXCR1/2-receptors on leukocytes and endothelial cells.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Mejilla/irrigación sanguínea , Proteínas de Insectos/farmacología , Psychodidae , Receptores de Interleucina-8A/efectos de los fármacos , Receptores de Interleucina-8B/efectos de los fármacos , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Células Cultivadas , Quimiotaxis de Leucocito/efectos de los fármacos , Cricetinae , Dextranos/metabolismo , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Colorantes Fluorescentes/metabolismo , Humanos , Proteínas de Insectos/genética , Proteínas de Insectos/aislamiento & purificación , Microscopía Fluorescente , Microscopía por Video , Mutación , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Psychodidae/química , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Proteínas Recombinantes/farmacología , Rodaminas/metabolismo , Factores de Tiempo , Vasodilatadores/aislamiento & purificación , Vénulas/efectos de los fármacos , Vénulas/metabolismo
20.
J Immunol ; 185(2): 1196-204, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20562262

RESUMEN

In many hemolytic disorders, such as malaria, the release of free heme has been involved in the triggering of oxidative stress and tissue damage. Patients presenting with severe forms of malaria commonly have impaired regulatory responses. Although intriguing, there is scarce data about the involvement of heme on the regulation of immune responses. In this study, we investigated the relation of free heme and the suppression of anti-inflammatory mediators such as PGE(2) and TGF-beta in human vivax malaria. Patients with severe disease presented higher hemolysis and higher plasma concentrations of Cu/Zn superoxide dismutase (SOD-1) and lower concentrations of PGE(2) and TGF-beta than those with mild disease. In addition, there was a positive correlation between SOD-1 concentrations and plasma levels of TNF-alpha. During antimalaria treatment, the concentrations of plasma SOD-1 reduced whereas PGE(2) and TGF-beta increased in the individuals severely ill. Using an in vitro model with human mononuclear cells, we demonstrated that the heme effect on the impairment of the production of PGE(2) and TGF-beta partially involves heme binding to CD14 and depends on the production of SOD-1. Aside from furthering the current knowledge about the pathogenesis of vivax malaria, the present results may represent a general mechanism for hemolytic diseases and could be useful for future studies of therapeutic approaches.


Asunto(s)
Dinoprostona/metabolismo , Hemo/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adolescente , Adulto , Células Cultivadas , Dinoprostona/sangre , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/inmunología , Eritrocitos/parasitología , Femenino , Citometría de Flujo , Hemólisis , Humanos , Inflamación/inmunología , Inflamación/parasitología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Receptores de Lipopolisacáridos/sangre , Receptores de Lipopolisacáridos/metabolismo , Malaria Vivax/sangre , Malaria Vivax/parasitología , Masculino , Persona de Mediana Edad , Plasmodium vivax/inmunología , Plasmodium vivax/fisiología , Interferencia de ARN , Superóxido Dismutasa/sangre , Superóxido Dismutasa/genética , Factor de Crecimiento Transformador beta/sangre , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA