Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Trends Immunol ; 44(12): 1014-1030, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37951789

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a fatal chronic interstitial lung disease (ILD) that affects lung mechanical functions and gas exchange. IPF is caused by increased fibroblast activity and collagen deposition that compromise the alveolar-capillary barrier. Identifying an effective therapy for IPF remains a clinical challenge. Chemokines are key proteins in cell communication that have functions in immunity as well as in tissue homeostasis, damage, and repair. Chemokine receptor signaling induces the activation and proliferation of lung-resident cells, including alveolar macrophages (AMs) and fibroblasts. AMs are an important source of chemokines and cytokines during IPF. We highlight the complexity of this system and, based on insights from genetic and transcriptomic studies, propose a new role for homeostatic chemokine imbalance in IPF, with implications for putative therapeutic targets.


Asunto(s)
Fibrosis Pulmonar Idiopática , Humanos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/metabolismo , Quimiocinas/metabolismo , Macrófagos Alveolares , Citocinas/metabolismo , Transducción de Señal , Pulmón
2.
Allergy ; 77(11): 3320-3336, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35615773

RESUMEN

BACKGROUND: Inflammasomes are large protein complexes that assemble in the cytosol in response to danger such as tissue damage or infection. Following activation, inflammasomes trigger cell death and the release of biologically active forms of pro-inflammatory cytokines interleukin (IL)-1ß and IL-18. NOD-like receptor family pyrin domain containing 6 (NLRP6) inflammasome is required for IL-18 secretion by intestinal epithelial cells, macrophages, and T cells, contributing to homeostasis and self-defense against pathogenic microbes. However, the involvement of NLRP6 in type 2 lung inflammation remains elusive. METHODS: Wild-type (WT) and Nlrp6-/- mice were used. Birch pollen extract (BPE)-induced allergic lung inflammation, eosinophil recruitment, Th2-related cytokine and chemokine production, airway hyperresponsiveness, and lung histopathology, Th2 cell differentiation, GATA3, and Th2 cytokines expression, were determined. Nippostrongylus brasiliensis (Nb) infection, worm count in intestine, type 2 innate lymphoid cell (ILC2), and Th2 cells in lungs were evaluated. RESULTS: We demonstrate in Nlrp6-/- mice that a mixed Th2/Th17 immune responses prevailed following birch pollen challenge with increased eosinophils, ILC2, Th2, and Th17 cell induction and reduced IL-18 production. Nippostrongylus brasiliensis infected Nlrp6-/- mice featured enhanced early expulsion of the parasite due to enhanced type 2 immune responses compared to WT hosts. In vitro, NLRP6 repressed Th2 polarization, as shown by increased Th2 cytokines and higher expression of the transcription factor GATA3 in the absence of NLRP6. Exogenous IL-18 administration partially reduced the enhanced airways inflammation in Nlrp6-/- mice. CONCLUSIONS: In summary, our data identify NLRP6 as a negative regulator of type 2 immune responses.


Asunto(s)
Inmunidad Innata , Neumonía , Animales , Ratones , Citocinas/metabolismo , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Linfocitos , Ratones Noqueados , Nippostrongylus , Neumonía/metabolismo , Células Th2
3.
Trends Immunol ; 40(8): 719-734, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31262653

RESUMEN

Self-DNA sensing by the immune system has emerged as a key contributing response in the pathogenesis of cancer and autoimmune diseases. Recent studies have established that release of nuclear and mitochondrial DNA can also drive lung inflammatory diseases. Here, we review the latest advances on self-DNA sensing and signaling, the influence of these pathways on lung inflammation, and how these findings contribute to our understanding of basic mechanisms of innate immunity. Within a dozen DNA sensors, the cGAS/STING, inflammasomes and Toll-Like Receptor pathways are central to nucleic acid sensing. We propose a key role for the STING pathway in self-DNA sensing in inflammatory lung conditions, and identify major remaining questions that may further our understanding and potential to control self-DNA sensing and innate immune activation.


Asunto(s)
ADN/inmunología , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno/inmunología , Neumonía/etiología , Neumonía/metabolismo , Animales , Autoinmunidad , Biomarcadores , Susceptibilidad a Enfermedades/inmunología , Humanos , Inmunidad Innata , Inflamasomas/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal
4.
J Neuroinflammation ; 17(1): 268, 2020 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917228

RESUMEN

BACKGROUND: Interleukin (IL)-33 is expressed in a healthy brain and plays a pivotal role in several neuropathologies, as protective or contributing to the development of cerebral diseases associated with cognitive impairments. However, the role of IL-33 in the brain is poorly understood, raising the question of its involvement in immunoregulatory mechanisms. METHODS: We administered recombinant IL-33 (rmIL-33) by intra-hippocampal injection to C57BL/6 J (WT) and IL-1αß deficient mice. Chronic minocycline administration was performed and cognitive functions were examined trough spatial habituation test. Hippocampal inflammatory responses were investigated by RT-qPCR. The microglia activation was assessed using immunohistological staining and fluorescence-activated cell sorting (FACS). RESULTS: We showed that IL-33 administration in mice led to a spatial memory performance defect associated with an increase of inflammatory markers in the hippocampus while minocycline administration limited the inflammatory response. Quantitative assessment of glial cell activation in situ demonstrated an increase of proximal intersections per radius in each part of the hippocampus. Moreover, rmIL-33 significantly promoted the outgrowth of microglial processes. Fluorescence-activated cell sorting analysis on isolated microglia, revealed overexpression of IL-1ß, 48 h post-rmIL-33 administration. This microglial reactivity was closely related to the onset of cognitive disturbance. Finally, we demonstrated that IL-1αß deficient mice were resistant to cognitive disorders after intra-hippocampal IL-33 injection. CONCLUSION: Thus, hippocampal IL-33 induced an inflammatory state, including IL-1ß overexpression by microglia cells, being causative of the cognitive impairment. These results highlight the pathological role for IL-33 in the central nervous system, independently of a specific neuropathological model.


Asunto(s)
Disfunción Cognitiva/metabolismo , Hipocampo/metabolismo , Inflamación/metabolismo , Interleucina-33/farmacología , Animales , Disfunción Cognitiva/etiología , Hipocampo/efectos de los fármacos , Inflamación/complicaciones , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/metabolismo , Minociclina/farmacología , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología
5.
Cell Commun Signal ; 18(1): 141, 2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32894139

RESUMEN

BACKGROUND: Low molecular weight carrageenan (Cg) is a seaweed-derived sulfated polysaccharide widely used as inflammatory stimulus in preclinical studies. However, the molecular mechanisms of Cg-induced inflammation are not fully elucidated. The present study aimed to investigate the molecular basis involved in Cg-induced macrophages activation and cytokines production. METHODS: Primary culture of mouse peritoneal macrophages were stimulated with Kappa Cg. The supernatant and cell lysate were used for ELISA, western blotting, immunofluorescence. Cg-induced mouse colitis was also developed. RESULTS: Here we show that Cg activates peritoneal macrophages to produce pro-inflammatory cytokines such as TNF and IL-1ß. While Cg-induced TNF production/secretion depends on TLR4/MyD88 signaling, the production of pro-IL-1ß relies on TLR4/TRIF/SYK/reactive oxygen species (ROS) signaling pathway. The maturation of pro-IL1ß into IL-1ß is dependent on canonical NLRP3 inflammasome activation via Pannexin-1/P2X7/K+ efflux signaling. In vivo, Cg-induced colitis was reduced in mice in the absence of NLRP3 inflammasome components. CONCLUSIONS: In conclusion, we unravel a critical role of the NLRP3 inflammasome in Cg-induced pro-inflammatory cytokines production and colitis, which is an important discovery on the pro-inflammatory properties of this sulfated polysaccharide for pre-clinical studies. Video abstract Carrageenan (Cg) is one the most used flogistic stimulus in preclinical studies. Nevertheless, the molecular basis of Cg-induced inflammation is not totally elucidated. Herein, Lopes et al. unraveled the molecular basis for Cg-induced macrophages production of biological active IL-1ß. The Cg-stimulated macrophages produces pro-IL-1ß depends on TLR4/TRIF/Syk/ROS, whereas its processing into mature IL-1ß is dependent on the canonical NLRP3 inflammasome.


Asunto(s)
Carragenina/inmunología , Citocinas/inmunología , Activación de Macrófagos , Macrófagos Peritoneales/inmunología , Animales , Células Cultivadas , Inflamasomas/inmunología , Inflamación/inmunología , Interleucina-1beta/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Factor de Necrosis Tumoral alfa/inmunología
6.
PLoS Pathog ; 13(4): e1006322, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28448579

RESUMEN

Cerebral malaria (CM) is associated with a high mortality rate and long-term neurocognitive impairment in survivors. The murine model of experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA (PbA)-infection reproduces several of these features. We reported recently increased levels of IL-33 protein in brain undergoing ECM and the involvement of IL-33/ST2 pathway in ECM development. Here we show that PbA-infection induced early short term and spatial memory defects, prior to blood brain barrier (BBB) disruption, in wild-type mice, while ST2-deficient mice did not develop cognitive defects. PbA-induced neuroinflammation was reduced in ST2-deficient mice with low Ifng, Tnfa, Il1b, Il6, CXCL9, CXCL10 and Cd8a expression, associated with an absence of neurogenesis defects in hippocampus. PbA-infection triggered a dramatic increase of IL-33 expression by oligodendrocytes, through ST2 pathway. In vitro, IL-33/ST2 pathway induced microglia expression of IL-1ß which in turn stimulated IL-33 expression by oligodendrocytes. These results highlight the IL-33/ST2 pathway ability to orchestrate microglia and oligodendrocytes responses at an early stage of PbA-infection, with an amplification loop between IL-1ß and IL-33, responsible for an exacerbated neuroinflammation context and associated neurological and cognitive defects.


Asunto(s)
Encéfalo/metabolismo , Disfunción Cognitiva/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Malaria Cerebral/complicaciones , Plasmodium berghei/fisiología , Animales , Encéfalo/parasitología , Encéfalo/fisiopatología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/genética , Disfunción Cognitiva/parasitología , Femenino , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-33/genética , Malaria Cerebral/genética , Malaria Cerebral/metabolismo , Malaria Cerebral/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/genética
7.
J Allergy Clin Immunol ; 139(5): 1650-1666, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27746240

RESUMEN

BACKGROUND: Protein kinase C (PKC) θ, a serine/threonine kinase, is involved in TH2 cell activation and proliferation. Type 2 innate lymphoid cells (ILC2s) resemble TH2 cells and produce the TH2 cytokines IL-5 and IL-13 but lack antigen-specific receptors. The mechanism by which PKC-θ drives innate immune cells to instruct TH2 responses in patients with allergic lung inflammation remains unknown. OBJECTIVES: We hypothesized that PKC-θ contributes to ILC2 activation and might be necessary for ILC2s to instruct the TH2 response. METHODS: PRKCQ gene expression was assessed in innate lymphoid cell subsets purified from human PBMCs and mouse lung ILC2s. ILC2 activation and eosinophil recruitment, TH2-related cytokine and chemokine production, lung histopathology, interferon regulatory factor 4 (IRF4) mRNA expression, and nuclear factor of activated T cells (NFAT1) protein expression were determined. Adoptive transfer of ILC2s from wild-type mice was performed in wild-type and PKC-θ-deficient (PKC-θ-/-) mice. RESULTS: Here we report that PKC-θ is expressed in both human and mouse ILC2s. Mice lacking PKC-θ had reduced ILC2 numbers, TH2 cell numbers and activation, airway hyperresponsiveness, and expression of the transcription factors IRF4 and NFAT1. Importantly, adoptive transfer of ILC2s restored eosinophil influx and IL-4, IL-5 and IL-13 production in lung tissue, as well as TH2 cell activation. The pharmacologic PKC-θ inhibitor (Compound 20) administered during allergen challenge reduced ILC2 numbers and activation, as well as airway inflammation and IRF4 and NFAT1 expression. CONCLUSIONS: Therefore our findings identify PKC-θ as a critical factor for ILC2 activation that contributes to TH2 cell differentiation, which is associated with IRF4 and NFAT1 expression in allergic lung inflammation.


Asunto(s)
Alérgenos/inmunología , Antígenos Dermatofagoides/inmunología , Asma/inmunología , Isoenzimas/inmunología , Linfocitos/inmunología , Proteína Quinasa C/inmunología , Animales , Líquido del Lavado Bronquioalveolar/citología , Diferenciación Celular , Citocinas/inmunología , Dipéptidos/farmacología , Femenino , Humanos , Inmunidad Innata , Factores Reguladores del Interferón/inmunología , Isoenzimas/genética , Recuento de Leucocitos , Pulmón/citología , Pulmón/inmunología , Pulmón/patología , Linfocitos/citología , Linfocitos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFATC/inmunología , Proteína Quinasa C/genética , Proteína Quinasa C-theta , Inhibidores de Proteínas Quinasas/farmacología
8.
Int J Mol Sci ; 19(7)2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973541

RESUMEN

Pleural tuberculosis is one of the most frequent forms of extra-pulmonary tuberculosis observed in patients infected with Mycobacterium tuberculosis. Tumor Necrosis Factor (TNF) is a crucial cytokine needed to control tuberculosis infection that remains a leading cause of morbidity and mortality worldwide. TNF blockade compromises host immunity and may increase the risk of reactivation of latent infection resulting in overt pulmonary, pleural and extra-pulmonary tuberculosis. While TNF signaling is mainly considered pro-inflammatory, its requirement for the anti-inflammation process involved in the resolution of infection and tissue repair is less explored. Our study analyzes the role of TNF and TNF receptors in the control of the inflammatory process associated with Bacillus Calmette-Guérin (BCG)-induced pleurisy. This study shows that the absence of TNF causes exacerbated inflammation in the pleural cavity of BCG-infected mice which is controlled by the transmembrane TNF (tmTNF) expression. The lack of TNF is associated with an impaired cellular expression and shedding of TNFR2 in the pleural cavity. The presence of tmTNF restores the normal expression of TNFR2 on myeloid cells during BCG-induced pleurisy. We also show that absence of TNFR1 affects the expression of TNFR2 on pleural cells and inflammation in the pleural cavity of BCG-infected mice. In conclusion, tmTNF but not soluble TNF prevents pleural cavity inflammation leading to attenuation and the resolution of the inflammatory process caused by mycobacterial pleurisy in association with the expression of TNFR2 on myeloid cells.


Asunto(s)
Inflamación/inmunología , Receptores del Factor de Necrosis Tumoral/metabolismo , Tuberculosis Pleural/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium bovis/inmunología , Cavidad Pleural/citología , Cavidad Pleural/patología , Receptores del Factor de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/genética
9.
J Infect Dis ; 216(7): 907-918, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-28973651

RESUMEN

Background: Recent evidence indicates a robust competition between the host and mycobacteria for iron acquisition during mycobacterial infection. Variable effects of iron supplementation on the susceptibility to mycobacterial infection have been reported. In this study, we revisited the effects of an experimental iron-enriched diet on Mycobacterium bovis bacille Calmette-Guerin (BCG) infection. Methods: Mice fed a standard diet or a diet moderately enriched with iron were infected with M. bovis BCG expressing green fluorescent protein. Colony-forming unit numbers, host myeloid cell counts, cell recruitment, cytokine production, and iron gene expression were determined at different stages of infection. Bone marrow-derived macrophages incubated with or without iron were also used to measure bacterial uptake, levels of inflammation markers, and iron gene expression. Results: In vivo analysis of BCG-infected mice revealed that moderate iron supplementation reduced inflammation, as measured by decreased proinflammatory cytokine levels and neutrophil recruitment and enhanced T-cell recruitment in granulomas, and decreased the bacterial load. Enhanced bacterial clearance in the liver correlated with upregulation of the gene encoding hepcidin, which is known to have antimicrobial proprieties, and with sequestration of iron in tissues. In cultured macrophages, iron supplementation induced reactive oxygen species and reduced uptake and intracellular growth of BCG. Conclusion: Moderate iron diet supplementation diminished inflammation and growth of M. bovis BCG via enhanced reactive oxygen species production, immune cell activation, and local hepcidin expression.


Asunto(s)
Citocinas/metabolismo , Hepcidinas/metabolismo , Hierro de la Dieta/farmacología , Mycobacterium bovis/inmunología , Linfocitos T/fisiología , Tuberculosis/microbiología , Animales , Citocinas/genética , Hepcidinas/genética , Hierro/metabolismo , Hígado/metabolismo , Hígado/microbiología , Pulmón/metabolismo , Pulmón/microbiología , Ratones , Tuberculosis/inmunología , Regulación hacia Arriba
10.
Am J Pathol ; 186(9): 2364-77, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27456129

RESUMEN

Tumor necrosis factor (TNF) is crucial to control Mycobacterium tuberculosis infection, which remains a leading cause of morbidity and mortality worldwide. TNF blockade compromises host immunity and may cause reactivation of latent infection, resulting in overt pulmonary, pleural, and extrapulmonary tuberculosis. Herein, we investigate the roles of TNF and TNF receptors in the control of Mycobacterium bovis bacillus Calmette-Guerin (BCG) pleural infection in a murine model. As controls, wild-type mice and those with a defective CCR5, a receptor that is crucial for control of viral infection but not for tuberculosis, were used. BCG-induced pleural infection was uncontrolled and progressive in absence of TNF or TNF receptor 1 (TNFR1)/TNFR2 (TNFR1R2) with increased inflammatory cell recruitment and bacterial load in the pleural cavity, and heightened levels of pleural and serum proinflammatory cytokines and chemokines, compared to wild-type control mice. The visceral pleura was thickened with chronic inflammation, which was prominent in TNF(-/-) and TNFR1R2(-/-) mice. The parietal pleural of TNF(-/-) and TNFR1R2(-/-) mice exhibited abundant inflammatory nodules containing mycobacteria, and these mice developed nonresolving inflammation and succumbed from disseminated BCG infection. By contrast, CCR5(-/-) mice survived and controlled pleural BCG infection as wild-type control mice. In conclusion, BCG-induced pleurisy was uncontrolled in the absence of TNF or TNF receptors with exacerbated inflammatory response, impaired bacterial clearance, and defective mesothelium repair, suggesting a critical role of TNF to control mycobacterial pleurisy.


Asunto(s)
Receptores del Factor de Necrosis Tumoral/inmunología , Tuberculosis Pleural/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mycobacterium bovis , Tuberculosis Pleural/patología
11.
FASEB J ; 30(1): 54-65, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26310268

RESUMEN

Neuropathic pain from injury to the peripheral and CNS represents a major health care issue. We have investigated the role of IL-33/IL-33 receptor (ST2) signaling in experimental models of neuropathic pain in mice. Chronic constriction injury (CCI) of the sciatic nerve induced IL-33 production in the spinal cord. IL-33/citrine reporter mice revealed that oligodendrocytes are the main cells expressing IL-33 within the spinal cord together with a minor expression by neurons, microglia. and astrocytes. CCI-induced mechanical hyperalgesia was reduced in IL-33R (ST2)(-/ -) mice compared with wild-type (WT) mice. Intrathecal treatment of WT mice with soluble IL-33 receptor (IL-33 decoy receptor) markedly reduced CCI-induced hyperalgesia. Consistent with these observations, intrathecal injection of IL-33 enhanced CCI hyperalgesia and induced hyperalgesia in naive mice. IL-33-mediated hyperalgesia during CCI was dependent on a reciprocal relationship with TNF-α and IL-1ß. IL-33-induced hyperalgesia was markedly attenuated by inhibitors of PI3K, mammalian target of rapamycin, MAPKs (p38, ERK, and JNK), NF-κB, and also by the inhibitors of glial cells (microglia and astrocytes). Furthermore, targeting these signaling pathways and cells inhibited IL-33-induced TNF-α and IL-1ß production in the spinal cord. Our study, therefore, reveals an important role of oligodendrocyte-derived IL-33 in neuropathic pain.


Asunto(s)
Alarminas/metabolismo , Hiperalgesia/metabolismo , Interleucina-33/metabolismo , Neuralgia/metabolismo , Oligodendroglía/metabolismo , Médula Espinal/metabolismo , Animales , Astrocitos/metabolismo , Ratones Noqueados , Microglía/metabolismo , Umbral del Dolor/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Médula Espinal/fisiopatología
12.
Eur J Immunol ; 45(5): 1354-65, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25682948

RESUMEN

Cerebral malaria, a severe complication of Plasmodium falciparum infection, can be modeled in murine Plasmodium berghei ANKA (PbA) infection. PbA-induced experimental cerebral malaria (ECM) is CD8(+) T-cell mediated, and influenced by TH 1/TH 2 balance. Here, we show that IL-33 expression is increased in brain undergoing ECM and we address the role of the IL-33/ST2 pathway in ECM development. ST2-deficient mice were resistant to PbA-induced neuropathology. They survived >20 days with no ECM neurological sign and a preserved cerebral microcirculation, while WT mice succumbed within 10 days with ECM, brain vascular leakage, distinct microvascular pathology obstruction, and hemorrhages. Parasitemia and brain parasite load were similar in ST2-deficient and WT mice. Protection was accompanied by reduced brain sequestration of activated CD4(+) T cells and perforin(+) CD8(+) T cells. While IFN-γ and T-cell-attracting chemokines CXCL9 and CXCL10 were not affected in the absence of functional ST2 pathway, the local expression of ICAM-1, CXCR3, and LT-α, crucial for ECM development, was strongly reduced, and this may explain the diminished pathogenic T-cell recruitment and resistance to ECM. Therefore, IL-33 is induced in PbA sporozoite infection, and the pathogenic T-cell responses with local microvascular pathology are dependent on IL-33/ST2 signaling, identifying IL-33 as a new actor in ECM development.


Asunto(s)
Malaria Cerebral/etiología , Plasmodium berghei , Receptores de Interleucina/metabolismo , Animales , Encéfalo/inmunología , Encéfalo/parasitología , Encéfalo/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Modelos Animales de Enfermedad , Femenino , Inflamación/etiología , Inflamación/inmunología , Inflamación/patología , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Interleucinas/metabolismo , Activación de Linfocitos , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium berghei/inmunología , Plasmodium berghei/patogenicidad , Receptores de Interleucina/deficiencia , Receptores de Interleucina/genética
13.
Clin Sci (Lond) ; 130(21): 1939-54, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27549113

RESUMEN

Glufosinate-ammonium (GLA), the active component of an herbicide, is known to cause neurotoxicity. GLA shares structural analogy with glutamate. It is a powerful inhibitor of glutamine synthetase (GS) and may bind to glutamate receptors. Since these potentials targets of GLA are present in lung and immune cells, we asked whether airway exposure to GLA may cause lung inflammation in mice. A single GLA exposure (1 mg/kg) induced seizures and inflammatory cell recruitment in the broncho-alveolar space, and increased myeloperoxidase (MPO), inducible NO synthase (iNOS), interstitial inflammation and disruption of alveolar septae within 6-24 h. Interleukin 1ß (IL-1ß) was increased and lung inflammation depended on IL-1 receptor 1 (IL-1R1). We demonstrate that glutamate receptor pathway is central, since the N-methyl-D-aspartate (NMDA) receptor inhibitor MK-801 prevented GLA-induced lung inflammation. Chronic exposure (0.2 mg/kg 3× per week for 4 weeks) caused moderate lung inflammation and enhanced airway hyperreactivity with significant increased airway resistance. In conclusion, GLA aerosol exposure causes glutamate signalling and IL-1R-dependent pulmonary inflammation with airway hyperreactivity in mice.


Asunto(s)
Aminobutiratos/toxicidad , Ácido Glutámico/inmunología , Herbicidas/toxicidad , Interleucina-1beta/inmunología , Neumonía/inmunología , Receptores de Interleucina-1/inmunología , Receptores de N-Metil-D-Aspartato/metabolismo , Aminobutiratos/inmunología , Animales , Herbicidas/inmunología , Humanos , Interleucina-1beta/genética , Ratones , Ratones Endogámicos C57BL , N-Metilaspartato , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Peroxidasa/genética , Peroxidasa/inmunología , Neumonía/etiología , Receptores de Interleucina-1/genética , Receptores de N-Metil-D-Aspartato/genética
14.
Biochim Biophys Acta ; 1841(1): 97-107, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24120921

RESUMEN

The nuclear receptor PPARγ acts as a key modulator of lipid metabolism, inflammation and pathogenesis in BCG-infected macrophages. However, the molecular mechanisms involved in PPARγ expression and functions during infection are not completely understood. Here, we investigate signaling pathways triggered by TLR2, the involvement of co-receptors and lipid rafts in the mechanism of PPARγ expression, lipid body formation and cytokine synthesis in macrophages during BCG infection. BCG induces NF-κB activation and increased PPARγ expression in a TLR2-dependent manner. Furthermore, BCG-triggered increase of lipid body biogenesis was inhibited by the PPARγ antagonist GW9662, but not by the NF-κB inhibitor JSH-23. In contrast, KC/CXCL1 production was largely dependent on NF-κB but not on PPARγ. BCG infection induced increased expression of CD36 in macrophages in vitro. Moreover, CD36 co-immunoprecipitates with TLR2 in BCG-infected macrophages, suggesting its interaction with TLR2 in BCG signaling. Pretreatment with CD36 neutralizing antibodies significantly inhibited PPARγ expression, lipid body formation and PGE2 production induced by BCG. Involvement of CD36 in lipid body formation was further confirmed by decreased BCG-induced lipid body formation in CD36 deficient macrophages. Similarly, CD14 and CD11b/CD18 blockage also inhibited BCG-induced lipid body formation, whereas TNF-α synthesis was not affected. Disruption of rafts recapitulates the latter result, inhibiting lipid body formation, but not TNF-α synthesis in BCG-infected macrophages. In conclusion, our results suggest that CD36-TLR2 cooperation and signaling compartmentalization within rafts, divert host response signaling through PPARγ-dependent and NF-κB-independent pathways, leading to increased macrophage lipid accumulation and down-modulation of macrophage response.


Asunto(s)
Quimiocina CXCL1/biosíntesis , Metabolismo de los Lípidos , Mycobacterium bovis , Transducción de Señal , Receptor Toll-Like 2/metabolismo , Tuberculosis , Factor de Necrosis Tumoral alfa/biosíntesis , Anilidas/farmacología , Animales , Antígeno CD11b/biosíntesis , Antígeno CD11b/genética , Antígenos CD18/biosíntesis , Antígenos CD18/genética , Antígenos CD36/biosíntesis , Antígenos CD36/genética , Quimiocina CXCL1/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Receptores de Lipopolisacáridos/biosíntesis , Receptores de Lipopolisacáridos/genética , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/patología , Microdominios de Membrana/genética , Microdominios de Membrana/metabolismo , Microdominios de Membrana/patología , Ratones , Ratones Noqueados , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , PPAR gamma/antagonistas & inhibidores , PPAR gamma/biosíntesis , PPAR gamma/genética , Fenilendiaminas/farmacología , Receptor Toll-Like 2/genética , Tuberculosis/metabolismo , Tuberculosis/patología , Tuberculosis/veterinaria , Factor de Necrosis Tumoral alfa/genética
15.
Infect Immun ; 83(9): 3612-23, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26123801

RESUMEN

Tumor necrosis factor (TNF) is an important cytokine for host defense against pathogens but is also associated with the development of human immunopathologies. TNF blockade effectively ameliorates many chronic inflammatory conditions but compromises host immunity to tuberculosis. The search for novel, more specific human TNF blockers requires the development of a reliable animal model. We used a novel mouse model with complete replacement of the mouse TNF gene by its human ortholog (human TNF [huTNF] knock-in [KI] mice) to determine resistance to Mycobacterium bovis BCG and M. tuberculosis infections and to investigate whether TNF inhibitors in clinical use reduce host immunity. Our results show that macrophages from huTNF KI mice responded to BCG and lipopolysaccharide similarly to wild-type macrophages by NF-κB activation and cytokine production. While TNF-deficient mice rapidly succumbed to mycobacterial infection, huTNF KI mice survived, controlling the bacterial burden and activating bactericidal mechanisms. Administration of TNF-neutralizing biologics disrupted the control of mycobacterial infection in huTNF KI mice, leading to an increased bacterial burden and hyperinflammation. Thus, our findings demonstrate that human TNF can functionally replace murine TNF in vivo, providing mycobacterial resistance that could be compromised by TNF neutralization. This new animal model will be helpful for the testing of specific biologics neutralizing human TNF.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Mycobacterium/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Animales , Western Blotting , Citocinas/biosíntesis , Citometría de Flujo , Técnicas de Sustitución del Gen/métodos , Humanos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/inmunología
17.
Eur J Immunol ; 43(10): 2683-95, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23780878

RESUMEN

Cerebral malaria is a severe complication of Plasmodium falciparum infection. Although T-cell activation and type II IFN-γ are required for Plasmodium berghei ANKA (PbA)-induced murine experimental cerebral malaria (ECM), the role of type I IFN-α/ß in ECM development remains unclear. Here, we address the role of the IFN-α/ß pathway in ECM devel-opment in response to hepatic or blood-stage PbA infection, using mice deficient for types I or II IFN receptors. While IFN-γR1⁻/⁻ mice were fully resistant, IFNAR1⁻/⁻ mice showed delayed and partial protection to ECM after PbA infection. ECM resistance in IFN-γR1⁻/⁻ mice correlated with unaltered cerebral microcirculation and absence of ischemia, while WT and IFNAR1⁻/⁻ mice developed distinct microvascular pathologies. ECM resistance appeared to be independent of parasitemia. Instead, key mediators of ECM were attenuated in the absence of IFNAR1, including PbA-induced brain sequestration of CXCR3⁺-activated CD8⁺ T cells. This was associated with reduced expression of Granzyme B, IFN-γ, IL-12Rß2, and T-cell-attracting chemokines CXCL9 and CXCL10 in IFNAR1⁻/⁻ mice, more so in the absence of IFN-γR1. Therefore, the type I IFN-α/ß receptor pathway contributes to brain T-cell responses and microvascular pathology, although it is not as essential as IFN-γ for the development of cerebral malaria upon hepatic or blood-stage PbA infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Cerebelo/inmunología , Interferón Tipo I/inmunología , Malaria Cerebral/inmunología , Plasmodium berghei/inmunología , Plasmodium falciparum/inmunología , Animales , Linfocitos T CD8-positivos/parasitología , Movimiento Celular/genética , Cerebelo/parasitología , Citotoxicidad Inmunológica/genética , Progresión de la Enfermedad , Humanos , Isquemia/genética , Malaria Cerebral/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación/genética , Modelos Animales , Receptores CXCR3/metabolismo , Receptores de Interferón/genética , Esporozoítos/inmunología
18.
J Immunol ; 188(4): 1905-14, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22238458

RESUMEN

A Th1 response is required for the development of Plasmodium berghei ANKA (PbA)-induced experimental cerebral malaria (ECM). The role of pro-Th1 IL-12 in malaria is complex and controversial. In this study, we addressed the role of IL-12Rß2 in ECM development. C57BL/6 mice deficient for IL-12Rß2, IL-12p40, or IL-12p35 were analyzed for ECM development after blood-stage PbA infection in terms of ischemia and blood flow by noninvasive magnetic resonance imaging and angiography, T cell recruitment, and gene expression. Without IL-12Rß2, no neurologic sign of ECM developed upon PbA infection. Although wild-type mice developed distinct brain microvascular pathology, ECM-resistant, IL-12Rß2-deficient mice showed unaltered cerebral microcirculation and the absence of ischemia after PbA infection. In contrast, mice deficient for IL-12p40 or IL-12p35 were sensitive to ECM development. The resistance of IL-12Rß2-deficient mice to ECM correlated with reduced recruitment of activated T cells and impaired overexpression of lymphotoxin-α, TNF-α, and IFN-γ in the brain after PbA infection. Therefore, IL-12Rß2 signaling is essential for ECM development but independent from IL-12p40 and IL-12p35. We document a novel link between IL-12Rß2 and lymphotoxin-α, TNF-α, and IFN-γ expression, key cytokines for ECM pathogenesis.


Asunto(s)
Subunidad beta 2 del Receptor de Interleucina-12/metabolismo , Malaria Cerebral/inmunología , Plasmodium berghei/inmunología , Células TH1/inmunología , Animales , Encéfalo/metabolismo , Encéfalo/microbiología , Encéfalo/patología , Linfocitos T CD8-positivos/inmunología , Interferón gamma/biosíntesis , Subunidad beta 2 del Receptor de Interleucina-12/deficiencia , Subunidad beta 2 del Receptor de Interleucina-12/genética , Subunidad p35 de la Interleucina-12/deficiencia , Subunidad p35 de la Interleucina-12/genética , Subunidad p35 de la Interleucina-12/inmunología , Subunidad p40 de la Interleucina-12/deficiencia , Subunidad p40 de la Interleucina-12/genética , Subunidad p40 de la Interleucina-12/inmunología , Activación de Linfocitos/inmunología , Linfotoxina-alfa/biosíntesis , Malaria Cerebral/parasitología , Malaria Cerebral/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/patogenicidad , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis
20.
Am J Pathol ; 180(1): 246-55, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22079430

RESUMEN

Cerebral malaria is a severe form of the disease that may result, in part, from an overt inflammatory response during infection by Plasmodium falciparum. The understanding of the pathogenesis of cerebral malaria may aid in the development of better therapeutic strategies for patients. The immune response in cerebral malaria involves elevation of circulating levels of cytokines and chemokines associated with leukocyte accumulation and breakdown of the blood-brain barrier in the central nervous system. Platelet-activating factor (PAF) is a mediator of inflammation shown to orchestrate inflammatory processes, including recruitment of leukocytes and increase of vascular permeability. Using mice lacking the PAF receptor (PAFR(-/-)), we investigated the relevance of this molecule for the outcome and the neuroinflammatory process triggered by P. berghei ANKA, an experimental model of cerebral malaria. In PAFR(-/-) mice, lethality was markedly delayed and brain inflammation was significantly reduced, as demonstrated by histology, accumulation, and activation of CD8(+) T cells, changes in vascular permeability and activation of caspase-3 on endothelial cells and leukocytes. Similarly, treatment with the PAFR antagonist UK-74,505 delayed lethality. Taken together, the results suggest that PAFR signaling is crucial for the development of experimental cerebral malaria. Mechanistically, PAFR activation is crucial for the cascade of events leading to changes in vascular permeability, accumulation, and activation of CD8(+) T cells and apoptosis of leukocytes and endothelial cells.


Asunto(s)
Malaria Cerebral/etiología , Glicoproteínas de Membrana Plaquetaria/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Química Encefálica , Caspasa 3/metabolismo , Quimiocinas/metabolismo , Citocinas/biosíntesis , Citocinas/metabolismo , Dihidropiridinas/farmacología , Imidazoles/farmacología , Leucocitos/fisiología , Activación de Linfocitos , Malaria Cerebral/prevención & control , Ratones , Ratones Endogámicos C57BL , Inhibidores de Agregación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Glicoproteínas de Membrana Plaquetaria/deficiencia , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA