Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
PLoS Pathog ; 10(7): e1004188, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24991816

RESUMEN

The purinergic P2X7 receptor (P2X7R) is a sensor of extracellular ATP, a damage-associated molecule that is released from necrotic cells and that induces pro-inflammatory cytokine production and cell death. To investigate whether the innate immune response to damage signals could contribute to the development of pulmonary necrotic lesions in severe forms of tuberculosis, disease progression was examined in C57BL/6 and P2X7R-/- mice that were intratracheally infected with highly virulent mycobacterial strains (Mycobacterium tuberculosis strain 1471 of the Beijing genotype family and Mycobacterium bovis strain MP287/03). The low-dose infection of C57BL/6 mice with bacteria of these strains caused the rapid development of extensive granulomatous pneumonia with necrotic areas, intense bacillus dissemination and anticipated animal death. In contrast, in P2X7R-/- mice, the lung pathology presented with moderate infiltrates of mononuclear leukocytes without visible signs of necrosis; the disease attenuation was accompanied by a delay in mortality. In vitro, the hypervirulent mycobacteria grew rapidly inside macrophages and induced death by a P2X7R-dependent mechanism that facilitated the release of bacilli. Furthermore, these bacteria were resistant to the protective mechanisms elicited in macrophages following extracellular ATP stimulation. Based on this study, we propose that the rapid intracellular growth of hypervirulent mycobacteria results in massive macrophage damage. The ATP released by damaged cells engages P2X7R and accelerates the necrotic death of infected macrophages and the release of bacilli. This vicious cycle exacerbates pneumonia and lung necrosis by promoting widespread cell destruction and bacillus dissemination. These findings suggest the use of drugs that have been designed to inhibit the P2X7R as a new therapeutic approach to treat the aggressive forms of tuberculosis.


Asunto(s)
Macrófagos , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Receptores Purinérgicos P2X7 , Tuberculosis Pulmonar , Adenosina Trifosfato/inmunología , Animales , Humanos , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Mycobacterium bovis/inmunología , Mycobacterium bovis/patogenicidad , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/inmunología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología
2.
Proc Natl Acad Sci U S A ; 110(35): E3321-30, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23942123

RESUMEN

NAIP5/NLRC4 (neuronal apoptosis inhibitory protein 5/nucleotide oligomerization domain-like receptor family, caspase activation recruitment domain domain-containing 4) inflammasome activation by cytosolic flagellin results in caspase-1-mediated processing and secretion of IL-1ß/IL-18 and pyroptosis, an inflammatory cell death pathway. Here, we found that although NLRC4, ASC, and caspase-1 are required for IL-1ß secretion in response to cytosolic flagellin, cell death, nevertheless, occurs in the absence of these molecules. Cytosolic flagellin-induced inflammasome-independent cell death is accompanied by IL-1α secretion and is temporally correlated with the restriction of Salmonella Typhimurium infection. Despite displaying some apoptotic features, this peculiar form of cell death do not require caspase activation but is regulated by a lysosomal pathway, in which cathepsin B and cathepsin D play redundant roles. Moreover, cathepsin B contributes to NAIP5/NLRC4 inflammasome-induced pyroptosis and IL-1α and IL-1ß production in response to cytosolic flagellin. Together, our data describe a pathway induced by cytosolic flagellin that induces a peculiar form of cell death and regulates inflammasome-mediated effector mechanisms of macrophages.


Asunto(s)
Citosol/metabolismo , Flagelina/metabolismo , Inflamasomas/metabolismo , Lisosomas/metabolismo , Macrófagos/inmunología , Animales , Apoptosis , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/microbiología , Salmonella typhimurium/patogenicidad , Receptor Toll-Like 5/genética
3.
J Immunol ; 190(7): 3629-38, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23460746

RESUMEN

Pathogens are detected by innate immune receptors that, upon activation, orchestrate an appropriate immune response. Recent studies revealed the intracellular signaling cascades involved in the TLR-initiated immune response to Brucella abortus infection. However, no report has elucidated the role of inflammasome receptors in Brucella recognition. Therefore, we decided to investigate the function of NLRC4, NLRP3, and AIM2 in sensing Brucella. In this study, we showed that NLRC4 is not required to induce caspase-1 activation and further secretion of IL-1ß by B. abortus in macrophages. In contrast, we determined that AIM2, which senses Brucella DNA, and NLRP3 are partially required for caspase-1 activation and IL-1ß secretion. Additionally, mitochondrial reactive oxygen species induced by Brucella were implicated in IL-1ß production. Furthermore, AIM2, NLRP3, ASC, and caspase-1 knockout mice were more susceptible to B. abortus infection than were wild-type animals, suggesting that multiple ASC-dependent inflammasomes contribute to host protection against infection. This protective effect is due to the inflammatory response caused by IL-1ß and IL-18 rather than pyroptosis, because we observed augmented bacterial burden in IL-1R and IL-18 knockout mice. Finally, we determined that bacterial type IV secretion system VirB and live, but not heat-killed, Brucella are required for full inflammasome activation in macrophages during infection. Taken together, our results indicate that Brucella is sensed by ASC inflammasomes that collectively orchestrate a robust caspase-1 activation and proinflammatory response.


Asunto(s)
Sistemas de Secreción Bacterianos , Brucella abortus/inmunología , Brucella abortus/metabolismo , Brucelosis/inmunología , Brucelosis/metabolismo , Caspasa 1/metabolismo , Proteínas del Citoesqueleto/metabolismo , Inflamasomas , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Adaptadoras de Señalización CARD , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/genética , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN , Activación Enzimática , Predisposición Genética a la Enfermedad , Granuloma/inmunología , Granuloma/metabolismo , Granuloma/microbiología , Inmunidad Innata , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Hígado/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo
4.
PLoS Pathog ; 8(5): e1002699, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615561

RESUMEN

MHC class Ia-restricted CD8(+) T cells are important mediators of the adaptive immune response against infections caused by intracellular microorganisms. Whereas antigen-specific effector CD8(+) T cells can clear infection caused by intracellular pathogens, in some circumstances, the immune response is suboptimal and the microorganisms survive, causing host death or chronic infection. Here, we explored the cellular and molecular mechanisms that could explain why CD8(+) T cell-mediated immunity during infection with the human protozoan parasite Trypanosoma cruzi is not optimal. For that purpose, we compared the CD8(+) T-cell mediated immune responses in mice infected with T. cruzi or vaccinated with a recombinant adenovirus expressing an immunodominant parasite antigen. Several functional and phenotypic characteristics of specific CD8(+) T cells overlapped. Among few exceptions was an accelerated expansion of the immune response in adenoviral vaccinated mice when compared to infected ones. Also, there was an upregulated expression of the apoptotic-signaling receptor CD95 on the surface of specific T cells from infected mice, which was not observed in the case of adenoviral-vaccinated mice. Most importantly, adenoviral vaccine provided at the time of infection significantly reduced the upregulation of CD95 expression and the proapoptotic phenotype of pathogen-specific CD8(+) cells expanded during infection. In parallel, infected adenovirus-vaccinated mice had a stronger CD8 T-cell mediated immune response and survived an otherwise lethal infection. We concluded that a suboptimal CD8(+) T-cell response is associated with an upregulation of CD95 expression and a proapoptotic phenotype. Both can be blocked by adenoviral vaccination.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neuraminidasa/inmunología , Vacunas Antiprotozoos/inmunología , Trypanosoma cruzi/inmunología , Receptor fas/biosíntesis , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Anticuerpos Antiprotozoarios/biosíntesis , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Apoptosis , Enfermedad de Chagas/inmunología , Enfermedad de Chagas/prevención & control , Interferón gamma/biosíntesis , Proteína 1 de la Membrana Asociada a los Lisosomas/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trypanosoma cruzi/patogenicidad , Vacunas Sintéticas/inmunología
5.
Methods ; 61(2): 110-6, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23454287

RESUMEN

Pyroptosis is a molecularly controlled form of cell death that exhibits some features of apoptosis as well of necrosis. Pyroptosis is induced by inflammasome-activated caspase-1 or caspase-11 (caspase-4 in humans), as a result of distinct pathogenic or damage stimuli. Although pyroptosis displays some morphological and biochemical features of apoptosis, it has an inflammatory outcome due to the loss of plasma membrane integrity and the consequent release of intracellular contents, reminiscent to necrosis. Here, we use cytosolic delivery of purified flagellin as an experimental tool to trigger pyroptosis and describe potential methods to study this form of cell death. Finally, we discuss the advantages and limitations of these methods.


Asunto(s)
Apoptosis/genética , Citosol/metabolismo , Flagelina/metabolismo , Inflamasomas/metabolismo , Macrófagos Peritoneales/metabolismo , Animales , Bacillus subtilis/química , Caspasa 1/deficiencia , Caspasa 1/genética , Citosol/ultraestructura , Activación Enzimática , Ácidos Grasos Monoinsaturados/química , Flagelina/farmacología , Humanos , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis/genética , Necrosis/patología , Transporte de Proteínas , Compuestos de Amonio Cuaternario/química , Transducción de Señal
6.
Mediators Inflamm ; 2014: 912965, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25104883

RESUMEN

Over the past 20 years, the immune effector mechanisms involved in the control of Trypanosoma cruzi, as well as the receptors participating in parasite recognition by cells of the innate immune system, have been largely described. However, the main questions on the physiopathology of Chagas disease remain unanswered: "Why does the host immune system fail to provide sterile immunity?" and "Why do only a proportion of infected individuals develop chronic pathology?" In this review, we describe the mechanisms proposed to explain the inability of the immune system to eradicate the parasite and the elements that allow the development of chronic heart disease. Moreover, we discuss the possibility that the inability of infected cardiomyocytes to sense intracellular T. cruzi contributes to parasite persistence in the heart and the development of chronic pathology.


Asunto(s)
Enfermedad de Chagas/inmunología , Animales , Enfermedad de Chagas/complicaciones , Cardiopatías/etiología , Cardiopatías/inmunología , Cardiopatías/parasitología , Humanos , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/parasitología , Trypanosoma cruzi/inmunología , Trypanosoma cruzi/patogenicidad
7.
Inflammation ; 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38329636

RESUMEN

Neutrophilic asthma is generally defined by poorly controlled symptoms and high levels of neutrophils in the lungs. Short-chain fatty acids (SCFAs) are proposed as nonpharmacological therapy for allergic asthma, but their impact on the neutrophilic asthma lacks evidence. SCFAs regulate immune cell responses and impact the inflammasome NLRP3, a potential pharmacological target for neutrophilic asthma. Here, we explored the capacity of SCFAs to mitigate murine-induced neutrophilic asthma and the contribution of NLRP3 to this asthma. The objective of this study is to analyze whether SCFAs can attenuate lung inflammation and tissue remodeling in murine neutrophilic asthma and NLRP3 contribution to this endotype. Wild-type (WT) C57BL6 mice orotracheally received 10 µg of HDM (house dust mite) in 80 µL of saline on days 0, 6-10. To explore SCFAs, each HDM group received 200 mM acetate, propionate, or butyrate. To explore NLRP3, Nlrp3 KO mice received the same protocol of HDM. On the 14th day, after euthanasia, bronchoalveolar lavage fluid (BALF) and lungs were collected to evaluate cellularity, inflammatory cytokines, and tissue remodeling. HDM group had increased BALF neutrophil influx, TNF-α, IFN-γ, IL-17A, collagen deposition, and mucus secretion compared to control. SCFAs distinctively attenuate lung inflammation. Only features of tissue remodeling were Nlrp3-dependent such as collagen deposition, mucus secretion, active TGF-ß cytokine, and IMs CD206+. SCFAs greatly decreased inflammatory cytokines and tissue remodeling. Only tissue remodeling was dependent on NLRP3. It reveals the potential of SCFAs to act as an additional therapy to mitigate neutrophilic asthma and the NLRP3 contribution to asthma.

8.
Proc Natl Acad Sci U S A ; 107(6): 2568-73, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20133793

RESUMEN

The peritoneal cavity (PerC) is a unique compartment within which a variety of immune cells reside, and from which macrophages (MØ) are commonly drawn for functional studies. Here we define two MØ subsets that coexist in PerC in adult mice. One, provisionally called the large peritoneal MØ (LPM), contains approximately 90% of the PerC MØ in unstimulated animals but disappears rapidly from PerC following lipopolysaccharide (LPS) or thioglycolate stimulation. These cells express high levels of the canonical MØ surface markers, CD11b and F4/80. The second subset, referred to as small peritoneal MØ (SPM), expresses substantially lower levels of CD11b and F4/80 but expresses high levels of MHC-II, which is not expressed on LPM. SPM, which predominates in PerC after LPS or thioglycolate stimulation, does not derive from LPM. Instead, it derives from blood monocytes that rapidly enter the PerC after stimulation and differentiate to mature SPM within 2 to 4 d. Both subsets show clear phagocytic activity and both produce nitric oxide (NO) in response to LPS stimulation in vivo. However, their responses to LPS show key differences: in vitro, LPS stimulates LPM, but not SPM, to produce NO; in vivo, LPS stimulates both subsets to produce NO, albeit with different response patterns. These findings extend current models of MØ heterogeneity and shed new light on PerC MØ diversity, development, and function. Thus, they introduce a new context for interpreting (and reinterpreting) data from ex vivo studies with PerC MØ.


Asunto(s)
Macrófagos Peritoneales/citología , Macrófagos Peritoneales/inmunología , Cavidad Peritoneal/citología , Animales , Antígenos de Diferenciación/metabolismo , Antígeno CD11b/metabolismo , Células Cultivadas , Escherichia coli/genética , Escherichia coli/inmunología , Escherichia coli/metabolismo , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Microscopía Confocal , Fagocitosis/inmunología , Tioglicolatos/farmacología
9.
J Biol Chem ; 285(42): 32087-95, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20702413

RESUMEN

Innate immune recognition of flagellin is shared by transmembrane TLR5 and cytosolic Nlrc4 (NOD-like receptor family CARD (caspase activation recruitment domain) domain containing 4)/Naip5 (neuronal apoptosis inhibitory protein 5). TLR5 activates inflammatory genes through MYD88 pathway, whereas Nlrc4 and Naip5 assemble multiprotein complexes called inflammasomes, culminating in caspase-1 activation, IL-1ß/IL-18 secretion, and pyroptosis. Although both TLR5 and Naip5/Nlrc4 pathways cooperate to clear infections, little is known about the relative anti-pathogen effector mechanisms operating through each of them. Here we show that the cytosolic flagellin (FLA-BSDot) was able to activate iNOS, an enzyme previously associated with TLR5 pathway. Using Nlrc4- or Naip5-deficient macrophages, we found that both receptors are involved in iNOS activation by FLA-BSDot. Moreover, distinct from extracellular flagellin (FLA-BS), iNOS activation by intracellular flagellin is completely abrogated in the absence of caspase-1. Interestingly, IL-1ß and IL-18 do not seem to be important for FLA-BSDot-mediated iNOS production. Together, our data defined an additional anti-pathogen effector mechanism operated through Naip5 and Nlrc4 inflammasomes and illustrated a novel signaling transduction pathway that activates iNOS.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Inflamación/inmunología , Complejos Multiproteicos/inmunología , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al Calcio/genética , Activación Enzimática , Femenino , Flagelina/inmunología , Flagelina/farmacología , Inmunidad Innata/inmunología , Interleucina-18/inmunología , Interleucina-1beta/inmunología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Proteína Inhibidora de la Apoptosis Neuronal/genética , Transducción de Señal/fisiología , Receptor Toll-Like 5/genética , Receptor Toll-Like 5/metabolismo
10.
Cell Mol Life Sci ; 67(10): 1643-51, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20229126

RESUMEN

Cells can die by distinct mechanisms with particular impacts on the immune response. In addition to apoptosis and necrosis, recent studies lead to characterization of a new pro-inflammatory form of cell death, pyroptosis. TLR and NLR, central innate immune sensors, can control infections by modulating host cell survival. In addition, TLRs can promote the induction of autophagy, thus promoting delivery of infecting pathogens to the lysosomes. On the other hand, activation of some NLR members, especially NLRC4 and NAIP5, leads to the infected cell death by pyroptosis, which is accompanied by secretion of the pro-inflammatory cytokines IL-1beta, IL-18, and IL-33. Data presented here illustrate how the compartmentalization of the innate immune sensors can influence the outcome of infections by controlling the fate of host cells.


Asunto(s)
Apoptosis/inmunología , Autofagia/inmunología , Infecciones/inmunología , Infecciones/patología , Inflamación/inmunología , Proteínas Adaptadoras de Señalización NOD/inmunología , Receptores Toll-Like/inmunología , Animales , Humanos
11.
Cell Death Dis ; 12(7): 692, 2021 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-34247195

RESUMEN

Chagas disease is a life-threatening disorder caused by the protozoan parasite Trypanosoma cruzi. Parasite-specific antibodies, CD8+ T cells, as well as IFN-γ and nitric oxide (NO) are key elements of the adaptive and innate immunity against the extracellular and intracellular forms of the parasite. Bim is a potent pro-apoptotic member of the Bcl-2 family implicated in different aspects of the immune regulation, such as negative selection of self-reactive thymocytes and elimination of antigen-specific T cells at the end of an immune response. Interestingly, the role of Bim during infections remains largely unidentified. To explore the role of Bim in Chagas disease, we infected WT, Bim+/-, Bim-/- mice with trypomastigotes forms of the Y strain of T. cruzi. Strikingly, our data revealed that Bim-/- mice exhibit a delay in the development of parasitemia followed by a deficiency in the control of parasite load in the bloodstream and a decreased survival compared to WT and Bim+/- mice. At the peak of parasitemia, peritoneal macrophages of Bim-/- mice exhibit decreased NO production, which correlated with a decrease in the pro-inflammatory Small Peritoneal Macrophage (SPM) subset. A similar reduction in NO secretion, as well as in the pro-inflammatory cytokines IFN-γ and IL-6, was also observed in Bim-/- splenocytes. Moreover, an impaired anti-T. cruzi CD8+ T-cell response was found in Bim-/- mice at this time point. Taken together, our results suggest that these alterations may contribute to the establishment of a delayed yet enlarged parasitic load observed at day 9 after infection of Bim-/- mice and place Bim as an important protein in the control of T. cruzi infections.


Asunto(s)
Proteína 11 Similar a Bcl2/deficiencia , Enfermedad de Chagas/parasitología , Trypanosoma cruzi/patogenicidad , Animales , Proteína 11 Similar a Bcl2/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/parasitología , Células Cultivadas , Enfermedad de Chagas/genética , Enfermedad de Chagas/inmunología , Enfermedad de Chagas/metabolismo , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Parásitos , Interferón gamma/metabolismo , Interleucina-6/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/parasitología , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Carga de Parásitos , Bazo/inmunología , Bazo/metabolismo , Bazo/parasitología , Factores de Tiempo , Trypanosoma cruzi/inmunología
12.
Cells ; 10(1)2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33440601

RESUMEN

This study evaluated the role of endogenous and exogenous annexin A1 (AnxA1) in the activation of the NLRP3 inflammasome in isolated peritoneal neutrophils. C57BL/6 wild-type (WT) and AnxA1 knockout mice (AnxA1-/-) received 0.3% carrageenan intraperitoneally and, after 3 h, the peritoneal exudate was collected. WT and AnxA1-/- neutrophils were then stimulated with lipopolysaccharide, followed by the NLRP3 agonists nigericin or ATP. To determine the exogenous effect of AnxA1, the neutrophils were pretreated with the AnxA1-derived peptide Ac2-26 followed by the NLRP3 agonists. Ac2-26 administration reduced NLRP3-derived IL-1ß production by WT neutrophils after nigericin and ATP stimulation. However, IL-1ß release was impaired in AnxA1-/- neutrophils stimulated by both agonists, and there was no further impairment in IL-1ß release with Ac2-26 treatment before stimulation. Despite this, ATP- and nigericin-stimulated AnxA1-/- neutrophils had increased levels of cleaved caspase-1. The lipidomics of supernatants from nigericin-stimulated WT and AnxA1-/- neutrophils showed potential lipid biomarkers of cell stress and activation, including specific sphingolipids and glycerophospholipids. AnxA1 peptidomimetic treatment also increased the concentration of phosphatidylserines and oxidized phosphocholines, which are lipid biomarkers related to the inflammatory resolution pathway. Together, our results indicate that exogenous AnxA1 negatively regulates NLRP3-derived IL-1ß production by neutrophils, while endogenous AnxA1 is required for the activation of the NLRP3 machinery.


Asunto(s)
Anexina A1/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neutrófilos/metabolismo , Animales , Inflamasomas/ultraestructura , Interleucina-1beta/metabolismo , Lípidos/química , Masculino , Ratones Endogámicos C57BL , Activación Neutrófila , Neutrófilos/ultraestructura
13.
Front Cell Infect Microbiol ; 11: 676183, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34123875

RESUMEN

Deficiency in memory formation and increased immunosenescence are pivotal features of Trypanosoma cruzi infection proposed to play a role in parasite persistence and disease development. The vaccination protocol that consists in a prime with plasmid DNA followed by the boost with a deficient recombinant human adenovirus type 5, both carrying the ASP2 gene of T. cruzi, is a powerful strategy to elicit effector memory CD8+ T-cells against this parasite. In virus infections, the inhibition of mTOR, a kinase involved in several biological processes, improves the response of memory CD8+ T-cells. Therefore, our aim was to assess the role of rapamycin, the pharmacological inhibitor of mTOR, in CD8+ T response against T. cruzi induced by heterologous prime-boost vaccine. For this purpose, C57BL/6 or A/Sn mice were immunized and daily treated with rapamycin for 34 days. CD8+ T-cells response was evaluated by immunophenotyping, intracellular staining, ELISpot assay and in vivo cytotoxicity. In comparison with vehicle-injection, rapamycin administration during immunization enhanced the frequency of ASP2-specific CD8+ T-cells and the percentage of the polyfunctional population, which degranulated (CD107a+) and secreted both interferon gamma (IFNγ) and tumor necrosis factor (TNF). The beneficial effects were long-lasting and could be detected 95 days after priming. Moreover, the effects were detected in mice immunized with ten-fold lower doses of plasmid/adenovirus. Additionally, the highly susceptible to T. cruzi infection A/Sn mice, when immunized with low vaccine doses, treated with rapamycin, and challenged with trypomastigote forms of the Y strain showed a survival rate of 100%, compared with 42% in vehicle-injected group. Trying to shed light on the biological mechanisms involved in these beneficial effects on CD8+ T-cells by mTOR inhibition after immunization, we showed that in vivo proliferation was higher after rapamycin treatment compared with vehicle-injected group. Taken together, our data provide a new approach to vaccine development against intracellular parasites, placing the mTOR inhibitor rapamycin as an adjuvant to improve effective CD8+ T-cell response.


Asunto(s)
Vacunas Antiprotozoos , Trypanosoma cruzi , Animales , Linfocitos T CD8-positivos , Ratones , Ratones Endogámicos C57BL , Sirolimus/farmacología , Vacunación
14.
Front Immunol ; 11: 536, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32328060

RESUMEN

Efficient induction of effector and long-term protective antigen-specific CD8+ T memory response by vaccination is essential to eliminate malignant and pathogen-infected cells. Intracellular infectious bacteria, including Listeria monocytogenes, have been considered potent vectors to carry multiple therapeutic proteins and generate antigen-specific CD8+ T cell responses. Although the role of molecules involved in inflammatory cell death pathways, such as necroptosis (RIPK3-mediated) and pyroptosis (Caspase-1/11-mediated), as effectors of immune response against intracellular bacteria are relatively well understood, their contribution to the adjuvant effect of recombinant bacterial vectors in the context of antigen-specific CD8+ T cell response remained obscure. Therefore, we evaluated the impact of RIPK3 and Caspase-1/11 (Casp-1/11) individual and combined deficiencies on the modulation of antigen-specific CD8+ T cell response during vaccination of mice with ovalbumin-expressing L. monocytogenes (LM-OVA). We observed that Casp-1/11 but not RIPK3 deficiency negatively impacts the capacity of mice to clear LM-OVA. Importantly, both RIPK3 and Casp-1/11 are necessary for optimal LM-OVA-mediated antigen-specific CD8+ T cell response, as measured by in vivo antigen-specific CD8+ T cell proliferation, target cell elimination, and cytokine production. Furthermore, Casp-1/11 and Casp-1/11/RIPK3 combined deficiencies restrict the early initiation of antigen-specific CD8+ T cell memory response. Taken together, our findings demonstrate that RIPK3 and Casp-1/11 influence the quality of CD8+ T cell responses induced by recombinant L. monocytogenes vectors.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Caspasa 1/inmunología , Caspasas Iniciadoras/inmunología , Vectores Genéticos/inmunología , Listeria monocytogenes/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Animales , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL
15.
Sci Adv ; 6(10): eaax6346, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32181339

RESUMEN

Placental malaria (PM) is associated with severe inflammation leading to abortion, preterm delivery, and intrauterine growth restriction. Innate immunity responses play critical roles, but the mechanisms underlying placental immunopathology are still unclear. Here, we investigated the role of inflammasome activation in PM by scrutinizing human placenta samples from an endemic area and ablating inflammasome components in a PM mouse model. The reduction in birth weight in babies from infected mothers is paralleled by increased placental expression of AIM2 and NLRP3 inflammasomes. Using genetic dissection, we reveal that inflammasome activation pathways are involved in the production and detrimental action of interleukin-1ß (IL-1ß) in the infected placenta. The IL-1R pharmacological antagonist Anakinra improved pregnancy outcomes by restoring fetal growth and reducing resorption in an experimental model. These findings unveil that IL-1ß-mediated signaling is a determinant of PM pathogenesis, suggesting that IL-1R antagonists can improve clinical outcomes of malaria infection in pregnancy.


Asunto(s)
Inflamasomas/efectos de los fármacos , Interleucina-1beta/inmunología , Malaria Falciparum/inmunología , Malaria/inmunología , Plasmodium falciparum/patogenicidad , Complicaciones Parasitarias del Embarazo/inmunología , Transducción de Señal/efectos de los fármacos , Animales , Caspasa 1/genética , Caspasa 1/inmunología , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Femenino , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Factores Inmunológicos/farmacología , Inflamasomas/genética , Inflamasomas/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/genética , Malaria/tratamiento farmacológico , Malaria/genética , Malaria/parasitología , Malaria Falciparum/genética , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Plasmodium berghei/inmunología , Plasmodium berghei/patogenicidad , Plasmodium falciparum/inmunología , Embarazo , Complicaciones Parasitarias del Embarazo/genética , Complicaciones Parasitarias del Embarazo/parasitología , Complicaciones Parasitarias del Embarazo/prevención & control , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología , Transducción de Señal/inmunología , Células THP-1 , Trofoblastos/efectos de los fármacos , Trofoblastos/inmunología , Trofoblastos/parasitología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
16.
Nat Rev Immunol ; 20(10): 594-602, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32913283

RESUMEN

The COVID-19 pandemic is shining a spotlight on the field of immunology like never before. To appreciate the diverse ways in which immunologists have contributed, Nature Reviews Immunology invited the president of the International Union of Immunological Societies and the presidents of 15 other national immunology societies to discuss how they and their members responded following the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).


Asunto(s)
COVID-19/epidemiología , Infecciones por Coronavirus/epidemiología , Cooperación Internacional , Pandemias , Neumonía Viral/epidemiología , Síndrome Respiratorio Agudo Grave/epidemiología , Sociedades Científicas/organización & administración , Antivirales/síntesis química , Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , Betacoronavirus/inmunología , Betacoronavirus/patogenicidad , COVID-19/inmunología , COVID-19/prevención & control , COVID-19/terapia , Vacunas contra la COVID-19 , Relaciones Comunidad-Institución , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/terapia , Salud Global/tendencias , Humanos , Educación del Paciente como Asunto/organización & administración , Equipo de Protección Personal/provisión & distribución , Neumonía Viral/inmunología , Neumonía Viral/terapia , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/inmunología , Síndrome Respiratorio Agudo Grave/terapia , Vacunas Virales/biosíntesis
17.
J Leukoc Biol ; 106(3): 531-540, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31071239

RESUMEN

Autophagy and inflammasome activation are cell-autonomous and cross-regulated processes involved in host resistance against infections. Our group previously described that NLRP3 inflammasome is required for the control of Trypanosoma cruzi, the causative agent of Chagas disease. However, the involvement of autophagy in this process was unclear. Here, we demonstrated that T. cruzi was able to induce an increase in LC3-II expression as well as autophagosome and autolysosome formation in peritoneal macrophages (PMs) from C57BL/6 wild-type mice. Moreover, the pharmacologic inhibition of autophagic machinery impaired the ability of PMs to control T. cruzi replication. Importantly, NLRP3 was required for the induction of a regular autophagic flux in response to T. cruzi, an effect mediated by its participation in the autolysosomes formation. Together, these results indicate autophagy as an effector mechanism mediated by NLRP3 to control T. cruzi infection.


Asunto(s)
Autofagia , Enfermedad de Chagas/metabolismo , Enfermedad de Chagas/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ácidos/metabolismo , Animales , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Lisosomas/metabolismo , Lisosomas/ultraestructura , Macrófagos/metabolismo , Macrófagos/parasitología , Ratones Endogámicos C57BL , Trypanosoma cruzi
18.
J Leukoc Biol ; 106(1): 201-207, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30997938

RESUMEN

Trypanossoma cruzi (T. cruzi), the causative protozoan of Chagas disease (CD) invades many cell types, including central nervous system (CNS) cells triggering local lesions and neurological impact. Previous work from our group described NLRP3 inflammasomes as central effectors for the parasite control by macrophages. Recent evidences demonstrate that NLRP3 can be activated in CNS cells with controversial consequences to the control of infections and inflammatory pathologies. However, the relative contribution of NLRP3 in different cell types remains to be elucidated. In this article, we described an effector response mediated by NLRP3 that works on microglia but not on astrocytes to control T. cruzi infection. Despite T. cruzi ability to invade astrocytes and microglia, astrocytes were clearly more permissive to parasite replication. Moreover, the absence of NLRP3 renders microglia but not astrocytes more permissive to T. cruzi replication. In fact, microglia but not astrocytes were able to secrete NLRP3-dependent IL-1ß and NO in response to T. cruzi. Importantly, the pharmacological inhibition of iNOS with aminoguanidine resulted in a significant increase in the numbers of amastigotes found in microglia from wild-type but not from NLRP3-/- mice, indicating the importance of NLRP3-mediated NO secretion to the infection control by these cells. Taken together, our findings revealed that T. cruzi differentially activates NLRP3 inflammasomes in astrocytes and microglia and established a role for these platforms in the control of a protozoan infection by glial cells from CNS.


Asunto(s)
Astrocitos/parasitología , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Óxido Nítrico/biosíntesis , Trypanosoma cruzi/fisiología , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Microglía/parasitología
19.
Front Immunol ; 9: 2379, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30459758

RESUMEN

Pattern Recognition Receptors (PRRs) are proteins capable of recognizing molecules frequently found in pathogens (the so-called Pathogen-Associated Molecular Patterns-PAMPs), or molecules released by damaged cells (the Damage-Associated Molecular Patterns-DAMPs). They emerged phylogenetically prior to the appearance of the adaptive immunity and, therefore, are considered part of the innate immune system. Signals derived from the engagement of PRRs on the immune cells activate microbicidal and pro-inflammatory responses required to eliminate or, at least, to contain infectious agents. Molecularly controlled forms of cell death are also part of a very ancestral mechanism involved in key aspects of the physiology of multicellular organism, including the elimination of unwanted, damaged or infected cells. Interestingly, each form of cell death has its particular effect on inflammation and on the development of innate and adaptive immune responses. In this review article, we discuss some aspects of the molecular interplay between the cell death machinery and signals initiated by the activation of PRRs by PAMPs and DAMPs.


Asunto(s)
Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Apoptosis , Muerte Celular , Interacciones Huésped-Patógeno , Humanos , Inflamación/etiología , Inflamación/metabolismo , Piroptosis
20.
Sci Rep ; 7: 41686, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28150715

RESUMEN

Nitric oxide synthase 2, inducible (Nos2) expression is necessary for the microbicidal activity of macrophages. However, NOS2 over-activation causes multiple inflammatory disorders, suggesting a tight gene regulation is necessary. Using cytosolic flagellin as a model for inflammasome-dependent NOS2 activation, we discovered a surprising new role for NLRC4/caspase-1 axis in regulating chromatin accessibility of the Nos2 promoter. We found that activation of two independent mechanisms is necessary for NOS2 expression by cytosolic flagellin: caspase-1 and NF-κB activation. NF-κB activation was necessary, but not sufficient, for NOS2 expression. Conversely, caspase-1 was necessary for NOS2 expression, but dispensable for NF-κB activation, indicating that this protease acts downstream NF-κB activation. We demonstrated that epigenetic regulation of Nos2 by caspase-1 involves cleavage of the chromatin regulator PARP1 (also known as ARTD1) and chromatin accessibility of the NF-κB binding sites located at the Nos2 promoter. Remarkably, caspase-1-mediated Nos2 transcription and NO production contribute to the resistance of macrophages to Salmonella typhimurium infection. Our results uncover the molecular mechanism behind the constricted regulation of Nos2 expression and open new therapeutic opportunities based on epigenetic activities of caspase-1 against infectious and inflammatory diseases.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Epigénesis Genética , Inflamasomas/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Caspasa 1/metabolismo , Citosol/metabolismo , Flagelina/metabolismo , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Modelos Biológicos , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Motivos de Nucleótidos , Regiones Promotoras Genéticas , Unión Proteica , Proteolisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA