Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Nat Commun ; 14(1): 1049, 2023 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-36828815

RESUMEN

Intracellular parasites from the Leishmania genus cause Leishmaniasis, a disease affecting millions of people worldwide. NLRP3 inflammasome is key for disease outcome, but the molecular mechanisms upstream of the inflammasome activation are still unclear. Here, we demonstrate that despite the absence of pyroptosis, Gasdermin-D (GSDMD) is active at the early stages of Leishmania infection in macrophages, allowing transient cell permeabilization, potassium efflux, and NLRP3 inflammasome activation. Further, GSDMD is processed into a non-canonical 25 kDa fragment. Gsdmd-/- macrophages and mice exhibit less NLRP3 inflammasome activation and are highly susceptible to infection by several Leishmania species, confirming the role of GSDMD for inflammasome-mediated host resistance. Active NLRP3 inflammasome and GSDMD are present in skin biopsies of patients, demonstrating activation of this pathway in human leishmaniasis. Altogether, our findings reveal that Leishmania subverts the normal functions of GSDMD, an important molecule to promote inflammasome activation and immunity in Leishmaniasis.


Asunto(s)
Leishmania , Leishmaniasis , Humanos , Ratones , Animales , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Gasderminas , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leishmania/metabolismo , Piroptosis/fisiología
2.
J Infect Dis ; 227(12): 1364-1375, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-36763010

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection triggers activation of the NLRP3 inflammasome, which promotes inflammation and aggravates severe COVID-19. Here, we report that SARS-CoV-2 induces upregulation and activation of human caspase-4/CASP4 (mouse caspase-11/CASP11), and this process contributes to NLRP3 activation. In vivo infections performed in transgenic hACE2 humanized mice, deficient or sufficient for Casp11, indicate that hACE2 Casp11-/- mice were protected from disease development, with the increased pulmonary parenchymal area, reduced clinical score of the disease, and reduced mortality. Assessing human samples from fatal cases of COVID-19, we found that CASP4 was expressed in patient lungs and correlated with the expression of inflammasome components and inflammatory mediators, including CASP1, IL1B, IL18, and IL6. Collectively, our data establish that CASP4/11 promotes NLRP3 activation and disease pathology, revealing a possible target for therapeutic interventions for COVID-19.


Asunto(s)
COVID-19 , Inflamasomas , Ratones , Animales , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Macrófagos/metabolismo , COVID-19/metabolismo , SARS-CoV-2/metabolismo , Ratones Transgénicos
3.
J Pathol ; 259(3): 291-303, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36441400

RESUMEN

A low-grade and persistent inflammation, which is the hallmark of obesity, requires the participation of NLRP3 and cell death. During Mycobacterium tuberculosis infection, NLRP3 signaling is important for bacterial killing by macrophages in vitro but was shown to be dispensable for host protection in vivo. We hypothesized that during obesity-tuberculosis (TB) comorbidity, NLRP3 signaling might play a detrimental role by inducing excessive inflammation. We employed a model of high-fat-diet-induced obesity, followed by M. tuberculosis infection in C57BL/6 mice. Obese mice presented increased susceptibility to infection and pulmonary immunopathology compared to lean mice. Using treatment with NLRP3 antagonist and Nlrp3-/- mice, we showed that NLRP3 signaling promoted cell death, with no effect in bacterial loads. The levels of palmitate were higher in the lungs of obese infected mice compared to lean counterparts, and we observed that this lipid increased M. tuberculosis-induced macrophage death in vitro, which was dependent on NLRP3 and caspase-1. At the chronic phase, although lungs of obese Nlrp3-/- mice showed an indication of granuloma formation compared to obese wild-type mice, there was no difference in the bacterial load. Our findings indicate that NLRP3 may be a potential target for host-directed therapy to reduce initial and severe inflammation-mediated disease and to treat comorbidity-associated TB. © 2022 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Ratones , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Palmitatos/metabolismo , Ratones Endogámicos C57BL , Tuberculosis/patología , Pulmón/patología , Inflamación/patología , Obesidad/metabolismo , Muerte Celular , Comorbilidad
4.
Eur J Immunol ; 53(2): e2249985, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36427489

RESUMEN

Flagellin-induced NAIP/NLRC4 inflammasome activation and pyroptosis are critical events restricting Legionella pneumophila infection. However, the cellular and molecular dynamics of the in vivo responses against this bacterium are still unclear. We have found temporal coordination of two independent innate immunity pathways in controlling Legionella infection, the inflammasome activation and the CCR2-mediated Mo-DC recruitment. Inflammasome activation was an important player at the early stage of infection by lowering the numbers of bacteria for an efficient bacterial clearance conferred by the Mo-DC at the late stage of the infection. Mo-DC emergence highly depended on CCR2-signaling and dispensed inflammasome activation and pyroptosis. Also, Mo-DC compartment did not rely on the inflammasome machinery to deliver proper immune responses and was the most abundant cytokine-producing among the monocyte-derived cells in the infected lung. Importantly, when the CCR2- and NLRC4-dependent axes of response were simultaneously ablated, we observed an aggravated bacterial burden in the lung of infected mice. Taken together, we showed that inflammasome activation and CCR2-mediated immune response interplay in distinct pathways to restrict pulmonary bacterial infection. These findings extend our understanding of the in vivo integration and cooperation of different innate immunity arms in controlling infectious agents.


Asunto(s)
Células Dendríticas , Inflamasomas , Legionella pneumophila , Enfermedad de los Legionarios , Monocitos , Animales , Ratones , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Células Dendríticas/metabolismo , Inflamasomas/genética , Inflamasomas/metabolismo , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/genética , Enfermedad de los Legionarios/inmunología , Macrófagos , Ratones Noqueados , Monocitos/metabolismo , Receptores CCR2/metabolismo
5.
Elife ; 112022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35666101

RESUMEN

COVID-19 is a disease of dysfunctional immune responses, but the mechanisms triggering immunopathogenesis are not established. The functional plasticity of macrophages allows this cell type to promote pathogen elimination and inflammation or suppress inflammation and promote tissue remodeling and injury repair. During an infection, the clearance of dead and dying cells, a process named efferocytosis, can modulate the interplay between these contrasting functions. Here, we show that engulfment of SARS-CoV-2-infected apoptotic cells exacerbates inflammatory cytokine production, inhibits the expression of efferocytic receptors, and impairs continual efferocytosis by macrophages. We also provide evidence supporting that lung monocytes and macrophages from severe COVID-19 patients have compromised efferocytic capacity. Our findings reveal that dysfunctional efferocytosis of SARS-CoV-2-infected cell corpses suppresses macrophage anti-inflammation and efficient tissue repair programs and provides mechanistic insights for the excessive production of pro-inflammatory cytokines and accumulation of tissue damage associated with COVID-19 immunopathogenesis.


Asunto(s)
COVID-19 , SARS-CoV-2 , Antiinflamatorios/farmacología , Apoptosis , Humanos , Macrófagos/metabolismo , Fagocitosis
6.
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
7.
Elife ; 102021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34151776

RESUMEN

Type I interferons (IFNs) are essential for anti-viral immunity, but often impair protective immune responses during bacterial infections. An important question is how type I IFNs are strongly induced during viral infections, and yet are appropriately restrained during bacterial infections. The Super susceptibility to tuberculosis 1 (Sst1) locus in mice confers resistance to diverse bacterial infections. Here we provide evidence that Sp140 is a gene encoded within the Sst1 locus that represses type I IFN transcription during bacterial infections. We generated Sp140-/- mice and found that they are susceptible to infection by Legionella pneumophila and Mycobacterium tuberculosis. Susceptibility of Sp140-/- mice to bacterial infection was rescued by crosses to mice lacking the type I IFN receptor (Ifnar-/-). Our results implicate Sp140 as an important negative regulator of type I IFNs that is essential for resistance to bacterial infections.


Asunto(s)
Infecciones Bacterianas/inmunología , Interferón Tipo I/metabolismo , Factores de Transcripción/metabolismo , Alelos , Animales , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Interferón Tipo I/genética , Macrófagos/fisiología , Masculino , Ratones , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Mycobacterium tuberculosis , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Organismos Libres de Patógenos Específicos , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/farmacología
8.
J Immunol ; 206(10): 2441-2452, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33941658

RESUMEN

Intestinal barrier is essential for dietary products and microbiota compartmentalization and therefore gut homeostasis. When this barrier is broken, cecal content overflows into the peritoneal cavity, leading to local and systemic robust inflammatory response, characterizing peritonitis and sepsis. It has been shown that IL-1ß contributes with inflammatory storm during peritonitis and sepsis and its inhibition has beneficial effects to the host. Therefore, we investigated the mechanisms underlying IL-1ß secretion using a widely adopted murine model of experimental peritonitis. The combined injection of sterile cecal content (SCC) and the gut commensal bacteria Bacteroides fragilis leads to IL-1ß-dependent peritonitis, which was mitigated in mice deficient in NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome components. Typically acting as a damage signal, SCC, but not B. fragilis, activates canonical pathway of NLRP3 promoting IL-1ß secretion in vitro and in vivo. Strikingly, absence of fiber in the SCC drastically reduces IL-1ß production, whereas high-fiber SCC conversely increases this response in an NLRP3-dependent manner. In addition, NLRP3 was also required for IL-1ß production induced by purified dietary fiber in primed macrophages. Extending to the in vivo context, IL-1ß-dependent peritonitis was worsened in mice injected with B. fragilis and high-fiber SCC, whereas zero-fiber SCC ameliorates the pathology. Corroborating with the proinflammatory role of dietary fiber, IL-1R-deficient mice were protected from peritonitis induced by B. fragilis and particulate bran. Overall, our study highlights a function, previously unknown, for dietary fibers in fueling peritonitis through NLRP3 activation and IL-1ß secretion outside the gut.


Asunto(s)
Infecciones por Bacteroides/inmunología , Bacteroides fragilis/inmunología , Fibras de la Dieta/efectos adversos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Peritonitis/inmunología , Animales , Infecciones por Bacteroides/microbiología , Dieta , Fibras de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Peritonitis/microbiología , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
9.
Life Sci ; 276: 119376, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33781826

RESUMEN

The severe forms and worsened outcomes of COVID-19 (coronavirus disease 19) are closely associated with hypertension and cardiovascular disease. Endothelial cells express Angiotensin-Converting Enzyme 2 (ACE2), which is the entrance door for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The hallmarks of severe illness caused by SARS-CoV-2 infection are increased levels of IL-6, C-reactive protein, D-dimer, ferritin, neutrophilia and lymphopenia, pulmonary intravascular coagulopathy and microthrombi of alveolar capillaries. The endothelial glycocalyx, a proteoglycan- and glycoprotein-rich layer covering the luminal side of endothelial cells, contributes to vascular homeostasis. It regulates vascular tonus and permeability, prevents thrombosis, and modulates leukocyte adhesion and inflammatory response. We hypothesized that cytokine production and reactive oxygen species (ROS) generation associated with COVID-19 leads to glycocalyx degradation. A cohort of 20 hospitalized patients with a confirmed COVID-19 diagnosis and healthy subjects were enrolled in this study. Mechanisms associated with glycocalyx degradation in COVID-19 were investigated. Increased plasma concentrations of IL-6 and IL1-ß, as well as increased lipid peroxidation and glycocalyx components were detected in plasma from COVID-19 patients compared to plasma from healthy subjects. Plasma from COVID-19 patients induced glycocalyx shedding in cultured human umbilical vein endothelial cells (HUVECs) and disrupted redox balance. Treatment of HUVECs with low molecular weight heparin inhibited the glycocalyx perturbation. In conclusion, plasma from COVID-19 patients promotes glycocalyx shedding and redox imbalance in endothelial cells, and heparin treatment potentially inhibits glycocalyx disruption.


Asunto(s)
COVID-19/sangre , COVID-19/patología , Glicocálix/patología , Heparina/farmacología , Anciano , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/virología , COVID-19/metabolismo , Prueba de COVID-19 , Estudios de Casos y Controles , Adhesión Celular/fisiología , Endotelio Vascular/metabolismo , Femenino , Glicocálix/metabolismo , Glicocálix/virología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-1beta/sangre , Interleucina-6/sangre , Masculino , Persona de Mediana Edad , Oxidación-Reducción , SARS-CoV-2 , Trombosis/metabolismo
10.
J Exp Med ; 217(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-32926098

RESUMEN

Severe COVID-19 patients develop acute respiratory distress syndrome that may progress to cytokine storm syndrome, organ dysfunction, and death. Considering that neutrophil extracellular traps (NETs) have been described as important mediators of tissue damage in inflammatory diseases, we investigated whether NETs would be involved in COVID-19 pathophysiology. A cohort of 32 hospitalized patients with a confirmed diagnosis of COVID-19 and healthy controls were enrolled. The concentration of NETs was augmented in plasma, tracheal aspirate, and lung autopsies tissues from COVID-19 patients, and their neutrophils released higher levels of NETs. Notably, we found that viable SARS-CoV-2 can directly induce the release of NETs by healthy neutrophils. Mechanistically, NETs triggered by SARS-CoV-2 depend on angiotensin-converting enzyme 2, serine protease, virus replication, and PAD-4. Finally, NETs released by SARS-CoV-2-activated neutrophils promote lung epithelial cell death in vitro. These results unravel a possible detrimental role of NETs in the pathophysiology of COVID-19. Therefore, the inhibition of NETs represents a potential therapeutic target for COVID-19.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Trampas Extracelulares/fisiología , Neumonía Viral/inmunología , Neumonía Viral/virología , Células A549 , Adulto , Enzima Convertidora de Angiotensina 2 , COVID-19 , Muerte Celular , Infecciones por Coronavirus/sangre , Infecciones por Coronavirus/patología , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Células HeLa , Humanos , Masculino , Activación Neutrófila , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/sangre , Neumonía Viral/patología , SARS-CoV-2 , Serina Proteasas/metabolismo , Succión , Tráquea/inmunología
11.
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
12.
Immunology ; 160(1): 78-89, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32107769

RESUMEN

Annexins are well-known Ca2+ phospholipid-binding proteins, which have a wide variety of cellular functions. The role of annexin A1 (AnxA1) in the innate immune system has focused mainly on the anti-inflammatory and proresolving properties through its binding to the formyl-peptide receptor 2 (FPR2)/ALX receptor. However, studies suggesting an intracellular role of AnxA1 are emerging. In this study, we aimed to understand the role of AnxA1 for interleukin (IL)-1ß release in response to activators of the nucleotide-binding domain leucine-rich repeat (NLR) and pyrin domain containing receptor 3 (NLRP3) inflammasome. Using AnxA1 knockout mice, we observed that AnxA1 is required for IL-1ß release in vivo and in vitro. These effects were due to reduction of transcriptional levels of IL-1ß, NLRP3 and caspase-1, a step called NLRP3 priming. Moreover, we demonstrate that AnxA1 co-localize and directly bind to NLRP3, suggesting the role of AnxA1 in inflammasome activation is independent of its anti-inflammatory role via FPR2. Therefore, AnxA1 regulates NLRP3 inflammasome priming and activation in a FPR2-independent manner.


Asunto(s)
Anexina A1/metabolismo , Inflamasomas/inmunología , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Administración Intranasal , Animales , Cartílago Articular , Caspasa 1/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Gota/inducido químicamente , Gota/inmunología , Gota/patología , Humanos , Inflamasomas/metabolismo , Inyecciones Intraarticulares , Pulmón/inmunología , Pulmón/patología , Macrófagos , Masculino , Ratones , Ratones Noqueados , Cultivo Primario de Células , Unión Proteica/inmunología , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/toxicidad , Silicosis/inmunología , Silicosis/patología , Transcripción Genética/inmunología , Ácido Úrico/administración & dosificación , Ácido Úrico/toxicidad
13.
Nat Commun ; 10(1): 5273, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31754185

RESUMEN

Leishmania RNA virus (LRV) is an important virulence factor associated with the development of mucocutaneous Leishmaniasis, a severe form of the disease. LRV-mediated disease exacerbation relies on TLR3 activation, but downstream mechanisms remain largely unexplored. Here, we combine human and mouse data to demonstrate that LRV triggers TLR3 and TRIF to induce type I IFN production, which induces autophagy. This process results in ATG5-mediated degradation of NLRP3 and ASC, thereby limiting NLRP3 inflammasome activation in macrophages. Consistent with the known restricting role of NLRP3 for Leishmania replication, the signaling pathway triggered by LRV results in increased parasite survival and disease progression. In support of this data, we find that lesions in patients infected with LRV+ Leishmania are associated with reduced inflammasome activation and the development of mucocutaneous disease. Our findings reveal the mechanisms triggered by LRV that contribute to the development of the debilitating mucocutaneous form of Leishmaniasis.


Asunto(s)
Inmunidad Innata/inmunología , Inflamasomas/inmunología , Leishmania/inmunología , Leishmaniasis Mucocutánea/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Virus ARN/inmunología , Receptor Toll-Like 3/inmunología , Animales , Autofagia/inmunología , Humanos , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Leishmania/fisiología , Leishmania/virología , Leishmaniasis Mucocutánea/parasitología , Leishmaniasis Mucocutánea/virología , Macrófagos/inmunología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Virus ARN/fisiología , Transducción de Señal/inmunología , Receptor Toll-Like 3/metabolismo
14.
PLoS Pathog ; 15(9): e1007934, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31479495

RESUMEN

Mayaro virus (MAYV) is an arbovirus that circulates in Latin America and is emerging as a potential threat to public health. Infected individuals develop Mayaro fever, a severe inflammatory disease characterized by high fever, rash, arthralgia, myalgia and headache. The disease is often associated with a prolonged arthralgia mediated by a chronic inflammation that can last months. Although the immune response against other arboviruses, such as chikungunya virus (CHIKV), dengue virus (DENV) and Zika virus (ZIKV), has been extensively studied, little is known about the pathogenesis of MAYV infection. In this study, we established models of MAYV infection in macrophages and in mice and found that MAYV can replicate in bone marrow-derived macrophages and robustly induce expression of inflammasome proteins, such as NLRP3, ASC, AIM2, and Caspase-1 (CASP1). Infection performed in macrophages derived from Nlrp3-/-, Aim2-/-, Asc-/-and Casp1/11-/-mice indicate that the NLRP3, but not AIM2 inflammasome is essential for production of inflammatory cytokines, such as IL-1ß. We also determined that MAYV triggers NLRP3 inflammasome activation by inducing reactive oxygen species (ROS) and potassium efflux. In vivo infections performed in inflammasome-deficient mice indicate that NLRP3 is involved with footpad swelling, inflammation and pain, establishing a role of the NLRP3 inflammasome in the MAYV pathogenesis. Accordingly, we detected higher levels of caspase1-p20, IL-1ß and IL-18 in the serum of MAYV-infected patients as compared to healthy individuals, supporting the participation of the NLRP3-inflammasome during MAYV infection in humans.


Asunto(s)
Infecciones por Alphavirus/inmunología , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Adulto , Anciano , Infecciones por Alphavirus/metabolismo , Animales , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Virus Chikungunya/metabolismo , Virus del Dengue/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamasomas/inmunología , Inflamación/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Especies Reactivas de Oxígeno/metabolismo , Togaviridae/patogenicidad , Virus Zika/metabolismo
15.
J Leukoc Biol ; 106(3): 619-629, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31392775

RESUMEN

This study investigates the participation of PI3Kγ in the development of joint inflammation and dysfunction in an experimental model of acute gout in mice. Acute gout was induced by injection of monosodium urate (MSU) crystals into the tibiofemoral joint of mice. The involvement of PI3Kγ was evaluated using a selective inhibitor and mice deficient for PI3Kγ (PI3Kγ-/- ) or with loss of kinase activity. Neutrophils recovered from the inflamed joint were quantified and stained for phosphorylated Akt (pAkt) and production of reactive oxygen species (ROS). The adherence of leukocytes to the joint microvasculature was assessed by intravital microscopy and cleaved caspase-1 by Western blot. Injection of MSU crystals induced massive accumulation of neutrophils expressing phosphorylated Akt. In the absence of PI3Kγ, there was reduction of pAkt expression, chemokine production, and neutrophil recruitment. Genetic or pharmacological inhibition of PI3Kγ reduced the adherence of leukocytes to the joint microvasculature, even in joints with established inflammation. Neutrophils from PI3Kγ-/- mice produced less ROS than wild-type neutrophils. There was decreased joint damage and dysfunction in the absence of PI3Kγ. In addition, in the absence of PI3Kγ activity, there was reduction of cleaved caspase-1 and IL-1ß production in synovial tissue after injection of MSU crystals and leukotriene B4 . Our studies suggest that PI3Kγ is crucial for MSU crystal-induced acute joint inflammation. It is necessary for regulating caspase-1 activation and for mediating neutrophil migration and activation. Drugs that impair PI3Kγ function may be useful to control acute gout inflammation.


Asunto(s)
Artritis Gotosa/enzimología , Artritis Gotosa/inmunología , Caspasa 1/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Infiltración Neutrófila , Enfermedad Aguda , Animales , Adhesión Celular , Movimiento Celular , Fosfatidilinositol 3-Quinasa Clase Ib/deficiencia , Citoplasma/metabolismo , Activación Enzimática , Inflamasomas/metabolismo , Inflamación/patología , Interleucina-1beta/metabolismo , Articulaciones/patología , Leucotrieno B4/metabolismo , Masculino , Ratones Endogámicos C57BL , Microvasos/patología , Neutrófilos/metabolismo , Nocicepción , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Membrana Sinovial/irrigación sanguínea , Ácido Úrico
16.
PLoS Pathog ; 15(6): e1007880, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31211814

RESUMEN

The largest ever recorded epidemic of the Chikungunya virus (CHIKV) broke out in 2004 and affected four continents. Acute symptomatic infections are typically associated with the onset of fever and often debilitating polyarthralgia/polyarthritis. In this study, a systems biology approach was adopted to analyze the blood transcriptomes of adults acutely infected with the CHIKV. Gene signatures that were associated with viral RNA levels and the onset of symptoms were identified. Among these genes, the putative role of the Eukaryotic Initiation Factor (eIF) family genes and apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC3A) in the CHIKV replication process were displayed. We further compared these signatures with signatures induced by the Dengue virus infection and rheumatoid arthritis. Finally, we demonstrated that the CHIKV in vitro infection of murine bone marrow-derived macrophages induced IL-1 beta production in a mechanism that is significantly dependent on the inflammasome NLRP3 activation. The observations provided valuable insights into virus-host interactions during the acute phase and can be instrumental in the investigation of new and effective therapeutic interventions.


Asunto(s)
Artritis/inmunología , Fiebre Chikungunya/inmunología , Virus Chikungunya/fisiología , Citidina Desaminasa/inmunología , Proteínas/inmunología , Replicación Viral/inmunología , Adulto , Animales , Artritis/patología , Artritis/virología , Fiebre Chikungunya/patología , Virus del Dengue/inmunología , Virus del Dengue/patogenicidad , Femenino , Fiebre/inmunología , Fiebre/patología , Fiebre/virología , Estudios de Seguimiento , Humanos , Interleucina-1beta/inmunología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología
17.
Curr Opin Microbiol ; 52: 70-76, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31229882

RESUMEN

The inflammasomes are multi-molecular platforms that are activated in host cell cytoplasm when the innate immune cells are infected with pathogens or exposed to damage signals. Many independent groups reported that Leishmania infection trigger activation of the NLRP3 inflammasome in macrophages for restriction of intracellular parasite replication. Accordingly, Leishmania can dampen NLRP3 activation as an evasion strategy. In vivo, the NLRP3 inflammasome can promote parasite clearance, but the failure to eliminate parasites in the tissues together with sustained inflammasome activation can promote IL-1ß-mediated disease pathology. In this review, we discuss the recent data regarding activation of the NLRP3 inflammasome in response to Leishmania and the beneficial and detrimental effects of the inflammasome during development of Leishmaniasis.


Asunto(s)
Interacciones Huésped-Patógeno , Evasión Inmune , Inmunidad Innata , Inflamasomas/metabolismo , Leishmania/inmunología , Animales , Humanos , Leishmania/crecimiento & desarrollo , Leishmaniasis/parasitología , Leishmaniasis/patología , Macrófagos/inmunología , Macrófagos/parasitología
18.
J Leukoc Biol ; 106(3): 631-640, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31063608

RESUMEN

The NLRP3 inflammasome is activated in response to multiple stimuli and triggers activation of caspase-1 (CASP1), IL-1ß production, and inflammation. NLRP3 activation requires two signals. The first leads to transcriptional regulation of specific genes related to inflammation, and the second is triggered when pathogens, toxins, or specific compounds damage cellular membranes and/or trigger the production of reactive oxygen species (ROS). Here, we assess the requirement of the first signal (priming) for the activation of the NLRP3 inflammasome in bone marrow-derived macrophages (BMDMs) infected with Leishmania amazonensis. We found that BMDMs express the inflammasome components NLRP3, ASC, and CASP1 at sufficient levels to enable the assembly and activation of NLRP3 inflammasome in response to infection. Therefore, priming was not required for the formation of ASC specks, CASP1 activation (measured by fluorescent dye FAM-YVAD), and restriction of L. amazonensis replication via the NLRP3 inflammasome. By contrast, BMDM priming was required for CASP1 cleavage (p20) and IL-1ß secretion, because priming triggers robust up-regulation of pro-IL-1ß and CASP11 that are important for efficient processing of CASP1 and IL-1ß. Taken together, our data shed light into the cellular and molecular processes involved in activation of the NLRP3 in macrophages by Leishmania, a process that is important for the outcome of Leishmaniasis.


Asunto(s)
Inflamasomas/metabolismo , Leishmania mexicana/fisiología , Macrófagos/parasitología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Proteínas Adaptadoras de Señalización CARD/metabolismo , Activación Enzimática , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Leishmania mexicana/crecimiento & desarrollo , Leishmaniasis Cutánea/enzimología , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Ligandos , Lipopolisacáridos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Parásitos/crecimiento & desarrollo , Receptores de Interleucina-1/metabolismo , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo , Regulación hacia Arriba
19.
Methods Mol Biol ; 1921: 305-319, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30694501

RESUMEN

Legionella pneumophila is a gram-negative bacterium that infects many species of unicellular protozoa in freshwater environments. The human infection is accidental, and the bacteria may not have evolved strategies to bypass innate immune signaling in mammalian macrophages. Thus, L. pneumophila triggers many innate immune pathways including inflammasome activation. The inflammasomes are multimolecular platforms assembled in the host cell cytoplasm and lead to activation of inflammatory caspases. Inflammasome activation leads to secretion of inflammatory cytokines, such as IL-1ß and IL-18, and an inflammatory form of cell death called pyroptosis, which initiates with the induction of a pore in the macrophage membranes. In this chapter we provide detailed protocols to evaluate Legionella-induced inflammasome activation in macrophages, including real-time pore formation assay, western blotting to detect activation of inflammatory caspases (cleavage and pulldown), and the measurement of inflammatory cytokines.


Asunto(s)
Interacciones Huésped-Patógeno , Inflamasomas/metabolismo , Legionella/fisiología , Legionelosis/metabolismo , Legionelosis/microbiología , Macrófagos/metabolismo , Macrófagos/microbiología , Animales , Caspasas/genética , Caspasas/metabolismo , Citocinas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Mediadores de Inflamación/metabolismo , Legionelosis/inmunología , Macrófagos/inmunología , Ratones , Ratones Noqueados
20.
Cell Rep ; 26(2): 429-437.e5, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30625325

RESUMEN

Activation of the NLRP3 inflammasome by Leishmania parasites is critical for the outcome of leishmaniasis, a disease that affects millions of people worldwide. We investigate the mechanisms involved in NLRP3 activation and demonstrate that caspase-11 (CASP11) is activated in response to infection by Leishmania species and triggers the non-canonical activation of NLRP3. This process accounts for host resistance to infection in macrophages and in vivo. We identify the parasite membrane glycoconjugate lipophosphoglycan (LPG) as the molecule involved in CASP11 activation. Cytosolic delivery of LPG in macrophages triggers CASP11 activation, and infections performed with Lpg1-/- parasites reduce CASP11/NLRP3 activation. Unlike bacterial LPS, purified LPG does not activate mouse CASP11 (or human Casp4) in vitro, suggesting the participation of additional molecules for LPG-mediated CASP11 activation. Our data identify a parasite molecule involved in CASP11 activation, thereby establishing the mechanisms underlying inflammasome activation in response to Leishmania species.


Asunto(s)
Caspasas Iniciadoras/metabolismo , Glicoesfingolípidos/metabolismo , Inflamasomas/metabolismo , Leishmania/metabolismo , Leishmania/patogenicidad , Leishmaniasis/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Células Cultivadas , Células HEK293 , Humanos , Leishmaniasis/parasitología , Macrófagos/metabolismo , Macrófagos/parasitología , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA