Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Differ ; 29(6): 1267-1282, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34916628

RESUMEN

Cytoplasmic recognition of microbial lipopolysaccharides (LPS) in human cells is elicited by the caspase-4 and caspase-5 noncanonical inflammasomes, which induce a form of inflammatory cell death termed pyroptosis. Here we show that LPS-mediated activation of caspase-4 also induces a stress response promoting cellular senescence, which is dependent on the caspase-4 substrate gasdermin-D and the tumor suppressor p53. Furthermore, we found that the caspase-4 noncanonical inflammasome is induced and assembled in response to oncogenic RAS signaling during oncogene-induced senescence (OIS). Moreover, targeting caspase-4 expression in OIS showed its critical role in the senescence-associated secretory phenotype and the cell cycle arrest induced in cellular senescence. Finally, we observed that caspase-4 induction occurs in vivo in mouse models of tumor suppression and ageing. Altogether, we are showing that cellular senescence is induced by cytoplasmic LPS recognition by the noncanonical inflammasome and that this pathway is conserved in the cellular response to oncogenic stress.


Asunto(s)
Caspasas Iniciadoras , Inflamasomas , Animales , Caspasas Iniciadoras/inmunología , Senescencia Celular/inmunología , Citoplasma/inmunología , Humanos , Inmunidad Innata , Inflamasomas/inmunología , Lipopolisacáridos/farmacología , Ratones
2.
Sci Immunol ; 6(62)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34734155

RESUMEN

Innate immune signaling pathways comprise multiple proteins that promote inflammation. This multistep means of information transfer suggests that complexity is a prerequisite for pathway design. Herein, we test this hypothesis by studying caspases that regulate inflammasome-dependent inflammation. Several caspases differ in their ability to recognize bacterial LPS and cleave interleukin-1ß (IL-1ß). No caspase is known to contain both activities, yet distinct caspases with complementary activities bookend an LPS-induced pathway to IL-1ß cleavage. Using caspase-1/4 hybrid proteins present in canines as a guide, we identified molecular determinants of IL-1ß cleavage specificity within caspase-1. This knowledge enabled the redesign of human caspase-4 to operate as a one-protein signaling pathway, which intrinsically links LPS detection to IL-1ß cleavage and release, independent of inflammasomes. We identified caspase-4 homologues in multiple carnivorans which display the activities of redesigned human caspase-4. These findings illustrate natural signaling pathway diversity and highlight how multistep innate immune pathways can be condensed into a single protein.


Asunto(s)
Caspasas Iniciadoras/metabolismo , Interleucina-1beta/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Animales , Caspasas Iniciadoras/inmunología , Células Cultivadas , Perros , Escherichia coli/química , Humanos , Interleucina-1beta/inmunología , Receptores de Lipopolisacáridos/inmunología , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Ratones , Ratones Noqueados
3.
Cell Immunol ; 370: 104425, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34800762

RESUMEN

Asthma is an inflammatory lung disorder characterized by mucus hypersecretion, cellular infiltration, and bronchial hyper-responsiveness. House dust mites (HDM) are the most prevalent cause of allergic sensitization. Canonical and noncanonical inflammasomes are multiprotein complexes that assemble in response to pathogen or danger-associated molecular patterns (PAMPs or DAMPs). Murine caspase-11 engages the noncanonical inflammasome. We addressed the role of caspase-11 in mediating host responses to HDM and subsequent allergic inflammation using caspase-11-/- mice, which lack caspase-11 while express caspase-1. We found that HDM induce caspase-11 expression in vitro. The presence of IL-4 and IL-13 promote caspase-11 expression. Additionally, caspase-11-/- macrophages show reduced release of IL-6, IL-12, and KC, and express lower levels of costimulatory molecules (e.g., CD40, CD86 and MHCII) in response to HDM stimulation. Notably, HDM sensitization of caspase-11-/- mice resulted in similar levels of IgE responses and hypothermia in response to nasal HDM challenge compared to WT. However, analysis of cell numbers and cytokines in bronchiolar alveolar lavage fluid (BALF) and histopathology of representative lung segments demonstrate altered inflammatory responses and reduced neutrophilia in the airways of the caspase-11-/- mice. These findings indicate that caspase-11 regulates airway inflammation in response to HDM exposure.


Asunto(s)
Caspasas Iniciadoras/inmunología , Hipersensibilidad/inmunología , Neumonía/inmunología , Pyroglyphidae/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
Int Immunopharmacol ; 99: 107914, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34246059

RESUMEN

OBJECTIVES: Acute respiratory distress syndrome (ARDS) is characterized by an excessive pulmonary inflammatory response. Pyroptosis is a newly form of programmed inflammatory cell death that is triggered by inflammatory caspases. Studies have shown that Luteolin has powerful anti-inflammation effects through activating the function of regulatory T cells (Tregs). The study aimed at investigating the effects of Luteolin on CLP-induced ALI. METHODS: In our study, we employed the mouse cecal ligation and puncture (CLP) model to explore whether Luteolin contributed to alleviated lung injury in vivo. H&E staining and wet/dry (W/D) weight ratios were used to evaluate the severity of lung injury. The serum and BALF of cytokines were assessed by ELISA. The number of neutrophils in the BALF was counted. Immunohistochemistry of IL-10 and MPO in lung tissue was detected. The ROS level in lung was tested by ROS Assay Kit and expression of Gpx4 in lung tissue was detected by qRT-PCR and Western blotting. The regulatory T cells (Treg) population was analyzed in spleen and Peripheral blood mononuclear cells (PBMCs). The levels of caspase-11 protein, caspase-1 protein, GSDMD protein, IL-1α and IL-1ß protein in the lung tissue was evaluated by Western blotting. RESULTS: We found Luteolin significantly inhibits inflammation and attenuated CLP-induced lung injury in vivo, and the levels of, caspase-11, caspase-1, GSDMD, IL-1α and IL-1ß protein in the lungs of CLP mice decreased significantly after pretreatment with Luteolin. Furthermore, the results showed that Luteolin could increase Treg frequencies and IL-10 levels in serum and BALF of CLP mice. It is noteworthy that depleting Tregs reverse Luteolin ameliorated lung injury, and IL-10 neutralizing antibodies treatment aggravated lung pyroptosis. CONCLUSIONS: Our study illustrated that Luteolin contributed to alleviated lung injury, and attenuated caspase-11-dependent pyroptosis in the lung tissue of the CLP-induced ALI mouse model. The mechanisms could be related to regulating the frequency of Tregs and the levels of Treg derived IL-10. Treg cells were show to produce IL-10 and could alleviating caspase-11-dependent lung pyroptosis.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Luteolina/uso terapéutico , Sepsis/tratamiento farmacológico , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Animales , Antiinflamatorios/farmacología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Caspasas Iniciadoras/inmunología , Interleucina-10/inmunología , Recuento de Leucocitos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Luteolina/farmacología , Masculino , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Piroptosis/efectos de los fármacos , Sepsis/complicaciones , Sepsis/inmunología , Sepsis/patología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
5.
Cell Rep ; 35(3): 109012, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33882312

RESUMEN

Caspase-11 sensing of intracellular lipopolysaccharide (LPS) plays critical roles during infections and sepsis. However, the key cell types that sense intracellular LPS and their contributions to the host responses at the organismal level are not completely clear. Here, we show that macrophage/monocyte-specific caspase-11 plays a dominant role in mediating the pathological manifestations of endotoxemia, including gasdermin D (GSDMD) activation, interleukin (IL)-1ß, IL-18, and damage-associated molecular pattern (DAMP) release, tissue damage, and death. Surprisingly, caspase-11 expression in CD11c+ cells and intestinal epithelial cells (IECs) plays minor detrimental roles in LPS shock. In contrast, caspase-11 expression in neutrophils is dispensable for LPS-induced lethality. Importantly, caspase-11 sensing of intracellular LPS in LyzM+ myeloid cells and MRP8+ neutrophils, but not CD11c+ cells and IECs, is necessary for bacterial clearance and host survival during intracellular bacterial infection. Thus, we reveal hierarchical cell-type-specific roles of caspase-11 that govern the host-protective and host-detrimental functions of the cytosolic LPS surveillance.


Asunto(s)
Caspasas Iniciadoras/genética , Lipopolisacáridos/toxicidad , Macrófagos Peritoneales/inmunología , Neutrófilos/inmunología , Choque Séptico/inmunología , Bazo/inmunología , Animales , Burkholderia/crecimiento & desarrollo , Burkholderia/patogenicidad , Antígenos CD11/genética , Antígenos CD11/inmunología , Calgranulina A/genética , Calgranulina A/inmunología , Caspasas Iniciadoras/inmunología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Femenino , Regulación de la Expresión Génica , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Hígado/inmunología , Hígado/microbiología , Macrófagos Peritoneales/microbiología , Masculino , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/inmunología , Monocitos/inmunología , Monocitos/microbiología , Neutrófilos/microbiología , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/inmunología , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/inmunología , Choque Séptico/genética , Choque Séptico/microbiología , Choque Séptico/mortalidad , Transducción de Señal , Bazo/microbiología , Análisis de Supervivencia
6.
Front Immunol ; 11: 585146, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329561

RESUMEN

The innate immune response to lipopolysaccharide is essential for host defense against Gram-negative bacteria. In response to bacterial infection, the TLR4/MD-2 complex that is expressed on the surface of macrophages, monocytes, dendritic, and epithelial cells senses picomolar concentrations of endotoxic LPS and triggers the production of various pro-inflammatory mediators. In addition, LPS from extracellular bacteria which is either endocytosed or transfected into the cytosol of host cells or cytosolic LPS produced by intracellular bacteria is recognized by cytosolic proteases caspase-4/11 and hosts guanylate binding proteins that are involved in the assembly and activation of the NLRP3 inflammasome. All these events result in the initiation of pro-inflammatory signaling cascades directed at bacterial eradication. However, TLR4-mediated signaling and caspase-4/11-induced pyroptosis are largely involved in the pathogenesis of chronic and acute inflammation. Both extra- and intracellular LPS receptors-TLR4/MD-2 complex and caspase-4/11, respectively-are able to directly bind the lipid A motif of LPS. Whereas the structural basis of lipid A recognition by the TLR4 complex is profoundly studied and well understood, the atomic mechanism of LPS/lipid A interaction with caspase-4/11 is largely unknown. Here we describe the LPS-induced TLR4 and caspase-4/11 mediated signaling pathways and their cross-talk and scrutinize specific structural features of the lipid A motif of diverse LPS variants that have been reported to activate caspase-4/11 or to induce caspase-4/11 mediated activation of NLRP3 inflammasome (either upon transfection of LPS in vitro or upon infection of cell cultures with intracellular bacteria or by LPS as a component of the outer membrane vesicles). Generally, inflammatory caspases show rather similar structural requirements as the TLR4/MD-2 complex, so that a "basic" hexaacylated bisphosphorylated lipid A architecture is sufficient for activation. However, caspase-4/11 can sense and respond to much broader variety of lipid A variants compared to the very "narrow" specificity of TLR4/MD-2 complex as far as the number and the length of lipid chains attached at the diglucosamine backbone of lipid A is concerned. Besides, modification of the lipid A phosphate groups with positively charged appendages such as phosphoethanolamine or aminoarabinose could be essential for the interaction of lipid A/LPS with inflammatory caspases and related proteins.


Asunto(s)
Caspasas Iniciadoras/inmunología , Caspasas/inmunología , Inmunidad Innata/inmunología , Lipopolisacáridos/inmunología , Receptor Toll-Like 4/inmunología , Animales , Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Humanos , Inflamasomas/inmunología , Inflamación/inmunología
7.
Nat Immunol ; 21(8): 880-891, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32541830

RESUMEN

Bacterial lipopolysaccharide triggers human caspase-4 (murine caspase-11) to cleave gasdermin-D and induce pyroptotic cell death. How lipopolysaccharide sequestered in the membranes of cytosol-invading bacteria activates caspases remains unknown. Here we show that in interferon-γ-stimulated cells guanylate-binding proteins (GBPs) assemble on the surface of Gram-negative bacteria into polyvalent signaling platforms required for activation of caspase-4. Caspase-4 activation is hierarchically controlled by GBPs; GBP1 initiates platform assembly, GBP2 and GBP4 control caspase-4 recruitment, and GBP3 governs caspase-4 activation. In response to cytosol-invading bacteria, activation of caspase-4 through the GBP platform is essential to induce gasdermin-D-dependent pyroptosis and processing of interleukin-18, thereby destroying the replicative niche for intracellular bacteria and alerting neighboring cells, respectively. Caspase-11 and GBPs epistatically protect mice against lethal bacterial challenge. Multiple antagonists of the pathway encoded by Shigella flexneri, a cytosol-adapted bacterium, provide compelling evolutionary evidence for the importance of the GBP-caspase-4 pathway in antibacterial defense.


Asunto(s)
Caspasas Iniciadoras/inmunología , Proteínas de Unión al GTP/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Inflamasomas/inmunología , Transducción de Señal/inmunología , Animales , Bacterias Gramnegativas/inmunología , Células HeLa , Humanos , Lipopolisacáridos/inmunología , Ratones , Piroptosis/inmunología
8.
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
9.
PLoS Pathog ; 16(4): e1008498, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32282854

RESUMEN

We investigated the role of the inflammasome effector caspases-1 and -11 during Salmonella enterica serovar Typhimurium infection of murine intestinal epithelial cells (IECs). Salmonella burdens were significantly greater in the intestines of caspase-1/11 deficient (Casp1/11-/-), Casp1-/- and Casp11-/- mice, as compared to wildtype mice. To determine if this reflected IEC-intrinsic inflammasomes, enteroid monolayers were derived and infected with Salmonella. Casp11-/- and wildtype monolayers responded similarly, whereas Casp1-/- and Casp1/11-/- monolayers carried significantly increased intracellular burdens, concomitant with marked decreases in IEC shedding and death. Pretreatment with IFN-γ to mimic inflammation increased caspase-11 levels and IEC death, and reduced Salmonella burdens in Casp1-/- monolayers, while high intracellular burdens and limited cell shedding persisted in Casp1/11-/- monolayers. Thus caspase-1 regulates inflammasome responses in IECs at baseline, while proinflammatory activation of IECs reveals a compensatory role for caspase-11. These results demonstrate the importance of IEC-intrinsic canonical and non-canonical inflammasomes in host defense against Salmonella.


Asunto(s)
Caspasa 1/inmunología , Caspasas Iniciadoras/inmunología , Inflamasomas/inmunología , Intestinos/enzimología , Intestinos/inmunología , Infecciones por Salmonella/enzimología , Salmonella typhimurium/inmunología , Animales , Células Epiteliales/enzimología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Femenino , Inmunidad Mucosa , Inflamasomas/metabolismo , Interferón gamma/inmunología , Mucosa Intestinal/enzimología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Intestinos/microbiología , Lipopolisacáridos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Salmonella/inmunología , Salmonella typhimurium/patogenicidad
10.
Nat Commun ; 11(1): 1055, 2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-32103022

RESUMEN

Activated caspase-1 and caspase-11 induce inflammatory cell death in a process termed pyroptosis. Here we show that Prostaglandin E2 (PGE2) inhibits caspase-11-dependent pyroptosis in murine and human macrophages. PGE2 suppreses caspase-11 expression in murine and human macrophages and in the airways of mice with allergic inflammation. Remarkably, caspase-11-deficient mice are strongly resistant to developing experimental allergic airway inflammation, where PGE2 is known to be protective. Expression of caspase-11 is elevated in the lung of wild type mice with allergic airway inflammation. Blocking PGE2 production with indomethacin enhances, whereas the prostaglandin E1 analog misoprostol inhibits lung caspase-11 expression. Finally, alveolar macrophages from asthma patients exhibit increased expression of caspase-4, a human homologue of caspase-11. Our findings identify PGE2 as a negative regulator of caspase-11-driven pyroptosis and implicate caspase-4/11 as a critical contributor to allergic airway inflammation, with implications for pathophysiology of asthma.


Asunto(s)
Asma/patología , Caspasas Iniciadoras/metabolismo , Dinoprostona/metabolismo , Macrófagos/inmunología , Piroptosis/fisiología , Animales , Antiinflamatorios no Esteroideos/farmacología , Asma/inmunología , Caspasas Iniciadoras/genética , Caspasas Iniciadoras/inmunología , Células Cultivadas , Sinergismo Farmacológico , Femenino , Humanos , Indometacina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Misoprostol/farmacología
11.
Cell ; 180(1): 64-78.e16, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31923400

RESUMEN

Enteric-associated neurons (EANs) are closely associated with immune cells and continuously monitor and modulate homeostatic intestinal functions, including motility and nutrient sensing. Bidirectional interactions between neuronal and immune cells are altered during disease processes such as neurodegeneration or irritable bowel syndrome. We investigated the effects of infection-induced inflammation on intrinsic EANs (iEANs) and the role of intestinal muscularis macrophages (MMs) in this context. Using murine models of enteric infections, we observed long-term gastrointestinal symptoms, including reduced motility and loss of excitatory iEANs, which was mediated by a Nlrp6- and Casp11-dependent mechanism, depended on infection history, and could be reversed by manipulation of the microbiota. MMs responded to luminal infection by upregulating a neuroprotective program via ß2-adrenergic receptor (ß2-AR) signaling and mediated neuronal protection through an arginase 1-polyamine axis. Our results identify a mechanism of neuronal death post-infection and point to a role for tissue-resident MMs in limiting neuronal damage.


Asunto(s)
Mucosa Intestinal/inmunología , Macrófagos/inmunología , Receptores Adrenérgicos beta 2/metabolismo , Adrenérgicos , Animales , Arginasa/metabolismo , Caspasas Iniciadoras/inmunología , Caspasas Iniciadoras/metabolismo , Sistema Nervioso Entérico/inmunología , Sistema Nervioso Entérico/metabolismo , Femenino , Enfermedades Gastrointestinales , Microbioma Gastrointestinal , Infecciones , Inflamación/inmunología , Mucosa Intestinal/metabolismo , Intestino Delgado/inmunología , Intestinos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microbiota , Neuronas/fisiología , Receptores Adrenérgicos beta 2/inmunología , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Transducción de Señal
12.
Front Immunol ; 10: 1904, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31440260

RESUMEN

Inflammasome activation can trigger an inflammatory and innate immune response through the release of cytokines and induction of pyroptosis. A dysfunctional inflammasome has been implicated in the development of human pathologies, including sepsis and septic shock. Here, we show that advanced glycosylation end-product specific receptor (AGER/RAGE) is required for caspase-11 inflammasome activation in macrophages. A nuclear damage-associated molecular pattern (nDAMP) complex, including high-mobility group box 1, histone, and DNA, can promote caspase-11-mediated gasdermin D cleavage, interleukin 1ß proteolytic maturation, and lactate dehydrogenase release. The inhibition of AGER-mediated lipid peroxidation via arachidonate 5-lipoxygenase (ALOX5) limits caspase-11 inflammasome activation and pyroptosis in macrophages in response to nDAMPs or cytosolic lipopolysaccharide. Importantly, the pharmacologic inhibition of the AGER-ALOX5 pathway or global depletion (Ager-/-) or conditional depletion of AGER in myeloid cells (AgerMye-/-) protects against lipopolysaccharide-induced septic death in poly(I:C)-primed mice. These data identify a molecular basis for caspase-11 inflammasome activation and provide a potential strategy to treat sepsis.


Asunto(s)
Caspasas Iniciadoras/inmunología , Muerte Celular/inmunología , Inflamasomas/inmunología , Peroxidación de Lípido/inmunología , Receptor para Productos Finales de Glicación Avanzada/inmunología , Sepsis/inmunología , Animales , Araquidonato 5-Lipooxigenasa/inmunología , Femenino , Inmunidad Innata/inmunología , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología
13.
J Invest Dermatol ; 139(6): 1306-1317, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30571969

RESUMEN

Inflammatory caspases, activated within the inflammasome, are responsible for the maturation and secretion of IL-1ß/IL-18. Although their expression in psoriasis was shown several years ago, little is known about the role of inflammatory caspases in the context of psoriasis. Here, we confirmed that caspases 1, 4, and 5 are activated in lesional skin from psoriasis patients. We showed in three psoriasis-like models that inflammatory caspases are activated, and accordingly, caspase 1/11 invalidation or pharmacological inhibition by Ac-YVAD-CMK (i.e., Ac-Tyr-Val-Ala-Asp-chloromethylketone) injection induced a decrease in ear thickness, erythema, scaling, inflammatory cytokine expression, and immune cell infiltration in mice. We observed that keratinocytes were primed to secrete IL-1ß when cultured in conditions mimicking psoriasis. Generation of chimeric mice by bone marrow transplantation was carried out to decipher the respective contribution of keratinocytes and/or immune cells in the activation of inflammatory caspases during psoriasis-like inflammatory response. Our data showed that the presence of caspase 1/11 in the immune system is sufficient for a fully inflammatory response, whereas the absence of caspase 1/11 in keratinocytes/fibroblasts had no impact. In summary, our study indicates that inflammatory caspases activated in immune cells are implicated in psoriasis pathogenesis.


Asunto(s)
Caspasa 1/deficiencia , Inhibidores de Caspasas/administración & dosificación , Caspasas Iniciadoras/deficiencia , Psoriasis/tratamiento farmacológico , Clorometilcetonas de Aminoácidos/administración & dosificación , Animales , Biopsia , Trasplante de Médula Ósea , Caspasa 1/genética , Caspasa 1/inmunología , Caspasas Iniciadoras/genética , Caspasas Iniciadoras/inmunología , Caspasas Iniciadoras/metabolismo , Células Cultivadas , Ensayos Clínicos como Asunto , Femenino , Humanos , Inyecciones Intraperitoneales , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Queratinocitos , Masculino , Ratones , Ratones Noqueados , Cultivo Primario de Células , Psoriasis/inmunología , Psoriasis/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Piel/inmunología , Piel/patología , Quimera por Trasplante
14.
J Biol Chem ; 293(30): 11878-11890, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-29875158

RESUMEN

The antiviral cGMP-AMP (cGAMP)-stimulator of interferon genes (STING) pathway is well characterized in mammalian cells. However, whether this pathway also plays a role in insect antiviral immunity is unknown. In this study, we found that cGAMP is produced in silkworm (Bombyx mori) cells infected with nucleopolyhedrovirus (NPV). In searches for STING-related sequences, we identified BmSTING, a potential cGAMP sensor in B. mori We observed that BmSTING overexpression effectively inhibits NPV replication in silkworm larvae, whereas dsRNA-mediated BmSTING knockdown resulted in higher viral load. Cleavage and nuclear translocation of BmRelish, a NF-κB-related transcription factor, was also observed when BmSTING was overexpressed and was enhanced by cGAMP stimulation or viral infection of B. mori larvae. Moreover, we identified a caspase-8-like protein (BmCasp8L) as a BmSTING-interacting molecule and as a suppressor of BmSTING-mediated BmRelish activation. Interestingly, cGAMP stimulation decreased BmCasp8L binding to BmSTING and increased BmRelish activity. Of note, an interaction between death-related ced-3/Nedd2-like caspase (BmDredd) and BmSTING promoted BmRelish cleavage for efficient antiviral signaling and protection of insect cells from viral infection. Our findings have uncovered BmSTING as a critical mediator of antiviral immunity in the model insect B. mori and have identified several BmSTING-interacting proteins that control antiviral defenses.


Asunto(s)
Bombyx/inmunología , Bombyx/virología , Caspasas Iniciadoras/inmunología , Proteínas de Insectos/inmunología , FN-kappa B/inmunología , Nucleopoliedrovirus/inmunología , Nucleótidos Cíclicos/inmunología , Animales , Inmunidad Innata , Transducción de Señal
15.
Front Immunol ; 9: 670, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29675024

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic fibro-proliferative disease characterized by poor prognosis, with a mean survival of ~2-3 years after definite diagnosis. The cause of IPF is still unknown but it is a heterogeneous condition in which the aberrant deposition of extracellular matrix leads to extensive lung remodeling. This remodeling is a consequence of inflammatory responses, but the mechanisms involved are poorly understood. In this study, we first analyzed a bleomycin-induced mouse model, which showed that higher expression of IL-1ß, but not IL-18, was correlated to pulmonary cell infiltration and fibrosis. Then, we found that peripheral blood mononuclear cells (PBMCs) from IPF patients released IL-1α and IL-18 in a NLRP3- and calpain-independent manner after LPS ± ATP stimulation. Instead, the activation of the absent in melanoma 2 (AIM2) inflammasome induced the release of IL-1α in a caspase-1-/caspase-8-independent manner; whereas IL-18 release was caspase-1 dependent. These effects correlated with the release of the pro-fibrotic TGF-ß, which was induced by AIM2 activation in a caspase-1- and TLR4-independent manner, but dependent on IL-1α. In this context, the activation of AIM2 induced the release of caspase-4 from IPF-derived PBMCs, which correlated with the mRNA levels of this caspase that was higher in IPF than in healthy PBMCs. In conclusion, our findings identify a novel molecular mechanism whereby the activation of AIM2 could lead to the activation of the non-canonical inflammasome (caspase-4 dependent) that induces the release of IL-1α responsible for the release of TGF-ß from PBMCs of IPF patients.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Fibrosis Pulmonar Idiopática/inmunología , Inflamasomas/inmunología , Leucocitos Mononucleares/inmunología , Adulto , Animales , Bleomicina , Caspasas Iniciadoras/inmunología , Citocinas/inmunología , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Ratones Endogámicos BALB C , Persona de Mediana Edad
16.
Nat Commun ; 6: 8761, 2015 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-26508369

RESUMEN

Monocytes promote the early host response to infection releasing key pro-inflammatory cytokines, such as IL-1ß. The biologically inactive IL-1ß precursor is processed to active form by inflammasomes, multi-protein complexes activating caspase-1. Human monocytes exhibit an unconventional one-step pathway of inflammasome activation in response to lipopolysaccharide (LPS) alone. Although this lineage-restricted mechanism is likely to contribute to the pathology of endotoxin shock, signalling pathways regulating this mechanism are currently unknown. Here we report that caspase-4 and caspase-5 mediate IL-1α and IL-1ß release from human monocytes after LPS stimulation. Although caspase-4 remains uncleaved, caspase-5 undergoes rapid processing upon LPS treatment. We also identify an additional caspase-5 cleavage product in LPS-stimulated monocytes, which correlates with IL-1 secretion. This one-step pathway requires Syk activity and Ca(2+) flux instigated by CD14/TLR4-mediated LPS internalization. Identification of caspase-4/5 as the key determinants of one-step inflammasome activation in human monocytes provides potential targets for therapeutic intervention in endotoxin shock.


Asunto(s)
Caspasas Iniciadoras/inmunología , Caspasas/inmunología , Inflamasomas/inmunología , Monocitos/enzimología , Caspasas/genética , Caspasas Iniciadoras/genética , Humanos , Inflamasomas/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Lipopolisacáridos/inmunología , Monocitos/inmunología , Transducción de Señal
17.
Eur J Immunol ; 45(10): 2918-26, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26173988

RESUMEN

Humans encode two inflammatory caspases that detect cytoplasmic LPS, caspase-4 and caspase-5. When activated, these trigger pyroptotic cell death and caspase-1-dependent IL-1ß production; however the mechanism underlying this process is not yet confirmed. We now show that a specific NLRP3 inhibitor, MCC950, prevents caspase-4/5-dependent IL-1ß production elicited by transfected LPS. Given that both caspase-4 and caspase-5 can detect cytoplasmic LPS, it is possible that these proteins exhibit some degree of redundancy. Therefore, we generated human monocytic cell lines in which caspase-4 and caspase-5 were genetically deleted either individually or together. We found that the deletion of caspase-4 suppressed cell death and IL-1ß production following transfection of LPS into the cytoplasm, or in response to infection with Salmonella typhimurium. Although deletion of caspase-5 did not confer protection against transfected LPS, cell death and IL-1ß production were reduced after infection with Salmonella. Furthermore, double deletion of caspase-4 and caspase-5 had a synergistic effect in the context of Salmonella infection. Our results identify the NLRP3 inflammasome as the specific platform for IL-1ß maturation, downstream of cytoplasmic LPS detection by caspase-4/5. We also show that both caspase-4 and caspase-5 are functionally important for appropriate responses to intracellular Gram-negative bacteria.


Asunto(s)
Proteínas Portadoras/inmunología , Caspasas Iniciadoras/inmunología , Caspasas/inmunología , Lipopolisacáridos/inmunología , Monocitos/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Línea Celular Tumoral , Humanos , Interleucina-1beta/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR
18.
Eur J Immunol ; 45(10): 2911-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26174085

RESUMEN

Inflammasome activation culminates in activation of caspase-1, which leads to the maturation and subsequent release of cytokines of the interleukin 1 (IL-1) family and results in a particular form of cell death known as pyroptosis. In addition, in the murine system, a so-called non-canonical inflammasome involving caspase-11 has been described that directly responds to cytosolic LPS. Here, we show that the human monocytic cell line THP1 activates the inflammasome in response to cytosolic LPS in a TLR4-independent fashion. This response is mediated by caspase-4 and accompanied by caspase-1 activation, pyroptosis, and IL-1ß maturation. In addition to caspase-4, efficient IL-1ß conversion upon intracellular LPS delivery relies on potassium efflux, NLRP3, ASC, and caspase-1, indicating that although caspase-4 activation alone is sufficient to induce pyroptosis, this process depends on the NLRP3 inflammasome activation to drive IL-1ß maturation. Altogether, this study provides evidence for the presence of a non-canonical inflammasome in humans and its dependence on caspase-4.


Asunto(s)
Proteínas Portadoras/inmunología , Caspasas Iniciadoras/inmunología , Inflamasomas/inmunología , Células Mieloides/inmunología , Proteínas Portadoras/genética , Caspasa 1/genética , Caspasa 1/inmunología , Caspasas Iniciadoras/genética , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Inflamasomas/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Lipopolisacáridos/farmacología , Células Mieloides/citología , Proteína con Dominio Pirina 3 de la Familia NLR
19.
Proc Natl Acad Sci U S A ; 112(21): 6688-93, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25964352

RESUMEN

Inflammasomes are critical for host defense against bacterial pathogens. In murine macrophages infected by gram-negative bacteria, the canonical inflammasome activates caspase-1 to mediate pyroptotic cell death and release of IL-1 family cytokines. Additionally, a noncanonical inflammasome controlled by caspase-11 induces cell death and IL-1 release. However, humans do not encode caspase-11. Instead, humans encode two putative orthologs: caspase-4 and caspase-5. Whether either ortholog functions similar to caspase-11 is poorly defined. Therefore, we sought to define the inflammatory caspases in primary human macrophages that regulate inflammasome responses to gram-negative bacteria. We find that human macrophages activate inflammasomes specifically in response to diverse gram-negative bacterial pathogens that introduce bacterial products into the host cytosol using specialized secretion systems. In primary human macrophages, IL-1ß secretion requires the caspase-1 inflammasome, whereas IL-1α release and cell death are caspase-1-independent. Instead, caspase-4 mediates IL-1α release and cell death. Our findings implicate human caspase-4 as a critical regulator of noncanonical inflammasome activation that initiates defense against bacterial pathogens in primary human macrophages.


Asunto(s)
Caspasas Iniciadoras/inmunología , Bacterias Gramnegativas/inmunología , Bacterias Gramnegativas/patogenicidad , Inflamasomas/inmunología , Animales , Caspasa 1/inmunología , Muerte Celular , Células Cultivadas , Humanos , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Legionella pneumophila/inmunología , Legionella pneumophila/patogenicidad , Lipopolisacáridos/toxicidad , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/patogenicidad
20.
Immunol Rev ; 265(1): 75-84, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25879285

RESUMEN

As a front line of defense against pathogenic microbes, our body employs a primitive, yet highly sophisticated and potent innate immune response pathway collectively referred to as the inflammasome. Innate immune cells, epithelial cells, and many other cell types are capable of detecting infection or tissue injury and mounting a coordinated molecular defense. For example, Gram-negative bacteria are specifically detected via a surveillance mechanism that involves activation of extracellular receptors such as Toll-like receptors (TLRs) followed by intracellular recognition and activation of pathways such as caspase-11 (caspase-4/5 in humans). Importantly, lipopolysaccharide (LPS), the major component of the outer membrane of Gram-negative bacteria, is a strong trigger of these pathways. Extracellular LPS primarily stimulates TLR4, which can serve as a priming signal for expression of inflammasome components. Intracellular LPS can then trigger caspase-11-dependent inflammasome activation in the cytoplasm. Here, we briefly review the burgeoning caspase-11-dependent non-canonical inflammasome field, focusing mainly on the innate sensing of LPS.


Asunto(s)
Infecciones Bacterianas/metabolismo , Caspasas Iniciadoras/metabolismo , Caspasas/metabolismo , Complejos Multiproteicos/metabolismo , Animales , Infecciones Bacterianas/inmunología , Caspasas/inmunología , Caspasas Iniciadoras/inmunología , Humanos , Inmunidad Innata , Lipopolisacáridos/inmunología , Ratones , Complejos Multiproteicos/inmunología , Transducción de Señal , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...