Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 252
Filtrar
1.
Nat Rev Immunol ; 24(7): 518-535, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38374299

RESUMEN

Inflammasomes are supramolecular complexes that form in the cytosol in response to pathogen-associated and damage-associated stimuli, as well as other danger signals that perturb cellular homoeostasis, resulting in host defence responses in the form of cytokine release and programmed cell death (pyroptosis). Inflammasome activity is closely associated with numerous human disorders, including rare genetic syndromes of autoinflammation, cardiovascular diseases, neurodegeneration and cancer. In recent years, a range of inflammasome components and their functions have been discovered, contributing to our knowledge of the overall machinery. Here, we review the latest advances in inflammasome biology from the perspective of structural and mechanistic studies. We focus on the most well-studied components of the canonical inflammasome - NAIP-NLRC4, NLRP3, NLRP1, CARD8 and caspase-1 - as well as caspase-4, caspase-5 and caspase-11 of the noncanonical inflammasome, and the inflammasome effectors GSDMD and NINJ1. These structural studies reveal important insights into how inflammasomes are assembled and regulated, and how they elicit the release of IL-1 family cytokines and induce membrane rupture in pyroptosis.


Asunto(s)
Inflamasomas , Piroptosis , Inflamasomas/inmunología , Inflamasomas/metabolismo , Humanos , Piroptosis/inmunología , Animales , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas Adaptadoras de Señalización CARD/inmunología , Proteínas Adaptadoras de Señalización CARD/genética , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Proteína Inhibidora de la Apoptosis Neuronal/inmunología , Proteína Inhibidora de la Apoptosis Neuronal/genética , Proteínas de Unión a Fosfato/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Caspasas/metabolismo , Caspasas/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas NLR/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/inmunología , Gasderminas
2.
Front Immunol ; 12: 755961, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867993

RESUMEN

Non-canonical inflammasome activation by mouse caspase-11 (or human CASPASE-4/5) is crucial for the clearance of certain gram-negative bacterial infections, but can lead to severe inflammatory damage. Factors that promote non-canonical inflammasome activation are well recognized, but less is known about the mechanisms underlying its negative regulation. Herein, we identify that the caspase-11 inflammasome in mouse and human macrophages (Mϕ) is negatively controlled by the zinc (Zn2+) regulating protein, metallothionein 3 (MT3). Upon challenge with intracellular lipopolysaccharide (iLPS), Mϕ increased MT3 expression that curtailed the activation of caspase-11 and its downstream targets caspase-1 and interleukin (IL)-1ß. Mechanistically, MT3 increased intramacrophage Zn2+ to downmodulate the TRIF-IRF3-STAT1 axis that is prerequisite for caspase-11 effector function. In vivo, MT3 suppressed activation of the caspase-11 inflammasome, while caspase-11 and MT3 synergized in impairing antibacterial immunity. The present study identifies an important yin-yang relationship between the non-canonical inflammasome and MT3 in controlling inflammation and immunity to gram-negative bacteria.


Asunto(s)
Caspasas/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Inflamasomas/inmunología , Macrófagos/inmunología , Metalotioneína 3/inmunología , Zinc/inmunología , Animales , Caspasas/metabolismo , Infecciones por Bacterias Gramnegativas/metabolismo , Humanos , Inflamasomas/metabolismo , Macrófagos/metabolismo , Metalotioneína 3/metabolismo , Ratones , Ratones Endogámicos C57BL , Zinc/metabolismo
3.
J Immunol Res ; 2021: 1125199, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34595242

RESUMEN

BACKGROUND: Acute lung injury (ALI) always leads to severe inflammation. As inflammation and oxidative stress are the common pathological basis of endotoxin-induced inflammatory injury and ischemic reperfusion injury (IRI), we speculate that remote ischemic preconditioning (RIPC) can be protective for ALI when used as remote inflammatory preconditioning (RInPC). METHOD: A total of 21 Sprague-Dawley rats were used for the animal experiments. Eighteen rats were equally and randomly divided into the control (NS injection), LPS (LPS injection), and RInPC groups. The RInPC was performed prior to the LPS injection via tourniquet blockage of blood flow to the right hind limb and adopted three cycles of 5 min tying followed by 5 min untying. Animals were sacrificed 24 hours later. There were 2 rats in the LPS group and 1 in the RInPC group who died before the end of the experiment. Supplementary experiments in the LPS and RInPC groups were conducted to ensure that 6 animals in each group reached the end of the experiment. RESULTS: In the present study, we demonstrated that the RInPC significantly attenuated the LPS-induced ALI in rats. Apoptotic cells were reduced significantly by the RInPC, with the simultaneous improvement of apoptosis-related proteins. Reduction of MPO and MDA and increasing of SOD activity were found significantly improved by the RInPC. Increasing of TNF-α, IL-1ß, and IL-6 induced by the LPS was inhibited, while IL-10 was significantly increased by RInPC, compared to the LPS group. CONCLUSION: RInPC could inhibit inflammation and attenuate oxidative stress, thereby reducing intrinsic apoptosis and providing lung protection in the LPS-induced ALI in rats.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Apoptosis/inmunología , Precondicionamiento Isquémico/métodos , Pulmón/inmunología , Transducción de Señal/inmunología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Animales , Caspasas/inmunología , Caspasas/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Lipopolisacáridos , Pulmón/metabolismo , Pulmón/patología , Malondialdehído/inmunología , Malondialdehído/metabolismo , Peroxidasa/inmunología , Peroxidasa/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas Sprague-Dawley , Superóxido Dismutasa/inmunología , Superóxido Dismutasa/metabolismo , Proteína X Asociada a bcl-2/inmunología , Proteína X Asociada a bcl-2/metabolismo
4.
Mol Immunol ; 137: 114-123, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34242920

RESUMEN

Toll-like receptors (TLRs) represent first line of host defence against microbes. Amongst different TLRs, TLR22 is exclusively expressed in non-mammalian vertebrates, including fish. The precise role of TLR22 in fish-immunity remains abstruse. Herein, we used headkidney macrophages (HKM) from Clarias gariepinus and deciphered its role in fish-immunity. Highest tlr22 expression was observed in the immunocompetent organ - headkidney; nonetheless expression in other tissues suggests its possible involvement in non-immune sites also. Aeromonas hydrophila infection up-regulates tlr22 expression in HKM. Our RNAi based study suggested TLR22 restricts intracellular survival of A. hydrophila. Inhibitor and RNAi studies further implicated TLR22 induces pro-inflammatory cytokines TNF-α and IL-1ß. We observed heightened caspase-1 activity and our results suggest the role of TLR22 in activating TNF-α/caspase-1/IL-1ß cascade leading to caspase-3 mediated apoptosis of A. hydrophila-infected HKM. We conclude, TLR22 plays critical role in immune-surveillance and triggers pro-inflammatory cytokines leading to caspase mediated HKM apoptosis and pathogen clearance.


Asunto(s)
Aeromonas hydrophila/inmunología , Apoptosis/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Inflamación/inmunología , Macrófagos/inmunología , Receptores Toll-Like/inmunología , Animales , Caspasas/inmunología , Bagres/inmunología , Bagres/microbiología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/microbiología , Proteínas de Peces/inmunología , Infecciones por Bacterias Gramnegativas/microbiología , Riñón Cefálico/inmunología , Riñón Cefálico/microbiología , Inflamación/microbiología , Interleucina-1beta/inmunología , Macrófagos/microbiología , Factor de Necrosis Tumoral alfa/inmunología
5.
Front Immunol ; 12: 644862, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093533

RESUMEN

NLRP3 inflammasome has emerged as a crucial regulator of inflammatory bowel disease (IBD) characterized by a chronic inflammatory disease of the gastrointestinal tract. The expression of MCT4 is significantly increased in intestinal mucosal tissue of IBD, which has been identified to regulate intestinal barrier function. However, the function of MCT4 in cell pyroptosis remained unknown. In this study, we have established a stable cell line with MCT4 overexpression in HT-29 and CaCO2 cells, respectively. Functional analysis revealed that ectopic expression of MCT4 in CaCO2 cells contributed to cell pyroptosis as evidenced by LDH assay, which is largely attributed to Caspase-1-mediated canonical pyroptosis, but not Caspase-4 and Caspase-5, leading to cleave pro-IL-1ß and IL-18 into mature form and release mediated by cleaved GSDMD. Mechanically, MCT4 overexpression in HT-29 and CaCO2 cell triggered the phosphorylation of ERK1/2 and NF-κB p65, while inhibition of MCT4 by MCT inhibitor α-Cyano-4-hydroxycinnamic acid (α-CHCA) in HT-29 and CaCO2 cells led to a significant downregulation of ERK1/2 and NF-κB activity. What's more, blockade of ERK1/2-NF-κB pathway could reverse the promotion effect of MCT4 on IL-1ß expression. Importantly, both MCT4 and Caspase-1, GSDMD were significantly increased in patients with IBD, and a positive clinical correlation between MCT4 and Caspase-1 expression was observed (p < 0.001). Taken together, these findings suggested that MCT4 promoted Caspase-1-mediated canonical cell pyroptosis to aggravate intestinal inflammation in intestinal epithelial cells (IECs) through the ERK1/2-NF-κB pathway.


Asunto(s)
Enfermedades Inflamatorias del Intestino/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Transportadores de Ácidos Monocarboxílicos/inmunología , Proteínas Musculares/inmunología , Piroptosis/inmunología , Células CACO-2 , Caspasas/inmunología , Células HT29 , Humanos , Inflamación/inmunología , Inflamación/patología , Enfermedades Inflamatorias del Intestino/patología , Interleucina-18/inmunología , Interleucina-1beta/inmunología , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Factor de Transcripción ReIA/inmunología
6.
mBio ; 12(3): e0105921, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34154417

RESUMEN

Burkholderia infections can result in serious diseases with high mortality, such as melioidosis, and they are difficult to treat with antibiotics. Innate immunity is critical for cell-autonomous clearance of intracellular pathogens like Burkholderia by regulating programmed cell death. Inflammasome-dependent inflammatory cytokine release and cell death contribute to host protection against Burkholderia pseudomallei and Burkholderia thailandensis; however, the contribution of apoptosis and necroptosis to protection is not known. Here, we found that bone marrow-derived macrophages (BMDMs) lacking key components of pyroptosis died via apoptosis during infection. BMDMs lacking molecules required for pyroptosis, apoptosis, and necroptosis (PANoptosis), however, were significantly resistant to B. thailandensis-induced cell death until later stages of infection. Consequently, PANoptosis-deficient BMDMs failed to limit B. thailandensis-induced cell-cell fusion, which permits increased intercellular spread and replication compared to wild-type or pyroptosis-deficient BMDMs. Respiratory B. thailandensis infection resulted in higher mortality in PANoptosis-deficient mice than in pyroptosis-deficient mice, indicating that, in the absence of pyroptosis, apoptosis is essential for efficient control of infection in vivo. Together, these findings suggest both pyroptosis and apoptosis are necessary for host-mediated control of Burkholderia infection. IMPORTANCEBurkholderia infections result in a high degree of mortality when left untreated; therefore, understanding the host immune response required to control infection is critical. In this study, we found a hierarchical cell death program utilized by infected cells to disrupt the intracellular niche of Burkholderia thailandensis, which limits bacterial intercellular spread, host cell-cell fusion, and bacterial replication. In macrophages, combined loss of key PANoptosis components results in extensive B. thailandensis infection-induced cell-cell fusion, bacterial replication, and increased cell death at later stages of infection compared with both wild-type (WT) and pyroptosis-deficient cells. During respiratory infection, mortality was increased in PANoptosis-deficient mice compared to pyroptosis-deficient mice, identifying an essential role for multiple cell death pathways in controlling B. thailandensis infection. These findings advance our understanding of the physiological role of programmed cell death in controlling Burkholderia infection.


Asunto(s)
Apoptosis/inmunología , Infecciones por Burkholderia/inmunología , Burkholderia/patogenicidad , Inmunidad Innata , Macrófagos/microbiología , Macrófagos/patología , Animales , Burkholderia/inmunología , Caspasas/clasificación , Caspasas/genética , Caspasas/inmunología , Femenino , Masculino , Ratones , Necroptosis/inmunología , Piroptosis/inmunología
7.
Biochem Soc Trans ; 49(3): 1311-1324, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34060593

RESUMEN

Innate immune responses are tightly regulated by various pathways to control infections and maintain homeostasis. One of these pathways, the inflammasome pathway, activates a family of cysteine proteases called inflammatory caspases. They orchestrate an immune response by cleaving specific cellular substrates. Canonical inflammasomes activate caspase-1, whereas non-canonical inflammasomes activate caspase-4 and -5 in humans and caspase-11 in mice. Caspases are highly specific enzymes that select their substrates through diverse mechanisms. During inflammation, caspase activity is responsible for the secretion of inflammatory cytokines and the execution of a form of lytic and inflammatory cell death called pyroptosis. This review aims to bring together our current knowledge of the biochemical processes behind inflammatory caspase activation, substrate specificity, and substrate signalling.


Asunto(s)
Caspasas/inmunología , Citocinas/inmunología , Inflamasomas/inmunología , Inflamación/inmunología , Transducción de Señal/inmunología , Animales , Caspasas/metabolismo , Citocinas/metabolismo , Activación Enzimática/inmunología , Humanos , Inflamasomas/metabolismo , Inflamación/metabolismo , Piroptosis/inmunología , Especificidad por Sustrato
8.
Viruses ; 12(11)2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33153187

RESUMEN

Progressive Multifocal Leukoencephalopathy (PML) is a fatal demyelinating disease of the CNS, resulting from the lytic infection of oligodendrocytes by the human neurotropic polyomavirus JC (JCPyV), typically associated with severe immunocompromised states and, in recent years, with the use of immunotherapies. Apoptosis is a homeostatic mechanism to dispose of senescent or damaged cells, including virally infected cells, triggered in the vast majority of viral infections of the brain. Previously, we showed upregulation of the normally dormant anti-apoptotic protein Survivin in cases of PML, which-in vitro-resulted in protection from apoptosis in JCPyV-infected primary cultures of astrocytes and oligodendrocytes. In the present study, we first demonstrate the absence of apoptotic DNA fragmentation and the lack of caspase activity in 16 cases of PML. We also identified the viral protein large T-Antigen as being responsible for the activation of the Survivin promoter. Chromatin Immunoprecipitation assay shows a direct binding between T-Antigen and the Survivin promoter DNA. Finally, we have identified the specific region of T-Antigen, spanning from amino acids 266 and 688, which binds to Survivin and translocates it to the nucleus, providing evidence of a mechanism that results in the efficient replication of JCPyV and a potential target for novel therapies.


Asunto(s)
Antígenos Virales de Tumores/genética , Apoptosis , Virus JC/genética , Regiones Promotoras Genéticas , Survivin/genética , Adulto , Anciano , Animales , Antígenos Virales de Tumores/inmunología , Astrocitos/virología , Caspasas/inmunología , Línea Celular Tumoral , Células Cultivadas , Niño , Fragmentación del ADN , Femenino , Humanos , Virus JC/inmunología , Virus JC/patogenicidad , Leucoencefalopatía Multifocal Progresiva , Masculino , Ratones , Persona de Mediana Edad , Oligodendroglía/virología , Adhesión en Parafina , Survivin/inmunología
9.
mBio ; 11(5)2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33024046

RESUMEN

Regulated macrophage death has emerged as an important mechanism to defend against intracellular pathogens. However, the importance and consequences of macrophage death during bacterial infection are poorly resolved. This is especially true for the recently described RIPK3-dependent lytic cell death, termed necroptosis. Salmonella enterica serovar Typhimurium is an intracellular pathogen that precisely regulates virulence expression within macrophages to evade and manipulate immune responses, which is a key factor in its ability to cause severe systemic infections. We combined genetic and pharmacological approaches to examine the importance of RIPK3 for S. Typhimurium-induced macrophage death using conditions that recapitulate bacterial gene expression during systemic infection in vivo Our findings indicate that noninvasive S. Typhimurium does not naturally induce macrophage necroptosis but does so in the presence of pan-caspase inhibition. Moreover, our data suggest that RIPK3 induction (following caspase inhibition) does not impact host survival following S. Typhimurium infection, which differs from previous findings based on inert lipopolysaccharide (LPS) injections. Finally, although necroptosis is typically characterized as highly inflammatory, our data suggest that RIPK3 skews the peritoneal myeloid population away from an inflammatory profile to that of a classically noninflammatory profile. Collectively, these data improve our understanding of S. Typhimurium-macrophage interactions, highlight the possibility that purified bacterial components may not accurately recapitulate the complexity of host-pathogen interactions, and reveal a potential and unexpected role for RIPK3 in resolving inflammation.IMPORTANCE Macrophages employ multiple strategies to limit pathogen infection. For example, macrophages may undergo regulated cell death, including RIPK3-dependent necroptosis, as a means of combatting intracellular bacterial pathogens. However, bacteria have evolved mechanisms to evade or exploit immune responses. Salmonella is an intracellular pathogen that avoids and manipulates immune detection within macrophages. We examined the contribution of RIPK3 to Salmonella-induced macrophage death. Our findings indicate that noninvasive Salmonella does not naturally induce necroptosis, but it does so when caspases are inhibited. Moreover, RIPK3 induction (following caspase inhibition) does not impact host survival following Salmonella systemic infection. Finally, our data show that RIPK3 induction results in recruitment of low-inflammatory myeloid cells, which was unexpected, as necroptosis is typically described as highly inflammatory. Collectively, these data improve our understanding of pathogen-macrophage interactions, including outcomes of regulated cell death during infection in vivo, and reveal a potential new role for RIPK3 in resolving inflammation.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Macrófagos/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Salmonelosis Animal/sangre , Animales , Inhibidores de Caspasas/farmacología , Caspasas/inmunología , Inflamasomas , Inflamación , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Necroptosis/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Salmonelosis Animal/microbiología , Salmonella typhimurium , Transducción de Señal
10.
J Fish Dis ; 43(4): 431-443, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32056240

RESUMEN

The caspase family proteins are aspartate-specific cysteine proteases that transmit extracellular signals to cells, ultimately cause apoptosis and therefore play a key role in cellular immunity. In this study, we cloned and characterized three caspases from Chinese black sleeper (Bostrychus sinensis), Bscasp-1, Bscasp-8 and Bscasp-9. Real-time PCR analysis showed that Bscasp-1, Bscasp-8 and Bscasp-9 were universally expressed in all tested tissues of B. sinensis. Expression analyses showed that after poly(I:C) stimulation and bacterial (Vibrio parahaemolyticus) infection, the three caspases were significantly upregulated. After poly(I:C) stimulation, the change of Bscasp-1 expression in the head kidney was the most obvious; peak expression was about 80.78-fold more than that of the control. In addition, the expression of Bscasp-8 and Bscasp-9 in the peripheral blood and liver was 167.99- and 17.98-fold higher than that in the control group, respectively. After V. parahaemolyticus infection, the expression peaks of Bscasp-1 and Bscasp-8 in the peripheral blood and spleen were 85.82-fold and 280.83-fold that of the control. However, the expression of Bscasp-9 in the peripheral blood was upregulated only 8.33-fold higher than that in the control group. These results indicate that Bscasp-1, Bscasp-8 and Bscasp-9 are likely involved in response to viral and bacterial infection.


Asunto(s)
Caspasas/genética , Caspasas/inmunología , Enfermedades de los Peces/inmunología , Peces/genética , Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Caspasas/química , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , Filogenia , Poli I-C/farmacología , Alineación de Secuencia/veterinaria , Vibriosis/inmunología , Vibriosis/veterinaria , Vibrio parahaemolyticus/fisiología
11.
PLoS Pathog ; 15(6): e1007872, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31194844

RESUMEN

Innate recognition of invading intracellular pathogens is essential for regulating robust and rapid CD4+ T cell effector function, which is critical for host-mediated immunity. The intracellular apicomplexan parasite, Toxoplasma gondii, is capable of infecting almost any nucleated cell of warm-blooded animals, including humans, and establishing tissue cysts that persist throughout the lifetime of the host. Recognition of T. gondii by TLRs is essential for robust IL-12 and IFN-γ production, two major cytokines involved in host resistance to the parasite. In the murine model of infection, robust IL-12 and IFN-γ production have been largely attributed to T. gondii profilin recognition by the TLR11 and TLR12 heterodimer complex, resulting in Myd88-dependent IL-12 production. However, TLR11 or TLR12 deficiency failed to recapitulate the acute susceptibility to T. gondii infection seen in Myd88-/- mice. T. gondii triggers inflammasome activation in a caspase-1-dependent manner resulting in cytokine release; however, it remains undetermined if parasite-mediated inflammasome activation impacts IFN-γ production and host resistance to the parasite. Using mice which lack different inflammasome components, we observed that the inflammasome played a limited role in host resistance when TLR11 remained functional. Strikingly, in the absence of TLR11, caspase-1 and -11 played a significant role for robust CD4+ TH1-derived IFN-γ responses and host survival. Moreover, we demonstrated that in the absence of TLR11, production of the caspase-1-dependent cytokine IL-18 was sufficient and necessary for CD4+ T cell-derived IFN-γ responses. Mechanistically, we established that T. gondii-mediated activation of the inflammasome and IL-18 were critical to maintain robust CD4+ TH1 IFN-γ responses during parasite infection in the absence of TLR11.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inmunidad Innata , Inflamasomas/inmunología , Interferón gamma/inmunología , Receptores Toll-Like/inmunología , Toxoplasma/inmunología , Toxoplasmosis Animal/inmunología , Animales , Linfocitos T CD4-Positivos/parasitología , Linfocitos T CD4-Positivos/patología , Caspasa 1/genética , Caspasa 1/inmunología , Caspasas/genética , Caspasas/inmunología , Caspasas Iniciadoras , Inflamasomas/genética , Interferón gamma/genética , Interleucina-18/genética , Interleucina-18/inmunología , Ratones , Ratones Noqueados , Receptores Toll-Like/genética , Toxoplasmosis Animal/genética , Toxoplasmosis Animal/patología
12.
Nat Immunol ; 20(5): 527-533, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30962589

RESUMEN

Monitoring of the cytosolic compartment by the innate immune system for pathogen-encoded products or pathogen activities often enables the activation of a subset of caspases. In most cases, the cytosolic surveillance pathways are coupled to activation of caspase-1 via canonical inflammasome complexes. A related set of caspases, caspase-11 in rodents and caspase-4 and caspase-5 in humans, monitors the cytosol for bacterial lipopolysaccharide (LPS). Direct activation of caspase-11, caspase-4 and caspase-5 by intracellular LPS elicits the lytic cell death called 'pyroptosis', which occurs in multiple cell types. The pyroptosis is executed by the pore-forming protein GSDMD, which is activated by cleavage mediated by caspase-11, caspase-4 or caspase-5. In monocytes, formation of GSDMD pores can induce activation of the NLRP3 inflammasome for maturation of the cytokines IL-1ß and IL-18. Caspase-11-mediated pyroptosis in response to cytosolic LPS is critical for antibacterial defense and septic shock. Here we review the emerging literature on the sensing of cytosolic LPS and its regulation and pathophysiological functions.


Asunto(s)
Caspasas/inmunología , Citosol/inmunología , Inmunidad Innata/inmunología , Lipopolisacáridos/inmunología , Animales , Caspasas/metabolismo , Citosol/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lipopolisacáridos/metabolismo , Modelos Inmunológicos , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , Proteínas de Unión a Fosfato , Piroptosis/inmunología
13.
PLoS One ; 14(4): e0214999, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30958862

RESUMEN

Processing of pro-interleukin (IL)-1ß and IL-18 is regulated by multiprotein complexes, known as inflammasomes. Inflammasome activation results in generation of bioactive IL-1ß and IL-18, which can exert potent pro-inflammatory effects. Our aim was to develop a whole blood-based assay to study the inflammasome in vitro and that also can be used as an assay in clinical studies. We show whole blood is a suitable milieu to study inflammasome activation in primary human monocytes. We demonstrated that unprocessed human blood cells can be stimulated to activate the inflammasome by the addition of adenosine 5'-triphosphate (ATP) within a narrow timeframe following lipopolysaccharide (LPS) priming. Stimulation with LPS resulted in IL-1ß release; however, addition of ATP is necessary for "full-blown" inflammasome stimulation resulting in high IL-1ß and IL-18 release. Intracellular cytokine staining demonstrated monocytes are the major producers of IL-1ß in human whole blood cultures, and this was associated with activation of caspase-1/4/5, as detected by a fluorescently labelled caspase-1/4/5 probe. By applying caspase inhibitors, we show that both the canonical inflammasome pathway (via caspase-1) as well as the non-canonical inflammasome pathway (via caspases-4 and 5) can be studied using this whole blood-based model.


Asunto(s)
Caspasas/inmunología , Inflamasomas/inmunología , Interleucina-1beta/inmunología , Modelos Inmunológicos , Monocitos/inmunología , Transducción de Señal/inmunología , Humanos , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Interleucina-18/inmunología , Lipopolisacáridos/toxicidad , Monocitos/patología , Transducción de Señal/efectos de los fármacos
14.
Nat Immunol ; 20(3): 276-287, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30692621

RESUMEN

Inflammatory caspases (caspase-1, caspase-4, caspase-5 and caspase-11 (caspase-1/-4/-5/-11)) mediate host defense against microbial infections, processing pro-inflammatory cytokines and triggering pyroptosis. However, precise checkpoints are required to prevent their unsolicited activation. Here we report that serpin family B member 1 (SERPINB1) limited the activity of those caspases by suppressing their caspase-recruitment domain (CARD) oligomerization and enzymatic activation. While the reactive center loop of SERPINB1 inhibits neutrophil serine proteases, its carboxy-terminal CARD-binding motif restrained the activation of pro-caspase-1/-4/-5/-11. Consequently, knockdown or deletion of SERPINB1 prompted spontaneous activation of caspase-1/-4/-5/-11, release of the cytokine IL-1ß and pyroptosis, inducing elevated inflammation after non-hygienic co-housing with pet-store mice and enhanced sensitivity to lipopolysaccharide- or Acinetobacter baumannii-induced endotoxemia. Our results reveal that SERPINB1 acts as a vital gatekeeper of inflammation by restraining neutrophil serine proteases and inflammatory caspases in a genetically and functionally separable manner.


Asunto(s)
Caspasas/inmunología , Mediadores de Inflamación/inmunología , Inflamación/inmunología , Serpinas/inmunología , Animales , Caspasas/genética , Caspasas/metabolismo , Línea Celular , Células Cultivadas , Activación Enzimática/inmunología , Células HEK293 , Humanos , Inflamación/genética , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/enzimología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Piroptosis/efectos de los fármacos , Piroptosis/inmunología , Células RAW 264.7 , Interferencia de ARN , Serina Proteasas/inmunología , Serina Proteasas/metabolismo , Serpinas/genética , Serpinas/metabolismo , Células THP-1 , Células U937
15.
Mol Med ; 24(1): 66, 2018 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-30587103

RESUMEN

BACKGROUND: Caspase-11, a cytosolic receptor of bacterial endotoxin (lipopolysaccharide: LPS), mediates immune responses and lethality in endotoxemia and experimental sepsis. However, the upstream pathways that regulate caspase-11 activation in endotoxemia and sepsis are not fully understood. The aim of this study is to test whether TIR-domain-containing adapter-inducing interferon-ß (TRIF) signaling is critical for caspase-11-dependent immune responses and lethality in endotoxemia. METHODS: Mice of indicated genotypes were subjected to endotoxemia or cecum ligation and puncture (CLP) and monitored daily by signs of a moribund state for lethality. Serum interleukin (IL)-1α, IL-1ß, IL-6 and tumor necrosis factor (TNF) were measured by ELISA. Data were analyzed by using student's t-test or one-way ANOVA followed by post-hoc Bonferroni test. Survival data were analyzed by using the log-rank test. RESULTS: Blockade of type 1 interferon signaling or genetic deletion of TRIF or guanylate-binding proteins (GBPs) prevented caspase-11-dependent immune responses, organ injury and lethality in endotoxemia and experimental sepsis. In vitro, deletion of GBPs blocked cytosolic LPS-induced caspase-11 activation in mouse macrophages. CONCLUSIONS: These findings demonstrate that TRIF signaling is required for caspase-11-dependent immune responses and lethality in endotoxemia and sepsis, and provide novel mechanistic insights into how LPS induces caspase-11 activation during bacterial infection.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/inmunología , Caspasas/inmunología , Endotoxemia/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Caspasas Iniciadoras , Endotoxemia/inducido químicamente , Femenino , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/inmunología , Interferón Tipo I/inmunología , Lipopolisacáridos , Macrófagos Peritoneales/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Cell Death Dis ; 9(12): 1182, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30518854

RESUMEN

Klebsiella pneumoniae is a Gram-negative bacterium responsible for severe cases of nosocomial pneumonia. During the infectious process, both neutrophils and monocytes migrate to the site of infection, where they carry out their effector functions and can be affected by different patterns of cell death. Our data show that clinical strains of K. pneumoniae have dissimilar mechanisms for surviving within macrophages; these mechanisms include modulation of microbicidal mediators and cell death. The A28006 strain induced high IL-1ß production and pyroptotic cell death in macrophages; by contrast, the A54970 strain induced high IL-10 production and low IL-1ß production by macrophages. Pyroptotic cell death induced by the A28006 strain leads to a significant increase in bacterial sensitivity to hydrogen peroxide, and efferocytosis of the pyroptotic cells results in efficient bacterial clearance both in vitro and in vivo. In addition, the A54970 strain was able to inhibit inflammasome activation and pyroptotic cell death by inducing IL-10 production. Here, for the first time, we present a K. pneumoniae strain able to inhibit inflammasome activation, leading to bacterial survival and dissemination in the host. The understanding of possible escape mechanisms is essential in the search for alternative treatments against multidrug-resistant bacteria.


Asunto(s)
Bacteriemia/microbiología , Interacciones Huésped-Patógeno/inmunología , Inflamasomas/inmunología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/patogenicidad , Piroptosis/inmunología , Animales , Bacteriemia/genética , Bacteriemia/inmunología , Bacteriemia/patología , Caspasa 1/deficiencia , Caspasa 1/genética , Caspasa 1/inmunología , Caspasas/deficiencia , Caspasas/genética , Caspasas/inmunología , Caspasas Iniciadoras , Femenino , Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Inflamasomas/genética , Interleucina-10/deficiencia , Interleucina-10/genética , Interleucina-10/inmunología , Infecciones por Klebsiella/genética , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/patología , Klebsiella pneumoniae/inmunología , Klebsiella pneumoniae/aislamiento & purificación , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/microbiología , Neutrófilos/inmunología , Neutrófilos/microbiología , Fagocitosis/genética , Piroptosis/genética
17.
Elife ; 72018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30526845

RESUMEN

Intracellular lipopolysaccharide (LPS) triggers the non-canonical inflammasome pathway, resulting in pyroptosis of innate immune cells. In addition to its well-known proinflammatory role, LPS can directly cause regression of some tumors, although the underlying mechanism has remained unknown. Here we show that secretoglobin(SCGB)3A2, a small protein predominantly secreted in airways, chaperones LPS to the cytosol through the cell surface receptor syndecan-1; this leads to pyroptotic cell death driven by caspase-11. SCGB3A2 and LPS co-treatment significantly induced pyroptosis of macrophage RAW264.7 cells and decreased cancer cell proliferation in vitro, while SCGB3A2 treatment resulted in reduced progression of xenograft tumors in mice. These data suggest a conserved function for SCGB3A2 in the innate immune system and cancer cells. These findings demonstrate a critical role for SCGB3A2 as an LPS delivery vehicle; they reveal one mechanism whereby LPS enters innate immune cells leading to pyroptosis, and they clarify the direct effect of LPS on cancer cells.


Asunto(s)
Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , Lipopolisacáridos/farmacología , Melanoma Experimental/tratamiento farmacológico , Secretoglobinas/genética , Sindecano-1/genética , Animales , Transporte Biológico , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/mortalidad , Caspasas/genética , Caspasas/inmunología , Caspasas Iniciadoras , Línea Celular Tumoral , Humanos , Inmunidad Innata , Metástasis Linfática , Masculino , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/mortalidad , Ratones , Ratones Transgénicos , Análisis por Matrices de Proteínas , Piroptosis/efectos de los fármacos , Piroptosis/genética , Piroptosis/inmunología , Células RAW 264.7 , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Secretoglobinas/antagonistas & inhibidores , Secretoglobinas/inmunología , Transducción de Señal , Análisis de Supervivencia , Sindecano-1/antagonistas & inhibidores , Sindecano-1/inmunología , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cell Death Dis ; 9(11): 1053, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30323167

RESUMEN

Activation of T cells, a major fraction of peripheral blood lymphocytes (PBLCS), is essential for the immune response. Genotoxic stress resulting from ionizing radiation (IR) and chemical agents, including anticancer drugs, has serious impact on T cells and, therefore, on the immune status. Here we compared the sensitivity of non-stimulated (non-proliferating) vs. CD3/CD28-stimulated (proliferating) PBLC to IR. PBLCs were highly sensitive to IR and, surprisingly, stimulation to proliferation resulted in resistance to IR. Radioprotection following CD3/CD28 activation was observed in different T-cell subsets, whereas stimulated CD34+ progenitor cells did not become resistant to IR. Following stimulation, PBLCs showed no significant differences in the repair of IR-induced DNA damage compared with unstimulated cells. Interestingly, ATM is expressed at high level in resting PBLCs and CD3/CD28 stimulation leads to transcriptional downregulation and reduced ATM phosphorylation following IR, indicating ATM to be key regulator of the high radiosensitivity of resting PBLCs. In line with this, pharmacological inhibition of ATM caused radioresistance of unstimulated, but not stimulated, PBLCs. Radioprotection was also achieved by inhibition of MRE11 and CHK1/CHK2, supporting the notion that downregulation of the MRN-ATM-CHK pathway following CD3/CD28 activation results in radioprotection of proliferating PBLCs. Interestingly, the crosslinking anticancer drug mafosfamide induced, like IR, more death in unstimulated than in stimulated PBLCs. In contrast, the bacterial toxin CDT, damaging DNA through inherent DNase activity, and the DNA methylating anticancer drug temozolomide induced more death in CD3/CD28-stimulated than in unstimulated PBLCs. Thus, the sensitivity of stimulated vs. non-stimulated lymphocytes to genotoxins strongly depends on the kind of DNA damage induced. This is the first study in which the killing response of non-proliferating vs. proliferating T cells was comparatively determined. The data provide insights on how immunotherapeutic strategies resting on T-cell activation can be impacted by differential cytotoxic effects resulting from radiation and chemotherapy.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/inmunología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Rayos gamma , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/inmunología , Clorometilcetonas de Aminoácidos/farmacología , Anticuerpos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/genética , Antígenos CD28/antagonistas & inhibidores , Antígenos CD28/genética , Complejo CD3/antagonistas & inhibidores , Complejo CD3/genética , Caspasas/genética , Caspasas/inmunología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Cromonas/farmacología , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/inmunología , Resistencia a Medicamentos/genética , Resistencia a Medicamentos/inmunología , Regulación de la Expresión Génica , Humanos , Isoxazoles/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/efectos de la radiación , Proteína Homóloga de MRE11/antagonistas & inhibidores , Proteína Homóloga de MRE11/genética , Proteína Homóloga de MRE11/inmunología , Morfolinas/farmacología , Cultivo Primario de Células , Pirazinas/farmacología , Pironas/farmacología , Tolerancia a Radiación/genética , Tolerancia a Radiación/inmunología , Transducción de Señal , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/efectos de la radiación , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/efectos de la radiación , Tiofenos/farmacología , Tioxantenos/farmacología
19.
Immunity ; 49(4): 740-753.e7, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30314759

RESUMEN

Caspase-11, a cytosolic endotoxin (lipopolysaccharide: LPS) receptor, mediates pyroptosis, a lytic form of cell death. Caspase-11-dependent pyroptosis mediates lethality in endotoxemia, but it is unclear how LPS is delivered into the cytosol for the activation of caspase-11. Here we discovered that hepatocyte-released high mobility group box 1 (HMGB1) was required for caspase-11-dependent pyroptosis and lethality in endotoxemia and bacterial sepsis. Mechanistically, hepatocyte-released HMGB1 bound LPS and targeted its internalization into the lysosomes of macrophages and endothelial cells via the receptor for advanced glycation end-products (RAGE). Subsequently, HMGB1 permeabilized the phospholipid bilayer in the acidic environment of lysosomes. This resulted in LPS leakage into the cytosol and caspase-11 activation. Depletion of hepatocyte HMGB1, inhibition of hepatocyte HMGB1 release, neutralizing extracellular HMGB1, or RAGE deficiency prevented caspase-11-dependent pyroptosis and death in endotoxemia and bacterial sepsis. These findings indicate that HMGB1 interacts with LPS to mediate caspase-11-dependent pyroptosis in lethal sepsis.


Asunto(s)
Caspasas/inmunología , Endotoxinas/inmunología , Proteína HMGB1/inmunología , Piroptosis/inmunología , Sepsis/inmunología , Animales , Caspasas/genética , Caspasas/metabolismo , Células Cultivadas , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Endotoxinas/metabolismo , Células HEK293 , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor para Productos Finales de Glicación Avanzada/inmunología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Sepsis/genética , Sepsis/metabolismo , Células THP-1
20.
Fish Shellfish Immunol ; 83: 76-83, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30195917

RESUMEN

Caspases are a conserved family of cysteine proteases characterized by specificity for aspartic acid and play an essential role in cell apoptosis. In the present study, a novel effector caspase (designated as EsCaspase-3/7-1) was identified from Chinese mitten crab Eriocheir sinensis. The open reading frame of EsCaspase-3/7-1 cDNA was of 972 bp, encoding a polypeptide of 323 amino acids. EsCaspase-3/7-1 contained an N-terminal prodomain and a conservative C-terminal CASc domain, with the conserved active site "QACRG". The mRNA transcripts of EsCaspase-3/7-1 were constitutively expressed in all the examined tissues with high expression level in hemocytes, hepatopancreas and gill. The EsCaspase-3/7-1 protein was mainly distributed in the cytoplasm of hemocytes. After Aeromonas hydrophila and lipopolysaccharide (LPS) stimulations, the mRNA expression level of EsCaspase-3/7-1 in hemocytes increased significantly. The mRNA expression level of EsCaspase-3/7-1 in hemocytes was significantly up-regulated after H2O2 treatment in vitro. The recombinant EsCaspase-3/7-1 protein (rEsCaspase-3/7-1) was capable of hydrolyzing the substrate Ac-DEVD-pNA rather than Ac-YVAD-pNA and Ac-VEID-pNA in vitro, and exhibited binding activity to LPS. These results demonstrated that EsCaspase-3/7-1 might act as an LPS receptor, and play an important role in the regulation of immune homeostasis of E. sinensis.


Asunto(s)
Proteínas de Artrópodos/genética , Braquiuros/inmunología , Caspasas/genética , Homeostasis/inmunología , Aeromonas hydrophila , Animales , Apoptosis/efectos de los fármacos , Proteínas de Artrópodos/inmunología , Braquiuros/genética , Caspasas/inmunología , Clonación Molecular , ADN Complementario , Regulación de la Expresión Génica , Hemocitos/metabolismo , Hepatopáncreas/metabolismo , Peróxido de Hidrógeno/farmacología , Inmunidad Innata , Lipopolisacáridos , Filogenia , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA