Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; : 107386, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38763335

RESUMEN

Inflammasomes serve as critical sensors for disruptions to cellular homeostasis, with inflammasome assembly leading to inflammatory caspase activation, gasdermin cleavage, and cytokine release. While the canonical pathways leading to priming, assembly, and pyroptosis are well characterized, recent work has begun to focus on the role of post-translational modifications (PTMs) in regulating inflammasome activity. A diverse array of PTMs, including phosphorylation, ubiquitination, SUMOylation, acetylation, and glycosylation, exert both activating and inhibitory influences on members of the inflammasome cascade through effects on protein-protein interactions, stability, and localization. Dysregulation of inflammasome activation is associated with a number of inflammatory diseases, and evidence is emerging that aberrant modification of inflammasome components contributes to this dysregulation. This review provides insight into PTMs within the NLRP3 inflammasome pathway and their functional consequences on the signaling cascade, and highlights outstanding questions that remain regarding the complex web of signals at play.

2.
Nat Commun ; 14(1): 5832, 2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37730693

RESUMEN

Macrophages infected with Gram-negative bacteria expressing Type III secretion system (T3SS) activate the NLRC4 inflammasome, resulting in Gasdermin D (GSDMD)-dependent, but GSDME independent IL-1ß secretion and pyroptosis. Here we examine inflammasome signaling in neutrophils infected with Pseudomonas aeruginosa strain PAO1 that expresses the T3SS effectors ExoS and ExoT. IL-1ß secretion by neutrophils requires the T3SS needle and translocon proteins and GSDMD. In macrophages, PAO1 and mutants lacking ExoS and ExoT (ΔexoST) require NLRC4 for IL-1ß secretion. While IL-1ß release from ΔexoST infected neutrophils is also NLRC4-dependent, infection with PAO1 is instead NLRP3-dependent and driven by the ADP ribosyl transferase activity of ExoS. Genetic and pharmacologic approaches using MCC950 reveal that NLRP3 is also essential for bacterial killing and disease severity in a murine model of P. aeruginosa corneal infection (keratitis). Overall, these findings reveal a function for ExoS ADPRT in regulating inflammasome subtype usage in neutrophils versus macrophages and an unexpected role for NLRP3 in P. aeruginosa keratitis.


Asunto(s)
Enfermedades de la Córnea , Pseudomonas aeruginosa , Animales , Ratones , Inflamasomas , Neutrófilos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Gravedad del Paciente
3.
Immunol Rev ; 314(1): 229-249, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36656082

RESUMEN

Pyroptosis is a proinflammatory mode of lytic cell death mediated by accumulation of plasma membrane (PM) macropores composed of gasdermin-family (GSDM) proteins. It facilitates two major functions in innate immunity: (i) elimination of intracellular replicative niches for pathogenic bacteria; and (ii) non-classical secretion of IL-1 family cytokines that amplify host-beneficial inflammatory responses to microbial infection or tissue damage. Physiological roles for gasdermin D (GSDMD) in pyroptosis and IL-1ß release during inflammasome signaling have been extensively characterized in macrophages. This involves cleavage of GSDMD by caspase-1 to generate GSDMD macropores that mediate IL-1ß efflux and progression to pyroptotic lysis. Neutrophils, which rapidly accumulate in large numbers at sites of tissue infection or damage, become the predominant local source of IL-1ß in coordination with their potent microbiocidal capacity. Similar to macrophages, neutrophils express GSDMD and utilize the same spectrum of diverse inflammasome platforms for caspase-1-mediated cleavage of GSDMD. Distinct from macrophages, neutrophils possess a remarkable capacity to resist progression to GSDMD-dependent pyroptotic lysis to preserve their viability for efficient microbial killing while maintaining GSDMD-dependent mechanisms for export of bioactive IL-1ß. Rather, neutrophils employ cell-specific mechanisms to conditionally engage GSDMD-mediated pyroptosis in response to bacterial pathogens that use neutrophils as replicative niches. GSDMD and pyroptosis have also been mechanistically linked to induction of NETosis, a signature neutrophil pathway that expels decondensed nuclear DNA into extracellular compartments for immobilization and killing of microbial pathogens. This review summarizes a rapidly growing number of recent studies that have produced new insights, unexpected mechanistic nuances, and some controversies regarding the regulation of, and roles for, neutrophil inflammasomes, pyroptosis, and GSDMs in diverse innate immune responses.


Asunto(s)
Inflamasomas , Piroptosis , Humanos , Piroptosis/fisiología , Inflamasomas/metabolismo , Neutrófilos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Gasderminas , Caspasa 1/metabolismo , Transducción de Señal
4.
J Biol Chem ; 299(2): 102908, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36642180

RESUMEN

When activated, gasdermin family members are thought to be pore-forming proteins that cause lytic cell death. Despite this, numerous studies have suggested that the threshold for lytic cell death is dependent on which gasdermin family member is activated. Determination of the propensity of various gasdermin family members to cause pyroptosis has been handicapped by the fact that for many of them, the mechanisms and timing of their activation are uncertain. In this article, we exploit the recently discovered exosite-mediated recognition of gasdermin D (GSDMD) by the inflammatory caspases to develop a system that activates gasdermin family members in an efficient and equivalent manner. We leverage this system to show that upon activation, GSDMD and gasdermin A (GSDMA) exhibit differential subcellular localization, differential plasma membrane permeabilization, and differential lytic cell death. While GSDMD localizes rapidly to both the plasma membrane and organelle membranes, GSDMA preferentially localizes to the mitochondria with delayed and diminished accumulation at the plasma membrane. As a consequence of this differential kinetics of subcellular localization, N-terminal GSDMA results in early mitochondrial dysfunction relative to plasma membrane permeabilization. This study thus challenges the assumption that gasdermin family members effect cell death through identical mechanisms and establishes that their activation in their respective tissues of expression likely results in different immunological outcomes.


Asunto(s)
Gasderminas , Piroptosis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Membrana Celular/metabolismo , Inflamasomas/metabolismo , Ingeniería de Proteínas
5.
Sci Signal ; 15(765): eabl6781, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36538593

RESUMEN

Pyroptosis is a mechanism of programmed, necrotic cell death mediated by gasdermins, a family of pore-forming proteins. Caspase-1 activates gasdermin D (GSDMD) under inflammatory conditions, whereas caspase-3 activates GSDME under apoptotic conditions, such as those induced by chemotherapy. These pathways are thought to be separate. However, we found that they are part of an integrated network of gatekeepers that enables pyroptotic cell death. We observed that GSDMD was the primary pyroptotic mediator in cultured blood cells in response to doxorubicin and etoposide, two common chemotherapies for hematopoietic malignancies. Upon treatment, the channel protein pannexin-1 (PANX1), which is stimulated by the initiation of apoptosis, increased membrane permeability to induce K+ efflux-driven activation of the NLRP3 inflammasome and GSDMD. However, either PANX1 or GSDME could also be the primary mediator of chemotherapy-induced pyroptosis when present at higher amounts. The most abundant pore-forming protein in acute myeloid leukemias from patients predicted the cell death pathway in response to chemotherapy. This interconnected network, a multistep switch that converts apoptosis to pyroptosis, could be clinically titratated to modulate cell death with regard to antitumor immunity or tumor lysis syndrome in patients.


Asunto(s)
Antineoplásicos , Neoplasias Hematológicas , Humanos , Gasderminas , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Apoptosis , Necrosis , Inflamasomas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Conexinas/genética , Conexinas/metabolismo
6.
J Orthop Res ; 39(3): 493-505, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32779803

RESUMEN

Wear particles from orthopedic implants cause aseptic loosening, the leading cause of implant revisions. The particles are phagocytosed by macrophages leading to activation of the nod-like receptor protein 3 (NLRP3) inflammasome and release of interleukin-1ß (IL-1ß) which then contributes to osteoclast differentiation and implant loosening. The mechanism of inflammasome activation by orthopedic particles is undetermined but other particles cause the cytosolic accumulation of the lysosomal cathepsin-family proteases which can activate the NLRP3 inflammasome. Here, we demonstrate that lysosome membrane disruption causes cathepsin release into the cytoplasm that drives both inflammasome activation and cell death but that these processes occur independently. Using wild-type and genetically-manipulated immortalized murine bone marrow derived macrophages and pharmacologic inhibitors, we found that NLRP3 and gasdermin D are required for particle-induced IL-1ß release but not for particle-induced cell death. In contrast, phagocytosis and lysosomal cathepsin release are critical for both IL-1ß release and cell death. Collectively, our findings identify the pan-cathepsin inhibitor Ca-074Me and the NLRP3 inflammasome inhibitor MCC950 as therapeutic interventions worth exploring in aseptic loosening of orthopedic implants. We also found that particle-induced activation of the NLRP3 inflammasome in pre-primed macrophages and cell death are not dependent on pathogen-associated molecular patterns adherent to the wear particles despite such pathogen-associated molecular patterns being critical for all other previously studied wear particle responses, including priming of the NLRP3 inflammasome.


Asunto(s)
Catepsinas/metabolismo , Lisosomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fagocitosis , Falla de Prótesis/etiología , Muerte Celular , Humanos , Interleucina-1beta/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos , Titanio
7.
J Clin Invest ; 130(8): 4218-4234, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32597834

RESUMEN

Gasdermin D (GSDMD) induces pyroptosis via the pore-forming activity of its N-terminal domain, cleaved by activated caspases associated with the release of IL-1ß. Here, we report a nonpyroptotic role of full-length GSDMD in guiding the release of IL-1ß-containing small extracellular vesicles (sEVs) from intestinal epithelial cells (IECs). In response to caspase-8 inflammasome activation, GSDMD, chaperoned by Cdc37/Hsp90, recruits the E3 ligase, NEDD4, to catalyze polyubiquitination of pro-IL-1ß, serving as a signal for cargo loading into secretory vesicles. GSDMD and IL-1ß colocalize with the exosome markers CD63 and ALIX intracellularly, and GSDMD and NEDD4 are required for release of CD63+ sEVs containing IL-1ß, GSDMD, NEDD4, and caspase-8. Importantly, increased expression of epithelial-derived GSDMD is observed both in patients with inflammatory bowel disease (IBD) and those with experimental colitis. While GSDMD-dependent release of IL-1ß-containing sEVs is detected in cultured colonic explants from colitic mice, GSDMD deficiency substantially attenuates disease severity, implicating GSDMD-mediated release of IL-1ß sEVs in the pathogenesis of intestinal inflammation, such as that observed in IBD.


Asunto(s)
Colitis/metabolismo , Células Epiteliales/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Interleucina-1beta/metabolismo , Mucosa Intestinal/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Animales , Línea Celular , Colitis/genética , Colitis/patología , Células Epiteliales/patología , Exosomas/genética , Exosomas/metabolismo , Exosomas/patología , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Interleucina-1beta/genética , Mucosa Intestinal/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Ubiquitina-Proteína Ligasas Nedd4/genética , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Proteínas de Unión a Fosfato/genética , Tetraspanina 30/genética , Tetraspanina 30/metabolismo
8.
Nat Commun ; 11(1): 2212, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32371889

RESUMEN

Gasdermin-D (GSDMD) in inflammasome-activated macrophages is cleaved by caspase-1 to generate N-GSDMD fragments. N-GSDMD then oligomerizes in the plasma membrane (PM) to form pores that increase membrane permeability, leading to pyroptosis and IL-1ß release. In contrast, we report that although N-GSDMD is required for IL-1ß secretion in NLRP3-activated human and murine neutrophils, N-GSDMD does not localize to the PM or increase PM permeability or pyroptosis. Instead, biochemical and microscopy studies reveal that N-GSDMD in neutrophils predominantly associates with azurophilic granules and LC3+ autophagosomes. N-GSDMD trafficking to azurophilic granules causes leakage of neutrophil elastase into the cytosol, resulting in secondary cleavage of GSDMD to an alternatively cleaved N-GSDMD product. Genetic analyses using ATG7-deficient cells indicate that neutrophils secrete IL-1ß via an autophagy-dependent mechanism. These findings reveal fundamental differences in GSDMD trafficking between neutrophils and macrophages that underlie neutrophil-specific functions during inflammasome activation.


Asunto(s)
Membrana Celular/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neutrófilos/metabolismo , Orgánulos/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Animales , Autofagosomas/metabolismo , Autofagia/genética , Caspasa 1/metabolismo , Permeabilidad de la Membrana Celular/genética , Humanos , Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Elastasa de Leucocito/genética , Elastasa de Leucocito/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión a Fosfato/genética , Transporte de Proteínas , Piroptosis/genética
9.
Methods Enzymol ; 629: 81-102, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31727258

RESUMEN

The efficacy of cancer chemotherapy is enhanced by induction of sustainable anti-tumor immune responses. Such responses involve accumulation of immunogenic mediators, such as extracellular ATP and ATP metabolites, within the tumor microenvironment. Recent studies have identified nucleotide-permeable plasma membrane channels or pores that are activated as early downstream consequences of different regulated cell death pathways: pannexin-1 channels in apoptosis, MLKL pores in necroptosis, and gasdermin-family pores in pyroptosis. This chapter describes the use of highly quantitative and semi-high-throughput methods based on the ATP sensor luciferase to measure dynamic changes in extracellular ATP, ADP, and AMP in tissue/cell culture models of cancer cells during various modes of regulated cell death in response to chemotherapeutic drugs, death receptors, or metabolic perturbation.


Asunto(s)
Adenosina Trifosfato/análisis , Luciferasas/química , Neoplasias/tratamiento farmacológico , Adenosina Difosfato/análisis , Adenosina Difosfato/inmunología , Adenosina Difosfato/metabolismo , Adenosina Monofosfato/análisis , Adenosina Monofosfato/inmunología , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/inmunología , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/inmunología , Espacio Extracelular/inmunología , Espacio Extracelular/metabolismo , Humanos , Muerte Celular Inmunogénica/efectos de los fármacos , Proteínas de Transporte de Membrana/metabolismo , Ratones , Neoplasias/inmunología , Neoplasias/patología , Cultivo Primario de Células , Piroptosis/efectos de los fármacos , Piroptosis/inmunología , Ratas
10.
J Leukoc Biol ; 106(6): 1197-1200, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31633233

RESUMEN

Discussion on LPS disruption of mitochondrial localization and autocrine purinergic signaling in neutrophil chemotaxis for control of E. coli infection.


Asunto(s)
Antiinfecciosos , Neutrófilos , Quimiotaxis , Comunicación , Escherichia coli , Lipopolisacáridos
11.
Infect Immun ; 87(8)2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31109947

RESUMEN

Little is known about whether pathogen invasion of neural tissue is affected by immune-based mechanisms in endothelial cells. We examined the effects of endothelial cell CD40 on Toxoplasma gondii invasion of the retina and brain, organs seeded hematogenously. T. gondii circulates in the bloodstream within infected leukocytes (including monocytes and dendritic cells) and as extracellular tachyzoites. After T. gondii infection, mice that expressed CD40 restricted to endothelial cells exhibited diminished parasite loads and histopathology in the retina and brain. These mice also had lower parasite loads in the retina and brain after intravenous (i.v.) injection of infected monocytes or dendritic cells. The protective effect of endothelial cell CD40 was not explained by changes in cellular or humoral immunity, reduced transmigration of leukocytes into neural tissue, or reduced invasion by extracellular parasites. Circulating T. gondii-infected leukocytes (dendritic cells used as a model) led to infection of neural endothelial cells. The number of foci of infection in these cells were reduced if endothelial cells expressed CD40. Infected dendritic cells and macrophages expressed membrane-associated inducible Hsp70. Infected leukocytes triggered Hsp70-dependent autophagy in CD40+ endothelial cells and anti-T. gondii activity dependent on ULK1 and beclin 1. Reduced parasite load in the retina and brain not only required CD40 expression in endothelial cells but was also dependent on beclin 1 and the expression of inducible Hsp70 in dendritic cells. These studies suggest that during endothelial cell-leukocyte interaction, CD40 restricts T. gondii invasion of neural tissue through a mechanism that appears mediated by endothelial cell anti-parasitic activity stimulated by Hsp70.


Asunto(s)
Encéfalo/parasitología , Antígenos CD40/fisiología , Células Endoteliales/inmunología , Retina/parasitología , Toxoplasma/patogenicidad , Animales , Autofagia , Movimiento Celular , Proteínas HSP70 de Choque Térmico/fisiología , Leucocitos/fisiología , Ratones , Ratones Endogámicos C57BL
12.
Clin Orthop Relat Res ; 476(12): 2442-2453, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30427314

RESUMEN

BACKGROUND: Orthopaedic wear particles activate the NLRP3 inflammasome to produce active interleukin 1ß (IL1ß). However, the NLRP3 inflammasome must be primed before it can be activated, and it is unknown whether wear particles induce priming. Toll-like receptors (TLRs) are thought to mediate particle bioactivity. It remains controversial whether pathogen-associated molecular patterns (PAMPs) and/or alarmins are responsible for TLR activation by wear particles. QUESTIONS/PURPOSES: (1) Does priming of the NLRP3 inflammasome by wear particles depend on adherent PAMPs? (2) Does priming of the NLRP3 inflammasome by wear particles depend on TLRs and TIRAP/Mal? (3) Does priming of the NLRP3 inflammasome by wear particles depend on cognate TLRs? (4) Does activation of the NLRP3 inflammasome by wear particles depend on adherent PAMPs? METHODS: Immortalized murine macrophages were stimulated by as-received titanium particles with adherent bacterial debris, endotoxin-free titanium particles, or titanium particles with adherent ultrapure lipopolysaccharide. To study priming, NLRP3 and IL1ß mRNA and IL1ß protein levels were assessed in wild-type, TLR4, TLR2, and TIRAP/Mal macrophages. To study activation, IL1ß protein secretion was assessed in wild-type macrophages preprimed with ultrapure lipopolysaccharide. RESULTS: Compared with titanium particles with adherent bacterial debris, endotoxin-free titanium particles induced 86% less NLRP3 mRNA (0.05 ± 0.03 versus 0.35 ± 0.01 NLRP3/GAPDH, p < 0.001) and 91% less IL1ß mRNA (0.02 ± 0.01 versus 0.22 ± 0.03 IL1ß/GAPDH, p < 0.001). ProIL1ß protein level was robustly increased in wild-type macrophages stimulated by particles with adherent PAMPs but was not detectably produced in macrophages stimulated by endotoxin-free particles. Adherence of ultrapure lipopolysaccharide to endotoxin-free particles reconstituted stimulation of NLRP3 and IL1ß mRNA. Particles with adherent bacterial debris induced 79% less NLRP3 mRNA (0.09 ± 0.004 versus 0.43 ± 0.13 NLRP3/GAPDH, p < 0.001) and 40% less IL1ß mRNA (0.09 ± 0.04 versus 0.15 ± 0.03 IL1ß/GAPDH, p = 0.005) in TLR4 macrophages than in wild-type. Similarly, those particles induced 49% less NLRP3 mRNA (0.22 ± 0.10 versus 0.43 ± 0.13 NLRP3/GAPDH, p = 0.004) and 47% less IL1ß mRNA (0.08 ± 0.02 versus 0.15 ± 0.03 IL1ß/GAPDH, p = 0.012) in TIRAP/Mal macrophages than in wild-type. Particles with adherent ultrapure lipopolysaccharide induced 96% less NLRP3 mRNA (0.012 ± 0.001 versus 0.27 ± 0.05 NLRP3/GAPDH, p = 0.003) and 91% less IL1ß mRNA (0.03 ± 0.01 versus 0.34 ± 0.07 IL1ß/GAPDH, p < 0.001) expression in TLR4 macrophages than in wild-type. In contrast, those particles did not induce less NLRP3 and IL1ß mRNA in TLR2 macrophages. IL1ß protein secretion was equivalently induced by particles with adherent bacterial debris or by endotoxin-free particles in a time-dependent manner in wild-type macrophages. For example, particles with adherent bacterial debris induced 99% ± 2% of maximal IL1ß secretion after 12 hours, whereas endotoxin-free particles induced 92% ± 11% (p > 0.5). CONCLUSIONS: This cell culture study showed that adherent PAMPs are required for priming of the NLRP3 inflammasome by wear particles and this process is dependent on their cognate TLRs and TIRAP/Mal. In contrast, activation of the NLRP3 inflammasome by titanium particles is not dependent on adherent PAMPs. Animal and implant retrieval studies are needed to determine whether wear particles have similar effects on the NLRP3 inflammasome in vivo. CLINICAL RELEVANCE: Our findings, together with recent findings that aseptic loosening associates with polymorphisms in the TIRAP/Mal locus, support that adherent PAMPs may contribute to aseptic loosening in patients undergoing arthroplasty.


Asunto(s)
Reactividad Cruzada/efectos de los fármacos , Macrófagos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Titanio/farmacología , Receptores Toll-Like/metabolismo , Animales , Interleucina-1beta/metabolismo , Ratones
13.
Sci Immunol ; 3(26)2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-30143556

RESUMEN

Dysregulation of inflammatory cell death is a key driver of many inflammatory diseases. Pyroptosis, a highly inflammatory form of cell death, uses intracellularly generated pores to disrupt electrolyte homeostasis and execute cell death. Gasdermin D, the pore-forming effector protein of pyroptosis, coordinates membrane lysis and the release of highly inflammatory molecules, such as interleukin-1ß, which potentiate the overactivation of the innate immune response. However, to date, there is no pharmacologic mechanism to disrupt pyroptosis. Here, we identify necrosulfonamide as a direct chemical inhibitor of gasdermin D, the pyroptotic pore-forming protein, which binds directly to gasdermin D to inhibit pyroptosis. Pharmacologic inhibition of pyroptotic cell death by necrosulfonamide is efficacious in sepsis models and suggests that gasdermin D inhibitors may be efficacious clinically in inflammatory diseases.


Asunto(s)
Acrilamidas/farmacología , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Piroptosis/efectos de los fármacos , Sulfonamidas/farmacología , Acrilamidas/uso terapéutico , Animales , Proteínas Reguladoras de la Apoptosis/fisiología , Citocinas/genética , Femenino , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lipopolisacáridos , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Proteínas de Neoplasias/fisiología , Proteínas de Unión a Fosfato , Pirina/fisiología , Infecciones por Salmonella/tratamiento farmacológico , Infecciones por Salmonella/inmunología , Salmonella typhimurium , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Sulfonamidas/uso terapéutico , Células THP-1
14.
Proc Natl Acad Sci U S A ; 115(26): 6792-6797, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29891674

RESUMEN

The inflammasomes are signaling platforms that promote the activation of inflammatory caspases such as caspases-1, -4, -5, and -11. Recent studies identified gasdermin D (GSDMD) as an effector for pyroptosis downstream of the inflammasome signaling pathways. Cleavage of GSDMD by inflammatory caspases allows its N-terminal domain to associate with membrane lipids and form pores that induce pyroptotic cell death. Despite the important role of GSDMD in pyroptosis, the molecular mechanisms of GSDMD recognition and cleavage by inflammatory caspases that trigger pyroptosis are poorly understood. Here, we demonstrate that the catalytic domains of inflammatory caspases can directly bind to both the full-length GSDMD and its cleavage site peptide, FLTD. A GSDMD-derived inhibitor, N-acetyl-Phe-Leu-Thr-Asp-chloromethylketone (Ac-FLTD-CMK), inhibits GSDMD cleavage by caspases-1, -4, -5, and -11 in vitro, suppresses pyroptosis downstream of both canonical and noncanonical inflammasomes, as well as reduces IL-1ß release following activation of the NLRP3 inflammasome in macrophages. By contrast, the inhibitor does not target caspase-3 or apoptotic cell death, suggesting that Ac-FLTD-CMK is a specific inhibitor for inflammatory caspases. Crystal structure of caspase-1 in complex with Ac-FLTD-CMK reveals extensive enzyme-inhibitor interactions involving both hydrogen bonds and hydrophobic contacts. Comparison with other caspase-1 structures demonstrates drastic conformational changes at the four active-site loops that assemble the catalytic groove. The present study not only contributes to our understanding of GSDMD recognition by inflammatory caspases but also reports a specific inhibitor for these caspases that can serve as a tool for investigating inflammasome signaling.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/química , Inhibidores de Caspasas/química , Proteínas de Neoplasias/química , Péptidos/química , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasa 3/química , Caspasa 3/metabolismo , Inhibidores de Caspasas/metabolismo , Dominio Catalítico , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Células Jurkat , Ratones , Proteínas de Neoplasias/metabolismo , Péptidos/metabolismo , Proteínas de Unión a Fosfato , Estructura Secundaria de Proteína , Células RAW 264.7 , Células THP-1
15.
Structure ; 26(5): 778-784.e3, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29576317

RESUMEN

Pyroptosis is an inflammatory form of programmed cell death that plays important roles in immune protection against infections and in inflammatory disorders. Gasdermin D (GSDMD) is an executor of pyroptosis upon cleavage by caspases-1/4/5/11 following canonical and noncanonical inflammasome activation. GSDMD N-terminal domain assembles membrane pores to induce cytolysis, whereas its C-terminal domain inhibits cell death through intramolecular association with the N domain. The molecular mechanisms of autoinhibition for GSDMD are poorly characterized. Here we report the crystal structures of the human and murine GSDMD C-terminal domains, which differ from those of the full-length murine GSDMA3 and the human GSDMB C-terminal domain. Mutations of GSDMD C-domain residues predicted to locate at its interface with the N-domain enhanced pyroptosis. Our results suggest that GSDMDs may employ a distinct mode of intramolecular domain interaction and autoinhibition, which may be relevant to its unique role in pyroptosis downstream of inflammasome activation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/química , Mutación , Proteínas de Neoplasias/química , Animales , Proteínas Reguladoras de la Apoptosis/genética , Cristalografía por Rayos X , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Modelos Moleculares , Proteínas de Neoplasias/genética , Proteínas de Unión a Fosfato , Dominios Proteicos , Estructura Secundaria de Proteína , Piroptosis
16.
J Clin Invest ; 128(3): 944-959, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29376892

RESUMEN

Coagulation factor XII (FXII) deficiency is associated with decreased neutrophil migration, but the mechanisms remain uncharacterized. Here, we examine how FXII contributes to the inflammatory response. In 2 models of sterile inflammation, FXII-deficient mice (F12-/-) had fewer neutrophils recruited than WT mice. We discovered that neutrophils produced a pool of FXII that is functionally distinct from hepatic-derived FXII and contributes to neutrophil trafficking at sites of inflammation. FXII signals in neutrophils through urokinase plasminogen activator receptor-mediated (uPAR-mediated) Akt2 phosphorylation at S474 (pAktS474). Downstream of pAkt2S474, FXII stimulation of neutrophils upregulated surface expression of αMß2 integrin, increased intracellular calcium, and promoted extracellular DNA release. The sum of these activities contributed to neutrophil cell adhesion, migration, and release of neutrophil extracellular traps in a process called NETosis. Decreased neutrophil signaling in F12-/- mice resulted in less inflammation and faster wound healing. Targeting hepatic F12 with siRNA did not affect neutrophil migration, whereas WT BM transplanted into F12-/- hosts was sufficient to correct the neutrophil migration defect in F12-/- mice and restore wound inflammation. Importantly, these activities were a zymogen FXII function and independent of FXIIa and contact activation, highlighting that FXII has a sophisticated role in vivo that has not been previously appreciated.


Asunto(s)
Factor XII/metabolismo , Neutrófilos/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Cicatrización de Heridas , Animales , Calcio/metabolismo , Adhesión Celular , Movimiento Celular , Células Cultivadas , Trampas Extracelulares , Femenino , Humanos , Inflamación , Leucocitos/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peritonitis/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal
17.
J Biol Chem ; 292(35): 14649-14658, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28726636

RESUMEN

Pyroptosis is a form of cell death important in defenses against pathogens that can also result in a potent and sometimes pathological inflammatory response. During pyroptosis, GSDMD (gasdermin D), the pore-forming effector protein, is cleaved, forms oligomers, and inserts into the membranes of the cell, resulting in rapid cell death. However, the potent cell death induction caused by GSDMD has complicated our ability to understand the biology of this protein. Studies aimed at visualizing GSDMD have relied on expression of GSDMD fragments in epithelial cell lines that naturally lack GSDMD expression and also lack the proteases necessary to cleave GSDMD. In this work, we performed mutagenesis and molecular modeling to strategically place tags and fluorescent proteins within GSDMD that support native pyroptosis and facilitate live-cell imaging of pyroptotic cell death. Here, we demonstrate that these fusion proteins are cleaved by caspases-1 and -11 at Asp-276. Mutations that disrupted the predicted p30-p20 autoinhibitory interface resulted in GSDMD aggregation, supporting the oligomerizing activity of these mutations. Furthermore, we show that these novel GSDMD fusions execute inflammasome-dependent pyroptotic cell death in response to multiple stimuli and allow for visualization of the morphological changes associated with pyroptotic cell death in real time. This work therefore provides new tools that not only expand the molecular understanding of pyroptosis but also enable its direct visualization.


Asunto(s)
Caspasa 1/metabolismo , Caspasas Iniciadoras/metabolismo , Caspasas/metabolismo , Inflamasomas/metabolismo , Macrófagos/citología , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Piroptosis , Sustitución de Aminoácidos , Animales , Línea Celular Transformada , Células HEK293 , Humanos , Inflamasomas/inmunología , Péptidos y Proteínas de Señalización Intracelular , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Microscopía Fluorescente , Microscopía por Video , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Proteínas de Unión a Fosfato , Mutación Puntual , Multimerización de Proteína , Transporte de Proteínas , Proteolisis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo
18.
Mol Pharmacol ; 92(1): 30-47, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28461585

RESUMEN

Pannexin-1 (Panx1) channels mediate the efflux of ATP and AMP from cancer cells in response to induction of extrinsic apoptosis by death receptors or intrinsic apoptosis by chemotherapeutic agents. We previously described the accumulation of extracellular ATP /AMP during chemotherapy-induced apoptosis in Jurkat human leukemia cells. In this study, we compared how different signaling pathways determine extracellular nucleotide pools in control Jurkat cells versus Jurkat lines that lack the Fas-associated death domain (FADD) or receptor-interacting protein kinase 1 (RIP1) cell death regulatory proteins. Tumor necrosis factor-α induced extrinsic apoptosis in control Jurkat cells and necroptosis in FADD-deficient cells; treatment of both lines with chemotherapeutic drugs elicited similar intrinsic apoptosis. Robust extracellular ATP/AMP accumulation was observed in the FADD-deficient cells during necroptosis, but not during apoptotic activation of Panx1 channels. Accumulation of extracellular ATP/AMP was similarly absent in RIP1-deficient Jurkat cells during apoptotic responses to chemotherapeutic agents. Apoptotic activation triggered equivalent proteolytic gating of Panx1 channels in all three Jurkat cell lines. The differences in extracellular ATP/AMP accumulation correlated with cell-line-specific expression of ectonucleotidases that metabolized the released ATP/AMP. CD73 mRNA, and αß-methylene-ADP-inhibitable ecto-AMPase activity were elevated in the FADD-deficient cells. In contrast, the RIP1-deficient cells were defined by increased expression of tartrate-sensitive prostatic acid phosphatase as a broadly acting ectonucleotidase. Thus, extracellular nucleotide accumulation during regulated tumor cell death involves interplay between ATP/AMP efflux pathways and different cell-autonomous ectonucleotidases. Differential expression of particular ectonucleotidases in tumor cell variants will determine whether chemotherapy-induced activation of Panx1 channels drives accumulation of immunostimulatory ATP versus immunosuppressive adenosine within the tumor microenvironment.


Asunto(s)
Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Antineoplásicos/toxicidad , Conexinas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/deficiencia , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Complejo Poro Nuclear/deficiencia , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/metabolismo , Humanos , Células Jurkat , Proteínas de Unión al ARN , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
19.
J Biol Chem ; 292(23): 9666-9679, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28404814

RESUMEN

The X-linked inhibitor of apoptosis (XIAP) protein has been identified as a key genetic driver of two distinct inflammatory disorders, X-linked lymphoproliferative syndrome 2 (XLP-2) and very-early-onset inflammatory bowel disease (VEO-IBD). Molecularly, the role of XIAP mutations in the pathogenesis of these disorders is unclear. Recent work has consistently shown XIAP to be critical for signaling downstream of the Crohn's disease susceptibility protein nucleotide-binding oligomerization domain-containing 2 (NOD2); however, the reported effects of XLP-2 and VEO-IBD XIAP mutations on cell death have been inconsistent. In this manuscript, we describe a CRISPR-mediated genetic system for cells of the myeloid lineage in which XIAP alleles can be replaced with disease-associated XIAP variants expressed at endogenous levels to simultaneously study inflammation-related cell death and NOD2 signaling. We show that, consistent with previous studies, NOD2 signaling is critically dependent on the BIR2 domain of XIAP. We further used this system to reconcile the aforementioned inconsistent XIAP cell death data to show that XLP-2 and VEO-IBD XIAP mutations that exhibit a loss-of-function NOD2 phenotype also lower the threshold for inflammatory cell death. Last, we identified and studied three novel patient XIAP mutations and used this system to characterize NOD2 and cell death phenotypes driven by XIAP. The results of this work support the role of XIAP in mediating NOD2 signaling while reconciling the role of XLP-2 and VEO-IBD XIAP mutations in inflammatory cell death and provide a set of tools and framework to rapidly test newly discovered XIAP variants.


Asunto(s)
Enfermedades Inflamatorias del Intestino/metabolismo , Trastornos Linfoproliferativos/metabolismo , Mutación , Proteína Adaptadora de Señalización NOD2/metabolismo , Transducción de Señal , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Línea Celular , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/patología , Proteína Adaptadora de Señalización NOD2/genética , Dominios Proteicos , Proteína Inhibidora de la Apoptosis Ligada a X/genética
20.
Diabetes ; 66(2): 483-493, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27474370

RESUMEN

Müller cells and macrophages/microglia are likely important for the development of diabetic retinopathy; however, the interplay between these cells in this disease is not well understood. An inflammatory process is linked to the onset of experimental diabetic retinopathy. CD40 deficiency impairs this process and prevents diabetic retinopathy. Using mice with CD40 expression restricted to Müller cells, we identified a mechanism by which Müller cells trigger proinflammatory cytokine expression in myeloid cells. During diabetes, mice with CD40 expressed in Müller cells upregulated retinal tumor necrosis factor-α (TNF-α), interleukin 1ß (IL-1ß), intracellular adhesion molecule 1 (ICAM-1), and nitric oxide synthase (NOS2), developed leukostasis and capillary degeneration. However, CD40 did not cause TNF-α or IL-1ß secretion in Müller cells. TNF-α was not detected in Müller cells from diabetic mice with CD40+ Müller cells. Rather, TNF-α was upregulated in macrophages/microglia. CD40 ligation in Müller cells triggered phospholipase C-dependent ATP release that caused P2X7-dependent production of TNF-α and IL-1ß by macrophages. P2X7-/- mice and mice treated with a P2X7 inhibitor were protected from diabetes-induced TNF-α, IL-1ß, ICAM-1, and NOS2 upregulation. Our studies indicate that CD40 in Müller cells is sufficient to upregulate retinal inflammatory markers and appears to promote experimental diabetic retinopathy and that Müller cells orchestrate inflammatory responses in myeloid cells through a CD40-ATP-P2X7 pathway.


Asunto(s)
Antígenos CD40/inmunología , Citocinas/inmunología , Diabetes Mellitus Experimental/inmunología , Retinopatía Diabética/inmunología , Células Ependimogliales/inmunología , Macrófagos/inmunología , Microglía/inmunología , Receptores Purinérgicos P2X7/inmunología , Animales , Antígenos CD40/genética , Capilares , Diabetes Mellitus Experimental/complicaciones , Retinopatía Diabética/etiología , Inflamación , Molécula 1 de Adhesión Intercelular/inmunología , Interleucina-1beta/inmunología , Leucostasis/inmunología , Masculino , Ratones , Ratones Noqueados , Células Mieloides/inmunología , Óxido Nítrico Sintasa de Tipo II/inmunología , Antagonistas del Receptor Purinérgico P2X/farmacología , Factor de Necrosis Tumoral alfa/inmunología , Fosfolipasas de Tipo C/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA