Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Mol Immunol ; 19(7): 777-790, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35468944

RESUMEN

The food colorant Red 40 is an environmental risk factor for colitis development in mice with increased expression of interleukin (IL)-23. This immune response is mediated by CD4+ T cells, but mechanistic insights into how these CD4+ T cells trigger and perpetuate colitis have remained elusive. Here, using single-cell transcriptomic analysis, we found that several CD4+ T-cell subsets are present in the intestines of colitic mice, including an interferon (IFN)-γ-producing subset. In vivo challenge of primed mice with Red 40 promoted rapid activation of CD4+ T cells and caused marked intestinal epithelial cell (IEC) apoptosis that was attenuated by depletion of CD4+ cells and blockade of IFN-γ. Ex vivo experiments showed that intestinal CD4+ T cells from colitic mice directly promoted apoptosis of IECs and intestinal enteroids. CD4+ T cell-mediated cytotoxicity was contact-dependent and required FasL, which promoted caspase-dependent cell death in target IECs. Genetic ablation of IFN-γ constrained IL-23- and Red 40-induced colitis development, and blockade of IFN-γ inhibited epithelial cell death in vivo. These results advance the understanding of the mechanisms regulating colitis development caused by IL-23 and food colorants and identify IFN-γ+ cytotoxic CD4+ T cells as a new potential therapeutic target for colitis.


Asunto(s)
Linfocitos T CD4-Positivos , Colitis , Colorantes de Alimentos , Interleucina-23 , Animales , Linfocitos T CD4-Positivos/inmunología , Colitis/inducido químicamente , Colitis/inmunología , Colorantes de Alimentos/efectos adversos , Interferón gamma/metabolismo , Interleucina-23/efectos adversos , Ratones , Ratones Endogámicos C57BL
2.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34887354

RESUMEN

SHARPIN, together with RNF31/HOIP and RBCK1/HOIL1, form the linear ubiquitin chain assembly complex (LUBAC) E3 ligase that catalyzes M1-linked polyubiquitination. Mutations in RNF31/HOIP and RBCK/HOIL1 in humans and Sharpin in mice lead to autoinflammation and immunodeficiency, but the mechanism underlying the immune dysregulation remains unclear. We now show that the phenotype of the Sharpincpdm/cpdm mice is dependent on CYLD, a deubiquitinase previously shown to mediate removal of K63-linked polyubiquitin chains. Dermatitis, disrupted splenic architecture, and loss of Peyer's patches in the Sharpincpdm/cpdm mice were fully reversed in Sharpincpdm/cpdm Cyld-/- mice. We observed enhanced association of RIPK1 with the death-signaling Complex II following TNF stimulation in Sharpincpdm/cpdm cells, a finding dependent on CYLD since we observed reversal in Sharpincpdm/cpdm Cyld-/- cells. Enhanced RIPK1 recruitment to Complex II in Sharpincpdm/cpdm cells correlated with impaired phosphorylation of CYLD at serine 418, a modification reported to inhibit its enzymatic activity. The dermatitis in the Sharpincpdm/cpdm mice was also ameliorated by the conditional deletion of Cyld using LysM-cre or Cx3cr1-cre indicating that CYLD-dependent death of myeloid cells is inflammatory. Our studies reveal that under physiological conditions, TNF- and RIPK1-dependent cell death is suppressed by the linear ubiquitin-dependent inhibition of CYLD. The Sharpincpdm/cpdm phenotype illustrates the pathological consequences when CYLD inhibition fails.


Asunto(s)
Enzima Desubiquitinante CYLD/metabolismo , Fibroblastos/metabolismo , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Muerte Celular , Enzima Desubiquitinante CYLD/genética , Embrión de Mamíferos/citología , Femenino , Regulación de la Expresión Génica/inmunología , Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Noqueados , Células Mieloides , Fosforilación , Enfermedades de la Piel , Ubiquitinación
3.
JCI Insight ; 6(24)2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34752416

RESUMEN

TNF ligation of TNF receptor 1 (TNFR1) promotes either inflammation and cell survival by (a) inhibiting RIPK1's death-signaling function and activating NF-κB or (b) causing RIPK1 to associate with the death-inducing signaling complex to initiate apoptosis or necroptosis. The cellular source of TNF that results in RIPK1-dependent cell death remains unclear. To address this, we employed in vitro systems and murine models of T cell-dependent transplant or tumor rejection in which target cell susceptibility to RIPK1-dependent cell death could be genetically altered. We show that TNF released by T cells is necessary and sufficient to activate RIPK1-dependent cell death in target cells and thereby mediate target cell cytolysis independently of T cell frequency. Activation of the RIPK1-dependent cell death program in target cells by T cell-derived TNF accelerates murine cardiac allograft rejection and synergizes with anti-PD1 administration to destroy checkpoint blockade-resistant murine melanoma. Together, the findings uncover a distinct immunological role for TNF released by cytotoxic effector T cells following cognate interactions with their antigenic targets. Manipulating T cell TNF and/or target cell susceptibility to RIPK1-dependent cell death can be exploited to either mitigate or augment T cell-dependent destruction of allografts and malignancies to improve outcomes.


Asunto(s)
Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factores de Transcripción TCF/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Muerte Celular , Humanos , Ratones
4.
Cell ; 184(17): 4447-4463.e20, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34363755

RESUMEN

TANK binding kinase 1 (TBK1) regulates IFN-I, NF-κB, and TNF-induced RIPK1-dependent cell death (RCD). In mice, biallelic loss of TBK1 is embryonically lethal. We discovered four humans, ages 32, 26, 7, and 8 from three unrelated consanguineous families with homozygous loss-of-function mutations in TBK1. All four patients suffer from chronic and systemic autoinflammation, but not severe viral infections. We demonstrate that TBK1 loss results in hypomorphic but sufficient IFN-I induction via RIG-I/MDA5, while the system retains near intact IL-6 induction through NF-κB. Autoinflammation is driven by TNF-induced RCD as patient-derived fibroblasts experienced higher rates of necroptosis in vitro, and CC3 was elevated in peripheral blood ex vivo. Treatment with anti-TNF dampened the baseline circulating inflammatory profile and ameliorated the clinical condition in vivo. These findings highlight the plasticity of the IFN-I response and underscore a cardinal role for TBK1 in the regulation of RCD.


Asunto(s)
Inflamación/enzimología , Proteínas Serina-Treonina Quinasas/deficiencia , Factor de Necrosis Tumoral alfa/farmacología , Células A549 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Autoinmunidad/efectos de los fármacos , Encéfalo/diagnóstico por imagen , Muerte Celular/efectos de los fármacos , Citocinas/metabolismo , Enzima Desubiquitinante CYLD/metabolismo , Femenino , Células HEK293 , Homocigoto , Humanos , Quinasa I-kappa B/metabolismo , Inmunofenotipificación , Inflamación/patología , Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Mutación con Pérdida de Función/genética , Masculino , Linaje , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Receptor Toll-Like 3/metabolismo , Transcriptoma/genética , Vesiculovirus/efectos de los fármacos , Vesiculovirus/fisiología
5.
Cell Death Dis ; 11(2): 94, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-32024820

RESUMEN

Adult T-cell leukemia/lymphoma (ATLL) is a malignancy of mature T cells associated with chronic infection by human T-cell lymphotropic virus type-1 (HTLV-1). ATLL patients with aggressive subtypes have dismal outcomes. We demonstrate that ATLL cells co-opt an early checkpoint within the tumor necrosis factor receptor 1 (TNFR1) pathway, resulting in survival advantage. This early checkpoint revolves around an interaction between the deubiquitinase CYLD and its target RIPK1. The status of RIPK1 K63-ubiquitination determines cell fate by creating either a prosurvival signal (ubiquitinated RIPK1) or a death signal (deubiquitinated RIPK1). In primary ATLL samples and in cell line models, an increased baseline level of CYLD phosphorylation was observed. We therefore tested the hypothesis that this modification of CYLD, which has been reported to inhibit its deubiquitinating function, leads to increased RIPK1 ubiquitination and thus provides a prosurvival signal to ATLL cells. CYLD phosphorylation can be pharmacologically reversed by IKK inhibitors, specifically by TBK1/IKKε and IKKß inhibitors (MRT67307 and TPCA). Both of the IKK sub-families can phosphorylate CYLD, and the combination of MRT67307 and TPCA have a marked effect in reducing CYLD phosphorylation and triggering cell death. ATLL cells overexpressing a kinase-inactive TBK1 (TBK1-K38A) demonstrate lower CYLD phosphorylation and subsequently reduced proliferation. IKK blockade reactivates CYLD, as evidenced by the reduction in RIPK1 ubiquitination, which leads to the association of RIPK1 with the death-inducing signaling complex (DISC) to trigger cell death. In the absence of CYLD, RIPK1 ubiquitination remains elevated following IKK blockade and it does not associate with the DISC. SMAC mimetics can similarly disrupt CYLD phosphorylation and lead to ATLL cell death through reduction of RIPK1 ubiquitination, which is CYLD dependent. These results identify CYLD as a crucial regulator of ATLL survival and point to its role as a potential novel target for pharmacologic modification in this disease.


Asunto(s)
Antineoplásicos/farmacología , Enzima Desubiquitinante CYLD/metabolismo , Quinasa I-kappa B/antagonistas & inhibidores , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/genética , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Enzima Desubiquitinante CYLD/genética , Regulación Leucémica de la Expresión Génica , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Células Jurkat , Leucemia-Linfoma de Células T del Adulto/genética , Leucemia-Linfoma de Células T del Adulto/metabolismo , Leucemia-Linfoma de Células T del Adulto/patología , Fosforilación , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Ubiquitinación
6.
Cell Death Dis ; 11(1): 60, 2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31974345

RESUMEN

The original version of this article contained an error in the name of one of the co-authors (Wim Declercq). This has been corrected in the PDF and HTML versions.

8.
Dev Cell ; 51(2): 277-291.e4, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31564612

RESUMEN

Quantifying cytostatic and cytotoxic outcomes are integral components of characterizing perturbagens used as research tools and in drug discovery pipelines. Furthermore, data-rich acquisition, coupled with robust methods for analysis, is required to properly assess the function and impact of these perturbagens. Here, we present a detailed and versatile method for single-cell and population-level analyses using real-time kinetic labeling (SPARKL). SPARKL integrates high-content live-cell imaging with automated detection and analysis of fluorescent reporters of cell death. We outline several examples of zero-handling, non-disruptive protocols for detailing cell death mechanisms and proliferation profiles. Additionally, we suggest several methods for mathematically analyzing these data to best utilize the collected kinetic data. Compared to traditional methods of detection and analysis, SPARKL is more sensitive, accurate, and high throughput while substantially eliminating sample processing and providing richer data.


Asunto(s)
Apoptosis/fisiología , Muerte Celular/fisiología , Proliferación Celular/fisiología , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Cinética
9.
Cell Death Dis ; 10(10): 692, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31534131

RESUMEN

The cytokine TNF promotes inflammation either directly by activating the MAPK and NF-κB signaling pathways, or indirectly by triggering cell death. A20 is a potent anti-inflammatory molecule, and mutations in the gene encoding A20 are associated with a wide panel of inflammatory pathologies, both in human and in the mouse. Binding of TNF to TNFR1 triggers the NF-κB-dependent expression of A20 as part of a negative feedback mechanism preventing sustained NF-κB activation. Apart from acting as an NF-κB inhibitor, A20 is also well-known for its ability to counteract the cytotoxic potential of TNF. However, the mechanism by which A20 mediates this function and the exact cell death modality that it represses have remained incompletely understood. In the present study, we provide in vitro and in vivo evidences that deletion of A20 induces RIPK1 kinase-dependent and -independent apoptosis upon single TNF stimulation. We show that constitutively expressed A20 is recruited to TNFR1 signaling complex (Complex I) via its seventh zinc finger (ZF7) domain, in a cIAP1/2-dependent manner, within minutes after TNF sensing. We demonstrate that Complex I-recruited A20 protects cells from apoptosis by stabilizing the linear (M1) ubiquitin network associated to Complex I, a process independent of its E3 ubiquitin ligase and deubiquitylase (DUB) activities and which is counteracted by the DUB CYLD, both in vitro and in vivo. In absence of linear ubiquitylation, A20 is still recruited to Complex I via its ZF4 and ZF7 domains, but this time protects the cells from death by deploying its DUB activity. Together, our results therefore demonstrate two distinct molecular mechanisms by which constitutively expressed A20 protect cells from TNF-induced apoptosis.


Asunto(s)
Receptores Tipo I de Factores de Necrosis Tumoral/efectos adversos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/uso terapéutico , Ubiquitina/efectos de los fármacos , Animales , Apoptosis , Humanos , Ratones , Transducción de Señal , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología
10.
Front Cell Dev Biol ; 7: 163, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31457011

RESUMEN

The clinical success of biologics that inhibit TNF (Tumor Necrosis Factor) in inflammatory bowel diseases (IBD), psoriasis and rheumatoid arthritis (RA) has clearly established a pathogenic role for this cytokine in these inflammatory disorders. TNF binding to its receptors activates NFκB and MAPK signaling, inducing the expression of downstream pro-inflammatory genes. This is thought to be the primary mechanism by which TNF elicits inflammation. TNF is also a well-known trigger of caspase-dependent apoptosis or caspase-independent necroptosis. Whether cell death has any role in TNF-mediated inflammation has been less clear. Emerging data from animal models now suggest that cellular demise caused by TNF may indeed provoke inflammation. The default response of most cells to TNF stimulation is survival, rather than death, due to the presence of two sequential cell death checkpoints. The early checkpoint is transcription-independent involving the non-degradative ubiquitination of RIPK1 to prevent RIPK1 from becoming a death-signaling molecule. The later checkpoint requires the induction of pro-survival genes by NFκB-mediated transcription. When the early checkpoint is disrupted, RIPK1 initiates cell death and we suggest the term ripoptocide to describe this manner of death (encompassing both apoptosis and necroptosis). The sensitivity of a cell to ripoptocide is determined by the balance between regulatory molecules that enforce and those that disassemble the early checkpoint. As there is evidence suggesting that ripoptocide is inflammatory, individuals may develop inflammation due to ripoptocide brought about by genetic, epigenetic or post-translational alteration of these checkpoint regulators. For these individuals, drugs that reinforce the early checkpoint and inhibit ripoptocide could be useful in ameliorating inflammation.

11.
Curr Opin Organ Transplant ; 24(1): 12-19, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30507704

RESUMEN

PURPOSE OF REVIEW: Regulated cell death (RCD) is likely to play a role in organ rejection but it is unclear how it may be invoked. A well-known trigger of regulated cell death is tumor necrosis factor-alpha (TNF), which activates both caspase-dependent apoptosis and caspase-independent necroptosis. TNF is best known as a pro-inflammatory cytokine because it activates NFκB and MAPK signaling to induce expression of pro-inflammatory genes. RECENT FINDINGS: Emerging data from animal models now suggest that TNF-induced cell death can also be inflammatory. Therefore, the role of cellular demise in regulating immunity should be considered. In transplantation, TNF could have a role in cellular injury or death from ischemia reperfusion (IR) injury and this may dictate organ survival. The default response to TNF in most cells is survival, rather than death, because of the presence of cell death checkpoints. However, cells succumb to TNF-driven death when these checkpoints are disrupted, and sensitivity to death likely reflects a reduction in molecules that fortify these checkpoints. We propose that a cell's propensity to die in response to TNF may underlie allograft rejection. SUMMARY: Genetic, epigenetic, and posttranslational control of death checkpoint regulators in donor tissues may determine graft survival. Therapeutically, drugs that prevent donor cell demise could be useful in preventing organ rejection.


Asunto(s)
Muerte Celular/genética , Factor de Necrosis Tumoral alfa/efectos adversos , Apoptosis , Humanos , Donantes de Tejidos
12.
Front Immunol ; 9: 2125, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30333819

RESUMEN

TACI signals activate B cell proliferation, isotype switch and antibody production in both normal immunity and autoimmune states. In contrast to murine TACI, the human TACI gene undergoes alternative splicing to produce short and long isoforms (TACI-S and TACI-L). In previous studies, we showed that transduction of the short, but not long isoform, into murine B cells or human pre-B cells lacking TACI, caused them to become transcriptional and morphologically identical to plasma cells. These data suggest that the expression of different isoforms in humans provides unique controls on B cell maturation. In these studies we show that TACI-S and TACI-L form complexes in a ligand-independent manner, not dependent on a single extracellular domain. Both TACI isoforms are detectable in the endosomal cellular compartment where they co-localize with MyD88, TRAF6, and the activated 65 kDa form of TLR9, depending on a conserved intracellular TACI sequence. In contrast to TACI-L expressing cells, or cells bearing both isoforms, TACI-S binds ligands BAFF and APRIL with substantially greater affinity and promotes enhanced NF-kB activation. Using isoform-specific monoclonal antibodies, we show that while TACI-L is predominant as a surface receptor surface on human B cells, significantly more TACI-S is noted in the intracellular compartment and also in marginal zone, isotype switched and plasmablast in resting B cells. TACI-S is increased in tonsillar B cells and also in the intracellular compartment of activated peripheral B cells. These data shows that alternative splicing of the human TACI gene leads to two isoforms both of which intersect with MyD88 and TRAF6 and form complexes with TLR9, but the two isoforms have different ligand binding capacities, subcellular locations and activation capabilities.


Asunto(s)
Factor Activador de Células B/inmunología , Células Plasmáticas/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Animales , Factor Activador de Células B/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Células Plasmáticas/citología , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/inmunología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
13.
Methods Mol Biol ; 1857: 1-9, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30136225

RESUMEN

Necroptosis is a more recently described form of regulated cell death (RCD) that occurs in a caspase-independent manner. This is a lytic form of cell death in which the cellular contents are released and these contents serve as damage-associated molecular patterns (DAMPs). DAMPs are endogenous ligands for pattern recognition receptors and therefore necroptosis is considered to be highly inflammatory and immunogenic. Members of the TNF family are the most well-studied triggers of necroptosis, though other immune receptors are also known to directly trigger this death pathway. Necroptosis is now defined to be dependent on the core signaling molecules of RIPK3 and MLKL. In the case of TNF, it also involves RIPK1, and induction of necroptosis often also requires inhibition of caspases. In this volume, a wide array of tools to study necroptosis are described. They include pharmacological, biochemical, cellular, and in vivo approaches. Combining multiple approaches in one's study is ideal for generating conclusive evidence for the involvement of necroptosis. The protocols presented in this chapter are highly useful in studying necroptosis, whose physiological and pathophysiological roles remain incompletely understood.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Modelos Biológicos , Necrosis , Proteínas Quinasas/metabolismo , Animales , Humanos , Transducción de Señal
14.
Methods Mol Biol ; 1857: 63-70, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30136230

RESUMEN

Necroptosis in macrophages is increasingly being recognized as a significant contributor to the inflammation seen in many pathologies. Here we describe a well-known method to obtain quiescent, mature macrophages that can be used to study the molecular mechanisms that regulate necroptosis. We also describe two ways to quantify this form of programmed cell death.


Asunto(s)
Apoptosis , Macrófagos/patología , Necrosis , Animales , Células Cultivadas , Macrófagos/efectos de los fármacos , Ratones , Transducción de Señal
15.
Methods Mol Biol ; 1857: 101-107, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30136234

RESUMEN

FAS-associated protein with death domain (FADD) is a signaling molecule required by members of the TNF receptor superfamily (TNFRSF) such as FAS and TNFR1 to induce apoptosis. FADD is a small adapter molecule that functions as a scaffold to recruit procaspase-8 and other regulators. The FADD-containing signaling complex that initiates the apoptotic cascade has been termed the death inducing signaling complex (DISC). In the absence of FADD, death receptors cannot induce apoptosis and in appropriate cell types, these death receptors then induce necroptosis. Necroptosis can also be induced by death receptors in FADD-sufficient cells when caspase-8 is inhibited, usually accomplished by the addition of caspase inhibitors. Under such necroptotic conditions, the immunoprecipitation of FADD to isolate the DISC can be utilized to examine components of this complex. Here, we describe the immunoprecipitation of FADD and subsequent western-blotting to identify RIPK1 in this complex during necroptosis.


Asunto(s)
Apoptosis , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Embrión de Mamíferos/patología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Fibroblastos/patología , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Ratones , Transducción de Señal
16.
Methods Mol Biol ; 1857: 181-188, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30136242

RESUMEN

Previous studies have demonstrated that CASPASE 8 can generate a prosurvival signal by inhibiting necroptosis via the cleavage of the deubiquitinating enzyme CYLD. Cleavage of CYLD at D215 results in the generation of a 25 kD N-terminal fragment and degradation of the C-terminal fragment containing the catalytic domain. Since CYLD is required for TNF-induced necroptosis, its proteolysis is necessary and sufficient to suppress necroptosis and generate a survival signal. Here we describe how to visualize CYLD proteolysis by western blot analysis, as a measure of CASPASE 8 activity and inhibition of necroptosis.


Asunto(s)
Apoptosis , Caspasa 8/metabolismo , Cisteína Endopeptidasas/metabolismo , Macrófagos/patología , Necrosis , Animales , Células Cultivadas , Enzima Desubiquitinante CYLD , Macrófagos/metabolismo , Ratones , Proteolisis , Transducción de Señal
18.
Cell Death Dis ; 9(3): 359, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29500402

RESUMEN

Receptor-interacting protein kinases 1 and 3 (RIPK1/3) have best been described for their role in mediating a regulated form of necrosis, referred to as necroptosis. During this process, RIPK3 phosphorylates mixed lineage kinase domain-like (MLKL) to cause plasma membrane rupture. RIPK3-deficient mice have recently been demonstrated to be protected in a series of disease models, but direct evidence for activation of necroptosis in vivo is still limited. Here, we sought to further examine the activation of necroptosis in kidney ischemia-reperfusion injury (IRI) and from TNFα-induced severe inflammatory response syndrome (SIRS), two models of RIPK3-dependent injury. In both models, MLKL-ko mice were significantly protected from injury to a degree that was slightly, but statistically significantly exceeding that of RIPK3-deficient mice. We also demonstrated, for the first time, accumulation of pMLKL in the necrotic tubules of human patients with acute kidney injury. However, our data also uncovered unexpected elevation of blood flow in MLKL-ko animals, which may be relevant to IRI and should be considered in the future. To further understand the mode of regulation of cell death by MLKL, we screened a panel of clinical plasma membrane channel blockers and we found phenytoin to inhibit necroptosis. However, we further found that phenytoin attenuated RIPK1 kinase activity in vitro, likely due to the hydantoin scaffold also present in necrostatin-1, and blocked upstream necrosome formation steps in the cells undergoing necroptosis. We further report that this clinically used anti-convulsant drug displayed protection from kidney IRI and TNFα-induces SIRS in vivo. Overall, our data reveal the relevance of RIPK3-pMLKL regulation for acute kidney injury and identifies an FDA-approved drug that may be useful for immediate clinical evaluation of inhibition of pro-death RIPK1/RIPK3 activities in human diseases.


Asunto(s)
Anticonvulsivantes/farmacología , Necrosis/prevención & control , Fenitoína/farmacología , Lesión Renal Aguda/patología , Animales , Biopsia , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Síndrome de Respuesta Inflamatoria Sistémica/inducido químicamente , Síndrome de Respuesta Inflamatoria Sistémica/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/farmacología
19.
Cell Rep ; 17(1): 221-232, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27681433

RESUMEN

Microvascular endothelial cells maintain a tight barrier to prevent passage of plasma and circulating immune cells into the extravascular tissue compartment, yet endothelial cells respond rapidly to vasoactive substances, including thrombin, allowing transient paracellular permeability. This response is a cornerstone of acute inflammation, but the mechanisms responsible are still incompletely understood. Here, we demonstrate that thrombin triggers MALT1 to proteolytically cleave cylindromatosis (CYLD). Fragmentation of CYLD results in microtubule disruption and a cascade of events leading to endothelial cell retraction and an acute permeability response. This finding reveals an unexpected role for the MALT1 protease, which previously has been viewed mostly as a driver of pro-inflammatory NF-κB signaling in lymphocytes. Thus, MALT1 not only promotes immune cell activation but also acutely regulates endothelial cell biology, actions that together facilitate tissue inflammation. Pharmacologic inhibition of MALT1 may therefore have synergistic impact by targeting multiple disparate steps in the overall inflammatory response.


Asunto(s)
Caspasas/inmunología , Cisteína Endopeptidasas/inmunología , Células Endoteliales/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Proteínas de Neoplasias/inmunología , Trombina/farmacología , Animales , Transporte Biológico , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/inmunología , Caspasas/genética , Línea Celular , Cisteína Endopeptidasas/genética , Enzima Desubiquitinante CYLD , Células Endoteliales/citología , Células Endoteliales/inmunología , Regulación de la Expresión Génica , Quinasa I-kappa B/genética , Quinasa I-kappa B/inmunología , Ratones , Ratones Transgénicos , Microtúbulos/ultraestructura , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , FN-kappa B/genética , FN-kappa B/inmunología , Proteínas de Neoplasias/genética , Permeabilidad/efectos de los fármacos , Cultivo Primario de Células , Receptor PAR-1/genética , Receptor PAR-1/inmunología , Transducción de Señal , Trombina/metabolismo
20.
Trends Immunol ; 37(8): 535-545, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27424290

RESUMEN

TNF is a master proinflammatory cytokine whose pathogenic role in inflammatory disorders has long been attributed to induction of proinflammatory mediators. TNF also activates cell survival and death pathways, and recent studies demonstrated that TNF also causes inflammation by inducing cell death. The default response of most cells to TNF is survival and NF-κB-mediated upregulation of prosurvival molecules is a well-documented protective mechanism downstream of TNFR1. Recent studies revealed the existence of an NF-κB-independent cell death checkpoint that restricts cell demise by inactivating RIPK1. Disruption of this checkpoint leads to RIPK1 kinase-dependent death and causes inflammation in vivo. These revelations bring complexity to the control of TNF-induced cell death, and suggest clinical benefit of RIPK1 inhibitors in TNF-driven human inflammatory disorders.


Asunto(s)
FN-kappa B/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Animales , Biomarcadores , Muerte Celular , Citocinas/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...