Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 348
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Rev Mol Cell Biol ; 24(5): 312-333, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36543934

RESUMEN

Regulated cell death (RCD) relies on activation and recruitment of pore-forming proteins (PFPs) that function as executioners of specific cell death pathways: apoptosis regulator BAX (BAX), BCL-2 homologous antagonist/killer (BAK) and BCL-2-related ovarian killer protein (BOK) for apoptosis, gasdermins (GSDMs) for pyroptosis and mixed lineage kinase domain-like protein (MLKL) for necroptosis. Inactive precursors of PFPs are converted into pore-forming entities through activation, membrane recruitment, membrane insertion and oligomerization. These mechanisms involve protein-protein and protein-lipid interactions, proteolytic processing and phosphorylation. In this Review, we discuss the structural rearrangements incurred by RCD-related PFPs and describe the mechanisms that manifest conversion from autoinhibited to membrane-embedded molecular states. We further discuss the formation and maturation of membrane pores formed by BAX/BAK/BOK, GSDMs and MLKL, leading to diverse pore architectures. Lastly, we highlight commonalities and differences of PFP mechanisms involving BAX/BAK/BOK, GSDMs and MLKL and conclude with a discussion on how, in a population of challenged cells, the coexistence of cell death modalities may have profound physiological and pathophysiological implications.


Asunto(s)
Apoptosis , Proteína X Asociada a bcl-2/metabolismo , Membrana Celular/metabolismo , Membranas/metabolismo
2.
Immunity ; 50(2): 348-361.e4, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30737145

RESUMEN

NF-κB (nuclear factor κB) signaling is considered critical for single positive (SP) thymocyte development because loss of upstream activators of NF-κB, such as the IKK complex, arrests their development. We found that the compound ablation of RelA, cRel, and p50, required for canonical NF-κB transcription, had no impact upon thymocyte development. While IKK-deficient thymocytes were acutely sensitive to tumor necrosis factor (TNF)-induced cell death, Rel-deficient cells remained resistant, calling into question the importance of NF-κB as the IKK target required for thymocyte survival. Instead, we found that IKK controlled thymocyte survival by repressing cell-death-inducing activity of the serine/threonine kinase RIPK1. We observed that RIPK1 expression was induced during development of SP thymocytes and that IKK was required to prevent RIPK1-kinase-dependent death of SPs in vivo. Finally, we showed that IKK was required to protect Rel-deficient thymocytes from RIPK1-dependent cell death, underscoring the NF-κB-independent function of IKK during thymic development.


Asunto(s)
Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Timocitos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Quinasa I-kappa B/genética , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Timocitos/citología , Timocitos/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
3.
Nature ; 607(7920): 784-789, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35859175

RESUMEN

The RNA-editing enzyme adenosine deaminase acting on RNA 1 (ADAR1) limits the accumulation of endogenous immunostimulatory double-stranded RNA (dsRNA)1. In humans, reduced ADAR1 activity causes the severe inflammatory disease Aicardi-Goutières syndrome (AGS)2. In mice, complete loss of ADAR1 activity is embryonically lethal3-6, and mutations similar to those found in patients with AGS cause autoinflammation7-12. Mechanistically, adenosine-to-inosine (A-to-I) base modification of endogenous dsRNA by ADAR1 prevents chronic overactivation of the dsRNA sensors MDA5 and PKR3,7-10,13,14. Here we show that ADAR1 also inhibits the spontaneous activation of the left-handed Z-nucleic acid sensor ZBP1. Activation of ZBP1 elicits caspase-8-dependent apoptosis and MLKL-mediated necroptosis of ADAR1-deficient cells. ZBP1 contributes to the embryonic lethality of Adar-knockout mice, and it drives early mortality and intestinal cell death in mice deficient in the expression of both ADAR and MAVS. The Z-nucleic-acid-binding Zα domain of ADAR1 is necessary to prevent ZBP1-mediated intestinal cell death and skin inflammation. The Zα domain of ADAR1 promotes A-to-I editing of endogenous Alu elements to prevent dsRNA formation through the pairing of inverted Alu repeats, which can otherwise induce ZBP1 activation. This shows that recognition of Alu duplex RNA by ZBP1 may contribute to the pathological features of AGS that result from the loss of ADAR1 function.


Asunto(s)
Adenosina Desaminasa , Inflamación , Proteínas de Unión al ARN , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Adenosina/metabolismo , Adenosina Desaminasa/química , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/metabolismo , Animales , Apoptosis , Enfermedades Autoinmunes del Sistema Nervioso , Caspasa 8/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/prevención & control , Inosina/metabolismo , Intestinos/patología , Ratones , Necroptosis , Malformaciones del Sistema Nervioso , Edición de ARN , ARN Bicatenario , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Piel/patología
4.
Immunity ; 49(2): 312-325.e5, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30076102

RESUMEN

Heterogeneity between different macrophage populations has become a defining feature of this lineage. However, the conserved factors defining macrophages remain largely unknown. The transcription factor ZEB2 is best described for its role in epithelial to mesenchymal transition; however, its role within the immune system is only now being elucidated. We show here that Zeb2 expression is a conserved feature of macrophages. Using Clec4f-cre, Itgax-cre, and Fcgr1-cre mice to target five different macrophage populations, we found that loss of ZEB2 resulted in macrophage disappearance from the tissues, coupled with their subsequent replenishment from bone-marrow precursors in open niches. Mechanistically, we found that ZEB2 functioned to maintain the tissue-specific identities of macrophages. In Kupffer cells, ZEB2 achieved this by regulating expression of the transcription factor LXRα, removal of which recapitulated the loss of Kupffer cell identity and disappearance. Thus, ZEB2 expression is required in macrophages to preserve their tissue-specific identities.


Asunto(s)
Macrófagos del Hígado/citología , Receptores X del Hígado/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Animales , Linaje de la Célula/inmunología , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Macrófagos del Hígado/inmunología , Hígado/citología , Receptores X del Hígado/metabolismo , Pulmón/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
5.
Nature ; 596(7871): 262-267, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34349263

RESUMEN

Regulated cell death is an integral part of life, and has broad effects on organism development and homeostasis1. Malfunctions within the regulated cell death process, including the clearance of dying cells, can manifest in diverse pathologies throughout various tissues including the gastrointestinal tract2. A long appreciated, yet elusively defined relationship exists between cell death and gastrointestinal pathologies with an underlying microbial component3-6, but the direct effect of dying mammalian cells on bacterial growth is unclear. Here we advance a concept that several Enterobacteriaceae, including patient-derived clinical isolates, have an efficient growth strategy to exploit soluble factors that are released from dying gut epithelial cells. Mammalian nutrients released after caspase-3/7-dependent apoptosis boosts the growth of multiple Enterobacteriaceae and is observed using primary mouse colonic tissue, mouse and human cell lines, several apoptotic triggers, and in conventional as well as germ-free mice in vivo. The mammalian cell death nutrients induce a core transcriptional response in pathogenic Salmonella, and we identify the pyruvate formate-lyase-encoding pflB gene as a key driver of bacterial colonization in three contexts: a foodborne infection model, a TNF- and A20-dependent cell death model, and a chemotherapy-induced mucositis model. These findings introduce a new layer to the complex host-pathogen interaction, in which death-induced nutrient release acts as a source of fuel for intestinal bacteria, with implications for gut inflammation and cytotoxic chemotherapy treatment.


Asunto(s)
Apoptosis , Enterobacteriaceae/crecimiento & desarrollo , Enterobacteriaceae/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Intestinos/citología , Intestinos/microbiología , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Animales , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/patología , Femenino , Enfermedades Transmitidas por los Alimentos/microbiología , Vida Libre de Gérmenes , Interacciones Huésped-Patógeno , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Masculino , Ratones , Mucositis/inducido químicamente , Salmonella/enzimología , Salmonella/genética , Salmonella/crecimiento & desarrollo , Salmonella/metabolismo , Transcriptoma , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
Immunity ; 47(1): 1-3, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28723543

RESUMEN

RIPK3 kinase-mediated phosphorylation of MLKL pseudokinase is the execution event of necroptosis. Two independent reports-in Immunity (Yoon et al., 2017) and Cell (Gong et al., 2017)-reveal that MLKL affects homeostatic membrane trafficking and necroptosis-enhanced bubble formation involving interaction with the ESCRT machinery.


Asunto(s)
Apoptosis , Membrana Celular/metabolismo , Vesículas Extracelulares/metabolismo , Necrosis , Proteínas Quinasas/metabolismo , Transporte de Proteínas , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Complejos de Clasificación Endosomal Requeridos para el Transporte , Humanos , Proteínas Quinasas/inmunología
7.
Nat Rev Mol Cell Biol ; 15(2): 135-47, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24452471

RESUMEN

Cell death research was revitalized by the understanding that necrosis can occur in a highly regulated and genetically controlled manner. Although RIPK1 (receptor-interacting protein kinase 1)- and RIPK3-MLKL (mixed lineage kinase domain-like)-mediated necroptosis is the most understood form of regulated necrosis, other examples of this process are emerging, including cell death mechanisms known as parthanatos, oxytosis, ferroptosis, NETosis, pyronecrosis and pyroptosis. Elucidating how these pathways of regulated necrosis are interconnected at the molecular level should enable this process to be therapeutically targeted.


Asunto(s)
Apoptosis/genética , Terapia Molecular Dirigida , Necrosis/genética , Transducción de Señal , Muerte Celular/genética , Humanos , Necrosis/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
8.
Mol Cell ; 72(3): 397-399, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388406

RESUMEN

In this issue of Molecular Cell, Ying et al. (2018) report on a novel function of MLKL in sciatic nerve regeneration after injury through myelin sheath destabilization. This function of MLKL is independent of necroptosis execution and requires phosphorylation at MLKL-S441.


Asunto(s)
Vaina de Mielina , Proteínas Quinasas , Apoptosis , Humanos , Necrosis , Regeneración Nerviosa , Proteína Serina-Treonina Quinasas de Interacción con Receptores
9.
Mol Cell ; 69(4): 566-580.e5, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29452637

RESUMEN

Tumor necrosis factor (TNF) can drive inflammation, cell survival, and death. While ubiquitylation-, phosphorylation-, and nuclear factor κB (NF-κB)-dependent checkpoints suppress the cytotoxic potential of TNF, it remains unclear whether ubiquitylation can directly repress TNF-induced death. Here, we show that ubiquitylation regulates RIPK1's cytotoxic potential not only via activation of downstream kinases and NF-kB transcriptional responses, but also by directly repressing RIPK1 kinase activity via ubiquitin-dependent inactivation. We find that the ubiquitin-associated (UBA) domain of cellular inhibitor of apoptosis (cIAP)1 is required for optimal ubiquitin-lysine occupancy and K48 ubiquitylation of RIPK1. Independently of IKK and MK2, cIAP1-mediated and UBA-assisted ubiquitylation suppresses RIPK1 kinase auto-activation and, in addition, marks it for proteasomal degradation. In the absence of a functional UBA domain of cIAP1, more active RIPK1 kinase accumulates in response to TNF, causing RIPK1 kinase-mediated cell death and systemic inflammatory response syndrome. These results reveal a direct role for cIAP-mediated ubiquitylation in controlling RIPK1 kinase activity and preventing TNF-mediated cytotoxicity.


Asunto(s)
Proteína 3 que Contiene Repeticiones IAP de Baculovirus/fisiología , Quinasa I-kappa B/metabolismo , Proteínas Inhibidoras de la Apoptosis/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Ubiquitina/metabolismo , Animales , Apoptosis , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Ubiquitinación
10.
Immunity ; 44(4): 807-20, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27037190

RESUMEN

Few apoptotic corpses are seen even in tissues with high cellular turnover, leading to the notion that the capacity for engulfment in vivo is vast. Whether corpse clearance can be enhanced in vivo for potential benefit is not known. In a colonic inflammation model, we noted that the expression of the phagocytic receptor Bai1 was progressively downmodulated. Consistent with this, BAI1-deficient mice had more pronounced colitis and lower survival, with many uncleared apoptotic corpses and inflammatory cytokines within the colonic epithelium. When we engineered and tested transgenic mice overexpressing BAI1, these had fewer apoptotic cells, reduced inflammation, and attenuated disease. Boosting BAI1-mediated uptake by intestinal epithelial cells (rather than myeloid cells) was important in attenuating inflammation. A signaling-deficient BAI1 transgene could not provide a similar benefit. Collectively, these complementary genetic approaches showed that cell clearance could be boosted in vivo, with potential to regulate tissue inflammation in specific contexts.


Asunto(s)
Proteínas Angiogénicas/genética , Apoptosis/inmunología , Colitis/inmunología , Células Epiteliales/inmunología , Mucosa Intestinal/inmunología , Animales , Línea Celular Tumoral , Colitis/inducido químicamente , Colon/inmunología , Colon/patología , Citocinas/inmunología , Sulfato de Dextran , Células HCT116 , Humanos , Inflamación/inmunología , Mucosa Intestinal/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal/inmunología
11.
Proc Natl Acad Sci U S A ; 119(2)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34996874

RESUMEN

Lethal toxin (LeTx)-mediated killing of myeloid cells is essential for Bacillus anthracis, the causative agent of anthrax, to establish systemic infection and induce lethal anthrax. The "LeTx-sensitive" NLRP1b inflammasome of BALB/c and 129S macrophages swiftly responds to LeTx intoxication with pyroptosis and secretion of interleukin (IL)-1ß. However, human NLRP1 is nonresponsive to LeTx, prompting us to investigate B. anthracis host-pathogen interactions in C57BL/6J (B6) macrophages and mice that also lack a LeTx-sensitive Nlrp1b allele. Unexpectedly, we found that LeTx intoxication and live B. anthracis infection of B6 macrophages elicited robust secretion of IL-1ß, which critically relied on the NLRP3 inflammasome. TNF signaling through both TNF receptor 1 (TNF-R1) and TNF-R2 were required for B. anthracis-induced NLRP3 inflammasome activation, which was further controlled by RIPK1 kinase activity and LeTx-mediated proteolytic inactivation of MAP kinase signaling. In addition to activating the NLRP3 inflammasome, LeTx-induced MAPKK inactivation and TNF production sensitized B. anthracis-infected macrophages to robust RIPK1- and caspase-8-dependent apoptosis. In agreement, purified LeTx triggered RIPK1 kinase activity- and caspase-8-dependent apoptosis only in macrophages primed with TNF or following engagement of TRIF-dependent Toll-like receptors. Consistently, genetic and pharmacological inhibition of RIPK1 inhibited NLRP3 inflammasome activation and apoptosis of LeTx-intoxicated and B. anthracis-infected macrophages. Caspase-8/RIPK3-deficient mice were significantly protected from B. anthracis-induced lethality, demonstrating the in vivo pathophysiological relevance of this cytotoxic mechanism. Collectively, these results establish TNF- and RIPK1 kinase activity-dependent NLRP3 inflammasome activation and macrophage apoptosis as key host-pathogen mechanisms in lethal anthrax.


Asunto(s)
Apoptosis , Bacillus anthracis/metabolismo , Caspasa 8/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Carbunco , Caspasa 8/genética , Interacciones Huésped-Patógeno/fisiología , Inflamasomas/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Piroptosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Transducción de Señal
12.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35105800

RESUMEN

Apoptosis is widely believed to be crucial for epithelial cell death and shedding in the intestine, thereby shaping the overall architecture of the gastrointestinal tract, but also regulating tolerance induction, pinpointing a role of apoptosis intestinal epithelial cell (IEC) turnover and maintenance of barrier function, and in maintaining immune homeostasis. To experimentally address this concept, we generated IEC-specific knockout mice that lack both executioner caspase-3 and caspase-7 (Casp3/7ΔIEC), which are the converging point of the extrinsic and intrinsic apoptotic pathway. Surprisingly, the overall architecture, cellular landscape, and proliferation rate remained unchanged in these mice. However, nonapoptotic cell extrusion was increased in Casp3/7ΔIEC mice, compensating apoptosis deficiency, maintaining the same physiological level of IEC shedding. Microbiome richness and composition stayed unaffected, bearing no sign of dysbiosis. Transcriptome and single-cell RNA sequencing analyses of IECs and immune cells revealed no differences in signaling pathways of differentiation and inflammation. These findings demonstrate that during homeostasis, apoptosis per se is dispensable for IEC turnover at the top of intestinal villi intestinal tissue dynamics, microbiome, and immune cell composition.


Asunto(s)
Apoptosis , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Células Epiteliales/enzimología , Homeostasis , Mucosa Intestinal/enzimología , Transducción de Señal , Animales , Caspasa 3/genética , Caspasa 7/genética , Ratones , Ratones Transgénicos
13.
Immunity ; 43(1): 200-9, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26163370

RESUMEN

Targeted mutagenesis in mice is a powerful tool for functional analysis of genes. However, genetic variation between embryonic stem cells (ESCs) used for targeting (previously almost exclusively 129-derived) and recipient strains (often C57BL/6J) typically results in congenic mice in which the targeted gene is flanked by ESC-derived passenger DNA potentially containing mutations. Comparative genomic analysis of 129 and C57BL/6J mouse strains revealed indels and single nucleotide polymorphisms resulting in alternative or aberrant amino acid sequences in 1,084 genes in the 129-strain genome. Annotating these passenger mutations to the reported genetically modified congenic mice that were generated using 129-strain ESCs revealed that nearly all these mice possess multiple passenger mutations potentially influencing the phenotypic outcome. We illustrated this phenotypic interference of 129-derived passenger mutations with several case studies and developed a Me-PaMuFind-It web tool to estimate the number and possible effect of passenger mutations in transgenic mice of interest.


Asunto(s)
Variación Genética/genética , Genoma/genética , Ratones Endogámicos C57BL/genética , Secuencia de Aminoácidos/genética , Animales , Caspasas/genética , Caspasas Iniciadoras , Mapeo Cromosómico , Hibridación Genómica Comparativa , Conexinas/genética , Genotipo , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 8 de la Matriz/genética , Ratones , Ratones Congénicos/genética , Ratones Noqueados , Mutación/genética , Proteínas del Tejido Nervioso/genética , Polimorfismo de Nucleótido Simple
14.
Cell ; 138(2): 229-32, 2009 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-19632174

RESUMEN

Protein kinases of the receptor interacting protein (RIP) family collaborate with death receptor proteins to regulate cell death. Recent studies (Cho et al., 2009; He et al., 2009; Zhang et al., 2009) reveal that the RIP3 kinase functions with RIP1 at the crossroads of apoptosis, necroptosis, and cell survival.


Asunto(s)
Muerte Celular , Supervivencia Celular , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Humanos , Ratones , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal
15.
Eur Respir J ; 61(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36549711

RESUMEN

BACKGROUND: Receptor-interacting protein kinase 1 (RIPK1) is a key mediator of regulated cell death (including apoptosis and necroptosis) and inflammation, both drivers of COPD pathogenesis. We aimed to define the contribution of RIPK1 kinase-dependent cell death and inflammation in the pathogenesis of COPD. METHODS: We assessed RIPK1 expression in single-cell RNA sequencing (RNA-seq) data from human and mouse lungs, and validated RIPK1 levels in lung tissue of COPD patients via immunohistochemistry. Next, we assessed the consequences of genetic and pharmacological inhibition of RIPK1 kinase activity in experimental COPD, using Ripk1 S25D/S25D kinase-deficient mice and the RIPK1 kinase inhibitor GSK'547. RESULTS: RIPK1 expression increased in alveolar type 1 (AT1), AT2, ciliated and neuroendocrine cells in human COPD. RIPK1 protein levels were significantly increased in airway epithelium of COPD patients compared with never-smokers and smokers without airflow limitation. In mice, exposure to cigarette smoke (CS) increased Ripk1 expression similarly in AT2 cells, and further in alveolar macrophages and T-cells. Genetic and/or pharmacological inhibition of RIPK1 kinase activity significantly attenuated airway inflammation upon acute and subacute CS exposure, as well as airway remodelling, emphysema, and apoptotic and necroptotic cell death upon chronic CS exposure. Similarly, pharmacological RIPK1 kinase inhibition significantly attenuated elastase-induced emphysema and lung function decline. Finally, RNA-seq on lung tissue of CS-exposed mice revealed downregulation of cell death and inflammatory pathways upon pharmacological RIPK1 kinase inhibition. CONCLUSIONS: RIPK1 kinase inhibition is protective in experimental models of COPD and may represent a novel promising therapeutic approach.


Asunto(s)
Enfisema , Enfermedad Pulmonar Obstructiva Crónica , Enfisema Pulmonar , Humanos , Ratones , Animales , Pulmón , Muerte Celular , Inflamación/metabolismo , Ratones Endogámicos C57BL , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
16.
Mol Cell ; 60(1): 63-76, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26344099

RESUMEN

TNF is a master pro-inflammatory cytokine. Activation of TNFR1 by TNF can result in both RIPK1-independent apoptosis and RIPK1 kinase-dependent apoptosis or necroptosis. These cell death outcomes are regulated by two distinct checkpoints during TNFR1 signaling. TNF-mediated NF-κB-dependent induction of pro-survival or anti-apoptotic molecules is a well-known late checkpoint in the pathway, protecting cells from RIPK1-independent death. On the other hand, the molecular mechanism regulating the contribution of RIPK1 to cell death is far less understood. We demonstrate here that the IKK complex phosphorylates RIPK1 at TNFR1 complex I and protects cells from RIPK1 kinase-dependent death, independent of its function in NF-κB activation. We provide in vitro and in vivo evidence that inhibition of IKKα/IKKß or its upstream activators sensitizes cells to death by inducing RIPK1 kinase-dependent apoptosis or necroptosis. We therefore report on an unexpected, NF-κB-independent role for the IKK complex in protecting cells from RIPK1-dependent death downstream of TNFR1.


Asunto(s)
Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Caspasa 8/metabolismo , Muerte Celular , Línea Celular , Embrión de Mamíferos/citología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Fosforilación , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología
17.
Proc Natl Acad Sci U S A ; 117(33): 19982-19993, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32753382

RESUMEN

The underlying mechanism of necroptosis in relation to cancer is still unclear. Here, MYC, a potent oncogene, is an antinecroptotic factor that directly suppresses the formation of the RIPK1-RIPK3 complex. Gene set enrichment analyses reveal that the MYC pathway is the most prominently down-regulated signaling pathway during necroptosis. Depletion or deletion of MYC promotes the RIPK1-RIPK3 interaction, thereby stabilizing the RIPK1 and RIPK3 proteins and facilitating necroptosis. Interestingly, MYC binds to RIPK3 in the cytoplasm and inhibits the interaction between RIPK1 and RIPK3 in vitro. Furthermore, MYC-nick, a truncated form that is mainly localized in the cytoplasm, prevented TNF-induced necroptosis. Finally, down-regulation of MYC enhances necroptosis in leukemia cells and suppresses tumor growth in a xenograft model upon treatment with birinapant and emricasan. MYC-mediated suppression of necroptosis is a mechanism of necroptosis resistance in cancer, and approaches targeting MYC to induce necroptosis represent an attractive therapeutic strategy for cancer.


Asunto(s)
Leucemia/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Leucemia/genética , Leucemia/fisiopatología , Ratones , Ratones Endogámicos BALB C , Necroptosis , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-myc/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal
18.
Int J Mol Sci ; 24(4)2023 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-36835566

RESUMEN

Circulating monocytes are recruited in damaged tissues to generate macrophages that modulate disease progression. Colony-stimulating factor-1 (CSF-1) promotes the generation of monocyte-derived macrophages, which involves caspase activation. Here, we demonstrate that activated caspase-3 and caspase-7 are located to the vicinity of the mitochondria in CSF1-treated human monocytes. Active caspase-7 cleaves p47PHOX at aspartate 34, which promotes the formation of the NADPH (nicotinamide adenine dinucleotide phosphate) oxidase complex NOX2 and the production of cytosolic superoxide anions. Monocyte response to CSF-1 is altered in patients with a chronic granulomatous disease, which are constitutively defective in NOX2. Both caspase-7 down-regulation and radical oxygen species scavenging decrease the migration of CSF-1-induced macrophages. Inhibition or deletion of caspases prevents the development of lung fibrosis in mice exposed to bleomycin. Altogether, a non-conventional pathway that involves caspases and activates NOX2 is involved in CSF1-driven monocyte differentiation and could be therapeutically targeted to modulate macrophage polarization in damaged tissues.


Asunto(s)
Caspasas , Factor Estimulante de Colonias de Macrófagos , Humanos , Animales , Ratones , Factor Estimulante de Colonias de Macrófagos/metabolismo , Caspasa 7/metabolismo , Caspasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Macrófagos/metabolismo , NADPH Oxidasas/metabolismo , Monocitos/metabolismo
19.
Immunity ; 38(2): 209-23, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23438821

RESUMEN

Regulated necrosis, termed necroptosis, is negatively regulated by caspase-8 and is dependent on the kinase activity of RIPK1 and RIPK3. Necroptosis leads to rapid plasma membrane permeabilization and to the release of cell contents and exposure of damage-associated molecular patterns (DAMPs). We are only beginning to identify the necroptotic DAMPs, their modifications, and their potential role in the regulation of inflammation. In this review, we discuss the physiological relevance of necroptosis and its role in the modulation of inflammation. For example, during viral infection, RIPK3-mediated necroptosis acts as a backup mechanism to clear pathogens. Necroptosis is also involved in apparently immunologically silent maintenance of T cell homeostasis. In contrast, the induction of necroptosis in skin, intestine, systemic inflammatory response syndrome, and ischemia reperfusion injury provoke a strong inflammatory response, which might be triggered by emission of DAMPs from necroptotic cells, showing the detrimental side of necroptosis.


Asunto(s)
Infecciones Bacterianas/metabolismo , Inflamación/metabolismo , Necrosis/metabolismo , Daño por Reperfusión/metabolismo , Virosis/metabolismo , Animales , Apoptosis/genética , Apoptosis/inmunología , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/fisiopatología , Caspasa 8/genética , Caspasa 8/inmunología , Humanos , Inflamación/inmunología , Inflamación/fisiopatología , Necrosis/inmunología , Necrosis/fisiopatología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Receptores de Reconocimiento de Patrones/genética , Receptores de Reconocimiento de Patrones/inmunología , Daño por Reperfusión/inmunología , Daño por Reperfusión/fisiopatología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Virosis/inmunología , Virosis/fisiopatología , Virus/crecimiento & desarrollo
20.
Nat Rev Mol Cell Biol ; 11(10): 700-14, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20823910

RESUMEN

For a long time, apoptosis was considered the sole form of programmed cell death during development, homeostasis and disease, whereas necrosis was regarded as an unregulated and uncontrollable process. Evidence now reveals that necrosis can also occur in a regulated manner. The initiation of programmed necrosis, 'necroptosis', by death receptors (such as tumour necrosis factor receptor 1) requires the kinase activity of receptor-interacting protein 1 (RIP1; also known as RIPK1) and RIP3 (also known as RIPK3), and its execution involves the active disintegration of mitochondrial, lysosomal and plasma membranes. Necroptosis participates in the pathogenesis of diseases, including ischaemic injury, neurodegeneration and viral infection, thereby representing an attractive target for the avoidance of unwarranted cell death.


Asunto(s)
Apoptosis/fisiología , Muerte Celular/fisiología , Receptores de Muerte Celular/fisiología , Animales , Membrana Celular/patología , Membrana Celular/fisiología , Humanos , Lisosomas/patología , Lisosomas/fisiología , Macrófagos/microbiología , Macrófagos/patología , Mitocondrias/patología , Mitocondrias/fisiología , Morfogénesis/fisiología , Necrosis , Proteínas Quinasas/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Shigella flexneri/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA