Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Autophagy ; : 1-34, 2024 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-38442890

RESUMEN

Macroautophagy/autophagy is a complex degradation process with a dual role in cell death that is influenced by the cell types that are involved and the stressors they are exposed to. Ferroptosis is an iron-dependent oxidative form of cell death characterized by unrestricted lipid peroxidation in the context of heterogeneous and plastic mechanisms. Recent studies have shed light on the involvement of specific types of autophagy (e.g. ferritinophagy, lipophagy, and clockophagy) in initiating or executing ferroptotic cell death through the selective degradation of anti-injury proteins or organelles. Conversely, other forms of selective autophagy (e.g. reticulophagy and lysophagy) enhance the cellular defense against ferroptotic damage. Dysregulated autophagy-dependent ferroptosis has implications for a diverse range of pathological conditions. This review aims to present an updated definition of autophagy-dependent ferroptosis, discuss influential substrates and receptors, outline experimental methods, and propose guidelines for interpreting the results.Abbreviation: 3-MA:3-methyladenine; 4HNE: 4-hydroxynonenal; ACD: accidentalcell death; ADF: autophagy-dependentferroptosis; ARE: antioxidant response element; BH2:dihydrobiopterin; BH4: tetrahydrobiopterin; BMDMs: bonemarrow-derived macrophages; CMA: chaperone-mediated autophagy; CQ:chloroquine; DAMPs: danger/damage-associated molecular patterns; EMT,epithelial-mesenchymal transition; EPR: electronparamagnetic resonance; ER, endoplasmic reticulum; FRET: Försterresonance energy transfer; GFP: green fluorescent protein;GSH: glutathione;IF: immunofluorescence; IHC: immunohistochemistry; IOP, intraocularpressure; IRI: ischemia-reperfusion injury; LAA: linoleamide alkyne;MDA: malondialdehyde; PGSK: Phen Green™ SK;RCD: regulatedcell death; PUFAs: polyunsaturated fatty acids; RFP: red fluorescentprotein;ROS: reactive oxygen species; TBA: thiobarbituricacid; TBARS: thiobarbituric acid reactive substances; TEM:transmission electron microscopy.

2.
Nat Cell Biol ; 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38424270

RESUMEN

Ferroptosis, an intricately regulated form of cell death characterized by uncontrolled lipid peroxidation, has garnered substantial interest since this term was first coined in 2012. Recent years have witnessed remarkable progress in elucidating the detailed molecular mechanisms that govern ferroptosis induction and defence, with particular emphasis on the roles of heterogeneity and plasticity. In this Review, we discuss the molecular ecosystem of ferroptosis, with implications that may inform and enable safe and effective therapeutic strategies across a broad spectrum of diseases.

3.
Arthritis Rheumatol ; 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38268500

RESUMEN

OBJECTIVE: The study objective was to assess the role of CCL19+ lymph node stromal cells of the joint-draining popliteal lymph node (pLN) for the development of arthritis. METHODS: CCL19+ lymph node stromal cells were spatiotemporally depleted for five days in the pLN before the onset of collagen-induced arthritis (CIA) using Ccl19-Cre × iDTR mice. In addition, therapeutic treatment with recombinant CCL19-immunoglobulin G (IgG), locally injected in the footpad, was used to confirm the results. RNA sequencing of lymph node stromal cells combined with T cell coculture assays using tropomyosin receptor kinase (Trk) family inhibitors together with in vivo local pLN small interfering RNA (siRNA) treatments were used to elucidate the pathway by which CCL19+ lymph node stromal cells initiate the onset of arthritis. RESULTS: Spatiotemporal depletion of CCL19+ lymph node stromal cells prevented disease onset in CIA mice. These inhibitory effects could be mimicked by local CCL19-IgG treatment. The messenger RNA sequencing analyses showed that CCL19+ lymph node stromal cells down-regulated the expression of the tropomyosin receptor kinase A (TrkA) just before disease onset. Blocking TrkA in lymph node stromal cells led to increased T cell proliferation in in vitro coculture assays. Similar effects were observed with the pan-Trk inhibitor larotrectinib in cocultures of lymph node stromal cells of patients with rheumatoid arthritis and T cells. Finally, local pLN treatment with TrkA inhibitor and TrkA siRNA led to exacerbated arthritis scores. CONCLUSION: CCL19+ lymph node stromal cells are crucially involved in the development of inflammatory arthritis. Therefore, targeting of CCL19+ lymph node stromal cells via TRK could provide a tool to prevent arthritis.

5.
Nat Rev Nephrol ; 19(5): 300-314, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36596919

RESUMEN

Cell death, particularly that of tubule epithelial cells, contributes critically to the pathophysiology of kidney disease. A body of evidence accumulated over the past 15 years has ascribed a central pathophysiological role to a particular form of regulated necrosis, termed necroptosis, to acute tubular necrosis, nephron loss and maladaptive renal fibrogenesis. Unlike apoptosis, which is a non-immunogenic process, necroptosis results in the release of cellular contents and cytokines, which triggers an inflammatory response in neighbouring tissue. This necroinflammatory environment can lead to severe organ dysfunction and cause lasting tissue injury in the kidney. Despite evidence of a link between necroptosis and various kidney diseases, there are no available therapeutic options to target this process. Greater understanding of the molecular mechanisms, triggers and regulators of necroptosis in acute and chronic kidney diseases may identify shortcomings in current approaches to therapeutically target necroptosis regulators and lead to the development of innovative therapeutic approaches.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Renal Crónica , Humanos , Necroptosis , Riñón/metabolismo , Apoptosis , Necrosis/complicaciones , Necrosis/metabolismo , Insuficiencia Renal Crónica/metabolismo , Lesión Renal Aguda/etiología
6.
Biochem Soc Trans ; 50(4): 1197-1205, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-36040212

RESUMEN

The RIP homotypic interaction motif (RHIM) is a conserved protein domain that is approximately 18-22 amino acids in length. In humans, four proteins carrying RHIM domains have been identified: receptor-interacting serine/threonine protein kinase (RIPK) 1, RIPK3, Z-DNA-binding protein 1 (ZBP1), and TIR domain-containing adapter-inducing IFN-ß (TRIF), which are all major players in necroptosis, a distinct form of regulated cell death. Necroptosis is mostly presumed to be a fail-safe form of cell death, occurring in cells in which apoptosis is compromised. Upon activation, RIPK1, ZBP1, and TRIF each hetero-oligomerize with RIPK3 and induce the assembly of an amyloid-like structure of RIPK3 homo-oligomers. These act as docking stations for the recruitment of the pseudokinase mixed-lineage kinase domain like (MLKL), the pore-forming executioner of necroptosis. As RHIM domain interactions are a vital component of the signaling cascade and can also be involved in apoptosis and pyroptosis activation, it is unsurprising that viral and bacterial pathogens have developed means of disrupting RHIM-mediated signaling to ensure survival. Moreover, as these mechanisms play an essential part of regulated cell death signaling, they have received much attention in recent years. Herein, we present the latest insights into the supramolecular structure of interacting RHIM proteins and their distinct signaling cascades in inflammation and infection. Their uncovering will ultimately contribute to the development of new therapeutic strategies in the regulation of lytic cell death.


Asunto(s)
Apoptosis , Necroptosis , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Apoptosis/fisiología , Proteínas Portadoras/metabolismo , Muerte Celular , Humanos , Transducción de Señal
7.
Cells ; 11(14)2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35883577

RESUMEN

Ten years after its initial description, ferroptosis has emerged as the most intensely studied entity among the non-apoptotic forms of regulated cell death. The molecular features of ferroptotic cell death and its functional role have been characterized in vitro and in an ever-growing number of animal studies, demonstrating that it exerts either highly detrimental or, depending on the context, occasionally beneficial effects on the organism. Consequently, two contrary therapeutic approaches are being explored to exploit our detailed understanding of this cell death pathway: the inhibition of ferroptosis to limit organ damage in disorders such as drug-induced toxicity or ischemia-reperfusion injury, and the induction of ferroptosis in cancer cells to ameliorate anti-tumor strategies. However, the path from basic science to clinical utility is rocky. Emphasizing ferroptosis inhibition, we review the success and failures thus far in the translational process from basic research in the laboratory to the treatment of patients.


Asunto(s)
Ferroptosis , Daño por Reperfusión , Animales , Muerte Celular , Daño por Reperfusión/metabolismo
8.
Cell Mol Life Sci ; 79(7): 387, 2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35763128

RESUMEN

Ferroptosis, a type of iron-dependent programmed cell death distinct from apoptosis, necroptosis, and other types of cell death, is characterized by lipid peroxidation, reactive oxygen species production, and mitochondrial dysfunction. Accumulating evidence has highlighted vital roles for ferroptosis in multiple diseases, including acute kidney injury. Therefore, ferroptosis has become a major focus for translational research. However, despite its involvement in pathological conditions, there are no pharmacologic inhibitors of ferroptosis in clinical use. In the context of drug repurposing, a strategy for identifying new uses for approved drugs outside the original medical application, we discovered that vitamin K1 is an efficient inhibitor of ferroptosis. Our findings are strengthened by the fact that the vitamin K antagonist phenprocoumon significantly exacerbated ferroptotic cell death in vitro and also massively worsened the course of acute kidney injury in vivo, which is of utmost clinical importance. We therefore assign vitamin K1 a novel role in preventing ferroptotic cell death in acute tubular necrosis during acute kidney injury. Since the safety, tolerability, pharmacokinetics, and pharmacodynamics of vitamin K1 formulations are well documented, this drug is primed for clinical application, and provides a new strategy for pharmacological control of ferroptosis and diseases associated with this mode of cell death.


Asunto(s)
Lesión Renal Aguda , Ferroptosis , Lesión Renal Aguda/tratamiento farmacológico , Humanos , Hierro/metabolismo , Fenprocumón , Vitamina K 1
9.
Biochem J ; 479(3): 259-272, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35015082

RESUMEN

Murine cytomegalovirus protein M45 contains a RIP homotypic interaction motif (RHIM) that is sufficient to confer protection of infected cells against necroptotic cell death. Mechanistically, the N-terminal region of M45 drives rapid self-assembly into homo-oligomeric amyloid fibrils, and interacts with the endogenous RHIM domains of receptor-interacting serine/threonine protein kinases (RIPK) 1, RIPK3, Z-DNA-binding protein 1, and Toll/interleukin-1 receptor domain-containing adaptor-inducing interferon-ß. Remarkably, all four aforementioned mammalian proteins harbouring such a RHIM domain are key components of inflammatory signalling and regulated cell death (RCD) processes. Immunogenic cell death by regulated necrosis causes extensive tissue damage in a wide range of diseases, including ischaemia reperfusion injury, myocardial infarction, sepsis, stroke, and solid organ transplantation. To harness the cell death suppression properties of M45 protein in a therapeutically usable manner, we developed a synthetic peptide encompassing only the RHIM domain of M45. To trigger delivery of RHIM into target cells, we fused the transactivator protein transduction domain of human immunodeficiency virus 1 to the N-terminus of the peptide. The fused peptide could efficiently penetrate eukaryotic cells, but unexpectedly it eradicated or destroyed all tested cancer cell lines and primary cells irrespective of species without further stimulus through a necrosis-like cell death. Typical inhibitors of different forms of RCD cannot impede this process, which appears to involve a direct disruption of biomembranes. Nevertheless, our finding has potential clinical relevance; reliable induction of a necrotic form of cell death distinct from all known forms of RCD may offer a novel therapeutic approach to combat resistant tumour cells.


Asunto(s)
Productos del Gen tat/química , Productos del Gen tat/metabolismo , Dominios Proteicos , Proteínas Recombinantes de Fusión/metabolismo , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/metabolismo , Transducción de Señal/genética , Proteínas Virales/química , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Amiloide/metabolismo , Animales , Productos del Gen tat/genética , VIH-1/química , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Necroptosis/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Ribonucleótido Reductasas/genética , Células U937 , Proteínas Virales/genética
10.
Cell Metab ; 33(12): 2355-2366.e8, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34847376

RESUMEN

Hexokinases (HK) catalyze the first step of glycolysis limiting its pace. HK2 is highly expressed in gut epithelium, contributes to immune responses, and is upregulated during inflammation. We examined the microbial regulation of HK2 and its impact on inflammation using mice lacking HK2 in intestinal epithelial cells (Hk2ΔIEC). Hk2ΔIEC mice were less susceptible to acute colitis. Analyzing the epithelial transcriptome from Hk2ΔIEC mice during colitis and using HK2-deficient intestinal organoids and Caco-2 cells revealed reduced mitochondrial respiration and epithelial cell death in the absence of HK2. The microbiota strongly regulated HK2 expression and activity. The microbially derived short-chain fatty acid (SCFA) butyrate repressed HK2 expression via histone deacetylase 8 (HDAC8) and reduced mitochondrial respiration in wild-type but not in HK2-deficient Caco-2 cells. Butyrate supplementation protected wild-type but not Hk2ΔIEC mice from colitis. Our findings define a mechanism how butyrate promotes intestinal homeostasis and suggest targeted HK2-inhibition as therapeutic avenue for inflammation.


Asunto(s)
Colitis , Hexoquinasa , Animales , Células CACO-2 , Muerte Celular/fisiología , Colitis/metabolismo , Colitis/microbiología , Células Epiteliales/metabolismo , Hexoquinasa/metabolismo , Histona Desacetilasas/metabolismo , Humanos , Ratones , Mitocondrias/metabolismo , Proteínas Represoras/metabolismo
11.
Cell Death Differ ; 28(5): 1610-1626, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33273695

RESUMEN

The receptor-interacting serine/threonine protein kinase 1 (RIPK1) is a key mediator of regulated cell death and inflammation. Recent studies suggest that RIPK1 inhibition would fundamentally improve the therapy of RIPK1-dependent organ damage in stroke, myocardial infarction, kidney failure, and systemic inflammatory response syndrome. Additionally, it could ameliorate or prevent multi-organ failure induced by cytokine release in the context of hyperinflammation, as seen in COVID-19 patients. Therefore, we searched for a RIPK1 inhibitor and present the aromatic antiepileptic and FDA-approved drug primidone (Liskantin®) as a potent inhibitor of RIPK1 activation in vitro and in a murine model of TNFα-induced shock, which mimics the hyperinflammatory state of cytokine release syndrome. Furthermore, we detected for the first time RIPK1 activation in the respiratory tract epithelium of hospitalized patients who tested positive for SARS-CoV-2 infection. Our data provide a strong rationale for evaluating the drug primidone in conditions of hyperinflammation in humans.


Asunto(s)
COVID-19/enzimología , Primidona/farmacología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , SARS-CoV-2/metabolismo , Animales , COVID-19/patología , Muerte Celular/efectos de los fármacos , Células HEK293 , Células HT29 , Humanos , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Inflamación/patología , Células Jurkat , Ratones , Células 3T3 NIH , Células U937 , Tratamiento Farmacológico de COVID-19
12.
Front Cell Dev Biol ; 8: 817, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32974349

RESUMEN

Necroptosis and pyroptosis are two forms of regulated cell death. They are executed by the proteins mixed-lineage kinase domain-like (MLKL) and gasdermin D (GSDMD), respectively. Once activated by numerous pathways, these proteins form membrane pores that allow the influx and efflux of various ions, proteins, and water, ultimately resulting in the death of the cell. These modalities of cell death are considered highly inflammatory because of the release of inflammatory cytokines and damage-associated molecular patterns, and are thereby not only deleterious for the dying cell itself, but also its environment or the entire organism. The relevance for these processes has been observed in various physiological and pathophysiological conditions, ranging from viral and bacterial infections over autoimmune and chronic inflammatory diseases to ischemic organ damage. In recent years, initial in vitro experiments have shed light on a range of connections between necroptosis and pyroptosis. Initial in vivo studies also indicate that, in many disease models, these two forms of cell death cannot be considered individually, as they demonstrate a complex interaction. In this article, we provide an overview of the currently known structure, pathways of activation, and functions of MLKL and GSDMD. With emerging evidence for an interconnection between necroptosis and pyroptosis in not only in vitro, but also in vivo models of disease, we highlight in particular the clinical relevance of the crosslinks between these two forms of inflammatory cell death and their implications for novel therapeutic strategies in a variety of diseases.

13.
Front Cell Dev Biol ; 7: 19, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30842945

RESUMEN

Necroptosis, initially identified as a backup cell death program when apoptosis is hindered, is a prominent feature in the etiology and progression of many human diseases, such as ischemic injury and sepsis. Receptor-interacting protein kinase 3 (RIPK3) is the cardinal regulator of this cell death modality, recruiting and phosphorylating the executioner mixed lineage kinase domain-like protein (MLKL) to signal necroptosis, which is terminated by a cellular plasma membrane rupture and the leakage of intracellular contents from dying cells. Experimental data to date indicate that RIPK3 and MLKL is the core machinery essential for all necroptotic cell death responses. By using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeat/CRISPR-associated protein 9) technology, we showed that Ripk3 and Mlkl knockout and Ripk3/Mlkl double-knockout in necroptosis-sensitive cell lines extensively block susceptibility to necroptosis, in each case to an indistinguishable degree. In vivo studies using Ripk3- or Mlkl-deficient mice validated kidney ischemia reperfusion injury and high-dose tumor necrosis factor (TNF) availability, as druggable targets in necroptotic-mediated pathologies. Here, we demonstrated that Ripk3 or Mlkl-deficient mice are protected to a similar extent from kidney ischemia reperfusion injury and TNF-induced toxicity. Remarkably, in contrast to each single knockout, Ripk3/Mlkl double-deficient mice did not have appreciable protection from either of the above necroptotic-mediated pathologies. Paradoxically, the double-knockout mice resembled, in each case, the vulnerable wild-type mice, revealing novel complexities in the mechanisms of inflammation-driven diseases, due to aberrant cell death.

14.
Cell Death Differ ; 26(9): 1631-1645, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30442947

RESUMEN

Different forms of regulated cell death-like apoptosis and necroptosis contribute to the pathophysiology of clinical conditions including ischemia-reperfusion injury, myocardial infarction, sepsis, and multiple sclerosis. In particular, the kinase activity of the receptor-interacting serine/threonine protein kinase 1 (RIPK1) is crucial for cell fate in inflammation and cell death. However, despite its involvement in pathological conditions, no pharmacologic inhibitor of RIPK1-mediated cell death is currently in clinical use. Herein, we screened a collection of clinical compounds to assess their ability to modulate RIPK1-mediated cell death. Our small-scale screen identified the anti-epilepsy drug Phenhydan® as a potent inhibitor of death receptor-induced necroptosis and apoptosis. Accordingly, Phenhydan® blocked activation of necrosome formation/activation as well as death receptor-induced NF-κB signaling by influencing the membrane function of cells, such as lipid raft formation, thus exerting an inhibitory effect on pathophysiologic cell death processes. By targeting death receptor signaling, the already FDA-approved Phenhydan® may provide new therapeutic strategies for inflammation-driven diseases caused by aberrant cell death.


Asunto(s)
Apoptosis/efectos de los fármacos , Inflamación/tratamiento farmacológico , Necroptosis/efectos de los fármacos , Fenitoína/farmacología , Animales , Anticonvulsivantes/farmacología , Apoptosis/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Células HT29 , Humanos , Inflamación/genética , Inflamación/patología , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/genética , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/genética , Células 3T3 NIH , Necroptosis/genética , Fenitoína/uso terapéutico , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Receptores de Muerte Celular/antagonistas & inhibidores , Receptores de Muerte Celular/genética , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/genética , Sepsis/tratamiento farmacológico , Sepsis/genética
15.
J Exp Med ; 215(11): 2868-2886, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30254094

RESUMEN

A coding variant of the inflammatory bowel disease (IBD) risk gene ATG16L1 has been associated with defective autophagy and deregulation of endoplasmic reticulum (ER) function. IL-22 is a barrier protective cytokine by inducing regeneration and antimicrobial responses in the intestinal mucosa. We show that ATG16L1 critically orchestrates IL-22 signaling in the intestinal epithelium. IL-22 stimulation physiologically leads to transient ER stress and subsequent activation of STING-dependent type I interferon (IFN-I) signaling, which is augmented in Atg16l1 ΔIEC intestinal organoids. IFN-I signals amplify epithelial TNF production downstream of IL-22 and contribute to necroptotic cell death. In vivo, IL-22 treatment in Atg16l1 ΔIEC and Atg16l1 ΔIEC/Xbp1 ΔIEC mice potentiates endogenous ileal inflammation and causes widespread necroptotic epithelial cell death. Therapeutic blockade of IFN-I signaling ameliorates IL-22-induced ileal inflammation in Atg16l1 ΔIEC mice. Our data demonstrate an unexpected role of ATG16L1 in coordinating the outcome of IL-22 signaling in the intestinal epithelium.


Asunto(s)
Proteínas Relacionadas con la Autofagia/inmunología , Proteínas Portadoras/inmunología , Interleucinas/inmunología , Mucosa Intestinal/inmunología , Proteínas de la Membrana/inmunología , Nucleotidiltransferasas/inmunología , Transducción de Señal/inmunología , Animales , Proteínas Relacionadas con la Autofagia/genética , Células CACO-2 , Proteínas Portadoras/genética , Variación Genética , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Interleucinas/genética , Mucosa Intestinal/patología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Nucleotidiltransferasas/genética , Transducción de Señal/genética , Interleucina-22
16.
Arch Toxicol ; 92(2): 759-775, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28975372

RESUMEN

Reactive oxygen species (ROS)-induced apoptosis has been extensively studied. Increasing evidence suggests that ROS, for instance, induced by hydrogen peroxide (H2O2), might also trigger regulated necrotic cell death pathways. Almost nothing is known about the cell death pathways triggered by tertiary-butyl hydroperoxide (t-BuOOH), a widely used inducer of oxidative stress. The lipid peroxidation products induced by t-BuOOH are involved in the pathophysiology of many diseases, such as cancer, cardiovascular diseases, or diabetes. In this study, we exposed murine fibroblasts (NIH3T3) or human keratinocytes (HaCaT) to t-BuOOH (50 or 200 µM, respectively) which induced a rapid necrotic cell death. Well-established regulators of cell death, i.e., p53, poly(ADP)ribose polymerase-1 (PARP-1), the stress kinases p38 and c-Jun N-terminal-kinases 1/2 (JNK1/2), or receptor-interacting serine/threonine protein kinase 1 (RIPK1) and 3 (RIPK3), were not required for t-BuOOH-mediated cell death. Using the selective inhibitors ferrostatin-1 (1 µM) and liproxstatin-1 (1 µM), we identified ferroptosis, a recently discovered cell death mechanism dependent on iron and lipid peroxidation, as the main cell death pathway. Accordingly, t-BuOOH exposure resulted in a ferrostatin-1- and liproxstatin-1-sensitive increase in lipid peroxidation and cytosolic ROS. Ferroptosis was executed independently from other t-BuOOH-mediated cellular damages, i.e., loss of mitochondrial membrane potential, DNA double-strand breaks, or replication block. H2O2 did not cause ferroptosis at equitoxic concentrations (300 µM) and induced a (1) lower and (2) ferrostatin-1- or liproxstatin-1-insensitive increase in lipid peroxidation. We identify that t-BuOOH and H2O2 produce a different pattern of lipid peroxidation, thereby leading to different cell death pathways and present t-BuOOH as a novel inducer of ferroptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Peroxidación de Lípido , terc-Butilhidroperóxido/toxicidad , Animales , Cardiolipinas/metabolismo , Línea Celular , Supervivencia Celular , Ciclohexilaminas/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Queratinocitos/citología , Potencial de la Membrana Mitocondrial , Ratones , Células 3T3 NIH , Fenilendiaminas/metabolismo , Quinoxalinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Compuestos de Espiro/metabolismo
17.
Cell Rep ; 21(12): 3427-3444, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-29262324

RESUMEN

The caspase activation and recruitment domain (CARD)-based inflammasome sensors NLRP1b and NLRC4 induce caspase-1-dependent pyroptosis independent of the inflammasome adaptor ASC. Here, we show that NLRP1b and NLRC4 trigger caspase-8-mediated apoptosis as an alternative cell death program in caspase-1-/- macrophages and intestinal epithelial organoids (IECs). The caspase-8 adaptor FADD was recruited to ASC specks, which served as cytosolic platforms for caspase-8 activation and NLRP1b/NLRC4-induced apoptosis. We further found that caspase-1 protease activity dominated over scaffolding functions in suppressing caspase-8 activation and induction of apoptosis of macrophages and IECs. Moreover, TLR-induced c-FLIP expression inhibited caspase-8-mediated apoptosis downstream of ASC speck assembly, but did not affect pyroptosis induction by NLRP1b and NLRC4. Moreover, unlike during pyroptosis, NLRP1b- and NLRC4-elicited apoptosis retained alarmins and the inflammasome-matured cytokines interleukin 1ß (IL-1ß) and IL-18 intracellularly. This work identifies critical mechanisms regulating apoptosis induction by the inflammasome sensors NLRP1b and NLRC4 and suggests converting pyroptosis into apoptosis as a paradigm for suppressing inflammation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proteínas de Unión al Calcio/metabolismo , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Piroptosis , Animales , Caspasa 8/metabolismo , Enterocitos/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores Toll-Like/metabolismo
18.
Cell Mol Life Sci ; 74(19): 3631-3645, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28551825

RESUMEN

Ferroptosis is a recently recognized caspase-independent form of regulated cell death that is characterized by the accumulation of lethal lipid ROS produced through iron-dependent lipid peroxidation. Considering that regulation of fatty acid metabolism is responsible for the membrane-resident pool of oxidizable fatty acids that undergo lipid peroxidation in ferroptotic processes, we examined the contribution of the key fatty acid metabolism enzyme, acyl-CoA synthetase long-chain family member 4 (ACSL4), in regulating ferroptosis. By using CRISPR/Cas9 technology, we found that knockout of Acsl4 in ferroptosis-sensitive murine and human cells conferred protection from erastin- and RSL3-induced cell death. In the same cell types, deletion of mixed lineage kinase domain-like (Mlkl) blocked susceptibility to necroptosis, as expected. Surprisingly, these studies also revealed ferroptosis and necroptosis are alternative, in that resistance to one pathway sensitized cells to death via the other pathway. These data suggest a mechanism by which one regulated necrosis pathway compensates for another when either ferroptosis or necroptosis is compromised. We verified the synergistic contributions of ferroptosis and necroptosis to tissue damage during acute organ failure in vivo. Interestingly, in the course of pathophysiological acute ischemic kidney injury, ACSL4 was initially upregulated and its expression level correlated with the severity of tissue damage. Together, our findings reveal ACSL4 to be a reliable biomarker of the emerging cell death modality of ferroptosis, which may also serve as a novel therapeutic target in preventing pathological cell death processes.


Asunto(s)
Lesión Renal Aguda/patología , Muerte Celular , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Animales , Línea Celular , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Eliminación de Gen , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
19.
Cell Rep ; 19(1): 175-187, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28380356

RESUMEN

Necroptosis is a form of regulated necrosis that results in cell death and content release after plasma membrane permeabilization. However, little is known about the molecular events responsible for the disruption of the plasma membrane. Here, we find that early increase in cytosolic calcium in TNF-induced necroptosis is mediated by treatment with a Smac mimetic via the TNF/RIP1/TAK1 survival pathway. This does not require the activation of the necrosome and is dispensable for necroptosis. Necroptosis induced by the activation of TLR3/4 pathways does not trigger early calcium flux. We also demonstrate that necroptotic plasma membrane rupture is mediated by osmotic forces and membrane pores around 4 nm in diameter. This late permeabilization step represents a hallmark in necroptosis execution that is cell and treatment independent and requires the RIP1/RIP3/MLKL core. In support of this, treatment with osmoprotectants reduces cell damage in an in vivo necroptosis model of ischemia-reperfusion injury.


Asunto(s)
Apoptosis , Calcio/metabolismo , Membrana Celular/metabolismo , Necrosis/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Biomimética , Células HEK293 , Células HT29 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Proteínas Mitocondriales/metabolismo , Células 3T3 NIH , Nanoporos , Ósmosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
20.
Cell Mol Life Sci ; 74(1): 173-181, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27535660

RESUMEN

Activated receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain like (MLKL) are essential components of the necroptotic pathway. Phosphorylated MLKL (pMLKL) is thought to induce membrane leakage, leading to cell swelling and disintegration of the cell membrane. However, the molecular identity of the necroptotic membrane pore remains unclear, and the role of pMLKL for membrane permeabilization is currently disputed. We observed earlier that the phospholipid scramblase and ion channel TMEM16F/anoctamin 6 cause large membrane currents, cell swelling, and cell death when activated by a strong increase in intracellular Ca2+. We, therefore, asked whether TMEM16F is also central to necroptotic cell death and other cellular events during necroptosis. Necroptosis was induced by TNFα, smac mimetic, and Z-VAD (TSZ) in NIH3T3 fibroblasts and the four additional cell lines HT29, 16HBE, H441, and L929. Time-dependent changes in intracellular Ca2+, cell morphology, and membrane currents were recorded. TSZ induced a small and only transient oscillatory rise in intracellular Ca2+, which was paralleled by the activation of outwardly rectifying Cl- currents, which were typical for TMEM16F/ANO6. Ca2+ oscillations were due to Ca2+ release from endoplasmic reticulum, and were independent of extracellular Ca2+. The initial TSZ-induced cell swelling was followed by cell shrinkage. Using typical channel blockers and siRNA-knockdown, the Cl- currents were shown to be due to the activation of ANO6. However, the knockdown of ANO6 or inhibitors of ANO6 did not inhibit necroptotic cell death. The present data demonstrate the activation of ANO6 during necroptosis, which, however, is not essential for cell death.


Asunto(s)
Señalización del Calcio , Membrana Celular/metabolismo , Necrosis/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Animales , Anoctaminas , Calcio/metabolismo , Línea Celular , Ratones , Células 3T3 NIH , Necrosis/patología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA