Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
Mol Metab ; : 101988, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-39004142

RESUMEN

OBJECTIVE: Receptor-interacting protein kinase 1 (RIPK1) orchestrates the decision between cell survival and cell death in response to tumor necrosis factor (TNF) and other cytokines. Whereas the scaffolding function of RIPK1 is crucial to prevent TNF-induced apoptosis and necroptosis, its kinase activity is required for necroptosis and partially for apoptosis. Although TNF is a proinflammatory cytokine associated with ß-cell loss in diabetes, the mechanism by which TNF induces ß-cell demise remains unclear. METHODS: Here, we dissected the contribution of RIPK1 scaffold versus kinase functions to ß-cell death regulation using mice lacking RIPK1 specifically in ß-cells (Ripk1ß-KO mice) or expressing a kinase-dead version of RIPK1 (Ripk1D138N mice), respectively. These mice were challenged with streptozotocin, a model of autoimmune diabetes. Moreover, Ripk1ß-KO mice were further challenged with a high-fat diet to induce hyperglycemia. For mechanistic studies, pancreatic islets were subjected to various killing and sensitising agents. RESULTS: Inhibition of RIPK1 kinase activity (Ripk1D138N mice) did not affect the onset and progression of hyperglycemia in a type 1 diabetes model. Moreover, the absence of RIPK1 expression in ß-cells did not affect normoglycemia under basal conditions or hyperglycemia under diabetic challenges. Ex vivo, primary pancreatic islets are not sensitised to TNF-induced apoptosis and necroptosis in the absence of RIPK1. Intriguingly, we found that pancreatic islets display high levels of the antiapoptotic cellular FLICE-inhibitory protein (cFLIP) and low levels of apoptosis (Caspase-8) and necroptosis (RIPK3) components. Cycloheximide treatment, which led to a reduction in cFLIP levels, rendered primary islets sensitive to TNF-induced cell death which was fully blocked by caspase inhibition. CONCLUSION: Unlike in many other cell types (e.g., epithelial, and immune), RIPK1 is not required for cell death regulation in ß-cells under physiological conditions or diabetic challenges. Moreover, in vivo and in vitro evidence suggest that pancreatic ß-cells do not undergo necroptosis but mainly caspase-dependent death in response to TNF. Last, our results show that ß-cells have a distinct mode of regulation of TNF-cytotoxicity that is independent of RIPK1 and that may be highly dependent on cFLIP.

2.
Autophagy ; : 1-16, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38873940

RESUMEN

Mesenchymal stem cells (MSCs) are used in cell therapy; nonetheless, their application is limited by their poor survival after transplantation in a proinflammatory microenvironment. Macroautophagy/autophagy activation in MSCs constitutes a stress adaptation pathway, promoting cellular homeostasis. Our proteomics data indicate that RUBCNL/PACER (RUN and cysteine rich domain containing beclin 1 interacting protein like), a positive regulator of autophagy, is also involved in cell death. Hence, we screened MSC survival upon various cell death stimuli under loss or gain of function of RUBCNL. MSCs were protected from TNF (tumor necrosis factor)-induced regulated cell death when RUBCNL was expressed. TNF promotes inflammation by inducing RIPK1 kinase-dependent apoptosis or necroptosis. We determine that MSCs succumb to RIPK1 kinase-dependent apoptosis upon TNF sensing and necroptosis when caspases are inactivated. We show that RUBCNL is a negative regulator of both RIPK1-dependent apoptosis and necroptosis. Furthermore, RUBCNL mutants that lose the ability to regulate autophagy, retain their function in negatively regulating cell death. We also found that RUBCNL forms a complex with RIPK1, which disassembles in response to TNF. In line with this finding, RUBCNL expression limits assembly of RIPK1-TNFRSF1A/TNFR1 complex I, suggesting that complex formation between RUBCNL and RIPK1 represses TNF signaling. These results provide new insights into the crosstalk between the RIPK1-mediated cell death and autophagy machineries and suggest that RUBCNL, due to its functional duality in autophagy and apoptosis/necroptosis, could be targeted to improve the therapeutic efficacy of MSCs. Abbreviations: BAF: bafilomycin A1; CASP3: caspase 3; Caspases: cysteine-aspartic proteases; cCASP3: cleaved CASP3; CQ: chloroquine; CHX: cycloheximide; cPARP: cleaved poly (ADP-ribose) polymerase; DEPs: differential expressed proteins; ETO: etoposide; MEF: mouse embryonic fibroblast; MLKL: mixed lineage kinase domain-like; MSC: mesenchymal stem cell; MTORC1: mechanistic target of rapamycin kinase complex 1; Nec1s: necrostatin 1s; NFKB/NF-kB: nuclear factor of kappa light polypeptide gene enhancer in B cells; PLA: proximity ligation assay; RCD: regulated cell death; RIPK1: receptor (TNFRSF)-interacting serine-threonine kinase 1; RIPK3: receptor-interacting serine-threonine kinase 3; RUBCNL/PACER: RUN and cysteine rich domain containing beclin 1 interacting protein like; siCtrl: small interfering RNA nonsense; siRNA: small interfering RNA; TdT: terminal deoxynucleotidyl transferase; Tm: tunicamycin; TNF: tumor necrosis factor; TNFRSF1A/TNFR1: tumor necrosis factor receptor superfamily, member 1a.

3.
ACS Photonics ; 11(6): 2236-2241, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38911845

RESUMEN

In this work, we present a photonic integrated platform based on buried InGaAs waveguides with InP cladding that operates over a large mid-infrared (mid-IR) spectral range. Thanks to wet-etch fabrication patterning and Fe doping, low propagation losses below 1.2 dB/cm (0.3 cm-1 loss coefficient) have been obtained between 4.6 and 11.2 µm wavelengths (890-1960 cm-1 wavenumber), in both transverse electric (TE) and transverse magnetic (TM) polarization modes. The possibility of monolithically integrating such waveguides with mid-IR sources offers promising perspectives for developing broadband, homogeneously integrated systems.

4.
EMBO Mol Med ; 15(12): e18028, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-38009521

RESUMEN

Tumor endothelial cells (TECs) actively repress inflammatory responses and maintain an immune-excluded tumor phenotype. However, the molecular mechanisms that sustain TEC-mediated immunosuppression remain largely elusive. Here, we show that autophagy ablation in TECs boosts antitumor immunity by supporting infiltration and effector function of T-cells, thereby restricting melanoma growth. In melanoma-bearing mice, loss of TEC autophagy leads to the transcriptional expression of an immunostimulatory/inflammatory TEC phenotype driven by heightened NF-kB and STING signaling. In line, single-cell transcriptomic datasets from melanoma patients disclose an enriched InflammatoryHigh /AutophagyLow TEC phenotype in correlation with clinical responses to immunotherapy, and responders exhibit an increased presence of inflamed vessels interfacing with infiltrating CD8+ T-cells. Mechanistically, STING-dependent immunity in TECs is not critical for the immunomodulatory effects of autophagy ablation, since NF-kB-driven inflammation remains functional in STING/ATG5 double knockout TECs. Hence, our study identifies autophagy as a principal tumor vascular anti-inflammatory mechanism dampening melanoma antitumor immunity.


Asunto(s)
Melanoma , Humanos , Ratones , Animales , Melanoma/patología , Células Endoteliales/metabolismo , Linfocitos T CD8-positivos , FN-kappa B/metabolismo , Autofagia , Inmunoterapia , Microambiente Tumoral
5.
Cell Death Dis ; 14(11): 755, 2023 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-37980412

RESUMEN

Plasma membrane permeabilization (PMP) is a defining feature of regulated necrosis. It allows the extracellular release of damage-associated molecular patterns (DAMPs) that trigger sterile inflammation. The pore forming molecules MLKL and GSDMs drive PMP in necroptosis and pyroptosis, respectively, but the process of PMP remains unclear in many other forms of regulated necrosis. Here, we identified NINJ1 as a crucial regulator of PMP and consequent DAMP release during ferroptosis, parthanatos, H2O2-induced necrosis and secondary necrosis. Importantly, the membrane-permeabilizing function of NINJ1 takes place after the metabolic death of the cells and is independent of the pore-forming molecules MLKL, GSDMD and GSDME. During ferroptosis, NINJ1 acts downstream of lipid peroxidation, which suggested a role for reactive oxygen species (ROS) in NINJ1 activation. Reactive oxygen species were however neither sufficient nor required to trigger NINJ1-dependent PMP. Instead, we found that NINJ1 oligomerization is induced by the swelling of the cell and that its permeabilizing potential still requires an addition, and yet to be discovered, activation mechanism.


Asunto(s)
Apoptosis , Peróxido de Hidrógeno , Humanos , Especies Reactivas de Oxígeno/metabolismo , Peróxido de Hidrógeno/metabolismo , Apoptosis/fisiología , Necrosis/metabolismo , Membrana Celular/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo
6.
Science ; 382(6669): 434-438, 2023 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-37883562

RESUMEN

Synthetic lattices in photonics enable the exploration of light states in new dimensions, transcending phenomena common only to physical space. We propose and demonstrate a quantum walk comb in synthetic frequency space formed by externally modulating a ring-shaped semiconductor laser with ultrafast recovery times. The initially ballistic quantum walk does not dissipate into low supermode states of the synthetic lattice; instead, the state stabilizes in a broad frequency comb, unlocking the full potential of the synthetic frequency lattice. Our device produces a low-noise, nearly flat broadband comb (reaching 100 per centimeter bandwidth) and offers a promising platform to generate broadband, tunable, and stable frequency combs.

7.
PLoS One ; 18(10): e0290241, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37792797

RESUMEN

INTRODUCTION: The Military Physical and Sports Training program was developed by the French Army in order to train, optimize, and maintain individual readiness. Although the health benefits of sport practice do not need to be demonstrated, such activities can cause acute musculoskeletal injuries that need to be addressed. The prevalence of lower limb injury is rather high in the French military population and, in particular, ranges from 15 to 45% during Special Forces selection courses. Thus, this project aims to investigate the efficiency of a body-centered program designed to enhance body awareness. The program seeks to train the mind to actively pay attention to body information, while the latter is viewed as a protective factor against fall injuries. We assume: (i) that postural control can be improved by enhancing the level of body awareness; and (ii) that greater postural awareness could be beneficial in reducing the risk of fall injuries. The body-centered prevention program is based on the Optimization of the Resources of the Armed Forces (ORAF) intervention, which focuses on mental preparation and recovery, and has been deployed in the French Army for many years. METHOD AND ANALYSES: The study focuses on five French Special Forces selection courses (400 soldiers/ participants). It is divided into two stages (year 1, year 2). The first year is dedicated to data collection from the control group (200 participants), while in the second year the ORAF intervention will be deployed. In both year, participants will be subjected to the same enrollment schedule (Fig 3). The main objective is to evaluate the effectiveness of the ORAF intervention in reducing the rate of fall injuries during military selection, based on a multidisciplinary method that captures demographic, biological, biometric, clinical, and para-clinical measures. TRIAL REGISTRATION: Registration number: IDRCB number 2021-A02108-33, Clinical Trial: NCT05451394.


Asunto(s)
Personal Militar , Enfermedades Musculoesqueléticas , Deportes , Humanos , Equilibrio Postural , Encuestas y Cuestionarios
8.
Trends Immunol ; 44(8): 628-643, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37357102

RESUMEN

Tumor necrosis factor (TNF) plays a central role in orchestrating mammalian inflammatory responses. It promotes inflammation either directly by inducing inflammatory gene expression or indirectly by triggering cell death. TNF-mediated cell death-driven inflammation can be beneficial during infection by providing cell-extrinsic signals that help to mount proper immune responses. Uncontrolled cell death caused by TNF is instead highly detrimental and is believed to cause several human autoimmune diseases. Death is not the default response to TNF sensing. Molecular brakes, or cell death checkpoints, actively repress TNF cytotoxicity to protect the organism from its detrimental consequences. These checkpoints therefore constitute essential safeguards against inflammatory diseases. Recent advances in the field have revealed the existence of several new and unexpected brakes against TNF cytotoxicity and pathogenicity.


Asunto(s)
Apoptosis , Transducción de Señal , Animales , Humanos , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Muerte Celular , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Inflamación/patología , Mamíferos
9.
Front Chem ; 11: 1160164, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37090247

RESUMEN

Receptor-Interacting serine/threonine-Protein Kinase 1 (RIPK1) emerged as an important driver of inflammation and, consequently, inflammatory pathologies. The enzymatic activity of RIPK1 is known to indirectly promote inflammation by triggering cell death, in the form of apoptosis, necroptosis and pyroptosis. Small molecule Receptor-Interacting serine/threonine-Protein Kinase 1 inhibitors have therefore recently entered clinical trials for the treatment of a subset of inflammatory pathologies. We previously identified GSK2656157 (GSK'157), a supposedly specific inhibitor of protein kinase R (PKR)-like ER kinase (PERK), as a much more potent type II Receptor-Interacting serine/threonine-Protein Kinase 1 inhibitor. We now performed further structural optimisation on the GSK'157 scaffold in order to develop a novel class of more selective Receptor-Interacting serine/threonine-Protein Kinase 1 inhibitors. Based on a structure-activity relationship (SAR) reported in the literature, we anticipated that introducing a substituent on the para-position of the pyridinyl ring would decrease the interaction with PERK. Herein, we report a series of novel GSK'157 analogues with different para-substituents with increased selectivity for Receptor-Interacting serine/threonine-Protein Kinase 1. The optimisation led to UAMC-3861 as the best compound of this series in terms of activity and selectivity for Receptor-Interacting serine/threonine-Protein Kinase 1 over PERK. The most selective compounds were screened in vitro for their ability to inhibit RIPK1-dependent apoptosis and necroptosis. With this work, we successfully synthesised a novel series of potent and selective type II Receptor-Interacting serine/threonine-Protein Kinase 1 inhibitors based on the GSK'157 scaffold.

10.
Autophagy ; 19(9): 2585-2589, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37014272

RESUMEN

The (macro)autophagy field is facing a paradigm shift after the recent discovery that cytosolic cargoes can still be selectively targeted to phagophores (the precursors to autophagosomes) even in the absence of LC3 or other Atg8-protein family members. Several in vitro studies have indeed reported on the existence of an unconventional selective autophagic pathway that involves the in-situ formation of an autophagosome around the cargo through the direct selective autophagy receptor-mediated recruitment of RB1CC1/FIP200, thereby bypassing the requirement of LC3. In an article recently published in Science, we demonstrate the physiological importance of this unconventional autophagic pathway in the context of TNF (tumor necrosis factor) signaling. We show that it promotes the degradation of the cytotoxic TNFRSF1A/TNFR1 (TNF receptor superfamily member 1A) complex II that assembles upon TNF sensing and thereby protects mice from TNFRSF1A-driven embryonic lethality and skin inflammation.Abbreviations: ATG: autophagy related; CASP: caspase; FIR: RB1CC1/FIP200-interacting region; LIR: LC3-interacting region; M1: linear; PAS: phagophore assembly site; PtdIns3K: phosphatidylinositol 3-kinase; TNF: tumor necrosis factor; TNFRSF1A: TNF receptor superfamily member 1A.


Asunto(s)
Autofagosomas , Autofagia , Ratones , Animales , Autofagia/fisiología , Autofagosomas/metabolismo , Familia de las Proteínas 8 Relacionadas con la Autofagia/metabolismo , Factores de Necrosis Tumoral/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo
11.
Cell Death Differ ; 30(5): 1097-1154, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37100955

RESUMEN

Apoptosis is a form of regulated cell death (RCD) that involves proteases of the caspase family. Pharmacological and genetic strategies that experimentally inhibit or delay apoptosis in mammalian systems have elucidated the key contribution of this process not only to (post-)embryonic development and adult tissue homeostasis, but also to the etiology of multiple human disorders. Consistent with this notion, while defects in the molecular machinery for apoptotic cell death impair organismal development and promote oncogenesis, the unwarranted activation of apoptosis promotes cell loss and tissue damage in the context of various neurological, cardiovascular, renal, hepatic, infectious, neoplastic and inflammatory conditions. Here, the Nomenclature Committee on Cell Death (NCCD) gathered to critically summarize an abundant pre-clinical literature mechanistically linking the core apoptotic apparatus to organismal homeostasis in the context of disease.


Asunto(s)
Apoptosis , Caspasas , Animales , Humanos , Apoptosis/genética , Muerte Celular , Caspasas/genética , Caspasas/metabolismo , Carcinogénesis , Mamíferos/metabolismo
12.
Mol Metab ; 69: 101681, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36707047

RESUMEN

OBJECTIVES: Type 1 diabetes (T1D) is caused by progressive immune-mediated loss of insulin-producing ß-cells. Inflammation is detrimental to ß-cell function and survival, moreover, both apoptosis and necrosis have been implicated as mechanisms of ß-cell loss in T1D. The receptor interacting serine/threonine protein kinase 1 (RIPK1) promotes inflammation by serving as a scaffold for NF-κB and MAPK activation, or by acting as a kinase that triggers apoptosis or necroptosis. It is unclear whether RIPK1 kinase activity is involved in T1D pathology. In the present study, we investigated if absence of RIPK1 activation would affect the susceptibility to immune-mediated diabetes or diet induced obesity (DIO). METHODS: The RIPK1 knockin mouse line carrying a mutation mimicking serine 25 phosphorylation (Ripk1S25D/S25D), which abrogates RIPK1 kinase activity, was utilized to assess the in vivo role of RIPK1 in immune-mediated diabetes or diet induced obesity (DIO). In vitro, ß-cell death and RIPK1 kinase activity was analysed in conditions known to induce RIPK1-dependent apoptosis/necroptosis. RESULTS: We demonstrate that Ripk1S25D/S25D mice presented normal glucose metabolism and ß-cell function. Furthermore, immune-mediated diabetes and DIO were not different between Ripk1S25D/S25D and Ripk1+/+ mice. Despite strong activation of RIPK1 kinase and other necroptosis effectors (RIPK3 and MLKL) by TNF+BV6+zVAD, no cell death was observed in mouse islets nor human ß-cells. CONCLUSION: Our results contrast recent literature showing that most cell types undergo necroptosis following RIPK1 kinase activation. This peculiarity may reflect an adaptation to the inability of ß-cells to proliferate and self-renewal.


Asunto(s)
Diabetes Mellitus Tipo 1 , Proteínas Quinasas , Ratones , Animales , Humanos , Proteínas Quinasas/metabolismo , Inflamación/metabolismo , Serina , Obesidad , Proteína Serina-Treonina Quinasas de Interacción con Receptores
13.
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
14.
Nat Rev Immunol ; 23(5): 289-303, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36380021

RESUMEN

Tumour necrosis factor (TNF) is a central cytokine in inflammatory reactions, and biologics that neutralize TNF are among the most successful drugs for the treatment of chronic inflammatory and autoimmune pathologies. In recent years, it became clear that TNF drives inflammatory responses not only directly by inducing inflammatory gene expression but also indirectly by inducing cell death, instigating inflammatory immune reactions and disease development. Hence, inhibitors of cell death are being considered as a new therapy for TNF-dependent inflammatory diseases.


Asunto(s)
Inflamación , Factor de Necrosis Tumoral alfa , Humanos , Inflamación/tratamiento farmacológico , Citocinas/metabolismo , Muerte Celular
15.
Commun Phys ; 6(1): 53, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38665410

RESUMEN

Fast (sub-second) spectroscopy with high spectral resolution is of vital importance for revealing quantum chemistry kinetics of complex chemical and biological reactions. Fourier transform (FT) spectrometers can achieve high spectral resolution and operate at hundreds of ms time scales in rapid-scan mode. However, the linear translation of a scanning mirror imposes stringent time-resolution limitations to these systems, which makes simultaneous high spectral and temporal resolution very difficult. Here, we demonstrate an FT spectrometer whose operational principle is based on continuous rotational motion of the scanning mirror, effectively decoupling the spectral resolution from the temporal one. Furthermore, we show that such rotational FT spectrometer can perform Mid-IR dual-comb spectroscopy with a single comb source, since the Doppler-shifted version of the comb serves as the second comb. In our realization, we combine the advantages of dual-comb and FT spectroscopy using a single quantum cascade laser frequency comb emitting at 8.2 µm as a light source. Our technique does not require any diffractive or dispersive optical elements and hence preserve the Jacquinot's-, Fellgett's-, and Connes'-advantages of FT spectrometers. By integrating mulitple broadband sources, such system could pave the way for applications where high speed, large optical bandwidth, and high spectral resolution are desired.

16.
Science ; 378(6625): 1201-1207, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36520901

RESUMEN

Cell death induced by tumor necrosis factor (TNF) can be beneficial during infection by helping to mount proper immune responses. However, TNF-induced death can also drive a variety of inflammatory pathologies. Protectives brakes, or cell-death checkpoints, normally repress TNF cytotoxicity to protect the organism from its potential detrimental consequences. Thus, although TNF can kill, this only occurs when one of the checkpoints is inactivated. Here, we describe a checkpoint that prevents apoptosis through the detoxification of the cytotoxic complex IIa that forms upon TNF sensing. We found that autophagy-related 9A (ATG9A) and 200kD FAK family kinase-interacting protein (FIP200) promote the degradation of this complex through a light chain 3 (LC3)-independent lysosomal targeting pathway. This detoxification mechanism was found to counteract TNF receptor 1 (TNFR1)-mediated embryonic lethality and inflammatory skin disease in mouse models.


Asunto(s)
Apoptosis , Proteínas Relacionadas con la Autofagia , Proteínas de la Membrana , Factor de Necrosis Tumoral alfa , Proteínas de Transporte Vesicular , Animales , Ratones , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Dermatitis/genética , Dermatitis/metabolismo , Dermatitis/patología , Modelos Animales de Enfermedad , Pérdida del Embrión/genética , Pérdida del Embrión/metabolismo , Pérdida del Embrión/patología , Lisosomas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
17.
Front Immunol ; 13: 1067164, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36532075

RESUMEN

The Inhibitor of Kappa B Kinase (IKK) complex is a critical regulator of NF-κB activation. More recently, IKK has also been shown to repress RIPK1 dependent extrinsic cell death pathways by directly phosphorylating RIPK1 at serine 25. In T cells, IKK expression is essential for normal development in the thymus, by promoting survival of thymocytes independently of NF-κB activation. RIPK1 undergoes extensive phosphorylation following TNF stimulation in T cells, though which targets are required to repress RIPK1 has not been defined. Here, we show that TNF induced phosphorylation of RIPK1 at S25 is IKK dependent. We test the relevance of this phosphorylation event in T cells using mice with a RIPK1S25D phosphomimetic point mutation to endogenous RIPK1. We find that this mutation protects T cells from TNF induced cell death when IKK activity is inhibited in vitro, and can rescues development of IKK deficient thymocytes in vivo to a degree comparable with kinase dead RIPK1D138N. Together, these data show that phosphorylation of RIPK1S25 by IKK represents a key regulatory event promoting survival of T cells by IKK.


Asunto(s)
FN-kappa B , Serina , Ratones , Animales , Fosforilación , FN-kappa B/metabolismo , Serina/metabolismo , Apoptosis , Factor de Necrosis Tumoral alfa/metabolismo , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Muerte Celular , Timocitos/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
18.
Opt Express ; 30(20): 36087-36095, 2022 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-36258545

RESUMEN

Optical frequency combs based on semiconductor lasers are a promising technology for monolithic integration of dual-comb spectrometers. However, the stabilization of offset frequency fceo remains a challenging feat due the lack of octave-spanning spectra. In a dual-comb configuration, the uncorrelated jitter of the offset frequencies leads to a non-periodic signal resulting in broadened beatnotes with a limited signal-to-noise ratio (SNR). Hence, expensive data acquisition schemes and complex signal processing are currently required. Here, we show that the offset frequencies of two frequency combs can be synchronized by optical injection locking, which allows full phase-stabilization when combined with electrical injection locking of both repetition frequencies frep. A single comb line isolated via an optical Vernier filter serves as Master oscillator for injection locking. The resulting dual-comb signal is periodic and stable over thousands of periods. This enables coherent averaging using analog electronics, which increases the SNR and reduces the data size by one and three orders of magnitude, respectively. The presented method will enable fully phase-stabilized dual-comb spectrometers by leveraging on integrated optical filters and provides access for comparing and stabilizing fceo to narrow-linewidth optical references.

19.
EMBO Rep ; 23(12): e55233, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36194667

RESUMEN

The anti-inflammatory protein A20 serves as a critical brake on NF-κB signaling and NF-κB-dependent inflammation. In humans, polymorphisms in or near the TNFAIP3/A20 gene have been associated with several inflammatory disorders, including rheumatoid arthritis (RA), and experimental studies in mice have demonstrated that myeloid-specific A20 deficiency causes the development of a severe polyarthritis resembling human RA. Myeloid A20 deficiency also promotes osteoclastogenesis in mice, suggesting a role for A20 in the regulation of osteoclast differentiation and bone formation. We show here that osteoclast-specific A20 knockout mice develop severe osteoporosis, but not inflammatory arthritis. In vitro, osteoclast precursor cells from A20 deficient mice are hyper-responsive to RANKL-induced osteoclastogenesis. Mechanistically, we show that A20 is recruited to the RANK receptor complex within minutes of ligand binding, where it restrains NF-κB activation independently of its deubiquitinating activity but through its zinc finger (ZnF) 4 and 7 ubiquitin-binding functions. Together, these data demonstrate that A20 acts as a regulator of RANK-induced NF-κB signaling to control osteoclast differentiation, assuring proper bone development and turnover.


Asunto(s)
FN-kappa B , Humanos , Animales , Ratones
20.
Cell Rep ; 40(7): 111205, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35977488

RESUMEN

Despite its crucial role in initiation of cytotoxic immune responses, the molecular pathways underlying antigen cross-presentation remain incompletely understood. The mechanism of antigen exit from endocytic compartments into the cytosol is a long-standing matter of controversy, confronting two main models: transfer through specific channels/transporters or rupture of endocytic membranes and leakage of luminal content. By monitoring the occurrence of intracellular damage in conventional dendritic cells (cDCs), we show that cross-presenting cDC1s display more frequent endomembrane injuries and increased recruitment of endosomal sorting complex required for transport (ESCRT)-III, the main repair system for intracellular membranes, relative to cDC2s. Silencing of CHMP2a or CHMP4b, two effector subunits of ESCRT-III, enhances cytosolic antigen export and cross-presentation. This phenotype is partially reversed by chemical inhibition of RIPK3, suggesting that endocytic damage is related to basal activation of the necroptosis pathway. Membrane repair therefore proves crucial in containing antigen export to the cytosol and cross-presentation in cDCs.


Asunto(s)
Reactividad Cruzada , Complejos de Clasificación Endosomal Requeridos para el Transporte , Presentación de Antígeno , Antígenos/metabolismo , Citosol/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...