Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 162(6): 1365-78, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26359988

RESUMEN

The cytokine TWEAK and its cognate receptor Fn14 are members of the TNF/TNFR superfamily and are upregulated in tumors. We found that Fn14, when expressed in tumors, causes cachexia and that antibodies against Fn14 dramatically extended lifespan by inhibiting tumor-induced weight loss although having only moderate inhibitory effects on tumor growth. Anti-Fn14 antibodies prevented tumor-induced inflammation and loss of fat and muscle mass. Fn14 signaling in the tumor, rather than host, is responsible for inducing this cachexia because tumors in Fn14- and TWEAK-deficient hosts developed cachexia that was comparable to that of wild-type mice. These results extend the role of Fn14 in wound repair and muscle development to involvement in the etiology of cachexia and indicate that Fn14 antibodies may be a promising approach to treat cachexia, thereby extending lifespan and improving quality of life for cancer patients.


Asunto(s)
Caquexia/tratamiento farmacológico , Neoplasias/patología , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/administración & dosificación , Atrofia/tratamiento farmacológico , Caquexia/patología , Muerte Celular , Neoplasias del Colon/tratamiento farmacológico , Citocina TWEAK , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Desarrollo de Músculos , Neoplasias/metabolismo , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/metabolismo , Alineación de Secuencia , Transducción de Señal , Receptor de TWEAK , Factores de Necrosis Tumoral/metabolismo
2.
Nature ; 609(7927): 590-596, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36002575

RESUMEN

Bacterial cell wall components provide various unique molecular structures that are detected by pattern recognition receptors (PRRs) of the innate immune system as non-self. Most bacterial species form a cell wall that consists of peptidoglycan (PGN), a polymeric structure comprising alternating amino sugars that form strands cross-linked by short peptides. Muramyl dipeptide (MDP) has been well documented as a minimal immunogenic component of peptidoglycan1-3. MDP is sensed by the cytosolic nucleotide-binding oligomerization domain-containing protein 24 (NOD2). Upon engagement, it triggers pro-inflammatory gene expression, and this functionality is of critical importance in maintaining a healthy intestinal barrier function5. Here, using a forward genetic screen to identify factors required for MDP detection, we identified N-acetylglucosamine kinase (NAGK) as being essential for the immunostimulatory activity of MDP. NAGK is broadly expressed in immune cells and has previously been described to contribute to the hexosamine biosynthetic salvage pathway6. Mechanistically, NAGK functions upstream of NOD2 by directly phosphorylating the N-acetylmuramic acid moiety of MDP at the hydroxyl group of its C6 position, yielding 6-O-phospho-MDP. NAGK-phosphorylated MDP-but not unmodified MDP-constitutes an agonist for NOD2. Macrophages from mice deficient in NAGK are completely deficient in MDP sensing. These results reveal a link between amino sugar metabolism and innate immunity to bacterial cell walls.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina , Proteína Adaptadora de Señalización NOD2 , Fosfotransferasas (Aceptor de Grupo Alcohol) , Acetilmuramil-Alanil-Isoglutamina/química , Acetilmuramil-Alanil-Isoglutamina/inmunología , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Acetilmuramil-Alanil-Isoglutamina/farmacología , Animales , Bacterias/química , Bacterias/inmunología , Pared Celular/química , Hexosaminas/biosíntesis , Inmunidad Innata , Macrófagos/enzimología , Macrófagos/inmunología , Ratones , Proteína Adaptadora de Señalización NOD2/agonistas , Proteína Adaptadora de Señalización NOD2/metabolismo , Peptidoglicano/química , Peptidoglicano/inmunología , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo
3.
Nature ; 594(7862): 246-252, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33845483

RESUMEN

The emergence and global spread of SARS-CoV-2 has resulted in the urgent need for an in-depth understanding of molecular functions of viral proteins and their interactions with the host proteome. Several individual omics studies have extended our knowledge of COVID-19 pathophysiology1-10. Integration of such datasets to obtain a holistic view of virus-host interactions and to define the pathogenic properties of SARS-CoV-2 is limited by the heterogeneity of the experimental systems. Here we report a concurrent multi-omics study of SARS-CoV-2 and SARS-CoV. Using state-of-the-art proteomics, we profiled the interactomes of both viruses, as well as their influence on the transcriptome, proteome, ubiquitinome and phosphoproteome of a lung-derived human cell line. Projecting these data onto the global network of cellular interactions revealed crosstalk between the perturbations taking place upon infection with SARS-CoV-2 and SARS-CoV at different levels and enabled identification of distinct and common molecular mechanisms of these closely related coronaviruses. The TGF-ß pathway, known for its involvement in tissue fibrosis, was specifically dysregulated by SARS-CoV-2 ORF8 and autophagy was specifically dysregulated by SARS-CoV-2 ORF3. The extensive dataset (available at https://covinet.innatelab.org ) highlights many hotspots that could be targeted by existing drugs and may be used to guide rational design of virus- and host-directed therapies, which we exemplify by identifying inhibitors of kinases and matrix metalloproteases with potent antiviral effects against SARS-CoV-2.


Asunto(s)
COVID-19/metabolismo , Interacciones Huésped-Patógeno , Proteoma/metabolismo , Proteómica , SARS-CoV-2/patogenicidad , Síndrome Respiratorio Agudo Grave/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Animales , Antivirales/farmacología , Autofagia/efectos de los fármacos , COVID-19/inmunología , COVID-19/virología , Línea Celular , Conjuntos de Datos como Asunto , Evaluación Preclínica de Medicamentos , Interacciones Huésped-Patógeno/inmunología , Humanos , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Fosforilación , Mapas de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional , Proteoma/química , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , SARS-CoV-2/inmunología , Síndrome Respiratorio Agudo Grave/inmunología , Síndrome Respiratorio Agudo Grave/virología , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitinación , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Viroporinas/metabolismo
4.
Immunity ; 45(3): 513-526, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27523270

RESUMEN

The kinases RIPK1 and RIPK3 and the pseudo-kinase MLKL have been identified as key regulators of the necroptotic cell death pathway, although a role for MLKL within the whole animal has not yet been established. Here, we have shown that MLKL deficiency rescued the embryonic lethality caused by loss of Caspase-8 or FADD. Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice were viable and fertile but rapidly developed severe lymphadenopathy, systemic autoimmune disease, and thrombocytopenia. These morbidities occurred more rapidly and with increased severity in Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice compared to Casp8(-/-)Ripk3(-/-) or Fadd(-/-)Ripk3(-/-) mice, respectively. These results demonstrate that MLKL is an essential effector of aberrant necroptosis in embryos caused by loss of Caspase-8 or FADD. Furthermore, they suggest that RIPK3 and/or MLKL may exert functions independently of necroptosis. It appears that non-necroptotic functions of RIPK3 contribute to the lymphadenopathy, autoimmunity, and excess cytokine production that occur when FADD or Caspase-8-mediated apoptosis is abrogated.


Asunto(s)
Apoptosis/fisiología , Enfermedades Autoinmunes/metabolismo , Muerte Celular/fisiología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Caspasa 8/metabolismo , Ratones , Ratones Endogámicos C57BL , Necrosis/metabolismo
5.
PLoS Biol ; 20(5): e3001636, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35576205

RESUMEN

The recent revolution in computational protein structure prediction provides folding models for entire proteomes, which can now be integrated with large-scale experimental data. Mass spectrometry (MS)-based proteomics has identified and quantified tens of thousands of posttranslational modifications (PTMs), most of them of uncertain functional relevance. In this study, we determine the structural context of these PTMs and investigate how this information can be leveraged to pinpoint potential regulatory sites. Our analysis uncovers global patterns of PTM occurrence across folded and intrinsically disordered regions. We found that this information can help to distinguish regulatory PTMs from those marking improperly folded proteins. Interestingly, the human proteome contains thousands of proteins that have large folded domains linked by short, disordered regions that are strongly enriched in regulatory phosphosites. These include well-known kinase activation loops that induce protein conformational changes upon phosphorylation. This regulatory mechanism appears to be widespread in kinases but also occurs in other protein families such as solute carriers. It is not limited to phosphorylation but includes ubiquitination and acetylation sites as well. Furthermore, we performed three-dimensional proximity analysis, which revealed examples of spatial coregulation of different PTM types and potential PTM crosstalk. To enable the community to build upon these first analyses, we provide tools for 3D visualization of proteomics data and PTMs as well as python libraries for data accession and processing.


Asunto(s)
Procesamiento Proteico-Postraduccional , Proteoma , Humanos , Espectrometría de Masas/métodos , Fosforilación , Proteómica/métodos
6.
Mol Cell Proteomics ; 21(9): 100279, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35944843

RESUMEN

Data-independent acquisition (DIA) methods have become increasingly attractive in mass spectrometry-based proteomics because they enable high data completeness and a wide dynamic range. Recently, we combined DIA with parallel accumulation-serial fragmentation (dia-PASEF) on a Bruker trapped ion mobility (IM) separated quadrupole time-of-flight mass spectrometer. This requires alignment of the IM separation with the downstream mass selective quadrupole, leading to a more complex scheme for dia-PASEF window placement compared with DIA. To achieve high data completeness and deep proteome coverage, here we employ variable isolation windows that are placed optimally depending on precursor density in the m/z and IM plane. This is implemented in the freely available py_diAID (Python package for DIA with an automated isolation design) package. In combination with in-depth project-specific proteomics libraries and the Evosep liquid chromatography system, we reproducibly identified over 7700 proteins in a human cancer cell line in 44 min with quadruplicate single-shot injections at high sensitivity. Even at a throughput of 100 samples per day (11 min liquid chromatography gradients), we consistently quantified more than 6000 proteins in mammalian cell lysates by injecting four replicates. We found that optimal dia-PASEF window placement facilitates in-depth phosphoproteomics with very high sensitivity, quantifying more than 35,000 phosphosites in a human cancer cell line stimulated with an epidermal growth factor in triplicate 21 min runs. This covers a substantial part of the regulated phosphoproteome with high sensitivity, opening up for extensive systems-biological studies.


Asunto(s)
Proteoma , Espectrometría de Masas en Tándem , Animales , Cromatografía Liquida/métodos , Factor de Crecimiento Epidérmico , Humanos , Mamíferos/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos
7.
Biochem Soc Trans ; 50(1): 13-20, 2022 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-35166321

RESUMEN

The tumour necrosis factor (TNF) is the most potent inducer of cell death amongst cytokines. It is crucial for processes including homeostasis, the development of the immune system and fighting infections. However, high levels of TNF due to genetic disorders or persistent infections can contribute to autoinflammatory and autoimmune diseases or life-threatening conditions like sepsis. These diseases generally display increased levels of cell death, which, downstream of the TNF receptor, can either be caspase-dependent (apoptosis) or caspase-independent (necroptosis). Significant efforts have been invested in unravelling and manipulating signalling mechanisms regulating these two different types of cell death. Here I discuss how modern proteomic approaches like phosphoproteomics and secretomics provide a novel perspective on this central cytokine and its effect on inflammation and cell survival.


Asunto(s)
Apoptosis , Proteómica , Apoptosis/fisiología , Caspasas , Muerte Celular , Citocinas , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
8.
Proc Natl Acad Sci U S A ; 111(42): 15072-7, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25288762

RESUMEN

Necroptosis is considered to be complementary to the classical caspase-dependent programmed cell death pathway, apoptosis. The pseudokinase Mixed Lineage Kinase Domain-Like (MLKL) is an essential effector protein in the necroptotic cell death pathway downstream of the protein kinase Receptor Interacting Protein Kinase-3 (RIPK3). How MLKL causes cell death is unclear, however RIPK3-mediated phosphorylation of the activation loop in MLKL trips a molecular switch to induce necroptotic cell death. Here, we show that the MLKL pseudokinase domain acts as a latch to restrain the N-terminal four-helix bundle (4HB) domain and that unleashing this domain results in formation of a high-molecular-weight, membrane-localized complex and cell death. Using alanine-scanning mutagenesis, we identified two clusters of residues on opposing faces of the 4HB domain that were required for the 4HB domain to kill cells. The integrity of one cluster was essential for membrane localization, whereas MLKL mutations in the other cluster did not prevent membrane translocation but prevented killing; this demonstrates that membrane localization is necessary, but insufficient, to induce cell death. Finally, we identified a small molecule that binds the nucleotide binding site within the MLKL pseudokinase domain and retards MLKL translocation to membranes, thereby preventing necroptosis. This inhibitor provides a novel tool to investigate necroptosis and demonstrates the feasibility of using small molecules to target the nucleotide binding site of pseudokinases to modulate signal transduction.


Asunto(s)
Apoptosis , Necrosis , Proteínas Quinasas/metabolismo , Adenosina Trifosfato/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Membrana Celular/metabolismo , Activación Enzimática , Concentración 50 Inhibidora , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Fosforilación , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
9.
Eur J Immunol ; 45(10): 2918-26, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26173988

RESUMEN

Humans encode two inflammatory caspases that detect cytoplasmic LPS, caspase-4 and caspase-5. When activated, these trigger pyroptotic cell death and caspase-1-dependent IL-1ß production; however the mechanism underlying this process is not yet confirmed. We now show that a specific NLRP3 inhibitor, MCC950, prevents caspase-4/5-dependent IL-1ß production elicited by transfected LPS. Given that both caspase-4 and caspase-5 can detect cytoplasmic LPS, it is possible that these proteins exhibit some degree of redundancy. Therefore, we generated human monocytic cell lines in which caspase-4 and caspase-5 were genetically deleted either individually or together. We found that the deletion of caspase-4 suppressed cell death and IL-1ß production following transfection of LPS into the cytoplasm, or in response to infection with Salmonella typhimurium. Although deletion of caspase-5 did not confer protection against transfected LPS, cell death and IL-1ß production were reduced after infection with Salmonella. Furthermore, double deletion of caspase-4 and caspase-5 had a synergistic effect in the context of Salmonella infection. Our results identify the NLRP3 inflammasome as the specific platform for IL-1ß maturation, downstream of cytoplasmic LPS detection by caspase-4/5. We also show that both caspase-4 and caspase-5 are functionally important for appropriate responses to intracellular Gram-negative bacteria.


Asunto(s)
Proteínas Portadoras/inmunología , Caspasas Iniciadoras/inmunología , Caspasas/inmunología , Lipopolisacáridos/inmunología , Monocitos/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Línea Celular Tumoral , Humanos , Interleucina-1beta/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR
10.
Blood ; 124(5): 737-49, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-24951427

RESUMEN

Differentiation of naïve CD4(+) T cells into effector (Th1, Th2, and Th17) and induced regulatory (iTreg) T cells requires lineage-specifying transcription factors and epigenetic modifications that allow appropriate repression or activation of gene transcription. The epigenetic silencing of cytokine genes is associated with the repressive H3K27 trimethylation mark, mediated by the Ezh2 or Ezh1 methyltransferase components of the polycomb repressive complex 2 (PRC2). Here we show that silencing of the Ifng, Gata3, and Il10 loci in naïve CD4(+) T cells is dependent on Ezh2. Naïve CD4(+) T cells lacking Ezh2 were epigenetically primed for overproduction of IFN-γ in Th2 and iTreg and IL-10 in Th2 cells. In addition, deficiency of Ezh2 accelerated effector Th cell death via death receptor-mediated extrinsic and intrinsic apoptotic pathways, confirmed in vivo for Ezh2-null IFN-γ-producing CD4(+) and CD8(+) T cells responding to Listeria monocytogenes infection. These findings demonstrate the key role of PRC2/Ezh2 in differentiation and survival of peripheral T cells and reveal potential immunotherapeutic targets.


Asunto(s)
Apoptosis/inmunología , Diferenciación Celular/inmunología , Silenciador del Gen/inmunología , Complejo Represivo Polycomb 2/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Supervivencia Celular/inmunología , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Humanos , Interferón gamma/inmunología , Interleucina-10/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/patología , Masculino , Ratones , Linfocitos T Colaboradores-Inductores/citología
11.
Biochem J ; 471(2): 255-65, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26283547

RESUMEN

The pseudokinase MLKL (mixed lineage kinase domain-like), has recently emerged as a critical component of the necroptosis cell death pathway. Although it is clear that phosphorylation of the activation loop in the MLKL pseudokinase domain by the upstream protein kinase RIPK3 (receptor-interacting protein kinase-3), is crucial to trigger MLKL activation, it has remained unclear whether other phosphorylation events modulate MLKL function. By reconstituting Mlkl(-/-), Ripk3(-/-) and Mlkl(-/-)Ripk3(-/-) cells with MLKL phospho-site mutants, we compared the function of known MLKL phosphorylation sites in regulating necroptosis with three phospho-sites that we identified by MS, Ser(158), Ser(228) and Ser(248). Expression of a phosphomimetic S345D MLKL activation loop mutant-induced stimulus-independent cell death in all knockout cells, demonstrating that RIPK3 phosphorylation of the activation loop of MLKL is sufficient to induce cell death. Cell death was also induced by S228A, S228E and S158A MLKL mutants in the absence of death stimuli, but was most profound in Mlkl(-/-)Ripk3(-/-) double knockout fibroblasts. These data reveal a potential role for RIPK3 as a suppressor of MLKL activation and indicate that phosphorylation can fine-tune the ability of MLKL to induce necroptosis.


Asunto(s)
Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Sustitución de Aminoácidos , Animales , Activación Enzimática/fisiología , Técnicas de Inactivación de Genes , Humanos , Ratones , Mutación Missense , Fosforilación/fisiología , Proteínas Quinasas/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Células U937
12.
Biochem J ; 457(3): 369-77, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24219132

RESUMEN

The pseudokinase MLKL (mixed lineage kinase domain-like) was identified recently as an essential checkpoint in the programmed necrosis or 'necroptosis' cell death pathway. In the present study, we report the crystal structure of the human MLKL pseudokinase domain at 1.7 Å (1 Å=0.1 nm) resolution and probe its nucleotide-binding mechanism by performing structure-based mutagenesis. By comparing the structures and nucleotide-binding determinants of human and mouse MLKL orthologues, the present study provides insights into the evolution of nucleotide-binding mechanisms among pseudokinases and their mechanistic divergence from conventional catalytically active protein kinases.


Asunto(s)
Adenosina Trifosfato/metabolismo , Evolución Molecular , Modelos Moleculares , Proteínas Quinasas/metabolismo , Adenosina Trifosfato/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Sitios de Unión , Cristalografía por Rayos X , Bases de Datos de Proteínas , Humanos , Lisina/química , Ratones , Conformación Molecular , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Ingeniería de Proteínas , Proteínas Quinasas/química , Proteínas Quinasas/genética , Proteínas Quinasas/aislamiento & purificación , Estabilidad Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Dispersión del Ángulo Pequeño , Alineación de Secuencia
13.
Front Cell Dev Biol ; 11: 1132696, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36846584

RESUMEN

Efferocytosis is a process by which phagocytes remove dead or dying cells. It is considered anti-inflammatory, as the removal process reduces potential inflammatory molecules originating from dead cells and results in the reprogramming of macrophages to an anti-inflammatory state. However, engulfment of infected dead cells, deregulated phagocytosis and perturbed digestion of apoptotic bodies induce inflammatory signalling pathways during efferocytosis. The affected inflammatory signalling molecules and the mechanism of activation are largely unknown. I discuss how the choice of dead cell cargo, the type of ingestion, and the digestion efficiency can influence phagocyte programming in the context of disease. I also present the latest findings, highlight knowledge gaps, and propose selected experimental approaches to fill them.

14.
Life Sci Alliance ; 5(4)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35027468

RESUMEN

Anti-TNF therapies are a core anti-inflammatory approach for chronic diseases such as rheumatoid arthritis and Crohn's Disease. Previously, we and others found that TNF blocks the emergence and function of alternative-activated or M2 macrophages involved in wound healing and tissue-reparative functions. Conceivably, anti-TNF drugs could mediate their protective effects in part by an altered balance of macrophage activity. To understand the mechanistic basis of how TNF regulates tissue-reparative macrophages, we used RNAseq, scRNAseq, ATACseq, time-resolved phospho-proteomics, gene-specific approaches, metabolic analysis, and signaling pathway deconvolution. We found that TNF controls tissue-reparative macrophage gene expression in a highly gene-specific way, dependent on JNK signaling via the type 1 TNF receptor on specific populations of alternative-activated macrophages. We further determined that JNK signaling has a profound and broad effect on activated macrophage gene expression. Our findings suggest that TNF's anti-M2 effects evolved to specifically modulate components of tissue and reparative M2 macrophages and TNF is therefore a context-specific modulator of M2 macrophages rather than a pan-M2 inhibitor.


Asunto(s)
Macrófagos , Transcripción Genética , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Inhibidores del Factor de Necrosis Tumoral/farmacología
15.
Nat Commun ; 12(1): 6053, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34663829

RESUMEN

Tumor necrosis factor (TNF) is one of the few cytokines successfully targeted by therapies against inflammatory diseases. However, blocking this well studied and pleiotropic ligand can cause dramatic side-effects. Here, we reason that a systems-level proteomic analysis of TNF signaling could dissect its diverse functions and offer a base for developing more targeted therapies. Therefore, we combine phosphoproteomics time course experiments with subcellular localization and kinase inhibitor analysis to identify functional modules of protein phosphorylation. The majority of regulated phosphorylation events can be assigned to an upstream kinase by inhibiting master kinases. Spatial proteomics reveals phosphorylation-dependent translocations of hundreds of proteins upon TNF stimulation. Phosphoproteome analysis of TNF-induced apoptosis and necroptosis uncovers a key role for transcriptional cyclin-dependent kinase activity to promote cytokine production and prevent excessive cell death downstream of the TNF signaling receptor. This resource of TNF-induced pathways and sites can be explored at http://tnfviewer.biochem.mpg.de/ .


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Proteoma/metabolismo , Transducción de Señal , Células A549 , Apoptosis , Muerte Celular , Línea Celular , Citocinas/metabolismo , Humanos , Necroptosis , Fosforilación , Factor de Necrosis Tumoral alfa/metabolismo , Células U937
16.
Nat Commun ; 12(1): 254, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33431886

RESUMEN

Protein ubiquitination is involved in virtually all cellular processes. Enrichment strategies employing antibodies targeting ubiquitin-derived diGly remnants combined with mass spectrometry (MS) have enabled investigations of ubiquitin signaling at a large scale. However, so far the power of data independent acquisition (DIA) with regards to sensitivity in single run analysis and data completeness have not yet been explored. Here, we develop a sensitive workflow combining diGly antibody-based enrichment and optimized Orbitrap-based DIA with comprehensive spectral libraries together containing more than 90,000 diGly peptides. This approach identifies 35,000 diGly peptides in single measurements of proteasome inhibitor-treated cells - double the number and quantitative accuracy of data dependent acquisition. Applied to TNF signaling, the workflow comprehensively captures known sites while adding many novel ones. An in-depth, systems-wide investigation of ubiquitination across the circadian cycle uncovers hundreds of cycling ubiquitination sites and dozens of cycling ubiquitin clusters within individual membrane protein receptors and transporters, highlighting new connections between metabolism and circadian regulation.


Asunto(s)
Ritmo Circadiano/fisiología , Proteoma/metabolismo , Ubiquitina/metabolismo , Células HEK293 , Humanos , Biblioteca de Péptidos , Proteómica , Reproducibilidad de los Resultados , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitinación
17.
Cell Rep ; 30(4): 1260-1270.e5, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31995763

RESUMEN

The inflammatory functions of the cytokine tumor necrosis factor (TNF) rely on its ability to induce cytokine production and to induce cell death. Caspase-dependent and caspase-independent pathways-apoptosis and necroptosis, respectively-regulate immunogenicity by the release of distinct sets of cellular proteins. To obtain an unbiased, systems-level understanding of this important process, we here applied mass spectrometry-based proteomics to dissect protein release during apoptosis and necroptosis. We report hundreds of proteins released from human myeloid cells in time course experiments. Both cell death types induce receptor shedding, but only apoptotic cells released nucleosome components. Conversely, necroptotic cells release lysosomal components by activating lysosomal exocytosis at early stages of necroptosis-induced membrane permeabilization and show reduced release of conventionally secreted cytokines.


Asunto(s)
Apoptosis , Caspasa 8/metabolismo , Citocinas/metabolismo , Necroptosis , Ácidos Pentanoicos/farmacología , Proteoma/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CCL24/metabolismo , Dipéptidos/farmacología , Exocitosis/efectos de los fármacos , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/metabolismo , Células HEK293 , Histonas/metabolismo , Humanos , Indoles/farmacología , Interleucina-8/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Espectrometría de Masas , Necroptosis/efectos de los fármacos
18.
ACS Chem Biol ; 15(10): 2702-2713, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-32902249

RESUMEN

Necroptosis is an inflammatory form of programmed cell death that has been implicated in various human diseases. Compound 2 is a more potent analogue of the published compound 1 and inhibits necroptosis in human and murine cells at nanomolar concentrations. Several target engagement strategies were employed, including cellular thermal shift assays (CETSA) and diazirine-mediated photoaffinity labeling via a bifunctional photoaffinity probe derived from compound 2. These target engagement studies demonstrate that compound 2 binds to all three necroptotic effector proteins (mixed lineage kinase domain-like protein (MLKL), receptor-interacting serine/threonine protein kinase 1 (RIPK1) and receptor-interacting serine/threonine protein kinase 3 (RIPK3)) at different levels in vitro and in cells. Compound 2 also shows efficacy in vivo in a murine model of systemic inflammatory response syndrome (SIRS).


Asunto(s)
Necroptosis/efectos de los fármacos , Compuestos de Fenilurea/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Sulfonamidas/uso terapéutico , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones Endogámicos C57BL , Compuestos de Fenilurea/metabolismo , Compuestos de Fenilurea/farmacocinética , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Sulfonamidas/metabolismo , Sulfonamidas/farmacocinética , Síndrome de Respuesta Inflamatoria Sistémica/tratamiento farmacológico
19.
Nat Commun ; 11(1): 3150, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32561755

RESUMEN

MLKL is the essential effector of necroptosis, a form of programmed lytic cell death. We have isolated a mouse strain with a single missense mutation, MlklD139V, that alters the two-helix 'brace' that connects the killer four-helix bundle and regulatory pseudokinase domains. This confers constitutive, RIPK3 independent killing activity to MLKL. Homozygous mutant mice develop lethal postnatal inflammation of the salivary glands and mediastinum. The normal embryonic development of MlklD139V homozygotes until birth, and the absence of any overt phenotype in heterozygotes provides important in vivo precedent for the capacity of cells to clear activated MLKL. These observations offer an important insight into the potential disease-modulating roles of three common human MLKL polymorphisms that encode amino acid substitutions within or adjacent to the brace region. Compound heterozygosity of these variants is found at up to 12-fold the expected frequency in patients that suffer from a pediatric autoinflammatory disease, chronic recurrent multifocal osteomyelitis (CRMO).


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Sistema Hematopoyético/patología , Necroptosis/genética , Proteínas Quinasas/genética , Animales , Animales Recién Nacidos , Enfermedades Autoinflamatorias Hereditarias , Humanos , Inflamación/genética , Ratones , Mutación Missense , Osteomielitis/genética , Proteínas Quinasas/metabolismo
20.
Pediatr Cardiol ; 30(6): 771-6, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19357907

RESUMEN

Patients who have undergone a Fontan procedure face an increased risk for thromboembolic complications. This study aimed to evaluate whether thromboelastography, a global whole-blood assay of coagulation, can be used to detect hypercoagulability in pediatric Fontan patients compared with healthy children. This prospective, cross-sectional study investigated 25 Fontan patients and 51 healthy children in three age groups: 1-5 years, 6-10 years, and 11-16 years. Kaolin-activated thromboelastography was performed on citrated samples. No statistically significant differences in thromboelastography parameters were found among the different age groups of the 51 healthy children. None of the 25 Fontan patients demonstrated evidence of hypercoagulability on thromboelastography (95% confidence interval, 0-7%), as defined by two standard deviations above or below the normal mean. The findings suggest that the percentage of Fontan patients demonstrating hypercoagulability on thromboelastography is substantially lower than the reported incidence of thromboembolic complications. Whether thromboelastography could be helpful in predicting patients at increased risk for thromboembolic complications or not still is not known. Further studies comparing the thromboelastography of Fontan patients with the thromboembolic complications of those without Fontan are needed to delineate these issues.


Asunto(s)
Coagulación Sanguínea/fisiología , Procedimiento de Fontan , Cardiopatías Congénitas/sangre , Cuidados Posoperatorios/métodos , Tromboelastografía/métodos , Trombosis/epidemiología , Adolescente , Niño , Preescolar , Estudios Transversales , Femenino , Estudios de Seguimiento , Cardiopatías Congénitas/cirugía , Humanos , Incidencia , Lactante , Masculino , Complicaciones Posoperatorias , Estudios Prospectivos , Valores de Referencia , Trombosis/sangre , Estados Unidos/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA