Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.883
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 184(10): 2618-2632.e17, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33836156

RESUMEN

The ongoing pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently affecting millions of lives worldwide. Large retrospective studies indicate that an elevated level of inflammatory cytokines and pro-inflammatory factors are associated with both increased disease severity and mortality. Here, using multidimensional epigenetic, transcriptional, in vitro, and in vivo analyses, we report that topoisomerase 1 (TOP1) inhibition suppresses lethal inflammation induced by SARS-CoV-2. Therapeutic treatment with two doses of topotecan (TPT), an FDA-approved TOP1 inhibitor, suppresses infection-induced inflammation in hamsters. TPT treatment as late as 4 days post-infection reduces morbidity and rescues mortality in a transgenic mouse model. These results support the potential of TOP1 inhibition as an effective host-directed therapy against severe SARS-CoV-2 infection. TPT and its derivatives are inexpensive clinical-grade inhibitors available in most countries. Clinical trials are needed to evaluate the efficacy of repurposing TOP1 inhibitors for severe coronavirus disease 2019 (COVID-19) in humans.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , ADN-Topoisomerasas de Tipo I/metabolismo , SARS-CoV-2/metabolismo , Inhibidores de Topoisomerasa I/farmacología , Topotecan/farmacología , Animales , COVID-19/enzimología , COVID-19/patología , Chlorocebus aethiops , Humanos , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Inflamación/patología , Inflamación/virología , Mesocricetus , Ratones , Ratones Transgénicos , Células THP-1 , Células Vero
2.
Cell ; 184(17): 4447-4463.e20, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34363755

RESUMEN

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


Asunto(s)
Inflamación/enzimología , Proteínas Serina-Treonina Quinasas/deficiencia , Factor de Necrosis Tumoral alfa/farmacología , Células A549 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Autoinmunidad/efectos de los fármacos , Encéfalo/diagnóstico por imagen , Muerte Celular/efectos de los fármacos , Citocinas/metabolismo , Enzima Desubiquitinante CYLD/metabolismo , Femenino , Células HEK293 , Homocigoto , Humanos , Quinasa I-kappa B/metabolismo , Inmunofenotipificación , Inflamación/patología , Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Mutación con Pérdida de Función/genética , Masculino , Linaje , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Receptor Toll-Like 3/metabolismo , Transcriptoma/genética , Vesiculovirus/efectos de los fármacos , Vesiculovirus/fisiología
3.
Cell ; 167(7): 1693-1704, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984721

RESUMEN

Caspases were originally identified as important mediators of inflammatory response and apoptosis. Recent discoveries, however, have unveiled their roles in mediating and suppressing two regulated forms of necrotic cell death, termed pyroptosis and necroptosis, respectively. These recent advances have significantly expanded our understanding of the roles of caspases in regulating development, adult homeostasis, and host defense response.


Asunto(s)
Caspasas/metabolismo , Necrosis/metabolismo , Animales , Apoptosis , Humanos , Infecciones/enzimología , Infecciones/metabolismo , Infecciones/patología , Inflamación/enzimología , Inflamación/metabolismo , Inflamación/patología , Necrosis/enzimología , Piroptosis
4.
Nature ; 627(8005): 865-872, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38509377

RESUMEN

Disease-associated astrocyte subsets contribute to the pathology of neurologic diseases, including multiple sclerosis and experimental autoimmune encephalomyelitis1-8 (EAE), an experimental model for multiple sclerosis. However, little is known about the stability of these astrocyte subsets and their ability to integrate past stimulation events. Here we report the identification of an epigenetically controlled memory astrocyte subset that exhibits exacerbated pro-inflammatory responses upon rechallenge. Specifically, using a combination of single-cell RNA sequencing, assay for transposase-accessible chromatin with sequencing, chromatin immunoprecipitation with sequencing, focused interrogation of cells by nucleic acid detection and sequencing, and cell-specific in vivo CRISPR-Cas9-based genetic perturbation studies we established that astrocyte memory is controlled by the metabolic enzyme ATP-citrate lyase (ACLY), which produces acetyl coenzyme A (acetyl-CoA) that is used by histone acetyltransferase p300 to control chromatin accessibility. The number of ACLY+p300+ memory astrocytes is increased in acute and chronic EAE models, and their genetic inactivation ameliorated EAE. We also detected the pro-inflammatory memory phenotype in human astrocytes in vitro; single-cell RNA sequencing and immunohistochemistry studies detected increased numbers of ACLY+p300+ astrocytes in chronic multiple sclerosis lesions. In summary, these studies define an epigenetically controlled memory astrocyte subset that promotes CNS pathology in EAE and, potentially, multiple sclerosis. These findings may guide novel therapeutic approaches for multiple sclerosis and other neurologic diseases.


Asunto(s)
Astrocitos , Encefalomielitis Autoinmune Experimental , Memoria Epigenética , Esclerosis Múltiple , Animales , Femenino , Humanos , Masculino , Ratones , Acetilcoenzima A/metabolismo , Astrocitos/enzimología , Astrocitos/metabolismo , Astrocitos/patología , ATP Citrato (pro-S)-Liasa/metabolismo , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Sistemas CRISPR-Cas , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Inflamación/enzimología , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Esclerosis Múltiple/enzimología , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Análisis de Expresión Génica de una Sola Célula , Transposasas/metabolismo
5.
Mol Cell ; 82(3): 527-541.e7, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35016033

RESUMEN

Citrulline can be converted into argininosuccinate by argininosuccinate synthetase (ASS1) in the urea cycle and the citrulline-nitric oxide cycle. However, the regulation and biological function of citrulline metabolism remain obscure in the immune system. Unexpectedly, we found that macrophage citrulline declines rapidly after interferon gamma (IFN-γ) and/or lipopolysaccharide (LPS) stimulation, which is required for efficient proinflammatory signaling activation. Mechanistically, IFN-γ and/or LPS stimulation promotes signal transducers and activators of transcription 1 (STAT1)-mediated ASS1 transcription and Janus kinase2 (JAK2)-mediated phosphorylation of ASS1 at tyrosine 87, thereby leading to citrulline depletion. Reciprocally, increased citrulline directly binds to JAK2 and inhibits JAK2-STAT1 signaling. Blockage of ASS1-mediated citrulline depletion suppresses the host defense against bacterial infection in vivo. We therefore define a central role for ASS1 in controlling inflammatory macrophage activation and antibacterial defense through depletion of cellular citrulline and, further, identify citrulline as an innate immune-signaling metabolite that engages a metabolic checkpoint for proinflammatory responses.


Asunto(s)
Argininosuccinato Sintasa/metabolismo , Citrulina/metabolismo , Inmunidad Innata , Inflamación/enzimología , Listeriosis/enzimología , Activación de Macrófagos , Macrófagos/enzimología , Animales , Argininosuccinato Sintasa/genética , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Inflamación/genética , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/genética , Listeriosis/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Células RAW 264.7 , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
6.
Nature ; 623(7985): 122-131, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37722602

RESUMEN

A fundamental and unresolved question in regenerative biology is how tissues return to homeostasis after injury. Answering this question is essential for understanding the aetiology of chronic disorders such as inflammatory bowel diseases and cancer1. We used the Drosophila midgut2 to investigate this and discovered that during regeneration a subpopulation of cholinergic3 neurons triggers Ca2+ currents among intestinal epithelial cells, the enterocytes, to promote return to homeostasis. We found that downregulation of the conserved cholinergic enzyme acetylcholinesterase4 in the gut epithelium enables acetylcholine from specific Egr5 (TNF in mammals)-sensing cholinergic neurons to activate nicotinic receptors in innervated enterocytes. This activation triggers high Ca2+, which spreads in the epithelium through Innexin2-Innexin7 gap junctions6, promoting enterocyte maturation followed by reduction of proliferation and inflammation. Disrupting this process causes chronic injury consisting of ion imbalance, Yki (YAP in humans) activation7, cell death and increase of inflammatory cytokines reminiscent of inflammatory bowel diseases8. Altogether, the conserved cholinergic pathway facilitates epithelial Ca2+ currents that heal the intestinal epithelium. Our findings demonstrate nerve- and bioelectric9-dependent intestinal regeneration and advance our current understanding of how a tissue returns to homeostasis after injury.


Asunto(s)
Señalización del Calcio , Calcio , Neuronas Colinérgicas , Drosophila melanogaster , Enterocitos , Intestinos , Animales , Humanos , Acetilcolina/metabolismo , Acetilcolinesterasa/metabolismo , Calcio/metabolismo , Neuronas Colinérgicas/metabolismo , Drosophila melanogaster/enzimología , Drosophila melanogaster/metabolismo , Enterocitos/metabolismo , Homeostasis , Inflamación/enzimología , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Intestinos/citología , Intestinos/metabolismo , Receptores Nicotínicos/metabolismo , Modelos Animales de Enfermedad
7.
Nature ; 620(7973): 374-380, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37532932

RESUMEN

Low-grade inflammation is a hallmark of old age and a central driver of ageing-associated impairment and disease1. Multiple factors can contribute to ageing-associated inflammation2; however, the molecular pathways that transduce aberrant inflammatory signalling and their impact in natural ageing remain unclear. Here we show that the cGAS-STING signalling pathway, which mediates immune sensing of DNA3, is a critical driver of chronic inflammation and functional decline during ageing. Blockade of STING suppresses the inflammatory phenotypes of senescent human cells and tissues, attenuates ageing-related inflammation in multiple peripheral organs and the brain in mice, and leads to an improvement in tissue function. Focusing on the ageing brain, we reveal that activation of STING triggers reactive microglial transcriptional states, neurodegeneration and cognitive decline. Cytosolic DNA released from perturbed mitochondria elicits cGAS activity in old microglia, defining a mechanism by which cGAS-STING signalling is engaged in the ageing brain. Single-nucleus RNA-sequencing analysis of microglia and hippocampi of a cGAS gain-of-function mouse model demonstrates that engagement of cGAS in microglia is sufficient to direct ageing-associated transcriptional microglial states leading to bystander cell inflammation, neurotoxicity and impaired memory capacity. Our findings establish the cGAS-STING pathway as a driver of ageing-related inflammation in peripheral organs and the brain, and reveal blockade of cGAS-STING signalling as a potential strategy to halt neurodegenerative processes during old age.


Asunto(s)
Envejecimiento , Encéfalo , Disfunción Cognitiva , Inflamación , Proteínas de la Membrana , Enfermedades Neurodegenerativas , Nucleotidiltransferasas , Animales , Humanos , Ratones , Envejecimiento/metabolismo , Envejecimiento/patología , Encéfalo/metabolismo , Encéfalo/patología , Efecto Espectador , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , ADN/inmunología , Inflamación/enzimología , Inflamación/metabolismo , Proteínas de la Membrana/metabolismo , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/metabolismo , Microglía/metabolismo , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/metabolismo , Nucleotidiltransferasas/metabolismo , Especificidad de Órganos , Transducción de Señal , Hipocampo/metabolismo , Hipocampo/patología
8.
Nature ; 609(7926): 348-353, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35978195

RESUMEN

The mammalian immune system uses various pattern recognition receptors to recognize invaders and host damage and transmits this information to downstream immunometabolic signalling outcomes. Laccase domain-containing 1 (LACC1) protein is an enzyme highly expressed in inflammatory macrophages and serves a central regulatory role in multiple inflammatory diseases such as inflammatory bowel diseases, arthritis and clearance of microbial infection1-4. However, the biochemical roles required for LACC1 functions remain largely undefined. Here we elucidated a shared biochemical function of LACC1 in mice and humans, converting L-citrulline to L-ornithine (L-Orn) and isocyanic acid and serving as a bridge between proinflammatory nitric oxide synthase (NOS2) and polyamine immunometabolism. We validated the genetic and mechanistic connections among NOS2, LACC1 and ornithine decarboxylase 1 (ODC1) in mouse models and bone marrow-derived macrophages infected by Salmonella enterica Typhimurium. Strikingly, LACC1 phenotypes required upstream NOS2 and downstream ODC1, and Lacc1-/- chemical complementation with its product L-Orn significantly restored wild-type activities. Our findings illuminate a previously unidentified pathway in inflammatory macrophages, explain why its deficiency may contribute to human inflammatory diseases and suggest that L-Orn could serve as a nutraceutical to ameliorate LACC1-associated immunological dysfunctions such as arthritis or inflammatory bowel disease.


Asunto(s)
Inflamación , Péptidos y Proteínas de Señalización Intracelular , Macrófagos , Óxido Nítrico Sintasa de Tipo II , Animales , Artritis/inmunología , Artritis/metabolismo , Citrulina/metabolismo , Cianatos/metabolismo , Humanos , Inflamación/enzimología , Inflamación/inmunología , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ornitina/metabolismo , Ornitina Descarboxilasa/metabolismo , Poliaminas/metabolismo , Salmonella typhimurium/inmunología
9.
Annu Rev Cell Dev Biol ; 30: 235-54, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25062361

RESUMEN

The rhomboid proteases were first discovered as regulators of Drosophila EGF receptor signaling; soon after, it was recognized that they represented the founder members of a widespread family of intramembrane serine proteases conserved in all kingdoms. More recently still, the family was promoted to a superfamily, encompassing a wide variety of distantly related proteins. One of the surprises has been that many members of the rhomboid-like superfamily are not active proteases. Given the size of this clan, and its relatively recent discovery, there is still much to learn. Nevertheless, we already understand much about how rhomboid proteases perform their surprising function of cleaving transmembrane domains. We also already know that members of the rhomboid-like superfamily participate in biological functions as diverse as growth factor signaling, mitochondrial dynamics, inflammation, parasite invasion, and the machinery of protein quality control. Their potential medical significance is now becoming apparent in several areas.


Asunto(s)
Proteínas de la Membrana/fisiología , Familia de Multigenes , Serina Proteasas/fisiología , Animales , Proteínas Portadoras/fisiología , Dominio Catalítico , Proteínas de Drosophila/fisiología , Humanos , Inflamación/enzimología , Mamíferos/metabolismo , Proteínas de la Membrana/clasificación , Mitocondrias/enzimología , Proteínas Mitocondriales/fisiología , Enfermedades Parasitarias/enzimología , Proteínas de Plantas/fisiología , Proteolisis , Serina Proteasas/clasificación , Terminología como Asunto
10.
Mol Cell ; 73(3): 413-428.e7, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30598363

RESUMEN

Receptor-interacting protein kinase (RIPK) 1 functions as a key mediator of tissue homeostasis via formation of Caspase-8 activating ripoptosome complexes, positively and negatively regulating apoptosis, necroptosis, and inflammation. Here, we report an unanticipated cell-death- and inflammation-independent function of RIPK1 and Caspase-8, promoting faithful chromosome alignment in mitosis and thereby ensuring genome stability. We find that ripoptosome complexes progressively form as cells enter mitosis, peaking at metaphase and disassembling as cells exit mitosis. Genetic deletion and mitosis-specific inhibition of Ripk1 or Caspase-8 results in chromosome alignment defects independently of MLKL. We found that Polo-like kinase 1 (PLK1) is recruited into mitotic ripoptosomes, where PLK1's activity is controlled via RIPK1-dependent recruitment and Caspase-8-mediated cleavage. A fine balance of ripoptosome assembly is required as deregulated ripoptosome activity modulates PLK1-dependent phosphorylation of downstream effectors, such as BUBR1. Our data suggest that ripoptosome-mediated regulation of PLK1 contributes to faithful chromosome segregation during mitosis.


Asunto(s)
Caspasa 8/metabolismo , Inestabilidad Cromosómica , Neoplasias del Colon/enzimología , Fibroblastos/enzimología , Mitosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Aneuploidia , Animales , Apoptosis , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasa 8/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Segregación Cromosómica , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Fibroblastos/patología , Células HT29 , Humanos , Inflamación/enzimología , Inflamación/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal , Quinasa Tipo Polo 1
11.
Development ; 149(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-34523672

RESUMEN

Sustained neutrophilic inflammation is detrimental for cardiac repair and associated with adverse outcomes following myocardial infarction (MI). An attractive therapeutic strategy to treat MI is to reduce or remove infiltrating neutrophils to promote downstream reparative mechanisms. CDK9 inhibitor compounds enhance the resolution of neutrophilic inflammation; however, their effects on cardiac repair/regeneration are unknown. We have devised a cardiac injury model to investigate inflammatory and regenerative responses in larval zebrafish using heartbeat-synchronised light-sheet fluorescence microscopy. We used this model to test two clinically approved CDK9 inhibitors, AT7519 and flavopiridol, examining their effects on neutrophils, macrophages and cardiomyocyte regeneration. We found that AT7519 and flavopiridol resolve neutrophil infiltration by inducing reverse migration from the cardiac lesion. Although continuous exposure to AT7519 or flavopiridol caused adverse phenotypes, transient treatment accelerated neutrophil resolution while avoiding these effects. Transient treatment with AT7519, but not flavopiridol, augmented wound-associated macrophage polarisation, which enhanced macrophage-dependent cardiomyocyte number expansion and the rate of myocardial wound closure. Using cdk9-/- knockout mutants, we showed that AT7519 is a selective CDK9 inhibitor, revealing the potential of such treatments to promote cardiac repair/regeneration.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Flavonoides/farmacología , Miocardio/enzimología , Neutrófilos/enzimología , Piperidinas/farmacología , Pirazoles/farmacología , Regeneración/efectos de los fármacos , Proteínas de Pez Cebra/antagonistas & inhibidores , Animales , Quinasa 9 Dependiente de la Ciclina/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Pez Cebra , Proteínas de Pez Cebra/metabolismo
12.
Nature ; 566(7745): 548-552, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30760924

RESUMEN

Singlet molecular oxygen (1O2) has well-established roles in photosynthetic plants, bacteria and fungi1-3, but not in mammals. Chemically generated 1O2 oxidizes the amino acid tryptophan to precursors of a key metabolite called N-formylkynurenine4, whereas enzymatic oxidation of tryptophan to N-formylkynurenine is catalysed by a family of dioxygenases, including indoleamine 2,3-dioxygenase 15. Under inflammatory conditions, this haem-containing enzyme is expressed in arterial endothelial cells, where it contributes to the regulation of blood pressure6. However, whether indoleamine 2,3-dioxygenase 1 forms 1O2 and whether this contributes to blood pressure control have remained unknown. Here we show that arterial indoleamine 2,3-dioxygenase 1 regulates blood pressure via formation of 1O2. We observed that in the presence of hydrogen peroxide, the enzyme generates 1O2 and that this is associated with the stereoselective oxidation of L-tryptophan to a tricyclic hydroperoxide via a previously unrecognized oxidative activation of the dioxygenase activity. The tryptophan-derived hydroperoxide acts in vivo as a signalling molecule, inducing arterial relaxation and decreasing blood pressure; this activity is dependent on Cys42 of protein kinase G1α. Our findings demonstrate a pathophysiological role for 1O2 in mammals through formation of an amino acid-derived hydroperoxide that regulates vascular tone and blood pressure under inflammatory conditions.


Asunto(s)
Presión Sanguínea/fisiología , Inflamación/sangre , Inflamación/fisiopatología , Oxígeno Singlete/metabolismo , Vasodilatadores/metabolismo , Animales , Línea Celular , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/antagonistas & inhibidores , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/química , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Cisteína/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Peróxido de Hidrógeno/química , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/química , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Inflamación/enzimología , Masculino , Oxidación-Reducción/efectos de los fármacos , Ratas , Transducción de Señal , Oxígeno Singlete/química , Triptófano/química , Triptófano/metabolismo
13.
Proc Natl Acad Sci U S A ; 119(33): e2123097119, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35939695

RESUMEN

Targeting nuclear factor-kappa B (NF-κB) represents a highly viable strategy against chemoresistance in cancers as well as cell death. Ubiquitination, including linear ubiquitination mediated by the linear ubiquitin chain assembly complex (LUBAC), is emerging as a crucial mechanism of overactivated NF-κB signaling. Ovarian tumor family deubiquitinase OTULIN is the only linear linkage-specific deubiquitinase; however, the molecular mechanisms of how it counteracts LUBAC-mediated NF-κB activation have been largely unknown. Here, we identify Lys64/66 of OTULIN for linear ubiquitination facilitated in a LUBAC-dependent manner as a necessary event required for OTULIN-LUBAC interaction under unstressed conditions, which becomes deubiquitinated by OTULIN itself in response to genotoxic stress. Furthermore, this self-deubiquitination of OTULIN occurs intermolecularly, mediated by OTULIN dimerization, resulting in the subsequent dissociation of OTULIN from the LUBAC complex and NF-κB overactivation. Oxidative stress induces OTULIN dimerization via cysteine-mediated covalent disulfide bonds. Our study reveals that the status of the physical interaction between OTULIN and LUBAC is a crucial determining factor for the genotoxic NF-κB signaling, as measured by cell survival and proliferation, while OTULIN loss of function resulting from its dimerization and deubiquitination leads to a dissociation of OTULIN from the LUBAC complex. Of note, similar molecular mechanisms apply to the inflammatory NF-κB signaling in response to tumor necrosis factor α. Hence, a fuller understanding of the detailed molecular mechanisms underlying the disruption of the OTULIN-LUBAC interaction will be instrumental for developing future therapeutic strategies against cancer chemoresistance and necroptotic processes pertinent to numerous human diseases.


Asunto(s)
Daño del ADN , Enzimas Desubicuitinizantes , Endopeptidasas , Inflamación , FN-kappa B , Complejos de Ubiquitina-Proteína Ligasa , Enzimas Desubicuitinizantes/genética , Enzimas Desubicuitinizantes/metabolismo , Resistencia a Antineoplásicos , Endopeptidasas/genética , Endopeptidasas/metabolismo , Humanos , Inflamación/enzimología , Inflamación/genética , FN-kappa B/metabolismo , Necroptosis , Multimerización de Proteína , Transducción de Señal , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ubiquitinación
14.
Blood ; 139(8): 1234-1245, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-34529778

RESUMEN

There is a critical need for cerebro-protective interventions to improve the suboptimal outcomes of patients with ischemic stroke who have been treated with reperfusion strategies. We found that nuclear pyruvate kinase muscle 2 (PKM2), a modulator of systemic inflammation, was upregulated in neutrophils after the onset of ischemic stroke in both humans and mice. Therefore, we determined the role of PKM2 in stroke pathogenesis by using murine models with preexisting comorbidities. We generated novel myeloid cell-specific PKM2-/- mice on wild-type (PKM2fl/flLysMCre+) and hyperlipidemic background (PKM2fl/flLysMCre+Apoe-/-). Controls were littermate PKM2fl/flLysMCre- or PKM2fl/flLysMCre-Apoe-/- mice. Genetic deletion of PKM2 in myeloid cells limited inflammatory response in peripheral neutrophils and reduced neutrophil extracellular traps after cerebral ischemia and reperfusion, suggesting that PKM2 promotes neutrophil hyperactivation in the setting of stroke. In the filament and autologous clot and recombinant tissue plasminogen activator stroke models, irrespective of sex, deletion of PKM2 in myeloid cells in either wild-type or hyperlipidemic mice reduced infarcts and enhanced long-term sensorimotor recovery. Laser speckle imaging revealed improved regional cerebral blood flow in myeloid cell-specific PKM2-deficient mice that was concomitant with reduced post-ischemic cerebral thrombo-inflammation (intracerebral fibrinogen, platelet [CD41+] deposition, neutrophil infiltration, and inflammatory cytokines). Mechanistically, PKM2 regulates post-ischemic inflammation in peripheral neutrophils by promoting STAT3 phosphorylation. To enhance the translational significance, we inhibited PKM2 nuclear translocation using a small molecule and found significantly reduced neutrophil hyperactivation and improved short-term and long-term functional outcomes after stroke. Collectively, these findings identify PKM2 as a novel therapeutic target to improve brain salvage and recovery after reperfusion.


Asunto(s)
Trombosis Intracraneal/enzimología , Accidente Cerebrovascular Isquémico/enzimología , Activación Neutrófila , Neutrófilos/enzimología , Piruvato Quinasa/metabolismo , Animales , Femenino , Inflamación/enzimología , Inflamación/genética , Trombosis Intracraneal/genética , Accidente Cerebrovascular Isquémico/genética , Masculino , Ratones , Ratones Noqueados para ApoE , Piruvato Quinasa/genética
15.
Nature ; 559(7712): 120-124, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29950720

RESUMEN

OTULIN (OTU deubiquitinase with linear linkage specificity) removes linear polyubiquitin from proteins that have been modified by LUBAC (linear ubiquitin chain assembly complex) and is critical for preventing auto-inflammatory disease1,2 and embryonic lethality during mouse development3. Here we show that OTULIN promotes rather than counteracts LUBAC activity by preventing its auto-ubiquitination with linear polyubiquitin. Thus, knock-in mice that express catalytically inactive OTULIN, either constitutively or selectively in endothelial cells, resembled LUBAC-deficient mice4 and died midgestation as a result of cell death mediated by TNFR1 (tumour necrosis factor receptor 1) and the kinase activity of RIPK1 (receptor-interacting protein kinase 1). Inactivation of OTULIN in adult mice also caused pro-inflammatory cell death. Accordingly, embryonic lethality and adult auto-inflammation were prevented by the combined loss of cell death mediators: caspase 8 for apoptosis and RIPK3 for necroptosis. Unexpectedly, OTULIN mutant mice that lacked caspase 8 and RIPK3 died in the perinatal period, exhibiting enhanced production of type I interferon that was dependent on RIPK1. Collectively, our results indicate that OTULIN and LUBAC function in a linear pathway, and highlight a previously unrecognized interaction between linear ubiquitination, regulators of cell death, and induction of type I interferon.


Asunto(s)
Muerte Celular , Enzimas Desubicuitinizantes/metabolismo , Endopeptidasas/metabolismo , Inflamación/metabolismo , Ubiquitina/química , Ubiquitina/metabolismo , Ubiquitinación , Animales , Caspasa 8/genética , Caspasa 8/metabolismo , Muerte Celular/genética , Enzimas Desubicuitinizantes/genética , Pérdida del Embrión/genética , Endopeptidasas/genética , Inflamación/enzimología , Inflamación/genética , Interferón Tipo I/biosíntesis , Ratones , Ratones Endogámicos C57BL , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Ubiquitinación/genética , Pérdida de Peso/genética
16.
Nature ; 555(7698): 673-677, 2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29562231

RESUMEN

Obesity-induced metabolic disease involves functional integration among several organs via circulating factors, but little is known about crosstalk between liver and visceral adipose tissue (VAT). In obesity, VAT becomes populated with inflammatory adipose tissue macrophages (ATMs). In obese humans, there is a close correlation between adipose tissue inflammation and insulin resistance, and in obese mice, blocking systemic or ATM inflammation improves insulin sensitivity. However, processes that promote pathological adipose tissue inflammation in obesity are incompletely understood. Here we show that obesity in mice stimulates hepatocytes to synthesize and secrete dipeptidyl peptidase 4 (DPP4), which acts with plasma factor Xa to inflame ATMs. Silencing expression of DPP4 in hepatocytes suppresses inflammation of VAT and insulin resistance; however, a similar effect is not seen with the orally administered DPP4 inhibitor sitagliptin. Inflammation and insulin resistance are also suppressed by silencing expression of caveolin-1 or PAR2 in ATMs; these proteins mediate the actions of DPP4 and factor Xa, respectively. Thus, hepatocyte DPP4 promotes VAT inflammation and insulin resistance in obesity, and targeting this pathway may have metabolic benefits that are distinct from those observed with oral DPP4 inhibitors.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Hepatocitos/metabolismo , Inflamación/enzimología , Resistencia a la Insulina , Grasa Intraabdominal/patología , Obesidad/enzimología , Administración Oral , Animales , Caveolina 1/deficiencia , Caveolina 1/genética , Caveolina 1/metabolismo , Dipeptidil Peptidasa 4/deficiencia , Dipeptidil Peptidasa 4/genética , Factor Xa/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Humanos , Inflamación/genética , Inflamación/metabolismo , Resistencia a la Insulina/genética , Grasa Intraabdominal/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Obesos , Obesidad/genética , Obesidad/metabolismo , Receptor PAR-2/deficiencia , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Fosfato de Sitagliptina/administración & dosificación , Fosfato de Sitagliptina/farmacología
17.
Nature ; 561(7721): 122-126, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30111836

RESUMEN

Immune recognition of pathogen-associated molecular patterns (PAMPs) by pattern recognition receptors often activates proinflammatory NF-κB signalling1. Recent studies indicate that the bacterial metabolite D-glycero-ß-D-manno-heptose 1,7-bisphosphate (HBP) can activate NF-κB signalling in host cytosol2-4, but it is unclear whether HBP is a genuine PAMP and the cognate pattern recognition receptor has not been identified. Here we combined a transposon screen in Yersinia pseudotuberculosis with biochemical analyses and identified ADP-ß-D-manno-heptose (ADP-Hep), which mediates type III secretion system-dependent NF-κB activation and cytokine expression. ADP-Hep, but not other heptose metabolites, could enter host cytosol to activate NF-κB. A CRISPR-Cas9 screen showed that activation of NF-κB by ADP-Hep involves an ALPK1 (alpha-kinase 1)-TIFA (TRAF-interacting protein with forkhead-associated domain) axis. ADP-Hep directly binds the N-terminal domain of ALPK1, stimulating its kinase domain to phosphorylate and activate TIFA. The crystal structure of the N-terminal domain of ALPK1 and ADP-Hep in complex revealed the atomic mechanism of this ligand-receptor recognition process. HBP was transformed by host adenylyltransferases into ADP-heptose 7-P, which could activate ALPK1 to a lesser extent than ADP-Hep. ADP-Hep (but not HBP) alone or during bacterial infection induced Alpk1-dependent inflammation in mice. Our findings identify ALPK1 and ADP-Hep as a pattern recognition receptor and an effective immunomodulator, respectively.


Asunto(s)
Azúcares de Adenosina Difosfato/inmunología , Burkholderia cenocepacia , Citosol , Inmunidad Innata , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Proteínas Quinasas/metabolismo , Yersinia pseudotuberculosis , Azúcares de Adenosina Difosfato/metabolismo , Animales , Infecciones por Burkholderia/enzimología , Infecciones por Burkholderia/inmunología , Infecciones por Burkholderia/patología , Burkholderia cenocepacia/genética , Burkholderia cenocepacia/inmunología , Burkholderia cenocepacia/metabolismo , Sistemas CRISPR-Cas , Cristalografía por Rayos X , Citocinas/biosíntesis , Citosol/enzimología , Citosol/inmunología , Disacáridos/metabolismo , Activación Enzimática , Femenino , Edición Génica , Factores Inmunológicos/inmunología , Factores Inmunológicos/metabolismo , Inmunomodulación , Inflamación/enzimología , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , FN-kappa B/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/metabolismo
18.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33766915

RESUMEN

Microglial-derived inflammation has been linked to a broad range of neurodegenerative and neuropsychiatric conditions, including amyotrophic lateral sclerosis (ALS). Using single-cell RNA sequencing, a class of Disease-Associated Microglia (DAMs) have been characterized in neurodegeneration. However, the DAM phenotype alone is insufficient to explain the functional complexity of microglia, particularly with regard to regulating inflammation that is a hallmark of many neurodegenerative diseases. Here, we identify a subclass of microglia in mouse models of ALS which we term RIPK1-Regulated Inflammatory Microglia (RRIMs). RRIMs show significant up-regulation of classical proinflammatory pathways, including increased levels of Tnf and Il1b RNA and protein. We find that RRIMs are highly regulated by TNFα signaling and that the prevalence of these microglia can be suppressed by inhibiting receptor-interacting protein kinase 1 (RIPK1) activity downstream of the TNF receptor 1. These findings help to elucidate a mechanism by which RIPK1 kinase inhibition has been shown to provide therapeutic benefit in mouse models of ALS and may provide an additional biomarker for analysis in ongoing phase 2 clinical trials of RIPK1 inhibitors in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Inflamación/enzimología , Microglía/enzimología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Proteínas de Ciclo Celular/genética , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Mutantes , Microglía/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Análisis de la Célula Individual , Superóxido Dismutasa-1/genética , Transcriptoma , Factor de Necrosis Tumoral alfa/metabolismo
19.
Crit Rev Biochem Mol Biol ; 56(4): 321-359, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33843388

RESUMEN

CK2 is a constitutively active protein kinase that assuring a constant level of phosphorylation to its numerous substrates supports many of the most important biological functions. Nevertheless, its activity has to be controlled and adjusted in order to cope with the varying needs of a cell, and several examples of a fine-tune regulation of its activity have been described. More importantly, aberrant regulation of this enzyme may have pathological consequences, e.g. in cancer, chronic inflammation, neurodegeneration, and viral infection. Our review aims at summarizing our current knowledge about CK2 regulation. In the first part, we have considered the most important stimuli shown to affect protein kinase CK2 activity/expression. In the second part, we focus on the molecular mechanisms by which CK2 can be regulated, discussing controversial aspects and future perspectives.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Transducción de Señal , Virosis/enzimología , Animales , Humanos , Inflamación/enzimología
20.
Nat Immunol ; 12(12): 1176-83, 2011 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-22057290

RESUMEN

Chronic inflammation has been strongly associated with tumor progression, but the underlying mechanisms remain elusive. Here we demonstrate that E3 ligase Itch and deubiquitinase Cyld formed a complex via interaction through 'WW-PPXY' motifs. The Itch-Cyld complex sequentially cleaved Lys63-linked ubiquitin chains and catalyzed Lys48-linked ubiquitination on the kinase Tak1 to terminate inflammatory signaling via tumor necrosis factor. Reconstitution of wild-type Cyld but not the mutant Cyld(Y485A), which cannot associate with Itch, blocked sustained Tak1 activation and proinflammatory cytokine production by Cyld(-/-) bone marrow-derived macrophages. Deficiency in Itch or Cyld led to chronic production of tumor-promoting cytokines by tumor-associated macrophages and aggressive growth of lung carcinoma. Thus, we have identified an Itch-Cyld-mediated regulatory mechanism in innate inflammatory cells.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Inflamación/enzimología , Quinasas Quinasa Quinasa PAM/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular Transformada , Línea Celular Tumoral , Cisteína Endopeptidasas/genética , Enzima Desubiquitinante CYLD , Activación Enzimática/genética , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/mortalidad , Neoplasias/patología , Unión Proteica , Alineación de Secuencia , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA