Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nature ; 578(7793): E9, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932732

RESUMEN

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Nature ; 574(7780): 686-690, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31645758

RESUMEN

Dietary habits and vascular risk factors promote both Alzheimer's disease and cognitive impairment caused by vascular factors1-3. Furthermore, accumulation of hyperphosphorylated tau, a microtubule-associated protein and a hallmark of Alzheimer's pathology4, is also linked to vascular cognitive impairment5,6. In mice, a salt-rich diet leads to cognitive dysfunction associated with a nitric oxide deficit in cerebral endothelial cells and cerebral hypoperfusion7. Here we report that dietary salt induces hyperphosphorylation of tau followed by cognitive dysfunction in mice, and that these effects are prevented by restoring endothelial nitric oxide production. The nitric oxide deficiency reduces neuronal calpain nitrosylation and results in enzyme activation, which, in turn, leads to tau phosphorylation by activating cyclin-dependent kinase 5. Salt-induced cognitive impairment is not observed in tau-null mice or in mice treated with anti-tau antibodies, despite persistent cerebral hypoperfusion and neurovascular dysfunction. These findings identify a causal link between dietary salt, endothelial dysfunction and tau pathology, independent of haemodynamic insufficiency. Avoidance of excessive salt intake and maintenance of vascular health may help to stave off the vascular and neurodegenerative pathologies that underlie dementia in the elderly.


Asunto(s)
Disfunción Cognitiva/inducido químicamente , Neuronas/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Proteínas tau/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Disfunción Cognitiva/metabolismo , Humanos , Ratones , Ratones Noqueados , Fosforilación , Cloruro de Sodio Dietético/farmacología
3.
Circ Res ; 130(8): 1145-1166, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35420912

RESUMEN

Stroke remains a leading cause of death and disability, with limited therapeutic options and suboptimal tools for diagnosis and prognosis. High throughput technologies such as proteomics generate large volumes of experimental data at once, thus providing an advanced opportunity to improve the status quo by facilitating identification of novel therapeutic targets and molecular biomarkers. Proteomics studies in animals are largely designed to decipher molecular pathways and targets altered in brain tissue after stroke, whereas studies in human patients primarily focus on biomarker discovery in biofluids and, more recently, in thrombi and extracellular vesicles. Here, we offer a comprehensive review of stroke proteomics studies conducted in both animal and human specimen and present our view on limitations, challenges, and future perspectives in the field. In addition, as a unique resource for the scientific community, we provide extensive lists of all proteins identified in proteomic studies as altered by stroke and perform postanalysis of animal data to reveal stroke-related cellular processes and pathways.


Asunto(s)
Proteómica , Accidente Cerebrovascular , Animales , Biomarcadores , Humanos , Pronóstico , Proteínas , Accidente Cerebrovascular/diagnóstico , Accidente Cerebrovascular/terapia
4.
Cell Mol Life Sci ; 78(5): 2169-2183, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32889561

RESUMEN

Cerebral ischemia-reperfusion increases intraneuronal levels of ubiquitinated proteins, but the factors driving ubiquitination and whether it results from altered proteostasis remain unclear. To address these questions, we used in vivo and in vitro models of cerebral ischemia-reperfusion, in which hippocampal slices were transiently deprived of oxygen and glucose to simulate ischemia followed by reperfusion, or the middle cerebral artery was temporarily occluded in mice. We found that post-ischemic ubiquitination results from two key steps: restoration of ATP at reperfusion, which allows initiation of protein ubiquitination, and free radical production, which, in the presence of sufficient ATP, increases ubiquitination above pre-ischemic levels. Surprisingly, free radicals did not augment ubiquitination through inhibition of the proteasome as previously believed. Although reduced proteasomal activity was detected after ischemia, this was neither caused by free radicals nor sufficient in magnitude to induce appreciable accumulation of proteasomal target proteins or ubiquitin-proteasome reporters. Instead, we found that ischemia-derived free radicals inhibit deubiquitinases, a class of proteases that cleaves ubiquitin chains from proteins, which was sufficient to elevate ubiquitination after ischemia. Our data provide evidence that free radical-dependent deubiquitinase inactivation rather than proteasomal inhibition drives ubiquitination following ischemia-reperfusion, and as such call for a reevaluation of the mechanisms of post-ischemic ubiquitination, previously attributed to altered proteostasis. Since deubiquitinase inhibition is considered an endogenous neuroprotective mechanism to shield proteins from oxidative damage, modulation of deubiquitinase activity may be of therapeutic value to maintain protein integrity after an ischemic insult.


Asunto(s)
Isquemia Encefálica/metabolismo , Enzimas Desubicuitinizantes/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinación , Adenosina Trifosfato/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Ubiquitina/metabolismo
5.
Nucleic Acids Res ; 43(20): 9889-904, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26476452

RESUMEN

Activation of NF-κB-dependent transcription represents an important hallmark of inflammation. While the acute inflammatory response is per se beneficial, it can become deleterious if its spatial and temporal profile is not tightly controlled. Classically, NF-κB activity is limited by cytoplasmic retention of the NF-κB dimer through binding to inhibitory IκB proteins. However, increasing evidence suggests that NF-κB activity can also be efficiently contained by direct ubiquitination of NF-κB subunits. Here, we identify the HECT-domain ubiquitin ligase HERC3 as novel negative regulator of NF-κB activity. We find that HERC3 restricts NF-κB nuclear import and DNA binding without affecting IκBα degradation. Instead HERC3 indirectly binds to the NF-κB RelA subunit after liberation from IκBα inhibitor leading to its ubiquitination and protein destabilization. Remarkably, the regulation of RelA activity by HERC3 is independent of its inherent ubiquitin ligase activity. Rather, we show that HERC3 and RelA are part of a multi-protein complex containing the proteasome as well as the ubiquitin-like protein ubiquilin-1 (UBQLN1). We present evidence that HERC3 and UBQLN1 provide a link between NF-κB RelA and the 26S proteasome, thereby facilitating RelA protein degradation. Our findings establish HERC3 as novel candidate regulating the inflammatory response initiated by NF-κB.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , FN-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo , Transcripción Genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/metabolismo , Dominio Catalítico , Bovinos , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , ADN/metabolismo , Células HEK293 , Humanos , Proteínas I-kappa B/metabolismo , Inhibidor NF-kappaB alfa , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
6.
J Neurosci ; 33(50): 19579-89, 2013 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-24336722

RESUMEN

Loss-of-function mutations of progranulin (PGRN) have been linked to frontotemporal dementia, but little is known about the effects of PGRN deficiency on the brain in health and disease. PGRN has been implicated in neurovascular development, inflammation, and Wnt signaling, a pathway involved in the formation of the blood-brain barrier (BBB). Because BBB alterations and inflammation contribute to ischemic brain injury, we examined the role of PGRN in the brain damage produced by ischemia-reperfusion. PGRN(+/-) and PGRN(-/-) mice underwent middle cerebral artery occlusion (MCAO) with monitoring of cerebral blood flow. Infarct volume and motor deficits were assessed 72 h later. Post-ischemic inflammation was examined by expression of inflammatory genes and flow cytometry. BBB structure and permeability were examined by electron microscopy (EM) and Evans blue (EB) extravasation, respectively. MCAO resulted in ~60% larger infarcts in PGRN(+/-) and PGRN(-/-) mice, an effect independent of hemodynamic factors or post-ischemic inflammation. Rather, massive hemorrhages and post-ischemic BBB disruption were observed, unrelated to degradation of tight junction (TJ) proteins or matrix metalloproteinases (MMPs). By EM, TJ were 30-52% shorter, fewer, and less interlocking, suggesting a weaker seal between endothelial cells. Intracerebral injection of platelet-derived growth factor-CC (PDGF-CC), which increases BBB permeability, resulted in a more severe BBB breakdown in PGRN(+/-) and PGRN(-/-) than wild-type mice. We describe a previously unrecognized involvement of PGRN in the expression of key ultrastructural features of the BBB. Such a novel vasoprotective role of PGRN may contribute to brain dysfunction and damage in conditions associated with reduced PGRN function.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Barrera Hematoencefálica/fisiopatología , Isquemia Encefálica/fisiopatología , Células Endoteliales/metabolismo , Granulinas , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/fisiopatología , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Noqueados , Progranulinas , Daño por Reperfusión/metabolismo , Daño por Reperfusión/fisiopatología , Accidente Cerebrovascular/fisiopatología
7.
J Biol Chem ; 288(1): 285-93, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23100252

RESUMEN

Phosphorylation of NF-κB plays an important role in modulating transcriptional activity of NF-κB independently of inhibitor of κB (IκB) proteins. For the p65 subunit, multiple phosphorylation sites have been mapped in and adjacent to both the N-terminal Rel homology domain and the C-terminal transactivation domain. Their impact on NF-κB-dependent transcription, however, has never been assessed at a broader level. In this study, we evaluate the importance of differential p65 phosphorylation on four serine acceptor sites in the Rel homology domain for the expression of an array of NF-κB-dependent genes in endothelial cells. We find that inhibition of p65 phosphorylation on these serine residues targets NF-κB activity to distinctive gene subsets in a κB enhancer element-specific context. We show that the phosphorylation-dependent alterations in gene and protein expression are reflective of the amount of p65 and phosphorylated RNA polymerase II (p-RNAP II) bound to respective gene promoter regions. Depending on the gene subset, impaired gene expression was either a result of decreased p65 promoter recruitment or of a failure of bound p65 to recruit p-RNAP II. In conclusion, our findings demonstrate that site-specific p65 phosphorylation targets NF-κB activity to particular gene subsets on a global level by influencing p65 and p-RNAP II promoter recruitment.


Asunto(s)
FN-kappa B/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , Animales , Sitios de Unión , Inmunoprecipitación de Cromatina , Células Endoteliales/citología , Citometría de Flujo , Regulación de la Expresión Génica , Humanos , Inflamación , Ratones , Ratones Transgénicos , Modelos Genéticos , Fosforilación , Plásmidos/metabolismo , Estructura Terciaria de Proteína , Factor de Transcripción ReIA/metabolismo , Transcripción Genética
8.
Commun Biol ; 7(1): 321, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38480905

RESUMEN

Ubiquitin modifications alter protein function and stability, thereby regulating cell homeostasis and viability, particularly under stress. Ischemic stroke induces protein ubiquitination at the ischemic periphery, wherein cells remain viable, however the identity of ubiquitinated proteins is unknown. Here, we employed a proteomics approach to identify these proteins in mice undergoing ischemic stroke. The data are available in a searchable web interface ( https://hochrainerlab.shinyapps.io/StrokeUbiOmics/ ). We detected increased ubiquitination of 198 proteins, many of which localize to the postsynaptic density (PSD) of glutamatergic neurons. Among these were proteins essential for maintaining PSD architecture, such as PSD95, as well as NMDA and AMPA receptor subunits. The largest enzymatic group at the PSD with elevated post-ischemic ubiquitination were kinases, such as CaMKII, PKC, Cdk5, and Pyk2, whose aberrant activities are well-known to contribute to post-ischemic neuronal death. Concurrent phospho-proteomics revealed altered PSD-associated phosphorylation patterns, indicative of modified kinase activities following stroke. PSD-located CaMKII, PKC, and Cdk5 activities were decreased while Pyk2 activity was increased after stroke. Removal of ubiquitin restored kinase activities to pre-stroke levels, identifying ubiquitination as the responsible molecular mechanism for post-ischemic kinase regulation. These findings unveil a previously unrecognized role of ubiquitination in the regulation of essential kinases involved in ischemic injury.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Ratones , Animales , Homólogo 4 de la Proteína Discs Large , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Quinasa 2 de Adhesión Focal , Densidad Postsináptica , Fosfotransferasas , Ubiquitinación , Isquemia , Ubiquitina
9.
Stroke ; 44(8): 2284-2291, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23743975

RESUMEN

BACKGROUND AND PURPOSE: Loss-of-function mutations of the lipoprotein receptor-related protein-6 (LRP6), a coreceptor in the Wingless-related integration site-ß-catenin prosurvival pathway, have been implicated in myocardial ischemia and neurodegeneration. However, it remains to be established whether LRP6 is also involved in ischemic brain injury. We used LRP6+/- mice to examine the role of this receptor in the mechanisms of focal cerebral ischemia. METHODS: Focal cerebral ischemia was induced by transient occlusion of the middle cerebral artery. Motor deficits and infarct volume were assessed 3 days later. Glycogen-synthase-kinase-3ß (GSK-3ß) phosphorylation was examined by Western blotting with phosphospecific antibodies, and the mitochondrial membrane potential changes induced by Ca2+ were also assessed. RESULTS: LRP6+/- mice have larger stroke and more severe motor deficits, effects that were independent of intraischemic cerebral blood flow, vascular factors, or cytosolic ß-catenin levels. Rather, LRP6 haploinsufficiency increased the activating phosphorylation and decreased the inhibitory phosphorylation of GSK-3ß, a kinase involved in proinflammatory signaling and mitochondrial dysfunction. Accordingly, postischemic inflammatory gene expression was enhanced in LRP6+/- mice. Furthermore, the association of mitochondria with activated GSK-3ß was increased in LRP6+/- mice, resulting in a reduction in the Ca2+ handling ability of mitochondria. The mitochondrial dysfunction was reversed by pharmacological inhibition of GSK-3ß. CONCLUSIONS: LRP6 activates an endogenous neuroprotective pathway that acts independently of ß-catenin by controlling GSK-3ß activity and preventing its deleterious mitochondrial and proinflammatory effects. The findings raise the possibility that emerging treatment strategies for diseases attributable to LRP6 loss-of-function mutations could also lead to new therapeutic avenues for ischemic stroke.


Asunto(s)
Isquemia Encefálica/prevención & control , Encéfalo/metabolismo , Encéfalo/patología , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/fisiología , Animales , Conducta Animal/fisiología , Encéfalo/fisiopatología , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Inflamación/genética , Inflamación/metabolismo , Inflamación/prevención & control , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/deficiencia , Ratones , Mitocondrias/genética , Actividad Motora/genética , Fosforilación/genética , Transducción de Señal/genética , beta Catenina/genética , beta Catenina/metabolismo , beta Catenina/fisiología
10.
Cell Mol Life Sci ; 69(12): 2057-73, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22261743

RESUMEN

Termination and resolution of inflammation are tightly linked to the inactivation of one of its strongest inducers, NF-κB. While canonical post-stimulus inactivation is achieved by upregulation of inhibitory molecules that relocate NF-κB complexes to the cytoplasm, termination of the NF-κB response can also be accomplished directly in the nucleus by posttranslational modifications, e.g., ubiquitination of the RelA subunit. Here we reveal a functional role for RelA monoubiquitination in regulating NF-κB activity. By employing serine-to-alanine mutants, we found that hypo-phosphorylated nuclear RelA is monoubiquitinated on multiple lysine residues. Ubiquitination was reversed by IκBα expression and was reduced when nuclear translocation was inhibited. RelA monoubiquitination decreased NF-κB transcriptional activity despite prolonged nuclear presence and independently of RelA degradation, possibly through decreased CREB-binding protein (CBP) co-activator binding. Polyubiquitin-triggered proteasomal degradation has been proposed as a model for RelA inactivation. However, here we show that proteasomal inhibition, similar to RelA hypo-phosphorylation, resulted in nuclear translocation and monoubiquitination of RelA. These findings indicate a degradation-independent mechanism for regulating the activity of nuclear RelA by ubiquitination.


Asunto(s)
FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factor de Transcripción ReIA/metabolismo , Ubiquitinación , Transporte Activo de Núcleo Celular , Línea Celular , Núcleo Celular/metabolismo , Humanos , Proteínas I-kappa B/biosíntesis , Mutación , Inhibidor NF-kappaB alfa , Factor de Transcripción ReIA/genética , Transcripción Genética
11.
bioRxiv ; 2023 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-37662420

RESUMEN

Ubiquitin modifications alter protein function and stability, thereby regulating cell homeostasis and viability, particularly under stress. Ischemic stroke induces protein ubiquitination at the ischemic periphery, wherein cells remain viable, however the identity of ubiquitinated proteins is unknown. Here, we employed a proteomics approach to identify these proteins in mice undergoing ischemic stroke. The data are available in a searchable web interface ( https://hochrainerlab.shinyapps.io/StrokeUbiOmics/ ). We detected increased ubiquitination of 198 proteins, many of which localize to the postsynaptic density (PSD) of glutamatergic neurons. Among these were proteins essential for maintaining PSD architecture, such as PSD95, as well as NMDA and AMPA receptor subunits. The largest enzymatic group at the PSD with elevated post-ischemic ubiquitination were kinases, such as CaMKII, PKC, Cdk5, and Pyk2, whose aberrant activities are well-known to contribute to post-ischemic neuronal death. Concurrent phospho-proteomics revealed altered PSD-associated phosphorylation patterns, indicative of modified kinase activities following stroke. PSD-located CaMKII, PKC, and Cdk5 activities were decreased while Pyk2 activity was increased after stroke. Removal of ubiquitin restored kinase activities to pre-stroke levels, identifying ubiquitination as the responsible molecular mechanism for post-ischemic kinase regulation. These findings unveil a previously unrecognized role of ubiquitination in the regulation of essential kinases involved in ischemic injury.

12.
Stroke ; 43(8): 2229-35, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22700531

RESUMEN

BACKGROUND AND PURPOSE: Cerebral ischemia leads to accumulation of ubiquitinated protein aggregates. However, the factors triggering ubiquitination and their impact on the outcome of cerebral ischemia remain poorly understood. Here we investigate the relationship between ubiquitin aggregation and duration of ischemia/reperfusion, infarct volume, and proteasomal activity in a mouse model of focal ischemia. METHODS: Free ubiquitin and ubiquitin aggregate levels were examined by Western blotting in the mouse neocortex and striatum after different periods of ischemia/reperfusion and permanent ischemia induced by middle cerebral artery occlusion. Infarct volumes were measured in thionin-stained brain sections. Proteasome activity was studied by fluorometric peptidase activity assay. RESULTS: Following transient ischemia, ubiquitin aggregates were detected in the ipsilateral neocortex and, to a lesser extent, striatum only after induction of reperfusion. In permanent ischemia, no ubiquitin aggregates were found. Shorter ischemic periods producing no or minimal tissue damage (10-15 minutes) resulted in ubiquitin aggregate levels similar to those produced by ischemia resulting in substantial infarction (30 minutes). Proteasomal impairment was greatest in ischemia without reperfusion, in which no ubiquitin aggregates were detected. CONCLUSIONS: The data demonstrate that reperfusion rather than ischemia leads to the appearance of ubiquitinated aggregates, which form in the absence of major tissue damage and are not correlated with decreased proteasomal peptidase activity. Ubiquitin aggregates may form in potentially viable brain tissue, which may be later recruited into infarction by factors independent of ubiquitination.


Asunto(s)
Isquemia Encefálica/patología , Infarto de la Arteria Cerebral Media/patología , Daño por Reperfusión/patología , Ubiquitina/metabolismo , Animales , Western Blotting , Encéfalo/patología , Isquemia Encefálica/metabolismo , Cuerpo Estriado/patología , Fluorometría , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neocórtex/patología , Péptido Hidrolasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Daño por Reperfusión/metabolismo , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/patología
13.
J Cell Mol Med ; 14(6B): 1555-68, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19656241

RESUMEN

A great variety of signalling pathways regulating inflammation, cell development and cell survival require NF-kappaB transcription factors, which are normally inactive due to binding to inhibitors, such as IkappaBalpha. The canonical activation pathway of NF-kappaB is initiated by phosphorylation of the inhibitor by an IkappaB kinase (IKK) complex triggering ubiquitination of IkappaB molecules by SCF-type E3-ligase complexes and rapid degradation by 26S-proteasomes. The ubiquitination machinery is regulated by the COP9 signalosome (CSN). We show that IkappaB kinases interact with the CSN-complex, as well as the SCF-ubiquitination machinery, providing an explanation for the rapid signalling-induced ubiquitination and degradation of IkappaBalpha. Furthermore, we reveal that IKK's phosphorylate not only IkappaBalpha, but also the CSN-subunit Csn5/JAB1 (c-Jun activation domain binding protein-1) and that IKK2 influences ubiquitination of Csn5/JAB1. Our observations imply that the CSN complex acts as an inhibitor of constitutive NF-kappaB activity in non-activated cells. Knock-down of Csn5/JAB1 clearly enhanced basal NF-kappaB activity and improved cell survival under stress. The inhibitory effect of Csn5/JAB1 requires a functional MPN(+) metalloprotease domain, which is responsible for cleaving ubiquitin-like Nedd8-modifications. Upon activation of cells with tumour necrosis factor-alpha, the CSN complex dissociates from IKK's allowing full and rapid activation of the NF-kappaB pathway by the concerted action of interacting protein complexes.


Asunto(s)
Quinasa I-kappa B/metabolismo , Complejos Multiproteicos/metabolismo , FN-kappa B/metabolismo , Péptido Hidrolasas/metabolismo , Transducción de Señal , Apoptosis , Complejo del Señalosoma COP9 , Línea Celular , Transferencia Resonante de Energía de Fluorescencia , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Ubiquitinación
14.
Nat Neurosci ; 23(9): 1079-1089, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32778793

RESUMEN

Cerebrovascular abnormalities have emerged as a preclinical manifestation of Alzheimer's disease and frontotemporal dementia, diseases characterized by the accumulation of hyperphosphorylated forms of the microtubule-associated protein tau. However, it is unclear whether tau contributes to these neurovascular alterations independent of neurodegeneration. We report that mice expressing mutated tau exhibit a selective suppression of neural activity-induced cerebral blood flow increases that precedes tau pathology and cognitive impairment. This dysfunction is attributable to a reduced vasodilatation of intracerebral arterioles and is reversible by reducing tau production. Mechanistically, the failure of neurovascular coupling involves a tau-induced dissociation of neuronal nitric oxide synthase (nNOS) from postsynaptic density 95 (PSD95) and a reduced production of the potent vasodilator nitric oxide during glutamatergic synaptic activity. These data identify glutamatergic signaling dysfunction and nitric oxide deficiency as yet-undescribed early manifestations of tau pathobiology, independent of neurodegeneration, and provide a mechanism for the neurovascular alterations observed in the preclinical stages of tauopathies.


Asunto(s)
Circulación Cerebrovascular/fisiología , Homólogo 4 de la Proteína Discs Large/metabolismo , Acoplamiento Neurovascular/fisiología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Proteínas tau/metabolismo , Animales , Humanos , Ratones , Ratones Transgénicos , Degeneración Nerviosa , Tauopatías/metabolismo
15.
J Neurosci ; 28(7): 1649-58, 2008 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-18272685

RESUMEN

CD36, a class-B scavenger receptor involved in multiple functions, including inflammatory signaling, may also contribute to ischemic brain injury through yet unidentified mechanisms. We investigated whether CD36 participates in the molecular events underlying the inflammatory reaction that accompanies cerebral ischemia and may contribute to the tissue damage. We found that activation of nuclear factor-kappaB, a transcription factor that coordinates postischemic gene expression, is attenuated in CD36-null mice subjected to middle cerebral artery occlusion. The infiltration of neutrophils and the glial reaction induced by cerebral ischemia were suppressed. Treatment with an inhibitor of inducible nitric oxide synthase, an enzyme that contributes to the tissue damage, reduced ischemic brain injury in wild-type mice, but not in CD36 nulls. In contrast to cerebral ischemia, the molecular and cellular inflammatory changes induced by intracerebroventricular injection of interleukin-1beta were not attenuated in CD36-null mice. The findings unveil a novel role of CD36 in early molecular events leading to nuclear factor-kappaB activation and postischemic inflammation. Inhibition of CD36 signaling may be a valuable therapeutic approach to counteract the deleterious effects of postischemic inflammation.


Asunto(s)
Antígenos CD36/metabolismo , Encefalitis/genética , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Secuencia de Aminoácidos , Animales , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Encefalitis/inducido químicamente , Encefalitis/metabolismo , Encefalitis/prevención & control , Expresión Génica , Guanidinas/farmacología , Interleucina-1beta , Ataque Isquémico Transitorio/etiología , Ataque Isquémico Transitorio/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Nitrobencenos/farmacología , Peroxidasa/metabolismo , ARN Mensajero/análisis , Especies Reactivas de Oxígeno/metabolismo , Sulfonamidas/farmacología
16.
Transl Stroke Res ; 9(2): 157-173, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28842824

RESUMEN

Post-translational protein modifications present an elegant and energy efficient way to dynamically reprogram cellular protein properties and functions in response to homeostatic imbalance. One such protein modification is the tagging of proteins with the small modifier ubiquitin that can have an impact on protein stability, localization, interaction dynamics, and function. Ubiquitination is vital to any eukaryotic cell under physiological conditions, but even more important under stress including oxidative, genotoxic, and heat stress, where ubiquitination levels are drastically increased. Elevated levels of ubiquitin-protein conjugates are also observed in the brain after focal and global cerebral ischemia. Post-ischemic ubiquitination is immediately induced with reperfusion and transiently detected in neurons with survival potential located in the peri-infarct area. This review aims to critically discuss current knowledge and controversies on protein ubiquitination after cerebral ischemia, with special emphasis on potential mechanisms leading to elevated ubiquitination and on target identification. Further, possible functional implications of post-ischemic ubiquitination, including a relationship to SUMOylation, a neuroprotective modification, will be highlighted. The elevation in ubiquitinated proteins following cerebral ischemia is a greatly under-explored research area, the better understanding of which may contribute to the development of novel stroke therapies.


Asunto(s)
Isquemia Encefálica/metabolismo , Modificación Traduccional de las Proteínas/fisiología , Daño por Reperfusión/metabolismo , Ubiquitina/metabolismo , Animales , Humanos
17.
Sci Rep ; 8(1): 2701, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29426953

RESUMEN

Protein aggregation critically affects cell viability in neurodegenerative diseases, but whether this also occurs in ischemic brain injury remains elusive. Prior studies report the post-ischemic aggregation of ubiquitin, small ubiquitin-related modifier (SUMO) and ribosomes, however whether other proteins are also affected is unknown. Here we employed a proteomic approach to identify the insoluble, aggregated proteome after cerebral ischemia. Mice underwent transient middle cerebral artery occlusion or sham-surgery. After 1-hour reperfusion, prior to apparent brain injury, mice were sacrificed and detergent-insoluble proteins were obtained and identified by nanoLC-MS/MS. Naturally existing insoluble proteins were determined in sham controls and aggregated proteins after cerebral ischemia/reperfusion were identified. Selected aggregated proteins found by proteomics were biochemically verified and aggregation propensities were studied during ischemia with or without reperfusion. We found that ischemia/reperfusion induces the aggregation of RNA-binding and heat-shock proteins, ubiquitin, SUMO and other proteins involved in cell signalling. RNA-binding proteins constitute the largest group of aggregating proteins in ischemia. These include TDP43, FUS, hnRNPA1, PSF/SFPQ and p54/NONO, all of which have been linked to neurodegeneration associated with amyotrophic lateral sclerosis and frontotemporal dementia. The aggregation of neurodegeneration-related disease proteins in cerebral ischemia unveils a previously unappreciated molecular overlap between neurodegenerative diseases and ischemic stroke.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Animales , Infarto Cerebral , Circulación Cerebrovascular/fisiología , Hipocampo/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/fisiopatología , Ataque Isquémico Transitorio , Masculino , Ratones , Ratones Endogámicos C57BL , Arteria Cerebral Media/fisiopatología , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Agregado de Proteínas/fisiología , Proteómica/métodos , Reperfusión , Daño por Reperfusión/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Accidente Cerebrovascular/metabolismo , Espectrometría de Masas en Tándem , Ubiquitina/metabolismo
18.
Sci Rep ; 8(1): 6802, 2018 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-29700368

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

19.
FEBS Lett ; 581(28): 5493-9, 2007 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-17991436

RESUMEN

Subcellular localization guided by IkappaBalpha is crucial for regulation of nuclear factor-kappaB function. Here, we show that p65 Rel homology domain phosphorylation mutants are transported into the nucleus after IkappaBalpha degradation, but as a consequence of lower IkappaBalpha levels their relocation to the cytosol is blocked. We demonstrate that phosphorylation of residues S205, S276, and S281 of p65 is not required for interaction between p65 and IkappaBalpha, but is pivotal for regulating cellular IkappaBalpha levels by positively affecting gene synthesis. Our findings indicate that reduction of phosphorylation leads to nuclear retention of p65, which might be partly responsible for altered transcriptional behavior of p65 serine mutants.


Asunto(s)
Núcleo Celular/metabolismo , Quinasa I-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , Línea Celular , Citosol/metabolismo , Humanos , Quinasa I-kappa B/genética , Ratones , Mutación/genética , Fosforilación , Serina/genética , Serina/metabolismo , Factor de Transcripción ReIA/deficiencia , Factor de Transcripción ReIA/genética
20.
Thromb Haemost ; 97(3): 370-7, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17334503

RESUMEN

Ubiquitin is a highly conserved 76-amino-acid peptide that becomes covalently attached to lysine residues of target proteins. Since ubiquitin itself contains seven lysine residues, ubiquitin molecules can generate different types of polyubiquitin chains. Lys48-linked polyubiquitylation is well-known as posttranslational tag for targeting proteins for degradation by the 26S proteasome. Recent studies have revealed several new functions of ubiquitin, e.g. activation of protein kinases, control of gene transcription, DNA repair and replication, intracellular trafficking and virus budding. These functions are mainly mediated by Lys63 polyubiquitin chains or attachment of a single ubiquitin molecule to one or several lysine residues within the target protein. Importantly, protein ubiquitylation exhibits inducibility, reversibilty and recognition by specialized ubiquitin-binding domains, features similar to protein phosphorylation. In this review we comprehensively describe regulations of protein ubiquitylation and their impact on distinct signaling pathways.


Asunto(s)
Endotelio Vascular/metabolismo , Inflamación/metabolismo , FN-kappa B/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Ubiquitina/metabolismo , Animales , Humanos , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Lisina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA