Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Nat Immunol ; 11(3): 265-72, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20139987

RESUMEN

Voltage-gated proton currents regulate generation of reactive oxygen species (ROS) in phagocytic cells. In B cells, stimulation of the B cell antigen receptor (BCR) results in the production of ROS that participate in B cell activation, but the involvement of proton channels is unknown. We report here that the voltage-gated proton channel HVCN1 associated with the BCR complex and was internalized together with the BCR after activation. BCR-induced generation of ROS was lower in HVCN1-deficient B cells, which resulted in attenuated BCR signaling via impaired BCR-dependent oxidation of the tyrosine phosphatase SHP-1. This resulted in less activation of the kinases Syk and Akt, impaired mitochondrial respiration and glycolysis and diminished antibody responses in vivo. Our findings identify unanticipated functions for proton channels in B cells and demonstrate the importance of ROS in BCR signaling and downstream metabolism.


Asunto(s)
Linfocitos B/inmunología , Canales Iónicos/inmunología , Especies Reactivas de Oxígeno/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Animales , Linfocitos B/enzimología , Activación Enzimática/inmunología , Immunoblotting , Péptidos y Proteínas de Señalización Intracelular/inmunología , Ratones , Ratones Noqueados , Microscopía Confocal , Mitocondrias/inmunología , Proteína Oncogénica v-akt/inmunología , Proteínas Tirosina Quinasas/inmunología , Transducción de Señal , Quinasa Syk
2.
PLoS Biol ; 16(4): e2003611, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29630591

RESUMEN

Nitric oxide (NO) regulates neuronal function and thus is critical for tuning neuronal communication. Mechanisms by which NO modulates protein function and interaction include posttranslational modifications (PTMs) such as S-nitrosylation. Importantly, cross signaling between S-nitrosylation and prenylation can have major regulatory potential. However, the exact protein targets and resulting changes in function remain elusive. Here, we interrogated the role of NO-dependent PTMs and farnesylation in synaptic transmission. We found that NO compromises synaptic function at the Drosophila neuromuscular junction (NMJ) in a cGMP-independent manner. NO suppressed release and reduced the size of available vesicle pools, which was reversed by glutathione (GSH) and occluded by genetic up-regulation of GSH-generating and de-nitrosylating glutamate-cysteine-ligase and S-nitroso-glutathione reductase activities. Enhanced nitrergic activity led to S-nitrosylation of the fusion-clamp protein complexin (cpx) and altered its membrane association and interactions with active zone (AZ) and soluble N-ethyl-maleimide-sensitive fusion protein Attachment Protein Receptor (SNARE) proteins. Furthermore, genetic and pharmacological suppression of farnesylation and a nitrosylation mimetic mutant of cpx induced identical physiological and localization phenotypes as caused by NO. Together, our data provide evidence for a novel physiological nitrergic molecular switch involving S-nitrosylation, which reversibly suppresses farnesylation and thereby enhances the net-clamping function of cpx. These data illustrate a new mechanistic signaling pathway by which regulation of farnesylation can fine-tune synaptic release.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurotransmisores/metabolismo , Óxido Nítrico/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Adaptadoras del Transporte Vesicular/genética , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Encéfalo/metabolismo , GMP Cíclico/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Glutamato-Cisteína Ligasa/genética , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/metabolismo , Larva/genética , Larva/metabolismo , Proteínas del Tejido Nervioso/genética , Unión Neuromuscular/citología , Unión Neuromuscular/metabolismo , Fenotipo , Prenilación , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Transmisión Sináptica , Vesículas Sinápticas/metabolismo
3.
Nature ; 518(7538): 236-9, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25607368

RESUMEN

In the healthy adult brain synapses are continuously remodelled through a process of elimination and formation known as structural plasticity. Reduction in synapse number is a consistent early feature of neurodegenerative diseases, suggesting deficient compensatory mechanisms. Although much is known about toxic processes leading to synaptic dysfunction and loss in these disorders, how synaptic regeneration is affected is unknown. In hibernating mammals, cooling induces loss of synaptic contacts, which are reformed on rewarming, a form of structural plasticity. We have found that similar changes occur in artificially cooled laboratory rodents. Cooling and hibernation also induce a number of cold-shock proteins in the brain, including the RNA binding protein, RBM3 (ref. 6). The relationship of such proteins to structural plasticity is unknown. Here we show that synapse regeneration is impaired in mouse models of neurodegenerative disease, in association with the failure to induce RBM3. In both prion-infected and 5XFAD (Alzheimer-type) mice, the capacity to regenerate synapses after cooling declined in parallel with the loss of induction of RBM3. Enhanced expression of RBM3 in the hippocampus prevented this deficit and restored the capacity for synapse reassembly after cooling. RBM3 overexpression, achieved either by boosting endogenous levels through hypothermia before the loss of the RBM3 response or by lentiviral delivery, resulted in sustained synaptic protection in 5XFAD mice and throughout the course of prion disease, preventing behavioural deficits and neuronal loss and significantly prolonging survival. In contrast, knockdown of RBM3 exacerbated synapse loss in both models and accelerated disease and prevented the neuroprotective effects of cooling. Thus, deficient synapse regeneration, mediated at least in part by failure of the RBM3 stress response, contributes to synapse loss throughout the course of neurodegenerative disease. The data support enhancing cold-shock pathways as potential protective therapies in neurodegenerative disorders.


Asunto(s)
Frío , Respuesta al Choque por Frío/fisiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Plasticidad Neuronal , Fármacos Neuroprotectores , Proteínas de Unión al ARN/metabolismo , Sinapsis/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Proteínas y Péptidos de Choque por Frío/metabolismo , Modelos Animales de Enfermedad , Hibernación/fisiología , Hipocampo/metabolismo , Masculino , Ratones , Priones/fisiología , Proteínas de Unión al ARN/genética , Regeneración
4.
Genes Dev ; 26(18): 2009-14, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22987635

RESUMEN

Aging is associated with impaired scavenging of reactive oxygen species (ROS). Here, we show that TAp73, a p53 family member, protects against aging by regulating mitochondrial activity and preventing ROS accumulation. TAp73-null mice show more pronounced aging with increased oxidative damage and senescence. TAp73 deletion reduces cellular ATP levels, oxygen consumption, and mitochondrial complex IV activity, with increased ROS production and oxidative stress sensitivity. We show that the mitochondrial complex IV subunit cytochrome C oxidase subunit 4 (Cox4i1) is a direct TAp73 target and that Cox4i1 knockdown phenocopies the cellular senescence of TAp73-null cells. Results indicate that TAp73 affects mitochondrial respiration and ROS homeostasis, thus regulating aging.


Asunto(s)
Envejecimiento/genética , Envejecimiento/metabolismo , Mitocondrias/metabolismo , Consumo de Oxígeno , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Animales , Células Cultivadas , Complejo IV de Transporte de Electrones/metabolismo , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Células HCT116 , Humanos , Ratones , Ratones Noqueados
5.
Nature ; 485(7399): 507-11, 2012 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-22622579

RESUMEN

The mechanisms leading to neuronal death in neurodegenerative disease are poorly understood. Many of these disorders, including Alzheimer's, Parkinson's and prion diseases, are associated with the accumulation of misfolded disease-specific proteins. The unfolded protein response is a protective cellular mechanism triggered by rising levels of misfolded proteins. One arm of this pathway results in the transient shutdown of protein translation, through phosphorylation of the α-subunit of eukaryotic translation initiation factor, eIF2. Activation of the unfolded protein response and/or increased eIF2α-P levels are seen in patients with Alzheimer's, Parkinson's and prion diseases, but how this links to neurodegeneration is unknown. Here we show that accumulation of prion protein during prion replication causes persistent translational repression of global protein synthesis by eIF2α-P, associated with synaptic failure and neuronal loss in prion-diseased mice. Further, we show that promoting translational recovery in hippocampi of prion-infected mice is neuroprotective. Overexpression of GADD34, a specific eIF2α-P phosphatase, as well as reduction of levels of prion protein by lentivirally mediated RNA interference, reduced eIF2α-P levels. As a result, both approaches restored vital translation rates during prion disease, rescuing synaptic deficits and neuronal loss, thereby significantly increasing survival. In contrast, salubrinal, an inhibitor of eIF2α-P dephosphorylation, increased eIF2α-P levels, exacerbating neurotoxicity and significantly reducing survival in prion-diseased mice. Given the prevalence of protein misfolding and activation of the unfolded protein response in several neurodegenerative diseases, our results suggest that manipulation of common pathways such as translational control, rather than disease-specific approaches, may lead to new therapies preventing synaptic failure and neuronal loss across the spectrum of these disorders.


Asunto(s)
Factor 2 Eucariótico de Iniciación/química , Factor 2 Eucariótico de Iniciación/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Fosfoproteínas/metabolismo , Priones/metabolismo , Biosíntesis de Proteínas , Proteínas Represoras/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Cinamatos/farmacología , Factor 2 Eucariótico de Iniciación/análisis , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/patología , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores , Fosfoproteínas/análisis , Fosforilación , Proteínas PrPSc/análisis , Proteínas PrPSc/metabolismo , Proteínas PrPSc/toxicidad , Enfermedades por Prión/patología , Priones/biosíntesis , Priones/genética , Biosíntesis de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Proteínas Represoras/análisis , Proteínas Represoras/química , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/patología , Transmisión Sináptica/efectos de los fármacos , Tiourea/análogos & derivados , Tiourea/farmacología , Respuesta de Proteína Desplegada/fisiología
6.
Proc Natl Acad Sci U S A ; 112(37): 11577-82, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26324887

RESUMEN

Short p63 isoform, ΔNp63, is crucial for epidermis formation, and it plays a pivotal role in controlling the turnover of basal keratinocytes by regulating the expression of a subset of genes involved in cell cycle and cell adhesion programs. The glycolytic enzyme hexokinase 2 (HK2) represents the first step of glucose utilization in cells. The family of HKs has four isoforms that differ mainly in their tissue and subcellular distribution. The preferential mitochondrial localization of HK2 at voltage-dependent anion channels provides access to ATP generated by oxidative phosphorylation and generates an ADP/ATP recycling mechanism to maintain high respiration rates and low electron leak. Here, we report that ΔNp63 depletion in human keratinocytes impairs mitochondrial basal respiration and increases mitochondrial membrane polarization and intracellular reactive oxygen species. We show ΔNp63-dependent regulation of HK2 expression, and we use ChIP, validated by p63-Chip sequencing genomewide profiling analysis, and luciferase assays to demonstrate the presence of one p63-specific responsive element within the 15th intronic region of the HK2 gene, providing evidence of a direct interaction. Our data support the notion of ΔNp63 as a master regulator in epithelial cells of a combined subset of molecular mechanisms, including cellular energy metabolism and respiration. The ΔNp63-HK2 axis is also present in epithelial cancer cells, suggesting that ΔNp63 could participate in cancer metabolic reprogramming.


Asunto(s)
Hexoquinasa/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proliferación Celular , Separación Celular , Citometría de Flujo , Silenciador del Gen , Glucólisis , Humanos , Peróxido de Hidrógeno/química , Queratinocitos/citología , Ratones , Microscopía Electrónica , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Células 3T3 NIH , Neoplasias/metabolismo , Fosforilación Oxidativa , Estrés Oxidativo , Oxígeno/química , Consumo de Oxígeno , Fenotipo , Proteína p53 Supresora de Tumor/metabolismo
7.
Proc Natl Acad Sci U S A ; 111(5): 1843-8, 2014 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-24449892

RESUMEN

The generation of viable sperm proceeds through a series of coordinated steps, including germ cell self-renewal, meiotic recombination, and terminal differentiation into functional spermatozoa. The p53 family of transcription factors, including p53, p63, and p73, are critical for many physiological processes, including female fertility, but little is known about their functions in spermatogenesis. Here, we report that deficiency of the TAp73 isoform, but not p53 or ΔNp73, results in male infertility because of severe impairment of spermatogenesis. Mice lacking TAp73 exhibited increased DNA damage and cell death in spermatogonia, disorganized apical ectoplasmic specialization, malformed spermatids, and marked hyperspermia. We demonstrated that TAp73 regulates the mRNA levels of crucial genes involved in germ stem/progenitor cells (CDKN2B), spermatid maturation/spermiogenesis (metalloproteinase and serine proteinase inhibitors), and steroidogenesis (CYP21A2 and progesterone receptor). These alterations of testicular histology and gene expression patterns were specific to TAp73 null mice and not features of mice lacking p53. Our work provides previously unidentified in vivo evidence that TAp73 has a unique role in spermatogenesis that ensures the maintenance of mitotic cells and normal spermiogenesis. These results may have implications for the diagnosis and management of human male infertility.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Fertilidad , Proteínas Nucleares/metabolismo , Espermatogénesis , Proteínas Supresoras de Tumor/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Envejecimiento/patología , Animales , Apoptosis/genética , Recuento de Células , Proliferación Celular , Daño del ADN/genética , Proteínas de Unión al ADN/deficiencia , Femenino , Fertilidad/genética , Regulación de la Expresión Génica , Humanos , Infertilidad Masculina/sangre , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Masculino , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Proteínas Nucleares/deficiencia , Estrés Oxidativo/genética , Progesterona/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo , Espermatogénesis/genética , Espermatozoides/metabolismo , Espermatozoides/patología , Testículo/metabolismo , Testículo/patología , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/deficiencia
8.
Hum Mol Genet ; 23(17): 4581-96, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24722203

RESUMEN

The cellular prion protein (PrP(C)) has been implicated in several neurodegenerative diseases as a result of protein misfolding. In humans, prion disease occurs typically with a sporadic origin where uncharacterized mechanisms induce spontaneous PrP(C) misfolding leading to neurotoxic PrP-scrapie formation (PrP(SC)). The consequences of misfolded PrP(C) signalling are well characterized but little is known about the physiological roles of PrP(C) and its involvement in disease. Here we investigated wild-type PrP(C) signalling in synaptic function as well as the effects of a disease-relevant mutation within PrP(C) (proline-to-leucine mutation at codon 101). Expression of wild-type PrP(C) at the Drosophila neuromuscular junction leads to enhanced synaptic responses as detected in larger miniature synaptic currents which are caused by enlarged presynaptic vesicles. The expression of the mutated PrP(C) leads to reduction of both parameters compared with wild-type PrP(C). Wild-type PrP(C) enhances synaptic release probability and quantal content but reduces the size of the ready-releasable vesicle pool. Partially, these changes are not detectable following expression of the mutant PrP(C). A behavioural test revealed that expression of either protein caused an increase in locomotor activities consistent with enhanced synaptic release and stronger muscle contractions. Both proteins were sensitive to proteinase digestion. These data uncover new functions of wild-type PrP(C) at the synapse with a disease-relevant mutation in PrP(C) leading to diminished functional phenotypes. Thus, our data present essential new information possibly related to prion pathogenesis in which a functional synaptic role of PrP(C) is compromised due to its advanced conversion into PrP(SC) thereby creating a lack-of-function scenario.


Asunto(s)
Priones/metabolismo , Probabilidad , Vesículas Sinápticas/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Calcio/farmacología , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/metabolismo , Endopeptidasa K/metabolismo , Larva/efectos de los fármacos , Larva/ultraestructura , Ratones , Actividad Motora/efectos de los fármacos , Proteínas Mutantes/metabolismo , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/patología , Unión Neuromuscular/ultraestructura , Vesículas Sinápticas/efectos de los fármacos , Vesículas Sinápticas/ultraestructura
9.
Proc Natl Acad Sci U S A ; 110(47): 18952-7, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24190996

RESUMEN

Total and N-terminal isoform selective p73 knockout mice show a variety of central nervous system defects. Here we show that TAp73 is a transcriptional activator of p75 neurotrophin receptor (p75(NTR)) and that p75(NTR) mRNA and protein levels are strongly reduced in the central and peripheral nervous systems of p73 knockout mice. In parallel, primary cortical neurons from p73 knockout mice showed a reduction in neurite outgrowth and in nerve growth factor-mediated neuronal differentiation, together with reduced miniature excitatory postsynaptic current frequencies and behavioral defects. p73 null mice also have impairments in the peripheral nervous system with reduced thermal sensitivity, axon number, and myelin thickness. At least some of these morphological and functional impairments in p73 null cells can be rescued by p75(NTR) re-expression. Together, these data demonstrate that loss of p75(NTR) contributes to the neurological phenotype of p73 knockout mice.


Asunto(s)
Malformaciones del Sistema Nervioso/genética , Neuritas/patología , Proteínas Nucleares/genética , Receptores de Factor de Crecimiento Nervioso/deficiencia , Animales , Western Blotting , Encéfalo/metabolismo , Biología Computacional , Ratones , Ratones Noqueados , Potenciales Postsinápticos Miniatura/genética , Vaina de Mielina/metabolismo , Malformaciones del Sistema Nervioso/patología , Neuritas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Activación Transcripcional/genética
10.
Blood ; 117(26): 7145-54, 2011 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-21562047

RESUMEN

Apoptosis in megakaryocytes results in the formation of platelets. The role of apoptotic pathways in platelet turnover and in the apoptotic-like changes seen after platelet activation is poorly understood. ABT-263 (Navitoclax), a specific inhibitor of antiapoptotic BCL2 proteins, which is currently being evaluated in clinical trials for the treatment of leukemia and other malignancies, induces a dose-limiting thrombocytopenia. In this study, the relationship between BCL2/BCL-X(L) inhibition, apoptosis, and platelet activation was investigated. Exposure to ABT-263 induced apoptosis but repressed platelet activation by physiologic agonists. Notably, ABT-263 induced an immediate calcium response in platelets and the depletion of intracellular calcium stores, indicating that on BCL2/BCL-X(L) inhibition platelet activation is abrogated because of a diminished calcium signaling. By comparing the effects of ABT-263 and its analog ABT-737 on platelets and leukemia cells from the same donor, we show, for the first time, that these BCL2/BCL-X(L) inhibitors do not offer any selective toxicity but induce apoptosis at similar concentrations in leukemia cells and platelets. However, reticulated platelets are less sensitive to apoptosis, supporting the hypothesis that treatment with ABT-263 induces a selective loss of older platelets and providing an explanation for the transient thrombocytopenia observed on ABT-263 treatment.


Asunto(s)
Compuestos de Anilina/farmacología , Apoptosis/efectos de los fármacos , Plaquetas/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Sulfonamidas/farmacología , Proteína bcl-X/antagonistas & inhibidores , Compuestos de Anilina/efectos adversos , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/metabolismo , Compuestos de Bifenilo/efectos adversos , Compuestos de Bifenilo/farmacología , Plaquetas/metabolismo , Plaquetas/ultraestructura , Expresión Génica , Homeostasis/efectos de los fármacos , Humanos , Cinética , Leucemia Linfocítica Crónica de Células B/sangre , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Terapia Molecular Dirigida , Nitrofenoles/efectos adversos , Nitrofenoles/farmacología , Piperazinas/efectos adversos , Piperazinas/farmacología , Inhibidores de Agregación Plaquetaria/efectos adversos , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonamidas/efectos adversos , Trombocitopenia/inducido químicamente , Trombopoyesis , Proteína bcl-X/metabolismo
11.
Hum Mol Genet ; 19(9): 1669-77, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20123860

RESUMEN

Mutations of thymidine kinase 2 (TK2), an essential component of the mitochondrial nucleotide salvage pathway, can give rise to mitochondrial DNA (mtDNA) depletion syndromes (MDS). These clinically heterogeneous disorders are characterized by severe reduction in mtDNA copy number in affected tissues and are associated with progressive myopathy, hepatopathy and/or encephalopathy, depending in part on the underlying nuclear genetic defect. Mutations of TK2 have previously been associated with an isolated myopathic form of MDS (OMIM 609560). However, more recently, neurological phenotypes have been demonstrated in patients carrying TK2 mutations, thus suggesting that loss of TK2 results in neuronal dysfunction. Here, we directly address the role of TK2 in neuronal homeostasis using a knockout mouse model. We demonstrate that in vivo loss of TK2 activity leads to a severe ataxic phenotype, accompanied by reduced mtDNA copy number and decreased steady-state levels of electron transport chain proteins in the brain. In TK2-deficient cerebellar neurons, these abnormalities are associated with impaired mitochondrial bioenergetic function, aberrant mitochondrial ultrastructure and degeneration of selected neuronal types. Overall, our findings demonstrate that TK2 deficiency leads to neuronal dysfunction in vivo, and have important implications for understanding the mechanisms of neurological impairment in MDS.


Asunto(s)
Enfermedades Mitocondriales/genética , Neuronas/citología , Neuronas/metabolismo , Timidina Quinasa/deficiencia , Análisis de Varianza , Animales , Ataxia/enzimología , Ataxia/etiología , Secuencia de Bases , Encéfalo/metabolismo , Variaciones en el Número de Copia de ADN , ADN Mitocondrial/genética , Metabolismo Energético , Vectores Genéticos , Immunoblotting , Inmunohistoquímica , Lentivirus , Ratones , Ratones Noqueados , Enfermedades Mitocondriales/complicaciones , Datos de Secuencia Molecular , Mutación/genética , Timidina Quinasa/genética
12.
Chem Res Toxicol ; 25(4): 828-37, 2012 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-22263782

RESUMEN

Amorphous silica nanoparticles (SiO2-NPs) have found broad applications in industry and are currently intensively studied for potential uses in medical and biomedical fields. Several studies have reported cytotoxic and inflammatory responses induced by SiO2-NPs in different cell types. The present study was designed to examine the association of oxidative stress markers with SiO2-NP induced cytotoxicity in human endothelial cells. We used pure monodisperse amorphous silica nanoparticles of two sizes (16 and 60 nm; S16 and S60) and a positive control, iron-doped nanosilica (16 nm; SFe), to study the generation of hydroxyl radicals (HO·) in cellular-free conditions and oxidative stress in cellular systems. We investigated whether SiO2-NPs could influence intracellular reduced glutathione (GSH) and oxidized glutathione (GSSG) levels, increase lipid peroxidation (malondialdehyde (MDA) and 4-hydroxyalkenal (HAE) concentrations), and up-regulate heme oxygenase-1 (HO-1) mRNA expression in the studied cells. None of the particles, except SFe, produced ROS in cell-free systems. We found significant modifications for all parameters in cells treated with SFe nanoparticles. At cytotoxic doses of S16 (40-50 µg/mL), we detected weak alterations of intracellular glutathione (4 h) and a marked induction of HO-1 mRNA (6 h). Cytotoxic doses of S60 elicited similar responses. Preincubation of cells being exposed to SiO2-NPs with an antioxidant (5 mM N-acetylcysteine, NAC) significantly reduced the cytotoxic activity of S16 and SFe (when exposed up to 25 and 50 µg/mL, respectively) but did not protect cells treated with S60. Preincubation with NAC significantly reduced HO-1 mRNA expression in cells treated with SFe but did not have any effect on HO-1 mRNA level in cell exposed to S16 and S60. Our study demonstrates that the chemical composition of the silica nanoparticles is a dominant factor in inducing oxidative stress.


Asunto(s)
Hierro/química , Nanopartículas/toxicidad , Estrés Oxidativo/efectos de los fármacos , Dióxido de Silicio/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Sistema Libre de Células , Regulación de la Expresión Génica , Glutatión/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Radical Hidroxilo/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Nanopartículas/química , Tamaño de la Partícula
13.
Cell Death Differ ; 29(1): 147-155, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34354257

RESUMEN

Cancer cells that are resistant to Bax/Bak-dependent intrinsic apoptosis can be eliminated by proteasome inhibition. Here, we show that proteasome inhibition induces the formation of high molecular weight platforms in the cytosol that serve to activate caspase-8. The activation complexes contain Fas-associated death domain (FADD) and receptor-interacting serine/threonine-protein kinase 1 (RIPK1). Furthermore, the complexes contain TRAIL-receptor 2 (TRAIL-R2) but not TRAIL-receptor 1 (TRAIL-R1). While RIPK1 inhibition or depletion did not affect proteasome inhibitor-induced cell death, TRAIL-R2 was found essential for efficient caspase-8 activation, since the loss of TRAIL-R2 expression abrogated caspase processing, significantly reduced cell death, and promoted cell re-growth after drug washout. Overall, our study provides novel insight into the mechanisms by which proteasome inhibition eliminates otherwise apoptosis-resistant cells, and highlights the crucial role of a ligand-independent but TRAIL-R2-dependent activation mechanism for caspase-8 in this scenario.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Apoptosis , Caspasa 8/metabolismo , Citosol/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
14.
J Cell Sci ; 122(Pt 18): 3330-9, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19706685

RESUMEN

Alterations in the autophagic pathway are associated with the onset and progression of various diseases. However, despite the therapeutic potential for pharmacological modulators of autophagic flux, few such compounds have been characterised. Here we show that clomipramine, an FDA-approved drug long used for the treatment of psychiatric disorders, and its active metabolite desmethylclomipramine (DCMI) interfere with autophagic flux. Treating cells with DCMI caused a significant and specific increase in autophagosomal markers and a concomitant blockage of the degradation of autophagic cargo. This observation might be relevant in therapy in which malignant cells exploit autophagy to survive stress conditions, rendering them more susceptible to the action of cytotoxic agents. In accordance, DCMI-mediated obstruction of autophagic flux increased the cytotoxic effect of chemotherapeutic agents. Collectively, our studies describe a new function of DCMI that can be exploited for the treatment of pathological conditions in which manipulation of autophagic flux is thought to be beneficial.


Asunto(s)
Autofagia/efectos de los fármacos , Clomipramina/análogos & derivados , Animales , Biomarcadores/metabolismo , Clomipramina/farmacología , Ensayo de Unidades Formadoras de Colonias , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/ultraestructura , Mutación Puntual/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo
15.
Nat Commun ; 12(1): 819, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33547302

RESUMEN

Regulated cell death is essential in development and cellular homeostasis. Multi-protein platforms, including the Death-Inducing Signaling Complex (DISC), co-ordinate cell fate via a core FADD:Caspase-8 complex and its regulatory partners, such as the cell death inhibitor c-FLIP. Here, using electron microscopy, we visualize full-length procaspase-8 in complex with FADD. Our structural analysis now reveals how the FADD-nucleated tandem death effector domain (tDED) helical filament is required to orientate the procaspase-8 catalytic domains, enabling their activation via anti-parallel dimerization. Strikingly, recruitment of c-FLIPS into this complex inhibits Caspase-8 activity by altering tDED triple helix architecture, resulting in steric hindrance of the canonical tDED Type I binding site. This prevents both Caspase-8 catalytic domain assembly and tDED helical filament elongation. Our findings reveal how the plasticity, composition and architecture of the core FADD:Caspase-8 complex critically defines life/death decisions not only via the DISC, but across multiple key signaling platforms including TNF complex II, the ripoptosome, and RIPK1/RIPK3 necrosome.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/química , Caspasa 8/química , Proteína de Dominio de Muerte Asociada a Fas/química , Proteína Serina-Treonina Quinasas de Interacción con Receptores/química , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Dominio Catalítico , Clonación Molecular , Microscopía por Crioelectrón , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/química , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/genética , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerización de Proteína , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Muerte Celular Regulada/genética , Factor de Necrosis Tumoral alfa/química , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
16.
J Physiol ; 588(Pt 23): 4683-93, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20937712

RESUMEN

There is a well-established link between hyperbilirubinaemia and hearing loss in paediatrics, but the cellular mechanisms have not been elucidated. Here we used the Gunn rat model of hyperbilirubinaemia to investigate bilirubin-induced hearing loss. In vivo auditory brainstem responses revealed that Gunn rats have severe auditory deficits within 18 h of exposure to high bilirubin levels. Using an in vitro preparation of the auditory brainstem from these rats, extracellular multi-electrode array recording from the medial nucleus of the trapezoid body (MNTB) showed longer latency and decreased amplitude of evoked field potentials following bilirubin exposure, suggestive of transmission failure at this synaptic relay. Whole-cell patch-clamp recordings confirmed that the electrophysiological properties of the postsynaptic MNTB neurons were unaffected by bilirubin, with no change in action potential waveforms or current-voltage relationships. However, stimulation of the trapezoid body was unable to elicit large calyceal EPSCs in MNTB neurons of hyperbilirubinaemic rats, indicative of damage at a presynaptic site. Multi-photon imaging of anterograde-labelled calyceal projections revealed axonal staining and presynaptic profiles around MNTB principal neuron somata. Following induction of hyperbilirubinaemia the giant synapses were largely destroyed. Electron microscopy confirmed loss of presynaptic calyceal terminals and supported the electrophysiological evidence for healthy postsynaptic neurons. MNTB neurons express high levels of neuronal nitric oxide synthase (nNOS). Nitric oxide has been implicated in mechanisms of bilirubin toxicity elsewhere in the brain, and antagonism of nNOS by 7-nitroindazole protected hearing during bilirubin exposure. We conclude that bilirubin-induced deafness is caused by degeneration of excitatory synaptic terminals in the auditory brainstem.


Asunto(s)
Pérdida Auditiva/etiología , Hiperbilirrubinemia/complicaciones , Hiperbilirrubinemia/metabolismo , Enfermedad Aguda , Animales , Femenino , Hiperbilirrubinemia/inducido químicamente , Masculino , Neuronas , Técnicas de Placa-Clamp , Ratas , Ratas Gunn , Ratas Wistar , Transducción de Señal , Sulfadimetoxina/toxicidad
17.
Haematologica ; 95(9): 1510-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20378569

RESUMEN

BACKGROUND: Bortezomib has been successfully used in the treatment of multiple myeloma and has been proposed as a potential treatment for chronic lymphocytic leukemia. In this study we investigated the mechanism by which bortezomib induces apoptosis in chronic lymphocytic leukemia cells. DESIGN AND METHODS: Using western blot analysis, we monitored the regulation of BCL2 family members, proteins of the unfolded protein response (endoplasmic reticulum stress response) and activation of caspases in relation to induction of apoptosis (measured by annexin-propidium iodide staining and loss of mitochondrial membrane potential) by bortezomib in chronic lymphocytic leukemia cells. RESULTS: Bortezomib induced apoptosis through activation of the mitochondrial pathway independently of changes associated with endoplasmic reticulum stress. Perturbation of mitochondria was regulated by a rapid and transcription-independent increase of NOXA protein, which preceded release of cytochrome c, HtrA2, Smac and activation of caspase-9 and -3. NOXA had a short half life (approximately 1-2 h) and was ubiquitinated on at least three primary lysine residues, resulting in proteasomal-dependent degradation. Down-regulation of NOXA, using short interfering RNA in chronic lymphocytic leukemia cells, decreased bortezomib-induced apoptosis. Finally bortezomib when combined with seliciclib resulted in a stronger and earlier increase in NOXA protein, caspase-3 cleavage and induction of apoptosis in chronic lymphocytic leukemia cells. CONCLUSIONS: These results highlight a critical role for NOXA in bortezomib-induced apoptosis in chronic lymphocytic leukemia cells and suggest that this drug may become more efficient for the treatment of chronic lymphocytic leukemia if combined with other agents able to interfere with the basal levels of MCL1.


Asunto(s)
Apoptosis/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/patología , Inhibidores de Proteasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Ubiquitinación , Ácidos Borónicos/farmacología , Bortezomib , Humanos , Leucemia Linfocítica Crónica de Células B/metabolismo , Inhibidores de Proteasas/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Pirazinas/farmacología , Células Tumorales Cultivadas
18.
J Cell Biol ; 168(4): 607-18, 2005 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-15716378

RESUMEN

Clostridial neurotoxins are bacterial endopeptidases that cleave the major SNARE proteins in peripheral motorneurons. Here, we show that disruption of synaptic architecture by botulinum neurotoxin C1 (BoNT/C) in central nervous system neurons activates distinct neurodegenerative programs in the axo-dendritic network and in the cell bodies. Neurites degenerate at an early stage by an active caspase-independent fragmentation characterized by segregation of energy competent mitochondria. Later, the cell body mitochondria release cytochrome c, which is followed by caspase activation, apoptotic nuclear condensation, loss of membrane potential, and, finally, cell swelling and lysis. Recognition and scavenging of dying processes by glia also precede the removal of apoptotic cell bodies, in line with a temporal and spatial segregation of different degenerative processes. Our results suggest that, in response to widespread synaptic damage, neurons first dismantle their connections and finally undergo apoptosis, when their spatial relationships are lost.


Asunto(s)
Apoptosis/fisiología , Toxinas Botulínicas/toxicidad , Mitocondrias/metabolismo , Neuronas/citología , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Cerebelo/citología , Citocromos c/metabolismo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Ratones , Microscopía Electrónica de Rastreo , Mitocondrias/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Neuritas , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neurotransmisores/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
19.
J Cell Biol ; 158(6): 1051-66, 2002 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-12235123

RESUMEN

Apoptosis depends critically on regulated cytoskeletal reorganization events in a cell. We demonstrate that death effector domain containing DNA binding protein (DEDD), a highly conserved and ubiquitous death effector domain containing protein, exists predominantly as mono- or diubiquitinated, and that diubiquitinated DEDD interacts with both the K8/18 intermediate filament network and pro-caspase-3. Early in apoptosis, both cytosolic DEDD and its close homologue DEDD2 formed filaments that colocalized with and depended on K8/18 and active caspase-3. Subsequently, these filamentous structures collapsed into intracellular inclusions that migrated into cytoplasmic blebs and contained DEDD, DEDD2, active caspase-3, and caspase-3-cleaved K18 late in apoptosis. Biochemical studies further confirmed that DEDD coimmunoprecipitated with both K18 and pro-caspase-3, and kinetic analyses placed apoptotic DEDD staining prior to caspase-3 activation and K18 cleavage. In addition, both caspase-3 activation and K18 cleavage was inhibited by expression of DEDDDeltaNLS1-3, a cytosolic form of DEDD that cannot be ubiquitinated. Finally, siRNA mediated DEDD knockdown cells exhibited inhibition of staurosporine-induced DNA degradation. Our data suggest that DEDD represents a novel scaffold protein that directs the effector caspase-3 to certain substrates facilitating their ordered degradation during apoptosis.


Asunto(s)
Apoptosis , Proteínas de Unión al ADN/fisiología , Filamentos Intermedios/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/metabolismo , Caspasa 3 , Caspasas/metabolismo , ADN/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Inhibidores Enzimáticos/farmacología , Precursores Enzimáticos/metabolismo , Femenino , Células HeLa , Humanos , Cuerpos de Inclusión/química , Cuerpos de Inclusión/fisiología , Cuerpos de Inclusión/ultraestructura , Filamentos Intermedios/ultraestructura , Células Jurkat , Queratinas/metabolismo , Cinética , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Proteínas Nucleares/metabolismo , ARN Interferente Pequeño , ARN no Traducido/metabolismo , Estaurosporina/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA