Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(5)2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38474278

RESUMEN

The small GTPase RAS acts as a plasma membrane-anchored intracellular neurotrophin counteracting neuronal degeneration in the brain, but the underlying molecular mechanisms are largely unknown. In transgenic mice expressing constitutively activated V12-Ha-RAS selectively in neurons, proteome analysis uncovered a 70% decrease in voltage-dependent anion channel-1 (VDAC-1) in the cortex and hippocampus. We observed a corresponding reduction in the levels of mRNA splicing variant coding for plasma membrane-targeted VDAC-1 (pl-VDAC-1) while mRNA levels for mitochondrial membrane VDAC-1 (mt-VDAC-1) remained constant. In primary cortical neurons derived from V12-Ha-RAS animals, a decrease in pl-VDAC-1 mRNA levels was observed, accompanied by a concomitant reduction in the ferricyanide reductase activity associated with VDAC-1 protein. Application of MEK inhibitor U0126 to transgenic cortical neurons reconstituted pl-VDAC-1 mRNA to reach wild-type levels. Excitotoxic glutamate-induced cell death was strongly attenuated in transgenic V12-Ha-RAS overexpressing cortical cultures. Consistently, a neuroprotective effect could also be achieved in wild-type cortical cultures by the extracellular application of channel-blocking antibody targeting the N-terminus of VDAC-1. These results may encourage novel therapeutic approaches toward blocking pl-VDAC-1 by monoclonal antibody targeting for complementary treatments in transplantation and neurodegenerative disease.


Asunto(s)
Enfermedades Neurodegenerativas , Canales Aniónicos Dependientes del Voltaje , Ratones , Animales , Canales Aniónicos Dependientes del Voltaje/metabolismo , Neuroprotección , Enfermedades Neurodegenerativas/metabolismo , Proteínas ras/metabolismo , Regulación hacia Abajo , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Membrana Celular/metabolismo , Ratones Transgénicos , ARN Mensajero/metabolismo , Canal Aniónico 2 Dependiente del Voltaje/metabolismo
2.
J Biol Chem ; 298(1): 101467, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34871548

RESUMEN

Bacillus anthracis lethal toxin and edema toxin are binary toxins that consist of a common cell-binding moiety, protective antigen (PA), and the enzymatic moieties, lethal factor (LF) and edema factor (EF). PA binds to either of two receptors, capillary morphogenesis protein-2 (CMG-2) or tumor endothelial marker-8 (TEM-8), which triggers the binding and cytoplasmic translocation of LF and EF. However, the distribution of functional TEM-8 and CMG-2 receptors during anthrax toxin intoxication in animals has not been fully elucidated. Herein, we describe an assay to image anthrax toxin intoxication in animals, and we use it to visualize TEM-8- and CMG-2-dependent intoxication in mice. Specifically, we generated a chimeric protein consisting of the N-terminal domain of LF fused to a nuclear localization signal-tagged Cre recombinase (LFn-NLS-Cre). When PA and LFn-NLS-Cre were coadministered to transgenic mice expressing a red fluorescent protein in the absence of Cre and a green fluorescent protein in the presence of Cre, intoxication could be visualized at single-cell resolution by confocal microscopy or flow cytometry. Using this assay, we found that: (a) CMG-2 is critical for intoxication in the liver and heart, (b) TEM-8 is required for intoxication in the kidney and spleen, (c) CMG-2 and TEM-8 are redundant for intoxication of some organs, (d) combined loss of CMG-2 and TEM-8 completely abolishes intoxication, and (e) CMG-2 is the dominant receptor on leukocytes. The novel assay will be useful for basic and clinical/translational studies of Bacillus anthracis infection and for clinical development of reengineered toxin variants for cancer treatment.


Asunto(s)
Carbunco , Antígenos Bacterianos , Bacillus anthracis , Toxinas Bacterianas , Animales , Carbunco/diagnóstico por imagen , Carbunco/metabolismo , Antígenos Bacterianos/química , Antígenos Bacterianos/toxicidad , Bacillus anthracis/metabolismo , Toxinas Bacterianas/toxicidad , Citoplasma/metabolismo , Ratones , Ratones Transgénicos
3.
Int J Mol Sci ; 21(18)2020 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-32911745

RESUMEN

The remote actuation of cellular processes such as migration or neuronal outgrowth is a challenge for future therapeutic applications in regenerative medicine. Among the different methods that have been proposed, the use of magnetic nanoparticles appears to be promising, since magnetic fields can act at a distance without interactions with the surrounding biological system. To control biological processes at a subcellular spatial resolution, magnetic nanoparticles can be used either to induce biochemical reactions locally or to apply forces on different elements of the cell. Here, we show that cell migration and neurite outgrowth can be directed by the forces produced by a switchable parallelized array of micro-magnetic pillars, following the passive uptake of nanoparticles. Using live cell imaging, we first demonstrate that adherent cell migration can be biased toward magnetic pillars and that cells can be reversibly trapped onto these pillars. Second, using differentiated neuronal cells we were able to induce events of neurite outgrowth in the direction of the pillars without impending cell viability. Our results show that the range of forces applied needs to be adapted precisely to the cellular process under consideration. We propose that cellular actuation is the result of the force on the plasma membrane caused by magnetically filled endo-compartments, which exert a pulling force on the cell periphery.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Magnetismo/métodos , Nanopartículas de Magnetita/uso terapéutico , Espacio Intracelular/fisiología , Campos Magnéticos , Nanopartículas de Magnetita/análisis , Fenómenos Mecánicos , Proyección Neuronal/efectos de los fármacos , Fenómenos Físicos , Medicina Regenerativa/métodos
5.
Hum Mol Genet ; 25(24): 5365-5382, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27794539

RESUMEN

Mutations in leucine-rich repeat kinase 2 (LRRK2) cause late-onset, autosomal dominant familial Parkinson`s disease (PD) and variation at the LRRK2 locus contributes to the risk for idiopathic PD. LRRK2 can function as a protein kinase and mutations lead to increased kinase activity. To elucidate the pathophysiological mechanism of the R1441C mutation in the GTPase domain of LRRK2, we expressed human wild-type or R1441C LRRK2 in dopaminergic neurons of Drosophila and observe reduced locomotor activity, impaired survival and an age-dependent degeneration of dopaminergic neurons thereby creating a new PD-like model. To explore the function of LRRK2 variants in vivo, we performed mass spectrometry and quantified 3,616 proteins in the fly brain. We identify several differentially-expressed cytoskeletal, mitochondrial and synaptic vesicle proteins (SV), including synaptotagmin-1, syntaxin-1A and Rab3, in the brain of this LRRK2 fly model. In addition, a global phosphoproteome analysis reveals the enhanced phosphorylation of several SV proteins, including synaptojanin-1 (pThr1131) and the microtubule-associated protein futsch (pSer4106) in the brain of R1441C hLRRK2 flies. The direct phosphorylation of human synaptojanin-1 by R1441C hLRRK2 could further be confirmed by in vitro kinase assays. A protein-protein interaction screen in the fly brain confirms that LRRK2 robustly interacts with numerous SV proteins, including synaptojanin-1 and EndophilinA. Our proteomic, phosphoproteomic and interactome study in the Drosophila brain provides a systematic analyses of R1441C hLRRK2-induced pathobiological mechanisms in this model. We demonstrate for the first time that the R1441C mutation located within the LRRK2 GTPase domain induces the enhanced phosphorylation of SV proteins in the brain.


Asunto(s)
Encéfalo/metabolismo , Neuronas Dopaminérgicas/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Enfermedad de Parkinson/genética , Proteoma/genética , Animales , Animales Modificados Genéticamente , Encéfalo/patología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/biosíntesis , Mutación , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Monoéster Fosfórico Hidrolasas/biosíntesis , Monoéster Fosfórico Hidrolasas/genética , Fosforilación , Mapas de Interacción de Proteínas , Vesículas Sinápticas/genética , Sinaptotagmina I/biosíntesis , Sinaptotagmina I/genética , Sintaxina 1/biosíntesis , Sintaxina 1/genética , Proteínas de Unión al GTP rab3/biosíntesis , Proteínas de Unión al GTP rab3/genética
6.
Biol Chem ; 404(10): 881-882, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37655951
7.
Biol Chem ; 399(7): 643-647, 2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29537215

RESUMEN

Mutations in human leucine-rich-repeat kinase 2 (LRRK2) have been found to be the most frequent cause of late-onset Parkinson's Disease (PD). LRRK2 is a large protein with two enzymatic domains, a GTPase and a kinase domain. A cluster of (auto)-phosphorylation sites within the N-terminus of LRRK2 have been shown to be crucial for the localization of LRRK2 and is important for PD pathogenesis. In addition, phosphorylation of sites within the G-domain of the protein affect GTPase activity. Here we discuss the role of these (auto)-phosphorylation sites of LRRK2 and their regulation by phosphatases and upstream kinases.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Fosfotransferasas/metabolismo
8.
Int J Mol Sci ; 19(12)2018 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-30558189

RESUMEN

Cellular activation of RAS GTPases into the GTP-binding "ON" state is a key switch for regulating brain functions. Molecular protein structural elements of rat sarcoma (RAS) and RAS homolog protein enriched in brain (RHEB) GTPases involved in this switch are discussed including their subcellular membrane localization for triggering specific signaling pathways resulting in regulation of synaptic connectivity, axonal growth, differentiation, migration, cytoskeletal dynamics, neural protection, and apoptosis. A beneficial role of neuronal H-RAS activity is suggested from cellular and animal models of neurodegenerative diseases. Recent experiments on optogenetic regulation offer insights into the spatiotemporal aspects controlling RAS/mitogen activated protein kinase (MAPK) or phosphoinositide-3 kinase (PI3K) pathways. As optogenetic manipulation of cellular signaling in deep brain regions critically requires penetration of light through large distances of absorbing tissue, we discuss magnetic guidance of re-growing axons as a complementary approach. In Parkinson's disease, dopaminergic neuronal cell bodies degenerate in the substantia nigra. Current human trials of stem cell-derived dopaminergic neurons must take into account the inability of neuronal axons navigating over a large distance from the grafted site into striatal target regions. Grafting dopaminergic precursor neurons directly into the degenerating substantia nigra is discussed as a novel concept aiming to guide axonal growth by activating GTPase signaling through protein-functionalized intracellular magnetic nanoparticles responding to external magnets.


Asunto(s)
Encéfalo/fisiología , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Proteínas ras/metabolismo , Animales , Diferenciación Celular , Movimiento Celular , Humanos , Neurogénesis , Optogenética , Transducción de Señal
9.
Biol Chem ; 398(5-6): 589-606, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28212107

RESUMEN

The small GTPase Rheb was originally detected as an immediate early response protein whose expression was induced by NMDA-dependent synaptic activity in the brain. Rheb's activity is highly regulated by its GTPase activating protein (GAP), the tuberous sclerosis complex protein, which stimulates the conversion from the active, GTP-loaded into the inactive, GDP-loaded conformation. Rheb has been established as an evolutionarily conserved molecular switch protein regulating cellular growth, cell volume, cell cycle, autophagy, and amino acid uptake. The subcellular localization of Rheb and its interacting proteins critically regulate its activity and function. In stem cells, constitutive activation of Rheb enhances differentiation at the expense of self-renewal partially explaining the adverse effects of deregulated Rheb in the mammalian brain. In the context of various cellular stress conditions such as oxidative stress, ER-stress, death factor signaling, and cellular aging, Rheb activation surprisingly enhances rather than prevents cellular degeneration. This review addresses cell type- and cell state-specific function(s) of Rheb and mainly focuses on neurons and their surrounding glial cells. Mechanisms will be discussed in the context of therapy that interferes with Rheb's activity using the antibiotic rapamycin or low molecular weight compounds.


Asunto(s)
Proteínas de Unión al GTP Monoméricas/metabolismo , Neuronas/citología , Animales , Apoptosis , Proliferación Celular , Humanos , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/metabolismo , Transporte de Proteínas
10.
Biol Chem ; 398(5-6): 577-588, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28475102

RESUMEN

Ras GTPases are key players in cellular signalling because they act as binary switches. These states manifest through toggling between an active (GTP-loaded) and an inactive (GDP-loaded) form. The hydrolysis and replenishing of GTP is controlled by two additional protein classes: GAP (GTPase-activating)- and GEF (Guanine nucleotide exchange factors)-proteins. The complex interplay of the proteins is known as the GTPase-cycle. Several point mutations of the Ras protein deregulate this cycle. Mutations in Ras are associated with up to one-third of human cancers. The three isoforms of Ras (H, N, K) exhibit high sequence similarity and mainly differ in a region called HVR (hypervariable region). The HVR governs the differential action and cellular distribution of the three isoforms. Rheb is a Ras-like GTPase that is conserved from yeast to mammals. Rheb is mainly involved in activation of cell growth through stimulation of mTORC1 activity. In this review, we summarise multidimensional NMR studies on Rheb and Ras carried out to characterise their structure-function relationship and explain how the activity of these small GTPases can be modulated by low molecular weight compounds. These might help to design GTPase-selective antagonists for treatment of cancer and brain disease.


Asunto(s)
Espectroscopía de Resonancia Magnética/métodos , Proteínas ras/química , Proteínas ras/metabolismo , Animales , Proteínas Activadoras de GTPasa/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Bibliotecas de Moléculas Pequeñas/farmacología
11.
Biol Chem ; 397(3): 207-14, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26641965

RESUMEN

Homer1a is upregulated by several different antidepressant measures, including non-pharmacological treatments, like sleep deprivation (SD) and electroconvulsive therapy (ECT) and antidepressant drugs, such as imipramine, fluoxetine and ketamine. Homer1a induction might thus be a crucial joint mechanism for antidepressant therapy in general. However, the upstream signaling pathways that regulate or induce Homer1a expression are still not well understood. The main focus of the present review is to offer an overview of the current knowledge about the potential role of Homer1a in depression and the signaling pathways responsible for Homer1a regulation. It is suggested here that a detailed characterization of the signaling mechanisms leading to Homer1a expression might provide novel therapeutic targets for antidepressant drug development.


Asunto(s)
Proteínas Portadoras/genética , Depresión/metabolismo , Regulación de la Expresión Génica , Transducción de Señal , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Depresión/tratamiento farmacológico , Depresión/genética , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/genética , Trastorno Depresivo/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Andamiaje Homer , Humanos , Receptor de Adenosina A1/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas ras/metabolismo
12.
Biol Chem ; 397(6): 541-54, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26894577

RESUMEN

Mutations in the gene coding for the multi-domain protein leucine-rich repeat kinase 2 (LRRK2) are the leading cause of genetically inherited Parkinson's disease (PD). Two of the common found mutations are the R1441C and G2019S. In this study we identified protein phosphatase 2A (PP2A) as an interacting partner of LRRK2. We were able to demonstrate that the Ras of complex protein (ROC) domain is sufficient to interact with the three subunits of PP2A in human neuroblastoma SH-SY5Y cells and in HeLa cells. The alpha subunit of PP2A is interacting with LRRK2 in the perinuclear region of HeLa cells. Silencing the catalytic subunit of PP2A by shRNA aggravated cellular degeneration induced by the pathogenic R1441C-LRRK2 mutant expressed in neuroblastoma SH-SY5Y cells. A similar enhancement of apoptotic nuclei was observed by downregulation of the catalytic subunit of PP2A in cultured cortical cells derived from neurons overexpressing the pathogenic mutant G2019S-LRRK2. Conversely, pharmacological activation of PP2A by sodium selenate showed a partial neuroprotection from R1441C-LRRK2-induced cellular degeneration. All these data suggest that PP2A is a new interacting partner of LRRK2 and reveal the importance of PP2A as a potential therapeutic target in PD.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Proteína Fosfatasa 2/metabolismo , Dominio Catalítico , Muerte Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/química , Neuronas/citología , Neuronas/efectos de los fármacos , Unión Proteica , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/genética , Ácido Selénico/farmacología
13.
Brain Behav Immun ; 52: 106-119, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26456693

RESUMEN

Cerebral white matter injury is a leading cause of adverse neurodevelopmental outcome in prematurely born infants involving cognitive deficits in later life. Despite increasing knowledge about the pathophysiology of perinatal brain injury, therapeutic options are limited. In the adult demyelinating disease multiple sclerosis the sphingosine-1-phosphate (S1P) receptor modulating substance fingolimod (FTY720) has beneficial effects. Herein, we evaluated the neuroprotective potential of FTY720 in a neonatal model of oxygen-toxicity, which is associated with hypomyelination and impaired neuro-cognitive outcome. A single dose of FTY720 (1mg/kg) at the onset of neonatal hyperoxia (24h 80% oxygen on postnatal day 6) resulted in improvement of neuro-cognitive development persisting into adulthood. This was associated with reduced microstructural white matter abnormalities 4 months after the insult. In search of the underlying mechanisms potential non-classical (i.e. lymphocyte-independent) pathways were analysed shortly after the insult, comprising modulation of oxidative stress and local inflammatory responses as well as myelination, oligodendrocyte degeneration and maturation. Treatment with FTY720 reduced hyperoxia-induced oxidative stress, microglia activation and associated pro-inflammatory cytokine expression. In vivo and in vitro analyses further revealed that oxygen-induced hypomyelination is restored to control levels, which was accompanied by reduced oligodendrocyte degeneration and enhanced maturation. Furthermore, hyperoxia-induced elevation of S1P receptor 1 (S1P1) protein expression on in vitro cultured oligodendrocyte precursor cells was reduced by activated FTY720 and protection from degeneration is abrogated after selective S1P1 blockade. Finally, FTY720s' classical mode of action (i.e. retention of immune cells within peripheral lymphoid organs) was analysed demonstrating that FTY720 diminished circulating lymphocyte counts independent from hyperoxia. Cerebral immune cell counts remained unchanged by hyperoxia and by FTY720 treatment. Taken together, these results suggest that beneficial effects of FTY720 in neonatal oxygen-induced brain injury may be rather attributed to its anti-oxidative and anti-inflammatory capacity acting in concert with a direct protection of developing oligodendrocytes than to a modulation of peripheral lymphocyte trafficking. Thus, FTY720 might be a potential new therapeutic option for the treatment of neonatal brain injury through reduction of white matter damage.


Asunto(s)
Trastornos del Conocimiento/prevención & control , Clorhidrato de Fingolimod/uso terapéutico , Hiperoxia/tratamiento farmacológico , Sustancia Blanca/efectos de los fármacos , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Imagen de Difusión por Resonancia Magnética , Femenino , Hiperoxia/patología , Lisofosfolípidos/metabolismo , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Fibras Nerviosas Mielínicas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/patología , Oxígeno/administración & dosificación , Embarazo , Distribución Aleatoria , Ratas , Ratas Wistar , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
14.
J Neurochem ; 130(4): 472-89, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24773031

RESUMEN

Dopamine replacement therapy in Parkinson's disease is associated with several unwanted effects, of which dyskinesia is the most disabling. The development of new therapeutic interventions to reduce the impact of dyskinesia in Parkinson's disease is therefore a priority need. This review summarizes the key molecular mechanisms that underlie dyskinesia. The role of dopamine receptors and their associated signaling mechanisms including dopamine-cAMP-regulated neuronal phosphoprotein, extracellular signal-regulated kinase, mammalian target of rapamycin, mitogen and stress-activated kinase-1 and Histone H3 are summarized, along with an evaluation of the role of cannabinoid and nicotinic acetylcholine receptors. The role of synaptic plasticity and animal behavioral results on dyskinesia are also evaluated. The most recent therapeutic advances to treat Parkinson's disease are discussed, with emphasis on the possibilities and limitations of non-pharmacological interventions such as physical activity, deep brain stimulation, transcranial magnetic field stimulation and cell replacement therapy. The review suggests new prospects for the management of Parkinson's disease-associated motor symptoms, especially the development of dyskinesia. This review aims at summarizing the key molecular mechanisms underlying dyskinesia and the most recent therapeutic advances to treat Parkinson's disease with emphasis on non-pharmacological interventions such as physical activity, deep brain stimulation (DBS), transcranial magnetic field stimulation (TMS) and cell replacement therapy. These new interventions are discussed from both the experimental and clinical point of view, describing their current strength and limitations.


Asunto(s)
Discinesias/etiología , Discinesias/terapia , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/terapia , Animales , Cannabinoides/farmacología , Trasplante de Células , Cromatina/efectos de los fármacos , Estimulación Encefálica Profunda , Dopaminérgicos/uso terapéutico , Fosfoproteína 32 Regulada por Dopamina y AMPc/fisiología , Discinesias/fisiopatología , Histonas/metabolismo , Humanos , Levodopa/farmacología , Levodopa/uso terapéutico , Actividad Motora/fisiología , Enfermedad de Parkinson/fisiopatología , Fosforilación , Desempeño Psicomotor/fisiología , Receptores de Dopamina D1/efectos de los fármacos , Receptores de Dopamina D1/metabolismo , Transducción de Señal/efectos de los fármacos , Estimulación Magnética Transcraneal
16.
Biol Chem ; 394(12): 1595-605, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23912219

RESUMEN

Methyl-CpG binding protein 2 (MeCP2), a nuclear protein highly expressed in neurons, was identified because of its ability to bind methylated DNA. In association with the transcriptional corepressor proteins Sin3a and histone deacetylases, it represses gene transcription. However, it has since become clear that MeCP2 is a multifunctional protein involved not only in transcriptional silencing but also in transcriptional activation, chromatin remodeling, and RNA splicing. Especially, its involvement in the X-linked neurologic disorder Rett syndrome emphasizes the importance of MeCP2 for normal development and maturation of the central nervous system. A number of animal models with complete or partial lack of MeCP2 functions have been generated to correlate the clinical phenotype of Rett syndrome, and studies involving different mutations of MeCP2 have shown similar effects. Animal model studies have further demonstrated that even the loss of a specific phosphorylation site of MeCP2 (S80, S421, and S424) disturbs normal maturation of the mammalian brain. This review covers recent findings regarding MeCP2 functions and its regulation by posttranslational modification, particularly MeCP2 phosphorylation and its effects on mammalian brain maturation, learning, and plasticity.


Asunto(s)
Encéfalo/fisiología , Aprendizaje/fisiología , Proteína 2 de Unión a Metil-CpG/metabolismo , Transcripción Genética , Animales , Encéfalo/crecimiento & desarrollo , Citosina/metabolismo , Metilación de ADN , Regulación de la Expresión Génica , Humanos , Memoria/fisiología , Proteína 2 de Unión a Metil-CpG/genética , Trastornos del Humor/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Trastornos Relacionados con Sustancias/metabolismo
17.
Int J Mol Sci ; 13(10): 12939-51, 2012 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23202931

RESUMEN

Autophagy is a self-degradative process that involves turnover and recycling of cytoplasmic components in healthy and diseased tissue. Autophagy has been shown to be protective at the early stages of programmed cell death but it can also promote apoptosis under certain conditions. Earlier we demonstrated that oxygen contributes to the pathogenesis of neonatal brain damage, which can be ameliorated by intervention with recombinant human erythropoietin (rhEpo). Extrinsic- and intrinsic apoptotic pathways are involved in oxygen induced neurotoxicity but the role of autophagy in this model is unclear. We analyzed the expression of autophagy activity markers in the immature rodent brain after exposure to elevated oxygen concentrations. We observed a hyperoxia-exposure dependent regulation of autophagy-related gene (Atg) proteins Atg3, 5, 12, Beclin-1, microtubule-associated protein 1 light chain 3 (LC3), LC3A-II, and LC3B-II which are all key autophagy activity proteins. Interestingly, a single injection with rhEpo at the onset of hyperoxia counteracted these oxygen-mediated effects. Our results indicate that rhEpo generates its protective effect by modifying the key autophagy activity proteins.


Asunto(s)
Autofagia/efectos de los fármacos , Encéfalo/metabolismo , Eritropoyetina/farmacología , Hipoxia , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 5 Relacionada con la Autofagia , Proteínas Relacionadas con la Autofagia , Beclina-1 , Encéfalo/efectos de los fármacos , Epoetina alfa , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Animales , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos
18.
J Biol Chem ; 285(44): 33979-91, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20685651

RESUMEN

Rheb is a homolog of Ras GTPase that regulates cell growth, proliferation, and regeneration via mammalian target of rapamycin (mTOR). Because of the well established potential of activated Ras to promote survival, we sought to investigate the ability of Rheb signaling to phenocopy Ras. We found that overexpression of lipid-anchored Rheb enhanced the apoptotic effects induced by UV light, TNFα, or tunicamycin in an mTOR complex 1 (mTORC1)-dependent manner. Knocking down endogenous Rheb or applying rapamycin led to partial protection, identifying Rheb as a mediator of cell death. Ras and c-Raf kinase opposed the apoptotic effects induced by UV light or TNFα but did not prevent Rheb-mediated apoptosis. To gain structural insight into the signaling mechanisms, we determined the structure of Rheb-GDP by NMR. The complex adopts the typical canonical fold of RasGTPases and displays the characteristic GDP-dependent picosecond to nanosecond backbone dynamics of the switch I and switch II regions. NMR revealed Ras effector-like binding of activated Rheb to the c-Raf-Ras-binding domain (RBD), but the affinity was 1000-fold lower than the Ras/RBD interaction, suggesting a lack of functional interaction. shRNA-mediated knockdown of apoptosis signal-regulating kinase 1 (ASK-1) strongly reduced UV or TNFα-induced apoptosis and suppressed enhancement by Rheb overexpression. In conclusion, Rheb-mTOR activation not only promotes normal cell growth but also enhances apoptosis in response to diverse toxic stimuli via an ASK-1-mediated mechanism. Pharmacological regulation of the Rheb/mTORC1 pathway using rapamycin should take the presence of cellular stress into consideration, as this may have clinical implications.


Asunto(s)
Apoptosis , Proteínas de Unión al GTP Monoméricas/metabolismo , Neuropéptidos/metabolismo , Secuencia de Aminoácidos , Retículo Endoplásmico/metabolismo , Células HeLa , Humanos , Espectroscopía de Resonancia Magnética/métodos , Diana Mecanicista del Complejo 1 de la Rapamicina , Conformación Molecular , Datos de Secuencia Molecular , Complejos Multiproteicos , Neuronas/metabolismo , Estrés Oxidativo , Proteínas , Proteína Homóloga de Ras Enriquecida en el Cerebro , Homología de Secuencia de Aminoácido , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Factores de Transcripción/metabolismo
19.
J Cell Sci ; 122(Pt 18): 3374-84, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19706687

RESUMEN

Multi-PDZ (PSD-95/Discs large/Zonula-occludens-1) domain proteins play a crucial role in the establishment and maintenance of cell polarization. The novel multi-PDZ domain protein FRMPD2 is a potential scaffolding protein consisting of an N-terminal KIND domain, a FERM domain and three PDZ domains. Here we show that FRMPD2 is localized in a polarized fashion in epithelial cells at the basolateral membrane and partially colocalizes with the tight-junction marker protein Zonula-occludens-1. Downregulation of FRMPD2 protein in Caco-2 cells is associated with an impairment of tight junction formation. We find that the FERM domain of FRMPD2 binds phosphatidylinositols and is sufficient for membrane localization. Moreover, we demonstrate that recruitment of FRMPD2 to cell-cell junctions is strictly E-cadherin-dependent, which is in line with our identification of catenin family proteins as binding partners for FRMPD2. We demonstrate that the FERM domain and binding of the PDZ2 domain to the armadillo protein p0071 are required for basolateral restriction of FRMPD2. Moreover, the PDZ2 domain of FRMPD2 is sufficient to partially redirect an apically localized protein to the basolateral membrane. Our results provide novel insights into the molecular function of FRMPD2 and into the targeting mechanism of peripheral membrane proteins in polarized epithelial cells.


Asunto(s)
Polaridad Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas de la Membrana/metabolismo , Dominios PDZ , Secuencia de Aminoácidos , Animales , Proteínas del Dominio Armadillo/metabolismo , Biomarcadores/metabolismo , Cadherinas/metabolismo , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Perros , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/química , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Fosfoproteínas/metabolismo , Unión Proteica , Transporte de Proteínas , Proteínas de Uniones Estrechas , Uniones Estrechas/metabolismo , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA