Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 11(17)2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-36078064

RESUMEN

Astrocytes, the main glial cells of the central nervous system, play a key role in brain volume control due to their intimate contacts with cerebral blood vessels and the expression of a distinctive equipment of proteins involved in solute/water transport. Among these is MLC1, a protein highly expressed in perivascular astrocytes and whose mutations cause megalencephalic leukoencephalopathy with subcortical cysts (MLC), an incurable leukodystrophy characterized by macrocephaly, chronic brain edema, cysts, myelin vacuolation, and astrocyte swelling. Although, in astrocytes, MLC1 mutations are known to affect the swelling-activated chloride currents (ICl,swell) mediated by the volume-regulated anion channel (VRAC), and the regulatory volume decrease, MLC1's proper function is still unknown. By combining molecular, biochemical, proteomic, electrophysiological, and imaging techniques, we here show that MLC1 is a Ca2+/Calmodulin-dependent protein kinase II (CaMKII) target protein, whose phosphorylation, occurring in response to intracellular Ca2+ release, potentiates VRAC-mediated ICl,swell. Overall, these findings reveal that MLC1 is a Ca2+-regulated protein, linking volume regulation to Ca2+ signaling in astrocytes. This knowledge provides new insight into the MLC1 protein function and into the mechanisms controlling ion/water exchanges in the brain, which may help identify possible molecular targets for the treatment of MLC and other pathological conditions caused by astrocyte swelling and brain edema.


Asunto(s)
Edema Encefálico , Quistes , Astrocitos/metabolismo , Edema Encefálico/patología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Cloruros/metabolismo , Quistes/metabolismo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias , Humanos , Proteínas de la Membrana/metabolismo , Proteómica , Canales Aniónicos Dependientes del Voltaje/metabolismo , Agua/metabolismo
2.
Front Immunol ; 12: 649475, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33936070

RESUMEN

In human primary dendritic cells (DC) rapamycin-an autophagy inducer and protein synthesis inhibitor-overcomes the autophagy block induced by Mycobacterium tuberculosis (Mtb) and promotes a Th1 response via IL-12 secretion. Here, the immunostimulatory activity of rapamycin in Mtb-infected DC was further investigated by analyzing both transcriptome and translatome gene profiles. Hundreds of differentially expressed genes (DEGs) were identified by transcriptome and translatome analyses of Mtb-infected DC, and some of these genes were found further modulated by rapamycin. The majority of transcriptome-associated DEGs overlapped with those present in the translatome, suggesting that transcriptionally stimulated mRNAs are also actively translated. In silico analysis of DEGs revealed significant changes in intracellular cascades related to cytokine production, cytokine-induced signaling and immune response to pathogens. In particular, rapamycin treatment of Mtb-infected DC caused an enrichment of IFN-ß, IFN-λ and IFN-stimulated gene transcripts in the polysome-associated RNA fraction. In addition, rapamycin led to an increase of IL-12, IL-23, IL-1ß, IL-6, and TNF-α but to a reduction of IL-10. Interestingly, upon silencing or pharmacological inhibition of GSK-3ß, the rapamycin-driven modulation of the pro- and anti-inflammatory cytokine balance was lost, indicating that, in Mtb-infected DC, GSK-3ß acts as molecular switch for the regulation of the cytokine milieu. In conclusion, our study sheds light on the molecular mechanism by which autophagy induction contributes to DC activation during Mtb infection and points to rapamycin and GSK-3ß modulators as promising compounds for host-directed therapy in the control of Mtb infection.


Asunto(s)
Autofagia/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Mycobacterium tuberculosis/inmunología , Sirolimus/farmacología , Tuberculosis/tratamiento farmacológico , Autofagia/genética , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/inmunología , Perfilación de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Cultivo Primario de Células , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/metabolismo , Tuberculosis/inmunología , Tuberculosis/microbiología
3.
Sci Rep ; 9(1): 5144, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30914688

RESUMEN

Amyloid protein misfolding results in a self-assembling aggregation process, characterized by the formation of typical aggregates. The attention is focused on pre-fibrillar oligomers (PFOs), formed in the early stages and supposed to be neurotoxic. PFOs structure may change due to their instability and different experimental protocols. Consequently, it is difficult to ascertain which aggregation species are actually neurotoxic. We used salmon Calcitonin (sCT) as an amyloid model whose slow aggregation rate allowed to prepare stable samples without photochemical cross-linking. Intracellular Ca2+ rise plays a fundamental role in amyloid protein-induced neurodegerations. Two paradigms have been explored: (i) the "membrane permeabilization" due to the formation of amyloid pores or other types of membrane damage; (ii) "receptor-mediated" modulation of Ca2+ channels. In the present paper, we tested the effects of native sCT PFOs- with respect to Monomer-enriched solutions in neurons characterized by an increasing degree of differentiation, in terms of -Ca2+-influx, cellular viability, -Long-Term Potentiation impairment, Post-Synaptic Densities and synaptophysin expression. Results indicated that PFOs-, but not Monomer-enriched solutions, induced abnormal -Ca2+-influx, which could only in part be ascribed to NMDAR activation. Thus, we propose an innovative neurotoxicity mechanism for amyloid proteins where "membrane permeabilization" and "receptor-mediated" paradigms coexist.


Asunto(s)
Amiloide/toxicidad , Calcitonina/toxicidad , Señalización del Calcio/efectos de los fármacos , Membrana Celular/metabolismo , Proteínas de Peces/toxicidad , Potenciación a Largo Plazo/efectos de los fármacos , N-Metilaspartato/metabolismo , Neuronas/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Multimerización de Proteína , Salmón , Amiloide/química , Animales , Calcitonina/química , Calcio/metabolismo , Línea Celular , Membrana Celular/patología , Proteínas de Peces/química , Ratones , Neuronas/patología , Síndromes de Neurotoxicidad/patología
4.
Biomolecules ; 10(1)2019 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-31905804

RESUMEN

To investigate the interaction between amyloid assemblies and "lipid-rafts", we performed functional and structural experiments on salmon calcitonin (sCT) solutions rich in prefibrillar oligomers, proto- and mature-fibers interacting with liposomes made of monosialoganglioside-GM1 (4%), DPPC (48%) and cholesterol (48%). To focus on the role played by electrostatic forces and considering that sCT is positive and GM1 is negative at physiologic pH, we compared results with those relative to GM1-free liposomes while, to assess membrane fluidity effects, with those relative to cholesterol-free liposomes. We investigated functional effects by evaluating Ca2+-influx in liposomes and viability of HT22-DIFF neurons. Only neurotoxic solutions rich in unstructured prefibrillar oligomers were able to induce Ca2+-influx in the "lipid-rafts" model, suggesting that the two phenomena were correlated. Thus, we investigated protein conformation and membrane modifications occurring during the interaction: circular dichroism showed that "lipid-rafts" fostered the formation of ß-structures and energy filtered-transmission electron microscopy that prefibrillar oligomers formed pores, similar to Aß did. We speculate that electrostatic forces between the positive prefibrillar oligomers and the negative GM1 drive the initial binding while the hydrophobic profile and flexibility of prefibrillar oligomers, together with the membrane fluidity, are responsible for the subsequent pore formation leading to Ca2+-influx and neurotoxicity.


Asunto(s)
Amiloide/metabolismo , Calcitonina/química , Calcitonina/toxicidad , Calcio/metabolismo , Microdominios de Membrana/metabolismo , Neuronas/metabolismo , Neuronas/patología , Amiloide/química , Amiloide/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Microdominios de Membrana/química , Microdominios de Membrana/efectos de los fármacos , Ratones , Modelos Biológicos , Neuronas/efectos de los fármacos
5.
PLoS One ; 11(5): e0156325, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27223470

RESUMEN

Duchenne Muscular Dystrophy, a genetic disorder that results in a gradual breakdown of muscle, is associated to mild to severe cognitive impairment in about one-third of dystrophic patients. The brain dysfunction is independent of the muscular pathology, occurs early, and is most likely due to defects in the assembly of the Dystrophin-associated Protein Complex (DPC) during embryogenesis. We have recently described the interaction of the DPC component ß-dystrobrevin with members of complexes that regulate chromatin dynamics, and suggested that ß-dystrobrevin may play a role in the initiation of neuronal differentiation. Since oxygen concentrations and miRNAs appear as well to be involved in the cellular processes related to neuronal development, we have studied how these factors act on ß-dystrobrevin and investigated the possibility of their functional interplay using the NTera-2 cell line, a well-established model for studying neurogenesis. We followed the pattern of expression and regulation of ß-dystrobrevin during the early stages of neuronal differentiation induced by exposure to retinoic acid (RA) under hypoxia as compared with normoxia, and found that ß-dystrobrevin expression is regulated during RA-induced differentiation of NTera-2 cells. We also found that ß-dystrobrevin pattern is delayed under hypoxic conditions, together with a delay in the differentiation and an increase in the proliferation rate of cells. We identified miRNA-143 as a direct regulator of ß-dystrobrevin expression, demonstrated that ß-dystrobrevin is expressed in the nucleus and showed that, in line with our previous in vitro results, ß-dystrobrevin is a repressor of synapsin I in live cells. Altogether the newly identified regulatory pathway miR-143/ß-dystrobrevin/synapsin I provides novel insights into the functions of ß-dystrobrevin and opens up new perspectives for elucidating the molecular mechanisms underlying the neuronal involvement in muscular dystrophy.


Asunto(s)
Proteínas Asociadas a la Distrofina/genética , Proteínas Asociadas a la Distrofina/metabolismo , MicroARNs/genética , Neurogénesis , Neuropéptidos/genética , Neuropéptidos/metabolismo , Regiones no Traducidas 3' , Diferenciación Celular , Hipoxia de la Célula , Línea Celular Tumoral , Núcleo Celular/genética , Proliferación Celular , Humanos , Neuronas/citología , Neuronas/efectos de los fármacos , Tretinoina/farmacología
6.
Biochim Biophys Acta ; 1842(9): 1622-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24932517

RESUMEN

Many proteins belonging to the amyloid family share the tendency to misfold and aggregate following common steps, and display similar neurotoxicity. In the aggregation pathway different kinds of species are formed, including several types of oligomers and eventually mature fibers. It is now suggested that the pathogenic aggregates are not the mature fibrils, but the intermediate, soluble oligomers. Many kinds of aggregates have been described to exist in a metastable state and in equilibrium with monomers. Up to now it is not clear whether a specific structure is at the basis of the neurotoxicity. Here we characterized, starting from the early aggregation stages, the oligomer populations formed by an amyloid protein, salmon calcitonin (sCT), chosen due to its very slow aggregation rate. To prepare different oligomer populations and characterize them by means of photoinduced cross-linking SDS-PAGE, Energy Filtered-Transmission Electron Microscopy (EF-TEM) and Circular Dichroism (CD) spectroscopy, we used Size Exclusion Chromatography (SEC), a technique that does not influence the aggregation process leaving the protein in the native state. Taking advantage of sCT low aggregation rate, we characterized the neurotoxic potential of the SEC-separated, non-crosslinked fractions in cultured primary hippocampal neurons, analyzing intracellular Ca(2+) influx and apoptotic trend. We provide evidence that native, globular, metastable, prefibrillar oligomers (dimers, trimers and tetramers) were the toxic species and that low concentrations of these aggregates in the population was sufficient to render the sample neurotoxic. Monomers and other kind of aggregates, such as annular or linear protofibers and mature fibers, were totally biologically inactive.


Asunto(s)
Amiloide/química , Amiloide/toxicidad , Encéfalo/patología , Hipocampo/patología , Animales , Encéfalo/efectos de los fármacos , Calcio/metabolismo , Células Cultivadas , Cromatografía en Gel , Dicroismo Circular , Reactivos de Enlaces Cruzados/farmacología , Dimerización , Electrofisiología , Hipocampo/efectos de los fármacos , Microscopía Electrónica de Transmisión , Fotoquímica , Ratas , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
7.
Oncotarget ; 4(12): 2271-87, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24345856

RESUMEN

LINE-1 elements make up the most abundant retrotransposon family in the human genome. Full-length LINE-1 elements encode a reverse transcriptase (RT) activity required for their own retrotranpsosition as well as that of non-autonomous Alu elements. LINE-1 are poorly expressed in normal cells and abundantly in cancer cells. Decreasing RT activity in cancer cells, by either LINE-1-specific RNA interference, or by RT inhibitory drugs, was previously found to reduce proliferation and promote differentiation and to antagonize tumor growth in animal models. Here we have investigated how RT exerts these global regulatory functions. We report that the RT inhibitor efavirenz (EFV) selectively downregulates proliferation of transformed cell lines, while exerting only mild effects on non-transformed cells; this differential sensitivity matches a differential RT abundance, which is high in the former and undetectable in the latter. Using CsCl density gradients, we selectively identify Alu and LINE-1 containing DNA:RNA hybrid molecules in cancer but not in normal cells. Remarkably, hybrid molecules fail to form in tumor cells treated with EFV under the same conditions that repress proliferation and induce the reprogramming of expression profiles of coding genes, microRNAs (miRNAs) and ultraconserved regions (UCRs). The RT-sensitive miRNAs and UCRs are significantly associated with Alu sequences. The results suggest that LINE-1-encoded RT governs the balance between single-stranded and double-stranded RNA production. In cancer cells the abundant RT reverse-transcribes retroelement-derived mRNAs forming RNA:DNA hybrids. We propose that this impairs the formation of double-stranded RNAs and the ensuing production of small regulatory RNAs, with a direct impact on gene expression. RT inhibition restores the 'normal' small RNA profile and the regulatory networks that depend on them. Thus, the retrotransposon-encoded RT drives a previously unrecognized mechanism crucial to the transformed state in tumor cells.


Asunto(s)
Elementos de Nucleótido Esparcido Largo , Neoplasias/genética , ADN Polimerasa Dirigida por ARN/genética , Diferenciación Celular/genética , Procesos de Crecimiento Celular/genética , Línea Celular Transformada , Línea Celular Tumoral , ADN de Neoplasias/genética , Humanos , Melanoma/enzimología , Melanoma/genética , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias/enzimología , ARN Neoplásico/genética , ADN Polimerasa Dirigida por ARN/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología , Transcriptoma/efectos de los fármacos , Transcriptoma/genética
8.
Exp Eye Res ; 116: 1-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23954924

RESUMEN

Dysbindin, the product of the DTNBP1 gene, was identified by yeast two hybrid assay as a binding partner of dystrobrevin, a cytosolic component of the dystrophin protein complex. Although its functional role has not yet been completely elucidated, the finding that dysbindin assembles into the biogenesis of lysosome related organelles complex 1 (BLOC-1) suggests that it participates in intracellular trafficking and biogenesis of organelles and vesicles. Dysbindin is ubiquitous and in brain is expressed primarily in neurons. Variations at the dysbindin gene have been associated with increased risk for schizophrenia. As anomalies in retinal function have been reported in patients suffering from neuropsychiatric disorders, we investigated the expression of dysbindin in the retina. Our results show that differentially regulated dysbindin isoforms are expressed in rat retina during postnatal maturation. Interestingly, we found that dysbindin is mainly localized in Müller cells. The identification of dysbindin in glial cells may open new perspectives for a better understanding of the functional involvement of this protein in visual alterations associated to neuropsychiatric disorders.


Asunto(s)
Proteínas Portadoras/genética , Células Ependimogliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/genética , ARN/genética , Retina/crecimiento & desarrollo , Animales , Western Blotting , Proteínas Portadoras/biosíntesis , Disbindina , Proteínas Asociadas a la Distrofina , Electroforesis , Células Ependimogliales/citología , Masculino , Proteínas del Tejido Nervioso/biosíntesis , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Retina/citología , Retina/metabolismo
9.
Biochim Biophys Acta ; 1833(1): 110-21, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23103755

RESUMEN

Phosphorylation and nitration of protein tyrosine residues are thought to play a role in signaling pathways at the nerve terminal and to affect functional properties of proteins involved in the synaptic vesicle (SV) exo-endocytotic cycle. We previously demonstrated that the tyrosine residues in the C-terminal domain of the SV protein Synaptophysin (SYP) are targets of peroxynitrite (PN). Here, we have characterized the association between SYP and c-src tyrosine kinase demonstrating that phosphorylation of Tyr(273) in the C-terminal domain of SYP is crucial in mediating SYP binding to and activation of c-src. SYP forms a complex with Dynamin I (DynI), a GTPase required for SV endocytosis, which may be regulated by tyrosine phosphorylation of SYP. We here report that, in rat brain synaptosomes treated with PN, the formation of SYP/DynI complex was impaired. Noteworthy, we found that DynI was also modified by PN. DynI tyrosine phosphorylation was down-regulated in a dose-dependent manner, while DynI tyrosine nitration increased. Using mass spectrometry analysis, we identified Tyr(354) as one nitration site in DynI. In addition, we tested DynI self-assembly and GTPase activity, which are enhanced by c-src-dependent tyrosine phosphorylation of DynI, and found that both were inhibited by PN. Our results suggest that the site-specific tyrosine residue modifications may modulate the association properties of SV proteins and serve as a regulator of DynI function via control of self-assembly, thus influencing the physiology of the exo-endocytotic cycle.


Asunto(s)
Dinamina I/metabolismo , Dinamina I/fisiología , Vesículas Sinápticas/metabolismo , Sinaptofisina/metabolismo , Sinaptofisina/fisiología , Secuencia de Aminoácidos , Animales , Dinamina I/química , Dinamina I/genética , Endocitosis/genética , Endocitosis/fisiología , Exocitosis/genética , Exocitosis/fisiología , Técnicas In Vitro , Datos de Secuencia Molecular , Nitratos/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Ratas , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Vesículas Sinápticas/fisiología , Sinaptofisina/química , Sinaptofisina/genética , Tirosina/metabolismo , Tirosina/fisiología
10.
FEBS J ; 279(22): 4131-44, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22978324

RESUMEN

Dystrobrevin family members (α and ß) are cytoplasmic components of the dystrophin-associated glycoprotein complex, a multimeric protein complex first isolated from skeletal muscle, which links the extracellular matrix to the actin cytoskeleton. Dystrobrevin shares high homology with the cysteine-rich and C-terminal domains of dystrophin and a common domain organization. The ß-dystrobrevin isoform is restricted to nonmuscle tissues, serves as a scaffold for signaling complexes, and may participate in intracellular transport through its interaction with kinesin heavy chain. We have previously characterized the molecular determinants affecting the ß-dystrobrevin-kinesin heavy chain interaction, among which is cAMP-dependent protein kinase [protein kinase A (PKA)] phosphorylation of ß-dystrobrevin. In this study, we have identified ß-dystrobrevin residues phosphorylated in vitro by PKA with pull-down assays, surface plasmon resonance measurements, and MS analysis. Among the identified phosphorylated residues, we demonstrated, by site-directed mutagenesis, that Thr11 is the regulatory site for the ß-dystrobrevin-kinesin interaction. As dystrobrevin may function as a signaling scaffold for kinases/phosphatases, we also investigated whether ß-dystrobrevin is phosphorylated in vitro by kinases other than PKA. Thr11 was phosphorylated by protein kinase C, suggesting that this represents a key residue modified by the activation of different signaling pathways.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Asociadas a la Distrofina/metabolismo , Cinesinas/metabolismo , Neuropéptidos/metabolismo , Proteína Quinasa C/metabolismo , Treonina/metabolismo , Secuencia de Aminoácidos , Western Blotting , Proteínas Asociadas a la Distrofina/genética , Humanos , Inmunoprecipitación , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , Neuropéptidos/genética , Fosforilación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Resonancia por Plasmón de Superficie , Treonina/genética
11.
J Immunol ; 189(6): 2833-42, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22904309

RESUMEN

Exosomes are nanovesicles released by normal and tumor cells, which are detectable in cell culture supernatant and human biological fluids, such as plasma. Functions of exosomes released by "normal" cells are not well understood. In fact, several studies have been carried out on exosomes derived from hematopoietic cells, but very little is known about NK cell exosomes, despite the importance of these cells in innate and adaptive immunity. In this paper, we report that resting and activated NK cells, freshly isolated from blood of healthy donors, release exosomes expressing typical protein markers of NK cells and containing killer proteins (i.e., Fas ligand and perforin molecules). These nanovesicles display cytotoxic activity against several tumor cell lines and activated, but not resting, immune cells. We also show that NK-derived exosomes undergo uptake by tumor target cells but not by resting PBMC. Exosomes purified from plasma of healthy donors express NK cell markers, including CD56+ and perforin, and exert cytotoxic activity against different human tumor target cells and activated immune cells as well. The results of this study propose an important role of NK cell-derived exosomes in immune surveillance and homeostasis. Moreover, this study supports the use of exosomes as an almost perfect example of biomimetic nanovesicles possibly useful in future therapeutic approaches against various diseases, including tumors.


Asunto(s)
Exosomas/inmunología , Exosomas/metabolismo , Células Asesinas Activadas por Linfocinas/inmunología , Células Asesinas Activadas por Linfocinas/metabolismo , Monitorización Inmunológica , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Subgrupos de Linfocitos B/patología , Linfoma de Burkitt/inmunología , Linfoma de Burkitt/patología , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/patología , Técnicas de Cocultivo , Exosomas/ultraestructura , Proteína Ligando Fas/biosíntesis , Humanos , Inmunofenotipificación , Células Jurkat , Células K562 , Células Asesinas Activadas por Linfocinas/ultraestructura , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Monitorización Inmunológica/métodos , Perforina/biosíntesis
12.
Hum Mol Genet ; 21(10): 2166-80, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22328087

RESUMEN

Megalencephalic leukoencephalopathy with subcortical cysts (MLC), a rare leukodystrophy characterized by macrocephaly, subcortical fluid cysts and myelin vacuolation, has been linked to mutations in the MLC1 gene. This gene encodes a membrane protein that is highly expressed in astrocytes. Based on MLC pathological features, it was proposed that astrocyte-mediated defects in ion and fluid homeostasis could account for the alterations observed in MLC-affected brains. However, the role of MLC1 and the effects of pathological mutations on astrocyte osmoregulatory functions have still to be demonstrated. Using human astrocytoma cells stably overexpressing wild-type MLC1 or three known MLC-associated pathological mutations, we investigated MLC1 involvement in astrocyte reaction to osmotic changes using biochemical, dynamic video imaging and immunofluorescence techniques. We have found that MLC1 overexpressed in astrocytoma cells is mainly localized in the plasma membrane, is part of the Na,K-ATPase-associated molecular complex that includes the potassium channel Kir4.1, syntrophin and aquaporin-4 and functionally interacts with the calcium permeable channel TRPV4 (transient receptor potential vanilloid-4 cation channel) which mediates swelling-induced cytosolic calcium increase and volume recovery in response to hyposmosis. Pathological MLC mutations cause changes in MLC1 expression and intracellular localization as well as in the astrocyte response to osmotic changes by altering MLC1 molecular interactions with the Na,K-ATPase molecular complex and abolishing the increase in calcium influx induced by hyposmosis and treatment with the TRPV4 agonist 4αPDD. These data demonstrate, for the first time, that MLC1 plays a role in astrocyte osmo-homeostasis and that defects in intracellular calcium dynamics may contribute to MLC pathogenesis.


Asunto(s)
Astrocitos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Canales Catiónicos TRPV/metabolismo , Calcio/metabolismo , Cationes Bivalentes , Quistes/genética , Quistes/metabolismo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Humanos , Mutación , Ósmosis , Transfección
13.
Hum Mol Genet ; 20(1): 90-103, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20926452

RESUMEN

Megalencephalic leucoencephalopathy with subcortical cysts (MLC) is a rare congenital leucodystrophy caused by mutations in MLC1, a membrane protein of unknown function. MLC1 expression in astrocyte end-feet contacting blood vessels and meninges, along with brain swelling, fluid cysts and myelin vacuolation observed in MLC patients, suggests a possible role for MLC1 in the regulation of fluid and ion homeostasis and cellular volume changes. To identify MLC1 direct interactors and dissect the molecular pathways in which MLC1 is involved, we used NH2-MLC1 domain as a bait to screen a human brain library in a yeast two-hybrid assay. We identified the ß1 subunit of the Na,K-ATPase pump as one of the interacting clones and confirmed it by pull-downs, co-fractionation assays and immunofluorescence stainings in human and rat astrocytes in vitro and in brain tissue. By performing ouabain-affinity chromatography on astrocyte and brain extracts, we isolated MLC1 and the whole Na,K-ATPase enzyme in a multiprotein complex that included Kir4.1, syntrophin and dystrobrevin. Because Na,K-ATPase is involved in intracellular osmotic control and volume regulation, we investigated the effect of hypo-osmotic stress on MLC1/Na,K-ATPase relationship in astrocytes. We found that hypo-osmotic conditions increased MLC1 membrane expression and favoured MLC1/Na,K-ATPase-ß1 association. Moreover, hypo-osmosis induced astrocyte swelling and the reversible formation of endosome-derived vacuoles, where the two proteins co-localized. These data suggest that through its interaction with Na,K-ATPase, MLC1 is involved in the control of intracellular osmotic conditions and volume regulation in astrocytes, opening new perspectives for understanding the pathological mechanisms of MLC disease.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Proteínas de la Membrana/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Células Cultivadas , Quistes/genética , Quistes/metabolismo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Humanos , Proteínas de la Membrana/genética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Complejos Multiproteicos/metabolismo , Ratas , Ratas Wistar , ATPasa Intercambiadora de Sodio-Potasio/genética
14.
J Biol Chem ; 285(32): 24740-50, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20530487

RESUMEN

alpha and beta dystrobrevins are cytoplasmic components of the dystrophin-associated protein complex that are thought to play a role as scaffold proteins in signal transduction and intracellular transport. In the search of new insights into the functions of beta-dystrobrevin, the isoform restricted to non-muscle tissues, we performed a two-hybrid screen of a mouse cDNA library to look for interacting proteins. Among the positive clones, one encodes iBRAF/HMG20a, a high mobility group (HMG)-domain protein that activates REST (RE-1 silencing transcription factor)-responsive genes, playing a key role in the initiation of neuronal differentiation. We characterized the beta-dystrobrevin-iBRAF interaction by in vitro and in vivo association assays, localized the binding region of one protein to the other, and assessed the kinetics of the interaction as one of high affinity. We also found that beta-dystrobrevin directly binds to BRAF35/HMG20b, a close homologue of iBRAF and a member of a co-repressor complex required for the repression of neural specific genes in neuronal progenitors. In vitro assays indicated that beta-dystrobrevin binds to RE-1 and represses the promoter activity of synapsin I, a REST-responsive gene that is a marker for neuronal differentiation. Altogether, our data demonstrate a direct interaction of beta-dystrobrevin with the HMG20 proteins iBRAF and BRAF35 and suggest that beta-dystrobrevin may be involved in regulating chromatin dynamics, possibly playing a role in neuronal differentiation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Asociadas a la Distrofina/fisiología , Proteínas del Grupo de Alta Movilidad/metabolismo , Neuronas/citología , Animales , Células COS , Proteínas de Ciclo Celular , Diferenciación Celular , Línea Celular Tumoral , Chlorocebus aethiops , Cromatina/química , Humanos , Cinética , Ratones , Distrofias Musculares/metabolismo , Ratas , Resonancia por Plasmón de Superficie
15.
Neurobiol Dis ; 37(3): 581-95, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19931615

RESUMEN

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare congenital leukodystrophy caused by mutations in the MLC1 gene that encodes a membrane protein of unknown function. In the brain MLC1 protein is mainly expressed in astrocyte end-feet, localizes in lipid rafts and associates with the dystrophin glycoprotein complex (DGC). Using pull-down and co-fractionation assays in cultured human and rat astrocytes, we show here that MLC1 intracellular domains pull-down the DGC proteins syntrophin, dystrobrevin, Kir4.1 and caveolin-1, the structural protein of caveolae, thereby supporting a role for DGC and caveolar structures in MLC1 function. By immunostaining and subcellular fractionation of cultured rat or human astrocytes treated with agents modulating caveolin-mediated trafficking, we demonstrate that MLC1 is also expressed in intracellular vesicles and endoplasmic reticulum and undergoes caveolae/raft-mediated endocytosis. Inhibition of endocytosis, cholesterol lowering and protein kinases A- and C-mediated MLC1 phosphorylation favour the expression of membrane-associated MLC1. Because pathological mutations prevent MLC1 membrane expression, the identification of substances regulating MLC1 intracellular trafficking is potentially relevant for the therapy of MLC.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Caveolas/metabolismo , Caveolina 1/metabolismo , Leucoencefalopatías/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/patología , Encéfalo/fisiopatología , Caveolas/ultraestructura , Línea Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Células Cultivadas , Colesterol/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Vesículas Citoplasmáticas/metabolismo , Vesículas Citoplasmáticas/ultraestructura , Complejo de Proteínas Asociado a la Distrofina/metabolismo , Endocitosis/fisiología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Humanos , Leucoencefalopatías/genética , Leucoencefalopatías/fisiopatología , Microdominios de Membrana/metabolismo , Microdominios de Membrana/ultraestructura , Fosforilación , Proteína Quinasa C/metabolismo , Transporte de Proteínas/fisiología , Ratas
16.
J Neurochem ; 111(3): 859-69, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19737347

RESUMEN

Peroxynitrite is a potent oxidant that contributes to tissue damage in neurodegenerative disorders. We have previously reported that treatment of rat brain synaptosomes with peroxynitrite induced post-translational modifications in pre- and post-synaptic proteins and stimulated soluble N-ethylmaleimide sensitive fusion proteins attachment receptor complex formation and endogenous glutamate release. In this study we show that, following peroxynitrite treatment, the synaptic vesicle protein synaptophysin (SYP) can be both phosphorylated and nitrated in a dose-dependent manner. We found that tyrosine-phosphorylated, but not tyrosine-nitrated, SYP bound to the src tyrosine kinase and enhanced its catalytic activity. These effects were mediated by direct and specific binding of the SYP cytoplasmic C-terminal tail with the src homology 2 domain. Using mass spectrometry analysis, we mapped the SYP C-terminal tail tyrosine residues modified by peroxynitrite and found one nitration site at Tyr250 and two phosphorylation sites at Tyr263 and Tyr273. We suggest that peroxynitrite-mediated modifications of SYP may be relevant in modulating src signalling of synaptic terminal in pathophysiological conditions.


Asunto(s)
Ácido Peroxinitroso/farmacología , Sinaptofisina/química , Sinaptofisina/metabolismo , Sinaptosomas/efectos de los fármacos , Tirosina/metabolismo , Dominios Homologos src/fisiología , Familia-src Quinasas/metabolismo , Animales , Encéfalo/ultraestructura , Masculino , Espectrometría de Masas/métodos , Unión Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Dominios Homologos src/genética
17.
Matrix Biol ; 27(4): 360-70, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18249103

RESUMEN

Keratoepithelin (KE) is an extracellular matrix protein that binds collagens, fibronectin, decorin, biglycan and integrins, interconnecting extracellular matrix components with resident cells in several tissues. KE has a molecular mass of 68 kDa and harbours four FAS1 domains named after those identified in the insect cell adhesion molecule fasciclin I. In humans, KE is preferentially expressed by the corneal epithelial layer and liberated towards the corneal stroma but it was also detected in the lung and in the bladder smooth muscle. No detailed information is available on the distribution of this protein in other human tissues. In this work, we have raised a polyclonal antibody against the recombinantly expressed human fourth FAS1 domain which is able to specifically detect KE in human skeletal muscle tissue extracts. Immunofluorescence experiments indicate that KE is localized around the perimysium and endomysium of each skeletal muscle fiber. The same kind of analysis shows that in muscle sections from patients affected by different forms of muscular dystrophy KE is upregulated and widely distributed in fibrotic tissues. The muscle specific expression of KE was also demonstrated by RT-PCR. In human skeletal muscle, KE may help to build up a bridge between collagen VI and yet unidentified muscle receptor(s), adding to the complexity of the adhesive molecular network established between muscle fibers and the surrounding basement membrane.


Asunto(s)
Proteínas de la Matriz Extracelular/inmunología , Proteínas de la Matriz Extracelular/metabolismo , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Secuencia de Aminoácidos , Animales , Córnea/metabolismo , Reacciones Cruzadas , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Peso Molecular , ARN Mensajero/genética , Ratas , Alineación de Secuencia , Factor de Crecimiento Transformador beta/química , Factor de Crecimiento Transformador beta/genética
18.
J Mol Biol ; 371(5): 1174-87, 2007 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-17610895

RESUMEN

The dystrophin-related and -associated protein dystrobrevin is a component of the dystrophin-associated protein complex, which directly links the cytoskeleton to the extracellular matrix. It is now thought that this complex also serves as a dynamic scaffold for signaling proteins, and dystrobrevin may play a role in this context. Since dystrobrevin involvement in signaling pathways seems to be dependent on its interaction with other proteins, we sought new insights and performed a two-hybrid screen of a mouse brain cDNA library using beta-dystrobrevin, the isoform expressed in non-muscle tissues, as bait. Among the positive clones characterized after the screen, one encodes the regulatory subunit RIalpha of the cAMP-dependent protein kinase A (PKA). We confirmed the interaction by in vitro and in vivo association assays, and mapped the binding site of beta-dystrobrevin on RIalpha to the amino-terminal region encompassing the dimerization/docking domain of PKA regulatory subunit. We also found that the domain of interaction for RIalpha is contained in the amino-terminal region of beta-dystrobrevin. We obtained evidence that beta-dystrobrevin also interacts directly with RIIbeta, and that not only beta-dystrobrevin but also alpha-dystrobrevin interacts with PKA regulatory subunits. We show that both alpha and beta-dystrobrevin are specific phosphorylation substrates for PKA and that protein phosphatase 2A (PP2A) is associated with dystrobrevins. Our results suggest a new role for dystrobrevin as a scaffold protein that may play a role in different cellular processes involving PKA signaling.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Asociadas a la Distrofina/química , Proteínas Asociadas a la Distrofina/fisiología , Animales , Sitios de Unión , Encéfalo/metabolismo , Células COS , Chlorocebus aethiops , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Matriz Extracelular , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Transducción de Señal , Técnicas del Sistema de Dos Híbridos
19.
J Neurosci Res ; 85(12): 2631-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17265465

RESUMEN

The dystrobrevins (alpha and beta) are components of the dystrophin-associated protein complex (DPC), which links the cytoskeleton to the extracellular matrix and serves as a scaffold for signaling proteins. The precise functions of the beta-dystrobrevin isoform, which is expressed in nonmuscle tissues, have not yet been determined. To gain further insights into the role of beta-dystrobrevin in brain, we performed a yeast two-hybrid screen and identified pancortin-2 as a novel beta-dystrobrevin-binding partner. Pancortins-1-4 are neuron-specific olfactomedin-related glycoproteins, highly expressed during brain development and widely distributed in the mature cerebral cortex of the mouse. Pancortins are important constituents of the extracellular matrix and are thought to play an essential role in neuronal differentiation. We characterized the interaction between pancortin-2 and beta-dystrobrevin by in vitro and in vivo association assays and mapped the binding site of pancortin-2 on beta-dystrobrevin to amino acids 202-236 of the beta-dystrobrevin molecule. We also found that the domain of interaction for beta-dystrobrevin is contained in the B part of pancortin-2, a central region that is common to all four pancortins. Our results indicate that beta-dystrobrevin could interact with all members of the pancortin family, implying that beta-dystrobrevin may be involved in brain development. We suggest that dystrobrevin, a motor protein receptor that binds kinesin heavy chain, might play a role in intracellular transport of pancortin to specific sites in the cell.


Asunto(s)
Proteínas Asociadas a la Distrofina/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Células COS , Chlorocebus aethiops , Clonación Molecular/métodos , Proteínas de la Matriz Extracelular/farmacocinética , Mutación , Unión Proteica , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Ratas , Isótopos de Azufre/farmacocinética , Transfección , Técnicas del Sistema de Dos Híbridos
20.
J Mol Biol ; 354(4): 872-82, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16288919

RESUMEN

Dystrobrevins are a family of widely expressed dystrophin-associated proteins that comprises alpha and beta isoforms and displays significant sequence homology with several protein-binding domains of the dystrophin C-terminal region. The complex distribution of the multiple dystrobrevin isoforms suggests that the variability of their composition may be important in mediating their function. We have recently identified kinesin as a novel dystrobrevin-interacting protein and localized the dystrobrevin-binding site on the cargo-binding domain of neuronal kinesin heavy chain (Kif5A). In the present study, we assessed the kinetics of the dystrobrevin-Kif5A interaction by quantitative pull-down assay and surface plasmon resonance (SPR) analysis and found that beta-dystrobrevin binds to kinesin with high affinity (K(D) approximately 40 nM). Comparison of the sensorgrams obtained with alpha and beta-dystrobrevin at the same concentration of analyte showed a lower affinity of alpha compared to that of beta-dystrobrevin, despite their functional domain homology and about 70% sequence identity. Analysis of the contribution of single dystrobrevin domains to the interaction revealed that the deletion of either the ZZ domain or the coiled-coil region decreased the kinetics of the interaction, suggesting that the tertiary structure of dystrobrevin may play a role in regulating the interaction of dystrobrevin with kinesin. In order to understand if structural changes induced by post-translational modifications could affect dystrobrevin affinity for kinesin, we phosphorylated beta-dystrobrevin in vitro and found that it showed reduced binding capacity towards kinesin. The interaction between the adaptor/scaffolding protein dystrobrevin and the motor protein kinesin may play a role in the transport and targeting of components of the dystrophin-associated protein complex to specific sites in the cell, with the differences in the binding properties of dystrobrevin isoforms reflecting their functional diversity within the same cell type. Phosphorylation events could have a regulatory role in this context.


Asunto(s)
Proteínas Asociadas a la Distrofina/química , Cinesinas/química , Animales , Proteínas Asociadas a la Distrofina/metabolismo , Cinesinas/metabolismo , Cinética , Ratones , Fosforilación , Unión Proteica , Conformación Proteica , Isoformas de Proteínas , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...