Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Soc Psychiatry Psychiatr Epidemiol ; 57(6): 1283-1289, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35279745

RESUMEN

PURPOSE: The aim of our study is to evaluate the number and the features of admissions to the emergency room (ER) requiring psychiatric consultation, in the period between May 4th and August 31st 2020. METHODS: We carried out a retrospective longitudinal observational study examining the 4 months following the initial lockdown imposed during the COVID-19 outbreak (May 4th and August 31st 2020). More specifically, the ER admissions leading to psychiatric referral were reviewed at all seven public hospitals of AUSL Romagna (Emilia Romagna region, Italy). Socio-demographic variables, history of medical comorbidities or psychiatric disorders, reason for ER admission, psychiatric diagnosis at discharge, and actions taken by the psychiatrist were collected. RESULTS: An 11.3% (p = 0.007) increase in psychiatric assessments was observed when compared with the same period of the previous year (2019). A positive personal history of psychiatric disorders (OR:0.68, CI: 0.53-0.87) and assessments leading to no indication for follow-up (OR: 0.22, CI: 0.13-0.39) were significantly less frequent, while there was a significant increase of cases featuring organic comorbidities (OR: 1.24, CI: 1.00-1.52) and suicidal ideation/self-harm/suicide attempt (OR: 1,71, CI: 1.19-2.45) or psychomotor agitation (OR: 1.46, CI: 1.02-2.07) as reason for admission. CONCLUSIONS: Our results showed an increase in ER psychiatric consultations compared to the previous year, underlying the increased psychological distress caused by the lockdown.


Asunto(s)
COVID-19 , COVID-19/epidemiología , Control de Enfermedades Transmisibles , Servicio de Urgencia en Hospital , Humanos , Italia/epidemiología , Pandemias , Estudios Retrospectivos
2.
Front Psychol ; 11: 1085, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32587546

RESUMEN

The heterogeneity of cognitive profiles among psychiatric patients has been reported to carry significant clinical information. However, how to best characterize such cognitive heterogeneity is still a matter of debate. Despite being well suited for clinical data, cluster analysis techniques, like the Two-Step and the Latent Class, received little to no attention in the literature. The present study aimed to test the validity of the cluster solutions obtained with Two-Step and Latent Class cluster analysis on the cognitive profile of a cross-diagnostic sample of 387 psychiatric inpatients. Two-Step and Latent Class cluster analysis produced similar and reliable solutions. The overall results reported that it is possible to group all psychiatric inpatients into Low and High Cognitive Profiles, with a higher degree of cognitive heterogeneity in schizophrenia and bipolar disorder patients than in depressive disorders and personality disorder patients.

3.
Front Immunol ; 9: 1186, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29896202

RESUMEN

Tumor microenvironment is fundamental for cancer progression and chemoresistance. Among stromal cells tumor-associated macrophages (TAMs) represent the largest population of infiltrating inflammatory cells in malignant tumors, promoting their growth, invasion, and immune evasion. M2-polarized TAMs are endowed with the nitric oxide (NO)-generating enzyme inducible nitric oxide synthase (iNOS). NO has divergent effects on tumors, since it can either stimulate tumor cells growth or promote their death depending on the source of it; likewise the role of iNOS in cancer differs depending on the cell type. The role of NO generated by TAMs has not been investigated. Using different tumor models in vitro and in vivo we found that NO generated by iNOS of M2-polarized TAMs is able to protect tumor cells from apoptosis induced by the chemotherapeutic agent cisplatin (CDDP). Here, we demonstrate that the protective effect of NO depends on the inhibition of acid sphingomyelinase (A-SMase), which is activated by CDDP in a pathway involving the death receptor CD95. Mechanistic insights indicate that NO actions occur via generation of cyclic GMP and activation of protein kinase G (PKG), inducing phosphorylation of syntaxin 4 (synt4), a SNARE protein responsible for A-SMase trafficking and activation. Noteworthy, phosphorylation of synt4 at serine 78 by PKG is responsible for the proteasome-dependent degradation of synt4, which limits the CDDP-induced exposure of A-SMase to the plasma membrane of tumor cells. This inhibits the cytotoxic mechanism of CDDP reducing A-SMase-triggered apoptosis. This is the first demonstration that endogenous NO system is a key mechanism through which TAMs protect tumor cells from chemotherapeutic drug-induced apoptosis. The identification of the pathway responsible for A-SMase activity downregulation in tumors leading to chemoresistance warrants further investigations as a means to identify new anti-cancer molecules capable of specifically inhibiting synt4 degradation.


Asunto(s)
Cisplatino/farmacología , Resistencia a Antineoplásicos/inmunología , Glioma/inmunología , Macrófagos/inmunología , Proteínas de Neoplasias/inmunología , Óxido Nítrico/inmunología , Proteínas Qa-SNARE/inmunología , Esfingomielina Fosfodiesterasa/inmunología , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Glioma/tratamiento farmacológico , Glioma/genética , Glioma/patología , Humanos , Macrófagos/patología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Óxido Nítrico/genética , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Proteínas Qa-SNARE/genética , Esfingomielina Fosfodiesterasa/genética
4.
Glia ; 64(8): 1437-60, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27270750

RESUMEN

The G protein-coupled receptor 17 (GPR17) plays crucial roles in myelination. It is highly expressed during transition of oligodendrocyte progenitor cells to immature oligodendrocytes, but, after this stage, it must be down-regulated to allow generation of mature myelinating cells. After endocytosis, GPR17 is sorted into lysosomes for degradation or recycled to the plasma membrane. Balance between degradation and recycling is important for modulation of receptor levels at the cell surface and thus for the silencing/activation of GPR17-signaling pathways that, in turn, affect oligodendrocyte differentiation. The molecular mechanisms at the basis of these processes are still partially unknown and their characterization will allow a better understanding of myelination and provide cues to interpret the consequences of GPR17 dysfunction in diseases. Here, we demonstrate that the endocytic trafficking of GPR17 is mediated by the interaction of a type I PDZ-binding motif located at the C-terminus of the receptor and SNX27, a recently identified protein of the endosome-associated retromer complex and whose functions in oligodendrocytes have never been studied. SNX27 knock-down significantly reduces GPR17 plasma membrane recycling in differentiating oligodendrocytes while accelerating cells' terminal maturation. Interestingly, trisomy-linked down-regulation of SNX27 expression in the brain of Ts65Dn mice, a model of Down syndrome, correlates with a decrease in GPR17(+) cells and an increase in mature oligodendrocytes, which, however, fail in reaching full maturation, eventually leading to hypomyelination. Our data demonstrate that SNX27 modulates GPR17 plasma membrane recycling and stability, and that disruption of the SNX27/GPR17 interaction might contribute to pathological oligodendrocyte differentiation defects. GLIA 2016. GLIA 2016;64:1437-1460.


Asunto(s)
Transporte Biológico/fisiología , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Nexinas de Clasificación/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Diferenciación Celular/fisiología , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/patología , Modelos Animales de Enfermedad , Síndrome de Down/metabolismo , Síndrome de Down/patología , Endocitosis/fisiología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligodendroglía/patología , Nexinas de Clasificación/deficiencia , Nexinas de Clasificación/genética
5.
Glia ; 63(2): 271-86, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25213035

RESUMEN

Oligodendrocyte progenitor cells (OPCs) persist in the adult central nervous system and guarantee oligodendrocyte turnover throughout life. It remains obscure how OPCs avoid exhaustion during adulthood. Similar to stem cells, OPCs could self-maintain by undergoing asymmetric divisions generating a mixed progeny either keeping a progenitor phenotype or proceeding to differentiation. To address this issue, we examined the distribution of stage-specific markers in sister OPCs during mitosis and later after cell birth, and assessed its correlation with distinct short-term fates. In both the adult and juvenile cerebral cortex a fraction of dividing OPCs gives rise to sister cells with diverse immunophenotypic profiles and short-term behaviors. Such heterogeneity appears as cells exit cytokinesis, but does not derive from the asymmetric segregation of molecules such as NG2 or PDGFRa expressed in the mother cell. Rather, rapid downregulation of OPC markers and upregulation of molecules associated with lineage progression contributes to generate early sister OPC asymmetry. Analyses during aging and upon exposure to physiological (i.e., increased motor activity) and pathological (i.e., trauma or demyelination) stimuli showed that both intrinsic and environmental factors contribute to determine the fraction of symmetric and asymmetric OPC pairs and the phenotype of the OPC progeny as soon as cells exit mitosis.


Asunto(s)
Envejecimiento , Mitosis/fisiología , Oligodendroglía/fisiología , Células Madre/fisiología , Análisis de Varianza , Animales , Antígenos/genética , Antígenos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Bromodesoxiuridina , Ciclo Celular/fisiología , Diferenciación Celular , Células Cultivadas , Sistema Nervioso Central/citología , Regulación de la Expresión Génica/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteoglicanos/genética , Proteoglicanos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
6.
J Cell Mol Med ; 18(9): 1785-96, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24909956

RESUMEN

GPR17 is a G(i) -coupled dual receptor activated by uracil-nucleotides and cysteinyl-leukotrienes. These mediators are massively released into hypoxic tissues. In the normal heart, GPR17 expression has been reported. By contrast, its role in myocardial ischaemia has not yet been assessed. In the present report, the expression of GPR17 was investigated in mice before and at early stages after myocardial infarction by using immunofluorescence, flow cytometry and RT-PCR. Before induction of ischaemia, results indicated the presence of the receptor in a population of stromal cells expressing the stem-cell antigen-1 (Sca-1). At early stages after ligation of the coronary artery, the receptor was expressed in Sca-1(+) cells, and cells stained with Isolectin-B4 and anti-CD45 antibody. GPR17(+) cells also expressed mesenchymal marker CD44. GPR17 function was investigated in vitro in a Sca-1(+)/CD31(-) cell line derived from normal hearts. These experiments showed a migratory function of the receptor by treatment with UDP-glucose and leukotriene LTD4, two GPR17 pharmacological agonists. The GPR17 function was finally assessed in vivo by treating infarcted mice with Cangrelor, a pharmacological receptor antagonist, which, at least in part, inhibited early recruitment of GPR17(+) and CD45(+) cells. These findings suggest a regulation of heart-resident mesenchymal cells and blood-borne cellular species recruitment following myocardial infarction, orchestrated by GPR17.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Infarto del Miocardio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Animales , Antígenos Ly/metabolismo , Movimiento Celular , Receptores de Hialuranos , Antígenos Comunes de Leucocito/metabolismo , Leucotrieno D4/farmacología , Leucotrieno D4/fisiología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Proteínas del Tejido Nervioso/agonistas , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Acoplados a Proteínas G/agonistas , Uridina Difosfato Glucosa/farmacología , Uridina Difosfato Glucosa/fisiología
7.
J Biol Chem ; 288(47): 33873-33883, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24108129

RESUMEN

Mutations in the CACNA1A gene, which encodes the pore-forming α1A subunit of the CaV2.1 voltage-gated calcium channel, cause a number of human neurologic diseases including familial hemiplegic migraine. We have analyzed the functional impact of the E1015K amino acid substitution located in the "synprint" domain of the α1A subunit. This variant was identified in two families with hemiplegic migraine and in one patient with migraine with aura. The wild type (WT) and the E1015K forms of the GFP-tagged α1A subunit were expressed in cultured hippocampal neurons and HEK cells to understand the role of the variant in the transport activity and physiology of CaV2.1. The E1015K variant does not alter CaV2.1 protein expression, and its transport to the cell surface and synaptic terminals is similar to that observed for WT channels. Electrophysiological data demonstrated that E1015K channels have increased current density and significantly altered inactivation properties compared with WT. Furthermore, the SNARE proteins syntaxin 1A and SNAP-25 were unable to modulate voltage-dependent inactivation of E1015K channels. Overall, our findings describe a genetic variant in the synprint site of the CaV2.1 channel which is characterized by a gain-of-function and associated with both hemiplegic migraine and migraine with aura in patients.


Asunto(s)
Canales de Calcio Tipo N , Hipocampo , Migraña con Aura , Mutación Missense , Proteínas del Tejido Nervioso , Terminales Presinápticos , Adolescente , Adulto , Sustitución de Aminoácidos , Animales , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Niño , Femenino , Células HEK293 , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Transporte Iónico/genética , Masculino , Persona de Mediana Edad , Migraña con Aura/genética , Migraña con Aura/metabolismo , Migraña con Aura/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Terminales Presinápticos/metabolismo , Terminales Presinápticos/patología , Conejos , Ratas , Proteína 25 Asociada a Sinaptosomas/genética , Proteína 25 Asociada a Sinaptosomas/metabolismo , Sintaxina 1/genética , Sintaxina 1/metabolismo
8.
Purinergic Signal ; 9(3): 451-62, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23801362

RESUMEN

Unveiling the mechanisms participating in the damage and repair of traumatic brain injury (TBI) is fundamental to develop new therapies. The P2Y-like GPR17 receptor has recently emerged as a sensor of damage and a key actor in lesion remodeling/repair in the rodent brain, but its role in humans is totally unknown. Here, we characterized GPR17 expression in brain specimens from seven intensive care unit TBI patients undergoing neurosurgery for contusion removal and from 28 autoptic TBI cases (and 10 control subjects of matched age and gender) of two university hospitals. In both neurosurgery and autoptic samples, GPR17 expression was strong inside the contused core and progressively declined distally according to a spatio-temporal gradient. Inside and around the core, GPR17 labeled dying neurons, reactive astrocytes, and activated microglia/macrophages. In peri-contused parenchyma, GPR17 decorated oligodendrocyte precursor cells (OPCs) some of which had proliferated, indicating re-myelination attempts. In autoptic cases, GPR17 expression positively correlated with death for intracranial complications and negatively correlated with patients' post-traumatic survival. Data indicate lesion-specific sequential involvement of GPR17 in the (a) death of irreversibly damaged neurons, (b) activation of microglia/macrophages remodeling the lesion, and (c) activation/proliferation of multipotent parenchymal progenitors (both reactive astrocytes and OPCs) starting repair processes. Data validate GPR17 as a target for neurorepair and are particularly relevant to setting up new therapies for TBI patients.


Asunto(s)
Lesiones Encefálicas/metabolismo , Regeneración Nerviosa/fisiología , Neuroglía/metabolismo , Neuronas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adulto , Anciano , Lesiones Encefálicas/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Microscopía Confocal , Persona de Mediana Edad , Neuroglía/patología , Neuronas/patología , Adulto Joven
9.
Purinergic Signal ; 9(3): 367-81, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23413038

RESUMEN

The rat adenine receptor (rAdeR) was the first member of a family of G protein-coupled receptors (GPCRs) activated by adenine and designated as P0-purine receptors. The present study aimed at gaining insights into structural aspects of ligand binding and function of the rAdeR. We exchanged amino acid residues predicted to be involved in ligand binding (Phe110(3.24), Asn115(3.29), Asn173(4.60), Phe179(45.39), Asn194(5.40), Phe195(5.41), Leu201(5.47), His252(6.54), and Tyr268(7.32)) for alanine and expressed them in Spodoptera frugiperda (Sf9) insect cells. Membrane preparations subjected to [(3)H]adenine binding studies revealed only minor effects indicating that none of the exchanged amino acids is part of the ligand binding pocket, at least in the inactive state of the receptor. Furthermore, we coexpressed the rAdeR and its mutants with mammalian Gi proteins in Sf9 insect cells to probe receptor activation. Two amino acid residues, Asn194(5.40) and Leu201(5.47), were found to be crucial for activation since their alanine mutants did not respond to adenine. Moreover we showed that-in contrast to most other rhodopsin-like GPCRs-the rAdeR does not contain essential disulfide bonds since preincubation with dithiothreitol neither altered adenine binding in Sf9 cell membranes, nor adenine-induced inhibition of adenylate cyclase in 1321N1 astrocytoma cells transfected with the rAdeR. To detect rAdeRs by Western blot analysis, we developed a specific antibody. Finally, we were able to show that the extended N-terminal sequence of the rAdeR constitutes a putative signal peptide of unknown function that is cleaved off in the mature receptor. Our results provide important insights into this new, poorly investigated family of purinergic receptors.


Asunto(s)
Receptores Purinérgicos/química , Receptores Purinérgicos/metabolismo , Animales , Sitios de Unión , Unión Competitiva , Western Blotting , Ligandos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Cuaternaria de Proteína , Ratas
10.
J Biol Chem ; 288(7): 5241-56, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23288840

RESUMEN

GPR17 is a G-protein-coupled receptor that is activated by two classes of molecules: uracil-nucleotides and cysteinyl-leukotrienes. GPR17 is required for initiating the differentiation of oligodendrocyte precursors but has to be down-regulated to allow cells to undergo terminal maturation. Although a great deal has been learned about GPR17 expression and signaling, no information is currently available about the trafficking of native receptors after the exposure of differentiating oligodendrocytes to endogenous agonists. Here, we demonstrate that neuron-conditioned medium induces the transcriptionally mediated, time-regulated expression of GPR17 in Oli-neu, an oligodendrocyte precursor cell line, making these cells suitable for studying the endocytic traffic of the native receptor. Agonist-induced internalization, intracellular trafficking, and membrane recycling of GPR17 were analyzed by biochemical and immunofluorescence assays using an ad hoc-developed antibody against the extracellular N-terminal of GPR17. Both UDP-glucose and LTD(4) increased GPR17 internalization, although with different efficiency. At early time points, internalized GPR17 co-localized with transferrin receptor, whereas at later times it partially co-localized with the lysosomal marker Lamp1, suggesting that a portion of GPR17 is targeted to lysosomes upon ligand binding. An analysis of receptor recycling and degradation demonstrated that a significant aliquot of GPR17 is recycled to the cell surface. Furthermore, internalized GPR17 displayed a co-localization with the marker of the "short loop" recycling endosomes, Rab4, while showing very minor co-localization with the "long loop" recycling marker, Rab11. Our results provide the first data on the agonist-induced trafficking of native GPR17 in oligodendroglial cells and may have implications for both physiological and pathological myelination.


Asunto(s)
Membrana Celular/metabolismo , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Clonación Molecular , Endocitosis , Humanos , Lisosomas/metabolismo , Ratones , Modelos Biológicos , Oligodendroglía/citología , Interferencia de ARN , Ratas
11.
Glia ; 59(12): 1958-73, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21956849

RESUMEN

NG2-expressing cells comprise a population of cycling precursors that can exit the cell cycle and differentiate into mature oligodendrocytes. As a whole, they display heterogeneous properties and behaviors that remain unresolved at the molecular level, although partly interpretable as distinct maturation stages. To address this issue, we analyzed the expression of the GPR17 receptor, recently shown to decorate NG2-expressing cells and to operate as an early sensor of brain damage, in immature and adult oligodendrocyte progenitors in the intact brain and after injury. In both the early postnatal and adult cerebral cortex, distinct GPR17 protein localizations and expression levels define different stages of oligodendroglial maturation, ranging from the precursor phase to the premyelinating phenotype. As soon as cells exit mitosis, a fraction of NG2-expressing cells displays accumulation of GPR17 protein in the Golgi apparatus. GPR17 expression is subsequently upregulated and distributed to processes of cells that stop dividing, progressively lose NG2 positivity and assume premyelinating features. Absence of colabeling with mature markers or myelin proteins indicates that GPR17 is downregulated when cells complete their final maturation. BrdU-based fate-mapping demonstrated that a significant fraction of newly generated oligodendrocyte progenitors transiently upregulates GPR17 during maturation. Importantly, we also found that GPR17 does not participate to the early reaction of NG2-expressing cells to damage, while it is induced at postacute stages after injury. These findings identify GPR17 as a marker for progenitor progression within the oligodendroglial lineage and highlight its participation to postacute reactivity of NG2 cells in different injury paradigms.


Asunto(s)
Antígenos/biosíntesis , Daño Encefálico Crónico/metabolismo , Lesiones Encefálicas/metabolismo , Diferenciación Celular/fisiología , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/metabolismo , Proteoglicanos/biosíntesis , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/metabolismo , Enfermedad Aguda , Animales , Antígenos/genética , Biomarcadores/metabolismo , Daño Encefálico Crónico/patología , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Aparato de Golgi/metabolismo , Aparato de Golgi/patología , Ratones , Ratones Endogámicos C57BL , Regeneración Nerviosa/fisiología , Oligodendroglía/patología , Cultivo Primario de Células , Proteoglicanos/genética , Células Madre/patología
12.
Glia ; 59(3): 363-78, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21264945

RESUMEN

The P2Y-like receptor GPR17 is expressed by adult neural progenitor cells, suggesting a role in lineage determination. Here, we characterized GPR17 expression and function in mouse cortical primary astrocytes/precursor cell cultures. GPR17 is expressed by a subpopulation of oligodendrocyte precursor cells (OPCs), but not by astrocytes. This expression pattern was also confirmed in vivo. In vitro, GPR17 expression was markedly influenced by culturing conditions. In the presence of growth factors (GFs), no significant GPR17 expression was found. When cultures were shifted to a differentiating medium, a dramatic, time-dependent increase in the number of highly branched GPR17-positive cells was observed. Under these conditions, GPR17 was induced in the totality of O4-positive immature oligodendrocytes. Instead, in cultures originally grown in the absence of GFs, GPR17 was already expressed in morphologically more mature OPCs. Shifting of these cultures to differentiating conditions induced GPR17 only in a subpopulation of O4-positive cells. Under both culture protocols, appearance of more mature CNPase- and MBP-positive cells was associated to a progressive loss of GPR17. GPR17 expression also sensitized cells to adenine nucleotide-induced cytotoxicity, whereas activation with uracil nucleotides promoted differentiation towards a more mature phenotype. We suggest that GFs may keep OPCs in a less differentiated stage by restraining GPR17 expression, and that, under permissive conditions, GPR17 contributes to OPCs differentiation. However, upon high extracellular adenine nucleotide concentrations, as during trauma and ischemia, GPR17 sensitizes cells to cytotoxicity. This double-edged sword role may be exploited to unveil new therapeutic approaches to acute and chronic brain disorders.


Asunto(s)
Adenosina Trifosfato/toxicidad , Diferenciación Celular/genética , Proteínas del Tejido Nervioso/genética , Oligodendroglía/citología , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Purinérgicos P2Y1/genética , Células Madre/citología , Células Madre/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Animales Recién Nacidos , Muerte Celular/genética , Células Cultivadas , Técnicas de Cocultivo , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/fisiología , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/fisiología , Receptores Purinérgicos P2Y1/biosíntesis
13.
J Biol Chem ; 286(12): 10593-604, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21209081

RESUMEN

The developing and mature central nervous system contains neural precursor cells expressing the proteoglycan NG2. Some of these cells continuously differentiate to myelin-forming oligodendrocytes; knowledge of the destiny of NG2(+) precursors would benefit from the characterization of new key functional players. In this respect, the G protein-coupled membrane receptor GPR17 has recently emerged as a new timer of oligodendrogliogenesis. Here, we used purified oligodendrocyte precursor cells (OPCs) to fully define the immunophenotype of the GPR17-expressing cells during OPC differentiation, unveil its native signaling pathway, and assess the functional consequences of GPR17 activation by its putative endogenous ligands, uracil nucleotides and cysteinyl leukotrienes (cysLTs). GPR17 presence was restricted to very early differentiation stages and completely segregated from that of mature myelin. Specifically, GPR17 decorated two subsets of slowly proliferating NG2(+) OPCs: (i) morphologically immature cells expressing other early proteins like Olig2 and PDGF receptor-α, and (ii) ramified preoligodendrocytes already expressing more mature factors, like O4 and O1. Thus, GPR17 is a new marker of these transition stages. In OPCs, GPR17 activation by either uracil nucleotides or cysLTs resulted in potent inhibition of intracellular cAMP formation. This effect was counteracted by GPR17 antagonists and receptor silencing with siRNAs. Finally, uracil nucleotides promoted and GPR17 inhibition, by either antagonists or siRNAs, impaired the normal program of OPC differentiation. These data have implications for the in vivo behavior of NG2(+) OPCs and point to uracil nucleotides and cysLTs as main extrinsic local regulators of these cells under physiological conditions and during myelin repair.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular , Regulación de la Expresión Génica/fisiología , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Células Madre/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/citología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Células Madre/citología , Nucleótidos de Uracilo/metabolismo , Nucleótidos de Uracilo/farmacología
14.
J Biol Chem ; 285(51): 40240-51, 2010 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-20956541

RESUMEN

Acid sphingomyelinase (A-SMase) is an important enzyme in sphingolipid metabolism and plays key roles in apoptosis, immunity, development, and cancer. In addition, it mediates cytotoxicity of cisplatin and some other chemotherapeutic drugs. The mechanism of A-SMase activation is still undefined. We now demonstrate that, upon CD95 stimulation, A-SMase is activated through translocation from intracellular compartments to the plasma membrane in an exocytic pathway requiring the t-SNARE protein syntaxin 4. Indeed, down-regulation of syntaxin 4 inhibits A-SMase translocation and activation induced by CD95 stimulation. This leads to inhibition of the CD95-triggered signaling events, including caspase 3 and 9 activation and apoptosis, activation of the survival pathway involving the protein kinase Akt, and important changes in cell cycle and proliferation. The molecular interaction between A-SMase and syntaxin 4 was not known and clarifies the mechanism of A-SMase activation. The novel actions of syntaxin 4 in sphingolipid metabolism and exocytosis we describe here define signaling mechanisms of broad relevance in cell pathophysiology.


Asunto(s)
Apoptosis/fisiología , Membrana Celular/enzimología , Exocitosis/fisiología , Proteínas Qa-SNARE/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Receptor fas/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Activación Enzimática/fisiología , Humanos , Transporte de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Esfingomielinas/metabolismo , Células U937
15.
Commun Integr Biol ; 3(4): 352-3, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20798824

RESUMEN

LIPIDS MAY AFFECT SYNAPTIC FUNCTION IN AT LEAST TWO WAYS: by acting as ligands for effector proteins [e.g., phosphatidylinositol (4,5) bisphosphate, diacylglycerol-mediated signaling] or by modifying the physicochemical properties and molecular organization of synaptic membranes. One that acts in the latter manner is cholesterol, an essential structural component of plasma membranes that is largely enriched in the membranes of synapses and synaptic vesicles, in which it may be involved in lipid-lipid and protein-lipid interactions. Cholesterol is an important constituent of the "membrane rafts" that may play a role in recruiting and organizing the specific proteins of the exocytic pathways. Furthermore, many synaptic proteins bind directly to cholesterol. The regulation of cholesterol and lipid levels may therefore influence the specific interactions and activity of synaptic proteins, and have a strong impact on synaptic functions.

16.
PLoS One ; 5(1): e8936, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-20126668

RESUMEN

Granins are major constituents of dense-core secretory granules in neuroendocrine cells, but their function is still a matter of debate. Work in cell lines has suggested that the most abundant and ubiquitously expressed granins, chromogranin A and B (CgA and CgB), are involved in granulogenesis and protein sorting. Here we report the generation and characterization of mice lacking chromogranin B (CgB-ko), which were viable and fertile. Unlike neuroendocrine tissues, pancreatic islets of these animals lacked compensatory changes in other granins and were therefore analyzed in detail. Stimulated secretion of insulin, glucagon and somatostatin was reduced in CgB-ko islets, in parallel with somewhat impaired glucose clearance and reduced insulin release, but normal insulin sensitivity in vivo. CgB-ko islets lacked specifically the rapid initial phase of stimulated secretion, had elevated basal insulin release, and stored and released twice as much proinsulin as wildtype (wt) islets. Stimulated release of glucagon and somatostatin was reduced as well. Surprisingly, biogenesis, morphology and function of insulin granules were normal, and no differences were found with regard to beta-cell stimulus-secretion coupling. We conclude that CgB is not required for normal insulin granule biogenesis or maintenance in vivo, but is essential for adequate secretion of islet hormones. Consequentially CgB-ko animals display some, but not all, hallmarks of human type-2 diabetes. However, the molecular mechanisms underlying this defect remain to be determined.


Asunto(s)
Cromogranina B/fisiología , Islotes Pancreáticos/metabolismo , Hormonas Pancreáticas/metabolismo , Animales , Cromogranina B/genética , Exocitosis , Insulina/metabolismo , Secreción de Insulina , Ratones , Ratones Noqueados
17.
J Cell Sci ; 123(Pt 4): 595-605, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20103534

RESUMEN

Cholesterol and sphingolipids are abundant in neuronal membranes, where they help the organisation of the membrane microdomains involved in major roles such as axonal and dendritic growth, and synapse and spine stability. The aim of this study was to analyse their roles in presynaptic physiology. We first confirmed the presence of proteins of the exocytic machinery (SNARES and Ca(v)2.1 channels) in the lipid microdomains of cultured neurons, and then incubated the neurons with fumonisin B (an inhibitor of sphingolipid synthesis), or with mevastatin or zaragozic acid (two compounds that affect the synthesis of cholesterol by inhibiting HMG-CoA reductase or squalene synthase). The results demonstrate that fumonisin B and zaragozic acid efficiently decrease sphingolipid and cholesterol levels without greatly affecting the viability of neurons or the expression of synaptic proteins. Electron microscopy showed that the morphology and number of synaptic vesicles in the presynaptic boutons of cholesterol-depleted neurons were similar to those observed in control neurons. Zaragozic acid (but not fumonisin B) treatment impaired synaptic vesicle uptake of the lipophilic dye FM1-43 and an antibody directed against the luminal epitope of synaptotagmin-1, effects that depended on the reduction in cholesterol because they were reversed by cholesterol reloading. The time-lapse confocal imaging of neurons transfected with ecliptic SynaptopHluorin showed that cholesterol depletion affects the post-depolarisation increase in fluorescence intensity. Taken together, these findings show that reduced cholesterol levels impair synaptic vesicle exocytosis in cultured neurons.


Asunto(s)
Colesterol/metabolismo , Exocitosis/fisiología , Vesículas Sinápticas/fisiología , Animales , Anticolesterolemiantes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Canales de Calcio Tipo N/metabolismo , Células Cultivadas , Exocitosis/efectos de los fármacos , Fumonisinas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inmunoglobulina G/metabolismo , Lovastatina/análogos & derivados , Lovastatina/farmacología , Microdominios de Membrana/metabolismo , Microscopía Electrónica de Transmisión , Modelos Neurológicos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neuronas/ultraestructura , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/fisiología , Terminales Presinápticos/ultraestructura , Ratas , Proteínas SNARE/metabolismo , Esfingolípidos/metabolismo , Vesículas Sinápticas/efectos de los fármacos , Vesículas Sinápticas/ultraestructura , Sinaptotagmina I/antagonistas & inhibidores , Sinaptotagmina I/inmunología , Sinaptotagmina I/metabolismo , Ácidos Tricarboxílicos/farmacología
19.
Brain ; 132(Pt 8): 2206-18, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19528093

RESUMEN

Upon central nervous system injury, the extracellular concentrations of nucleotides and cysteinyl-leukotrienes, two unrelated families of endogenous signalling molecules, are markedly increased at the site of damage, suggesting that they may act as 'danger signals' to alert responses to tissue damage and start repair. Here we show that, in non-injured spinal cord parenchyma, GPR17, a P2Y-like receptor responding to both uracil nucleotides (e.g. UDP-glucose) and cysteinyl-leukotrienes (e.g. LTD4 and LTC4), is present on a subset of neurons and of oligodendrocytes at different stages of maturation, whereas it is not expressed by astrocytes. GPR17 immunoreactivity was also found on ependymal cells lining the central canal that still retain some of the characteristics of stem/progenitor cells during adulthood. Induction of spinal cord injury (SCI) by acute compression resulted in marked cell death of GPR17+ neurons and oligodendrocytes inside the lesion followed by the appearance of proliferating GPR17+ microglia/macrophages migrating to and infiltrating into the lesioned area. Moreover, 72 h after SCI, GPR17+ ependymal cells started to proliferate and to express GFAP, suggesting their activation and 'de-differentiation' to pluripotent progenitor cells. The in vivo knock down of GPR17 by an antisense oligonucleotide strategy during SCI induction markedly reduced tissue damage and related histological and motor deficits, thus confirming the crucial role played by this receptor in the early phases of tissue damage development. Taken together, our findings suggest a dual and spatiotemporal-dependent role for GPR17 in SCI. At very early times after injury, GPR17 mediates neuronal and oligodendrocyte death inside the lesioned area. At later times, GPR17+ microglia/macrophages are recruited from distal parenchymal areas and move toward the lesioned zone, to suggest a role in orchestrating local remodelling responses. At the same time, the induction of the stem cell marker GFAP in GPR17+ ependymal cells suggests initiation of repair mechanisms. Thus, GPR17 may act as a 'sensor' of damage that is activated by nucleotides and cysteinyl-leukotrienes released in the lesioned area, and could also participate in post-injury responses. Moreover, its presence on spinal cord pre-oligodendrocytes and precursor-like cells suggests GPR17 as a novel target for therapeutic manipulation to foster remyelination and functional repair in SCI.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Biomarcadores/metabolismo , Movimiento Celular/fisiología , Proliferación Celular , Epéndimo/metabolismo , Técnicas de Silenciamiento del Gen , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos , Microglía/metabolismo , Microglía/patología , Trastornos del Movimiento/metabolismo , Trastornos del Movimiento/patología , Trastornos del Movimiento/prevención & control , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/fisiología , Neuronas/metabolismo , Oligodendroglía/metabolismo , Oligodesoxirribonucleótidos Antisentido , Células Madre Pluripotentes/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/fisiología , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/prevención & control
20.
PLoS One ; 3(10): e3579, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18974869

RESUMEN

Deciphering the mechanisms regulating the generation of new neurons and new oligodendrocytes, the myelinating cells of the central nervous system, is of paramount importance to address new strategies to replace endogenous damaged cells in the adult brain and foster repair in neurodegenerative diseases. Upon brain injury, the extracellular concentrations of nucleotides and cysteinyl-leukotrienes (cysLTs), two families of endogenous signaling molecules, are markedly increased at the site of damage, suggesting that they may act as "danger signals" to alert responses to tissue damage and start repair. Here we show that, in brain telencephalon, GPR17, a recently deorphanized receptor for both uracil nucleotides and cysLTs (e.g., UDP-glucose and LTD(4)), is normally present on neurons and on a subset of parenchymal quiescent oligodendrocyte precursor cells. We also show that induction of brain injury using an established focal ischemia model in the rodent induces profound spatiotemporal-dependent changes of GPR17. In the lesioned area, we observed an early and transient up-regulation of GPR17 in neurons expressing the cellular stress marker heat shock protein 70. Magnetic Resonance Imaging in living mice showed that the in vivo pharmacological or biotechnological knock down of GPR17 markedly prevents brain infarct evolution, suggesting GPR17 as a mediator of neuronal death at this early ischemic stage. At later times after ischemia, GPR17 immuno-labeling appeared on microglia/macrophages infiltrating the lesioned area to indicate that GPR17 may also acts as a player in the remodeling of brain circuitries by microglia. At this later stage, parenchymal GPR17+ oligodendrocyte progenitors started proliferating in the peri-injured area, suggesting initiation of remyelination. To confirm a specific role for GPR17 in oligodendrocyte differentiation, the in vitro exposure of cortical pre-oligodendrocytes to the GPR17 endogenous ligands UDP-glucose and LTD(4) promoted the expression of myelin basic protein, confirming progression toward mature oligodendrocytes. Thus, GPR17 may act as a "sensor" that is activated upon brain injury on several embryonically distinct cell types, and may play a key role in both inducing neuronal death inside the ischemic core and in orchestrating the local remodeling/repair response. Specifically, we suggest GPR17 as a novel target for therapeutic manipulation to foster repair of demyelinating wounds, the types of lesions that also occur in patients with multiple sclerosis.


Asunto(s)
Hipoxia Encefálica/patología , Proteínas del Tejido Nervioso/fisiología , Receptores Acoplados a Proteínas G/fisiología , Cicatrización de Heridas/genética , Animales , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Diferenciación Celular/efectos de los fármacos , Clonación Molecular , Perfilación de la Expresión Génica , Hipoxia Encefálica/genética , Hipoxia Encefálica/metabolismo , Leucotrieno D4/farmacología , Ratones , Modelos Biológicos , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Purinérgicos/genética , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células Madre/fisiología , Uridina Difosfato Glucosa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA