Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Epilepsia ; 64(10): 2827-2840, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37543852

RESUMO

OBJECTIVE: Posttranscriptional mechanisms are increasingly recognized as important contributors to the formation of hyperexcitable networks in epilepsy. Messenger RNA (mRNA) polyadenylation is a key regulatory mechanism governing protein expression by enhancing mRNA stability and translation. Previous studies have shown large-scale changes in mRNA polyadenylation in the hippocampus of mice during epilepsy development. The cytoplasmic polyadenylation element-binding protein CPEB4 was found to drive epilepsy-induced poly(A) tail changes, and mice lacking CPEB4 develop a more severe seizure and epilepsy phenotype. The mechanisms controlling CPEB4 function and the downstream pathways that influence the recurrence of spontaneous seizures in epilepsy remain poorly understood. METHODS: Status epilepticus was induced in wild-type and CPEB4-deficient male mice via an intra-amygdala microinjection of kainic acid. CLOCK binding to the CPEB4 promoter was analyzed via chromatin immunoprecipitation assay and melatonin levels via high-performance liquid chromatography in plasma. RESULTS: Here, we show increased binding of CLOCK to recognition sites in the CPEB4 promoter region during status epilepticus in mice and increased Cpeb4 mRNA levels in N2A cells overexpressing CLOCK. Bioinformatic analysis of CPEB4-dependent genes undergoing changes in their poly(A) tail during epilepsy found that genes involved in the regulation of circadian rhythms are particularly enriched. Clock transcripts displayed a longer poly(A) tail length in the hippocampus of mice post-status epilepticus and during epilepsy. Moreover, CLOCK expression was increased in the hippocampus in mice post-status epilepticus and during epilepsy, and in resected hippocampus and cortex of patients with drug-resistant temporal lobe epilepsy. Furthermore, CPEB4 is required for CLOCK expression after status epilepticus, with lower levels in CPEB4-deficient compared to wild-type mice. Last, CPEB4-deficient mice showed altered circadian function, including altered melatonin blood levels and altered clustering of spontaneous seizures during the day. SIGNIFICANCE: Our results reveal a new positive transcriptional-translational feedback loop involving CPEB4 and CLOCK, which may contribute to the regulation of the sleep-wake cycle during epilepsy.


Assuntos
Proteínas CLOCK , Epilepsia Resistente a Medicamentos , Epilepsia do Lobo Temporal , Melatonina , Proteínas de Ligação a RNA , Estado Epiléptico , Animais , Humanos , Masculino , Camundongos , Epilepsia do Lobo Temporal/metabolismo , Hipocampo , Melatonina/sangue , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Convulsões , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/genética , Fatores de Transcrição/metabolismo , Proteínas CLOCK/genética
2.
Br J Pharmacol ; 180(13): 1710-1729, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36637008

RESUMO

BACKGROUND AND PURPOSE: Neonatal seizures represent a clinical emergency. However, current anti-seizure medications fail to resolve seizures in ~50% of infants. The P2X7 receptor (P2X7R) is an important driver of inflammation, and evidence suggests that P2X7R contributes to seizures and epilepsy in adults. However, no genetic proof has yet been provided to determine what contribution P2X7R makes to neonatal seizures, its effects on inflammatory signalling during neonatal seizures, and the therapeutic potential of P2X7R-based treatments on long-lasting brain excitability. EXPERIMENTAL APPROACH: Neonatal seizures were induced by global hypoxia in 7-day-old mouse pups (P7). The role of P2X7Rs during seizures was analysed in P2X7R-overexpressing and knockout mice. Treatment of wild-type mice after hypoxia with the P2X7R antagonist JNJ-47965567 was used to determine the effects of the P2X7R on long-lasting brain hyperexcitability. Cell type-specific P2X7R expression was analysed in P2X7R-EGFP reporter mice. RNA sequencing was used to monitor P2X7R-dependent hippocampal downstream signalling. KEY RESULTS: P2X7R deletion reduced seizure severity, whereas P2X7R overexpression exacerbated seizure severity and reduced responsiveness to anti-seizure medication. P2X7R deficiency led to an anti-inflammatory phenotype in microglia, and treatment of mice with a P2X7R antagonist reduced long-lasting brain hyperexcitability. RNA sequencing identified several pathways altered in P2X7R knockout mice after neonatal hypoxia, including a down-regulation of genes implicated in inflammation and glutamatergic signalling. CONCLUSION AND IMPLICATIONS: Treatments based on targeting the P2X7R may represent a novel therapeutic strategy for neonatal seizures with P2X7Rs contributing to the generation of neonatal seizures, driving inflammatory processes and long-term hyperexcitability states.


Assuntos
Receptores Purinérgicos P2X7 , Convulsões , Animais , Camundongos , Animais Recém-Nascidos , Encéfalo/metabolismo , Epilepsia/induzido quimicamente , Epilepsia/metabolismo , Hipóxia/complicações , Inflamação/tratamento farmacológico , Camundongos Knockout , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo , Convulsões/metabolismo
3.
Epilepsia ; 64(2): 511-523, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36507708

RESUMO

OBJECTIVE: The P2X7 receptor (P2X7R) is an important contributor to neuroinflammation, responding to extracellularly released adenosine triphosphate. Expression of the P2X7R is increased in the brain in experimental and human epilepsy, and genetic or pharmacologic targeting of the receptor can reduce seizure frequency and severity in preclinical models. Experimentally induced seizures also increase levels of the P2X7R in blood. Here, we tested 18 F-JNJ-64413739, a positron emission tomography (PET) P2X7R antagonist, as a potential noninvasive biomarker of seizure-damage and epileptogenesis. METHODS: Status epilepticus was induced via an intra-amygdala microinjection of kainic acid. Static PET studies (30 min duration, initiated 30 min after tracer administration) were conducted 48 h after status epilepticus via an intravenous injection of 18 F-JNJ-64413739. PET images were coregistered with a brain magnetic resonance imaging atlas, tracer uptake was determined in the different brain regions and peripheral organs, and values were correlated to seizure severity during status epilepticus. 18 F-JNJ-64413739 was also applied to ex vivo human brain slices obtained following surgical resection for intractable temporal lobe epilepsy. RESULTS: P2X7R radiotracer uptake correlated strongly with seizure severity during status epilepticus in brain structures including the cerebellum and ipsi- and contralateral cortex, hippocampus, striatum, and thalamus. In addition, a correlation between radiotracer uptake and seizure severity was also evident in peripheral organs such as the heart and the liver. Finally, P2X7R radiotracer uptake was found elevated in brain sections from patients with temporal lobe epilepsy when compared to control. SIGNIFICANCE: Taken together, our data suggest that P2X7R-based PET imaging may help to identify seizure-induced neuropathology and temporal lobe epilepsy patients with increased P2X7R levels possibly benefitting from P2X7R-based treatments.


Assuntos
Epilepsia do Lobo Temporal , Estado Epiléptico , Camundongos , Humanos , Masculino , Animais , Epilepsia do Lobo Temporal/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Receptores Purinérgicos P2X7/uso terapêutico , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/diagnóstico por imagem , Estado Epiléptico/metabolismo , Convulsões/tratamento farmacológico
4.
An. R. Acad. Nac. Farm. (Internet) ; 88(número extraordinario): 189-197, diciembre 2022. ilus
Artigo em Espanhol | IBECS | ID: ibc-225776

RESUMO

Estudios previos han mostrado un papel clave de las células microgliales en los procesos neuroinflamatorios asociados con algunas enfermedades neurodegenerativas, como la enfermedad de Alzheimer (EA). La microglía detecta varios tipos de moléculas difusibles que regulan el múltiple repertorio de funciones microgliales. Entre ellos, los nucleótidos extracelulares, actuando sobre los receptores P2 microgliales, llevan a cabo un papel central. En este sentido, el receptor P2X7 ionotrópico ha sido reconocido como un regulador clave de las respuestas inflamatorias mediadas por la microglia. Se sabe que la microglía libera ATP y otros nucleótidos al medio extracelular. Aunque se han propuesto varios mecanismos, tales como la liberación a través de conexinas o panexinas, no se puede descartar un origen vesicular para estos nucleótidos liberados, basándose en la actividad del transportador vesicular de nucleótidos (VNUT).En este trabajo hemos analizado si la expresión de VNUT y el receptor P2X7, así como la liberación de ATP, podrían modificarse en la microglía reactiva. Para lograr la activación de la microglía estimulamos las células con el lipopolisacárido (LPS). Además, analizamos el efecto del péptido β1-amiloide, β1-42, que puede activar también las células microgliales, sobre la expresión de VNUT y la liberación de ATP en la microglía. (AU)


Previous studies have shown a key role of microglial cells in the neuroinflammatory processes associated with some neurodegenerative diseases, such as Alzheimer’s disease (AD). Microglia sense several types of diffusible molecules that regulate the multiple repertoire of microglial functions. Among them, extracellular nucleotides, acting on microglial P2 receptors, have central roles. In this sense, the ionotropic P2X7 receptor has gained recognition as a key regulator of microglial-mediated inflammatory responses. It is known that microglia releases ATP and other nucleotides to the extracellular medium. Although several mechanisms, such as release trough conexins or panexins, has been proposed, a vesicular origin for this released nucleotides, relying on the activity of the vesicular nucleotide transporter (VNUT), cannot be ruled out.In this work we evaluated whether the expression of VNUT and the P2X7 receptor, as well as the ATP release, could be modified in the reactive microglia. To achieve microglia activation we stimulated the cells with the lipopolysaccharide (LPS). Moreover, we analyzed the effect of the β-amyloid peptide β1-42, which is also able to activate the microglial cells, on the expression of VNUT and the ATP release in the microglia. (AU)


Assuntos
Humanos , Peptídeos beta-Amiloides , Receptores Purinérgicos , Microglia
5.
Br J Pharmacol ; 179(12): 2986-3006, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34962289

RESUMO

BACKGROUND AND PURPOSE: Refractory status epilepticus is a clinical emergency associated with high mortality and morbidity. Increasing evidence suggests neuroinflammation contributes to the development of drug-refractoriness during status epilepticus. Here, we have determined the contribution of the ATP-gated P2X7 receptor, previously linked to inflammation and increased hyperexcitability, to drug-refractory status epilepticus and its therapeutic potential. EXPERIMENTAL APPROACH: Status epilepticus was induced via a unilateral microinjection of kainic acid into the amygdala in adult mice. Severity of status epilepticus was compared in animals with overexpressing or knock-out of the P2X7 receptor, after inflammatory priming by pre-injection of bacterial lipopolysaccharide (LPS) and in mice treated with P2X7 receptor-targeting and anti-inflammatory drugs. KEY RESULTS: Mice overexpressing P2X7 receptors were unresponsive to several anticonvulsants (lorazepam, midazolam, phenytoin and carbamazepine) during status epilepticus. P2X7 receptor expression increased in microglia during status epilepticus, at times when responses to anticonvulsants were reduced. Overexpression of P2X7 receptors induced a pro-inflammatory phenotype in microglia during status epilepticus and the anti-inflammatory drug minocycline restored normal responses to anticonvulsants in mice overexpressing P2X7 receptors. Pretreatment of wild-type mice with LPS increased P2X7 receptor levels in the brain and reduced responsiveness to anticonvulsants during status epilepticus, which was overcome by either genetic deletion of P2X7 receptors or treatment with the P2X7 receptor antagonists, AFC-5128 or ITH15004. CONCLUSION AND IMPLICATIONS: Our results demonstrate that P2X7 receptor-induced pro-inflammatory effects contribute to resistance to pharmacotherapy during status epilepticus. Therapies targeting P2X7 receptors could be novel adjunctive treatments for drug-refractory status epilepticus.


Assuntos
Receptores Purinérgicos P2X7 , Estado Epiléptico , Trifosfato de Adenosina/metabolismo , Animais , Anticonvulsivantes/efeitos adversos , Convulsivantes/efeitos adversos , Lipopolissacarídeos/farmacologia , Camundongos , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/metabolismo
6.
STAR Protoc ; 2(4): 100964, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34841278

RESUMO

Low-density cell culture of the postnatal cerebellum, combined with live imaging and single-cell tracking, allows the behavior of postnatal cerebellar neural stem cells (NSCs) and their progeny to be monitored. Cultured cerebellar NSCs maintain their neurogenic nature giving rise, in the same relative proportions that exist in vivo, to the neuronal progeny generated by the three postnatal cerebellar neurogenic niches. This protocol describes the identification of the nature of the progeny through both post-imaging immunocytochemistry and patch-clamp recordings. For complete details on the use and execution of this protocol, please refer to Paniagua-Herranz et al. (2020b).


Assuntos
Cerebelo/citologia , Técnicas Citológicas/métodos , Células-Tronco Neurais/citologia , Animais , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Cells ; 10(9)2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34572093

RESUMO

Circulating molecules have potential as biomarkers to support the diagnosis of epilepsy and to assist with differential diagnosis, for example, in conditions resembling epilepsy, such as in psychogenic non-epileptic seizures (PNES). The P2X7 receptor (P2X7R) is an important regulator of inflammation and mounting evidence supports its activation in the brain during epilepsy. Whether the P2X7R or P2X7R-dependent signaling molecules can be used as biomarkers of epilepsy has not been reported. P2X7R levels were analyzed by quantitative ELISA using plasma samples from controls and patients with temporal lobe epilepsy (TLE) or PNES. Moreover, blood cell P2X7R expression and P2X7R-dependent cytokine signature was measured following status epilepticus in P2X7R-EGFP reporter, wildtype, and P2X7R-knockout mice. P2X7R plasma levels were higher in TLE patients when compared with controls and patients with PNES. Plasma levels of the broad inflammatory marker protein C-Reactive protein (CRP) were similar between the three groups. Using P2X7R-EGFP reporter mice, we identified monocytes as the main blood cell type expressing P2X7R after experimentally evoked seizures. Finally, cytokine array analysis in P2X7R-deficient mice identified KC/GRO as a potential P2X7R-dependent plasma biomarker following status epilepticus and during epilepsy. Our data suggest that P2X7R signaling components may be a promising subclass of circulating biomarkers to support the diagnosis of epilepsy.


Assuntos
Biomarcadores/sangue , Encéfalo/metabolismo , Epilepsia do Lobo Temporal/diagnóstico , Receptores Purinérgicos P2X7/sangue , Adulto , Animais , Estudos de Casos e Controles , Epilepsia do Lobo Temporal/sangue , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
Front Mol Neurosci ; 14: 732199, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34566578

RESUMO

Background: Evidence suggests that earlier diagnosis and initiation of treatment immediately after birth is critical for improved neurodevelopmental outcomes following neonatal encephalopathy (NE). Current diagnostic tests are, however, mainly restricted to clinical diagnosis with no molecular tests available. Purines including adenosine are released during brain injury such as hypoxia and are also present in biofluids. Whether blood purine changes can be used to diagnose NE has not been investigated to date. Methods: Blood purines were measured in a mouse model of neonatal hypoxia and infants with NE using a novel point-of-care diagnostic technology (SMARTChip) based on the summated electrochemical detection of adenosine and adenosine metabolites in the blood. Results: Blood purine concentrations were ∼2-3-fold elevated following hypoxia in mice [2.77 ± 0.48 µM (Control) vs. 7.57 ± 1.41 µM (post-hypoxia), p = 0.029]. Data in infants with NE had a 2-3-fold elevation when compared to healthy controls [1.63 ± 0.47 µM (Control, N = 5) vs. 4.87 ± 0.92 µM (NE, N = 21), p = 0.0155]. ROC curve analysis demonstrates a high sensitivity (81%) and specificity (80%) for our approach to identify infants with NE. Moreover, blood purine concentrations were higher in infants with NE and seizures [8.13 ± 3.23 µM (with seizures, N = 5) vs. 3.86 ± 0.56 µM (without seizures, N = 16), p = 0.044]. Conclusion: Our data provides the proof-of-concept that measurement of blood purine concentrations via SMARTChip technology may offer a low-volume bedside test to support a rapid diagnosis of NE.

9.
Stem Cell Reports ; 15(5): 1080-1094, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33065045

RESUMO

Little is known about the intrinsic specification of postnatal cerebellar neural stem cells (NSCs) and to what extent they depend on information from their local niche. Here, we have used an adapted cell preparation of isolated postnatal NSCs and live imaging to demonstrate that cerebellar progenitors maintain their neurogenic nature by displaying hallmarks of NSCs. Furthermore, by using this preparation, all the cell types produced postnatally in the cerebellum, in similar relative proportions to those observed in vivo, can be monitored. The fact that neurogenesis occurs in such organized manner in the absence of signals from the local environment, suggests that cerebellar lineage progression is to an important extent governed by cell-intrinsic or pre-programmed events. Finally, we took advantage of the absence of the niche to assay the influence of the vesicular nucleotide transporter inhibition, which dramatically reduced the number of NSCs in vitro by promoting their progression toward neurogenesis.


Assuntos
Cerebelo/metabolismo , Células-Tronco Neurais/citologia , Neurogênese , Proteínas de Transporte de Nucleotídeos/fisiologia , Imagem com Lapso de Tempo , Animais , Ciclo Celular , Diferenciação Celular , Divisão Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Microscopia , Análise de Célula Única
10.
Int J Mol Sci ; 21(21)2020 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-33105750

RESUMO

Neonatal seizures are one of the most common comorbidities of neonatal encephalopathy, with seizures aggravating acute injury and clinical outcomes. Current treatment can control early life seizures; however, a high level of pharmacoresistance remains among infants, with increasing evidence suggesting current anti-seizure medication potentiating brain damage. This emphasises the need to develop safer therapeutic strategies with a different mechanism of action. The purinergic system, characterised by the use of adenosine triphosphate and its metabolites as signalling molecules, consists of the membrane-bound P1 and P2 purinoreceptors and proteins to modulate extracellular purine nucleotides and nucleoside levels. Targeting this system is proving successful at treating many disorders and diseases of the central nervous system, including epilepsy. Mounting evidence demonstrates that drugs targeting the purinergic system provide both convulsive and anticonvulsive effects. With components of the purinergic signalling system being widely expressed during brain development, emerging evidence suggests that purinergic signalling contributes to neonatal seizures. In this review, we first provide an overview on neonatal seizure pathology and purinergic signalling during brain development. We then describe in detail recent evidence demonstrating a role for purinergic signalling during neonatal seizures and discuss possible purine-based avenues for seizure suppression in neonates.


Assuntos
Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P2/metabolismo , Convulsões/etiologia , Convulsões/terapia , Animais , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Modelos Animais de Doenças , Humanos , Hipotermia Induzida/métodos , Lactente , Recém-Nascido , Terapia de Alvo Molecular , Antagonistas do Receptor Purinérgico P2/farmacologia , Purinas/metabolismo , Convulsões/tratamento farmacológico
11.
Neurosci Bull ; 36(11): 1242-1258, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32895896

RESUMO

Mounting evidence suggests that the ATP-gated P2X7 receptor contributes to increased hyperexcitability in the brain. While increased expression of P2X7 in the hippocampus and cortex following status epilepticus and during epilepsy has been repeatedly demonstrated, the cell type-specific expression of P2X7 and its expression in extra-hippocampal brain structures remains incompletely explored. In this study, P2X7 expression was visualized by using a transgenic mouse model overexpressing P2X7 fused to the fluorescent protein EGFP. The results showed increased P2X7-EGFP expression after status epilepticus induced by intra-amygdala kainic acid and during epilepsy in different brain regions including the hippocampus, cortex, striatum, thalamus and cerebellum, and this was most evident in microglia and oligodendrocytes. Co-localization of P2X7-EGFP with cell type-specific markers was not detected in neurons or astrocytes. These data suggest that P2X7 activation is a common pathological hallmark across different brain structures, possibly contributing to brain inflammation and neurodegeneration following acute seizures and during epilepsy.


Assuntos
Epilepsia , Receptores Purinérgicos P2X7/metabolismo , Estado Epiléptico , Trifosfato de Adenosina , Animais , Encéfalo/metabolismo , Epilepsia/metabolismo , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Oligodendroglia/metabolismo , Estado Epiléptico/metabolismo
12.
Front Cell Neurosci ; 13: 224, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31156398

RESUMO

Vesicular storage of neurotransmitters, which allows their subsequent exocytotic release, is essential for chemical transmission in the central nervous system. Neurotransmitter uptake into secretory vesicles is carried out by vesicular transporters, which use the electrochemical proton gradient generated by a vacuolar H+-ATPase to drive neurotransmitter vesicular accumulation. ATP and other nucleotides are relevant extracellular signaling molecules that participate in a variety of biological processes. Although the active transport of nucleotides into secretory vesicles has been characterized from the pharmacological and biochemical point of view, the protein responsible for such vesicular accumulation remained unidentified for some time. In 2008, the human SLC17A9 gene, the last identified member of the SLC17 transporters, was found to encode the vesicular nucleotide transporter (VNUT). VNUT is expressed in various ATP-secreting cells and is able to transport a wide variety of nucleotides in a vesicular membrane potential-dependent manner. VNUT knockout mice lack vesicular storage and release of ATP, resulting in blockage of the purinergic transmission. This review summarizes the current studies on VNUT and analyzes the physiological relevance of the vesicular nucleotide transport in the central nervous system. The possible role of VNUT in the development of some pathological processes, such as chronic neuropathic pain or glaucoma is also discussed. The putative involvement of VNUT in these pathologies raises the possibility of the use of VNUT inhibitors for therapeutic purposes.

13.
An Real Acad Farm ; 85(2): 189-197, abr.-jun. 2019. graf
Artigo em Inglês | IBECS | ID: ibc-186176

RESUMO

Previous studies have shown a key role of microglial cells in the neuroinflammatory processes associated with some neurodegenerative diseases, such as Alzheimer’s disease (AD). Microglia sense several types of diffusible molecules that regulate the multiple repertoire of microglial functions. Among them, extracellular nucleotides, acting on microglial P2 receptors, have central roles. In this sense, the ionotropic P2X7 receptor has gained recognition as a key regulator of microglial-mediated inflammatory responses. It is known that microglia releases ATP and other nucleotides to the extracellular medium. Although several mechanisms, such as release trough conexins or panexins, has been proposed, a vesicular origin for this released nucleotides, relying on the activity of the vesicular nucleotide transporter (VNUT), cannot be ruled out. In this work we evaluated whether the expression of VNUT and the P2X7 receptor, as well as the ATP release, could be modified in the reactive microglia. To achieve microglia activation we stimulated the cells with the lipopolysaccharide (LPS). Moreover, we analyzed the effect of the b-amyloid peptide b1-42, which is also able to activate the microglial cells, on the expression of VNUT and the ATP release in the microgli


Estudios previos han mostrado un papel clave de las células microgliales en los procesos neuroinflamatorios asociados con algunas enfermedades neurodegenerativas, como la enfermedad de Alzheimer (EA). La microglía detecta varios tipos de moléculas difusibles que regulan el múltiple repertorio de funciones microgliales. Entre ellos, los nucleótidos extracelulares, actuando sobre los receptores P2 microgliales, llevan a cabo un papel central. En este sentido, el receptor P2X7 ionotrópico ha sido reconocido como un regulador clave de las respuestas inflamatorias mediadas por la microglia. Se sabe que la microglía libera ATP y otros nucleótidos al medio extracelular. Aunque se han propuesto varios mecanismos, tales como la liberación a través de conexinas o panexinas, no se puede descartar un origen vesicular para estos nucleótidos liberados, basándose en la actividad del transportador vesicular de nucleótidos (VNUT). En este trabajo hemos analizado si la expresión de VNUT y el receptor P2X7, así como la liberación de ATP, podrían modificarse en la microglía reactiva. Para lograr la activación de la microglía estimulamos las células con el lipopolisacárido (LPS). Además, analizamos el efecto del péptido (R)1-amiloide, (R)1-42, que puede activar también las células microgliales, sobre la expresión de VNUT y la liberación de ATP en la microglía


Assuntos
Humanos , Peptídeos beta-Amiloides/fisiologia , Microglia/metabolismo , Microglia/patologia , Degeneração Neural/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Reação em Cadeia da Polimerase em Tempo Real , Western Blotting , Imunofluorescência , Células Cultivadas , Transdução de Sinais
14.
An Real Acad Farm ; 84(1): 16-38, ene.-mar. 2018. ilus, graf, tab
Artigo em Espanhol | IBECS | ID: ibc-178047

RESUMO

El almacenamiento vesicular de los neurotransmisores, que permite su subsecuente liberación exocitótica, es un proceso esencial para la transmisión química en neuronas y células endocrinas. La acumulación de los neurotransmisores en vesículas de secreción se lleva a cabo por medio de transportadores vesiculares, que utilizan el gradiente electroquímico de protones generado por una ATPasa vacuolar como fuerza impulsora del transporte. El ATP, así como otros nucleótidos y dinucleótidos, son importantes moléculas señalizadoras que intervienen en una gran variedad de procesos biológicos. Aunque el transporte activo de nucleótidos se ha caracterizado desde el punto de vista bioquímico y farmacológico en una variedad de vesículas de secreción, la proteína responsable de esta acumulación vesicular permaneció durante mucho tiempo desconocida. En 2008, se demostró que SLC17A9, el último miembro identificado de la familia de transportadores SLC17, codifica el transportador vesicular de nucleótidos (VNUT). VNUT se expresa en una variedad de células que liberan ATP y ha mostrado ser capaz de transportar varios nucleótidos de manera dependiente del potencial de membrana vesicular. Ratones deficientes en VNUT pierden la capacidad de almacenar y liberar ATP de neuronas y células neuroendocrinas, lo que resulta en un bloqueo de la transmisión química purinérgica. En esta revisión se pretende resumir los estudios llevados a cabo hasta la fecha sobre VNUT y analizar la relevancia del transporte vesicular de nucleótidos en distintos tipos celulares y tejidos. Asimismo, se discute el posible uso de inhibidores de VNUT, así como de ARNs de interferencia que reduzcan su expresión, con fines terapéuticos


Vesicular storage of neurotransmitters, which allows their subsequent exocytotic release, is essential for chemical transmission in neurons and endocrine cells. Neurotransmitter uptake to secretory vesicles is carried out by vesicular transporters, which use the electrochemical gradient of protons generated by a vacuolar proton-ATPase as transport driving force. ATP and other nucleotides and dinucleotides are relevant signaling molecules that participate in a variety of biological process. Although the active transport of nucleotides has been pharmacologically and biochemically characterized in a diversity of secretory vesicles, the protein responsible for such vesicular accumulation remained unidentified for some time. In 2008, SLC17A9, the last identified member of the SLC17 transporter family, was found to encode the vesicular nucleotide transporter (VNUT). VNUT is expressed in various ATP-secreting cells and is able to transport several nucleotides in a vesicular membrane potential- dependent fashion. VNUT knockout mice lack vesicular storage and release of ATP from neurons and neuroendocrine cells, resulting in blockage of the purinergic chemical transmission. This review summarizes the current studies on VNUT and analyzes the relevance of vesicular nucleotide transport in different cells types and tissues. The possible use of VNUT inhibitors and interference RNA to reduce VNUT gene expression for therapeutic purposes is also discussed


Assuntos
Humanos , Proteínas Vesiculares de Transporte de Neurotransmissores/química , Sistemas Neurossecretores , Sistema Nervoso Central , Proteínas Vesiculares de Transporte de Neurotransmissores , Fotomicrografia
15.
J Vis Exp ; (130)2017 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-29286427

RESUMO

Understanding the mechanisms that control critical biological events of neural cell populations, such as proliferation, differentiation, or cell fate decisions, will be crucial to design therapeutic strategies for many diseases affecting the nervous system. Current methods to track cell populations rely on their final outcomes in still images and they generally fail to provide sufficient temporal resolution to identify behavioral features in single cells. Moreover, variations in cell death, behavioral heterogeneity within a cell population, dilution, spreading, or the low efficiency of the markers used to analyze cells are all important handicaps that will lead to incomplete or incorrect read-outs of the results. Conversely, performing live imaging and single cell tracking under appropriate conditions represents a powerful tool to monitor each of these events. Here, a time-lapse video-microscopy protocol, followed by post-processing, is described to track neural populations with single cell resolution, employing specific software. The methods described enable researchers to address essential questions regarding the cell biology and lineage progression of distinct neural populations.


Assuntos
Biologia Celular/instrumentação , Linhagem da Célula/fisiologia , Rastreamento de Células/métodos , Microscopia de Vídeo/métodos , Neurônios/ultraestrutura , Análise de Célula Única/métodos , Animais , Diferenciação Celular/fisiologia , Técnicas Citológicas/métodos , Humanos , Monitorização Fisiológica , Neurônios/citologia
16.
Front Pharmacol ; 8: 951, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29311945

RESUMO

Adenosine triphosphate (ATP) is an important extracellular neurotransmitter that participates in several critical processes like cell differentiation, neuroprotection or axon guidance. Prior to its exocytosis, ATP must be stored in secretory vesicles, a process that is mediated by the Vesicular Nucleotide Transporter (VNUT). This transporter has been identified as the product of the SLC17A9 gene and it is prominently expressed in discrete brain areas, including the cerebellum. The main population of cerebellar neurons, the glutamatergic granule neurons, depends on purinergic signaling to trigger neuroprotective responses. However, while nucleotide receptors like P2X7 and P2Y13 are known to be involved in neuroprotection, the mechanisms that regulate ATP release in relation to such events are less clearly understood. In this work, we demonstrate that cerebellar granule cells express a functional VNUT that is involved in the regulation of ATP exocytosis. Numerous vesicles loaded with this nucleotide can be detected in these granule cells and are staining by the fluorescent ATP-marker, quinacrine. High potassium stimulation reduces quinacrine fluorescence in granule cells, indicating they release ATP via calcium dependent exocytosis. Specific subcellular markers were used to assess the localization of VNUT in granule cells, and the transporter was detected in both the axonal and somatodendritic compartments, most predominantly in the latter. However, co-localization with the specific lysosomal marker LAMP-1 indicated that VNUT can also be found in non-synaptic vesicles, such as lysosomes. Interestingly, the weak co-localization between VNUT and VGLUT1 suggests that the ATP and glutamate vesicle pools are segregated, as also observed in the cerebellar cortex. During post-natal cerebellar development, VNUT is found in granule cell precursors, co-localizing with markers of immature cells like doublecortin, suggesting that this transporter may be implicated in the initial stages of granule cell development.

17.
Purinergic Signal ; 11(2): 239-49, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25847073

RESUMO

Before being released, nucleotides are stored in secretory vesicles through the vesicular nucleotide transporter (VNUT). Once released, extracellular ATP participates in neuronal differentiation processes. Thus, the expression of a functional VNUT could be an additional component of the purinergic system which regulates neuronal differentiation and axonal elongation. In vitro expression of VNUT decreases neuritogenesis in N2a cells differentiated by retinoic acid treatment, whereas silencing of VNUT expression increases the number and length of neurites in these cells. These results highlight the role of VNUT in the neuritogenic process because this transporter regulates the ATP content in neurosecretory vesicles.


Assuntos
Trifosfato de Adenosina/metabolismo , Diferenciação Celular/fisiologia , Neurônios/citologia , Proteínas de Transporte de Nucleotídeos/metabolismo , Animais , Transporte Biológico , Células Cultivadas , Espaço Extracelular/metabolismo , Camundongos , Nucleotídeos/metabolismo
18.
FEBS J ; 279(17): 3022-32, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22748038

RESUMO

Within the last 10 years, the use of different RNases as therapeutic agents for various diseases has been pursued. Furthermore, the advancements of recombinant technology have allowed the assembly of proteins with different functions. In this regard, immunoribonucleases (immunoRNases) stand out as some of the most promising therapeutic candidates given their enzymatic and non-mutagenic character. Accordingly, the work reported here describes fusing RNase T1, one of the most studied members of the microbial RNase family, to the single-chain variable fragment (scFv) of a monoclonal antibody that targets the glycoprotein A33 antigen (GPA33) from human colon cancer cells. A heterologous production system, which employs the yeast Pichia pastoris, has been optimized to produce this immunoRNase (scFvA33T1) with yields of ∼ 5-10 mg · L(-1). The purified protein appears to be correctly folded as it retains its antigen specificity and ribonucleolytic activity. Finally, it also shows specific binding to, internalization into and toxicity against GPA33-positive cell lines compared with the control, GPA33-negative cells. Overall, it can be concluded that scFvA33T1 is a promising therapeutic fusion protein with the additional advantage that presumably it can be produced and purified in large amounts using an easily scalable yeast-based system.


Assuntos
Neoplasias do Colo/enzimologia , Ribonuclease T1/metabolismo , Anticorpos de Cadeia Única , Dicroísmo Circular , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Eletroforese em Gel de Poliacrilamida , Endocitose , Humanos , Espectrofotometria Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...