Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 300(4): 107138, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38447794

RESUMEN

Short tandem repeats are inherently unstable during DNA replication depending on repeat length, and the expansion of the repeat length in the human genome is responsible for repeat expansion disorders. Pentanucleotide AAGGG and ACAGG repeat expansions in intron 2 of the gene encoding replication factor C subunit 1 (RFC1) cause cerebellar ataxia, neuropathy, vestibular areflexia syndrome (CANVAS) and other phenotypes of late-onset cerebellar ataxia. Herein, we reveal the structural polymorphism of the RFC1 repeats associated with CANVAS in vitro. Single-stranded AAGGG repeat DNA formed a hybrid-type G-quadruplex, whereas its RNA formed a parallel-type G-quadruplex with three layers. The RNA of the ACAGG repeat formed hairpin structure comprising C-G and G-C base pairs with A:A and GA:AG mismatched repeats. Furthermore, both pathogenic repeat RNAs formed more rigid structures than those of the nonpathogenic repeat RNAs. These findings provide novel insights into the structural polymorphism of the RFC1 repeats, which may be closely related to the disease mechanism of CANVAS.


Asunto(s)
Ataxia Cerebelosa , Expansión de las Repeticiones de ADN , Enfermedades del Sistema Nervioso Periférico , Proteína de Replicación C , Enfermedades Vestibulares , Humanos , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/metabolismo , G-Cuádruplex , Repeticiones de Microsatélite , Polimorfismo Genético , Proteína de Replicación C/genética , Proteína de Replicación C/metabolismo , Proteína de Replicación C/química , ARN/química , ARN/genética , ARN/metabolismo , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/metabolismo , Enfermedades Vestibulares/genética , Enfermedades Vestibulares/metabolismo
2.
J Clin Invest ; 133(22)2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37707954

RESUMEN

Expansion of CAG and CTG (CWG) triplet repeats causes several inherited neurological diseases. The CWG repeat diseases are thought to involve complex pathogenic mechanisms through expanded CWG repeat-derived RNAs in a noncoding region and polypeptides in a coding region, respectively. However, an effective therapeutic approach has not been established for the CWG repeat diseases. Here, we show that a CWG repeat DNA-targeting compound, cyclic pyrrole-imidazole polyamide (CWG-cPIP), suppressed the pathogenesis of coding and noncoding CWG repeat diseases. CWG-cPIP bound to the hairpin form of mismatched CWG DNA, interfering with transcription elongation by RNA polymerase through a preferential activity toward repeat-expanded DNA. We found that CWG-cPIP selectively inhibited pathogenic mRNA transcripts from expanded CWG repeats, reducing CUG RNA foci and polyglutamine accumulation in cells from patients with myotonic dystrophy type 1 (DM1) and Huntington's disease (HD). Treatment with CWG-cPIP ameliorated behavioral deficits in adeno-associated virus-mediated CWG repeat-expressing mice and in a genetic mouse model of HD, without cytotoxicity or off-target effects. Together, we present a candidate compound that targets expanded CWG repeat DNA independently of its genomic location and reduces both pathogenic RNA and protein levels. CWG-cPIP may be used for the treatment of CWG repeat diseases and improvement of clinical outcomes.


Asunto(s)
Enfermedad de Huntington , Distrofia Miotónica , Humanos , Animales , Ratones , ARN/genética , Expansión de Repetición de Trinucleótido/genética , Nylons/farmacología , Distrofia Miotónica/genética , Repeticiones de Trinucleótidos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , ADN , Imidazoles/farmacología
3.
CNS Neurosci Ther ; 29(6): 1547-1560, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36786129

RESUMEN

AIMS: We previously found that a decoy peptide derived from the C-terminal sequence of α-Synuclein (αSyn) prevents cytotoxic αSyn aggregation caused by fatty acid-binding protein 3 (FABP3) in vitro. In this study, we continued to utilize αSyn-derived peptides to further validate their effects on αSyn neurotoxicity and behavioral impairments in αSyn preformed fibrils (PFFs)-injected mouse model of Parkinson's disease (PD). METHODS: Mice were injected with αSyn PFFs in the bilateral olfactory bulb (OB) and then were subjected to behavioral analysis at 2-week intervals post-injection. Peptides nasal administration was initiated one week after injection. Changes in phosphorylation of αSyn and neuronal damage in the OB were measured using immunostaining at week 4. The effect of peptides on the interaction between αSyn and FABP3 was examined using co-immunoprecipitation. RESULTS: αSyn PFF-injected mice showed significant memory loss but no motor function impairment. Long-term nasal treatment with peptides effectively prevented memory impairment. In peptide-treated αSyn PFF-injected mice, the peptides entered the OB smoothly through the nasal cavity and were mainly concentrated in neurons in the mitral cell layer, significantly suppressing the excessive phosphorylation of αSyn and reducing the formation of αSyn-FABP3 oligomers, thereby preventing neuronal death. The addition of peptides also blocked the interaction of αSyn and FABP3 at the recombinant protein level, and its effect was strongest at molar concentrations comparable to those of αSyn and FABP3. CONCLUSIONS: Our findings suggest that the αSyn decoy peptide represents a novel therapeutic approach for reducing the accumulation of toxic αSyn-FABP3 oligomers in the brain, thereby preventing the progression of synucleinopathies.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Ratones , Animales , alfa-Sinucleína/metabolismo , Neuronas/metabolismo , Encéfalo/metabolismo , Amnesia/metabolismo , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/prevención & control , Trastornos de la Memoria/metabolismo
4.
Sci Adv ; 9(8): eade2035, 2023 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-36827365

RESUMEN

Consecutive guanine RNA sequences can adopt quadruple-stranded structures, termed RNA G-quadruplexes (rG4s). Although rG4-forming sequences are abundant in transcriptomes, the physiological roles of rG4s in the central nervous system remain poorly understood. In the present study, proteomics analysis of the mouse forebrain identified DNAPTP6 as an RNA binding protein with high affinity and selectivity for rG4s. We found that DNAPTP6 coordinates the assembly of stress granules (SGs), cellular phase-separated compartments, in an rG4-dependent manner. In neurons, the knockdown of DNAPTP6 diminishes the SG formation under oxidative stress, leading to synaptic dysfunction and neuronal cell death. rG4s recruit their mRNAs into SGs through DNAPTP6, promoting RNA self-assembly and DNAPTP6 phase separation. Together, we propose that the rG4-dependent phase separation of DNAPTP6 plays a critical role in neuronal function through SG assembly.


Asunto(s)
G-Cuádruplex , ARN , Animales , Ratones , ARN/química , Gránulos de Estrés , ARN Mensajero/genética , Neuronas/metabolismo
5.
Nihon Yakurigaku Zasshi ; 158(1): 15, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-36596479
6.
Nihon Yakurigaku Zasshi ; 158(1): 30-33, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-36596485

RESUMEN

Repeat expansion diseases are caused by the aberrant repeat expansions within specific genes. RNAs derived from aberrant repeat sequences form non-canonical secondary structures, contributing to induce cell toxicity. In particular, RNA G-quadruplexes (G4RNAs) formed in guanine-rich repeat expanded RNAs trigger neurodegeneration. We have previously shown that the expanded CGG repeat-derived G4RNAs initiate aggregation of FMRpolyG, a neuropathogenic protein generated by repeat-associated non-AUG (RAN) translation in Fragile X-associated tremor/ataxia syndrome (FXTAS). In this review, we describe the neuropathological mechanism attributed to G4RNAs in guanine-rich repeat expansion diseases, including FXTAS.


Asunto(s)
Síndrome del Cromosoma X Frágil , Expansión de Repetición de Trinucleótido , Humanos , Expansión de Repetición de Trinucleótido/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/patología , Ataxia/genética , Ataxia/metabolismo , Ataxia/patología , ARN
7.
Clin Epigenetics ; 13(1): 204, 2021 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-34774111

RESUMEN

BACKGROUND: GGC repeat expansions in NOTCH2NLC are associated with neuronal intranuclear inclusion disease. Very recently, asymptomatic carriers with NOTCH2NLC repeat expansions were reported. In these asymptomatic individuals, the CpG island in NOTCH2NLC is hypermethylated, suggesting that two factors repeat length and DNA methylation status should be considered to evaluate pathogenicity. Long-read sequencing can be used to simultaneously profile genomic and epigenomic alterations. We analyzed four sporadic cases with NOTCH2NLC repeat expansion and their phenotypically normal parents. The native genomic DNA that retains base modification was sequenced on a per-trio basis using both PacBio and Oxford Nanopore long-read sequencing technologies. A custom workflow was developed to evaluate DNA modifications. With these two technologies combined, long-range DNA methylation information was integrated with complete repeat DNA sequences to investigate the genetic origins of expanded GGC repeats in these sporadic cases. RESULTS: In all four families, asymptomatic fathers had longer expansions (median: 522, 390, 528 and 650 repeats) compared with their affected offspring (median: 93, 117, 162 and 140 repeats, respectively). These expansions are much longer than the disease-causing range previously reported (in general, 41-300 repeats). Repeat lengths were extremely variable in the father, suggesting somatic mosaicism. Instability is more frequent in alleles with uninterrupted pure GGCs. Single molecule epigenetic analysis revealed complex DNA methylation patterns and epigenetic heterogeneity. We identified an aberrant gain-of-methylation region (2.2 kb in size beyond the CpG island and GGC repeats) in asymptomatic fathers. This methylated region was unmethylated in the normal allele with bilateral transitional zones with both methylated and unmethylated CpG dinucleotides, which may be protected from methylation to ensure NOTCH2NLC expression. CONCLUSIONS: We clearly demonstrate that the four sporadic NOTCH2NLC-related cases are derived from the paternal GGC repeat contraction associated with demethylation. The entire genetic and epigenetic landscape of the NOTCH2NLC region was uncovered using the custom workflow of long-read sequence data, demonstrating the utility of this method for revealing epigenetic/mutational changes in repetitive elements, which are difficult to characterize by conventional short-read/bisulfite sequencing methods. Our approach should be useful for biomedical research, aiding the discovery of DNA methylation abnormalities through the entire genome.


Asunto(s)
Relaciones Padre-Hijo , Antecedentes Genéticos , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas del Tejido Nervioso/genética , Metilación de ADN/genética , Metilación de ADN/fisiología , Epigénesis Genética/genética , Epigénesis Genética/fisiología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/estadística & datos numéricos , Humanos , Péptidos y Proteínas de Señalización Intercelular/análisis , Proteínas del Tejido Nervioso/análisis
8.
J Biol Chem ; 296: 100663, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33862084

RESUMEN

α-synuclein (αSyn) is a protein known to form intracellular aggregates during the manifestation of Parkinson's disease. Previously, it was shown that αSyn aggregation was strongly suppressed in the midbrain region of mice that did not possess the gene encoding the lipid transport protein fatty acid binding protein 3 (FABP3). An interaction between these two proteins was detected in vitro, suggesting that FABP3 may play a role in the aggregation and deposition of αSyn in neurons. To characterize the molecular mechanisms that underlie the interactions between FABP3 and αSyn that modulate the cellular accumulation of the latter, in this report, we used in vitro fluorescence assays combined with fluorescence microscopy, transmission electron microscopy, and quartz crystal microbalance assays to characterize in detail the process and consequences of FABP3-αSyn interaction. We demonstrated that binding of FABP3 to αSyn results in changes in the aggregation mechanism of the latter; specifically, a suppression of fibrillar forms of αSyn and also the production of aggregates with an enhanced cytotoxicity toward mice neuro2A cells. Because this interaction involved the C-terminal sequence region of αSyn, we tested a peptide derived from this region of αSyn (αSynP130-140) as a decoy to prevent the FABP3-αSyn interaction. We observed that the peptide competitively inhibited binding of αSyn to FABP3 in vitro and in cultured cells. We propose that administration of αSynP130-140 might be used to prevent the accumulation of toxic FABP3-αSyn oligomers in cells, thereby preventing the progression of Parkinson's disease.


Asunto(s)
Amiloide/antagonistas & inhibidores , Proteína 3 de Unión a Ácidos Grasos/metabolismo , Neuroblastoma/patología , Fragmentos de Péptidos/farmacología , Agregación Patológica de Proteínas/prevención & control , alfa-Sinucleína/metabolismo , Amiloide/metabolismo , Animales , Proteína 3 de Unión a Ácidos Grasos/genética , Humanos , Ratones , Neuroblastoma/genética , Neuroblastoma/metabolismo , Células Tumorales Cultivadas , alfa-Sinucleína/antagonistas & inhibidores , alfa-Sinucleína/genética
9.
Sci Adv ; 7(3)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33523882

RESUMEN

Fragile X-related tremor/ataxia syndrome (FXTAS) is a neurodegenerative disease caused by CGG triplet repeat expansions in FMR1, which elicit repeat-associated non-AUG (RAN) translation and produce the toxic protein FMRpolyG. We show that FMRpolyG interacts with pathogenic CGG repeat-derived RNA G-quadruplexes (CGG-G4RNA), propagates cell to cell, and induces neuronal dysfunction. The FMRpolyG polyglycine domain has a prion-like property, preferentially binding to CGG-G4RNA. Treatment with 5-aminolevulinic acid, which is metabolized to protoporphyrin IX, inhibited RAN translation of FMRpolyG and CGG-G4RNA-induced FMRpolyG aggregation, ameliorating aberrant synaptic plasticity and behavior in FXTAS model mice. Thus, we present a novel therapeutic strategy to target G4RNA prionoids.


Asunto(s)
Síndrome del Cromosoma X Frágil , G-Cuádruplex , Enfermedades Neurodegenerativas , Animales , Ataxia/genética , Ataxia/metabolismo , Ataxia/patología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Ratones , Temblor/genética , Temblor/metabolismo
10.
Acta Physiol (Oxf) ; 232(1): e13613, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33393208

RESUMEN

AIMS: Adult hippocampal neurogenesis plays an important role in neuronal plasticity and maintenance in mammals. Low-threshold voltage-gated T-type calcium channels produce calcium spikes that increase fast action potentials in newborn cells in the hippocampal dentate gyrus (DG); however, their role in adult hippocampal neurogenesis remains unclear. Here, we demonstrate impaired adult hippocampal neurogenesis in Cav3.1T-type calcium channel knockout mice. METHODS AND RESULTS: Cav3.1T-type calcium channel was predominantly localized in neuronal progenitor cells of the mouse hippocampal DG. By counting the number of 5-bromo-2'-deoxyuridine-labeled cells, decreased proliferation and survival of newly generated cells were observed in the adult hippocampal DG in Cav3.1 knockout mice as compared to wild-type (WT) mice. Moreover, the degree of maturation of doublecortin-positive cells in Cav3.1 knockout mice was lower than that in WT mice, suggesting that Cav3.1 deletion may impair neuronal differentiation. Consistent with impaired hippocampal neurogenesis, Cav3.1 knockout mice showed decreased social interaction. Reduced phosphorylation levels of calcium/calmodulin-dependent protein kinase II and protein kinase B were closely associated with impaired hippocampal neurogenesis in Cav3.1 knockout mice. Moreover, the mRNA and protein expression levels of brain-derived neurotrophic factor, important for neurogenesis, were significantly decreased in Cav3.1 knockout mice. Finally, gene ontology analysis revealed alterations in genes related to the promotion of cell death/apoptosis and suppression of cell proliferation/neuronal differentiation pathways, including Bdnf. CONCLUSION: These results suggest that the Cav3.1T-type calcium channel may be a key molecule required for cell proliferation, survival and neuronal differentiation in newly generated cells of the adult mouse hippocampus.


Asunto(s)
Canales de Calcio Tipo T , Animales , Canales de Calcio Tipo T/metabolismo , Proliferación Celular , Supervivencia Celular , Giro Dentado , Hipocampo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurogénesis , Neuronas/metabolismo
11.
Int J Mol Sci ; 22(1)2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33401521

RESUMEN

In synucleinopathies, while motor symptoms are thought to be attributed to the accumulation of misfolded α-synuclein (αSyn) in nigral dopaminergic neurons, it remains to be elucidated how cognitive decline arises. Here, we investigated the effects of distinct αSyn strains on cognition and the related neuropathology in the medial septum/diagonal band (MS/DB), a key region for cognitive processing. Bilateral injection of αSyn fibrils into the dorsal striatum potently impaired cognition in mice. The cognitive decline was accompanied by accumulation of phosphorylated αSyn at Ser129 and reduction of gamma-aminobutyric acid (GABA)-ergic but not cholinergic neurons in the MS/DB. Since we have demonstrated that fatty acid-binding protein 3 (FABP3) is critical for αSyn neurotoxicity in nigral dopaminergic neurons, we investigated whether FABP3 also participates in αSyn pathology in the MS/DB and cognitive decline. FABP3 was highly expressed in GABAergic but rarely in cholinergic neurons in the MS/DB. Notably, Fabp3 deletion antagonized the accumulation of phosphorylated αSyn, decrease in GABAergic neurons, and cognitive impairment caused by αSyn fibrils. Overall, the present study indicates that FABP3 mediates αSyn neurotoxicity in septal GABAergic neurons and the resultant cognitive impairment, and that FABP3 in this subpopulation could be a therapeutic target for dementia in synucleinopathies.


Asunto(s)
Disfunción Cognitiva/etiología , Proteína 3 de Unión a Ácidos Grasos/metabolismo , Neuronas GABAérgicas/metabolismo , Neuroprotección , Sinucleinopatías/complicaciones , Animales , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/prevención & control , Proteína 3 de Unión a Ácidos Grasos/fisiología , Neuronas GABAérgicas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Sinucleinopatías/fisiopatología , alfa-Sinucleína
12.
Front Pharmacol ; 11: 583291, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33281604

RESUMEN

A previous study reported that scabronine G methyl ester (SG-ME) potentially enhances the in vitro secretion of neurotrophic factors such as nerve growth factor via the protein kinase C (PKC)-ζ pathway. However, it remains unknown whether SG-ME can improve cognitive dysfunctions in olfactory bulbectomized (OBX) mice. To address this question, we evaluated SG-ME-treated and untreated OBX mice in a passive avoidance test. We also investigated potential effects of SG-ME on several parameters: cell proliferation and cAMP response element-binding protein (CREB) phosphorylation in the hippocampal dentate gyrus by immunohistochemistry, brain-derived neurotrophic factor (BDNF) levels in the hippocampus by Western blotting, p-CREB levels in the hippocampus by MapAnalyzer, and long-term potentiation (LTP) by electrophysiology. On the 14th day after surgery OBX mice showed altered passive avoidance and decreases in both cell proliferation and long-term potentiation in the hippocampus, while these changes were reversed by SG-ME (20 µg/mouse) 24 h after the treatment. The improvement in memory deficits was prevented when SG-ME was co-administeredwith either zeta inhibitory peptide (PKC-ζ inhibitor), anti-BDNF antibody, ANA-12 (TrkB antagonist), U0126 (MEK inhibitor), H-89 (PKA inhibitor), LY294002 (PI3K inhibitor) or KN-93 (CaMKII inhibitor). We found that SG-ME enhanced brain-derived neurotrophic factor and p-CREB levels in the hippocampus while p-CREB was localized in neurons, but not in astrocytes nor microglial cells. These findings revealed the potential of SG-ME in improving memory impairments by enhancing cell proliferation and LTP via activation of the BDNF/CREB signaling pathway in neurons.

13.
Yakugaku Zasshi ; 140(10): 1207-1212, 2020.
Artículo en Japonés | MEDLINE | ID: mdl-32999199

RESUMEN

T-type calcium channels are low-threshold voltage-gated calcium channel and characterized by unique electrophysiological properties such as fast inactivation and slow deactivation kinetics. All subtypes of T-type calcium channel (Cav3.1, 3.2 and 3.3) are widely expressed in the central nerve system, and they have an important role in homeostasis of sleep, pain response, and development of epilepsy. Recently, several reports suggest that T-type calcium channels may mediate neuronal plasticity in the mouse brain. We succeeded to develop T-type calcium channel enhancer ethyl 8'-methyl-2',4-dioxo-2-(piperidin-1-yl)-2'H-spiro[cyclopentane-1,3'-imidazo[1,2-a]pyridine]-2-ene-3-carboxylate (SAK3) which enhances Cav3.1 and 3.3 currents in each-channel expressed neuro2A cells. SAK3 can promote acetylcholine (ACh) release in the mouse hippocampus via enhancing T-type calcium channel. In this review, we have introduced the role of T-type calcium channel, especially Cav3.1 channel in the mouse hippocampus based on our previous data using SAK3 and Cav3.1 knockout mice.


Asunto(s)
Canales de Calcio Tipo T/efectos de los fármacos , Canales de Calcio Tipo T/fisiología , Imidazoles/farmacología , Neuronas/fisiología , Compuestos de Espiro/farmacología , Acetilcolina/metabolismo , Animales , Encéfalo/fisiología , Canales de Calcio Tipo T/genética , Canales de Calcio Tipo T/metabolismo , Células Cultivadas , Sistema Nervioso Central/metabolismo , Fenómenos Electrofisiológicos , Epilepsia/etiología , Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Homeostasis , Ratones , Plasticidad Neuronal , Dolor/etiología , Ratas , Sueño/fisiología
14.
Int J Mol Sci ; 21(15)2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32752296

RESUMEN

Recently, we developed the fatty acid-binding protein 3 (FABP3) ligand MF1 (4-(2-(1-(2-chlorophenyl)-5-phenyl-1H-pyrazol-3-yl)phenoxy) butanoic acid) as a therapeutic candidate for α-synucleinopathies. MF1 shows affinity towards γ-aminobutyric acid type-A (GABAA) receptor, but its effect on the receptor remains unclear. Here, we investigate the pharmacological properties of MF1 on the GABAA receptor overexpressed in Neuro2A cells. While MF1 (1-100 µm) alone failed to evoke GABA currents, MF1 (1 µm) promoted GABA currents during GABA exposure (1 and 10 µm). MF1-promoted GABA currents were blocked by flumazenil (10 µm) treatment, suggesting that MF1 enhances receptor function via the benzodiazepine recognition site. Acute and chronic administration of MF1 (0.1, 0.3 and 1.0 mg/kg, p.o.) significantly attenuated status epilepticus (SE) and the mortality rate in pilocarpine (PILO: 300 mg/kg, i.p.)-treated mice, similar to diazepam (DZP: 5.0 mg/kg, i.p.). The anti-epileptic effects of DZP (5.0 mg/kg, i.p.) and MF1 (0.3 mg/kg, p.o.) were completely abolished by flumazenil (25 mg/kg, i.p.) treatment. Pentylenetetrazol (PTZ: 90 mg/kg, i.p.)-induced seizures in mice were suppressed by DZP (5.0 mg/kg, i.p.), but not MF1. Collectively, this suggests that MF1 is a mild enhancer of the GABAA receptor and exercises anti-epileptic effects through the receptor's benzodiazepine recognition site in PILO-induced SE models.


Asunto(s)
Anticonvulsivantes/farmacología , Benzodiazepinas/farmacología , Proteína 3 de Unión a Ácidos Grasos/metabolismo , Receptores de GABA-A/metabolismo , Estado Epiléptico/tratamiento farmacológico , Animales , Anticonvulsivantes/química , Anticonvulsivantes/metabolismo , Benzodiazepinas/metabolismo , Sitios de Unión , Línea Celular Tumoral , Diazepam/metabolismo , Diazepam/farmacología , Flumazenil/metabolismo , Flumazenil/farmacología , Ligandos , Masculino , Ratones Endogámicos ICR , Pentilenotetrazol/metabolismo , Pentilenotetrazol/farmacología , Convulsiones/tratamiento farmacológico , Convulsiones/metabolismo , Estado Epiléptico/metabolismo
15.
Int J Mol Sci ; 21(6)2020 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-32210174

RESUMEN

Oligomerization and/or aggregation of α-synuclein (α-Syn) triggers α-synucleinopathies such as Parkinson's disease and dementia with Lewy bodies. It is known that α-Syn can spread in the brain like prions; however, the mechanism remains unclear. We demonstrated that fatty acid binding protein 3 (FABP3) promotes propagation of α-Syn in mouse brain. Animals were injected with mouse or human α-Syn pre-formed fibrils (PFF) into the bilateral substantia nigra pars compacta (SNpc). Two weeks after injection of mouse α-Syn PFF, wild-type (WT) mice exhibited motor and cognitive deficits, whereas FABP3 knock-out (Fabp3-/-) mice did not. The number of phosphorylated α-Syn (Ser-129)-positive cells was significantly decreased in Fabp3-/- mouse brain compared to that in WT mice. The SNpc was unilaterally infected with AAV-GFP/FABP3 in Fabp3-/- mice to confirm the involvement of FABP3 in the development of α-Syn PFF toxicity. The number of tyrosine hydroxylase (TH)- and phosphorylated α-Syn (Ser-129)-positive cells following α-Syn PFF injection significantly decreased in Fabp3-/- mice and markedly increased by AAV-GFP/FABP3 infection. Finally, we confirmed that the novel FABP3 inhibitor MF1 significantly antagonized motor and cognitive impairments by preventing α-Syn spreading following α-Syn PFF injection. Overall, FABP3 enhances α-Syn spreading in the brain following α-Syn PFF injection, and the FABP3 ligand MF1 represents an attractive therapeutic candidate for α-synucleinopathy.


Asunto(s)
Encéfalo/metabolismo , Proteína 3 de Unión a Ácidos Grasos/metabolismo , alfa-Sinucleína/metabolismo , Animales , Encéfalo/patología , Cognición , Modelos Animales de Enfermedad , Proteína 3 de Unión a Ácidos Grasos/antagonistas & inhibidores , Proteína 3 de Unión a Ácidos Grasos/genética , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Neuronas/metabolismo , Fosforilación , Sinucleinopatías/etiología , Sinucleinopatías/metabolismo , Sinucleinopatías/patología , Sinucleinopatías/psicología , Tirosina 3-Monooxigenasa/metabolismo , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/efectos adversos
16.
Biochem Biophys Res Commun ; 531(1): 67-74, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-32035619

RESUMEN

Guanine-rich DNA and RNA can form a four-stranded structure, termed G-quadruplexes (G4s) in vitro as well as in cells. The formation of G4 is implicated in many physiological events, such as gene transcription, translation, and epigenetics. However, the presence of G4 has not been revealed in the brain. Here, we demonstrate the localization of G4 in the mouse brain by immunohistochemical analysis. In cultured mouse forebrain neurons, numerous punctate G4 foci were observed in nuclei as well as in cytoplasmic areas, including axons, dendrites, and postsynapses. Interestingly, the G4 foci in nuclei show more marked co-localizations with the bright spots of DAPI-positive heterochromatin clusters in mature neurons compared to immature ones. In slices from adult mouse brain, the G4 foci were distributed throughout the brain but were particularly prominent in the hippocampus, olfactory bulb, and cerebellum. In the hippocampus, G4 foci were strongly expressed in neurons and weakly in astrocytes. Consistent with the results in cultured neurons, the nuclear G4 foci were co-localized with heterochromatin in calbindin-positive mature granule cells but less in doublecortin-positive neuronal progenitor cells in the dentate gyrus. Electron microscopic immunolabeling revealed G4 foci on nucleolus-associated chromosomal domains (NADs) and cytoplasm in the adult mouse hippocampal CA1 region. These observations suggest potentially critical roles of G4 in neuronal developmental stages through regulating chromatin structures and cytoplasmic metabolism of RNA.


Asunto(s)
Química Encefálica , Encéfalo/citología , G-Cuádruplex , Neuronas/citología , Animales , Encéfalo/ultraestructura , Células Cultivadas , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/ultraestructura
17.
Neuropharmacology ; 168: 107975, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31991146

RESUMEN

Autism spectrum disorders (ASDs) constitute a neurodevelopmental disorder characterized by social deficits, repetitive behaviors, and learning disability. Oxidative stress and mitochondrial dysfunction are associated with ASD brain pathology. Here, we used oxidative stress in prenatal valproic acid (VPA)-exposed rats as an ASD model. After maternal VPA exposure (600 mg/kg, p.o.) on embryonic day (E) 12.5, temporal analyses of oxidative stress in the brain using an anti-4-hydroxy-2-nonenal antibody revealed that oxidative stress was increased in the hippocampus after birth. This was accompanied by aberrant enzymatic activity in the mitochondrial electron transport chain and reduced adenosine triphosphate (ATP) levels in the hippocampus. VPA-exposed rats exhibited impaired spatial reference and object recognition memory alongside impaired social behaviors and repetitive behaviors. ASD-like behaviors including learning and memory were rescued by chronic oral administration of 5-aminolevulinic acid (5-ALA; 30 mg/kg/day) and intranasal administration of oxytocin (OXT; 12 µg/kg/day), a neuropeptide that improves social behavior in ASD patients. 5-ALA but not OXT treatment ameliorated oxidative stress and mitochondrial dysfunction in the hippocampus of VPA-exposed rats. Fewer parvalbumin-positive interneurons were observed in VPA-exposed rats. Both 5-ALA and OXT treatments augmented the number of parvalbumin-positive interneurons. Collectively, our results indicate that oral 5-ALA administration ameliorated oxidative stress and mitochondrial dysfunction, suggesting that 5-ALA administration improves ASD-like neuropathology and behaviors via mechanisms different to those of OXT.


Asunto(s)
Ácido Aminolevulínico/uso terapéutico , Anticonvulsivantes/toxicidad , Trastorno Autístico/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Ácido Valproico/toxicidad , Ácido Aminolevulínico/farmacología , Animales , Trastorno Autístico/inducido químicamente , Trastorno Autístico/metabolismo , Relación Dosis-Respuesta a Droga , Reposicionamiento de Medicamentos/métodos , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Femenino , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Estrés Oxidativo/fisiología , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Ratas Sprague-Dawley
18.
Biochem Biophys Res Commun ; 531(1): 51-55, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31980177

RESUMEN

Genomic regions with guanine (G)-rich sequences make non-Watson-Crick base pairs, which result in the formation of unique nucleic acid structures called G-quadruplexes (G4s) in cells. Studies have suggested that abnormal G4s are involved in neurological diseases. For example, the formation of G4s caused by expansion of G-rich sequences is implicated in C9orf72-mediated amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD), and fragile X-related tremor/ataxia syndrome (FXTAS). In addition, the disruption and/or mutation of G4 binding proteins (G4BPs), such as heterogeneous nuclear ribonucleoproteins (hnRNPs) and DNA/RNA helicases, is related to neurological diseases. For instance, mutations in a G4BP called ATRX lead to a neurodevelopmental disorder, ATR-X syndrome, which is associated with intellectual disability. We found that porphyrins are potential candidate drugs for treating ATR-X syndrome through their G4 binding ability. Importantly, intracellular porphyrins are produced from 5-aminolevulinic acid (5-ALA) in vivo. Oral administration of 5-ALA improved cognitive dysfunction in an ATR-X syndrome model mouse, and language ability in an ATR-X syndrome patient. In this review, we suggest a novel therapeutic strategy targeting G4s using porphyrins in neurological diseases.


Asunto(s)
G-Cuádruplex/efectos de los fármacos , Discapacidad Intelectual Ligada al Cromosoma X/tratamiento farmacológico , Porfirinas/farmacología , Talasemia alfa/tratamiento farmacológico , Animales , Descubrimiento de Drogas/métodos , Humanos , Discapacidad Intelectual Ligada al Cromosoma X/genética , Terapia Molecular Dirigida/métodos , Porfirinas/química , Talasemia alfa/genética
19.
Nihon Yakurigaku Zasshi ; 154(6): 294-300, 2019.
Artículo en Japonés | MEDLINE | ID: mdl-31787679

RESUMEN

The most common form of DNA is a right-handed helix, the B-form DNA. DNA can also adopt a variety of alternative conformations, termed non-B-form DNA secondary structures, including the G-quadruplex (G4). Furthermore, non-canonical RNA G4 secondary structures are also observed. Recent bioinformatics analysis revealed genomic positions of G4. In addition, G4 formation may be associated with various biological functions, including DNA replication, transcription, epigenetic modification, and RNA metabolism. In this review, we focus on G4 structures in neuronal functions, which may have important roles reveal mechanisms underlying neurological disorders. In addition, we discuss the potential of G4s as a therapeutic target for neurological diseases.


Asunto(s)
G-Cuádruplex , Terapia Molecular Dirigida , Enfermedades del Sistema Nervioso/tratamiento farmacológico , ADN , Humanos , Conformación de Ácido Nucleico , ARN
20.
J Pharmacol Sci ; 140(3): 263-272, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31474557

RESUMEN

Atypical antipsychotics improve positive and negative symptoms but are not effective for treating cognitive impairments in patients with schizophrenia. We previously reported that cognitive impairments in neonatal ventral hippocampus (NVH)-lesioned rats show resistance to atypical antipsychotics risperidone and are associated with reduced calcium/calmodulin-dependent protein kinase II (CaMKII) and protein kinase C (PKC) signaling in memory-related regions. The cognitive enhancer ST101 (spiro[imi-dazo[1,2-a]pyridine-3,2-indan]-2(3H)-one) stimulates CaMKII activity in the hippocampus and medial prefrontal cortex (mPFC). We thus tested ST101 on cognitive impairments in NVH-lesioned rats. Chronic ST101 administration (0.1 and/or 0.5 mg/kg, p.o.) significantly improved deficits in prepulse inhibition (PPI), social interaction, and cognitive function in NVH-lesioned rats. ST101 administration (0.5 mg/kg, p.o.) significantly restored the decreased CaMKII autophosphorylation (Thr-286) in the mPFC and hippocampal CA1 regions of NVH-lesioned rats when assessed by immunohistochemistry. Chronic ST101 administration (0.1 mg/kg, p.o.) improved the decline in phosphorylation levels of CaMKII (Thr-286), PKCα (Ser-657), α-amino-3-hydroxy-5-methyl-4-isoxazol- propionic acid (AMPA)-type glutamate receptor subunit 1 (GluA1: Ser-831), and N-methyl-d-aspartate (NMDA) receptor subunit 1 (GluN1: Ser-896) in the mPFC and hippocampal CA1 regions. Taken together, these results suggest that ST101 improves schizophrenia-like behaviors and cognitive impairment by enhancing CaMKII/PKCα signaling in the mPFC and hippocampus in NVH-lesioned rats.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Trastornos del Conocimiento/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Indanos/farmacología , Proteína Quinasa C-alfa/metabolismo , Esquizofrenia/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Compuestos de Espiro/farmacología , Animales , Animales Recién Nacidos , Antipsicóticos/farmacología , Cognición/efectos de los fármacos , Trastornos del Conocimiento/metabolismo , Femenino , Hipocampo/metabolismo , Masculino , Memoria/efectos de los fármacos , Fosforilación/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Risperidona/farmacología , Esquizofrenia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA