Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38950903

RESUMO

In higher eukaryotes, tRNA methyltransferase 10A (TRMT10A) is responsible for N1-methylguanosine modification at position nine of various cytoplasmic tRNAs. Pathogenic mutations in TRMT10A cause intellectual disability, microcephaly, diabetes, and short stature in humans, and generate cytotoxic tRNA fragments in cultured cells; however, it is not clear how TRMT10A supports codon translation or brain functions. Here, we generated Trmt10a null mice and showed that tRNAGln(CUG) and initiator methionine tRNA levels were universally decreased in various tissues; the same was true in a human cell line lacking TRMT10A. Ribosome profiling of mouse brain revealed that dysfunction of TRMT10A causes ribosome slowdown at the Gln(CAG) codon and increases translation of Atf4 due to higher frequency of leaky scanning of its upstream open reading frames. Broadly speaking, translation of a subset of mRNAs, especially those for neuronal structures, is perturbed in the mutant brain. Despite not showing discernable defects in the pancreas, liver, or kidney, Trmt10a null mice showed lower body weight and smaller hippocampal postsynaptic densities, which is associated with defective synaptic plasticity and memory. Taken together, our study provides mechanistic insight into the roles of TRMT10A in the brain, and exemplifies the importance of universal tRNA modification during translation of specific codons.

2.
J Biol Chem ; 300(4): 107138, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38447794

RESUMO

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.


Assuntos
Ataxia Cerebelar , Expansão das Repetições de DNA , Doenças do Sistema Nervoso Periférico , Proteína de Replicação C , Doenças Vestibulares , Humanos , Ataxia Cerebelar/genética , Ataxia Cerebelar/metabolismo , Quadruplex G , Repetições de Microssatélites , Polimorfismo Genético , Proteína de Replicação C/genética , Proteína de Replicação C/metabolismo , Proteína de Replicação C/química , RNA/química , RNA/genética , RNA/metabolismo , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças Vestibulares/genética , Doenças Vestibulares/metabolismo
3.
Nucleic Acids Res ; 50(14): 8143-8153, 2022 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-35801908

RESUMO

Tandem repeats of guanine-rich sequences in RNA often form thermodynamically stable four-stranded RNA structures. Such RNA G-quadruplexes have long been considered to be linked to essential biological processes, yet their physiological significance in cells remains unclear. Here, we report a approach that permits the detection of RNA G-quadruplex structures that modulate protein translation in mammalian cells. The approach combines antibody arrays and RGB-1, a small molecule that selectively stabilizes RNA G-quadruplex structures. Analysis of the protein and mRNA products of 84 cancer-related human genes identified Nectin-4 and CapG as G-quadruplex-controlled genes whose mRNAs harbor non-canonical G-quadruplex structures on their 5'UTR region. Further investigations revealed that the RNA G-quadruplex of CapG exhibits a structural polymorphism, suggesting a possible mechanism that ensures the translation repression in a KCl concentration range of 25-100 mM. The approach described in the present study sets the stage for further discoveries of RNA G-quadruplexes.


Assuntos
Quadruplex G , Regiões 5' não Traduzidas , Animais , Guanina/química , Humanos , Mamíferos/genética , Biossíntese de Proteínas , RNA Mensageiro/metabolismo
4.
J Neurosci ; 39(38): 7551-7563, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31371425

RESUMO

Mental disorders are caused by genetic and environmental factors. We here show that deficiency of an isoform of dopamine D2 receptor (D2R), D2LR, causes stress vulnerability in mouse. This occurs through dysfunction of serotonin [5-hydroxytryptamine (5-HT)] 1A receptor (5-HT1AR) on serotonergic neurons in the mouse brain. Exposure to forced swim stress significantly increased anxiety- and depressive-like behaviors in D2LR knock-out (KO) male mice compared with wild-type mice. Treatment with 8-OH-DPAT, a 5-HT1AR agonist, failed to alleviate the stress-induced behaviors in D2LR-KO mice. In forced swim-stressed D2LR-KO mice, 5-HT efflux in the medial prefrontal cortex was elevated and the expression of genes related to 5-HT levels was upregulated by the transcription factor PET1 in the dorsal raphe nucleus. Notably, D2LR formed a heteromer with 5-HT1AR in serotonergic neurons, thereby suppressing 5-HT1AR-activated G-protein-activated inwardly rectifying potassium conductance in D2LR-KO serotonergic neurons. Finally, D2LR overexpression in serotonergic neurons in the dorsal raphe nucleus alleviated stress vulnerability observed in D2LR-KO mice. Together, we conclude that disruption of the negative feedback regulation by the D2LR/5-HT1A heteromer causes stress vulnerability.SIGNIFICANCE STATEMENT Etiologies of mental disorders are multifactorial, e.g., interactions between genetic and environmental factors. In this study, using a mouse model, we showed that genetic depletion of an isoform of dopamine D2 receptor, D2LR, causes stress vulnerability associated with dysfunction of serotonin 1A receptor, 5-HT1AR in serotonergic neurons. The D2LR/5-HT1AR inhibitory G-protein-coupled heteromer may function as a negative feedback regulator to suppress psychosocial stress.


Assuntos
Encéfalo/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Receptores de Dopamina D2/deficiência , Neurônios Serotoninérgicos/metabolismo , Estresse Psicológico/metabolismo , Animais , Masculino , Camundongos , Camundongos Knockout
5.
Biochem Biophys Res Commun ; 531(1): 51-55, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31980177

RESUMO

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.


Assuntos
Quadruplex G/efeitos dos fármacos , Deficiência Intelectual Ligada ao Cromossomo X/tratamento farmacológico , Porfirinas/farmacologia , Talassemia alfa/tratamento farmacológico , Animais , Descoberta de Drogas/métodos , Humanos , Deficiência Intelectual Ligada ao Cromossomo X/genética , Terapia de Alvo Molecular/métodos , Porfirinas/química , Talassemia alfa/genética
6.
Biochem Biophys Res Commun ; 531(1): 67-74, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-32035619

RESUMO

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.


Assuntos
Química Encefálica , Encéfalo/citologia , Quadruplex G , Neurônios/citologia , Animais , Encéfalo/ultraestrutura , Células Cultivadas , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/ultraestrutura
7.
Int J Mol Sci ; 21(15)2020 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-32752296

RESUMO

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.


Assuntos
Anticonvulsivantes/farmacologia , Benzodiazepinas/farmacologia , Proteína 3 Ligante de Ácido Graxo/metabolismo , Receptores de GABA-A/metabolismo , Estado Epiléptico/tratamento farmacológico , Animais , Anticonvulsivantes/química , Anticonvulsivantes/metabolismo , Benzodiazepinas/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Diazepam/metabolismo , Diazepam/farmacologia , Flumazenil/metabolismo , Flumazenil/farmacologia , Ligantes , Masculino , Camundongos Endogâmicos ICR , Pentilenotetrazol/metabolismo , Pentilenotetrazol/farmacologia , Convulsões/tratamento farmacológico , Convulsões/metabolismo , Estado Epiléptico/metabolismo
8.
Int J Mol Sci ; 21(6)2020 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-32210174

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Proteína 3 Ligante de Ácido Graxo/metabolismo , alfa-Sinucleína/metabolismo , Animais , Encéfalo/patologia , Cognição , Modelos Animais de Doenças , Proteína 3 Ligante de Ácido Graxo/antagonistas & inibidores , Proteína 3 Ligante de Ácido Graxo/genética , Imunofluorescência , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Fosforilação , Sinucleinopatias/etiologia , Sinucleinopatias/metabolismo , Sinucleinopatias/patologia , Sinucleinopatias/psicologia , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/administração & dosagem , alfa-Sinucleína/efeitos adversos
9.
J Pharmacol Sci ; 139(2): 51-58, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30600144

RESUMO

Low-threshold Ca2+ spikes are mediated by T-type Ca2+ channels, which have electrophysiological properties of fast inactivation and slow deactivation kinetics. A low membrane potential of approximately -60 mV is sufficient to trigger channel opening. We recently introduced a novel T-type Ca2+ channel enhancer that improves cognition and inhibits amyloid beta aggregation in an Alzheimer's disease (AD) mouse model. The enhancer stimulates ACh release, Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity, and neurogenesis in the hippocampus. Then, we discuss how T-type Ca2+ channel enhancer improves cognition and impaired neurogenesis and how CaMKII signaling in neurodegenerative diseases reduces amyloid beta aggregation. We provide a perspective of the potential AD therapies to target CaMKII signaling. In this context, we overview our attempts leading to the development of a T-type Ca2+ channel enhancer as cognitive enhancer, the action of which has been associated with CaMKII and presumably proteasome activity.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Canais de Cálcio Tipo T/fisiologia , Imidazóis/uso terapêutico , Compostos de Espiro/uso terapêutico , Acetilcolina/metabolismo , Doença de Alzheimer/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Hipocampo/fisiologia , Humanos , Imidazóis/farmacologia , Neurogênese , Complexo de Endopeptidases do Proteassoma/metabolismo , Compostos de Espiro/farmacologia
10.
Int J Mol Sci ; 20(12)2019 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-31200506

RESUMO

The most common form of DNA is a right-handed helix or the B-form DNA. DNA can also adopt a variety of alternative conformations, non-B-form DNA secondary structures, including the DNA G-quadruplex (DNA-G4). Furthermore, besides stem-loops that yield A-form double-stranded RNA, non-canonical RNA G-quadruplex (RNA-G4) secondary structures are also observed. Recent bioinformatics analysis of the whole-genome and transcriptome obtained using G-quadruplex-specific antibodies and ligands, revealed genomic positions of G-quadruplexes. In addition, accumulating evidence pointed to the existence of these structures under physiologically- and pathologically-relevant conditions, with functional roles in vivo. In this review, we focused on DNA-G4 and RNA-G4, which may have important roles in neuronal function, and reveal mechanisms underlying neurological disorders related to synaptic dysfunction. In addition, we mention the potential of G-quadruplexes as therapeutic targets for neurological diseases.


Assuntos
Quadruplex G , Doenças do Sistema Nervoso/genética , Animais , Terapia Genética/métodos , Humanos , Doenças do Sistema Nervoso/terapia
11.
Cell Physiol Biochem ; 51(3): 1461-1479, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30485861

RESUMO

BACKGROUND/AIMS: The migration of mesenchymal cells is a fundamental cellular process that has been implicated in many pathophysiological conditions and is induced by chemoattractants such as platelet-derived growth factors (PDGFs). However, the regulatory mechanisms shaping this migration remain to be elucidated. METHODS: Here, we prepared mouse skin fibroblasts inactivated for different PDGF receptor genes and systematically measured their chemotactic responses within a gradient of different chemoattractants. RESULTS: We found that PDGFRαß and PDGFRßß dimers were strong inducers of random and directionally-persistent migration, respectively, that was sustained for up to 24 h. MAPK and PI3K were necessary to mediate random and directional migration, respectively. Directional migration was accompanied by abundant ventral stress fiber formation and consistent cell shape with less frequent formation of branch-like processes. CONCLUSION: This is the first systematic study that characterized the chemotaxis mediated by three-different types of PDGFR dimers in mesenchymal cell migration. Our data demonstrate that PDGFR dimer formation is the critical step to determine the specific mode of fibroblast chemotaxis, while the accompanying cytoskeletal remodeling might contribute to migration persistence.


Assuntos
Movimento Celular , Fibroblastos/citologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Quimiotaxia , Fibroblastos/metabolismo , Técnicas de Inativação de Genes , Camundongos , Multimerização Proteica , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais , Pele/citologia , Pele/metabolismo
12.
Int J Mol Sci ; 19(9)2018 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-30231518

RESUMO

α-thalassemia X-linked intellectual disability (ATR-X) syndrome is caused by mutations in ATRX. An ATR-X model mouse lacking Atrx exon 2 displays phenotypes that resemble symptoms in the human intellectual disability: cognitive defects and abnormal dendritic spine formation. We herein target activation of sigma-1 receptor (Sig-1R) that can induce potent neuroprotective and neuroregenerative effects by promoting the activity of neurotrophic factors, such as brain-derived neurotrophic factor (BDNF). We demonstrated that treatment with SA4503, a potent activator of Sig-1R, reverses axonal development and dendritic spine abnormalities in cultured cortical neurons from ATR-X model mice. Moreover, the SA4503 treatment rescued cognitive deficits exhibited by the ATR-X model mice. We further found that significant decreases in the BDNF-protein level in the medial prefrontal cortex of ATR-X model mice were recovered with treatment of SA4503. These results indicate that the rescue of dendritic spine abnormalities through the activation of Sig-1R has a potential for post-diagnostic therapy in ATR-X syndrome.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Deficiência Intelectual Ligada ao Cromossomo X/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Piperazinas/uso terapêutico , Receptores sigma/metabolismo , Talassemia alfa/tratamento farmacológico , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/fisiopatologia , Modelos Animais de Doenças , Ligantes , Masculino , Deficiência Intelectual Ligada ao Cromossomo X/complicações , Deficiência Intelectual Ligada ao Cromossomo X/fisiopatologia , Camundongos Endogâmicos C57BL , Talassemia alfa/complicações , Talassemia alfa/fisiopatologia , Receptor Sigma-1
13.
J Pharmacol Sci ; 2017 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-29107444

RESUMO

Dopamine receptor family proteins include seven transmembrane and trimeric GTP-binding protein-coupled receptors (GPCRs). Among them, the dopamine D2 receptor (D2R) is most extensively studied. All clinically used antipsychotic drugs serve as D2R antagonists in the mesolimbic dopamine system, and their ability to block D2R signaling is positively correlated with antipsychotic efficiency. Human genetic studies also show a significant association of DRD2 polymorphisms with disorders including schizophrenia and Parkinson's disease. D2R exists as two alternatively spliced isoforms, the long isoform (D2LR) and the short isoform (D2SR), which differ in a 29-amino acid (AA) insert in the third cytoplasmic loop. Importantly, previous reports demonstrate functional diversity between the two isoforms in humans. In this review, we focus on binding proteins that specifically interact with the D2LR 29AA insert. We discuss how D2R activities are mediated not only by heterotrimeric G proteins but by D2LR-interacting proteins, which in part regulate diverse D2R activities.

14.
Int J Mol Sci ; 19(1)2017 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-29271887

RESUMO

Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII), a multifunctional serine (Ser)/threonine (Thr) protein kinase, regulates diverse activities related to Ca2+-mediated neuronal plasticity in the brain, including synaptic activity and gene expression. Among its regulators, protein phosphatase-1 (PP1), a Ser/Thr phosphatase, appears to be critical in controlling CaMKII-dependent neuronal signaling. In postsynaptic densities (PSDs), CaMKII is required for hippocampal long-term potentiation (LTP), a cellular process correlated with learning and memory. In response to Ca2+ elevation during hippocampal LTP induction, CaMKIIα, an isoform that translocates from the cytosol to PSDs, is activated through autophosphorylation at Thr286, generating autonomous kinase activity and a prolonged Ca2+/CaM-bound state. Moreover, PP1 inhibition enhances Thr286 autophosphorylation of CaMKIIα during LTP induction. By contrast, CaMKII nuclear import is regulated by Ser332 phosphorylation state. CaMKIIδ3, a nuclear isoform, is dephosphorylated at Ser332 by PP1, promoting its nuclear translocation, where it regulates transcription. In this review, we summarize physio-pathological roles of CaMKII/PP1 signaling in neurons. CaMKII and PP1 crosstalk and regulation of gene expression is important for neuronal plasticity as well as survival and/or differentiation.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Proteína Fosfatase 1/metabolismo , Transdução de Sinais , Transporte Ativo do Núcleo Celular , Animais , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Humanos , Potenciação de Longa Duração , Plasticidade Neuronal
15.
J Biol Chem ; 290(35): 21663-75, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26163515

RESUMO

We have reported previously that dopamine D2 receptor stimulation activates calcium/calmodulin-dependent protein kinase II (CaMKII) δ3, a CaMKII nuclear isoform, increasing BDNF gene expression. However, the mechanisms underlying that activity remained unclear. Here we report that CaMKIIδ3 is dephosphorylated at Ser(332) by protein phosphatase 1 (PP1), promoting CaMKIIδ3 nuclear translocation. Neuro-2a cells transfected with CaMKIIδ3 showed cytoplasmic and nuclear staining, but the staining was predominantly nuclear when CaMKIIδ3 was coexpressed with PP1. Indeed, PP1 and CaMKIIδ3 coexpression significantly increased nuclear CaMKII activity and enhanced BDNF expression. In support of this idea, chronic administration of the dopamine D2 receptor partial agonist aripiprazole increased PP1 activity and promoted nuclear CaMKIIδ3 translocation and BDNF expression in the rat brain substantia nigra. Moreover, aripiprazole treatment enhanced neurite extension and inhibited cell death in cultured dopaminergic neurons, effects blocked by PP1γ knockdown. Taken together, nuclear translocation of CaMKIIδ3 following dephosphorylation at Ser(332) by PP1 likely accounts for BDNF expression and subsequent neurite extension and survival of dopaminergic neurons.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Núcleo Celular/metabolismo , Neurônios Dopaminérgicos/metabolismo , Proteína Fosfatase 1/metabolismo , Sequência de Aminoácidos , Animais , Aripiprazol/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/química , Núcleo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Neurônios Dopaminérgicos/efeitos dos fármacos , Masculino , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Fosfopeptídeos/metabolismo , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/metabolismo , Serina/metabolismo , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo
16.
Chembiochem ; 17(10): 928-35, 2016 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-26991472

RESUMO

ATR-X (α-thalassemia/mental retardation X-linked) syndrome is caused by mutations in chromatin remodeler ATRX. ATRX can bind the variable number of tandem repeats (VNTR) sequence in the promoter region of the α-globin gene cluster. The VNTR sequence, which contains the potential G-quadruplex-forming sequence CGC(GGGGCGGGG)n , is involved in the downregulation of α-globin expression. We investigated G-quadruplex and i-motif formation in single-stranded DNA and long double-stranded DNA. The promoter region without the VNTR sequence showed approximately twofold higher luciferase activity than the promoter region harboring the VNTR sequence. G-quadruplex stabilizers hemin and TMPyP4 reduced the luciferase activity, whereas expression of ATRX led to a recovery in reporter activity. Our results demonstrate that stable G-quadruplex formation by the VNTR sequence downregulates the expression of α-globin genes and that ATRX might bind to and resolve the G-quadruplex.


Assuntos
DNA Helicases/metabolismo , Quadruplex G , Regulação da Expressão Gênica/genética , Repetições Minissatélites/genética , alfa-Globinas/genética , Sequência de Bases , Dicroísmo Circular , Regulação para Baixo , Genes Reporter , Hemina/química , Hemina/metabolismo , Humanos , Microscopia de Força Atômica , Porfirinas/química , Porfirinas/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , alfa-Globinas/metabolismo
18.
J Biol Chem ; 289(27): 18957-65, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24855640

RESUMO

α-Synuclein (αSyn) accumulation in dopaminergic (DA) neurons is partly regulated by long-chain polyunsaturated fatty acids. We found that fatty acid-binding protein 3 (FABP3, H-FABP), a factor critical for arachidonic acid (AA) transport and metabolism in brain, is highly expressed in DA neurons. Fabp3 knock-out (Fabp3(-/-)) mice were resistant to 1-methyl-1,2,3,6-tetrahydropiridine-induced DA neurodegeneration in the substantia nigra pars compacta and showed improved motor function. Interestingly, FABP3 interacted with αSyn in the substantia nigra pars compacta, and αSyn accumulation following 1-methyl-1,2,3,6-tetrahydropiridine treatment was attenuated in Fabp3(-/-) compared with wild-type mice. We confirmed that FABP3 overexpression aggravates AA-induced αSyn oligomerization and promotes cell death in PC12 cells, whereas overexpression of a mutant form of FABP3 lacking fatty-acid binding capacity did not. Taken together, αSyn oligomerization in DA neurons is likely aggravated by AA through FABP3 in Parkinson disease pathology.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Neurônios Dopaminérgicos/efeitos dos fármacos , Proteínas de Ligação a Ácido Graxo/metabolismo , Intoxicação por MPTP/metabolismo , Neurotoxinas/toxicidade , Multimerização Proteica , alfa-Sinucleína/química , Animais , Ácido Araquidônico/farmacologia , Morte Celular/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Proteína 3 Ligante de Ácido Graxo , Proteínas de Ligação a Ácido Graxo/deficiência , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Intoxicação por MPTP/patologia , Intoxicação por MPTP/fisiopatologia , Camundongos , Atividade Motora/efeitos dos fármacos , Neostriado/citologia , Células PC12 , Ligação Proteica , Multimerização Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína , Ratos , Substância Negra/citologia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
19.
Biochim Biophys Acta ; 1840(12): 3320-34, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25175561

RESUMO

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a disease caused by motor neuron degeneration. Recently, a novel SIGMAR1 gene variant (p.E102Q) was discovered in some familial ALS patients. METHODS: We address mechanisms underlying neurodegeneration caused by the mutation using Neuro2A cells overexpressing σ1R(E102Q), a protein of a SIGMAR1 gene variant (p.E102Q) and evaluate potential amelioration by ATP production via methyl pyruvate (MP) treatment. RESULTS: σ1R(E102Q) overexpression promoted dissociation of the protein from the endoplasmic reticulum (ER) membrane and cytoplasmic aggregation, which in turn impaired mitochondrial ATP production and proteasome activity. Under ER stress conditions, overexpression of wild-type σ1R suppressed ER stress-induced mitochondrial injury, whereas σ1R(E102Q) overexpression aggravated mitochondrial damage and induced autophagic cell death. Moreover, σ1R(E102Q)-overexpressing cells showed aberrant extra-nuclear localization of the TAR DNA-binding protein (TDP-43), a condition exacerbated by ER stress. Treatment of cells with the mitochondrial Ca(2+) transporter inhibitor Ru360 mimicked the effects of σ1R(E102Q) overexpression, indicating that aberrant σ1R-mediated mitochondrial Ca(2+) transport likely underlies TDP-43 extra-nuclear localization, segregation in inclusion bodies, and ubiquitination. Finally, enhanced ATP production promoted by methyl pyruvate (MP) treatment rescued proteasome impairment and TDP-43 extra-nuclear localization caused by σ1R(E102Q) overexpression. CONCLUSIONS: Our observations suggest that neurodegeneration seen in some forms of ALS are due in part to aberrant mitochondrial ATP production and proteasome activity as well as TDP-43 mislocalization resulting from the SIGMAR1 mutation. GENERAL SIGNIFICANCE: ATP supplementation by MP represents a potential therapeutic strategy to treat ALS caused by SIGMAR1 mutation.

20.
J Biol Chem ; 287(28): 23318-31, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22619170

RESUMO

The σ1 receptor (σ(1)R) regulates endoplasmic reticulum (ER)/mitochondrial interorganellar Ca(2+) mobilization through the inositol 1,4,5-trisphosphate receptor (IP(3)R). Here, we observed that expression of a novel splice variant of σ(1)R, termed short form σ(1)R (σ(1)SR), has a detrimental effect on mitochondrial energy production and cell survival. σ(1)SR mRNA lacks 47 ribonucleotides encoding exon 2, resulting in a frameshift and formation of a truncated receptor. σ(1)SR localizes primarily in the ER at perinuclear regions and forms a complex with σ(1)R but not with IP(3)R in the mitochondrion-associated ER membrane. Overexpression of both σ(1)R and the truncated isoform promotes mitochondrial elongation with increased ER mitochondrial contact surface. σ(1)R overexpression increases the efficiency of mitochondrial Ca(2+) uptake in response to IP(3)R-driven stimuli, whereas σ(1)SR overexpression reduces it. Most importantly, σ(1)R promotes ATP production via increased mitochondrial Ca(2+) uptake, promoting cell survival in the presence of ER stress. By contrast, σ(1)SR suppresses ATP production following ER stress, enhancing cell death. Taken together, the newly identified σ(1)SR isoform interferes with σ(1)R function relevant to mitochondrial energy production under ER stress conditions, promoting cellular apoptosis.


Assuntos
Apoptose , Retículo Endoplasmático/metabolismo , Mitocôndrias/metabolismo , Receptores sigma/metabolismo , Trifosfato de Adenosina/metabolismo , Processamento Alternativo , Animais , Encéfalo/metabolismo , Cálcio/metabolismo , Linhagem Celular Tumoral , Immunoblotting , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Dados de Sequência Molecular , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , Receptores sigma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA