Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Elife ; 122024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38529532

RESUMO

Increased levels of lactate, an end-product of glycolysis, have been proposed as a potential surrogate marker for metabolic changes during neuronal excitation. These changes in lactate levels can result in decreased brain pH, which has been implicated in patients with various neuropsychiatric disorders. We previously demonstrated that such alterations are commonly observed in five mouse models of schizophrenia, bipolar disorder, and autism, suggesting a shared endophenotype among these disorders rather than mere artifacts due to medications or agonal state. However, there is still limited research on this phenomenon in animal models, leaving its generality across other disease animal models uncertain. Moreover, the association between changes in brain lactate levels and specific behavioral abnormalities remains unclear. To address these gaps, the International Brain pH Project Consortium investigated brain pH and lactate levels in 109 strains/conditions of 2294 animals with genetic and other experimental manipulations relevant to neuropsychiatric disorders. Systematic analysis revealed that decreased brain pH and increased lactate levels were common features observed in multiple models of depression, epilepsy, Alzheimer's disease, and some additional schizophrenia models. While certain autism models also exhibited decreased pH and increased lactate levels, others showed the opposite pattern, potentially reflecting subpopulations within the autism spectrum. Furthermore, utilizing large-scale behavioral test battery, a multivariate cross-validated prediction analysis demonstrated that poor working memory performance was predominantly associated with increased brain lactate levels. Importantly, this association was confirmed in an independent cohort of animal models. Collectively, these findings suggest that altered brain pH and lactate levels, which could be attributed to dysregulated excitation/inhibition balance, may serve as transdiagnostic endophenotypes of debilitating neuropsychiatric disorders characterized by cognitive impairment, irrespective of their beneficial or detrimental nature.


Assuntos
Disfunção Cognitiva , Endofenótipos , Animais , Camundongos , Humanos , Encéfalo/metabolismo , Disfunção Cognitiva/metabolismo , Modelos Animais de Doenças , Lactatos/metabolismo , Concentração de Íons de Hidrogênio
2.
Mol Biol Cell ; 34(11): ar110, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37585286

RESUMO

Alcadein α (Alcα) and amyloid-ß protein precursor (APP) are cargo receptors that associate vesicles with kinesin-1. These vesicles, which contain either Alcα or APP, transport various proteins/cargo molecules into axon nerve terminals. Here, we analyzed immune-isolated Alcα- and APP-containing vesicles of adult mouse brains with LC-MS/MS and identified proteins present in vesicles that contained either Alcα or APP. Among these proteins, Frizzled-5 (Fzd5), a Wnt receptor, was detected mainly in Alcα vesicles. Although colocalization ratios of Fzd5 with Alcα are low in the neurites of differentiating neurons by a low expression of Fzd5 in embryonic brains, the suppression of Alcα expression decreased the localization of Fzd5 in neurites of primary cultured neurons. Furthermore, Fzd5-EGFP expressed in primary cultured neurons was preferentially transported in axons with the transport velocities of Alcα vesicles. In synaptosomal fractions of adult-mice brains that express higher levels of Fzd5, the amount of Fzd5 and the phosphorylation level of calcium/calmodulin-dependent protein kinase-II were reduced in the Alcα-deficient mice. These results suggest that reduced transport of Fzd5 by Alcα-containing vesicles associated with kinesin-1 in axon terminals may impair the response to Wnt ligands in the noncanonical Ca2+-dependent signal transduction pathway at nerve terminals of mature neurons.


Assuntos
Transporte Axonal , Cinesinas , Animais , Camundongos , Precursor de Proteína beta-Amiloide/metabolismo , Transporte Axonal/fisiologia , Cromatografia Líquida , Cinesinas/metabolismo , Espectrometria de Massas em Tandem
3.
Behav Brain Res ; 452: 114590, 2023 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-37499910

RESUMO

Synaptic dysfunction underlies many neurodevelopmental disorders (NDDs). The membrane-associated mucin domain-containing glycosylphosphatidylinositol anchor proteins (MDGAs) regulate synaptic development by modulating neurexin-neuroligin complex formation. Since understanding the neurodevelopmental profile and the sex-based differences in the manifestation of the symptoms of NDDs is important for their early diagnosis, we tested a mouse model haploinsufficient for MDGA2 (MDGA2+/-) on a neurodevelopmental test battery, containing sensory, motor, and cognitive measures, as well as ultrasonic vocalizations. When male and female MDGA2+/- and wildtype (WT) C57BL/6 J mice were examined from 2 to 23 days of age using this test battery, genotype and sex differences in body weight, sensory-motor processes, and ultrasonic vocalizations were observed. The auditory startle reflex appeared earlier in the MDGA2+/- than in WT mice and the MDGA2+/- mice produced fewer ultrasonic vocalizations. The MDGA2+/- mice showed reduced locomotion and rearing than WT mice in the open field after 17 days of age and spent less time investigating a novel object than WT mice at 21 days of age. Female MDGA2+/- mice weighed less than WT females and showed lower grip strength, indicating a delay in sensory-motor development in MDGA2+/- mice, which appears to be more pronounced in females than males. The behavioural phenotypes resulting from MDGA2 haploinsufficiency suggests that it shows delayed development of motor behaviour, grip strength and exploratory behaviour, non-social phenotypes of NDDs.


Assuntos
Transtornos do Neurodesenvolvimento , Camundongos , Feminino , Masculino , Animais , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Proteínas de Membrana , Reflexo de Sobressalto , Moléculas de Adesão de Célula Nervosa/metabolismo , Proteínas Ligadas por GPI/metabolismo
4.
EMBO Mol Med ; 15(5): e17052, 2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-36994913

RESUMO

We propose a new therapeutic strategy for Alzheimer's disease (AD). Brain peptide p3-Alcß37 is generated from the neuronal protein alcadein ß through cleavage of γ-secretase, similar to the generation of amyloid ß (Aß) derived from Aß-protein precursor/APP. Neurotoxicity by Aß oligomers (Aßo) is the prime cause prior to the loss of brain function in AD. We found that p3-Alcß37 and its shorter peptide p3-Alcß9-19 enhanced the mitochondrial activity of neurons and protected neurons against Aßo-induced toxicity. This is due to the suppression of the Aßo-mediated excessive Ca2+ influx into neurons by p3-Alcß. Successful transfer of p3-Alcß9-19 into the brain following peripheral administration improved the mitochondrial viability in the brain of AD mice model, in which the mitochondrial activity is attenuated by increasing the neurotoxic human Aß42 burden, as revealed through brain PET imaging to monitor mitochondrial function. Because mitochondrial dysfunction is common in the brain of AD patients alongside increased Aß and reduced p3-Alcß37 levels, the administration of p3-Alcß9-19 may be a promising treatment for restoring, protecting, and promoting brain functions in patients with AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Camundongos , Animais , Humanos , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Neurônios/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo
5.
Neurosci Res ; 186: 3-9, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36244569

RESUMO

Ischemic stroke is one of the leading causes of death and disability worldwide. The inhibition of cerebral blood flow triggers intertwined pathological events, resulting in cell death and loss of brain function. Interestingly, animals pre-exposed to short-term ischemia can tolerate subsequent severe ischemia. This phenomenon is called ischemic tolerance and is also triggered by other noxious stimuli. However, whether short-term exposure to non-noxious stimuli can induce ischemic tolerance remains unknown. Recently, we found that pre-exposing mice to an enriched environment for 40 min is sufficient to facilitate cell survival after a subsequent stroke. The neuroprotective process depends on the neuronal activity soon before stroke, of which the activity-dependent transcription factor Npas4 is essential. Excessive Ca2+ influx triggers Npas4 expression in ischemic neurons, leading to the activation of neuroprotective programs. Pre-induction of Npas4 in the normal brain effectively supports cell survival after stroke. Furthermore, our study revealed that Npas4 regulates L-type voltage-gated Ca2+ channels through expression of the small Ras-like GTPase Gem in ischemic neurons. Ischemic tolerance is a good model for understanding how to promote neuroprotective mechanisms in the normal and injured brain. Here, we highlight activity-dependent ischemic tolerance and discuss its role in promoting neuroprotection against stroke.


Assuntos
Isquemia Encefálica , Fármacos Neuroprotetores , Acidente Vascular Cerebral , Camundongos , Animais , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Acidente Vascular Cerebral/metabolismo , Isquemia/metabolismo , Isquemia/patologia , Neuroproteção , Fármacos Neuroprotetores/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos
6.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1867(12): 159222, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35988872

RESUMO

N-Acyl-phosphatidylethanolamines (NAPEs), a minor class of membrane glycerophospholipids, accumulate along with their bioactive metabolites, N-acylethanolamines (NAEs) during ischemia. NAPEs can be formed through N-acylation of phosphatidylethanolamine by cytosolic phospholipase A2ε (cPLA2ε, also known as PLA2G4E) or members of the phospholipase A and acyltransferase (PLAAT) family. However, the enzyme responsible for the NAPE production in brain ischemia has not yet been clarified. Here, we investigated a possible role of cPLA2ε using cPLA2ε-deficient (Pla2g4e-/-) mice. As analyzed with brain homogenates of wild-type mice, the age dependency of Ca2+-dependent NAPE-forming activity showed a bell-shape pattern being the highest at the first week of postnatal life, and the activity was completely abolished in Pla2g4e-/- mice. However, liquid chromatography-tandem mass spectrometry revealed that the NAPE levels of normal brain were similar between wild-type and Pla2g4e-/- mice. In contrast, post-mortal accumulations of NAPEs and most species of NAEs were only observed in decapitated brains of wild-type mice. These results suggested that cPLA2ε is responsible for Ca2+-dependent formation of NAPEs in the brain as well as the accumulation of NAPEs and NAEs during ischemia, while other enzyme(s) appeared to be involved in the maintenance of basal NAPE levels.


Assuntos
Isquemia Encefálica , Fosfatidiletanolaminas , Aciltransferases/metabolismo , Animais , Isquemia Encefálica/genética , Modelos Animais de Doenças , Glicerofosfolipídeos , Camundongos , Fosfatidiletanolaminas/metabolismo , Fosfolipases A , Fosfolipases A2 Citosólicas , Espiperona/análogos & derivados
7.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34349016

RESUMO

Ischemic stroke, which results in loss of neurological function, initiates a complex cascade of pathological events in the brain, largely driven by excitotoxic Ca2+ influx in neurons. This leads to cortical spreading depolarization, which induces expression of genes involved in both neuronal death and survival; yet, the functions of these genes remain poorly understood. Here, we profiled gene expression changes that are common to ischemia (modeled by middle cerebral artery occlusion [MCAO]) and to experience-dependent activation (modeled by exposure to an enriched environment [EE]), which also induces Ca2+ transients that trigger transcriptional programs. We found that the activity-dependent transcription factor Npas4 was up-regulated under MCAO and EE conditions and that transient activation of cortical neurons in the healthy brain by the EE decreased cell death after stroke. Furthermore, both MCAO in vivo and oxygen-glucose deprivation in vitro revealed that Npas4 is necessary and sufficient for neuroprotection. We also found that this protection involves the inhibition of L-type voltage-gated Ca2+ channels (VGCCs). Next, our systematic search for Npas4-downstream genes identified Gem, which encodes a Ras-related small GTPase that mediates neuroprotective effects of Npas4. Gem suppresses the membrane localization of L-type VGCCs to inhibit excess Ca2+ influx, thereby protecting neurons from excitotoxic death after in vitro and in vivo ischemia. Collectively, our findings indicate that Gem expression via Npas4 is necessary and sufficient to promote neuroprotection in the injured brain. Importantly, Gem is also induced in human cerebral organoids cultured under an ischemic condition, revealing Gem as a new target for drug discovery.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , AVC Isquêmico/fisiopatologia , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neurônios/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Morte Celular , Células HEK293 , Humanos , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , AVC Isquêmico/genética , AVC Isquêmico/mortalidade , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/genética , Neurônios/patologia , Organoides
8.
Brain Res ; 1768: 147595, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34332964

RESUMO

The present study aimed to assess spinal tract formation in neurons originating from cervical (C7), brachial (C14), and thoracic (T4) regions, with the lumbar (LS2) region as a reference, in a chick embryo. For the assessment of the spinal tracts, we introduced a vector expressing human placental alkaline phosphatase into progenitor cells generated after neural tube closure and belonging to the above segments, using in ovo electroporation. The ascending axons took primarily similar paths: dorsal commissural, ventral commissural, and dorsal non-commissural paths, with some variance depending on their originating segments. Some populations of non-commissural neurons later extended their axons following a ventral path. The elongation rates of these axons are primarily constant and tended to increase over time; however, some variations depending on the originating segments were also observed. Some of the dorsally ascending axons entered into the developing cerebellum, and spinocerebellar neurons originating from T4 projected their axons into the cortex of the cerebellum differently from those from LS2. These results unveil an overall picture of early ascending spinal tract formation.


Assuntos
Fosfatase Alcalina/metabolismo , Isoenzimas/metabolismo , Medula Espinal/fisiologia , Coluna Vertebral/embriologia , Fosfatase Alcalina/fisiologia , Animais , Axônios/fisiologia , Encéfalo/embriologia , Encéfalo/fisiologia , Cerebelo/fisiologia , Embrião de Galinha , Eletroporação , Proteínas Ligadas por GPI/metabolismo , Proteínas Ligadas por GPI/fisiologia , Isoenzimas/fisiologia , Vias Neurais , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Medula Espinal/embriologia , Coluna Vertebral/metabolismo
10.
Neuropsychopharmacol Rep ; 40(4): 371-375, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33089673

RESUMO

AIM: Fear conditioning tests are intended to elucidate a subject's ability to associate a conditioned stimulus with an aversive, unconditioned stimulus, such as footshock. Among these tests, a paradigm related to precise cortical functions would be increasingly important in drug screening for disorders such as schizophrenia and dementia. Therefore, we established a new fear conditioning paradigm using a visual cue in mice. In addition, the validity of the test was evaluated using a genetically engineered mouse, heterozygous deficient in Mdga1 (Mdga1+/-), which is related to schizophrenia. RESULTS: Mice were given footshocks associated with a visual cue of moving gratings at training in 25-minute sessions. The mice showed the conditioned response of freezing behavior to the visual stimulus at testing 24 hours after the footshocks. In the test for validation, the Mdga1+/- deficient mice showed significantly less freezing than wild-type mice. CONCLUSION: The visually cued fear conditioning paradigm with moving gratings has been established, which is experimentally useful to evaluate animal cortical functions. The validity of the test was confirmed for Mdga1-deficient mice with possible deficiency in cortical functions.


Assuntos
Condicionamento Operante/fisiologia , Sinais (Psicologia) , Medo/fisiologia , Transtornos da Memória/fisiopatologia , Percepção de Movimento/fisiologia , Córtex Visual/fisiologia , Animais , Estimulação Elétrica/efeitos adversos , Medo/psicologia , Feminino , Transtornos da Memória/psicologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estimulação Luminosa/métodos
11.
J Biol Chem ; 295(28): 9650-9662, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-32467230

RESUMO

Alzheimer's disease (AD) is a very common neurodegenerative disorder, chiefly caused by increased production of neurotoxic ß-amyloid (Aß) peptide generated from proteolytic cleavage of ß-amyloid protein precursor (APP). Except for familial AD arising from mutations in the APP and presenilin (PSEN) genes, the molecular mechanisms regulating the amyloidogenic processing of APP are largely unclear. Alcadein α/calsyntenin1 (ALCα/CLSTN1) is a neuronal type I transmembrane protein that forms a complex with APP, mediated by the neuronal adaptor protein X11-like (X11L or MINT2). Formation of the ALCα-X11L-APP tripartite complex suppresses Aß generation in vitro, and X11L-deficient mice exhibit enhanced amyloidogenic processing of endogenous APP. However, the role of ALCα in APP metabolism in vivo remains unclear. Here, by generating ALCα-deficient mice and using immunohistochemistry, immunoblotting, and co-immunoprecipitation analyses, we verified the role of ALCα in the suppression of amyloidogenic processing of endogenous APP in vivo We observed that ALCα deficiency attenuates the association of X11L with APP, significantly enhances amyloidogenic ß-site cleavage of APP, especially in endosomes, and increases the generation of endogenous Aß in the brain. Furthermore, we noted amyloid plaque formation in the brains of human APP-transgenic mice in an ALCα-deficient background. These results unveil a potential role of ALCα in protecting cerebral neurons from Aß-dependent pathogenicity in AD.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Proteínas de Ligação ao Cálcio/deficiência , Complexos Multiproteicos/metabolismo , Processamento de Proteína Pós-Traducional , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Camundongos , Camundongos Knockout , Complexos Multiproteicos/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo
12.
Cell Death Dis ; 11(3): 166, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32127528

RESUMO

Maintenance of retinal ganglion cells (RGCs) activity is relied on axonal transport conveying materials required for their survival such as neurotrophic factors. Kinesin-1 undergoes anterograde transport in axons, and Alcadein α (Alcα; also called calsyntenin-1) is a major cargo adaptor protein that can drive kinesin-1 to transport vesicles containing Alcα. The long-term effects of Alcα-deficiency on retinal morphology and survival of RGCs during postnatal development were examined in Alcα knockout mice. At 1.5, 3, 6, and 15 months postnatal, the number of retrogradely labeled RGCs was determined in flat-mounted retinas of Alcα-deficient and wild-type mice. Retinal damage was assessed histologically by determining the retinal thickness. Intraocular pressure (IOP) was measured with a Tonolab tonometer. At 1.5 months postnatal, the number of retrogradely labeled RGCs was not different between wild-type and Alcα-deficient mice. However, at 3, 6, and 15 months postnatal, the number of RGCs was significantly lower in Alcα deficient mice than those of wild-type mice (143 ± 41.1 cells/mm2 vs. 208 ± 28.4 cells/mm2, respectively, at 3 months; P < 0.01). No differences were seen in retinal thickness or IOP between the two types of mice at any postnatal age. Alcα-deficient mice showed spontaneous loss of RGCs but no elevation in IOP. These mice mimic normal-tension glaucoma and will be useful for investigating the mechanism of neurodegeneration in this disorder and for developing treatments for RGC loss that does not involve changes in IOP.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Axônios/metabolismo , Retina/metabolismo , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Animais , Transporte Axonal/fisiologia , Axônios/patologia , Modelos Animais de Doenças , Pressão Intraocular/fisiologia , Cinesinas/deficiência , Cinesinas/metabolismo , Glaucoma de Baixa Tensão/metabolismo , Camundongos Knockout , Vesículas Transportadoras/metabolismo
13.
Neurosci Lett ; 716: 134677, 2020 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-31812551

RESUMO

MDGA1 (MAM domain-containing glycosylphosphatidylinositol anchor) has recently been linked to schizophrenia and bipolar disorder. Dysregulation of dopamine (DA) and serotonin (5-HT) systems has long been associated with schizophrenia and other neuropsychiatric disorders. Here, we measured prepulse inhibition (PPI) of the startle response and ex vivo tissue content of monoamines and their metabolites in the frontal cortex, striatum and hippocampus of Mdga1 homozygous (Mdga1-KO), Mdga1 heterozygous (Mdga1-HT) and wild-type (WT) male mice. We found that Mdga1-KO mice exhibited statistically significant impairment of PPI, and had higher levels of homovanillic acid in all three brain regions studied compared with Mdga1-HT and WT mice (P < 0.05), while levels of norepinephrine, DA and its metabolites 3,4-dihydroxyphenylacetic acid and 3-methoxytyramine remained unchanged. Mdga1-KO mice also had a lower 5-hydroxyindoleacetic acid level in the striatum (P < 0.05) compared with WT mice. 5-HT levels remained unchanged with the exception of a significant increase in the level in the cortex. These data are the first evidence suggesting that MDGA1 deficiency leads to a pronounced deficit in PPI and plays an important role in perturbation of DA and 5-HT metabolism in mouse brain; such changes may contribute to a range of neuropsychiatric disorders.


Assuntos
Encéfalo/metabolismo , Dopamina/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Inibição Pré-Pulso/fisiologia , Serotonina/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reflexo de Sobressalto/fisiologia
14.
Alzheimers Dement (N Y) ; 5: 740-750, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31754625

RESUMO

INTRODUCTION: Neuronal p3-Alcß peptides are generated from the precursor protein Alcadein ß (Alcß) through cleavage by α- and γ-secretases of the amyloid ß (Aß) protein precursor (APP). To reveal whether p3-Alcß is involved in Alzheimer's disease (AD) contributes for the development of novel therapy and/or drug targets. METHODS: We developed new sandwich enzyme-linked immunosorbent assay (sELISA) systems to quantitate levels of p3-Alcß in the cerebrospinal fluid (CSF). RESULTS: In monkeys, CSF p3-Alcß decreases with age, and the aging is also accompanied by decreased brain expression of Alcß. In humans, CSF p3-Alcß levels decrease to a greater extent in those with AD than in age-matched controls. Subjects carrying presenilin gene mutations show a significantly lower CSF p3-Alcß level. A cell study with an inverse modulator of γ-secretase remarkably reduces the generation of p3-Alcß37 while increasing the production of Aß42. DISCUSSION: Aging decreases the generation of p3-Alcß, and further significant decrease of p3-Alcß caused by aberrant γ-secretase activity may accelerate pathogenesis in AD.

15.
Cell Rep ; 21(13): 3637-3645, 2017 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-29281813

RESUMO

Synaptopathies contributing to neurodevelopmental disorders are linked to mutations in synaptic organizing molecules, including postsynaptic neuroligins, presynaptic neurexins, and MDGAs, which regulate their interaction. The role of MDGA1 in suppressing inhibitory versus excitatory synapses is controversial based on in vitro studies. We show that genetic deletion of MDGA1 in vivo elevates hippocampal CA1 inhibitory, but not excitatory, synapse density and transmission. Furthermore, MDGA1 is selectively expressed by pyramidal neurons and regulates perisomatic, but not distal dendritic, inhibitory synapses. Mdga1-/- hippocampal networks demonstrate muted responses to neural excitation, and Mdga1-/- mice are resistant to induced seizures. Mdga1-/- mice further demonstrate compromised hippocampal long-term potentiation, consistent with observed deficits in spatial and context-dependent learning and memory. These results suggest that mutations in MDGA1 may contribute to cognitive deficits through altered synaptic transmission and plasticity by loss of suppression of inhibitory synapse development in a subcellular domain- and cell-type-selective manner.


Assuntos
Cognição , Rede Nervosa/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Inibição Neural , Sinapses/metabolismo , Animais , Região CA1 Hipocampal/patologia , Deleção de Genes , Potenciação de Longa Duração , Camundongos Endogâmicos C57BL , Camundongos Knockout , Moléculas de Adesão de Célula Nervosa/deficiência , Sinapses/ultraestrutura , Transmissão Sináptica
16.
Neuron ; 91(5): 1052-1068, 2016 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-27608760

RESUMO

Mutations in a synaptic organizing pathway contribute to autism. Autism-associated mutations in MDGA2 (MAM domain containing glycosylphosphatidylinositol anchor 2) are thought to reduce excitatory/inhibitory transmission. However, we show that mutation of Mdga2 elevates excitatory transmission, and that MDGA2 blocks neuroligin-1 interaction with neurexins and suppresses excitatory synapse development. Mdga2(+/-) mice, modeling autism mutations, demonstrated increased asymmetric synapse density, mEPSC frequency and amplitude, and altered LTP, with no change in measures of inhibitory synapses. Behavioral assays revealed an autism-like phenotype including stereotypy, aberrant social interactions, and impaired memory. In vivo voltage-sensitive dye imaging, facilitating comparison with fMRI studies in autism, revealed widespread increases in cortical spontaneous activity and intracortical functional connectivity. These results suggest that mutations in MDGA2 contribute to altered cortical processing through the dual disadvantages of elevated excitation and hyperconnectivity, and indicate that perturbations of the NRXN-NLGN pathway in either direction from the norm increase risk for autism.


Assuntos
Moléculas de Adesão Celular Neuronais/fisiologia , Córtex Cerebral/fisiologia , Cognição/fisiologia , Proteínas Ligadas por GPI/fisiologia , Haploinsuficiência/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Células Cultivadas , Córtex Cerebral/metabolismo , Proteína 4 Homóloga a Disks-Large , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteínas Ligadas por GPI/biossíntese , Proteínas Ligadas por GPI/genética , Guanilato Quinases/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/fisiologia , Moléculas de Adesão de Célula Nervosa/biossíntese , Moléculas de Adesão de Célula Nervosa/genética , Receptores de AMPA/metabolismo , Receptores de AMPA/fisiologia , Sinapses/metabolismo
17.
J Stroke Cerebrovasc Dis ; 25(9): 2184-8, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27342700

RESUMO

BACKGROUND: The present study investigates the effects of d-allose, a rare sugar, on the inflammatory response after transient forebrain ischemia in the gerbil and whether it reduces oxidative stress (8-hydroxyl-2'-deoxyguanosine levels) and behavioral deficits. METHODS: Transient forebrain ischemia was induced by occlusion of the bilateral common carotid arteries for 5 minutes. d-Allose was intraperitoneally injected immediately after ischemia (400 mg/kg). Inflammatory cytokines and oxidative damage in the hippocampus and behavioral deficits were examined 3 days after ischemia. RESULTS: d-Allose administration reduced ischemia-induced cytokine production, oxidative stress, and behavioral deficits (motor and memory related). CONCLUSIONS: The present results suggest that d-allose reduces brain injury after transient global ischemia by suppressing inflammation as well as by inhibiting oxidative stress.


Assuntos
Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Edulcorantes/uso terapêutico , 8-Hidroxi-2'-Desoxiguanosina , Análise de Variância , Animais , Glicemia/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Gerbillinae , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos dos Movimentos/tratamento farmacológico , Transtornos dos Movimentos/etiologia , Traumatismo por Reperfusão/complicações , Fatores de Tempo
18.
Brain Res Bull ; 122: 45-53, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26930163

RESUMO

Ischemic tolerance (IT) is induced by a variety of insults to the brain (e.g., nonfatal ischemia, heat and hypoxia) and it provides a strong neuroprotective effect. Although the mechanisms are still not fully elucidated, Ca(2+) is regarded as a key mediator of IT. Ryanodine receptors (RyRs) are located in the sarcoplasmic/endoplasmic reticulum membrane and are responsible for the release of Ca(2+) from intracellular stores. In brain, neuronal RyRs are thought to play a role in various neuropathological conditions, including ischemia. The purpose of the present study was to investigate the involvement of RyRs in IT. Pretreatment with a RyR antagonist, dantrolene (25mg/kg, i.p), blocked IT in a gerbil global ischemia model, while a RyR agonist, caffeine (100mg/kg, i.p), stimulated the production of IT. In vitro, using rat hippocampal cells, short-term oxygen/glucose deprivation induced preconditioning and RyR antagonists, dantrolene (50 and 100 µM) and ryanodine (100 and 200 µM) prevented it. RyR protein and mRNA levels were transiently decreased after induction of IT. These results suggest that RyRs are involved in the induction of ischemic tolerance.


Assuntos
Isquemia Encefálica/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Cafeína/farmacologia , Cálcio/metabolismo , Agonistas dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio , Células Cultivadas , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Gerbillinae , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Neurônios/efeitos dos fármacos , Ratos , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo
19.
J Biol Chem ; 290(2): 987-95, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25406318

RESUMO

The neural type I membrane protein Alcadein α (Alcα), is primarily cleaved by amyloid ß-protein precursor (APP) α-secretase to generate a membrane-associated carboxyl-terminal fragment (Alcα CTF), which is further cleaved by γ-secretase to secrete p3-Alcα peptides and generate an intracellular cytoplasmic domain fragment (Alcα ICD) in the late secretory pathway. By association with the neural adaptor protein X11L (X11-like), Alcα and APP form a ternary complex that suppresses the cleavage of both Alcα and APP by regulating the transport of these membrane proteins into the late secretory pathway where secretases are active. However, it has not been revealed how Alcα and APP are directed from the ternary complex formed largely in the Golgi into the late secretory pathway to reach a nerve terminus. Using a novel transgenic mouse line expressing excess amounts of human Alcα CTF (hAlcα CTF) in neurons, we found that expression of hAlcα CTF induced excess production of hAlcα ICD, which facilitated APP transport into the nerve terminus and enhanced APP metabolism, including Aß generation. In vitro cell studies also demonstrated that excess expression of Alcα ICD released both APP and Alcα from the ternary complex. These results indicate that regulated intramembrane proteolysis of Alcα by γ-secretase regulates APP trafficking and the production of Aß in vivo.


Assuntos
Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Proteínas de Ligação ao Cálcio/genética , Secretases da Proteína Precursora do Amiloide/química , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Caderinas , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte , Citoplasma/metabolismo , Complexo de Golgi/metabolismo , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso , Estrutura Terciária de Proteína , Proteólise , Via Secretória/genética
20.
Mol Biol Cell ; 25(22): 3569-80, 2014 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-25165140

RESUMO

Alzheimer's ß-amyloid precursor protein (APP) associates with kinesin-1 via JNK-interacting protein 1 (JIP1); however, the role of JIP1 in APP transport by kinesin-1 in neurons remains unclear. We performed a quantitative analysis to understand the role of JIP1 in APP axonal transport. In JIP1-deficient neurons, we find that both the fast velocity (∼2.7 µm/s) and high frequency (66%) of anterograde transport of APP cargo are impaired to a reduced velocity (∼1.83 µm/s) and a lower frequency (45%). We identified two novel elements linked to JIP1 function, located in the central region of JIP1b, that interact with the coiled-coil domain of kinesin light chain 1 (KLC1), in addition to the conventional interaction of the JIP1b 11-amino acid C-terminal (C11) region with the tetratricopeptide repeat of KLC1. High frequency of APP anterograde transport is dependent on one of the novel elements in JIP1b. Fast velocity of APP cargo transport requires the C11 domain, which is regulated by the second novel region of JIP1b. Furthermore, efficient APP axonal transport is not influenced by phosphorylation of APP at Thr-668, a site known to be phosphorylated by JNK. Our quantitative analysis indicates that enhanced fast-velocity and efficient high-frequency APP anterograde transport observed in neurons are mediated by novel roles of JIP1b.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Precursor de Proteína beta-Amiloide/genética , Transporte Axonal/genética , Neurônios/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células COS , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Chlorocebus aethiops , Regulação da Expressão Gênica , Hipocampo/citologia , Hipocampo/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/citologia , Fosforilação , Plasmídeos , Cultura Primária de Células , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Transdução de Sinais , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...