Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neuroinflammation ; 19(1): 246, 2022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-36199097

RESUMO

Differential microglial inflammatory responses play a role in regulation of differentiation and maturation of oligodendrocytes (OLs) in brain white matter. How microglia-OL crosstalk is altered by traumatic brain injury (TBI) and its impact on axonal myelination and neurological function impairment remain poorly understood. In this study, we investigated roles of a Na+/H+ exchanger (NHE1), an essential microglial pH regulatory protein, in microglial proinflammatory activation and OL survival and differentiation in a murine TBI model induced by controlled cortical impact. Similar TBI-induced contusion volumes were detected in the Cx3cr1-CreERT2 control (Ctrl) mice and selective microglial Nhe1 knockout (Cx3cr1-CreERT2;Nhe1flox/flox, Nhe1 cKO) mice. Compared to the Ctrl mice, the Nhe1 cKO mice displayed increased resistance to initial TBI-induced white matter damage and accelerated chronic phase of OL regeneration at 30 days post-TBI. The cKO brains presented increased anti-inflammatory phenotypes of microglia and infiltrated myeloid cells, with reduced proinflammatory transcriptome profiles. Moreover, the cKO mice exhibited accelerated post-TBI sensorimotor and cognitive functional recovery than the Ctrl mice. These phenotypic outcomes in cKO mice were recapitulated in C57BL6J wild-type TBI mice receiving treatment of a potent NHE1 inhibitor HOE642 for 1-7 days post-TBI. Taken together, these findings collectively demonstrated that blocking NHE1 protein stimulates restorative microglial activation in oligodendrogenesis and neuroprotection, which contributes to accelerated brain repair and neurological function recovery after TBI.


Assuntos
Lesões Encefálicas Traumáticas , Substância Branca , Animais , Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Oligodendroglia , Recuperação de Função Fisiológica
2.
J Neurosci ; 40(2): 424-446, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31694961

RESUMO

Persistent endoplasmic reticulum (ER) stress in neurons is associated with activation of inflammatory cells and subsequent neuroinflammation following traumatic brain injury (TBI); however, the underlying mechanism remains elusive. We found that induction of neuronal-ER stress, which was mostly characterized by an increase in phosphorylation of a protein kinase R-like ER kinase (PERK) leads to release of excess interferon (IFN)ß due to atypical activation of the neuronal-STING signaling pathway. IFNß enforced activation and polarization of the primary microglial cells to inflammatory M1 phenotype with the secretion of a proinflammatory chemokine CXCL10 due to activation of STAT1 signaling. The secreted CXCL10, in turn, stimulated the T-cell infiltration by serving as the ligand and chemoattractant for CXCR3+ T-helper 1 (Th1) cells. The activation of microglial cells and infiltration of Th1 cells resulted in white matter injury, characterized by impaired myelin basic protein and neurofilament NF200, the reduced thickness of corpus callosum and external capsule, and decline of mature oligodendrocytes and oligodendrocyte precursor cells. Intranasal delivery of CXCL10 siRNA blocked Th1 infiltration but did not fully rescue microglial activation and white matter injury after TBI. However, impeding PERK-phosphorylation through the administration of GSK2656157 abrogated neuronal induction of IFNß, switched microglial polarization to M2 phenotype, prevented Th1 infiltration, and increased Th2 and Treg levels. These events ultimately attenuated the white matter injury and improved anxiety and depressive-like behavior following TBI.SIGNIFICANCE STATEMENT A recent clinical study showed that human brain trauma patients had enhanced expression of type-1 IFN; suggests that type-1 IFN signaling may potentially influence clinical outcome in TBI patients. However, it was not understood how TBI leads to an increase in IFNß and whether induction of IFNß has any influence on neuroinflammation, which is the primary reason for morbidity and mortality in TBI. Our study suggests that induction of PERK phosphorylation, a characteristic feature of ER stress is responsible for an increase in neuronal IFNß, which, in turn, activates microglial cells and subsequently manifests the infiltration of T cells to induce neuroinflammation and subsequently white matter injury. Blocking PERK phosphorylation using GSK2656157 (or PERK knockdown) the whole cascade of neuroinflammation was attenuated and improved cognitive function after TBI.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Estresse do Retículo Endoplasmático/fisiologia , Microglia/metabolismo , Linfócitos T , Substância Branca/fisiopatologia , eIF-2 Quinase/metabolismo , Animais , Feminino , Interferon beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Substância Branca/lesões
3.
J Neurosci ; 37(24): 5900-5911, 2017 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-28522733

RESUMO

The PKR-like ER kinase (PERK), a transmembrane protein, resides in the endoplasmic reticulum (ER). Its activation serves as a key sensor of ER stress, which has been implicated in traumatic brain injury (TBI). The loss of memory is one of the most common symptoms after TBI, but the precise role of PERK activation in memory impairment after TBI has not been well elucidated. Here, we have shown that blocking the activation of PERK using GSK2656157 prevents the loss of dendritic spines and rescues memory deficits after TBI. To elucidate the molecular mechanism, we found that activated PERK phosphorylates CAMP response element binding protein (CREB) and PSD95 directly at the S129 and T19 residues, respectively. Phosphorylation of CREB protein prevents its interaction with a coactivator, CREB-binding protein, and subsequently reduces the BDNF level after TBI. Conversely, phosphorylation of PSD95 leads to its downregulation in pericontusional cortex after TBI in male mice. Treatment with either GSK2656157 or overexpression of a kinase-dead mutant of PERK (PERK-K618A) rescues BDNF and PSD95 levels in the pericontusional cortex by reducing phosphorylation of CREB and PSD95 proteins after TBI. Similarly, administration of either GSK2656157 or overexpression of PERK-K618A in primary neurons rescues the loss of dendritic outgrowth and number of synapses after treatment with a PERK activator, tunicamycin. Therefore, our study suggests that inhibition of PERK phosphorylation could be a potential therapeutic target to restore memory deficits after TBI.SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) is the leading cause of death and disability around the world and affects 1.7 million Americans each year. Here, we have shown that TBI-activated PKR-like ER kinase (PERK) is responsible for memory deficiency, which is the most common problem in TBI patients. A majority of PERK's biological activities have been attributed to its function as an eIF2α kinase. However, our study suggests that activated PERK mediates its function via increasing phosphorylation of CAMP response element binding protein (CREB) and PSD95 after TBI. Blocking PERK phosphorylation rescues spine loss and memory deficits independently of phosphorylation of eIF2α. Therefore, our study suggests that CREB and PSD95 are novel substrates of PERK, so inhibition of PERK phosphorylation using GSK2656157 would be beneficial against memory impairment after TBI.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Guanilato Quinases/metabolismo , Proteínas de Membrana/metabolismo , Transtornos da Memória/metabolismo , eIF-2 Quinase/metabolismo , Animais , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/patologia , Espinhas Dendríticas/patologia , Proteína 4 Homóloga a Disks-Large , Regulação para Baixo , Ativação Enzimática , Masculino , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos C57BL
4.
Neurobiol Dis ; 118: 108-116, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30010002

RESUMO

Cell cycle activation has been associated with varying types of neurological disorders including brain injury. Cyclin D1 is a critical modulator of cell cycle activation and upregulation of Cyclin D1 in neurons contributes to the pathology associated with traumatic brain injury (TBI). Mitochondrial mass is a critical factor to maintain the mitochondrial function, and it can be regulated by different signaling cascades and transcription factors including NRF1. However, the underlying mechanism of how TBI leads to impairment of mitochondrial mass following TBI remains obscure. Our results indicate that augmentation of CyclinD1 attenuates mitochondrial mass formation following TBI. To elucidate the molecular mechanism, we found that Cyclin D1 interacts with a transcription factor NRF1 in the nucleus and prevents NRF1's interaction with p300 in the pericontusional cortex following TBI. As a result, the acetylation level of NRF1 was decreased, and its transcriptional activity was attenuated. This event leads to a loss of mitochondrial mass in the pericontusional cortex following TBI. Intranasal delivery of Cyclin D1 RNAi immediately after TBI rescues transcriptional activation of NRF1 and recovers mitochondrial mass after TBI.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Ciclina D1/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Animais , Lesões Encefálicas Traumáticas/genética , Ciclina D1/antagonistas & inibidores , Ciclina D1/genética , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/genética , Interferência de RNA/fisiologia
5.
Neurochem Res ; 39(1): 150-60, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24277080

RESUMO

The synaptic glutamate level homeostasis is mainly maintained by the astrocytes membrane bound glutamate transporter type-1 (GLT-1/EAAT2). Alterations in its expression during development and aging and the underlying mechanisms are not well studied. Here, we report that NF-κB interaction was highest in both cerebral and cerebellar cortices at day 15 when compared with that at day 0 during development, and it further declined significantly in day 45, and remained unchanged in 20 and 70 weeks mice. On the other hand, N-myc interaction was highest at 0 day which significantly declined at 15-day and interestingly remained unaltered at later ages in both the cortices. This age dependent reciprocal pattern of NF-κB and N-myc interactions with their cognate GLT-1 promoter sequences was further correlated with GLT-1 protein and transcript levels. We found that higher NF-κB interaction with its cognate GLT-1 promoter sequences correlates with up-regulation whereas the higher N-myc interaction correlates with down-regulation of GLT-1 expression during postnatal developmental age up to 15 day, however, such phenomenon was not found in the higher ages from day 45 to 70 weeks. Thus our data suggests a postnatal development- and age dependent differential interaction of transcription factors NF-κB and N-myc to their respective sequences and they act as positive and negative regulator, respectively of GLT-1 gene expression in the brain during early developmental period in both cerebral and cerebellar cortices which might be different in aging of mice.


Assuntos
Córtex Cerebelar/crescimento & desenvolvimento , Córtex Cerebral/crescimento & desenvolvimento , Transportador 2 de Aminoácido Excitatório/biossíntese , NF-kappa B/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Animais , Córtex Cerebelar/metabolismo , Córtex Cerebral/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , Camundongos , Regiões Promotoras Genéticas/fisiologia
6.
Artigo em Inglês | MEDLINE | ID: mdl-37002525

RESUMO

Neonatal exposure to decabromodiphenyl ether (PBDE-209), a widely used flame retardant, affects cognitive performances in the later stage of life in a sex-dependent manner. PBDE-209 interferes with glutamatergic signaling and N-methyl-D-aspartate receptor (NMDAR) subunits with unresolved regulatory mechanisms. This study exposed male and female mice pups through postnatal day (PND) 3-10 to PBDE-209 (oral dose: 0, 6, or 20 mg/kg body weight). The frontal cortex and hippocampus, collected from neonate (PND 11) and young (PND 60) mice, were analyzed for cAMP response element-binding protein (CREB) and RE1-silencing transcription factor/ Neuron-restrictive silencer factor (REST/NRSF) binding to NMDAR1 promoter and expression of NMDAR1 gene by electrophoretic mobility shift assay and semi-quantitative RT-PCR respectively. Behavioral changes were assessed using spontaneous alternation behavior and novel object recognition tests in young mice. In neonates, the binding of CREB was increased, while REST/NRSF was decreased significantly to their cognate NMDAR1 promoter sequences at the high dose of PBDE-209 in both the sexes. This reciprocal pattern of CREB and REST/NRSF interactions correlates with the up-regulation of NMDAR1 expression. Young males followed a similar pattern of CREB and REST/NRSF binding and NMDAR1 expression as in neonates. Surprisingly, young females did not show any alteration when compared to age-matched controls. Also, we found that only young males showed working and recognition memory deficits. These results indicate that early exposure to PBDE-209 interferes with CREB- and REST/NRSF-dependent regulation of the NMDAR1 gene in an acute setting. However, long-term effects persist only in young males that could be associated with cognitive impairment.

7.
PLoS One ; 16(9): e0257784, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34582497

RESUMO

Drug repurposing has the potential to bring existing de-risked drugs for effective intervention in an ongoing pandemic-COVID-19 that has infected over 131 million, with 2.8 million people succumbing to the illness globally (as of April 04, 2021). We have used a novel `gene signature'-based drug repositioning strategy by applying widely accepted gene ranking algorithms to prioritize the FDA approved or under trial drugs. We mined publically available RNA sequencing (RNA-Seq) data using CLC Genomics Workbench 20 (QIAGEN) and identified 283 differentially expressed genes (FDR<0.05, log2FC>1) after a meta-analysis of three independent studies which were based on severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) infection in primary human airway epithelial cells. Ingenuity Pathway Analysis (IPA) revealed that SARS-CoV-2 activated key canonical pathways and gene networks that intricately regulate general anti-viral as well as specific inflammatory pathways. Drug database, extracted from the Metacore and IPA, identified 15 drug targets (with information on COVID-19 pathogenesis) with 46 existing drugs as potential-novel candidates for repurposing for COVID-19 treatment. We found 35 novel drugs that inhibit targets (ALPL, CXCL8, and IL6) already in clinical trials for COVID-19. Also, we found 6 existing drugs against 4 potential anti-COVID-19 targets (CCL20, CSF3, CXCL1, CXCL10) that might have novel anti-COVID-19 indications. Finally, these drug targets were computationally prioritized based on gene ranking algorithms, which revealed CXCL10 as the common and strongest candidate with 2 existing drugs. Furthermore, the list of 283 SARS-CoV-2-associated proteins could be valuable not only as anti-COVID-19 targets but also useful for COVID-19 biomarker development.


Assuntos
Tratamento Farmacológico da COVID-19 , Reposicionamento de Medicamentos/métodos , SARS-CoV-2/genética , Antivirais/uso terapêutico , Avaliação Pré-Clínica de Medicamentos/métodos , Células Epiteliais/efeitos dos fármacos , Epitélio/efeitos dos fármacos , Humanos , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Mucosa Respiratória/virologia , Sistema Respiratório/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/patogenicidade
8.
Free Radic Biol Med ; 134: 630-643, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30790655

RESUMO

Traumatic Brain Injury (TBI) affects more than 1.7 million Americans each year and about 30% of TBI-patients having visual impairments. The loss of retinal ganglion cells (RGC) in the retina and axonal degeneration in the optic nerve have been attributed to vision impairment following TBI; however, the molecular mechanism has not been elucidated. Here we have shown that an increase in histone di-methylation at lysine 9 residue (H3K9Me2), synthesized by the catalytic activity of a histone methyltransferase, G9a is responsible for RGC loss and axonal degeneration in the optic nerve following TBI. To elucidate the molecular mechanism, we found that an increase in H3K9Me2 results in the induction of oxidative stress both in the RGC and optic nerve by decreasing the mRNA level of antioxidants such as Superoxide dismutase (sod) and catalase through impairing the transcriptional activity of Nuclear factor E2-related factor 2 (Nrf2) via direct interaction. The induction of oxidative stress is associated with death in RGC and oligodendrocyte precursor cells (OPCs). The death in OPCs is correlated with a reduction in myelination, and the expression of myelin binding protein (MBP) in association with degeneration of neurofilaments in the optic nerve. This event allied to an impairment of the retrograde transport of axons and loss of nerve fiber layer in the optic nerve following TBI. An administration of G9a inhibitor, UNC0638 attenuates the induction of H3K9Me2 both in RGC and optic nerve and subsequently activates Nrf2 to reduce oxidative stress. This event was concomitant with the rescue in the loss of retinal thickness, attenuation in optic nerve degeneration and improvement in the retrograde transport of axons following TBI.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Histonas/metabolismo , Lisina/metabolismo , Nervo Óptico/patologia , Estresse Oxidativo , Células Ganglionares da Retina/patologia , Transtornos da Visão/etiologia , Animais , Lesões Encefálicas Traumáticas/patologia , Modelos Animais de Doenças , Histonas/genética , Lisina/genética , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Nervo Óptico/metabolismo , Células Ganglionares da Retina/metabolismo , Transtornos da Visão/metabolismo , Transtornos da Visão/patologia
9.
J Neurotrauma ; 36(23): 3284-3296, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31169064

RESUMO

An enduring deficit in neurogenesis largely contributes to the development of severe post-traumatic psychiatric disorders such as anxiety, depression, and memory impairment following traumatic brain injury (TBI); however, the mechanism remains obscure. Here we have shown that an imbalance in the generation of γ-aminobutyric acid (GABA)ergic and glutamatergic neurons due to aberrant induction of vesicular glutamate transporter 1 (vGlut1)-positive glutamatergic cells is responsible for impaired neuronal differentiation in the hippocampus following TBI. To elucidate the molecular mechanism, we found that TBI activates a transcription factor, Pax3, by increasing its acetylation status, and subsequently induces Ngn2 transcription. This event, in turn, augments the vGlut1-expressing glutamatergic neurons and accumulation of excess glutamate in the hippocampus that can affect neuronal differentiation. In our study the acetylation of Pax3 was increased due to loss of its interaction with a deacetylase, histone deacetylase 4 (HDAC4), which was downregulated after TBI. TBI-induced activation of GSK3ß was responsible for the degradation of HDAC4. We also showed that overexpression of HDAC4 before TBI reduces Pax3 acetylation by restoring an interaction between HDAC4 and Pax3 in the hippocampus. This event prevents the aberrant induction of vGlut1-positive glutamatergic neurons by decreasing the Ngn2 level and subsequently reinforces the balance between GABAergic and glutamatergic neurons following TBI. Further, we found that overexpression of HDAC4 in the hippocampus improves anxiety, depressive-like behavior, and memory functions following TBI.


Assuntos
Lesões Encefálicas Traumáticas/enzimologia , Regulação para Baixo/fisiologia , Histona Desacetilases/metabolismo , Transtornos Mentais/enzimologia , Neurogênese/fisiologia , Animais , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/psicologia , Feminino , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos Mentais/patologia , Transtornos Mentais/psicologia , Camundongos , Camundongos Endogâmicos C57BL
10.
Rev Neurosci ; 27(1): 93-100, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26352199

RESUMO

Traumatic brain injury (TBI), a major global health and socioeconomic problem, is now established as a chronic disease process with a broad spectrum of pathophysiological symptoms followed by long-term disabilities. It triggers multiple and multidirectional biochemical events that lead to neurodegeneration and cognitive impairment. Recent studies have presented strong evidence that patients with TBI history have a tendency to develop proteinopathy, which is the pathophysiological feature of neurodegenerative disorders such as Alzheimer disease (AD), chronic traumatic encephalopathy (CTE), and amyotrophic lateral sclerosis (ALS). This review mainly focuses on mechanisms related to AD, CTE, and ALS that are induced after TBI and their relevance to the advancement of these neurodegenerative diseases. This review encompasses acute effects and chronic neurodegenerative consequences after TBI for a better understanding of TBI-induced neuronal death and to design therapies that will effectively treat patients in the primary or secondary progressive stages.


Assuntos
Doença de Alzheimer/etiologia , Esclerose Lateral Amiotrófica/etiologia , Lesões Encefálicas/complicações , Doença de Alzheimer/epidemiologia , Doença de Alzheimer/metabolismo , Esclerose Lateral Amiotrófica/epidemiologia , Esclerose Lateral Amiotrófica/metabolismo , Animais , Lesões Encefálicas/metabolismo , Humanos
11.
Mol Neurobiol ; 53(5): 3377-3388, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26081154

RESUMO

Traumatic brain injury (TBI) is one of the major risk factors of dementia, aging, and cognitive impairments, etc. We have previously reported that expression of the astrocytic glutamate transporter GLT-1/EAAT2 is downregulated in the pericontusional cortex of adult and old mice in post-TBI time-dependent manner, and the process of decline starts before in old than in adult TBI mice. However, relationship between age- and TBI-dependent alterations in GLT-1/EAAT2 expression and interactions of transcription factors NF-κB and N-myc with their cognate GLT-1/EAAT2 promoter sequences, an important step of its transcriptional control, is not known. To understand this, we developed TBI mouse model by modified chronic head injury (CHI) method, analyzed expression of GFAP, TNF-α, and AQP4 by RT-PCR for its validation, and analyzed interactions of NF-κB and N-myc with GLT-1/EAAT2 promoter sequences by electrophoretic mobility shift assay (EMSA). Our EMSA data revealed that interactions of NF-κB and N-myc with GLT-1/EAAT2 promoter sequences was significantly elevated in the ipsi-lateral cortex of both adult and old TBI mice in post-TBI time-dependent manner; however, these interactions started immediately in the old compared to that in adult TBI mice, which could be attributed to our previously reported age- and post-TBI time-dependent differential expression of GLT-1/EAAT2 in the pericontusional cortex.


Assuntos
Envelhecimento/patologia , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/patologia , Córtex Cerebral/patologia , Transportador 2 de Aminoácido Excitatório/genética , Proteína Proto-Oncogênica N-Myc/metabolismo , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Animais , Aquaporina 4/genética , Aquaporina 4/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Transportador 2 de Aminoácido Excitatório/metabolismo , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Camundongos , Ligação Proteica/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-26413122

RESUMO

CDRI-08 is a standardized bacoside enriched ethanolic extract of Bacopa monnieri, a nootropic plant. We reported that CDRI-08 attenuated oxidative stress and memory impairment in mice, induced by a flame retardant, PBDE-209. In order to explore the mechanism, present study was designed to examine the role of CDRI-08 on the expression of NMDAR1 (NR1) and the binding of REST/NRSF to NR1 promoter against postnatal exposure of PBDE-209. Male mice pups were orally supplemented with CDRI-08 at the doses of 40, 80, or 120 mg/kg along with PBDE-209 (20 mg/kg) during PND 3-10 and frontal cortex and hippocampus were collected at PND 11 and 60 to study the expression and regulation of NR1 by RT-PCR and electrophoretic mobility shift assay, respectively. The findings showed upregulated expression of NR1 and decreased binding of REST/NRSF to NR1 promoter after postnatal exposure of PBDE-209. Interestingly, supplementation with CDRI-08 significantly restored the expression of NR1 and binding of REST/NRSF to NR1 promoter near to the control value at the dose of 120 mg/kg. In conclusion, the results suggest that CDRI-08 possibly acts on glutamatergic system through expression and regulation of NR1 and may restore memory, impaired by PBDE-209 as reported in our previous study.

13.
Age (Dordr) ; 35(1): 59-67, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22057895

RESUMO

Glial cells, besides participating as passive supporting matrix, are also proposed to be involved in the optimization of the interstitial space for synaptic transmission by tight control of ionic and water homeostasis. In adult mouse brain, inwardly rectifying K+ (Kir4.1) and aquaporin-4 (AQP4) channels localize to astroglial endfeets in contact with brain microvessels and glutamate synapses, optimizing clearance of extracellular K(+) and water from the synaptic layers. However, it is still unclear whether there is an age-dependent difference in the expressions of Kir4.1 and AQP4 channels specifically during postnatal development and aging when various marked changes occur in brain and if these changes region specific. RT-PCR and immunoblotting was conducted to compare the relative expression of Kir4.1 and AQP4 mRNA and protein in the early and mature postnatal (0-, 15-, 45-day), adult (20-week), and old age (70-week) mice cerebral and cerebellar cortices. Expressions of Kir4.1 and AQP4 mRNA and protein are very low at 0-day. A pronounced and continuous increase was observed by mature postnatal ages (15-, 45-days). However, in the 70-week-old mice, expressions are significantly up-regulated as compared to 20-week-old mice. Both genes follow the same age-related pattern in both cerebral and cerebellar cortices. The time course and expression pattern suggests that Kir4.1 and AQP4 channels may play an important role in brain K(+) and water homeostasis in early postnatal weeks after birth and during aging.


Assuntos
Envelhecimento/genética , Encéfalo/crescimento & desenvolvimento , Neuroglia/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , RNA Mensageiro/genética , Regulação para Cima , Animais , Eletroforese em Gel de Poliacrilamida , Masculino , Camundongos , Camundongos Endogâmicos AKR , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA