Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 19(2): e0297289, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38315685

RESUMO

Alzheimer's disease (AD) is characterized by cognitive and memory impairments and neuropathological abnormalities. AD has no cure, inadequate treatment options, and a limited understanding of possible prevention measures. Previous studies have demonstrated that AD model mice that received a diet high in the essential nutrient choline had reduced amyloidosis, cholinergic deficits, and gliosis, and increased neurogenesis. In this study, we investigated the lifelong effects of perinatal choline supplementation on behavior, cognitive function, and amyloidosis in AppNL-G-F AD model mice. Pregnant and lactating mice were given a diet containing either 1.1 g/kg (control) or 5 g/kg (supplemented) of choline chloride until weaning and subsequently, all offspring received the control diet throughout their life. At 3, 6, 9, and 12 months of age, animals were behaviorally tested in the Open Field Test, Elevated Plus Maze, Barnes Maze, and in a contextual fear conditioning paradigm. Immunohistochemical analysis of Aß42 was also conducted on the brains of these mice. AppNL-G-F mice displayed hippocampal-dependent spatial learning deficits starting at 3-months-old that persisted until 12-months-old. These spatial learning deficits were fully prevented by perinatal choline supplementation at young ages (3 and 6 months) but not in older mice (12 months). AppNL-G-F mice also had impaired fearful learning and memory at 9- and 12-months-old that were diminished by choline supplementation. Perinatal choline supplementation reduced Aß42 deposition in the amygdala, cortex, and hippocampus of AppNL-G-F mice. Together, these results demonstrate that perinatal choline supplementation is capable of preventing cognitive deficits and dampening amyloidosis in AppNL-G-F mice and suggest that ensuring adequate choline consumption during early life may be a valuable method to prevent or reduce AD dementia and neuropathology.


Assuntos
Doença de Alzheimer , Amiloidose , Gravidez , Feminino , Camundongos , Animais , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/patologia , Camundongos Transgênicos , Lactação , Modelos Animais de Doenças , Encéfalo/metabolismo , Amiloidose/patologia , Colina/farmacologia , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/prevenção & controle , Transtornos da Memória/patologia , Aprendizagem em Labirinto , Suplementos Nutricionais , Peptídeos beta-Amiloides/metabolismo
2.
Noncoding RNA ; 7(1)2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33535543

RESUMO

The neurological damage of Alzheimer's disease (AD) is thought to be irreversible upon onset of dementia-like symptoms, as it takes years to decades for occult pathologic changes to become symptomatic. It is thus necessary to identify individuals at risk for the development of the disease before symptoms manifest in order to provide early intervention. Surrogate markers are critical for early disease detection, stratification of patients in clinical trials, prediction of disease progression, evaluation of response to treatment, and also insight into pathomechanisms. Here, we review the evidence for a number of microRNAs that may serve as biomarkers with possible mechanistic insights into the AD pathophysiologic processes, years before the clinical manifestation of the disease.

3.
J Alzheimers Dis ; 68(1): 357-365, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30775993

RESUMO

Genome-wide association studies identified a single nucleotide polymorphism (SNP) in the MSRB3 gene encoding Methionine Sulfoxide Reductase-B3 (MsrB3) to be associated with the risk for low hippocampal volume and late onset Alzheimer's disease (AD). Subsequently, we identified AD-associated abnormal patterns of neuronal and vascular MsrB3 expression in postmortem hippocampi. The present study investigated the relationship between the MSRB3 SNP rs61921502, G (minor/risk allele) and MRI measures of brain injury including total brain volume, hippocampal volume, and white matter hyperintensities using linear regression models; the presence of brain infarcts using logistic regression models; and the incidence of stroke, dementia, and AD using Cox proportional hazards models in 2,038 Framingham Heart Study Offspring participants with MRI administered close to examination cycle 7 (1998-2001). Participants with neurological conditions that impede evaluation of vascular pathology by MRI, i.e., brain tumors, multiple sclerosis, and major head trauma, were excluded from the study. When adjusted for age and age squared at MRI exam, sex, and presence of Apolipoproteinɛ4 allele (APOE4), individuals with MSRB3 rs61921502 minor allele had increased odds for brain infarcts on MRI compared to those with no minor allele. However, in stratified analyses, MSRB3 rs61921502 minor allele was significantly associated with increased odds for MRI brain infarcts only in the absence of APOE4.


Assuntos
Alelos , Infarto Encefálico/genética , Demência/genética , Predisposição Genética para Doença , Hipocampo/diagnóstico por imagem , Metionina Sulfóxido Redutases/genética , Polimorfismo de Nucleotídeo Único , Idoso , Infarto Encefálico/diagnóstico por imagem , Infarto Encefálico/epidemiologia , Demência/diagnóstico por imagem , Demência/epidemiologia , Feminino , Humanos , Incidência , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade
5.
PLoS One ; 12(1): e0170450, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28103298

RESUMO

Prevention of Alzheimer's disease (AD) is a major goal of biomedical sciences. In previous studies we showed that high intake of the essential nutrient, choline, during gestation prevented age-related memory decline in a rat model. In this study we investigated the effects of a similar treatment on AD-related phenotypes in a mouse model of AD. We crossed wild type (WT) female mice with hemizygous APPswe/PS1dE9 (APP.PS1) AD model male mice and maintained the pregnant and lactating dams on a control AIN76A diet containing 1.1 g/kg of choline or a choline-supplemented (5 g/kg) diet. After weaning all offspring consumed the control diet. As compared to APP.PS1 mice reared on the control diet, the hippocampus of the perinatally choline-supplemented APP.PS1 mice exhibited: 1) altered levels of amyloid precursor protein (APP) metabolites-specifically elevated amounts of ß-C-terminal fragment (ß-CTF) and reduced levels of solubilized amyloid Aß40 and Aß42 peptides; 2) reduced number and total area of amyloid plaques; 3) preserved levels of choline acetyltransferase protein (CHAT) and insulin-like growth factor II (IGF2) and 4) absence of astrogliosis. The data suggest that dietary supplementation of choline during fetal development and early postnatal life may constitute a preventive strategy for AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/prevenção & controle , Precursor de Proteína beta-Amiloide/genética , Amiloidose/prevenção & controle , Colina O-Acetiltransferase/metabolismo , Colina/administração & dosagem , Suplementos Nutricionais , Hipocampo/metabolismo , Presenilina-1/genética , Doença de Alzheimer/dietoterapia , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/patologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Masculino , Camundongos , Camundongos Mutantes , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neurogênese/efeitos dos fármacos , Gravidez , Presenilina-1/metabolismo
6.
Sci Transl Med ; 4(134): 134ra60, 2012 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22593173

RESUMO

Blast exposure is associated with traumatic brain injury (TBI), neuropsychiatric symptoms, and long-term cognitive disability. We examined a case series of postmortem brains from U.S. military veterans exposed to blast and/or concussive injury. We found evidence of chronic traumatic encephalopathy (CTE), a tau protein-linked neurodegenerative disease, that was similar to the CTE neuropathology observed in young amateur American football players and a professional wrestler with histories of concussive injuries. We developed a blast neurotrauma mouse model that recapitulated CTE-linked neuropathology in wild-type C57BL/6 mice 2 weeks after exposure to a single blast. Blast-exposed mice demonstrated phosphorylated tauopathy, myelinated axonopathy, microvasculopathy, chronic neuroinflammation, and neurodegeneration in the absence of macroscopic tissue damage or hemorrhage. Blast exposure induced persistent hippocampal-dependent learning and memory deficits that persisted for at least 1 month and correlated with impaired axonal conduction and defective activity-dependent long-term potentiation of synaptic transmission. Intracerebral pressure recordings demonstrated that shock waves traversed the mouse brain with minimal change and without thoracic contributions. Kinematic analysis revealed blast-induced head oscillation at accelerations sufficient to cause brain injury. Head immobilization during blast exposure prevented blast-induced learning and memory deficits. The contribution of blast wind to injurious head acceleration may be a primary injury mechanism leading to blast-related TBI and CTE. These results identify common pathogenic determinants leading to CTE in blast-exposed military veterans and head-injured athletes and additionally provide mechanistic evidence linking blast exposure to persistent impairments in neurophysiological function, learning, and memory.


Assuntos
Traumatismos por Explosões/complicações , Traumatismos por Explosões/patologia , Lesão Encefálica Crônica/complicações , Lesão Encefálica Crônica/patologia , Militares/psicologia , Veteranos/psicologia , Aceleração , Adolescente , Adulto , Animais , Atletas , Axônios/patologia , Comportamento Animal , Traumatismos por Explosões/fisiopatologia , Concussão Encefálica/complicações , Concussão Encefálica/patologia , Concussão Encefálica/fisiopatologia , Lesão Encefálica Crônica/fisiopatologia , Modelos Animais de Doenças , Cabeça/patologia , Cabeça/fisiopatologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Hipocampo/ultraestrutura , Humanos , Pressão Intracraniana , Potenciação de Longa Duração , Masculino , Camundongos , Pessoa de Meia-Idade , Fosforilação , Mudanças Depois da Morte , Transmissão Sináptica , Adulto Jovem , Proteínas tau/metabolismo
7.
Brain Res ; 1413: 84-97, 2011 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-21840511

RESUMO

Choline is a vital nutrient needed during early development for both humans and rodents. Severe dietary choline deficiency during pregnancy leads to birth defects, while more limited deficiency during mid- to late pregnancy causes deficits in hippocampal plasticity in adult rodent offspring that are accompanied by cognitive deficits only when task demands are high. Because prenatal choline supplementation confers neuroprotection of the adult hippocampus against a variety of neural insults and aids memory, we hypothesized that prenatal choline deficiency may enhance vulnerability to neural injury. To examine this, adult offspring of rat dams either fed a control diet (CON) or one deficient in choline (DEF) during embryonic days 12-17 were given multiple injections (i.p.) of saline (control) or kainic acid to induce seizures and were euthanized 16 days later. Perhaps somewhat surprisingly, DEF rats were not more susceptible to seizure induction and showed similar levels of seizure-induced hippocampal histopathology, GAD expression loss, upregulated hippocampal GFAP and growth factor expression, and increased dentate cell and neuronal proliferation as that seen in CON rats. Although prenatal choline deficiency compromises adult hippocampal plasticity in the intact brain, it does not appear to exacerbate the neuropathological response to seizures in the adult hippocampus at least shortly after excitotoxic injury.


Assuntos
Deficiência de Colina/metabolismo , Colina/administração & dosagem , Hipocampo/metabolismo , Ácido Caínico/toxicidade , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Convulsões/metabolismo , Fatores Etários , Animais , Deficiência de Colina/induzido quimicamente , Suscetibilidade a Doenças , Feminino , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Masculino , Fármacos Neuroprotetores/administração & dosagem , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente
8.
PLoS One ; 6(6): e21166, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21695154

RESUMO

BACKGROUND: Cholinergic projection from the septum to the hippocampus is crucial for normal cognitive function and degeneration of cells and nerve fibers within the septohippocampal pathway contributes to the pathophysiology of Alzheimer's disease. Bone morphogenetic protein (BMP) 9 is a cholinergic differentiating factor during development both in vivo and in vitro. METHODOLOGY/PRINCIPAL FINDINGS: To determine whether BMP9 could protect the adult cholinergic septohippocampal pathway from axotomy-evoked loss of the cholinergic phenotype, we performed unilateral fimbria-fornix transection in mice and treated them with a continuous intracerebroventricular infusion of BMP9 for six days. The number of choline acetyltransferase (CHAT)-positive cells was reduced by 50% in the medial septal nucleus ipsilateral to the lesion as compared to the intact, contralateral side, and BMP9 infusion prevented this loss in a dose-dependent manner. Moreover, BMP9 prevented most of the decline of hippocampal acetylcholine levels ipsilateral to the lesion, and markedly increased CHAT, choline transporter CHT, NGF receptors p75 (NGFR-p75) and TrkA (NTRK1), and NGF protein content in both the lesioned and unlesioned hippocampi. In addition, BMP9 infusion reduced bilaterally hippocampal levels of basic FGF (FGF2) protein. CONCLUSIONS/SIGNIFICANCE: These data indicate that BMP9 administration can prevent lesion-evoked impairment of the cholinergic septohippocampal neurons in adult mice and, by inducing NGF, establishes a trophic environment for these cells.


Assuntos
Acetilcolina/metabolismo , Fatores de Diferenciação de Crescimento/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenótipo , Septo do Cérebro/citologia , Acetilcolina/biossíntese , Animais , Axotomia , Biomarcadores/metabolismo , Colina O-Acetiltransferase/metabolismo , Fórnice/cirurgia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Fator 2 de Diferenciação de Crescimento , Fatores de Diferenciação de Crescimento/administração & dosagem , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Bombas de Infusão , Masculino , Camundongos , Fator de Crescimento Neural/metabolismo , Neurônios/enzimologia , Receptor de Fator de Crescimento Neural/metabolismo , Receptor trkA/metabolismo , Regulação para Cima/efeitos dos fármacos
9.
BMC Cell Biol ; 12: 20, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21586144

RESUMO

BACKGROUND: The amyloid precursor protein (APP) is cleaved by ß- and γ-secretases to generate toxic amyloid ß (Aß) peptides. Alternatively, α-secretases cleave APP within the Aß domain, precluding Aß formation and releasing the soluble ectodomain, sAPPα. We previously showed that inhibition of the GTPase dynamin reduced APP internalization and increased release of sAPPα, apparently by prolonging the interaction between APP and α-secretases at the plasma membrane. This was accompanied by a reduction in Aß generation. In the present study, we investigated whether surface expression of the α-secretase ADAM (a disintegrin and metalloprotease)10 is also regulated by dynamin-dependent endocytosis. RESULTS: Transfection of human embryonic kidney (HEK) cells stably expressing M3 muscarinic receptors with a dominant negative dynamin I mutant (dyn I K44A), increased surface expression of both immature, and mature, catalytically active forms of co-expressed ADAM10. Surface levels of ADAM10 were unaffected by activation of protein kinase C (PKC) or M3 receptors, indicating that receptor-coupled shedding of the ADAM substrate APP is unlikely to be mediated by inhibition of ADAM10 endocytosis in this cell line. Dyn I K44A strongly increased the formation of a C-terminal fragment of ADAM10, consistent with earlier reports that the ADAM10 ectodomain is itself a target for sheddases. The abundance of this fragment was increased in the presence of a γ-secretase inhibitor, but was not affected by M3 receptor activation. The dynamin mutant did not affect the distribution of ADAM10 and its C-terminal fragment between raft and non-raft membrane compartments. CONCLUSIONS: Surface expression and limited proteolysis of ADAM10 are regulated by dynamin-dependent endocytosis, but are unaffected by activation of signaling pathways that upregulate shedding of ADAM substrates such as APP. Modulation of ADAM10 internalization could affect cellular behavior in two ways: by altering the putative signaling activity of the ADAM10 C-terminal fragment, and by regulating the biological function of ADAM10 substrates such as APP and N-cadherin.


Assuntos
Proteínas ADAM/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Dinamina I/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/metabolismo , Carbacol/farmacologia , Carbamatos/farmacologia , Linhagem Celular , Dipeptídeos/farmacologia , Dinamina I/genética , Dinamina I/metabolismo , Endocitose , Humanos , Proteína Quinase C/metabolismo , Receptor Muscarínico M3/metabolismo
10.
Hippocampus ; 21(6): 584-608, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20232399

RESUMO

Status epilepticus (SE) in adulthood dramatically alters the hippocampus and produces spatial learning and memory deficits. Some factors, like environmental enrichment and exercise, may promote functional recovery from SE. Prenatal choline supplementation (SUP) also protects against spatial memory deficits observed shortly after SE in adulthood, and we have previously reported that SUP attenuates the neuropathological response to SE in the adult hippocampus just 16 days after SE. It is unknown whether SUP can ameliorate longer-term cognitive and neuropathological consequences of SE, whether repeatedly engaging the injured hippocampus in a cognitive task might facilitate recovery from SE, and whether our prophylactic prenatal dietary treatment would enable the injured hippocampus to more effectively benefit from cognitive rehabilitation. To address these issues, adult offspring from rat dams that received either a control (CON) or SUP diet on embryonic days 12-17 first received training on a place learning water maze task (WM) and were then administered saline or kainic acid (KA) to induce SE. Rats then either remained in their home cage, or received three additional WM sessions at 3, 6.5, and 10 weeks after SE to test spatial learning and memory retention. Eleven weeks after SE, the brains were analyzed for several hippocampal markers known to be altered by SE. SUP attenuated SE-induced spatial learning deficits and completely rescued spatial memory retention by 10 weeks post-SE. Repeated WM experience prevented SE-induced declines in glutamic acid decarboxylase (GAD) and dentate gyrus neurogenesis, and attenuated increased glial fibrilary acidic protein (GFAP) levels. Remarkably, SUP alone was similarly protective to an even greater extent, and SUP rats that were water maze trained after SE showed reduced hilar migration of newborn neurons. These findings suggest that prophylactic SUP is protective against the long-term cognitive and neuropathological effects of KA-induced SE, and that rehabilitative cognitive enrichment may be partially beneficial.


Assuntos
Colina/administração & dosagem , Hipocampo , Ácido Caínico/efeitos adversos , Fenômenos Fisiológicos da Nutrição Pré-Natal/fisiologia , Estado Epiléptico , Animais , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Glutamato Descarboxilase/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/patologia , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Neurônios/fisiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Sprague-Dawley , Retenção Psicológica/efeitos dos fármacos , Percepção Espacial/efeitos dos fármacos , Percepção Espacial/fisiologia , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/dietoterapia , Estado Epiléptico/patologia , Estado Epiléptico/prevenção & controle
11.
Nutr Res ; 30(10): 722-30, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21056288

RESUMO

The hypothesis of this study is that a folate-deficient diet (FD) has a greater effect on cholinergic system in the peripheral nervous system than in the brain, and that this effect escalates with age. It was tested by comparing choline and acetylcholine levels in male Sprague Dawley rats fed either control or folate-deficient diets for 10 weeks, starting at age 4 weeks (the young group) or 9 months (the adult group). Folate-deficient diet consumption resulted in depletion of plasma folate in both age groups. In young folate-deficient rats, liver and lung choline levels were significantly lower than those in the respective controls. No other significant effects of FD on choline and acetylcholine metabolism were found in young rats. In adult rats, FD consumption markedly decreased choline levels in the liver, kidneys, and heart; furthermore, choline levels in the cortex and striatum were moderately elevated, although hippocampal choline levels were not affected. Acetylcholine levels were higher in the heart, cortex, and striatum but lower in the hippocampus in adult folate-deficient rats, as compared to controls. Higher acetylcholine levels in the striatum in adult folate-deficient rats were also associated with higher dopamine release in the striatal slices. Thus, both age groups showed higher cholinergic metabolic sensitivity to FD in the peripheral nervous system than in the brain. However, compensatory abilities appeared to be better in the young group, implicating the adult group as a preferred model for further investigation of folate-choline-acetylcholine interactions and their role in brain plasticity and cognitive functions.


Assuntos
Acetilcolina/metabolismo , Encéfalo/metabolismo , Deficiência de Colina/metabolismo , Colina/metabolismo , Deficiência de Ácido Fólico/metabolismo , Sistema Nervoso Periférico/metabolismo , Envelhecimento/metabolismo , Animais , Dieta , Dopamina/metabolismo , Ácido Fólico/administração & dosagem , Ácido Fólico/sangue , Rim/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley , Complexo Vitamínico B/administração & dosagem , Complexo Vitamínico B/sangue
12.
FASEB J ; 24(8): 2752-61, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20371614

RESUMO

Choline dehydrogenase (CHDH) catalyzes the conversion of choline to betaine, an important methyl donor and organic osmolyte. We have previously identified single nucleotide polymorphisms (SNPs) in the human CHDH gene that, when present, seem to alter the activity of the CHDH enzyme. These SNPs occur frequently in humans. We created a Chdh(-/-) mouse to determine the functional effects of mutations that result in decreased CHDH activity. Chdh deletion did not affect fetal viability or alter growth or survival of these mice. Only one of eleven Chdh(-/-) males was able to reproduce. Loss of CHDH activity resulted in decreased testicular betaine and increased choline and PCho concentrations. Chdh(+/+) and Chdh(-/-) mice produced comparable amounts of sperm; the impaired fertility was due to diminished sperm motility in the Chdh(-/-) males. Transmission electron microscopy revealed abnormal mitochondrial morphology in Chdh(-/-) sperm. ATP content, total mitochondrial dehydrogenase activity and inner mitochondrial membrane polarization were all significantly reduced in sperm from Chdh(-/-) animals. Mitochondrial changes were also detected in liver, kidney, heart, and testis tissues. We suggest that men who have SNPs in CHDH that decrease the activity of the CHDH enzyme could have decreased sperm motility and fertility.


Assuntos
Colina Desidrogenase/deficiência , Motilidade dos Espermatozoides , Animais , Betaína/análise , Colina/análise , Colina Desidrogenase/genética , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/patologia , Mitocôndrias/ultraestrutura , Mutação , Polimorfismo de Nucleotídeo Único , Testículo/química
13.
Brain Res ; 1237: 153-66, 2008 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-18778697

RESUMO

Altered dietary choline availability early in life leads to persistent changes in spatial memory and hippocampal plasticity in adulthood. Developmental programming by early choline nutrition may determine the range of adult choline intake that is optimal for the types of neural plasticity involved in cognitive function. To test this, male Sprague-Dawley rats were exposed to a choline chloride deficient (DEF), sufficient (CON), or supplemented (SUP) diet during embryonic days 12-17 and then returned to a control diet (1.1 g choline chloride/kg). At 70 days of age, we found that DEF and SUP rats required fewer choices to locate 8 baited arms of a 12-arm radial maze than CON rats. When switched to a choline-deficient diet (0 g/kg), SUP rats showed impaired performance while CON and DEF rats were unaffected. In contrast, when switched to a choline-supplemented diet (5.0 g/kg), DEF rats' performance was significantly impaired while CON and SUP rats were less affected. These changes in performance were reversible when the rats were switched back to a control diet. In a second experiment, DEF, CON, and SUP rats were either maintained on a control diet, or the choline-supplemented diet. After 12 weeks, DEF rats were significantly impaired by choline supplementation on a matching-to-place water-maze task, which was also accompanied by a decrease in dentate cell proliferation in DEF rats only. IGF-1 levels were elevated by both prenatal and adult choline supplementation. Taken together, these findings suggest that the in utero availability of an essential nutrient, choline, causes differential behavioral and neuroplastic sensitivity to the adult choline supply.


Assuntos
Colina/administração & dosagem , Hipocampo/efeitos dos fármacos , Memória/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Nootrópicos/administração & dosagem , Fenômenos Fisiológicos da Nutrição Pré-Natal , Animais , Comportamento Animal , Bromodesoxiuridina/metabolismo , Proliferação de Células/efeitos dos fármacos , Deficiência de Colina/patologia , Deficiência de Colina/fisiopatologia , Suplementos Nutricionais , Feminino , Hipocampo/fisiologia , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Gravidez , Ratos , Ratos Sprague-Dawley , Percepção Espacial/efeitos dos fármacos , Percepção Espacial/fisiologia
14.
Brain Res ; 1237: 110-23, 2008 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-18786518

RESUMO

Supplemental choline in the maternal diet produces a lasting enhancement in memory in offspring that resists age-related decline and is accompanied by neuroanatomical, neurophysiological and neurochemical changes in the hippocampus. The present study was designed to examine: 1) if prenatal choline supplementation alters behaviors that contribute to risk or resilience in cognitive aging, and 2) whether, at old age (25 months), prenatally choline-supplemented rats show evidence of preserved hippocampal plasticity. A longitudinal design was used to look at exploration of an open field, with and without objects, at 1 and 24 months of age in male and female rats whose mothers were fed a diet supplemented with choline (SUP; 5 mg/kg choline chloride) or not supplemented (CON; 1.1 mg/kg choline chloride) on embryonic days 12-17. Aging caused a significant decline in open field exploration that was more pronounced in males but interest in novel objects was maintained in both sexes. Prenatal choline supplementation attenuated, but did not prevent age-related decline in exploration in males and increased object exploration in young females. Following behavioral assessment, rats were euthanized to assess markers of hippocampal plasticity. Aged SUP males and females had more newly proliferated cells in the hippocampal dentate gyrus and protein levels of vascular endothelial growth factor (VEGF) and neurotrophin-3 (NT-3) were significantly elevated in female SUP rats in comparison to all other groups. Taken together, these findings provide the first evidence that prenatal choline supplementation causes changes in exploratory behaviors over the lifespan and preserves some features of hippocampal plasticity that can be seen even at 2 years of age.


Assuntos
Colina/administração & dosagem , Comportamento Exploratório/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Nootrópicos/administração & dosagem , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Comportamento Animal , Bromodesoxiuridina/metabolismo , Proliferação de Células , Corticosterona/farmacologia , Comportamento Exploratório/fisiologia , Feminino , Hipocampo/fisiologia , Masculino , Aprendizagem em Labirinto , Plasticidade Neuronal/fisiologia , Gravidez , Ratos , Ratos Sprague-Dawley , Fatores Sexuais , Estresse Psicológico/tratamento farmacológico
15.
J Cell Biochem ; 104(5): 1818-31, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18348264

RESUMO

Muscarinic receptors subserve many functions in both peripheral and central nervous systems. Some of these processes depend on increases in protein synthesis, which may be achieved by activation of mammalian target of rapamycin (mTOR), a kinase that regulates protein translation capacity. Here, we examined the regulation of mTOR-dependent signaling pathways by muscarinic receptors in SK-N-SH human neuroblastoma cells, and in human embryonic kidney (HEK) cell lines transfected with individual muscarinic receptor subtypes. In SK-N-SH cells, the acetylcholine analog carbachol stimulated phosphorylation of the ribosomal S6 protein, a downstream target of mTOR. The sensitivity of the response to subtype-selective muscarinic receptor antagonists indicated that it was mediated by M3 receptors. Carbachol-evoked S6 phosphorylation was blocked by the mTOR inhibitor rapamycin, but was independent of phosphoinositide 3-kinase activation. The response was significantly reduced by the mitogen-activated protein kinase kinase (MEK) inhibitor U0126, which also inhibited carbachol-evoked S6 phosphorylation in HEK cells expressing M2 receptors, but was ineffective in M3 receptor-expressing HEK cells, although carbachol activated MAPK in both transfected lines. The p90 ribosomal S6 kinase has been implicated in mTOR regulation by phorbol esters, but was not activated by carbachol in any of the cell lines tested. The protein kinase C inhibitor bisindolylmaleimide I reduced carbachol-stimulated S6 phosphorylation in SK-N-SH cells, and in HEK cells expressing M3 receptors, but not in HEK cells expressing M2 receptors. The results demonstrate that multiple muscarinic receptor subtypes regulate mTOR, and that both MAPK-dependent and -independent mechanisms may mediate the response in a cell context-specific manner.


Assuntos
Proteínas Quinases/metabolismo , Receptores Muscarínicos/metabolismo , Proteína S6 Ribossômica/metabolismo , Carbacol/farmacologia , Linhagem Celular Tumoral , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Quinase C/metabolismo , Receptor Muscarínico M2/metabolismo , Receptor Muscarínico M3/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Serina-Treonina Quinases TOR , Acetato de Tetradecanoilforbol/farmacologia , Transfecção
16.
Neurobiol Dis ; 30(2): 255-69, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18353663

RESUMO

Prenatal choline supplementation (SUP) protects adult rats against spatial memory deficits observed after excitotoxin-induced status epilepticus (SE). To examine the mechanism underlying this neuroprotection, we determined the effects of SUP on a variety of hippocampal markers known to change in response to SE and thought to underlie ensuing cognitive deficits. Adult offspring from rat dams that received either a control or SUP diet on embryonic days 12-17 were administered saline or kainic acid (i.p.) to induce SE and were euthanized 16 days later. SUP markedly attenuated seizure-induced hippocampal neurodegeneration, dentate cell proliferation, and hippocampal GFAP mRNA expression levels, prevented the loss of hippocampal GAD65 protein and mRNA expression, and altered growth factor expression patterns. SUP also enhanced pre-seizure hippocampal levels of BDNF, NGF, and IGF-1, which may confer a neuroprotective hippocampal microenvironment that dampens the neuropathological response to and/or helps facilitate recovery from SE to protect cognitive function.


Assuntos
Colina/administração & dosagem , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Cuidado Pré-Natal/métodos , Estado Epiléptico/patologia , Estado Epiléptico/prevenção & controle , Animais , Feminino , Masculino , Gravidez , Fenômenos Fisiológicos da Nutrição Pré-Natal/fisiologia , Ratos , Ratos Sprague-Dawley , Estado Epiléptico/dietoterapia
17.
J Biol Chem ; 282(43): 31777-88, 2007 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-17724018

RESUMO

During gestation there is a high demand for the essential nutrient choline. Adult rats supplemented with choline during embryonic days (E) 11-17 have improved memory performance and do not exhibit age-related memory decline, whereas prenatally choline-deficient animals have memory deficits. Choline, via betaine, provides methyl groups for the production of S-adenosylmethionine, a substrate of DNA methyltransferases (DNMTs). We describe an apparently adaptive epigenomic response to varied gestational choline supply in rat fetal liver and brain. S-Adenosylmethionine levels increased in both organs of E17 fetuses whose mothers consumed a choline-supplemented diet. Surprisingly, global DNA methylation increased in choline-deficient animals, and this was accompanied by overexpression of Dnmt1 mRNA. Previous studies showed that the prenatal choline supply affects the expression of multiple genes, including insulin-like growth factor 2 (Igf2), whose expression is regulated in a DNA methylation-dependent manner. The differentially methylated region 2 of Igf2 was hypermethylated in the liver of E17 choline-deficient fetuses, and this as well as Igf2 mRNA levels correlated with the expression of Dnmt1 and with hypomethylation of a regulatory CpG within the Dnmt1 locus. Moreover, mRNA expression of brain and liver Dnmt3a and methyl CpG-binding domain 2 (Mbd2) protein as well as cerebral Dnmt3l was inversely correlated to the intake of choline. Thus, choline deficiency modulates fetal DNA methylation machinery in a complex fashion that includes hypomethylation of the regulatory CpGs within the Dnmt1 gene, leading to its overexpression and the resultant increased global and gene-specific (e.g. Igf2) DNA methylation. These epigenomic responses to gestational choline supply may initiate the long term developmental changes observed in rats exposed to varied choline intake in utero.


Assuntos
Deficiência de Colina/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Regulação da Expressão Gênica no Desenvolvimento/genética , Fator de Crescimento Insulin-Like II/genética , Animais , Estudos de Coortes , DNA (Citosina-5-)-Metiltransferase 1 , Feminino , Inativação Gênica , Fator de Crescimento Insulin-Like II/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima
18.
Eur J Neurosci ; 25(8): 2473-82, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17445242

RESUMO

Increased dietary intake of choline early in life improves performance of adult rats on memory tasks and prevents their age-related memory decline. Because neurogenesis in the adult hippocampus also declines with age, we investigated whether prenatal choline availability affects hippocampal neurogenesis in adult Sprague-Dawley rats and modifies their neurogenic response to environmental stimulation. On embryonic days (ED) 12-17, pregnant rats ate a choline-supplemented (SUP-5 g/kg), choline sufficient (SFF-1.1 g/kg), or choline-free (DEF) semisynthetic diet. Adult offspring either remained in standard housing or were given 21 daily visits to explore a maze. On the last ten exploration days, all rats received daily injections of 5-bromo-2-deoxyuridine (BrdU, 100 mg/kg). The number of BrdU+ cells was significantly greater in the dentate gyrus in SUP rats compared to SFF or DEF rats. While maze experience increased the number of BrdU+ cells in SFF rats to the level seen in the SUP rats, this enriching experience did not alter cell proliferation in DEF rats. Similar patterns of cell proliferation were obtained with immunohistochemical staining for neuronal marker doublecortin, confirming that diet and exploration affected hippocampal neurogenesis. Moreover, hippocampal levels of the brain-derived neurotrophic factor (BDNF) were increased in SUP rats as compared to SFF and DEF animals. We conclude that prenatal choline intake has enduring effects on adult hippocampal neurogenesis, possibly via up-regulation of BDNF levels, and suggest that these alterations of neurogenesis may contribute to the mechanism of life-long changes in cognitive function governed by the availability of choline during gestation.


Assuntos
Colina/metabolismo , Dieta , Hipocampo/crescimento & desenvolvimento , Neurônios/fisiologia , Efeitos Tardios da Exposição Pré-Natal , Animais , Comportamento Animal/fisiologia , Bromodesoxiuridina/metabolismo , Colina/administração & dosagem , Suplementos Nutricionais , Proteína Duplacortina , Comportamento Exploratório/fisiologia , Feminino , Hipocampo/citologia , Hipocampo/fisiologia , Masculino , Neurônios/citologia , Gravidez , Ratos , Ratos Sprague-Dawley
19.
Brain Res ; 1088(1): 49-56, 2006 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-16626664

RESUMO

Basal forebrain cholinergic neurons play critical roles in the organization of brain cortical structures and in processes such as learning and memory. We have previously shown that bone morphogenetic protein (BMP) 9, a member of the transforming growth factor (TGF) beta superfamily of cytokines, is a differentiating factor for cholinergic central nervous system neurons. However, whereas the basic signal transduction pathways for most known members of the TGF-beta superfamily have been well characterized in brain and other organs, nothing is known about the signal transduction pathway of BMP9 in the brain. Here, we describe the pattern of expression of BMP receptors, including Bmpr-Ia, Bmpr-Ib, Bmpr-II, Actr-I. Actr-Ib, Actr-II and Actr-IIb, Alk-1, and Smad proteins (Smads 1-5 and Smad8) in the septal region of the basal forebrain during mouse development. Using cultured basal forebrain cells derived from embryonic day (E) 14 mice, we show that BMP9 causes phosphorylation of Smad1 and Smad5, formation of a complex of Smad4 with Samd1 and/or Smad5, and translocation of these proteins into the nucleus. These data show that BMP9 activates the canonical BMP signaling pathway and suggest that this could be one of the mechanisms responsible for the induction of the cholinergic phenotype by BMP9 in the basal forebrain.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Prosencéfalo/fisiologia , Proteínas Smad/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting/métodos , Receptores de Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/farmacologia , Células Cultivadas , Embrião de Mamíferos , Ativação Enzimática/efeitos dos fármacos , Fator 2 de Diferenciação de Crescimento , Camundongos , Prosencéfalo/citologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Proteínas Smad/genética
20.
J Cell Biochem ; 98(3): 672-84, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16440311

RESUMO

The discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase that is highly expressed in breast carcinoma cells. Upon binding to collagen, DDR1 undergoes autophosphorylation followed by limited proteolysis to generate a tyrosine phosphorylated C-terminal fragment (CTF). Although it was postulated that this fragment is formed as a result of shedding of the N-terminal ectodomain, collagen-dependent release of the DDR1 extracellular domain has not been demonstrated. We now report that, in conjunction with CTF formation, collagen type I stimulates concentration-dependent, saturable shedding of the DDR1 ectodomain from two carcinoma cell lines, and from transfected cells. In contrast, collagen did not promote cleavage of other transmembrane proteins including the amyloid precursor protein (APP), ErbB2, and E-cadherin. Collagen-dependent tyrosine phosphorylation and proteolysis of DDR1 in carcinoma cells were reduced by a pharmacologic Src inhibitor. Moreover, expression of a dominant negative Src mutant protein in human embryonic kidney cells inhibited collagen-dependent phosphorylation and shedding of co-transfected DDR1. The hydroxamate-based metalloproteinase inhibitor TAPI-1 (tumor necrosis factor-alpha protease inhibitor-1), and tissue inhibitor of metalloproteinase (TIMP)-3, also blocked collagen-evoked DDR1 shedding, but did not reduce levels of the phosphorylated CTF. Neither shedding nor CTF formation were affected by the gamma-secretase inhibitor, L-685,458. The results demonstrate that collagen-evoked ectodomain cleavage of DDR1 is mediated in part by Src-dependent activation or recruitment of a matrix- or disintegrin metalloproteinase, and that CTF formation can occur independently of ectodomain shedding. Delayed shedding of the DDR1 ectodomain may represent a mechanism that limits DDR1-dependent cell adhesion and migration on collagen matrices.


Assuntos
Colágeno Tipo I/farmacologia , Receptores Proteína Tirosina Quinases/química , Receptores Proteína Tirosina Quinases/metabolismo , Quinases da Família src/metabolismo , Secretases da Proteína Precursora do Amiloide , Ácido Aspártico Endopeptidases , Células Cultivadas , Receptor com Domínio Discoidina 1 , Endopeptidases/metabolismo , Humanos , Fosfotirosina/metabolismo , Estrutura Terciária de Proteína/efeitos dos fármacos , Fatores de Tempo , Inibidor Tecidual de Metaloproteinase-3/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...