Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 142(1): 52-64, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20603014

RESUMO

Cancer is influenced by its microenvironment, yet broader, environmental effects also play a role but remain poorly defined. We report here that mice living in an enriched housing environment show reduced tumor growth and increased remission. We found this effect in melanoma and colon cancer models, and that it was not caused by physical activity alone. Serum from animals held in an enriched environment (EE) inhibited cancer proliferation in vitro and was markedly lower in leptin. Hypothalamic brain-derived neurotrophic factor (BDNF) was selectively upregulated by EE, and its genetic overexpression reduced tumor burden, whereas BDNF knockdown blocked the effect of EE. Mechanistically, we show that hypothalamic BDNF downregulated leptin production in adipocytes via sympathoneural beta-adrenergic signaling. These results suggest that genetic or environmental activation of this BDNF/leptin axis may have therapeutic significance for cancer.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Neoplasias do Colo/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Melanoma/metabolismo , Transdução de Sinais , Meio Social , Adipócitos/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Neoplasias do Colo/genética , Neoplasias do Colo/fisiopatologia , Genes APC , Abrigo para Animais , Hipotálamo/citologia , Imunocompetência , Melanoma/genética , Melanoma/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Processos Neoplásicos , Distribuição Aleatória , Receptores Adrenérgicos beta/metabolismo
2.
Epilepsia ; 62(12): 3091-3104, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34617595

RESUMO

OBJECTIVE: An attractive target to interfere with epileptic brain hyperexcitability is the enhancement of γ-aminobutyric acidergic (GABAergic) inhibition by inactivation of the GABA-metabolizing enzyme GABA aminotransferase (GABA-AT). GABA-AT inactivators were designed to control seizures by raising brain GABA levels. OV329, a novel drug candidate for the treatment of epilepsy and addiction, has been shown in vitro to be substantially more potent as a GABA-AT inactivator than vigabatrin, an antiseizure drug approved as an add-on therapy for adult patients with refractory complex partial seizures and monotherapy for pediatric patients with infantile spasms. Thus, we hypothesized that OV329 should produce pronounced anticonvulsant effects in two different rat seizure models. METHODS: We therefore examined the effects of OV329 (5, 20, and 40 mg/kg ip) on the seizure threshold of female Wistar Unilever rats, using the timed intravenous pentylenetetrazole (ivPTZ) seizure threshold model as a seizure test particularly sensitive to GABA-potentiating manipulations, and amygdala-kindled rats as a model of difficult-to-treat temporal lobe epilepsy. RESULTS: GABA-AT inactivation by OV329 clearly increased the threshold of both ivPTZ-induced and amygdala-kindled seizures. OV329 further showed a 30-fold greater anticonvulsant potency on ivPTZ-induced myoclonic jerks and clonic seizures compared to vigabatrin investigated previously. Notably, all rats were responsive to OV329 in both seizure models. SIGNIFICANCE: These results reveal an anticonvulsant profile of OV329 that appears to be superior in both potency and efficacy to vigabatrin and highlight OV329 as a highly promising candidate for the treatment of seizures and pharmacoresistant epilepsies.


Assuntos
Epilepsia , Excitação Neurológica , Tonsila do Cerebelo , Animais , Anticonvulsivantes/efeitos adversos , Epilepsia/tratamento farmacológico , Feminino , Humanos , Excitação Neurológica/fisiologia , Pentilenotetrazol/efeitos adversos , Ratos , Ratos Wistar , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Transaminases/efeitos adversos , Vigabatrina/efeitos adversos , Ácido gama-Aminobutírico/farmacologia
3.
Epilepsia ; 62(11): 2845-2857, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34510432

RESUMO

OBJECTIVE: Dravet syndrome is a severe developmental and epileptic encephalopathy (DEE) most often caused by de novo pathogenic variants in SCN1A. Individuals with Dravet syndrome rarely achieve seizure control and have significantly elevated risk for sudden unexplained death in epilepsy (SUDEP). Heterozygous deletion of Scn1a in mice (Scn1a+/- ) recapitulates several core phenotypes, including temperature-dependent and spontaneous seizures, SUDEP, and behavioral abnormalities. Furthermore, Scn1a+/- mice exhibit a similar clinical response to standard anticonvulsants. Cholesterol 24-hydroxlase (CH24H) is a brain-specific enzyme responsible for cholesterol catabolism. Recent research has indicated the therapeutic potential of CH24H inhibition for diseases associated with neural excitation, including seizures. METHODS: In this study, the novel compound soticlestat, a CH24H inhibitor, was administered to Scn1a+/- mice to investigate its ability to improve Dravet-like phenotypes in this preclinical model. RESULTS: Soticlestat treatment reduced seizure burden, protected against hyperthermia-induced seizures, and completely prevented SUDEP in Scn1a+/- mice. Video-electroencephalography (EEG) analysis confirmed the ability of soticlestat to reduce occurrence of electroclinical seizures. SIGNIFICANCE: This study demonstrates that soticlestat-mediated inhibition of CH24H provides therapeutic benefit for the treatment of Dravet syndrome in mice and has the potential for treatment of DEEs.


Assuntos
Epilepsias Mioclônicas , Epilepsia , Piperidinas , Piridinas , Convulsões Febris , Morte Súbita Inesperada na Epilepsia , Animais , Colesterol 24-Hidroxilase/antagonistas & inibidores , Epilepsias Mioclônicas/complicações , Epilepsias Mioclônicas/tratamento farmacológico , Epilepsias Mioclônicas/genética , Epilepsia/genética , Síndromes Epilépticas , Camundongos , Mortalidade Prematura , Mutação , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Piperidinas/farmacologia , Piridinas/farmacologia , Convulsões/etiologia , Convulsões/genética , Convulsões Febris/tratamento farmacológico , Morte Súbita Inesperada na Epilepsia/etiologia
4.
Annu Rev Neurosci ; 35: 331-45, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22462541

RESUMO

A focus of much cancer research is at the molecular and cellular levels. In contrast, the effects of social interactions and psychological state are less investigated, and considered by many a "soft" science. Yet several highly rigorous studies have begun to tease out biochemical pathways by which the brain can influence the development and growth of cancer. Previous reviews have discussed the concept of stress and cancer. Here, we discuss recent work showing environments that are more complex and challenging, but not stressful per se, and that have robust effects on peripheral cancer by activating a specific neuroendocrine brain-adipocyte axis. These enriched environments lead to activation of the sympathetic innervation of fat tissue, suppression of leptin, and a reduction in cancer proliferation by inducing hypothalamic BDNF expression. We summarize this work and discuss how these data integrate into the body of literature regarding stress, the environment, and cancer.


Assuntos
Adipócitos/fisiologia , Encéfalo/fisiologia , Neoplasias/fisiopatologia , Animais , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Meio Ambiente , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Leptina/metabolismo , Solidão , Modelos Biológicos , Obesidade/fisiopatologia , Transdução de Sinais/fisiologia , Apoio Social , Estresse Psicológico/fisiopatologia , Sistema Nervoso Simpático/fisiologia
5.
Proc Natl Acad Sci U S A ; 114(42): 11211-11216, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-28973921

RESUMO

Optogenetic strategies to restore vision in patients who are blind from end-stage retinal degenerations aim to render remaining retinal cells light sensitive once photoreceptors are lost. Here, we assessed long-term functional outcomes following subretinal delivery of the human melanopsin gene (OPN4) in the rd1 mouse model of retinal degeneration using an adeno-associated viral vector. Ectopic expression of OPN4 using a ubiquitous promoter resulted in cellular depolarization and ganglion cell action potential firing. Restoration of the pupil light reflex, behavioral light avoidance, and the ability to perform a task requiring basic image recognition were restored up to 13 mo following injection. These data suggest that melanopsin gene therapy via a subretinal route may be a viable and stable therapeutic option for the treatment of end-stage retinal degeneration in humans.


Assuntos
Terapia Genética/métodos , Degeneração Retiniana/terapia , Opsinas de Bastonetes/genética , Animais , Dependovirus , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C3H , Visão Ocular
6.
Int J Mol Sci ; 21(21)2020 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-33182365

RESUMO

Ischaemic brain damage induces autoimmune responses, including the production of autoantibodies with potential neuroprotective effects. Platelets share unexplained similarities with neurons, and the formation of anti-platelet antibodies has been documented in neurological disorders. The aim of this study was to investigate the presence of anti-platelet antibodies in the peripheral blood of patients after ischaemic stroke and determine any clinical correlations. Using a flow cytometry-based platelet immunofluorescence method, we detected platelet-reactive antibodies in 15 of 48 (31%) stroke patients and two of 50 (4%) controls (p < 0.001). Western blotting revealed heterogeneous reactivities with platelet proteins, some of which overlapped with brain proteins. Stroke patients who carried anti-platelet antibodies presented with larger infarcts and more severe neurological dysfunction, which manifested as higher scores on the National Institutes of Health Stroke Scale (NIHSS; p = 0.009), but they had a greater recovery in the NIHSS by the time of hospital discharge (day 7 ± 2) compared with antibody-negative patients (p = 0.043). Antibodies from stroke sera reacted more strongly with activated platelets (p = 0.031) and inhibited platelet aggregation by up to 30.1 ± 2.8% (p < 0.001), suggesting the potential to interfere with thrombus formation. In conclusion, platelet-reactive antibodies can be found in patients soon after ischaemic stroke and correlate with better short-term outcomes, suggesting a potential novel mechanism limiting thrombosis.


Assuntos
Autoanticorpos/imunologia , Plaquetas/imunologia , Isquemia Encefálica/imunologia , AVC Isquêmico/imunologia , Idoso , Autoimunidade/imunologia , Coagulação Sanguínea/imunologia , Feminino , Humanos , Masculino , Agregação Plaquetária/imunologia , Contagem de Plaquetas/métodos , Trombose/imunologia
7.
J Neurosci ; 37(28): 6729-6740, 2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28607169

RESUMO

Dysregulation of cell cycle machinery is implicated in a number of neuronal death contexts, including stroke. Increasing evidence suggests that cyclin-dependent kinases (Cdks) are inappropriately activated in mature neurons under ischemic stress conditions. We previously demonstrated a functional role for the cyclin D1/Cdk4/pRb (retinoblastoma tumor suppressor protein) pathway in delayed neuronal death induced by ischemia. However, the molecular signals leading to cyclin D/Cdk4/pRb activation following ischemic insult are presently not clear. Here, we investigate the cell division cycle 25 (Cdc25) dual-specificity phosphatases as potential upstream regulators of ischemic neuronal death and Cdk4 activation. We show that a pharmacologic inhibitor of Cdc25 family members (A, B, and C) protects mouse primary neurons from hypoxia-induced delayed death. The major contributor to the death process appears to be Cdc25A. shRNA-mediated knockdown of Cdc25A protects neurons in a delayed model of hypoxia-induced death in vitro Similar results were observed in vivo following global ischemia in the rat. In contrast, neurons singly or doubly deficient for Cdc25B/C were not significantly protective. We show that Cdc25A activity, but not level, is upregulated in vitro following hypoxia and global ischemic insult in vivo Finally, we show that shRNA targeting Cdc25A blocks Ser795 pRb phosphorylation. Overall, our results indicate a role for Cdc25A in delayed neuronal death mediated by ischemia.SIGNIFICANCE STATEMENT A major challenge in stroke is finding an effective neuroprotective strategy to treat cerebral ischemic injury. Cdc25 family member A (Cdc25A) is a phosphatase normally activated during cell division in proliferating cells. We found that Cdc25A is activated in neurons undergoing ischemic stress mediated by hypoxia in vitro and global cerebral ischemia in rats in vivo We show that pharmacologic or genetic inhibition of Cdc25A activity protects neurons from delayed death in vitro and in vivo Downregulation of Cdc25A led to reduction in retinoblastoma tumor suppressor protein (pRb) phosphorylation. An increase in pRb phosphorylation has been previously linked to ischemic neuronal death. Our results identify Cdc25A as a potential target for neuroprotectant strategy for the treatment of delayed ischemic neuronal death.


Assuntos
Apoptose , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosfatases cdc25/metabolismo , Animais , Células Cultivadas , Ativação Enzimática , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley
8.
Platelets ; 28(8): 799-811, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28277064

RESUMO

GluN1 is a mandatory component of N-methyl-D-aspartate receptors (NMDARs) best known for their roles in the brain, but with increasing evidence for relevance in peripheral tissues, including platelets. Certain anti-GluN1 antibodies reduce brain infarcts in rodent models of ischaemic stroke. There is also evidence that human anti-GluN1 autoantibodies reduce neuronal damage in stroke patients, but the underlying mechanism is unclear. This study investigated whether anti-GluN1-mediated neuroprotection involves inhibition of platelet function. Four commercial anti-GluN1 antibodies were screened for their abilities to inhibit human platelet aggregation. Haematological parameters were examined in rats vaccinated with GluN1. Platelet effects of a mouse monoclonal antibody targeting the glycine-binding region of GluN1 (GluN1-S2) were tested in assays of platelet activation, aggregation and thrombus formation. The epitope of anti-GluN1-S2 was mapped and the mechanism of antibody action modelled using crystal structures of GluN1. Our work found that rats vaccinated with GluN1 had a mildly prolonged bleeding time and carried antibodies targeting mostly GluN1-S2. The monoclonal anti-GluN1-S2 antibody (from BD Biosciences) inhibited activation and aggregation of human platelets in the presence of adrenaline, adenosine diphosphate, collagen, thrombin and a protease-activated receptor 1-activating peptide. When human blood was flowed over collagen-coated surfaces, anti-GluN1-S2 impaired thrombus growth and stability. The epitope of anti-GluN1-S2 was mapped to α-helix H located within the glycine-binding clamshell of GluN1, where the antibody binding was computationally predicted to impair opening of the NMDAR channel. Our results indicate that anti-GluN1-S2 inhibits function of human platelets, including dense granule release and thrombus growth. Findings add to the evidence that platelet NMDARs regulate thrombus formation and suggest a novel mechanism by which anti-GluN1 autoantibodies limit stroke-induced neuronal damage.


Assuntos
Autoanticorpos/sangue , Plaquetas/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Trombose/genética , Animais , Humanos , Masculino , Ratos , Ratos Wistar
9.
J Biol Chem ; 289(26): 18202-13, 2014 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-24828495

RESUMO

Inappropriate activation of cell cycle proteins, in particular cyclin D/Cdk4, is implicated in neuronal death induced by various pathologic stresses, including DNA damage and ischemia. Key targets of Cdk4 in proliferating cells include members of the E2F transcription factors, which mediate the expression of cell cycle proteins as well as death-inducing genes. However, the presence of multiple E2F family members complicates our understanding of their role in death. We focused on whether E2F4, an E2F member believed to exhibit crucial control over the maintenance of a differentiated state of neurons, may be critical in ischemic neuronal death. We observed that, in contrast to E2F1 and E2F3, which sensitize to death, E2F4 plays a crucial protective role in neuronal death evoked by DNA damage, hypoxia, and global ischemic insult both in vitro and in vivo. E2F4 occupies promoter regions of proapoptotic factors, such as B-Myb, under basal conditions. Following stress exposure, E2F4-p130 complexes are lost rapidly along with the presence of E2F4 at E2F-containing B-Myb promoter sites. In contrast, the presence of E2F1 at B-Myb sites increases with stress. Furthermore, B-Myb and C-Myb expression increases with ischemic insult. Taken together, we propose a model by which E2F4 plays a protective role in neurons from ischemic insult by forming repressive complexes that prevent prodeath factors such as Myb from being expressed.


Assuntos
Fator de Transcrição E2F4/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Neurônios/citologia , Proteína p130 Retinoblastoma-Like/metabolismo , Animais , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Morte Celular , Fator de Transcrição E2F4/genética , Humanos , Hipóxia-Isquemia Encefálica/genética , Hipóxia-Isquemia Encefálica/fisiopatologia , Masculino , Camundongos Knockout , Neurônios/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Ratos Wistar , Proteína p130 Retinoblastoma-Like/genética , Transativadores/genética , Transativadores/metabolismo
10.
Lancet ; 383(9923): 1129-37, 2014 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-24439297

RESUMO

BACKGROUND: Choroideremia is an X-linked recessive disease that leads to blindness due to mutations in the CHM gene, which encodes the Rab escort protein 1 (REP1). We assessed the effects of retinal gene therapy with an adeno-associated viral (AAV) vector encoding REP1 (AAV.REP1) in patients with this disease. METHODS: In a multicentre clinical trial, six male patients (aged 35-63 years) with choroideremia were administered AAV.REP1 (0·6-1·0×10(10) genome particles, subfoveal injection). Visual function tests included best corrected visual acuity, microperimetry, and retinal sensitivity tests for comparison of baseline values with 6 months after surgery. This study is registered with ClinicalTrials.gov, number NCT01461213. FINDINGS: Despite undergoing retinal detachment, which normally reduces vision, two patients with advanced choroideremia who had low baseline best corrected visual acuity gained 21 letters and 11 letters (more than two and four lines of vision). Four other patients with near normal best corrected visual acuity at baseline recovered to within one to three letters. Mean gain in visual acuity overall was 3·8 letters (SE 4·1). Maximal sensitivity measured with dark-adapted microperimetry increased in the treated eyes from 23·0 dB (SE 1·1) at baseline to 25·3 dB (1·3) after treatment (increase 2·3 dB [95% CI 0·8-3·8]). In all patients, over the 6 months, the increase in retinal sensitivity in the treated eyes (mean 1·7 [SE 1·0]) was correlated with the vector dose administered per mm(2) of surviving retina (r=0·82, p=0·04). By contrast, small non-significant reductions (p>0·05) were noted in the control eyes in both maximal sensitivity (-0·8 dB [1·5]) and mean sensitivity (-1·6 dB [0·9]). One patient in whom the vector was not administered to the fovea re-established variable eccentric fixation that included the ectopic island of surviving retinal pigment epithelium that had been exposed to vector. INTERPRETATION: The initial results of this retinal gene therapy trial are consistent with improved rod and cone function that overcome any negative effects of retinal detachment. These findings lend support to further assessment of gene therapy in the treatment of choroideremia and other diseases, such as age-related macular degeneration, for which intervention should ideally be applied before the onset of retinal thinning. FUNDING: UK Department of Health and Wellcome Trust.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/administração & dosagem , Coroideremia/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenoviridae/genética , Adulto , Idoso , Coroideremia/fisiopatologia , Fluorescência , Técnicas de Transferência de Genes , Humanos , Injeções Intraoculares , Masculino , Pessoa de Meia-Idade , Descolamento Retiniano/fisiopatologia , Descolamento Retiniano/terapia , Transgenes/genética , Acuidade Visual/fisiologia
11.
Nat Med ; 13(11): 1333-40, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17982462

RESUMO

Anorexia and weight loss are part of the wasting syndrome of late-stage cancer, are a major cause of morbidity and mortality in cancer, and are thought to be cytokine mediated. Macrophage inhibitory cytokine-1 (MIC-1) is produced by many cancers. Examination of sera from individuals with advanced prostate cancer showed a direct relationship between MIC-1 abundance and cancer-associated weight loss. In mice with xenografted prostate tumors, elevated MIC-1 levels were also associated with marked weight, fat and lean tissue loss that was mediated by decreased food intake and was reversed by administration of antibody to MIC-1. Additionally, normal mice given systemic MIC-1 and transgenic mice overexpressing MIC-1 showed hypophagia and reduced body weight. MIC-1 mediates its effects by central mechanisms that implicate the hypothalamic transforming growth factor-beta receptor II, extracellular signal-regulated kinases 1 and 2, signal transducer and activator of transcription-3, neuropeptide Y and pro-opiomelanocortin. Thus, MIC-1 is a newly defined central regulator of appetite and a potential target for the treatment of both cancer anorexia and weight loss, as well as of obesity.


Assuntos
Anorexia/metabolismo , Citocinas/fisiologia , Família Multigênica/imunologia , Neoplasias da Próstata/metabolismo , Redução de Peso , Animais , Anorexia/genética , Anorexia/imunologia , Anorexia/fisiopatologia , Anticorpos/administração & dosagem , Anticorpos/fisiologia , Linhagem Celular Tumoral , Citocinas/sangue , Citocinas/genética , Citocinas/imunologia , Fator 15 de Diferenciação de Crescimento , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Neoplasias da Próstata/sangue , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/fisiopatologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/fisiologia , Redução de Peso/genética , Redução de Peso/imunologia
12.
Learn Mem ; 20(2): 75-9, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23322555

RESUMO

Narp knockout (KO) mice demonstrate an impaired extinction of morphine conditioned place preference (CPP). Because the medial prefrontal cortex (mPFC) has been implicated in extinction learning, we tested whether Narp cells in this region play a role in the extinction of morphine CPP. We found that intracranial injections of adenoassociated virus (AAV) expressing wild-type (WT) Narp into the mPFC of Narp KO mice rescued the extinction and the injection of AAV expressing a dominant negative form of Narp (NarpN) into the mPFC of WT mice impaired the extinction of morphine CPP. These findings suggest that Narp in the mPFC mediates the extinction of morphine CPP.


Assuntos
Proteína C-Reativa/metabolismo , Condicionamento Operante/fisiologia , Extinção Psicológica/fisiologia , Morfina/administração & dosagem , Entorpecentes/administração & dosagem , Proteínas do Tecido Nervoso/metabolismo , Córtex Pré-Frontal/metabolismo , Análise de Variância , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteína C-Reativa/deficiência , Dependovirus/genética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microinjeções , Proteínas do Tecido Nervoso/deficiência
13.
Stroke ; 44(8): 2212-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23723305

RESUMO

BACKGROUND AND PURPOSE: Antibodies against neuronal antigens develop in patients after stroke and some may serve as biomarkers of neuronal injury. We aimed to determine whether antibodies against subunit 1 (GluN1) of the N-methyl-D-aspartate receptor also develop after stroke and if so, whether they correlate with stroke characteristics. METHODS: Forty-eight patients with ischemic stroke and 96 healthy controls were tested for the presence of serum antibodies targeting GluN1. Testing was conducted using 20-kDa recombinant GluN1-S2 peptide (by ELISA and Western blotting) and on rat brain tissue (by Western blotting and immunohistochemistry). Clinical examinations and computed tomographic brain scans were performed to assess clinical state and infarct size and location. RESULTS: Of the 48 patients with ischemic stroke, 21 (44%) had antibodies that reacted with the recombinant GluN1-S2. There was no evidence of antibody binding to intact GluN1 in brain tissue. Western blot appearances suggested reactivity with GluN1 degradation products. Patients with anti-GluN1-S2 antibodies were more likely to have higher National Institutes of Health Stroke Scale scores, larger infarcts, and more frequent cortical involvement. Of the 96 controls, only 3 (3%), all aged>50 years, had antibodies that reacted with GluN1-S2 at low levels. CONCLUSIONS: Antibodies that bind recombinant GluN1-S2 peptides (but not the intact GluN1 protein) develop transiently in patients after stroke in proportion to infarct size, suggesting that these antibodies are raised secondarily to neuronal damage. The anti-GluN1-S2 antibodies may provide useful information about the presence and severity of cerebral infarction. This will require confirmation in larger studies.


Assuntos
Autoanticorpos/biossíntese , Proteínas do Tecido Nervoso/imunologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios , Receptores de N-Metil-D-Aspartato/imunologia , Receptores de N-Metil-D-Aspartato/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Autoanticorpos/sangue , Biomarcadores , Isquemia Encefálica/sangue , Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Feminino , Humanos , Infarto da Artéria Cerebral Média/imunologia , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Neurônios/imunologia , Neurônios/patologia , Ratos , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/patologia
14.
Behav Pharmacol ; 24(4): 332-6, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23751516

RESUMO

The medial prefrontal cortex (mPFC) plays a key role in extinction learning. Previously, we found that expression of a neuronal activity-regulated pentraxin (Narp) dominant-negative construct in the mPFC of mice blocked extinction of morphine-conditioned place preference. To further investigate the role of mPFC Narp in the extinction of drug seeking, we tested whether mPFC Narp is necessary for the extinction of heroin self-administration in rats. Specifically, we injected an adeno-associated viral vector expressing a dominant-negative form of Narp (NarpN) into the infralimbic region of the mPFC of rats and compared lever presses during extinction to those of rats injected with a control virus. In contrast to our previous study, we found that injection of NarpN did not affect extinction of heroin self-administration. Our findings suggest that mPFC Narp is necessary for extinction of opiate seeking in the Pavlovian-conditioned place preference paradigm but not in the operant paradigm of drug self-administration.


Assuntos
Proteína C-Reativa/metabolismo , Extinção Psicológica/efeitos dos fármacos , Heroína/administração & dosagem , Entorpecentes/administração & dosagem , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Córtex Pré-Frontal/citologia , Análise de Variância , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteína C-Reativa/genética , Condicionamento Clássico/efeitos dos fármacos , Dependovirus/genética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Autoadministração , Fatores de Tempo , Transdução Genética
15.
Nat Med ; 12(6): 677-81, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16715092

RESUMO

Glutamatergic signaling and intracellular calcium mobilization in the spinal cord are crucial for the development of nociceptive plasticity, which is associated with chronic pathological pain. Long-form Homer proteins anchor glutamatergic receptors to sources of calcium influx and release at synapses, which is antagonized by the short, activity-dependent splice variant Homer1a. We show here that Homer1a operates in a negative feedback loop to regulate the excitability of the pain pathway in an activity-dependent manner. Homer1a is rapidly and selectively upregulated in spinal cord neurons after peripheral inflammation in an NMDA receptor-dependent manner. Homer1a strongly attenuates calcium mobilization as well as MAP kinase activation induced by glutamate receptors and reduces synaptic contacts on spinal cord neurons that process pain inputs. Preventing activity-induced upregulation of Homer1a using shRNAs in mice in vivo exacerbates inflammatory pain. Thus, activity-dependent uncoupling of glutamate receptors from intracellular signaling mediators is a novel, endogenous physiological mechanism for counteracting sensitization at the first, crucial synapse in the pain pathway. Furthermore, we observed that targeted gene transfer of Homer1a to specific spinal segments in vivo reduces inflammatory hyperalgesia. Thus, Homer1 function is crucially involved in pain plasticity and constitutes a promising therapeutic target for the treatment of chronic inflammatory pain.


Assuntos
Proteínas de Transporte/metabolismo , Inflamação/fisiopatologia , Neurônios/metabolismo , Dor/metabolismo , Isoformas de Proteínas/metabolismo , Transdução de Sinais/fisiologia , Sinapses/fisiologia , Animais , Cálcio/metabolismo , Proteínas de Transporte/genética , Doença Crônica , Dependovirus/genética , Dependovirus/metabolismo , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Retroalimentação Fisiológica , Proteínas de Arcabouço Homer , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Conformação de Ácido Nucleico , Dor/fisiopatologia , Isoformas de Proteínas/genética , RNA/química , RNA/metabolismo , Ratos , Ratos Wistar , Receptores de Glutamato/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Medula Espinal/citologia , Sinapses/ultraestrutura
16.
Ann Plast Surg ; 70(6): 726-31, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23403543

RESUMO

BACKGROUND: The combination of gene therapy and plastic surgery may have the potential to improve the specificity that is needed to achieve clinically applicable treatment regimens. Our goal was to develop a method for gene modification that would yield sustainable production of gene products but would be less time consuming than existing protocols. METHODS: An adenoassociated virus was used to deliver gene products to pectoralis muscle flaps. Gene modification was accomplished via either direct injection or novel fat grafting techniques. RESULTS: The production of gene product was observable by both in vivo imaging and immunohistochemical staining. Gene products were not detected in tissues that were not in contact with the fat grafts that were incubated with the viral vector, indicating that the transduction stayed local to the flap. CONCLUSIONS: Using novel recombinant adenoassociated virus vectors, we have developed a method for gene delivery that is highly efficient and applicable to muscle flaps.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Procedimentos de Cirurgia Plástica/métodos , Gordura Subcutânea/transplante , Retalhos Cirúrgicos , Animais , Vetores Genéticos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gordura Subcutânea/virologia , Retalhos Cirúrgicos/virologia , Transfecção/métodos
17.
Nat Genet ; 36(8): 827-35, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15258583

RESUMO

An enriched environment is associated with hippocampal plasticity, including improved cognitive performance and increased neurogenesis. Here, we show that hippocampal expression of vascular endothelial growth factor (VEGF) is increased by both an enriched environment and performance in a spatial maze. Hippocampal gene transfer of VEGF in adult rats resulted in approximately 2 times more neurogenesis associated with improved cognition. In contrast, overexpression of placental growth factor, which signals through Flt1 but not kinase insert domain protein receptors (KDRs), had negative effects on neurogenesis and inhibited learning, although it similarly increased endothelial cell proliferation. Expression of a dominant-negative mutant KDR inhibited basal neurogenesis and impaired learning. Coexpression of mutant KDR antagonized VEGF-enhanced neurogenesis and learning without inhibiting endothelial cell proliferation. Furthermore, inhibition of VEGF expression by RNA interference completely blocked the environmental induction of neurogenesis. These data support a model in which VEGF, acting through KDR, mediates the effect of the environment on neurogenesis and cognition.


Assuntos
Hipocampo/fisiologia , Aprendizagem , Memória , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Divisão Celular , Endotélio Vascular/fisiologia , Hipocampo/metabolismo , Plasticidade Neuronal , Neurônios/fisiologia , Fator de Crescimento Placentário , Proteínas da Gravidez/fisiologia , Ratos
18.
J Neurophysiol ; 105(2): 923-8, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20980546

RESUMO

Deletion of N-methyl-D-aspartate receptors (NMDARs) early in development results in an increase in the number of synaptic AMPA receptors (AMPARs), suggesting a role for NMDARs in negatively regulating AMPAR trafficking at developing synapses. Substantial evidence has shown that AMPAR subunits function differentially in AMPAR trafficking. However, the role of AMPAR subunits in the enhancement of AMPARs following NMDAR ablation remains unknown. We have now performed single-cell genetic deletions in double-floxed mice in which the deletion of GluN1 is combined with the deletion of GluA1 or GluA2. We find that the AMPAR enhancement following NMDAR deletion requires the GluA2 subunit, but not the GluA1 subunit, indicating a key role for GluA2 in the regulation of AMPAR trafficking in developing synapses.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciação de Longa Duração/fisiologia , Células Piramidais/fisiologia , Receptores de AMPA/química , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/fisiologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Camundongos , Camundongos Transgênicos , Subunidades Proteicas , Relação Estrutura-Atividade
19.
Nat Med ; 9(9): 1173-9, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12925848

RESUMO

Glucagon-like peptide-1 (GLP-1) is a gut peptide that, together with its receptor, GLP-1R, is expressed in the brain. Here we show that intracerebroventricular (i.c.v.) GLP-1 and [Ser(2)]exendin(1-9) (HSEGTFTSD; homologous to a conserved domain in the glucagon/GLP-1 family) enhance associative and spatial learning through GLP-1R. [Ser(2)]exendin(1-9), but not GLP-1, is also active when administered peripherally. GLP-1R-deficient mice have a phenotype characterized by a learning deficit that is restored after hippocampal Glp1r gene transfer. In addition, rats overexpressing GLP-1R in the hippocampus show improved learning and memory. GLP-1R-deficient mice also have enhanced seizure severity and neuronal injury after kainate administration, with an intermediate phenotype in heterozygotes and phenotypic correction after Glp1r gene transfer in hippocampal somatic cells. Systemic administration of [Ser(2)]exendin(1-9) in wild-type animals prevents kainate-induced apoptosis of hippocampal neurons. Brain GLP-1R represents a promising new target for both cognitive-enhancing and neuroprotective agents.


Assuntos
Glucagon/farmacologia , Hipocampo/fisiologia , Aprendizagem/fisiologia , Fragmentos de Peptídeos/farmacologia , Precursores de Proteínas/farmacologia , Receptores de Glucagon/genética , Receptores de Glucagon/metabolismo , Administração Intranasal , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Cognição/efeitos dos fármacos , Expressão Gênica , Técnicas de Transferência de Genes , Glucagon/administração & dosagem , Peptídeo 1 Semelhante ao Glucagon , Receptor do Peptídeo Semelhante ao Glucagon 1 , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Ácido Caínico/efeitos adversos , Aprendizagem/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/administração & dosagem , Precursores de Proteínas/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores de Glucagon/efeitos dos fármacos , Convulsões/induzido quimicamente , Convulsões/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais
20.
Proc Natl Acad Sci U S A ; 105(14): 5597-602, 2008 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-18375768

RESUMO

How the billions of synapses in the adult mammalian brain are precisely specified remains one of the fundamental questions of neuroscience. Although a genetic program is likely to encode the basic neural blueprint, much evidence suggests that experience-driven activity through NMDA receptors wires up neuronal circuits by inducing a process similar to long-term potentiation. To test this notion directly, we eliminated NMDA receptors before and during synaptogenesis in single cells in vitro and in vivo. Although the prevailing model would predict that NMDA receptor deletion should strongly inhibit the maturation of excitatory circuits, we find that genetic ablation of NMDA receptor function profoundly increases the number of functional synapses between neurons. Conversely, reintroduction of NMDA receptors into NR1-deficient neurons reduces the number of functional inputs, a process requiring network activity and NMDA receptor function. Although NMDA receptor deletion increases the strength of unitary connections, it does not alter neuronal morphology, suggesting that basal NMDA receptor activation blocks the recruitment of AMPA receptors to silent synapses. Based on these results we suggest a new model for the maturation of excitatory synapses in which ongoing activation of NMDA receptors prevents premature synaptic maturation by ensuring that only punctuated bursts of activity lead to the induction of a functional synapse for the activity-dependent wiring of neural circuitry.


Assuntos
Encéfalo/crescimento & desenvolvimento , Receptores de N-Metil-D-Aspartato/fisiologia , Sinapses/fisiologia , Animais , Encéfalo/fisiologia , Hipocampo/citologia , Potenciação de Longa Duração , Camundongos , Camundongos Endogâmicos , Neurônios/citologia , Neurônios/fisiologia , Receptores de AMPA , Receptores de N-Metil-D-Aspartato/deficiência , Transmissão Sináptica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA