Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Drug Dev Res ; 84(6): 1085-1095, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37291958

RESUMO

The decades-old cholinergic hypothesis of Alzheimer's disease (AD) led to clinical testing and FDA approval of acetylcholinesterase inhibitor drugs. Subsequently, the α7 nicotinic acetylcholine receptor (α7nAChR) was proposed as a new drug target for enhancing cholinergic neurotransmission. Nearly simultaneously, soluble amyloid ß1-42 (Aß42 ) was shown to bind α7nAChR with picomolar affinity to activate kinases that hyperphosphorylate tau, the precursor to tau-containing tangles. Multiple biopharmaceutical companies explored α7nAChR as a drug target for AD, mostly to enhance neurotransmission. Directly targeting α7nAChR proved to be a drug development challenge. The ultra-high-affinity interaction between Aß42 and α7nAChR posed a significant hurdle for direct competition in the AD brain. The receptor rapidly desensitizes, undermining efficacy of agonists. Drug discovery approaches therefore included partial agonists and allosteric modulators of α7nAChR. After substantial effort, numerous drug candidates were abandoned due to lack of efficacy or drug-related toxicities. As alternatives, proteins interacting with α7nAChR were sought. In 2016, a novel nAChR regulator was identified, but no drug candidates have emerged from this effort. In 2012, the interaction of filamin A with α7nAChR was shown to be critical to Aß42 's toxic signaling via α7nAChR, presenting a new drug target. The novel drug candidate simufilam disrupts the filamin A-α7nAChR interaction, reduces Aß42 's high-affinity binding to α7nAChR, and suppresses Aß42 's toxic signaling. Early clinical trials of simufilam showed improvements in experimental CSF biomarkers and indications of cognitive improvement in mild AD patients at 1 year. Simufilam is currently in phase 3 clinical trials as a disease-modifying treatment for AD.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Peptídeos beta-Amiloides/metabolismo , Filaminas/metabolismo , Acetilcolinesterase/metabolismo , Inibidores da Colinesterase/uso terapêutico , Desenvolvimento de Medicamentos
2.
Int J Mol Sci ; 24(18)2023 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-37762230

RESUMO

Simufilam is a novel oral drug candidate in Phase 3 clinical trials for Alzheimer's disease (AD) dementia. This small molecule binds an altered form of filamin A (FLNA) that occurs in AD. This drug action disrupts FLNA's aberrant linkage to the α7 nicotinic acetylcholine receptor (α7nAChR), thereby blocking soluble amyloid beta1-42 (Aß42)'s signaling via α7nAChR that hyperphosphorylates tau. Here, we aimed to clarify simufilam's mechanism. We now show that simufilam reduced Aß42 binding to α7nAChR with a 10-picomolar IC50 using time-resolved fluorescence resonance energy transfer (TR-FRET), a robust technology to detect highly sensitive molecular interactions. We also show that FLNA links to multiple inflammatory receptors in addition to Toll-like receptor 4 (TLR4) in postmortem human AD brains and in AD transgenic mice: TLR2, C-X-C chemokine receptor type 4 (CXCR4), C-C chemokine receptor type 5 (CCR5), and T-cell co-receptor cluster of differentiation 4 (CD4). These aberrant FLNA linkages, which can be induced in a healthy control brain by Aß42 incubation, were disrupted by simufilam. Simufilam reduced inflammatory cytokine release from Aß42-stimulated human astrocytes. In the AD transgenic mice, CCR5-G protein coupling was elevated, indicating persistent activation. Oral simufilam reduced both the FLNA-CCR5 linkage and the CCR5-G protein coupling in these mice, while restoring CCR5's responsivity to C-C chemokine ligand 3 (CCL3). By disrupting aberrant FLNA-receptor interactions critical to AD pathogenic pathways, simufilam may promote brain health.


Assuntos
Doença de Alzheimer , Camundongos , Humanos , Animais , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Filaminas/metabolismo , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo
3.
Ann Neurol ; 88(3): 513-525, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32557841

RESUMO

OBJECTIVE: To examine associations of molecular markers of brain insulin signaling with Alzheimer disease (AD) and cognition among older persons with or without diabetes. METHODS: This clinical-pathologic study was derived from a community-based cohort study, the Religious Orders Study. We studied 150 individuals (mean age at death =87 years, 48% women): 75 with and 75 without diabetes (matched by sex on age at death and education). Using enzyme-linked immunosorbent assay, immunohistochemistry, and ex vivo stimulation of brain tissue with insulin, we assessed insulin signaling in the postmortem middle frontal gyrus cortex. Postmortem data documented AD neuropathology. Clinical evaluations documented cognitive function proximate to death, based on 17 neuropsychological tests. In adjusted regression analyses, we examined associations of brain insulin signaling with diabetes, AD, and level of cognition. RESULTS: Brain insulin receptor substrate-1 (IRS1) phosphorylation (pS307 IRS1/total IRS1) and serine/threonine-protein kinase (AKT) phosphorylation (pT308 AKT1/total AKT1) were similar in persons with or without diabetes. AKT phosphorylation was associated with the global AD pathology score (p = 0.001). In contrast, IRS1 phosphorylation was not associated with AD (p = 0.536). No other associations of insulin signaling were found with the global AD score, including when using the ex vivo brain insulin stimulation method. In secondary analyses, normalized pT308 AKT1 was positively correlated with both the amyloid burden and tau tangle density, and no other associations of brain insulin signaling with neuropathology were observed. Moreover, normalized pT308 AKT1 was associated with a lower level of global cognitive function (estimate = -0.212, standard error = 0.097; p = 0.031). INTERPRETATION: Brain AKT phosphorylation, a critical node in the signaling of insulin and other growth factors, is associated with AD neuropathology and lower cognitive function. ANN NEUROL 2020;88:513-525.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Cognição/fisiologia , Insulina/metabolismo , Idoso de 80 Anos ou mais , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Feminino , Humanos , Masculino , Transdução de Sinais
4.
Mol Psychiatry ; 25(4): 750-760, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-30214040

RESUMO

Multiple lines of evidence point to glutamatergic signaling in the postsynaptic density (PSD) as a pathophysiologic mechanism in schizophrenia. Integral to PSD glutamatergic signaling is reciprocal interplay between GluN and mGluR5 signaling. We examined agonist-induced mGluR5 signaling in the postmortem dorsolateral prefrontal cortex (DLPFC) derived from 17 patients and age-matched and sex-matched controls. The patient group showed a striking reduction in mGluR5 signaling, manifested by decreases in Gq/11 coupling and association with PI3K and Homer compared to controls (p < 0.01 for all). This was accompanied by increases in serine and tyrosine phosphorylation of mGluR5, which can decrease mGluR5 activity via desensitization (p < 0.01). In addition, we find altered protein-protein interaction (PPI) of mGluR5 with RGS4, norbin, Preso 1 and tamalin, which can also attenuate mGluR5 activity. We previously reported molecular underpinnings of GluN hypofunction (decreased GluN2 phosphorylation) and here we show those of reduced mGluR5 signaling in schizophrenia. We find that reduced GluN2 phosphorylation can be precipitated by attenuated mGluR5 activity and that increased mGluR5 phosphorylation can result from decreased GluN function, suggesting a reciprocal interplay between the two pathways in schizophrenia. Interestingly, the patient group showed decreased mGluR5-GluN association (p < 0.01), a mechanistic basis for the reciprocal facilitation. In sum, we present the first direct evidence for mGluR5 hypoactivity, propose a reciprocal interplay between GluN and mGluR5 pathways as integral to glutamatergic dysregulation and suggest protein-protein interactions in mGluR5-GluN complexes as potential targets for intervention in schizophrenia.


Assuntos
Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/metabolismo , Idoso , Idoso de 80 Anos ou mais , Antipsicóticos/uso terapêutico , Encéfalo/metabolismo , Fármacos Atuantes sobre Aminoácidos Excitatórios/metabolismo , Feminino , Humanos , Masculino , Proteínas de Membrana/metabolismo , Fosforilação , Densidade Pós-Sináptica/metabolismo , Córtex Pré-Frontal/metabolismo , Receptor de Glutamato Metabotrópico 5/fisiologia , Transdução de Sinais/efeitos dos fármacos
5.
Alzheimers Dement ; 10(1 Suppl): S12-25, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24529520

RESUMO

Alzheimer's disease (AD) is an age-related neurodegenerative disease leading over the course of decades to the most common form of dementia. Many of its pathologic features and cognitive deficits may be due in part to brain insulin resistance recently demonstrated in the insulin receptor→insulin receptor substrate-1 (IRS-1) signaling pathway. The proximal cause of such resistance in AD dementia and amnestic mild cognitive impairment (aMCI) appears to be serine inhibition of IRS-1, a phenomenon likely due to microglial release of inflammatory cytokines triggered by oligomeric Aß. Studies on animal models of AD and on human brain tissue from MCI cases at high risk of AD dementia have shown that brain insulin resistance and many other pathologic features and symptoms of AD may be greatly reduced or even reversed by treatment with FDA-approved glucagon-like peptide-1 (GLP-1) analogs such as liraglutide (Victoza). These findings call attention to the need for further basic, translational, and clinical studies on GLP-1 analogs as promising AD therapeutics.


Assuntos
Doença de Alzheimer , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Resistência à Insulina , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Humanos
6.
J Neurosci ; 32(29): 9773-84, 2012 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-22815492

RESUMO

PTI-125 is a novel compound demonstrating a promising new approach to treating Alzheimer's disease (AD), characterized by neurodegeneration and amyloid plaque and neurofibrillary pathologies. We show that the toxic signaling of amyloid-ß(42) (Aß(42)) by the α7-nicotinic acetylcholine receptor (α7nAChR), which results in tau phosphorylation and formation of neurofibrillary tangles, requires the recruitment of the scaffolding protein filamin A (FLNA). By binding FLNA with high affinity, PTI-125 prevents Aß(42)'s toxic cascade, decreasing phospho-tau and Aß aggregates and reducing the dysfunction of α7nAChRs, NMDARs, and insulin receptors. PTI-125 prevents Aß(42) signaling by drastically reducing its affinity for α7nAChRs and can even dissociate existing Aß(42)-α7nAChR complexes. Additionally, PTI-125 prevents Aß-induced inflammatory cytokine release by blocking FLNA recruitment to toll-like receptor 4, illustrating an anti-inflammatory effect. PTI-125's broad spectrum of beneficial effects is demonstrated here in an intracerebroventricular Aß(42) infusion mouse model of AD and in human postmortem AD brain tissue.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/efeitos dos fármacos , Proteínas Contráteis/antagonistas & inibidores , Proteínas dos Microfilamentos/antagonistas & inibidores , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas Contráteis/metabolismo , Citocinas/metabolismo , Feminino , Filaminas , Humanos , Masculino , Camundongos , Proteínas dos Microfilamentos/metabolismo , Emaranhados Neurofibrilares/efeitos dos fármacos , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Fosforilação/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Nicotínicos/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptor Nicotínico de Acetilcolina alfa7 , Proteínas tau/metabolismo
7.
Nat Med ; 12(7): 824-8, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16767099

RESUMO

Recent molecular genetics studies implicate neuregulin 1 (NRG1) and its receptor erbB in the pathophysiology of schizophrenia. Among NRG1 receptors, erbB4 is of particular interest because of its crucial roles in neurodevelopment and in the modulation of N-methyl-D-aspartate (NMDA) receptor signaling. Here, using a new postmortem tissue-stimulation approach, we show a marked increase in NRG1-induced activation of erbB4 in the prefrontal cortex in schizophrenia. Levels of NRG1 and erbB4, however, did not differ between schizophrenia and control groups. To evaluate possible causes for this hyperactivation of erbB4 signaling, we examined the association of erbB4 with PSD-95 (postsynaptic density protein of 95 kDa), as this association has been shown to facilitate activation of erbB4. Schizophrenia subjects showed substantial increases in erbB4-PSD-95 interactions. We found that NRG1 stimulation suppresses NMDA receptor activation in the human prefrontal cortex, as previously reported in the rodent cortex. NRG1-induced suppression of NMDA receptor activation was more pronounced in schizophrenia subjects than in controls, consistent with enhanced NRG1-erbB4 signaling seen in this illness. Therefore, these findings suggest that enhanced NRG1 signaling may contribute to NMDA hypofunction in schizophrenia.


Assuntos
Encéfalo/fisiopatologia , Receptores ErbB/fisiologia , Neuregulina-1/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Esquizofrenia/fisiopatologia , Animais , Encéfalo/patologia , Cadáver , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C3H , Córtex Pré-Frontal/patologia , Córtex Pré-Frontal/fisiopatologia , Receptor ErbB-4 , Esquizofrenia/patologia , Transdução de Sinais
8.
Front Aging ; 4: 1175601, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37457922

RESUMO

Introduction: Implicated in both aging and Alzheimer's disease (AD), mammalian target of rapamycin (mTOR) is overactive in AD brain and lymphocytes. Stimulated by growth factors such as insulin, mTOR monitors cell health and nutrient needs. A small molecule oral drug candidate for AD, simufilam targets an altered conformation of the scaffolding protein filamin A (FLNA) found in AD brain and lymphocytes that induces aberrant FLNA interactions leading to AD neuropathology. Simufilam restores FLNA's normal shape to disrupt its AD-associated protein interactions. Methods: We measured mTOR and its response to insulin in lymphocytes of AD patients before and after oral simufilam compared to healthy control lymphocytes. Results: mTOR was overactive and its response to insulin reduced in lymphocytes from AD versus healthy control subjects, illustrating another aspect of insulin resistance in AD. After oral simufilam, lymphocytes showed normalized basal mTOR activity and improved insulin-evoked mTOR activation in mTOR complex 1, complex 2, and upstream and downstream signaling components (Akt, p70S6K and phosphorylated Rictor). Suggesting mechanism, we showed that FLNA interacts with the insulin receptor until dissociation by insulin, but this linkage was elevated and its dissociation impaired in AD lymphocytes. Simufilam improved the insulin-mediated dissociation. Additionally, FLNA's interaction with Phosphatase and Tensin Homolog deleted on Chromosome 10 (PTEN), a negative regulator of mTOR, was reduced in AD lymphocytes and improved by simufilam. Discussion: Reducing mTOR's basal overactivity and its resistance to insulin represents another mechanism of simufilam to counteract aging and AD pathology. Simufilam is currently in Phase 3 clinical trials for AD dementia.

9.
J Neurosci ; 31(30): 11044-54, 2011 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-21795553

RESUMO

Repetitive transcranial magnetic stimulation (rTMS) induces neuronal long-term potentiation or depression. Although brain-derived neurotrophic factor (BDNF) and its cognate tyrosine receptor kinase B (TrkB) contribute to the effects of rTMS, their precise role and underlying mechanism remain poorly understood. Here we show that daily 5 Hz rTMS for 5 d improves BDNF-TrkB signaling in rats by increasing the affinity of BDNF for TrkB, which results in higher tyrosine-phosphorylated TrkB, increased recruitment of PLC-γ1 and shc/N-shc to TrkB, and heightened downstream ERK2 and PI-3K activities in prefrontal cortex and in lymphocytes. The elevated BDNF-TrkB signaling is accompanied by an increased association between the activated TrkB and NMDA receptor (NMDAR). In normal human subjects, 5 d rTMS to motor cortex decreased resting motor threshold, which correlates with heightened BDNF-TrkB signaling and intensified TrkB-NMDAR association in lymphocytes. These findings suggest that rTMS to cortex facilitates BDNF-TrkB-NMDAR functioning in both cortex and lymphocytes.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/líquido cefalorraquidiano , Encéfalo/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Linfócitos/efeitos da radiação , Receptor trkB/líquido cefalorraquidiano , Transdução de Sinais/efeitos da radiação , Estimulação Magnética Transcraniana , Adulto , Análise de Variância , Animais , Encéfalo/metabolismo , Estudos Cross-Over , Método Duplo-Cego , Eletromiografia , Potencial Evocado Motor/fisiologia , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Técnicas In Vitro , Linfócitos/metabolismo , Masculino , Músculo Esquelético/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/fisiologia , Estatística como Assunto , Adulto Jovem
10.
Physiol Behav ; 234: 113286, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33321142

RESUMO

Emotional contagion refers to the sharing of emotional states between individuals and can cause depressive behaviors in healthy persons who live with depressed individuals. Negative emotional contagion has been observed in animal models, but the vast majority of studies are short-term and bear little resemblance to long-term human relationships. Thus, the first aim of this study was to establish an animal model of stress-induced negative emotional contagion that develops across time and between pairs. To accomplish this, we tested the hypothesis that sedentary male rats that cohabitate for five weeks with a stress-exposed female will exhibit a depression-like phenotype that is observable on behavioral and physiological measures. In addition, drawing from a comprehensive literature that describes the beneficial effects of prior exercise on stress-related behavior, we tested our second hypothesis that in males that were paired with a stressed female, prior voluntary exercise will diminish the impact of negative emotional contagion. We found that pair housing a healthy male with a stressed female led to emotional contagion; males gained less body weight, were anhedonic, demonstrated heightened anxiety-like behavior, had lower serum brain-derived neurotrophic factor (BDNF) levels, had decreased hippocampal BDNF-stimulated tyrosine receptor kinase B (TrkB) signaling and had increased pro-inflammatory cytokine expression in the hippocampus. For the most part, the five-week exercise window that occurred prior to pair housing had few effects in non-stress paired rats, but had partial, yet substantial protective effects in rats that were pair-housed with a stressed female. Specifically, stress-paired, exercised rats showed less depressive-like behavior, had partially preserved hippocampal BDNF-stimulated TrkB signaling, had normalized serum BDNF concentration, and had hippocampal cytokine and immediate early gene levels that were equivalent to controls. These preclinical findings introduce a new model of negative emotional contagion between dyads of male-female rats and support the view that inclusion of exercise programs would be beneficial for persons that may, in the future, be susceptible to negative emotional contagion.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Receptor trkB , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Feminino , Hipocampo/metabolismo , Masculino , Ratos , Receptor trkB/metabolismo , Estresse Psicológico , Tirosina
11.
Acta Neuropathol Commun ; 9(1): 71, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33858515

RESUMO

Insulin is an important hormone for brain function, and alterations in insulin metabolism may be associated with neuropathology. We examined associations of molecular markers of brain insulin signaling with cerebrovascular disease. Participants were enrolled in the Religious Orders Study (ROS), an ongoing epidemiologic community-based, clinical-pathologic study of aging from across the United States. Using cross-sectional analyses, we studied a subset of ROS: 150 persons with or without diabetes, matched 1:1 by sex on age-at-death and education. We used ELISA, immunohistochemistry, and ex vivo stimulation with insulin, to document insulin signaling in postmortem midfrontal gyrus cortex tissue. Postmortem neuropathologic data identified cerebrovascular disease including brain infarcts, classified by number (as none for the reference; one; and more than one), size (gross and microscopic infarcts), and brain region/location (cortical and subcortical). Cerebral vessel pathologies were assessed, including severity of atherosclerosis, arteriolosclerosis, and amyloid angiopathy. In separate regression analyses, greater AKT1 phosphorylation at T308 following ex vivo stimulation with insulin (OR = 1.916; estimate = 0.650; p = 0.007) and greater pS616IRS1 immunolabeling in neuronal cytoplasm (OR = 1.610; estimate = 0.476; p = 0.013), were each associated with a higher number of brain infarcts. Secondary analyses showed consistent results for gross infarcts and microinfarcts separately, but no other association including by infarct location (cortical or subcortical). AKT S473 phosphorylation following insulin stimulation was associated with less amyloid angiopathy severity, but not with other vessel pathology including atherosclerosis and arteriolosclerosis. In summary, insulin resistance in the human brain, even among persons without diabetes, is associated with cerebrovascular disease and especially infarcts. The underlying pathophysiologic mechanisms need further elucidation. Because brain infarcts are known to be associated with lower cognitive function and dementia, these data are relevant to better understanding the link between brain metabolism and brain function.


Assuntos
Encéfalo/fisiologia , Transtornos Cerebrovasculares/patologia , Resistência à Insulina , Idoso , Idoso de 80 Anos ou mais , Autopsia , Transtornos Cerebrovasculares/epidemiologia , Estudos Transversais , Complicações do Diabetes/epidemiologia , Complicações do Diabetes/patologia , Diabetes Mellitus , Feminino , Humanos , Insulina/metabolismo , Masculino , Transdução de Sinais/fisiologia
12.
Neuroscience ; 473: 142-158, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34298123

RESUMO

Exposure to intense or repeated stressors can lead to depression or post-traumatic stress disorder (PTSD). Neurological changes induced by stress include impaired neurotrophin signaling, which is known to influence synaptic integrity and plasticity. The present study used an ex vivo approach to examine the impact of acute or repeated stress on BDNF-stimulated TrkB signaling in hippocampus (HIPPO) and prefrontal cortex (PFC). Rats in an acute multiple stressor group experienced five stressors in one day whereas rats in a repeated unpredictable stressor group experienced 20 stressors across 10 days. After stress exposure, slices were incubated with vehicle or BDNF, followed by immunoprecipitation and immunoblot assays to assess protein levels, activation states and protein-protein linkage associated with BDNF-TrkB signaling. Three key findings are (1) exposure to stressors significantly diminished BDNF-stimulated TrkB signaling in HIPPO and PFC such that reductions in TrkB activation, diminished recruitment of adaptor proteins to TrkB, reduced activation of downstream signaling molecules, disruption of TrkB-NMDAr linkage, and changes in basal and BDNF-stimulated Arc expression were observed. (2) After stress, BDNF stimulation enhanced TrkB-NMDAr linkage in PFC, suggestive of compensatory mechanisms in this region. (3) We discovered an uncoupling between TrkB signaling, TrkB-NMDAr linkage and Arc expression in PFC and HIPPO. In addition, a robust surge in pro-inflammatory cytokines was observed in both regions after repeated exposure to stressors. Collectively, these data provide therapeutic targets for future studies that investigate how to reverse stress-induced downregulation of BDNF-TrkB signaling and underscore the need for functional studies that examine stress-related TrkB-NMDAr activities in PFC.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Receptores de N-Metil-D-Aspartato , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Ratos , Receptor trkB/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais
13.
Mol Neurodegener ; 16(1): 26, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33863362

RESUMO

BACKGROUND: Apolipoprotein E4 (APOE4) is associated with a greater response to neuroinflammation and the risk of developing late-onset Alzheimer's disease (AD), but the mechanisms for this association are not clear. The activation of calcium-dependent cytosolic phospholipase A2 (cPLA2) is involved in inflammatory signaling and is elevated within the plaques of AD brains. The relation between APOE4 genotype and cPLA2 activity is not known. METHODS: Mouse primary astrocytes, mouse and human brain samples differing by APOE genotypes were collected for measuring cPLA2 expression, phosphorylation, and activity in relation to measures of inflammation and oxidative stress. RESULTS: Greater cPLA2 phosphorylation, cPLA2 activity and leukotriene B4 (LTB4) levels were identified in ApoE4 compared to ApoE3 in primary astrocytes, brains of ApoE-targeted replacement (ApoE-TR) mice, and in human brain homogenates from the inferior frontal cortex of patients with AD carrying APOE3/E4 compared to APOE3/E3. Greater cPLA2 phosphorylation was also observed in human postmortem frontal cortical synaptosomes and primary astrocytes after treatment with recombinant ApoE4 ex vivo. In ApoE4 astrocytes, the greater levels of LTB4, reactive oxygen species (ROS), and inducible nitric oxide synthase (iNOS) were reduced after cPLA2 inhibition. CONCLUSIONS: Our findings implicate greater activation of cPLA2 signaling system with APOE4, which could represent a potential drug target for mitigating the increased neuroinflammation with APOE4 and AD.


Assuntos
Apolipoproteína E4/metabolismo , Cálcio/farmacologia , Córtex Cerebral/enzimologia , Sistema de Sinalização das MAP Quinases , Fosfolipases A2 Citosólicas/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacologia , Apolipoproteína E4/genética , Apolipoproteína E4/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Córtex Cerebral/patologia , Ativação Enzimática/efeitos dos fármacos , Heterozigoto , Humanos , Inflamassomos , Inflamação , Leucotrieno B4/biossíntese , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Estresse Oxidativo , Fragmentos de Peptídeos/farmacologia , Fosforilação , Processamento de Proteína Pós-Traducional , Espécies Reativas de Oxigênio , Sinaptossomos/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
14.
J Neurosci ; 29(35): 10961-73, 2009 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-19726654

RESUMO

Alzheimer's disease (AD) is characterized by synaptic dysfunction and cardinal neuropathological features including amyloid plaques and neurofibrillary tangles. Soluble amyloid-beta (Abeta) can suppress synaptic activities by interacting with alpha7 nicotinic acetylcholine receptors (alpha7nAChRs). Here, we show that alpha7nAChR and NMDA glutamatergic receptor (NMDAR) activities are severely compromised in synaptosomes prepared from AD and Abeta(1-42) (Abeta42)-exposed control frontal cortex slices from postmortem tissue. Whereas Abeta(12-28) prevents Abeta42 from binding to alpha7nAChRs, 2-[2-(4-bromophenyl)-2-oxoethyl]-1-methyl pyridinium (S 24795), a novel alpha7nAChR partial agonist, facilitates release of Abeta42 from Abeta42-alpha7nAChR and -Abeta42 complexes. S 24795 interacts with alpha7nAChR and Abeta(15-20) region of the Abeta42 to enable partial recovery of the alpha7nAChR and NMDAR channel function. These findings suggest that the Abeta-alpha7nAChR interaction may be an upstream pathogenic event in AD and demonstrate that some recovery of neuronal channel activities may be achieved in AD brains by removing Abeta from alpha7nAChRs.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/fisiologia , Encéfalo/fisiologia , Sistemas de Liberação de Medicamentos/métodos , Compostos de Piridínio/administração & dosagem , Receptores de N-Metil-D-Aspartato/fisiologia , Receptores Nicotínicos/fisiologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/antagonistas & inibidores , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Sistemas de Liberação de Medicamentos/tendências , Feminino , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Receptor Nicotínico de Acetilcolina alfa7
15.
J Neurosci ; 29(19): 6308-19, 2009 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-19439608

RESUMO

Prenatal cocaine exposure produces sustained neurobehavioral and brain synaptic changes closely resembling those of animals with defective AMPA receptors (AMPARs). We hypothesized that prenatal cocaine exposure attenuates AMPAR signaling by interfering with AMPAR synaptic targeting. AMPAR function is governed by receptor cycling on and off the synaptic membrane through its interaction with glutamate receptor-interacting protein (GRIP), a PDZ domain protein that is regulated by reversible phosphorylation. Our results show that prenatal cocaine exposure markedly reduces AMPAR synaptic targeting and attenuates AMPAR-mediated synaptic long-term depression in the frontal cortex of 21-d-old rats. This cocaine effect is the result of reduced GRIP-AMPAR interaction caused by persistent phosphorylation of GRIP by protein kinase C (PKC) and Src tyrosine kinase. These data support the restoration of AMPAR activation via suppressing excessive PKC-mediated GRIP phosphorylation as a novel therapeutic approach to treat the neurobehavioral consequences of prenatal cocaine.


Assuntos
Proteínas de Transporte/metabolismo , Fármacos do Sistema Nervoso Central/toxicidade , Cocaína/toxicidade , Proteínas do Tecido Nervoso/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Receptores de AMPA/metabolismo , Sinapses/efeitos dos fármacos , Animais , Feminino , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/crescimento & desenvolvimento , Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/fisiologia , Masculino , Fosforilação/efeitos dos fármacos , Gravidez , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Sinapses/metabolismo , Quinases da Família src/metabolismo
16.
Biochem Pharmacol ; 176: 113814, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31954716

RESUMO

Brain tumors, particularly high-grade glioblastomas, are a crucial public health issue due to poor prognosis and an extremely low survival rate. The glioblastoma multiforme (GBM) grows rapidly within its unique microenvironment that is characterized by active neural communications. Therefore, diverse neurotransmitters not only maintain normal brain functions but also influence glioma progression. To fully appreciate the relationship between neurotransmitters and glioma progression, we reviewed potential neurotransmitter contributors in human GBM and the much less aggressive Low-grade glioma (LGG) by combining previously published data from gene-mutation/mRNA sequencing databases together with protein-protein interaction (PPI) network analysis results. The summarized results indicate that glutamatergic and calcium signaling may provide positive feedback to promote glioma formation through 1) metabolic reprogramming and genetic switching to accelerate glioma duplication and progression; 2) upregulation of cytoskeleton proteins and elevation of intracellular Ca2+ levels to increase glutamate release and facilitate formation of synaptic-like connections with surrounding cells in their microenvironment. The upregulated glutamatergic neuronal activities in turn stimulate glioma growth and signaling. Importantly, the enhanced electrical and molecular signals from both neurons and glia propagate out to enable glioma symptoms such as epilepsy and migraine. The elevated intracellular Ca2+ also activates nitric oxide synthase to produce nitric oxide (NO) that can either promote or inhibit tumorigenesis. By analyzing the network effects for complex interaction among neurotransmitters such as glutamate, Ca2+ and NO in brain tumor progression, especially GBM, we identified the glutamatergic signaling as the potential therapeutic targets and suggest manipulation of glutamatergic signaling may be an effective treatment strategy for this aggressive brain cancer.


Assuntos
Neoplasias Encefálicas/metabolismo , Cálcio/metabolismo , Glioblastoma/metabolismo , Ácido Glutâmico/metabolismo , Transdução de Sinais , Animais , Neoplasias Encefálicas/patologia , Progressão da Doença , Glioblastoma/patologia , Humanos , Neurônios/metabolismo , Neurônios/patologia , Óxido Nítrico/metabolismo
17.
Dev Neurosci ; 31(1-2): 71-5, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19372688

RESUMO

Alterations in dendritic spine density following prenatal cocaine exposure were examined in the present study. Timed pregnant rats were injected daily with 30 mg/kg cocaine or saline during gestation. At postnatal day 21, male and female animals were separated and spine density was assessed following Golgi impregnation. In prenatal cocaine-exposed rats, significant increases in dendritic spine density were observed on pyramidal cells in the CA1 region of the hippocampus, basal dendrites of layer II/III of the medial prefrontal cortex, medium spiny neurons of the striatum and the core of the nucleus accumbens, as well as in neurons of the ventromedial hypothalamic nucleus. No differences were observed in either apical or basal dendrites of pyramidal cells in layer III of the sensory cortex or layer V of the medial prefrontal cortex, or in apical dendrites of layer II/III pyramidal cells of the medial prefrontal cortex. Moreover, there were no sex differences in any region examined. These results demonstrate that prenatal cocaine exposure increases spine density in many brain regions at postnatal day 21, and this effect is independent of sex.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Cocaína/toxicidade , Espinhas Dendríticas/efeitos dos fármacos , Animais , Feminino , Masculino , Gravidez , Células Piramidais/efeitos dos fármacos , Ratos , Caracteres Sexuais
18.
Crit Rev Oncog ; 24(3): 243-250, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32422022

RESUMO

Glioblastoma multiforme (GBM) is the most common and malignant glial tumor. Although pro-growth, pro-survival, and pro-metastasis insulin signaling has been proposed to be a prominent driver of GBM progression, the insulin receptor (IR) signaling cascade in GBM has not been fully elucidated. Upon binding of the insulin and insulin-like growth factor-1 (IGF-1), IR is activated by increasing the levels of tyrosine-phosphorylated (pY) IRP on tyrosine 960, 1150, and 1151 residues as well as IRS-1 recruitment to IRß. This leads to activation of the downstream PI3K/AKT/GSK3 or mTORC1/ERK, many of which are implicated in tumorigenesis including breast and liver carcinomas. Here, we directly compare insulin signaling in U87 MG human glioblastoma to primary human astrocytes by assessing the levels of activated IRß, IRS-1 recruitment to IRß, as well as downstream activated mitogenic ERK2 and pro-survival AKT1 under nonstimulated conditions and induced by 1 nM insulin. Our results show insulin receptor and its downstream signaling molecules are robustly hyperactivated. This mechanism renders a reduced insulin-induced response. Our findings provide a mechanism through which GBM develops and grows aggressively even without insulin.


Assuntos
Antígenos CD/metabolismo , Glioblastoma/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Insulina/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Linhagem Celular Tumoral , Glioblastoma/patologia , Humanos , Transdução de Sinais/fisiologia
19.
Neurobiol Aging ; 84: 119-130, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31539648

RESUMO

Aberrant insulin and adipokine signaling has been implicated in cognitive decline associated with both type 2 diabetes mellitus and neurodegenerative diseases. We established methods that reliably measure insulin, adiponectin and leptin signaling, and their crosstalk, in thawed postmortem mid-frontal cortical tissue from cognitively normal older subjects with a short postmortem interval. Insulin-evoked insulin receptor (IR) activation increases activated, tyrosine-phosphorylated IRß on tyrosine residues 960, 1150, and 1151, insulin receptor substrate-1 recruitment to IRß and phosphorylated RAC-α-serine/threonine-protein kinase. Adiponectin augments, but leptin inhibits, insulin signaling. Adiponectin activates adiponectin receptors to induce APPL1 binding to adiponectin receptor 1 and 2 and T-cadherin and downstream adenosine monophosphate-dependent protein kinase phosphorylation. Insulin inhibited adiponectin-induced signaling. In addition, leptin-induced leptin receptor (OB-R) signaling promotes Janus kinase 2 recruitment to OB-R and Janus kinase 2 and downstream signal transducer and activator of transcription 3 phosphorylation. Insulin enhanced leptin signaling. These data demonstrate insulin and adipokine signaling interactions in human brain. Future studies can use these methods to examine insulin, adiponectin, and leptin metabolic dysregulation in aging and disease states, such as type 2 diabetes and Alzheimer's disease-related dementias.


Assuntos
Adipocinas/metabolismo , Encéfalo/patologia , Insulina/metabolismo , Transdução de Sinais , Envelhecimento/metabolismo , Encéfalo/metabolismo , Humanos , Leptina/metabolismo , Mudanças Depois da Morte
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA