Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 14(6)2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38927051

RESUMO

Manganese (Mn) is an essential heavy metal in the human body, while excess Mn leads to neurotoxicity, as observed in this study, where 100 µM of Mn was administered to the human neuroblastoma (SH-SY5Y) cell model of dopaminergic neurons in neurodegenerative diseases. We quantitated pathway and gene changes in homeostatic cell-based adaptations to Mn exposure. Utilizing the Gene Expression Omnibus, we accessed the GSE70845 dataset as a microarray of SH-SY5Y cells published by Gandhi et al. (2018) and applied statistical significance cutoffs at p < 0.05. We report 74 pathway and 10 gene changes with statistical significance. ReactomeGSA analyses demonstrated upregulation of histones (5 out of 10 induced genes) and histone deacetylases as a neuroprotective response to remodel/mitigate Mn-induced DNA/chromatin damage. Neurodegenerative-associated pathway changes occurred. NF-κB signaled protective responses via Sirtuin-1 to reduce neuroinflammation. Critically, Mn activated three pathways implicating deficits in purine metabolism. Therefore, we validated that urate, a purine and antioxidant, mitigated Mn-losses of viability in SH-SY5Y cells. We discuss Mn as a hypoxia mimetic and trans-activator of HIF-1α, the central trans-activator of vascular hypoxic mitochondrial dysfunction. Mn induced a 3-fold increase in mRNA levels for antioxidant metallothionein-III, which was induced 100-fold by hypoxia mimetics deferoxamine and zinc.


Assuntos
Manganês , Neuroblastoma , Humanos , Manganês/toxicidade , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neuroblastoma/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Biomarcadores/metabolismo
2.
J Alzheimers Dis ; 96(1): 41-45, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37781810

RESUMO

Familial Alzheimer's disease (fAD) mutations in the amyloid-ß protein precursor (AßPP) enhance brain AßPP C-Terminal Fragment (CTF) levels to inhibit lysosomal v-ATPase. Consequent disrupted acidification of the endolysosomal pathway may trigger brain iron deficiencies and mitochondrial dysfunction. The iron responsive element (IRE) in the 5'Untranslated-region of AßPP mRNA should be factored into this cycle where reduced bioavailable Fe-II would decrease IRE-dependent AßPP translation and levels of APP-CTFß in a cycle to adaptively restore iron homeostasis while increases of transferrin-receptors is evident. In healthy younger individuals, Fe-dependent translational modulation of AßPP is part of the neuroprotective function of sAßPPα with its role in iron transport.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Ferro/metabolismo , Biossíntese de Proteínas , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Lisossomos/metabolismo , Peptídeos beta-Amiloides/metabolismo
3.
J Neurosci Res ; 100(1): 297-308, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-31721270

RESUMO

The rewarding effect of opiates is mediated through dissociable neural systems in drug naïve and drug-dependent states. Neuroadaptations associated with chronic drug use are similar to those produced by chronic pain, suggesting that opiate reward could also involve distinct mechanisms in chronic pain and pain-naïve states. We tested this hypothesis by examining the effect of dopamine (DA) antagonism on morphine reward in a rat model of neuropathic pain.Neuropathic pain was induced in male Sprague-Dawley rats through chronic constriction (CCI) of the sciatic nerve; reward was assessed in the conditioned place preference (CPP) paradigm in separate groups at early (4-8 days post-surgery) and late (11-15 days post-surgery) phases of neuropathic pain. Minimal effective doses of morphine that produced a CPP in early and late phases of neuropathic pain were 6 mg/kg and 2 mg/kg respectively. The DA D1 receptor antagonist, SCH23390, blocked a morphine CPP in sham, but not CCI, rats at a higher dose (0.5 mg/kg), but had no effect at a lower dose (0.1 mg/kg). The DA D2 receptor antagonist, eticlopride (0.1 and 0.5 mg/kg), had no effect on a morphine CPP in sham or CCI rats, either in early or late phases of neuropathic pain. In the CPP paradigm, morphine reward involves DA D1 mechanisms in pain-naïve but not chronic pain states. This could reflect increased sensitivity to drug effects in pain versus no pain conditions and/or differential mediation of opiate reward in these two states.


Assuntos
Dor Crônica , Morfina , Animais , Dor Crônica/tratamento farmacológico , Masculino , Morfina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1 , Recompensa
4.
eNeuro ; 7(5)2020.
Artigo em Inglês | MEDLINE | ID: mdl-32859725

RESUMO

µ-Opioid receptors (MORs) are densely expressed in different brain regions known to mediate reward. One such region is the striatum where MORs are densely expressed, yet the role of these MOR populations in modulating reward is relatively unknown. We have begun to address this question by using a series of genetically engineered mice based on the Cre recombinase/loxP system to selectively delete MORs from specific neurons enriched in the striatum: dopamine 1 (D1) receptors, D2 receptors, adenosine 2a (A2a) receptors, and choline acetyltransferase (ChAT). We first determined the effects of each deletion on opioid-induced locomotion, a striatal and dopamine-dependent behavior. We show that MOR deletion from D1 neurons reduced opioid (morphine and oxycodone)-induced hyperlocomotion, whereas deleting MORs from A2a neurons resulted in enhanced opioid-induced locomotion, and deleting MORs from D2 or ChAT neurons had no effect. We also present the effect of each deletion on opioid intravenous self-administration. We first assessed the acquisition of this behavior using remifentanil as the reinforcing opioid and found no effect of genotype. Mice were then transitioned to oxycodone as the reinforcer and maintained here for 9 d. Again, no genotype effect was found. However, when mice underwent 3 d of extinction training, during which the drug was not delivered, but all cues remained as during the maintenance phase, drug-seeking behavior was enhanced when MORs were deleted from A2a or ChAT neurons. These findings show that these selective MOR populations play specific roles in reward-associated behaviors.


Assuntos
Analgésicos Opioides , Receptores Opioides mu , Analgésicos Opioides/farmacologia , Animais , Camundongos , Morfina , Neurônios , Receptores Opioides mu/genética , Recompensa
5.
Learn Mem ; 27(9): 395-413, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32817306

RESUMO

A set of common-acting iron-responsive 5'untranslated region (5'UTR) motifs can fold into RNA stem loops that appear significant to the biology of cognitive declines of Parkinson's disease dementia (PDD), Lewy body dementia (LDD), and Alzheimer's disease (AD). Neurodegenerative diseases exhibit perturbations of iron homeostasis in defined brain subregions over characteristic time intervals of progression. While misfolding of Aß from the amyloid-precursor-protein (APP), alpha-synuclein, prion protein (PrP) each cause neuropathic protein inclusions in the brain subregions, iron-responsive-like element (IRE-like) RNA stem-loops reside in their transcripts. APP and αsyn have a role in iron transport while gene duplications elevate the expression of their products to cause rare familial cases of AD and PDD. Of note, IRE-like sequences are responsive to excesses of brain iron in a potential feedback loop to accelerate neuronal ferroptosis and cognitive declines as well as amyloidosis. This pathogenic feedback is consistent with the translational control of the iron storage protein ferritin. We discuss how the IRE-like RNA motifs in the 5'UTRs of APP, alpha-synuclein and PrP mRNAs represent uniquely folded drug targets for therapies to prevent perturbed iron homeostasis that accelerates AD, PD, PD dementia (PDD) and Lewy body dementia, thus preventing cognitive deficits. Inhibition of alpha-synuclein translation is an option to block manganese toxicity associated with early childhood cognitive problems and manganism while Pb toxicity is epigenetically associated with attention deficit and later-stage AD. Pathologies of heavy metal toxicity centered on an embargo of iron export may be treated with activators of APP and ferritin and inhibitors of alpha-synuclein translation.


Assuntos
Regiões 5' não Traduzidas/efeitos dos fármacos , Peptídeos beta-Amiloides/metabolismo , Ferritinas/metabolismo , Ferroptose/fisiologia , Intoxicação por Metais Pesados/metabolismo , Proteínas Reguladoras de Ferro/metabolismo , Transtornos Neurocognitivos/metabolismo , Doenças Neurodegenerativas/metabolismo , Fármacos Neuroprotetores/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , alfa-Sinucleína/metabolismo , Peptídeos beta-Amiloides/efeitos dos fármacos , Animais , Ferritinas/efeitos dos fármacos , Ferroptose/efeitos dos fármacos , Intoxicação por Metais Pesados/tratamento farmacológico , Intoxicação por Metais Pesados/fisiopatologia , Humanos , Proteínas Reguladoras de Ferro/efeitos dos fármacos , Transtornos Neurocognitivos/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , alfa-Sinucleína/efeitos dos fármacos
6.
Int J Mol Sci ; 20(4)2019 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-30823541

RESUMO

The therapeutic value of inhibiting translation of the amyloid precursor protein (APP) offers the possibility to reduce neurotoxic amyloid formation, particularly in cases of familial Alzheimer's disease (AD) caused by APP gene duplications (Dup⁻APP) and in aging Down syndrome individuals. APP mRNA translation inhibitors such as the anticholinesterase phenserine, and high throughput screened molecules, selectively inhibited the uniquely folded iron-response element (IRE) sequences in the 5'untranslated region (5'UTR) of APP mRNA and this class of drug continues to be tested in a clinical trial as an anti-amyloid treatment for AD. By contrast, in younger age groups, APP expression is not associated with amyloidosis, instead it acts solely as a neuroprotectant while facilitating cellular ferroportin-dependent iron efflux. We have reported that the environmental metallotoxins Lead (Pb) and manganese (Mn) cause neuronal death by interfering with IRE dependent translation of APP and ferritin. The loss of these iron homeostatic neuroprotectants thereby caused an embargo of iron (Fe) export from neurons as associated with excess unstored intracellular iron and the formation of toxic reactive oxidative species (ROS). We propose that APP 5'UTR directed translation activators can be employed therapeutically to protect neurons exposed to high acute Pb and/or Mn exposure. Certainly, high potency APP translation activators, exemplified by the Food and Drug Administration (FDA) pre-approved M1 muscarinic agonist AF102B and high throughput-screened APP 5'UTR translation activators, are available for drug development to treat acute toxicity caused by Pb/Mn exposure to neurons. We conclude that APP translation activators can be predicted to prevent acute metal toxicity to neurons by a mechanism related to the 5'UTR specific yohimbine which binds and targets the canonical IRE RNA stem loop as an H-ferritin translation activator.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Ferritinas/genética , Proteínas Reguladoras de Ferro/genética , Intoxicação do Sistema Nervoso por Chumbo/tratamento farmacológico , Intoxicação por Manganês/tratamento farmacológico , Agonistas Muscarínicos/uso terapêutico , Quinuclidinas/uso terapêutico , Elementos de Resposta/fisiologia , Tiofenos/uso terapêutico , Regiões 5' não Traduzidas/efeitos dos fármacos , Doença Aguda , Doença de Alzheimer/metabolismo , Animais , Síndrome de Down/metabolismo , Humanos , Ferro/metabolismo , Camundongos , Agonistas Muscarínicos/farmacologia , Neurônios/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Quinuclidinas/farmacologia , RNA Mensageiro/genética , Ratos , Tiofenos/farmacologia
7.
J Neurochem ; 147(6): 831-848, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30152072

RESUMO

For more than 150 years, it is known that occupational overexposure of manganese (Mn) causes movement disorders resembling Parkinson's disease (PD) and PD-like syndromes. However, the mechanisms of Mn toxicity are still poorly understood. Here, we demonstrate that Mn dose- and time-dependently blocks the protein translation of amyloid precursor protein (APP) and heavy-chain Ferritin (H-Ferritin), both iron homeostatic proteins with neuroprotective features. APP and H-Ferritin are post-transcriptionally regulated by iron responsive proteins, which bind to homologous iron responsive elements (IREs) located in the 5'-untranslated regions (5'-UTRs) within their mRNA transcripts. Using reporter assays, we demonstrate that Mn exposure repressed the 5'-UTR-activity of APP and H-Ferritin, presumably via increased iron responsive proteins-iron responsive elements binding, ultimately blocking their protein translation. Using two specific Fe2+ -specific probes (RhoNox-1 and IP-1) and ion chromatography inductively coupled plasma mass spectrometry (IC-ICP-MS), we show that loss of the protective axis of APP and H-Ferritin resulted in unchecked accumulation of redox-active ferrous iron (Fe2+ ) fueling neurotoxic oxidative stress. Enforced APP expression partially attenuated Mn-induced generation of cellular and lipid reactive oxygen species and neurotoxicity. Lastly, we could validate the Mn-mediated suppression of APP and H-Ferritin in two rodent in vivo models (C57BL6/N mice and RjHan:SD rats) mimicking acute and chronic Mn exposure. Together, these results suggest that Mn-induced neurotoxicity is partly attributable to the translational inhibition of APP and H-Ferritin resulting in impaired iron metabolism and exacerbated neurotoxic oxidative stress. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Assuntos
Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Apoferritinas/antagonistas & inibidores , Ferro/metabolismo , Intoxicação por Manganês/metabolismo , Regiões 5' não Traduzidas , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Apoferritinas/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Modificação Traducional de Proteínas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
8.
Neurotherapeutics ; 15(1): 156-175, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29340929

RESUMO

S-Adenosyl methionine (SAMe), as a major methyl donor, exerts its influence on central nervous system function through cellular transmethylation pathways, including the methylation of DNA, histones, protein phosphatase 2A, and several catecholamine moieties. Based on available evidence, this review focuses on the lifelong range of severe neuropsychiatric and neurodegenerative diseases and their associated neuropathologies, which have been linked to the deficiency/load of SAMe production or/and the disturbance in transmethylation pathways. Also included in this review are the present-day applications of SAMe in the treatment in these diseases in each age group.


Assuntos
Transtornos Mentais/metabolismo , Doenças Neurodegenerativas/metabolismo , S-Adenosilmetionina/metabolismo , Animais , Metilação de DNA , Epigênese Genética , Humanos , Transtornos Mentais/genética , Metilação , Doenças Neurodegenerativas/genética , Transdução de Sinais
9.
Nat Med ; 23(3): 355-360, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28134928

RESUMO

Opiates are essential for treating pain, but termination of opiate therapy can cause a debilitating withdrawal syndrome in chronic users. To alleviate or avoid the aversive symptoms of withdrawal, many of these individuals continue to use opiates. Withdrawal is therefore a key determinant of opiate use in dependent individuals, yet its underlying mechanisms are poorly understood and effective therapies are lacking. Here, we identify the pannexin-1 (Panx1) channel as a therapeutic target in opiate withdrawal. We show that withdrawal from morphine induces long-term synaptic facilitation in lamina I and II neurons within the rodent spinal dorsal horn, a principal site of action for opiate analgesia. Genetic ablation of Panx1 in microglia abolished the spinal synaptic facilitation and ameliorated the sequelae of morphine withdrawal. Panx1 is unique in its permeability to molecules up to 1 kDa in size and its release of ATP. We show that Panx1 activation drives ATP release from microglia during morphine withdrawal and that degrading endogenous spinal ATP by administering apyrase produces a reduction in withdrawal behaviors. Conversely, we found that pharmacological inhibition of ATP breakdown exacerbates withdrawal. Treatment with a Panx1-blocking peptide (10panx) or the clinically used broad-spectrum Panx1 blockers, mefloquine or probenecid, suppressed ATP release and reduced withdrawal severity. Our results demonstrate that Panx1-mediated ATP release from microglia is required for morphine withdrawal in rodents and that blocking Panx1 alleviates the severity of withdrawal without affecting opiate analgesia.


Assuntos
Comportamento Animal/efeitos dos fármacos , Conexinas/genética , Microglia/efeitos dos fármacos , Morfina/efeitos adversos , Entorpecentes/efeitos adversos , Proteínas do Tecido Nervoso/genética , Células do Corno Posterior/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/genética , Trifosfato de Adenosina/metabolismo , Animais , Apirase/farmacologia , Western Blotting , Técnicas de Cultura de Células , Técnicas de Cocultura , Conexinas/antagonistas & inibidores , Conexinas/metabolismo , Mefloquina/farmacologia , Camundongos , Microglia/metabolismo , Naloxona/farmacologia , Antagonistas de Entorpecentes/efeitos adversos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nociceptividade/efeitos dos fármacos , Células do Corno Posterior/metabolismo , Probenecid/farmacologia , Ratos , Síndrome de Abstinência a Substâncias/etiologia , Síndrome de Abstinência a Substâncias/metabolismo
10.
J Neurochem ; 138(3): 479-94, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27206843

RESUMO

Iron supplementation ameliorates the neurotoxicity of the environmental contaminant lead (Pb); however, the mechanism remains undefined. Iron is an essential nutrient but high levels are toxic due to the catalytic generation of destructive hydroxyl radicals. Using human neuroblastoma SH-SY5Y cells to model human neurons, we investigated the effect of Pb on proteins of iron homeostasis: the Alzheimer's amyloid precursor protein (APP), which stabilizes the iron exporter ferroportin 1; and, the heavy subunit of the iron-storage protein, ferritin (FTH). Lead (Pb(II) and Pb(IV) inhibited APP translation and raised cytosolic iron(II). Lead also increased iron regulatory protein-1 binding to the cognate 5'untranslated region-specific iron-responsive element (IRE) of APP and FTH mRNAs. Concurrent iron treatment rescued cells from Pb toxicity by specifically restoring APP synthesis, i.e. levels of the APP-related protein, APLP-2, were unchanged. Significantly, iron/IRE-independent over-expression of APP695  protected SH-SY5Y cells from Pb toxicity, demonstrating that APP plays a key role in maintaining safe levels of intracellular iron. Overall, our data support a model of neurotoxicity where Pb enhances iron regulatory protein/IRE-mediated repression of APP and FTH translation. We propose novel treatment options for Pb poisoning to include chelators and the use of small molecules to maintain APP and FTH translation. We propose the following cascade for Lead (Pb) toxicity to neurons; by targeting the interaction between Iron regulatory protein-1 and Iron-responsive elements, Pb caused translational repression of proteins that control intracellular iron homeostasis, including the Alzheimer's amyloid precursor protein (APP) that stabilizes the iron exporter ferroportin, and the ferroxidase heavy subunit of the iron-storage protein, ferritin. When unregulated, IRE-independent over-expression of APP695 protected SH-SY5Y neurons from Pb toxicity. There is a novel and key role for APP in maintaining safe levels of intracellular iron pertinent to lead toxicity.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Homeostase/efeitos dos fármacos , Chumbo/toxicidade , Neurônios/efeitos dos fármacos , Síndromes Neurotóxicas/tratamento farmacológico , Precursor de Proteína beta-Amiloide/genética , Desferroxamina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Ferro/metabolismo , Quelantes de Ferro/farmacologia , Neuroblastoma/metabolismo , Neurônios/metabolismo , Síndromes Neurotóxicas/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos
11.
PLoS One ; 11(1): e0145184, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26799482

RESUMO

The innate immune response is characterized by activation of transcription factors, nuclear factor kappa B and activator protein-1 and their downstream targets, the pro-inflammatory cytokines including interleukin 1ß and interleukin 6. Normal development of this response in the intestine is critical to survival of the human neonate and delays can cause the onset of devastating inflammatory diseases such as necrotizing enterocolitis. Previous studies have addressed the role of nuclear factor kappa B in the development of the innate immune response in the enterocyte, however despite its central role in the control of multiple pro-inflammatory cytokine genes, little is known on the role of Activator Protein 1 in this response in the enterocyte. Here we show that the canonical Activator Protein 1 members, cJun and cFos and their upstream kinases JNK and p38 play an essential role in the regulation of interleukin 6 in the immature enterocyte. Our data supports a model whereby the cFos/cJun heterodimer and the more potent cJun homodimer downstream of JNK are replaced by less efficient JunD containing dimers, contributing to the decreased responsiveness to interleukin 1ß and decreased interleukin 6 secretion observed in the mature enterocyte. The tissue specific expression of JunB in colonocytes and colon derived tissues together with its ability to repress Interleukin-1ß induction of an Interleukin-6 gene reporter in the NCM-460 colonocyte suggests that induction of JunB containing dimers may offer an attractive therapeutic strategy for the control of IL-6 secretion during inflammatory episodes in this area of the intestine.


Assuntos
Enterócitos/fisiologia , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Linhagem Celular , Enterócitos/citologia , Enterócitos/efeitos dos fármacos , Xenoenxertos , Humanos , Interleucina-1beta/farmacologia , Interleucina-6/genética , Intestinos/citologia , Intestinos/embriologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos SCID , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Técnicas de Cultura de Órgãos , Fosforilação , Elementos de Resposta , Transdução de Sinais , Fator de Transcrição AP-1/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Urology ; 85(5): 1025-1033, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25917728

RESUMO

Intravesical local anesthetics, in a wide variety of combinations, are increasingly used to treat patients with interstitial cystitis-bladder pain syndrome (IC/BPS). Lidocaine has demonstrated properties that block the neuroinflammatory cycle associated with IC/BPS at many of the interactive points in this cycle. Intravesical lidocaine has been shown to assist in identifying the bladder as the source of pain in patients with pelvic pain. An appreciation of these anti-inflammatory effects and of the pharmacokinetics of intravesical lidocaine in patients with IC/BPS could lead to a safe and effective diagnosis and treatment for an as yet unidentified subset of patients in the IC/BPS spectrum.


Assuntos
Anestésicos Locais/uso terapêutico , Cistite Intersticial/diagnóstico , Cistite Intersticial/tratamento farmacológico , Lidocaína/uso terapêutico , Feminino , Humanos
13.
J Mol Cell Biol ; 7(2): 168-79, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25701657

RESUMO

Otoferlin, an integral membrane protein implicated in a late stage of exocytosis, has been reported to play a critical role in hearing although the underlying mechanisms remain elusive. However, its widespread tissue distribution infers a more ubiquitous role in synaptic vesicle trafficking. Glutamate, an excitatory neurotransmitter, is converted to its inhibitory counterpart, γ-aminobutyric acid (GABA), by L-glutamic acid decarboxylase (GAD), which exists in soluble (GAD67) and membrane-bound (GAD65) forms. For the first time, we have revealed a close association between otoferlin and GAD65 in both HEK293 and neuronal cells, including SH-SY5Y neuroblastoma and primary rat hippocampus cells, showing a direct interaction between GAD65 and otoferlin's C2 domains. In primary rat hippocampus cells, otoferlin and GAD65 co-localized in a punctate pattern within the cell body, as well as in the axon along the path of vesicular traffic. Significantly, GABA is virtually abolished in otoferlin-knockdown neuronal cells whereas otoferlin overexpression markedly increases endogenous GABA. GABA attenuation in otoferlin-knockdown primary cells is correlated with diminished L-type calcium current. This previously unknown and close correlation demonstrates that otoferlin, through GAD65, modulates GABAergic activity. The discovery of otoferlin-GAD65 functional coupling provides a new avenue for understanding the molecular mechanism by which otoferlin functions in neurological pathways.


Assuntos
Neurônios GABAérgicos/fisiologia , Glutamato Descarboxilase/fisiologia , Proteínas de Membrana/fisiologia , Animais , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio , Linhagem Celular Tumoral , Células Cultivadas , Células HEK293 , Hipocampo/citologia , Humanos , Transporte Proteico , Ratos Sprague-Dawley , Ácido gama-Aminobutírico/biossíntese
14.
Behav Pharmacol ; 24(3): 207-13, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23591124

RESUMO

In addition to sensory disturbances, neuropathic pain is associated with an ongoing and persistent negative affective state. This condition may be reflected as altered sensitivity to rewarding stimuli. We examined this hypothesis by testing whether the rewarding properties of morphine are altered in a rat model of neuropathic pain. Neuropathic pain was induced by chronic constriction of the common sciatic nerve. Drug reward was assessed using an unbiased, three-compartment conditioned place preference (CPP) paradigm. The rats underwent two habituation sessions beginning 6 days after surgery. Over the next 8 days, they were injected with drug or vehicle and were confined to one CPP compartment for 30 min. On the following test day, the rats had access to all three compartments for 30 min. Consistent with the literature, systemic administration of morphine dose-dependently increased the CPP in pain-naive animals. In rats with neuropathic pain, however, the dose-dependent effects of morphine were in a bell-shaped curve, with a low dose of morphine (2 mg/kg) producing a greater CPP than a higher dose of morphine (8 mg/kg). In a separate group of animals, acute administration of morphine reversed mechanical allodynia in animals with neuropathic pain at the same doses that produced a CPP. The increased potency of systemic morphine to produce a CPP in animals with neuropathic pain suggests that the motivation for opioid-induced reward is different in the two states.


Assuntos
Analgésicos Opioides/uso terapêutico , Condicionamento Operante/efeitos dos fármacos , Morfina/uso terapêutico , Neuralgia/tratamento farmacológico , Neuralgia/psicologia , Recompensa , Análise de Variância , Animais , Modelos Animais de Doenças , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Masculino , Neuralgia/fisiopatologia , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Long-Evans , Fatores de Tempo
15.
Anesth Analg ; 114(3): 690-3, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22190548

RESUMO

We evaluated the antinociceptive effects of acute and chronic morphine administered spinally via lumbar puncture in intrathecally catheterized and sham-surgery rats. The effects of acute morphine did not differ between groups. Catheterized rats developed tolerance to chronic morphine more rapidly, compared with sham and naive rats. Therefore, catheter presence facilitated development of opioid antinociceptive tolerance. Spinal astrogliosis, determined by measurement of 3-dimensional cell volumes, was observed in catheterized rats as indicated by significantly larger cell volumes compared with surgery-naive controls. Gliosis induced by chronic intrathecal morphine administered to surgery-naive animals was comparable to that observed in saline-treated catheterized rats.


Assuntos
Cateteres de Demora , Tolerância a Medicamentos/fisiologia , Morfina/administração & dosagem , Medição da Dor/efeitos dos fármacos , Raquianestesia/métodos , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Cateterismo/métodos , Injeções Espinhais , Masculino , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley
16.
J Neural Transm (Vienna) ; 118(3): 493-507, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21221670

RESUMO

Increased brain α-synuclein (SNCA) protein expression resulting from gene duplication and triplication can cause a familial form of Parkinson's disease (PD). Dopaminergic neurons exhibit elevated iron levels that can accelerate toxic SNCA fibril formation. Examinations of human post mortem brain have shown that while mRNA levels for SNCA in PD have been shown to be either unchanged or decreased with respect to healthy controls, higher levels of insoluble protein occurs during PD progression. We show evidence that SNCA can be regulated via the 5'untranslated region (5'UTR) of its transcript, which we modeled to fold into a unique RNA stem loop with a CAGUGN apical loop similar to that encoded in the canonical iron-responsive element (IRE) of L- and H-ferritin mRNAs. The SNCA IRE-like stem loop spans the two exons that encode its 5'UTR, whereas, by contrast, the H-ferritin 5'UTR is encoded by a single first exon. We screened a library of 720 natural products (NPs) for their capacity to inhibit SNCA 5'UTR driven luciferase expression. This screen identified several classes of NPs, including the plant cardiac glycosides, mycophenolic acid (an immunosuppressant and Fe chelator), and, additionally, posiphen was identified to repress SNCA 5'UTR conferred translation. Western blotting confirmed that Posiphen and the cardiac glycoside, strophanthidine, selectively blocked SNCA expression (~1 µM IC(50)) in neural cells. For Posiphen this inhibition was accelerated in the presence of iron, thus providing a known APP-directed lead with potential for use as a SNCA blocker for PD therapy. These are candidate drugs with the potential to limit toxic SNCA expression in the brains of PD patients and animal models in vivo.


Assuntos
Antibacterianos/farmacologia , Encéfalo/metabolismo , Cardenolídeos/farmacologia , Dicloxacilina/farmacologia , Ferro/metabolismo , Emaranhados Neurofibrilares/metabolismo , Estrofantidina/farmacologia , alfa-Sinucleína/metabolismo , Regiões 5' não Traduzidas/genética , Western Blotting , Encéfalo/patologia , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Emaranhados Neurofibrilares/efeitos dos fármacos , Emaranhados Neurofibrilares/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , alfa-Sinucleína/genética
17.
J Biol Chem ; 285(41): 31217-32, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20558735

RESUMO

Iron influx increases the translation of the Alzheimer amyloid precursor protein (APP) via an iron-responsive element (IRE) RNA stem loop in its 5'-untranslated region. Equal modulated interaction of the iron regulatory proteins (IRP1 and IRP2) with canonical IREs controls iron-dependent translation of the ferritin subunits. However, our immunoprecipitation RT-PCR and RNA binding experiments demonstrated that IRP1, but not IRP2, selectively bound the APP IRE in human neural cells. This selective IRP1 interaction pattern was evident in human brain and blood tissue from normal and Alzheimer disease patients. We computer-predicted an optimal novel RNA stem loop structure for the human, rhesus monkey, and mouse APP IREs with reference to the canonical ferritin IREs but also the IREs encoded by erythroid heme biosynthetic aminolevulinate synthase and Hif-2α mRNAs, which preferentially bind IRP1. Selective 2'-hydroxyl acylation analyzed by primer extension analysis was consistent with a 13-base single-stranded terminal loop and a conserved GC-rich stem. Biotinylated RNA probes deleted of the conserved CAGA motif in the terminal loop did not bind to IRP1 relative to wild type probes and could no longer base pair to form a predicted AGA triloop. An AGU pseudo-triloop is key for IRP1 binding to the canonical ferritin IREs. RNA probes encoding the APP IRE stem loop exhibited the same high affinity binding to rhIRP1 as occurs for the H-ferritin IRE (35 pm). Intracellular iron chelation increased binding of IRP1 to the APP IRE, decreasing intracellular APP expression in SH-SY5Y cells. Functionally, shRNA knockdown of IRP1 caused increased expression of neural APP consistent with IRP1-APP IRE-driven translation.


Assuntos
Regiões 5' não Traduzidas , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Proteína 1 Reguladora do Ferro/metabolismo , Conformação de Ácido Nucleico , Biossíntese de Proteínas , Receptores de Superfície Celular/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/patologia , Linhagem Celular Tumoral , Humanos , Ferro/metabolismo , Proteína 1 Reguladora do Ferro/genética , Macaca mulatta , Camundongos , Nexinas de Proteases , Receptores de Superfície Celular/genética
18.
J Neuroinflammation ; 7: 27, 2010 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-20398373

RESUMO

Tumor necrosis factor alpha (TNF-alpha) was discovered more than a century ago, and its known roles have extended from within the immune system to include a neuro-inflammatory domain in the nervous system. Neuropathic pain is a recognized type of pathological pain where nociceptive responses persist beyond the resolution of damage to the nerve or its surrounding tissue. Very often, neuropathic pain is disproportionately enhanced in intensity (hyperalgesia) or altered in modality (hyperpathia or allodynia) in relation to the stimuli. At time of this writing, there is as yet no common consensus about the etiology of neuropathic pain - possible mechanisms can be categorized into peripheral sensitization and central sensitization of the nervous system in response to the nociceptive stimuli. Animal models of neuropathic pain based on various types of nerve injuries (peripheral versus spinal nerve, ligation versus chronic constrictive injury) have persistently implicated a pivotal role for TNF-alpha at both peripheral and central levels of sensitization. Despite a lack of success in clinical trials of anti-TNF-alpha therapy in alleviating the sciatic type of neuropathic pain, the intricate link of TNF-alpha with other neuro-inflammatory signaling systems (e.g., chemokines and p38 MAPK) has indeed inspired a systems approach perspective for future drug development in treating neuropathic pain.


Assuntos
Dor/patologia , Doenças do Sistema Nervoso Periférico/patologia , Fator de Necrose Tumoral alfa/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Citocinas/metabolismo , Humanos , Inflamação/patologia , Neuroglia/fisiologia , Plasticidade Neuronal/fisiologia , Dor/etiologia , Traumatismos dos Nervos Periféricos , Doenças do Sistema Nervoso Periférico/complicações , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Biochim Biophys Acta ; 1790(7): 615-28, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19166904

RESUMO

Recent studies that alleles in the hemochromatosis gene may accelerate the onset of Alzheimer's disease by five years have validated interest in the model in which metals (particularly iron) accelerate disease course. Biochemical and biophysical measurements demonstrated the presence of elevated levels of neurotoxic copper zinc and iron in the brains of AD patients. Intracellular levels of APP holoprotein were shown to be modulated by iron by a mechanism that is similar to the translation control of the ferritin L- and H mRNAs by iron-responsive element (IRE) RNA stem loops in their 5' untranslated regions (5'UTRs). More recently a putative IRE-like sequence was hypothesized present in the Parkinsons's alpha synuclein (ASYN) transcript (see [A.L. Friedlich, R.E. Tanzi, J.T. Rogers, The 5'-untranslated region of Parkinson's disease alpha-synuclein messenger RNA contains a predicted iron responsive element, Mol. Psychiatry 12 (2007) 222-223. [6]]). Together with the demonstration of metal dependent translation of APP mRNA, the involvement of metals in the plaque of AD patients and of increased iron in striatal neurons in the substantia nigra (SN) of Parkinson's disease patients have stimulated the development of metal attenuating agents and iron chelators as a major new therapeutic strategy for the treatment of these neurodegenerative diseases. In the case of AD, metal based therapeutics may ultimately prove more cost effective than the use of an amyloid vaccine as the preferred anti-amyloid therapeutic strategy to ameliorate the cognitive decline of AD patients.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Ferro/metabolismo , Doenças Neurodegenerativas/genética , alfa-Sinucleína/genética , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Sequência de Bases , Quelantes/uso terapêutico , Ferritinas/genética , Homeostase , Humanos , Inflamação , Ferro/uso terapêutico , Dados de Sequência Molecular , Doença de Parkinson/genética , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Receptores da Transferrina/genética , Proteínas tau/metabolismo
20.
Biochem Soc Trans ; 36(Pt 6): 1282-7, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19021541

RESUMO

The essential metals iron, zinc and copper deposit near the Abeta (amyloid beta-peptide) plaques in the brain cortex of AD (Alzheimer's disease) patients. Plaque-associated iron and zinc are in neurotoxic excess at 1 mM concentrations. APP (amyloid precursor protein) is a single transmembrane metalloprotein cleaved to generate the 40-42-amino-acid Abetas, which exhibit metal-catalysed neurotoxicity. In health, ubiquitous APP is cleaved in a non-amyloidogenic pathway within its Abeta domain to release the neuroprotective APP ectodomain, APP(s). To adapt and counteract metal-catalysed oxidative stress, as during reperfusion from stroke, iron and cytokines induce the translation of both APP and ferritin (an iron storage protein) by similar mechanisms. We reported that APP was regulated at the translational level by active IL (interleukin)-1 (IL-1-responsive acute box) and IRE (iron-responsive element) RNA stem-loops in the 5' untranslated region of APP mRNA. The APP IRE is homologous with the canonical IRE RNA stem-loop that binds the iron regulatory proteins (IRP1 and IRP2) to control intracellular iron homoeostasis by modulating ferritin mRNA translation and transferrin receptor mRNA stability. The APP IRE interacts with IRP1 (cytoplasmic cis-aconitase), whereas the canonical H-ferritin IRE RNA stem-loop binds to IRP2 in neural cell lines, and in human brain cortex tissue and in human blood lysates. The same constellation of RNA-binding proteins [IRP1/IRP2/poly(C) binding protein] control ferritin and APP translation with implications for the biology of metals in AD.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Ferritinas/genética , Ferro/metabolismo , Neurônios/patologia , Estresse Oxidativo , Biossíntese de Proteínas , Doença de Alzheimer/genética , Humanos , Neurônios/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA