Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Molecules ; 26(9)2021 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-33921982

RESUMO

Fourteen coumarin-derived compounds modified at the C3 carbon of coumarin with an α,ß-unsaturated ketone were synthesized. These compounds may be designated as chalcocoumarins (3-cinnamoyl-2H-chromen-2-ones). Both chalcones and coumarins are recognized scaffolds in medicinal chemistry, showing diverse biological and pharmacological properties among which neuroprotective activities and multiple enzyme inhibition, including mitochondrial enzyme systems, stand out. The evaluation of monoamine oxidase B (MAO-B) inhibitors has aroused considerable interest as therapeutic agents for neurodegenerative diseases such as Parkinson's. Of the fourteen chalcocumarins evaluated here against MAO-B, ChC4 showed the strongest activity in vitro, with IC50 = 0.76 ± 0.08 µM. Computational docking, molecular dynamics and MM/GBSA studies, confirm that ChC4 binds very stably to the active rMAO-B site, explaining the experimental inhibition data.


Assuntos
Chalconas/química , Cumarínicos/química , Inibidores da Monoaminoxidase/química , Inibidores da Monoaminoxidase/farmacologia , Monoaminoxidase/química , Animais , Sítios de Ligação , Relação Dose-Resposta a Droga , Humanos , Ligantes , Conformação Molecular , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Estrutura Molecular , Ligação Proteica , Ratos , Relação Estrutura-Atividade
2.
J Neurochem ; 137(4): 506-17, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26875993

RESUMO

The generation of abnormally high levels of reactive oxygen species (ROS) is linked to cellular dysfunction, including neuronal toxicity and neurodegeneration. However, physiological ROS production modulates redox-sensitive roles of several molecules such as transcription factors, signaling proteins, and cytoskeletal components. Changes in the functions of redox-sensitive proteins may be important for defining key aspects of stem cell proliferation and differentiation, neuronal maturation, and neuronal plasticity. In neurons, most of the studies have been focused on the pathological implications of such modifications and only very recently their essential roles in neuronal development and plasticity has been recognized. In this review, we discuss the participation of NADPH oxidases (NOXs) and a family of protein-methionine sulfoxide oxidases, named molecule interacting with CasLs, as regulated enzymatic sources of ROS production in neurons, and describes the contribution of ROS signaling to neurogenesis and differentiation, neurite outgrowth, and neuronal plasticity. We review the role of reactive oxygen species (ROS) in neurogenesis, axon growth, and guidance and NMDA-receptor-mediated plasticity, LTP, and memory. ROS participation is presented in the context of NADPH oxidase and MICAL functions and their importance for brain functions.


Assuntos
Neurogênese/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos , Oxirredução
3.
Antioxidants (Basel) ; 10(1)2021 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-33419006

RESUMO

Iron accumulation and neuroinflammation are pathological conditions found in several neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Iron and inflammation are intertwined in a bidirectional relationship, where iron modifies the inflammatory phenotype of microglia and infiltrating macrophages, and in turn, these cells secrete diffusible mediators that reshape neuronal iron homeostasis and regulate iron entry into the brain. Secreted inflammatory mediators include cytokines and reactive oxygen/nitrogen species (ROS/RNS), notably hepcidin and nitric oxide (·NO). Hepcidin is a small cationic peptide with a central role in regulating systemic iron homeostasis. Also present in the cerebrospinal fluid (CSF), hepcidin can reduce iron export from neurons and decreases iron entry through the blood-brain barrier (BBB) by binding to the iron exporter ferroportin 1 (Fpn1). Likewise, ·NO selectively converts cytosolic aconitase (c-aconitase) into the iron regulatory protein 1 (IRP1), which regulates cellular iron homeostasis through its binding to iron response elements (IRE) located in the mRNAs of iron-related proteins. Nitric oxide-activated IRP1 can impair cellular iron homeostasis during neuroinflammation, triggering iron accumulation, especially in the mitochondria, leading to neuronal death. In this review, we will summarize findings that connect neuroinflammation and iron accumulation, which support their causal association in the neurodegenerative processes observed in AD and PD.

4.
Front Neurosci ; 13: 48, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30809110

RESUMO

Iron and calcium share the common feature of being essential for normal neuronal function. Iron is required for mitochondrial function, synaptic plasticity, and the development of cognitive functions whereas cellular calcium signals mediate neurotransmitter exocytosis, axonal growth and synaptic plasticity, and control the expression of genes involved in learning and memory processes. Recent studies have revealed that cellular iron stimulates calcium signaling, leading to downstream activation of kinase cascades engaged in synaptic plasticity. The relationship between calcium and iron is Janus-faced, however. While under physiological conditions iron-mediated reactive oxygen species generation boosts normal calcium-dependent signaling pathways, excessive iron levels promote oxidative stress leading to the upsurge of unrestrained calcium signals that damage mitochondrial function, among other downstream targets. Similarly, increases in mitochondrial calcium to non-physiological levels result in mitochondrial dysfunction and a predicted loss of iron homeostasis. Hence, if uncontrolled, the iron/calcium self-feeding cycle becomes deleterious to neuronal function, leading eventually to neuronal death. Here, we review the multiple cell-damaging responses generated by the unregulated iron/calcium self-feeding cycle, such as excitotoxicity, free radical-mediated lipid peroxidation, and the oxidative modification of crucial components of iron and calcium homeostasis/signaling: the iron transporter DMT1, plasma membrane, and intracellular calcium channels and pumps. We discuss also how iron-induced dysregulation of mitochondrial calcium contributes to the generation of neurodegenerative conditions, including Alzheimer's disease (AD) and Parkinson's disease (PD).

5.
Prog Neurobiol ; 74(6): 323-49, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15649580

RESUMO

This review is focused on the structure and function of Alzheimer's amyloid deposits. Amyloid formation is a process in which normal well-folded cellular proteins undergo a self-assembly process that leads to the formation of large and ordered protein structures. Amyloid deposition, oligomerization, and higher order polymerization, and the structure adopted by these assemblies, as well as their functional relationship with cell biology are underscored. Numerous efforts have been directed to elucidate these issues and their relation with senile dementia. Significant advances made in the last decade in amyloid structure, dynamics and cell biology are summarized and discussed. The mechanism of amyloid neurotoxicity is discussed with emphasis on the Wnt signaling pathway. This review is focused on Alzheimer's amyloid fibrils in general and has been divided into two parts dealing with the structure and function of amyloid.


Assuntos
Doença de Alzheimer/metabolismo , Amiloide/química , Amiloide/metabolismo , Placa Amiloide/metabolismo , Doença de Alzheimer/fisiopatologia , Amiloide/toxicidade , Animais , Apoptose/fisiologia , Humanos , Modelos Biológicos , Modelos Moleculares , Necrose/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/toxicidade , Transdução de Sinais/fisiologia
6.
Fertil Steril ; 106(2): 393-401, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27117373

RESUMO

OBJECTIVE: To evaluate divalent metal transporter-1 (DMT1) expression in healthy women's and endometriosis patients' endometrium and to analyze DMT1 and ferritin light chain (Fn-L) expression modulation by iron overload and IL-1ß in endometrial stromal cells (ESCs). DESIGN: Observational and experimental study. SETTING: University hospital research laboratory. PATIENT(S): Thirty-one healthy women and 24 endometriosis patients. INTERVENTION(S): Menstrual, proliferative, and secretory endometrial biopsies. Isolated ESCs from seven endometrial biopsies incubated with IL-1ß or FeSO4 overload for 24 hours. MAIN OUTCOME MEASURE(S): Divalent metal transporter-1 endometrial protein expression assessed by immunohistochemistry and Western blot. Divalent metal transporter-1 and Fn-L proteins expression in stimulated ESCs evaluated by Western blot. RESULT(S): Divalent metal transporter-1 is expressed throughout the menstrual cycle in human endometrium. Four endometrial DMT1 variants were identified accordingly to their molecular weight: DMT-80, -65, -55, and -50. Endometrial expression of DMT-80 and -55 is higher in endometriosis patients than in healthy women. In ESCs, iron overload induces an overexpression of DMT-80, DMT-50, and Fn-L, whereas IL-1ß increases DMT-80 and -50 expressions and decreases Fn-L expression. CONCLUSION(S): Divalent metal transporter-1 overexpression in endometriosis patients' endometrium can increase iron influx to endometrial cells, inducing oxidative stress-mediated proinflammatory signaling. In turn, endometriosis-related conditions, as iron overload and inflammation (IL-1ß), enhance endometriosis patients endometrial DMT1 expression, creating a vicious circle on DMT-1-modulated pathways.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Endometriose/metabolismo , Endométrio/metabolismo , Células Estromais/metabolismo , Apoferritinas/metabolismo , Biópsia , Estudos de Casos e Controles , Endometriose/patologia , Endometriose/fisiopatologia , Endométrio/efeitos dos fármacos , Endométrio/patologia , Endométrio/fisiopatologia , Feminino , Compostos Ferrosos/farmacologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Interleucina-1beta/farmacologia , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/patologia , Ciclo Menstrual , Estresse Oxidativo , Transdução de Sinais , Células Estromais/efeitos dos fármacos , Células Estromais/patologia , Técnicas de Cultura de Tecidos
7.
Biochem Pharmacol ; 69(11): 1559-66, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15896335

RESUMO

Hereditary hemochromatosis (HH) is a condition in which intestinal iron absorption is greatly elevated. Present treatment is weekly phlebotomy, affecting quality of life and leading to recurrent infections. The iron transporter divalent metal transporter-1 (DMT-1) of enterocytes is responsible for iron uptake from the intestinal lumen; iron is further extruded into the blood by the basolateral transporter ferroportin-1. A therapeutic approach for HH could start with a long-term reduction of iron transport by reduction of DMT-1 levels. We designed an AAV vector coding for a short antisense RNA (AAV-DMT-1-AS) against DMT-1, which reduced iron uptake by 50-60% in human intestinal cells (Caco-2). At low infection levels, DMT-1 mRNA virtually disappeared, suggesting RNAi-like and/or RNase H antisense effects. DMT-1 mRNA levels returned to normal at higher infection levels, indicating that an additional mechanism of mRNA occupation, able to block DMT-1 translation and to avoid feedback regulation by iron responsive elements (IRE), also exists. Cell morphology was normal in all cases and no increases in the interferon-related responses, measured by (a) 2'-5' A oligo synthetase (b) IFITM1 and (c) ISGF3gamma mRNA levels, were observed. Studies presented herein indicate that enterocyte targeting with a gene coding for a short antisense against iron transport blocks enterocyte iron uptake, which may have therapeutic value.


Assuntos
Dependovirus/genética , Marcação de Genes/métodos , Hemocromatose/metabolismo , Mucosa Intestinal/metabolismo , Ferro/antagonistas & inibidores , RNA Antissenso/administração & dosagem , Células CACO-2 , Linhagem Celular , Vetores Genéticos , Hemocromatose/genética , Hemocromatose/terapia , Humanos , Intestinos/efeitos dos fármacos , Ferro/metabolismo , RNA Antissenso/genética
8.
Fertil Steril ; 103(2): 439-47, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25500022

RESUMO

OBJECTIVE: To evaluate the effect of iron overload on nuclear factor kappa-B (NF-κB) activation in human endometrial stromal cells (ESCs). DESIGN: Experimental study. SETTING: University hospital research laboratory. PATIENT(S): Ten healthy women. INTERVENTION(S): Isolated ESCs from endometrial biopsies were incubated with 50 µM FeSO(4) or vehicle. The NF-κB inhibitor [5-(p-fluorophenyl)-2-ureido] thiophene-3-carboxamide (TPCA-1), which inhibits IKKß, the kinase of IκBα (inhibitory protein of NF-κB), was used to prevent iron overload-stimulated NF-κB changes in ESCs. MAIN OUTCOME MEASURE(S): NF-κB activation was assessed by p65:DNA-binding activity immunodetection assay. IκBα, p65, and intercellular adhesion molecule (ICAM)-1 proteins expression was evaluated by Western blots. ESC soluble ICAM (sICAM)-1 secretion was measured by ELISA using conditioned medium. RESULT(S): Iron overload increased p65:DNA-binding activity and decreased IκBα and p65 cytoplasmic expression in ESCs after 30 minutes of incubation as compared with the basal condition. ESC ICAM-1 expression and sICAM-1 secretion were higher after 24 hours of iron overload treatment than in the absence of treatment. TPCA-1 prevented the iron overload-induced increase of p65:DNA binding and IκBα degradation. CONCLUSION(S): Iron overload activates IKKß in ESCs, stimulating the NF-κB pathway and increasing ICAM-1 expression and sICAM-1 secretion. These results suggest that iron overload induces a proendometriotic phenotype on healthy ESCs, which could participate in endometriosis pathogenesis and development.


Assuntos
Endometriose/metabolismo , Endométrio/citologia , Endométrio/metabolismo , Sobrecarga de Ferro/metabolismo , NF-kappa B/metabolismo , Adulto , Amidas/farmacologia , Células Cultivadas , Endometriose/induzido quimicamente , Endométrio/efeitos dos fármacos , Feminino , Compostos Férricos/toxicidade , Humanos , Sobrecarga de Ferro/induzido quimicamente , NF-kappa B/antagonistas & inibidores , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Tiofenos/farmacologia
9.
Neurotox Res ; 17(3): 238-47, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19655216

RESUMO

Iron is essential for crucial neuronal functions but is also highly toxic in excess. Neurons acquire iron through transferrin receptor-mediated endocytosis and via the divalent metal transporter 1 (DMT1). The N-terminus (1A, 1B) and C-terminus (+IRE, -IRE) splice variants of DMT1 originate four protein isoforms, all of which supply iron to cells. Diverse physiological or pathological conditions induce differential DMT1 variant expression, which are cell-type dependent. Hence, it becomes relevant to ascertain if activation of neuronal plasticity processes that require functional N-methyl D: -aspartate (NMDA) receptors, including in vitro stimulation of NMDA receptor-mediated signaling and spatial memory training, selectively modify DMT1 variant expression. Here, we report for the first time that brief (5 min) exposure of primary hippocampal cultures to NMDA (50 muM) increased 24 h later the expression of DMT1-1B and DMT1+IRE, but not of DMT1-IRE mRNA. In contrast, endogenous DMT1 mRNA levels remained unaffected following 6 h incubation with brain-derived nerve factor. NMDA (25-50 muM) also enhanced DMT1 protein expression 24-48 h later; this enhancement was abolished by the transcription inhibitor actinomycin D and by the NMDA receptor antagonist MK-801, implicating NMDA receptors in de novo DMT1 expression. Additionally, spatial memory training enhanced DMT1-1B and DMT1+IRE expression and increased DMT1 protein content in rat hippocampus, where the exon1A variant was not found. These results suggest that NMDA receptor-dependent plasticity processes stimulate expression of the iron transporter DMT1-1B+IRE isoform, which presumably plays a significant role in hippocampal spatial memory formation.


Assuntos
Proteínas de Transporte de Cátions/genética , Regulação da Expressão Gênica/fisiologia , Hipocampo/metabolismo , Memória/fisiologia , RNA Mensageiro/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Proteínas de Transporte de Cátions/metabolismo , Dactinomicina/farmacologia , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , N-Metilaspartato/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Técnicas de Cultura de Tecidos
10.
Biol Res ; 39(1): 103-4, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16629170

RESUMO

Brain cells have a highly active oxidative metabolism, yet they contain only low to moderate superoxide dismutase and catalase activities. Thus, their antioxidant defenses rely mainly on cellular reduced glutathione levels. In this work, in cortical neurons we characterized viability and changes in reduced and oxidized glutathione levels in response to a protocol of iron accumulation. We found that massive death occurred after 2 days in culture with 10 microM Fe. Surviving cells developed an adaptative response that included increased synthesis of GSH and the maintenance of a glutathione-based reduction potential. These results highlight the fundamental role of glutathione homeostasis in the antioxidant response and provide novel insights into the adaptative mechanisms of neurons subjected to progressive iron loads.


Assuntos
Córtex Cerebral/citologia , Glutationa/metabolismo , Ferro/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Animais , Morte Celular/efeitos dos fármacos , Córtex Cerebral/metabolismo , Dissulfeto de Glutationa/metabolismo , Homeostase , Ferro/farmacologia , Neurônios/química , Oxirredução , Ratos , Fatores de Tempo
11.
Biol Res ; 39(1): 199-201, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16629182

RESUMO

There is increasing evidence that accumulation of redox-active iron in mitochondria leads to oxidative damage and contributes to various neurodegenerative diseases, such as Friedreich's ataxia and Parkinsons disease. In this work, we examined the existence of regulatory mechanisms for mitochondrial iron uptake and storage. To that end, we used rhodamine B-[(1,10-phenanthrolin-5-yl)amino carbonyl] benzyl ester, a new fluorescent iron-sensitive probe that is targeted specifically to the mitochondrion. We found that extracellular iron was incorporated readily into mitochondria in an apparently saturable process. Moreover, the rate of iron incorporation responded to the Fe status of the cell, an indication that the mitochondrion actively regulates its iron content.


Assuntos
Corantes Fluorescentes , Ferro/metabolismo , Mitocôndrias/metabolismo , Rodaminas , Linhagem Celular , Humanos , Fatores de Tempo
12.
Biometals ; 16(1): 215-23, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12572681

RESUMO

Oxidative stress phenomena have been related with the onset of neurodegenerative diseases. Particularly in Alzheimer Disease (AD), oxygen reactive species (ROS) and its derivatives can be found in brain samples of postmortem AD patients. However, the mechanisms by which oxygen reactive species can alter neuronal function are still not elucidated. There is a growing amount of evidence pointing to a role for mitochondrial damage as the source of free radicals involved in oxidative stress. Among the species that participate in the production of oxygen reactive radicals, transition metals are one of the most important. Several reports have implicated the involvement of redox-active metals with the onset of different neurodegenerative diseases such as Alzheimer's Disease (AD), Progressive Supranuclear Palsy (PSP), Amyotrophic Lateral Sclerosis (ALS) and Parkinson's Disease (PD). On the other hand, our previous studies have indicated that A beta-induced deregulation of the protein kinase Cdk5 associated with tau protein hyperphosphorylation constitute a critical pathway toward neurodegeneration. In the current paper we have shown that iron induces an imbalance in the function of Cdk5/p25 system of hippocampal neurons, resulting in a marked decrease in tau phosphorylation at the typical Alzheimer's epitopes. The loss of phosphorylated tau epitopes correlated with an increase in 4-hydroxy-nonenal (HNE) adducts revealing damage by oxidative stress. This effects on tau phosphorylation patterns seems to be a consequence of a decrease in the Cdk5/p25 complex activity that appears to result from a depletion of the activator p25, a mechanism in which calcium transients could be implicated.


Assuntos
Hipocampo/fisiologia , Ferro/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Proteínas tau/metabolismo , Animais , Células Cultivadas , Embrião de Mamíferos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Modelos Animais , Modelos Neurológicos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Estresse Oxidativo/fisiologia , Fosforilação , Ratos
13.
Biol. Res ; 39(1): 103-104, 2006. ilus
Artigo em Inglês | LILACS | ID: lil-430703

RESUMO

Brain cells have a highly active oxidative metabolism, yet they contain only low to moderate superoxide dismutase and catalase activities. Thus, their antioxidant defenses rely mainly on cellular reduced glutathione levels. In this work, in cortical neurons we characterized viability and changes in reduced and oxidized glutathione levels in response to a protocol of iron accumulation. We found that massive death occurred after 2 days in culture with 10 mM Fe. Surviving cells developed an adaptative response that included increased synthesis of GSH and the maintenance of a glutathione-based reduction potential. These results highlight the fundamental role of glutathione homeostasis in the antioxidant response and provide novel insights into the adaptative mechanisms of neurons subjected to progressive iron loads.


Assuntos
Animais , Ratos , Córtex Cerebral/citologia , Glutationa/metabolismo , Ferro/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Morte Celular/efeitos dos fármacos , Córtex Cerebral/metabolismo , Dissulfeto de Glutationa/metabolismo , Homeostase , Ferro/farmacologia , Neurônios/química , Oxirredução , Fatores de Tempo
14.
Biol. Res ; 39(1): 199-201, 2006. ilus
Artigo em Inglês | LILACS | ID: lil-430715

RESUMO

There is increasing evidence that accumulation of redox-active iron in mitochondria leads to oxidative damage and contributes to various neurodegenerative diseases, such as Friedreich's ataxia and Parkinson's disease. In this work, we examined the existence of regulatory mechanisms for mitochondrial iron uptake and storage. To that end, we used rhodamine B- [(1,10-phenanthrolin-5-yl)amino carbonyl ] benzyl ester, a new fluorescent iron-sensitive probe that is targeted specifically to the mitochondrion. We found that extracellular iron was incorporated readily into mitochondria in an apparently saturable process. Moreover, the rate of iron incorporation responded to the Fe status of the cell, an indication that the mitochondrion actively regulates its iron content.


Assuntos
Humanos , Corantes Fluorescentes , Ferro/metabolismo , Mitocôndrias/metabolismo , Rodaminas , Linhagem Celular , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA