Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 39(20): e103791, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32865299

RESUMO

The link between cholesterol homeostasis and cleavage of the amyloid precursor protein (APP), and how this relationship relates to Alzheimer's disease (AD) pathogenesis, is still unknown. Cellular cholesterol levels are regulated through crosstalk between the plasma membrane (PM), where most cellular cholesterol resides, and the endoplasmic reticulum (ER), where the protein machinery that regulates cholesterol levels resides. The intracellular transport of cholesterol from the PM to the ER is believed to be activated by a lipid-sensing peptide(s) in the ER that can cluster PM-derived cholesterol into transient detergent-resistant membrane domains (DRMs) within the ER, also called the ER regulatory pool of cholesterol. When formed, these cholesterol-rich domains in the ER maintain cellular homeostasis by inducing cholesterol esterification as a mechanism of detoxification while attenuating its de novo synthesis. In this manuscript, we propose that the 99-aa C-terminal fragment of APP (C99), when delivered to the ER for cleavage by γ-secretase, acts as a lipid-sensing peptide that forms regulatory DRMs in the ER, called mitochondria-associated ER membranes (MAM). Our data in cellular AD models indicates that increased levels of uncleaved C99 in the ER, an early phenotype of the disease, upregulates the formation of these transient DRMs by inducing the internalization of extracellular cholesterol and its trafficking from the PM to the ER. These results suggest a novel role for C99 as a mediator of cholesterol disturbances in AD, potentially explaining early hallmarks of the disease.


Assuntos
Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Retículo Endoplasmático/metabolismo , Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Animais , Linhagem Celular , Colesterol/biossíntese , Retículo Endoplasmático/genética , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Células-Tronco Pluripotentes Induzidas , Metabolismo dos Lipídeos , Lipidômica , Camundongos , Mitocôndrias/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Presenilina-2/genética , Presenilina-2/metabolismo , Domínios Proteicos , RNA Interferente Pequeno , Esfingomielina Fosfodiesterase/metabolismo
2.
EMBO J ; 36(22): 3356-3371, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29018038

RESUMO

In the amyloidogenic pathway associated with Alzheimer disease (AD), the amyloid precursor protein (APP) is cleaved by ß-secretase to generate a 99-aa C-terminal fragment (C99) that is then cleaved by γ-secretase to generate the ß-amyloid (Aß) found in senile plaques. In previous reports, we and others have shown that γ-secretase activity is enriched in mitochondria-associated endoplasmic reticulum (ER) membranes (MAM) and that ER-mitochondrial connectivity and MAM function are upregulated in AD We now show that C99, in addition to its localization in endosomes, can also be found in MAM, where it is normally processed rapidly by γ-secretase. In cell models of AD, however, the concentration of unprocessed C99 increases in MAM regions, resulting in elevated sphingolipid turnover and an altered lipid composition of both MAM and mitochondrial membranes. In turn, this change in mitochondrial membrane composition interferes with the proper assembly and activity of mitochondrial respiratory supercomplexes, thereby likely contributing to the bioenergetic defects characteristic of AD.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Retículo Endoplasmático/metabolismo , Membranas Intracelulares/metabolismo , Mitocôndrias/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Linhagem Celular , Respiração Celular , Retículo Endoplasmático/ultraestrutura , Humanos , Membranas Intracelulares/ultraestrutura , Camundongos , Mitocôndrias/ultraestrutura , Mutação/genética , Consumo de Oxigênio , Presenilinas/genética , Transporte Proteico , Esfingolipídeos/metabolismo , Regulação para Cima
3.
Int J Mol Sci ; 22(16)2021 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-34445139

RESUMO

Recent evidence pinpoints extracellular vesicles (EVs) as key players in intercellular communication. Given the importance of cholesterol and sphingomyelin in EV biology, and the relevance of mitochondria-associated endoplasmic reticulum membranes (MAMs) in cholesterol/sphingomyelin homeostasis, we evaluated if MAMs and sphingomyelinases (SMases) could participate in ethanol-induced EV release. EVs were isolated from the extracellular medium of BV2 microglia treated or not with ethanol (50 and 100 mM). Radioactive metabolic tracers combined with thin layer chromatography were used as quantitative methods to assay phospholipid transfer, SMase activity and cholesterol uptake/esterification. Inhibitors of SMase (desipramine and GW4869) and MAM (cyclosporin A) activities were also utilized. Our data show that ethanol increases the secretion and inflammatory molecule concentration of EVs. Ethanol also upregulates MAM activity and alters lipid metabolism by increasing cholesterol uptake, cholesterol esterification and SMase activity in microglia. Notably, the inhibition of either SMase or MAM activity prevented the ethanol-induced increase in EV secretion. Collectively, these results strongly support a lipid-driven mechanism, specifically via SMases and MAM, to explain the effect of ethanol on EV secretion in glial cells.


Assuntos
Retículo Endoplasmático/efeitos dos fármacos , Etanol/farmacologia , Vesículas Extracelulares/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Membranas Mitocondriais/efeitos dos fármacos , Esfingomielina Fosfodiesterase/metabolismo , Compostos de Anilina/farmacologia , Animais , Compostos de Benzilideno/farmacologia , Células Cultivadas , Colesterol/metabolismo , Ciclosporina/farmacologia , Retículo Endoplasmático/metabolismo , Vesículas Extracelulares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Fosfolipídeos/metabolismo
4.
Neurobiol Dis ; 145: 105062, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32866617

RESUMO

The discovery of contact sites was a breakthrough in cell biology. We have learned that an organelle cannot function in isolation, and that many cellular functions depend on communication between two or more organelles. One such contact site results from the close apposition of the endoplasmic reticulum (ER) and mitochondria, known as mitochondria-associated ER membranes (MAMs). These intracellular lipid rafts serve as hubs for the regulation of cellular lipid and calcium homeostasis, and a growing body of evidence indicates that MAM domains modulate cellular function in both health and disease. Indeed, MAM dysfunction has been described as a key event in Alzheimer disease (AD) pathogenesis. Our most recent work shows that, by means of its affinity for cholesterol, APP-C99 accumulates in MAM domains of the ER and induces the uptake of extracellular cholesterol as well as its trafficking from the plasma membrane to the ER. As a result, MAM functionality becomes chronically upregulated while undergoing continual turnover. The goal of this review is to discuss the consequences of C99 elevation in AD, specifically the upregulation of cholesterol trafficking and MAM activity, which abrogate cellular lipid homeostasis and disrupt the lipid composition of cellular membranes. Overall, we present a novel framework for AD pathogenesis that can be linked to the many complex alterations that occur during disease progression, and that may open a door to new therapeutic strategies.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Colesterol/metabolismo , Retículo Endoplasmático/metabolismo , Mitocôndrias/metabolismo , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/patologia , Animais , Transporte Biológico/fisiologia , Retículo Endoplasmático/patologia , Humanos , Mitocôndrias/patologia
5.
Curr Opin Clin Nutr Metab Care ; 23(2): 68-75, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32004239

RESUMO

PURPOSE OF REVIEW: The purpose of this brief review is to gain an understanding on the multiple roles that lipids exert on the brain, and to highlight new ideas in the impact of lipid homeostasis in the regulation of synaptic transmission. RECENT FINDINGS: Recent data underline the crucial function of lipid homeostasis in maintaining neuronal function and synaptic plasticity. Moreover, new advances in analytical approaches to study lipid classes and species is opening a new door to understand and monitor how alterations in lipid pathways could shed new light into the pathogenesis of neurodegeneration. SUMMARY: Lipids are one of the most essential elements of the brain. However, our understanding of the role of lipids within the central nervous system is still largely unknown. Identifying the molecular mechanism (s) by which lipids can regulate neuronal transmission represents the next frontier in neuroscience, and a new challenge in our understanding of the brain and the mechanism(s) behind neurological disorders.


Assuntos
Encéfalo/metabolismo , Metabolismo dos Lipídeos/fisiologia , Lipídeos/fisiologia , Doenças do Sistema Nervoso/metabolismo , Neurônios/metabolismo , Animais , Homeostase , Humanos , Plasticidade Neuronal
6.
J Neuroinflammation ; 16(1): 136, 2019 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-31272469

RESUMO

BACKGROUND: Current evidence indicates that extracellular vesicles (EVs) participate in intercellular signaling, and in the regulation and amplification of neuroinflammation. We have previously shown that ethanol activates glial cells through Toll-like receptor 4 (TLR4) by triggering neuroinflammation. Here, we evaluate if ethanol and the TLR4 response change the release and inflammatory content of astrocyte-derived EVs, and whether these vesicles are capable of communicating with neurons by spreading neuroinflammation. METHODS: Cortical neurons and astrocytes in culture were used. EVs were isolated from the extracellular medium of the primary culture of the WT and TLR4-KO astrocytes treated with or without ethanol (40 mM) for 24 h. Flow cytometry, nanoparticle tracking analysis technology, combined with exosomal molecular markers (tetraspanins) along with electron microscopy, were used to characterize and quantify EVs. The content of EVs in inflammatory proteins, mRNA, and miRNAs was analyzed by Western blot and RT-PCR in both astrocyte-derived EVs and the neurons incubated or not with these EVs. Functional analyses of miRNAs were also performed. RESULTS: We show that ethanol increases the number of secreted nanovesicles and their content by raising the levels of both inflammatory-related proteins (TLR4, NFκB-p65, IL-1R, caspase-1, NLRP3) and by changing miRNAs (mir-146a, mir-182, and mir-200b) in the EVs from the WT-astrocytes compared with those from the untreated WT cells. No changes were observed in either the number of isolated EVs or their content between the untreated and ethanol-treated TLR4-KO astrocytes. We also show that astrocyte-derived EVs could be internalized by naïve cortical neurons to increase the neuronal levels of inflammatory protein (COX-2) and miRNAs (e.g., mir-146a) and to compromise their survival. The functional analysis of miRNAs revealed the regulatory role of the expressed miRNAs in some genes involved in several inflammatory pathways. CONCLUSIONS: These results suggest that astrocyte-derived EVs could act as cellular transmitters of inflammation signaling by spreading and amplifying the neuroinflammatory response induced by ethanol through TLR4 activation.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Etanol/toxicidade , Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Inflamação/induzido quimicamente , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mapas de Interação de Proteínas/efeitos dos fármacos , Mapas de Interação de Proteínas/fisiologia , Receptor 4 Toll-Like/agonistas
7.
J Neurosci Res ; 96(5): 765-780, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29214654

RESUMO

Adolescence is a critical stage of brain maturation in which important plastic and dynamic processes take place in different brain regions, leading to development of the adult brain. Ethanol drinking in adolescence disrupts brain plasticity and causes structural and functional changes in immature brain areas (prefrontal cortex, limbic system) that result in cognitive and behavioral deficits. These changes, along with secretion of sexual and stress-related hormones in adolescence, may impact self-control, decision making, and risk-taking behaviors that contribute to anxiety and initiation of alcohol consumption. New data support the participation of the neuroimmune system in the effects of ethanol on the developing and adult brain. This article reviews the potential pathological bases that underlie the effects of alcohol on the adolescent brain, such as the contribution of genetic background, the perturbation of epigenetic programming, and the influence of the neuroimmune response. Special emphasis is given to the actions of ethanol in the innate immune receptor toll-like receptor 4 (TLR4), since recent studies have demonstrated that by activating the inflammatory TLR4/NFκB signaling response in glial cells, binge drinking of ethanol triggers the release of cytokines/chemokines and free radicals, which exacerbate the immune response that causes neuroinflammation/neural damage as well as short- and long-term neurophysiological, cognitive, and behavioral dysfunction. Finally, potential treatments that target the neuroimmune response to treat the neuropathological and behavioral consequences of adolescent alcohol abuse are discussed.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/imunologia , Consumo Excessivo de Bebidas Alcoólicas/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Adolescente , Encéfalo/crescimento & desenvolvimento , Cognição/efeitos dos fármacos , Humanos , Sistema Imunitário/efeitos dos fármacos
8.
J Neuroinflammation ; 14(1): 145, 2017 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-28738878

RESUMO

BACKGROUND: Inflammation during brain development participates in the pathogenesis of early brain injury and cognitive dysfunctions. Prenatal ethanol exposure affects the developing brain and causes neural impairment, cognitive and behavioral effects, collectively known as fetal alcohol spectrum disorders (FASD). Our previous studies demonstrate that ethanol activates the innate immune response and TLR4 receptor and causes neuroinflammation, brain damage, and cognitive defects in the developmental brain stage of adolescents. We hypothesize that by activating the TLR4 response, maternal alcohol consumption during pregnancy triggers the release of cytokines and chemokines in both the maternal sera and brains of fetuses/offspring, which impairs brain ontogeny and causes cognitive dysfunction. METHODS: WT and TLR4-KO female mice treated with or without 10% ethanol in the drinking water during gestation and lactation were used. Cytokine/chemokine levels were determined by ELISA in the amniotic fluid, maternal serum, and cerebral cortex, as well as in the offspring cerebral cortex. Microglial and neuronal markers (evaluated by western blotting), myelin proteins (immunohistochemical and western blotting) and synaptic parameters (western blotting and electron microscopy) were assessed in the cortices of the WT and TLR4-KO pups on PND 0, 20, and 66. Behavioral tests (elevated plus maze and passive avoidance) were performed in the WT and TLR4-KO mice on PND 66 exposed or not to ethanol. RESULTS: We show that alcohol intake during gestation and lactation increases the levels of several cytokines/chemokines (IL-1ß, IL-17, MIP-1α, and fractalkine) in the maternal sera, amniotic fluid, and brains of fetuses and offspring. The upregulation of cytokines/chemokines is associated with an increase in activated microglia markers (CD11b and MHC-II), and with a reduction in some synaptic (synaptotagmin, synapsin IIa) and myelin (MBP, PLP) proteins in the brains of offspring on days 0, 20, and 66 (long-term effects). These changes are associated with long-term behavioral impairments, in the 66-day-old alcohol-exposed pups. TLR4-deficient mice are protected against ethanol-induced cytokine/chemokine production in alcohol-treated dams and offspring, along with synaptic and myelin alterations, and the log-term behavioral dysfunction induced by ethanol in offspring. CONCLUSIONS: These results suggest that the immune system activation, through the TLR4 response, might play an important role in the neurodevelopmental defects in FASD.


Assuntos
Depressores do Sistema Nervoso Central/toxicidade , Deficiências do Desenvolvimento/etiologia , Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Receptor 4 Toll-Like/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Aprendizagem da Esquiva , Peso Corporal/fisiologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/ultraestrutura , Citocinas/metabolismo , Deficiências do Desenvolvimento/genética , Modelos Animais de Doenças , Feminino , Transtornos do Espectro Alcoólico Fetal/etiologia , Transtornos do Espectro Alcoólico Fetal/patologia , Masculino , Comportamento Materno/fisiologia , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Proteínas da Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Receptor 4 Toll-Like/genética
9.
Alcohol Clin Exp Res ; 41(7): 1257-1270, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28493563

RESUMO

BACKGROUND: We previously showed that, by activating innate immune receptors Toll-like 4 (TLR4), adolescent intermittent ethanol (EtOH) exposure causes neuroinflammation, myelin damage, and behavioral dysfunctions. Recent findings reveal that clinically used opioid antagonists naltrexone (NT) and naloxone (NX) inhibit opioid-induced TLR4 signaling and that NT, NX, and nalmefene (NF), the 6-methylene derivative of NX, are able to reduce alcohol drinking escalation. METHODS: NF (0.1 mg/kg, intraperitoneally) was injected 1 hour prior to EtOH (3 g/kg, intraperitoneally) following intermittent treatment in female (PND35) adolescent mice. Inflammatory molecules, myelin proteins, and apoptotic markers were assessed in the prefrontal cortex (PFC) and striatum/nucleus accumbens (STR/NAcc). The effect of NF on alcohol drinking preference was evaluated in both the wild-type and TLR4 knockout (KO) adolescent mice. Using astroglial cells, the inhibitory potential of NT, NX, and NF on lipopolysaccharide (LPS), or the EtOH-triggered TLR4 response, was compared. RESULTS: Our findings indicate that NF prevents the up-regulation of cytokines (IL-1ß, IL-17A, TNF-α), chemokines (MCP-1, MIP-1, KC), and pro-inflammatory mediators (iNOS, COX-2), along with myelin damage and apoptotic events, in both PFC and STR/NAcc. NF also abolishes EtOH-induced escalation of alcohol preference/consumption, but has no effect when administered to TLR4-KO mice. In vitro experiments indicate that NX and NF inhibit TLR4 activation upon LPS or EtOH stimulation. Immunofluorescence studies and lipid rafts isolation show that NF is able to prevent TLR4 translocation to lipid rafts/caveolae in astrocytes. CONCLUSIONS: These results suggest that NF prevents neuroinflammation and brain damage by blocking the TLR4 response and also support the role of central pro-inflammatory immune signaling in the modulation of alcohol consumption/addiction.


Assuntos
Consumo de Bebidas Alcoólicas/prevenção & controle , Transtornos Relacionados ao Uso de Álcool/prevenção & controle , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/uso terapêutico , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Morte Celular/efeitos dos fármacos , Quimiocinas/metabolismo , Avaliação Pré-Clínica de Medicamentos , Etanol/efeitos adversos , Feminino , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Bainha de Mielina/efeitos dos fármacos , Naloxona/farmacologia , Naloxona/uso terapêutico , Naltrexona/farmacologia , Naltrexona/uso terapêutico , Antagonistas de Entorpecentes/farmacologia , Cultura Primária de Células , Receptor 4 Toll-Like/metabolismo
10.
Addict Biol ; 22(6): 1829-1841, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27699959

RESUMO

Heavy binge drinking in adolescence can cause long-term cognitive and behavioral dysfunctions. Recent experimental evidence indicates the participation of immune system activation in the effects of ethanol in the adolescent brain and suggests gender differences. The present study aims to assess plasma cytokine and chemokine levels in male and female adolescents and young adults during acute alcohol intoxication and to correlate these results with the toll-like receptor 4 (TLR4) response. The potential role of the TLR4 signaling response was also assessed in plasma and prefrontal cortex (PFC) of adolescent wild-type and TLR4-knockout male and female mice with binge ethanol treatment. The results showed that alcohol intoxication increased the plasma levels of several cytokine and chemokine [interferon-γ, interleukin (IL)-10, IL-17A, IL-1ß, IL-2, IL-4, IL-6, IL-8, fractalkine, monocyte chemoattractant protein 1 (MCP-1) and macrophage inflammatory protein 1α (MIP-1α)] and the upregulation of TLR4 mRNA levels occurred in intoxicated females, while elevation of colony-stimulating factor was only observed in the plasma of males. In wild-type female adolescent mice, intermittent ethanol treatment increased the levels of several cytokines (IL-17A and IL-1ß) and chemokines (MCP-1, MIP-1α and fractalkine) in PFC and in serum (IL-17A, MCP-1 and MIP-1α), but significant differences in the fractalkine levels in PFC were observed only in male mice. No changes in serum or prefrontal cortex cytokine and chemokine levels were noted in ethanol-treated male or female TLR4-knockout mice. Our findings revealed that females are more vulnerable than males to inflammatory effects of binge ethanol drinking and suggested that TLR4 is an important target of ethanol-induced inflammation and neuroinflammation in adolescence.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/sangue , Depressores do Sistema Nervoso Central/farmacologia , Citocinas/sangue , Etanol/farmacologia , Consumo de Álcool por Menores , Adolescente , Adulto , Animais , Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Depressores do Sistema Nervoso Central/sangue , Quimiocinas/sangue , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Etanol/sangue , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Fatores Sexuais , Espanha , Adulto Jovem
11.
Brain Behav Immun ; 53: 159-171, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26686767

RESUMO

Studies in humans and experimental animals have demonstrated the vulnerability of the adolescent brain to actions of ethanol and the long-term consequences of binge drinking, including the behavioral and cognitive deficits that result from alcohol neurotoxicity, and increased risk to alcohol abuse and dependence. Although the mechanisms that participate in these effects are largely unknown, we have shown that ethanol by activating innate immune receptors, toll-like receptor 4 (TLR4), induces neuroinflammation, impairs myelin proteins and causes cognitive dysfunctions in adolescent mice. Since neuroimmune signaling is also involved in alcohol abuse, the aim of this study was to assess whether ethanol treatment in adolescence promotes the long-term synaptic and molecular events associated with alcohol abuse and addiction. Using wild-type (WT) and TLR4-deficient (TLR4-KO) adolescent mice treated intermittently with ethanol (3g/kg) for 2 weeks, we showed that binge-like ethanol treatment in adolescent mice promotes short- and long-term alterations in synaptic plasticity and epigenetic changes in the promoter region of bdnf and fosb, which increased their expression in the mPFC of young adult animals. These molecular events were associated with long-term rewarding and anxiogenic-related behavioral effects, along with increased alcohol preference. Our results further showed the participation of neuroimmune system activation and the TLR4 signaling response since deficient mice in TLR4 (TLR4-KO) are protected against molecular and behavioral alterations of ethanol in the adolescent brain. Our results highlight a new role of the neuroimmune function and open up new avenues to develop pharmacological treatments that can normalize the immune signaling responsible for long-term effects in adolescence, including alcohol abuse and related disorders.


Assuntos
Alcoolismo/genética , Consumo Excessivo de Bebidas Alcoólicas/genética , Etanol/toxicidade , Receptor 4 Toll-Like/genética , Fatores Etários , Alcoolismo/metabolismo , Animais , Ansiedade/induzido quimicamente , Ansiedade/genética , Ansiedade/metabolismo , Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/genética , Transtornos Cognitivos/metabolismo , Epigênese Genética , Etanol/administração & dosagem , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas da Mielina/metabolismo , Recompensa , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/metabolismo
12.
Alcohol Clin Exp Res ; 40(11): 2260-2270, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27650785

RESUMO

The innate immune response in the central nervous system (CNS) participates in both synaptic plasticity and neural damage. Emerging evidence from human and animal studies supports the role of the neuroimmune system response in many actions of ethanol (EtOH) on the CNS. Research studies have shown that alcohol stimulates brain immune cells, microglia, and astrocytes, by activating innate immune receptors Toll-like receptors (TLRs) and NOD-like receptors (inflammasome NLRs) triggering signaling pathways, which culminate in the production of pro-inflammatory cytokines and chemokines that lead to neuroinflammation. This review focuses on evidence that indicates the participation of TLRs and the inflammasome NLRs signaling response in many effects of EtOH on the CNS, such as neuroinflammation associated with brain damage, cognitive and behavioral dysfunction, and adolescent brain development alterations. It also reviews findings that indicate the role of TLR4-dependent signaling immune molecules in alcohol consumption, reward, and addiction. The research data suggest that overactivation of TLR4 or NLRs increases pro-inflammatory cytokines and mediators to cause neural damage in the cerebral cortex and hippocampus, while modest TLR4 activation, along with the generation of certain cytokines and chemokines in specific brain areas (e.g., amygdala, ventral tegmental area), modulate neurotransmission, alcohol drinking, and alcohol rewards. Elimination of TLR4 and NLRP3 abolishes many neuroimmune effects of EtOH. Despite much progress being made in this area, there are some research gaps and unanswered questions that this review discusses. Finally, potential therapies that target neuroimmune pathways to treat neuropathological and behavioral consequences of alcohol abuse are also evaluated.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Sistema Nervoso Central/efeitos dos fármacos , Etanol/farmacologia , Imunidade Inata/efeitos dos fármacos , Neuroimunomodulação/efeitos dos fármacos , Adolescente , Desenvolvimento do Adolescente , Consumo de Bebidas Alcoólicas/imunologia , Consumo de Bebidas Alcoólicas/metabolismo , Alcoolismo/metabolismo , Animais , Biomarcadores/metabolismo , Sistema Nervoso Central/metabolismo , Depressores do Sistema Nervoso Central/imunologia , Etanol/imunologia , Humanos , Proteínas NLR/metabolismo , Receptores Toll-Like/metabolismo
13.
Brain Behav Immun ; 45: 233-44, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25486089

RESUMO

The adolescent brain undergoes important dynamic and plastic cell changes, including overproduction of axons and synapses, followed by rapid pruning along with ongoing axon myelination. These developmental changes make the adolescent brain particularly vulnerable to neurotoxic and behavioral effects of alcohol. Although the mechanisms of these effects are largely unknown, we demonstrated that ethanol by activating innate immune receptors toll-like receptor 4 (TLR4), induces neuroinflammation and brain damage in adult mice. The present study aims to evaluate whether intermittent ethanol treatment in adolescence promotes TLR4-dependent pro-inflammatory processes, leading to myelin and synaptic dysfunctions, and long-term cognitive impairments. Using wild-type (WT) and TLR4-deficient (TLR4-KO) adolescent mice treated intermittently with ethanol (3.0g/kg) for 2weeks, we show that binge-like ethanol treatment activates TLR4 signaling pathways (MAPK, NFκB) leading to the up-regulation of cytokines and pro-inflammatory mediators (COX-2, iNOS, HMGB1), impairing synaptic and myelin protein levels and causing ultrastructural alterations. These changes were associated with long-lasting cognitive dysfunctions in young adult mice, as demonstrated with the object recognition, passive avoidance and olfactory behavior tests. Notably, elimination of TLR4 receptors prevented neuroinflammation along with synaptic and myelin derangements, as well as long-term cognitive alterations. These results support the role of the neuroimmune response and TLR4 signaling in the neurotoxic and behavioral effects of ethanol in adolescence.


Assuntos
Transtornos Relacionados ao Uso de Álcool/genética , Depressores do Sistema Nervoso Central/farmacologia , Transtornos Cognitivos/genética , Cognição/efeitos dos fármacos , Etanol/farmacologia , Bainha de Mielina/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Animais , Depressores do Sistema Nervoso Central/efeitos adversos , Transtornos Cognitivos/induzido quimicamente , Ciclo-Oxigenase 2/efeitos dos fármacos , Ciclo-Oxigenase 2/imunologia , Etanol/efeitos adversos , Proteína HMGB1/efeitos dos fármacos , Proteína HMGB1/imunologia , Camundongos , Camundongos Knockout , Quinases de Proteína Quinase Ativadas por Mitógeno/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/imunologia , Proteínas da Mielina/efeitos dos fármacos , Proteínas da Mielina/metabolismo , Bainha de Mielina/genética , Bainha de Mielina/ultraestrutura , NF-kappa B/efeitos dos fármacos , NF-kappa B/imunologia , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/imunologia , Transdução de Sinais/efeitos dos fármacos , Sinapses/genética , Sinapses/ultraestrutura
14.
Addict Biol ; 20(4): 756-72, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24854157

RESUMO

The treatment for cocaine use constitutes a clinical challenge because of the lack of appropriate therapies and the high rate of relapse. Recent evidence indicates that the immune system might be involved in the pathogenesis of cocaine addiction and its co-morbid psychiatric disorders. This work examined the plasma pro-inflammatory cytokine and chemokine profile in abstinent cocaine users (n = 82) who sought outpatient cocaine treatment and age/sex/body mass-matched controls (n = 65). Participants were assessed with the diagnostic interview Psychiatric Research Interview for Substance and Mental Diseases according to the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision (DSM-IV-TR). Tumor necrosis factor-alpha, chemokine (C-C motif) ligand 2/monocyte chemotactic protein-1 and chemokine (C-X-C motif) ligand 12 (CXCL12)/stromal cell-derived factor-1 (SDF-1) were decreased in cocaine users, although all cytokines were identified as predictors of a lifetime pathological use of cocaine. Interleukin-1 beta (IL-1ß), chemokine (C-X3-C motif) ligand 1 (CX3CL1)/fractalkine and CXCL12/SDF-1 positively correlated with the cocaine symptom severity when using the DSM-IV-TR criteria for cocaine abuse/dependence. These cytokines allowed the categorization of the outpatients into subgroups according to severity, identifying a subgroup of severe cocaine users (9-11 criteria) with increased prevalence of co-morbid psychiatric disorders [mood (54%), anxiety (32%), psychotic (30%) and personality (60%) disorders]. IL-1ß was observed to be increased in users with such psychiatric disorders relative to those users with no diagnosis. In addition to these clinical data, studies in mice demonstrated that plasma IL-1ß, CX3CL1 and CXCL12 were also affected after acute and chronic cocaine administration, providing a preclinical model for further research. In conclusion, cocaine exposure modifies the circulating levels of pro-inflammatory mediators. Plasma cytokine/chemokine monitoring could improve the stratification of cocaine consumers seeking treatment and thus facilitate the application of appropriate interventions, including management of heightened risk of psychiatric co-morbidity. Further research is necessary to elucidate the role of the immune system in the etiology of cocaine addiction.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/sangue , Citocinas/metabolismo , Adolescente , Adulto , Idoso , Assistência Ambulatorial , Animais , Estudos de Casos e Controles , Quimiocina CX3CL1/metabolismo , Quimiocina CXCL12/metabolismo , Quimiocinas/metabolismo , Transtornos Relacionados ao Uso de Cocaína/complicações , Transtornos Relacionados ao Uso de Cocaína/terapia , Estudos Transversais , Diagnóstico Duplo (Psiquiatria) , Feminino , Humanos , Interleucina-1beta/metabolismo , Masculino , Transtornos Mentais/sangue , Transtornos Mentais/complicações , Camundongos , Pessoa de Meia-Idade , Adulto Jovem
15.
J Neurochem ; 129(3): 448-62, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24345077

RESUMO

Toll-like receptor 4 (TLR4) activation and signalling in glial cells play critical roles in neurological disorders and in alcohol-induced brain damage. TLR4 endocytosis upon lipopolysaccharide (LPS) stimulation regulates which signalling pathway is activated, the MyD88-dependent or the TIR-domain-containing adapter-inducing interferon-ß (TRIF)-dependent pathway. However, it remains elusive whether ethanol-induced TLR4 signalling is associated with receptor internalization and trafficking, and which endocytic pathway(s) are used in cortical astrocytes. Using the adenoviral over-expression of TLR4(GFP) , confocal microscopy and the imagestream technique, we show that upon ethanol or LPS stimulation, TLR4 co-localizes with markers of the clathrin and caveolin endocytic pathways, and that this endocytosis is dependent on dynamin. Using chlorpromazin and filipin as inhibitors of the clathrin and rafts/caveolae endocytic pathways, respectively, we demostrate that TRIF-dependent signalling relies on an intact clathrin pathway, whereas disruption of rafts/caveolae inhibits the MyD88- and TRIF-dependent signalling pathways. Immunofluorescence studies also suggest that lipid rafts and clathrin cooperate for appropriate TLR4 internalization. We also show that ethanol can trigger similar endocytic pathways as LPS does, although ethanol delays clathrin internalization and alters TLR4 vesicular trafficking. Our results provide new insights into the effects of ethanol or LPS on TLR4 signalling in cortical astrocytes, events that may underlie neuroinflammation and brain damage. The results demonstrate that ethanol or lipopolysaccharide (LPS) triggers toll-like receptor 4 (TLR4) endocytosis by caveolae and clathrin-dependent pathways in astrocytes. We proposed that while clathrin is the protein responsible for TLR4 internalization, caveolin-1/lipid rafts membrane microdomains are required for TLR4 signaling. The results provide new insights into the effects of ethanol on TLR4 signalling in astrocytes, events that may underlie neuroinflammation.


Assuntos
Astrócitos/metabolismo , Endocitose/efeitos dos fármacos , Etanol/toxicidade , Lipopolissacarídeos/toxicidade , Receptor 4 Toll-Like/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Western Blotting , Cavéolas/efeitos dos fármacos , Cavéolas/metabolismo , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Vesículas Revestidas por Clatrina/efeitos dos fármacos , Vesículas Revestidas por Clatrina/metabolismo , Endocitose/fisiologia , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Imunoprecipitação , Microscopia Confocal , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
16.
J Neurochem ; 126(2): 261-73, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23600947

RESUMO

Alcohol consumption can induce brain damage, demyelination, and neuronal death, although the mechanisms are poorly understood. Toll-like receptors are sensors of the innate immune system and their activation induces inflammatory processes. We have reported that ethanol activates and recruits Toll-like receptor (TLR)4 receptors within the lipid rafts of glial cells, triggering the production of inflammatory mediators and causing neuroinflammation. Since TLR2 can also participate in the glial response and in the neuroinflammation, we investigate the effects of ethanol on TLR4/TLR2 responses. Here, we demonstrate that ethanol up-regulates TLR4 and TLR2 expression in microglial cells, inducing the production of inflammatory mediators which triggers reactive oxygen species generation and neuronal apoptosis. Ethanol also promotes TLR4/TLR2 recruitment into lipid rafts-caveolae, mimicking their activation by their ligands, lipopolysaccharide, and lipoteichoic acid (LTA). Immunoprecipitation and confocal microscopy studies reveal that ethanol induces a physical association between TLR2 and TLR4 receptors, suggesting the formation of heterodimers. Using microglia from either TLR2 or TLR4 knockout mice, we show that TLR2 potentiates the effects of ethanol on the TLR4 response reflected by the activation of MAPKs and inducible NO synthase. In summary, we provide evidence for a mechanism by which ethanol triggers TLR4/TLR2 association contributing to the neuroinflammation and neurodegeneration associated with alcohol abuse.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Neuroglia/efeitos dos fármacos , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Cavéolas/efeitos dos fármacos , Cavéolas/metabolismo , Células Cultivadas , Córtex Cerebral , Citocinas/metabolismo , Transferência de Energia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Inflamação , Lipopolissacarídeos/farmacologia , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neuroglia/citologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Óxido Nítrico Sintase/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Receptor 2 Toll-Like/deficiência , Receptor 4 Toll-Like/deficiência
17.
EBioMedicine ; 92: 104628, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37245481

RESUMO

BACKGROUND: The most common form of neuronal ceroid lipofuscinosis (NCL) is juvenile CLN3 disease (JNCL), a currently incurable neurodegenerative disorder caused by mutations in the CLN3 gene. Based on our previous work and on the premise that CLN3 affects the trafficking of the cation-independent mannose-6 phosphate receptor and its ligand NPC2, we hypothesised that dysfunction of CLN3 leads to the aberrant accumulation of cholesterol in the late endosomes/lysosomes (LE/Lys) of JNCL patients' brains. METHODS: An immunopurification strategy was used to isolate intact LE/Lys from frozen autopsy brain samples. LE/Lys isolated from samples of JNCL patients were compared with age-matched unaffected controls and Niemann-Pick Type C (NPC) disease patients. Indeed, mutations in NPC1 or NPC2 result in the accumulation of cholesterol in LE/Lys of NPC disease samples, thus providing a positive control. The lipid and protein content of LE/Lys was then analysed using lipidomics and proteomics, respectively. FINDINGS: Lipid and protein profiles of LE/Lys isolated from JNCL patients were profoundly altered compared to controls. Importantly, cholesterol accumulated in LE/Lys of JNCL samples to a comparable extent than in NPC samples. Lipid profiles of LE/Lys were similar in JNCL and NPC patients, except for levels of bis(monoacylglycero)phosphate (BMP). Protein profiles detected in LE/Lys of JNCL and NPC patients appeared identical, except for levels of NPC1. INTERPRETATION: Our results support that JNCL is a lysosomal cholesterol storage disorder. Our findings also support that JNCL and NPC disease share pathogenic pathways leading to aberrant lysosomal accumulation of lipids and proteins, and thus suggest that the treatments available for NPC disease may be beneficial to JNCL patients. This work opens new avenues for further mechanistic studies in model systems of JNCL and possible therapeutic interventions for this disorder. FUNDING: San Francisco Foundation.


Assuntos
Doenças por Armazenamento dos Lisossomos , Doença de Niemann-Pick Tipo C , Humanos , Doença de Niemann-Pick Tipo C/genética , Doença de Niemann-Pick Tipo C/metabolismo , Doença de Niemann-Pick Tipo C/patologia , Colesterol/metabolismo , Doenças por Armazenamento dos Lisossomos/metabolismo , Proteínas/metabolismo , Lisossomos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/genética
18.
Methods Cell Biol ; 155: 33-44, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32183966

RESUMO

Organelles within cells are interconnected by physical associations or contact sites. In the last decade, many reports have shown that these interactions are functional domains that maintain cellular homeostasis. One of the best studied interactions is between endoplasmic reticulum (ER) and mitochondria via mitochondria-associated membranes or MAMs. MAMs are lipid rafts in the ER in close apposition to mitochondria, where multiple enzymatic activities converge to coordinately regulate cellular functions such as: the import of phosphatidylserine into mitochondria from the ER for decarboxylation to phosphatidylethanolamine, cholesterol esterification, calcium signaling, mitochondrial shape and motility, autophagy and apoptosis. In this chapter, we describe and discuss some of the methods to isolate and assay this interesting cellular region.


Assuntos
Técnicas Citológicas/métodos , Retículo Endoplasmático/metabolismo , Membranas Intracelulares/metabolismo , Mitocôndrias/metabolismo , Animais , Centrifugação com Gradiente de Concentração , Humanos , Microdomínios da Membrana/metabolismo
19.
Methods Cell Biol ; 155: 321-335, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32183965

RESUMO

Mitochondria and their associated membranes actively participate in biosynthesis, trafficking, and degradation of cellular phospholipids. Two crucial lipid biosynthetic activities of mitochondria include (i) the decarboxylation of phosphatidylserine to phosphatidylethanolamine and (ii) the de novo synthesis of cardiolipin. Here we describe protocols to measure these two activities, applying isotope-labeled or exogenous substrates in combination with thin-layer chromatography or mass spectrometry.


Assuntos
Mitocôndrias/metabolismo , Fosfolipídeos/biossíntese , Animais , Cardiolipinas/biossíntese , Células Cultivadas , Drosophila melanogaster/metabolismo , Fosfatidiletanolaminas/biossíntese , Fosfatidilserinas/biossíntese
20.
Int Rev Neurobiol ; 154: 235-278, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32739006

RESUMO

Inter-organelle communication is a rapidly-expanding field that has transformed our understanding of cell biology and pathology. Organelle-organelle contact sites can generate transient functional domains that act as enzymatic hubs involved in the regulation of cellular metabolism and intracellular signaling. One of these hubs is located in areas of the endoplasmic reticulum (ER) connected to mitochondria, called mitochondria-associated ER membranes (MAM). These MAM are transient lipid rafts intimately involved in cholesterol and phospholipid metabolism, calcium homeostasis, and mitochondrial function and dynamics. In addition, γ-secretase-mediated proteolysis of the amyloid precursor protein 99-aa C-terminal fragment (C99) to form amyloid ß also occurs at the MAM. Our most recent data indicates that in Alzheimer's disease, increases in uncleaved C99 levels at the MAM provoke the upregulation of MAM-resident functions, resulting in the loss of lipid homeostasis, and mitochondrial dysfunction. Here, we discuss the relevance of these findings in the field, and the contribution of C99 and MAM dysfunction to Alzheimer's disease neuropathology.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Colesterol/metabolismo , Retículo Endoplasmático/metabolismo , Mitocôndrias/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA