Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Am J Drug Alcohol Abuse ; 49(3): 345-358, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-36345683

RESUMO

Background: Hippocampal and cerebellar neuropathology occurs in individuals with alcohol use disorders (AUD), resulting in impaired cognitive and motor function.Objectives: Evaluate the effects of ethanol on the expression of pro- and anti-inflammatory molecules, as well as the effects of the anti-inflammatory PPAR-γ agonist pioglitazone in suppressing ethanol-induced neuroinflammation.Methods: Adult male and female mice were treated chronically with ethanol for just under a month followed by a single acute binge dose of ethanol. Animals were provided liquid diet in the absence of ethanol (Control; n = 18, 9 M/9F), liquid diet containing ethanol (ethanol; n = 22, 11 M/11F), or liquid diet containing ethanol plus gavage administration of 30.0 mg/kg pioglitazone (ethanol + pioglitazone; n = 20, 10 M/10F). The hippocampus and cerebellum were isolated 24 h following the binge dose of ethanol, mRNA was isolated, and pro- and anti-inflammatory molecules were quantified by qRT-PCR.Results: Ethanol significantly (p < .05) increased the expression of pro-inflammatory molecules IL-1ß, TNF-α, CCL2, and COX2; increased the expression of inflammasome-related molecules NLRP3 and Casp1 but decreased IL-18; and altered the expression of anti-inflammatory molecules including TGFßR1 in the hippocampus and cerebellum, though some differences were observed between males and females and the two brain regions. The anti-inflammatory pioglitazone inhibited ethanol-induced alterations in the expression of most, but not all, inflammation-related molecules.Conclusion: Chronic plus binge administration of ethanol induced the expression of inflammatory molecules in adult mice and pioglitazone suppressed ethanol-induced neuroinflammation.


Assuntos
Alcoolismo , Etanol , Camundongos , Feminino , Masculino , Animais , Etanol/farmacologia , Pioglitazona/metabolismo , Pioglitazona/farmacologia , Doenças Neuroinflamatórias , Hipocampo , Cerebelo/metabolismo
2.
J Neurosci Res ; 99(8): 1973-1985, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-32959429

RESUMO

Ethanol exposure to the fetus during pregnancy can result in fetal alcohol spectrum disorders (FASD). These disorders vary in severity, can affect multiple organ systems, and can lead to lifelong disabilities. Damage to the central nervous system (CNS) is common in FASD, and can result in altered behavior and cognition. The incidence of FASD is alarmingly high, resulting in significant personal and societal costs. There are no cures for FASD. Alcohol can directly alter the function of neurons in the developing CNS. In addition, ethanol can alter the function of CNS glial cells including microglia and astrocytes which normally maintain homeostasis in the CNS. These glial cells can function as resident immune cells in the CNS to protect against pathogens and other insults. However, activation of glia can also damage CNS cells and lead to aberrant CNS function. Ethanol exposure to the developing brain can result in the activation of glia and neuroinflammation, which may contribute to the pathology associated with FASD. This suggests that anti-inflammatory agents may be effective in the treatment of FASD.


Assuntos
Astrócitos/metabolismo , Sistema Nervoso Central/metabolismo , Etanol/farmacologia , Transtornos do Espectro Alcoólico Fetal/metabolismo , Microglia/metabolismo , Doenças Neuroinflamatórias/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Astrócitos/imunologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/fisiopatologia , Feminino , Transtornos do Espectro Alcoólico Fetal/imunologia , Transtornos do Espectro Alcoólico Fetal/fisiopatologia , Humanos , Microglia/imunologia , Doenças Neuroinflamatórias/imunologia , Doenças Neuroinflamatórias/fisiopatologia , Gravidez
3.
J Neurosci Res ; 99(8): 1986-2007, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33533128

RESUMO

Fetal alcohol spectrum disorders (FASD) are alarmingly common, result in significant personal and societal loss, and there is no effective treatment for these disorders. Cerebellar neuropathology is common in FASD and causes aberrant cognitive and motor function. Ethanol-induced neuroinflammation is believed to contribute to neuropathological sequelae of FASD, and was previously demonstrated in the cerebellum in animal models of FASD. We now demonstrate neuroinflammation persists in the cerebellum several days following cessation of ethanol treatment in an early postnatal mouse model, with meaningful implications for timing of therapeutic intervention in FASD. We also demonstrate by Sholl analysis that ethanol decreases ramification of microglia cell processes in cells located near the Purkinje cell layer but not those near the external granule cell layer. Ethanol did not alter the expression of anti-inflammatory molecules or molecules that constitute NLRP1 and NLRP3 inflammasomes. Interestingly, ethanol decreased the expression of IL-23a (P19) and IL-12Rß1 suggesting that ethanol may suppress IL-12 and IL-23 signaling. Fractalkine-fractalkine receptor (CX3CL1-CX3CR1) signaling is believed to suppress microglial activation and our demonstration that ethanol decreases CX3CL1 expression suggests that ethanol modulation of CX3CL1-CX3CR1 signaling may contribute to cerebellar neuroinflammation and neuropathology. We demonstrate ethanol alters the expression of specific molecules in the cerebellum understudied in FASD, but crucial for immune responses. Ethanol increases the expression of NOX-2 and NGP and decreases the expression of RAG1, NOS1, CD59a, S1PR5, PTPN22, GPR37, and Serpinb1b. These molecules represent a new horizon as potential targets for development of FASD therapy.


Assuntos
Cerebelo/metabolismo , Transtornos do Espectro Alcoólico Fetal/metabolismo , Microglia/metabolismo , Doenças Neuroinflamatórias/metabolismo , Animais , Cerebelo/patologia , Quimiocina CX3CL1/metabolismo , Citocinas/metabolismo , Feminino , Expressão Gênica , Inflamassomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , Gravidez
4.
Alcohol Clin Exp Res ; 45(7): 1408-1423, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34060105

RESUMO

BACKGROUND: The developing hippocampus and cerebellum, unique among brain regions, exhibit a secondary surge in neurogenesis during the third trimester of pregnancy. Ethanol (EtOH) exposure during this period is results in a loss of tissue volume and associated neurobehavioral deficits. However, mechanisms that link EtOH exposure to teratology in these regions are not well understood. We therefore analyzed transcriptomic adaptations to EtOH exposure to identify mechanistic linkages. METHODS: Hippocampi and cerebella were microdissected at postnatal day (P)10, from control C57BL/6J mouse pups, and pups treated with 4 g/kg of EtOH from P4 to P9. RNA was isolated and RNA-seq analysis was performed. We compared gene expression in EtOH- and vehicle-treated control neonates and performed biological pathway-overrepresentation analysis. RESULTS: While EtOH exposure resulted in the general induction of genes associated with the S-phase of mitosis in both cerebellum and hippocampus, overall there was little overlap in differentially regulated genes and associated biological pathways between these regions. In cerebellum, EtOH additionally induced gene expression associated with the G2/M-phases of the cell cycle and sonic hedgehog signaling, while in hippocampus, EtOH-induced the pathways for ribosome biogenesis and protein translation. Moreover, EtOH inhibited the transcriptomic identities associated with inhibitory interneuron subpopulations in the hippocampus, while in the cerebellum there was a more pronounced inhibition of transcripts across multiple oligodendrocyte maturation stages. CONCLUSIONS: These data indicate that during the delayed neurogenic period, EtOH may stimulate the cell cycle, but it otherwise results in widely divergent molecular effects in the hippocampus and cerebellum. Moreover, these data provide evidence for region- and cell-type-specific vulnerability, which may contribute to the pathogenic effects of developmental EtOH exposure.


Assuntos
Animais Recém-Nascidos/crescimento & desenvolvimento , Cerebelo/crescimento & desenvolvimento , Etanol/efeitos adversos , Hipocampo/crescimento & desenvolvimento , Neurogênese/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Animais , Apoptose/genética , Ciclo Celular/genética , Cerebelo/metabolismo , Etanol/administração & dosagem , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hipocampo/metabolismo , Interneurônios/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , RNA Mensageiro/análise
5.
Alcohol Clin Exp Res ; 44(2): 435-444, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31872887

RESUMO

BACKGROUND: Prenatal ethanol exposure (PE) impairs midbrain dopaminergic (DA) neuron function, which might contribute to various cognitive and behavioral deficits, including attention deficits and increased addiction risk, often observed in individuals with fetal alcohol spectrum disorders. Currently, the underlying mechanisms for PE-induced deficits are unclear. PE could lead to neuroinflammation by activating microglia, which play an important role in synaptic function. In the present study, we investigated PE effects on microglial activation and DA neuron density and morphology in the ventral tegmental area (VTA). Since postnatal environmental enrichment can reduce neuroinflammation and ameliorate several PE-induced behavioral deficits, we examined if a postnatal environmental intervention strategy using neonatal handling and postweaning complex housing could reverse PE effects on VTA DA neurons and microglia. METHODS: Pregnant rats received 0 or 6 g/kg/d ethanol by 2 intragastric intubations on gestation days 8 to 20. After birth, rats were reared in the standard laboratory or enriched condition. Male adult rats (8 to 12 weeks old) were used for immunocytochemistry. RESULTS: The results showed that PE decreased VTA DA neuron body size in standardly housed rats. Moreover, there was a significant decrease in numbers of VTA microglial branches and junctions in PE rats, suggesting morphological activation of microglia and possible neuroinflammation. The PE effects on microglia were normalized by postnatal environmental intervention, which also decreased the numbers of microglial branches and junctions in control animals, possibly via reduced stress. CONCLUSIONS: Our findings show an association between PE-induced morphological activation of microglia and impaired DA neuron morphology in the VTA. Importantly, postnatal environmental intervention rescues possible PE-induced microglial activation. These data support that environmental intervention can be effective in ameliorating cognitive and behavioral deficits associated with VTA DA neuron dysfunctions, such as attention deficits and increased addiction risk.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Meio Ambiente , Etanol/toxicidade , Microglia/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/terapia , Área Tegmentar Ventral/efeitos dos fármacos , Fatores Etários , Animais , Animais Recém-Nascidos , Neurônios Dopaminérgicos/patologia , Etanol/administração & dosagem , Feminino , Abrigo para Animais , Masculino , Microglia/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/patologia , Ratos , Ratos Sprague-Dawley , Área Tegmentar Ventral/patologia
6.
Brain Behav Immun ; 64: 320-329, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28167117

RESUMO

Binge alcohol drinking has emerged as a typical phenomenon in young people. This pattern of drinking, repeatedly leading to extremely high blood and brain alcohol levels and intoxication is associated with severe risks of neurodegeneration and cognitive damage. Mechanisms involved in excitotoxicity and neuroinflammation are pivotal elements in alcohol-induced neurotoxicity. Evidence has demonstrated that PPARγ receptor activation shows anti-inflammatory and neuroprotective properties. Here we examine whether treatment with the PPARγ agonist pioglitazone is beneficial in counteracting neurodegeneration, neuroinflammation and cognitive damage produced by binge alcohol intoxication. Adult Wistar rats were subjected to a 4-day binge intoxication procedure, which is commonly used to model excessive alcohol consumption in humans. Across the 4-day period, pioglitazone (0, 30, 60mg/kg) was administered orally twice daily at 12-h intervals. Degenerative cells were detected by fluoro-jade B (FJ-B) immunostaining in brain regions where expression of pro-inflammatory cytokines was also determined. The effects of pioglitazone on cognitive function were assessed in an operant reversal learning task and the Morris water maze task. Binge alcohol exposure produced selective neuronal degeneration in the hippocampal dentate gyrus and the adjacent entorhinal cortex. Pioglitazone reduced FJ-B positive cells in both regions and prevented alcohol-induced expression of pro-inflammatory cytokines. Pioglitazone also rescued alcohol-impaired reversal learning in the operant task and spatial learning deficits in the Morris water maze. These findings demonstrate that activation of PPARγ protects against neuronal and cognitive degeneration elicited by binge alcohol exposure. The protective effect of PPARγ agonist appears to be linked to inhibition of pro-inflammatory cytokines.


Assuntos
Comportamento Animal/efeitos dos fármacos , Etanol/toxicidade , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , PPAR gama/agonistas , Tiazolidinedionas/administração & dosagem , Animais , Concentração Alcoólica no Sangue , Citocinas/metabolismo , Etanol/administração & dosagem , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Atividade Motora/efeitos dos fármacos , Neurônios/patologia , Pioglitazona , Ratos Wistar , Aprendizagem Espacial/efeitos dos fármacos , Memória Espacial/efeitos dos fármacos
7.
Alcohol Clin Exp Res ; 39(3): 445-54, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25703036

RESUMO

BACKGROUND: Fetal alcohol spectrum disorders (FASD) result from fetal exposure to alcohol and are the leading cause of mental retardation in the United States. There is currently no effective treatment that targets the causes of these disorders. Thus, novel therapies are critically needed to limit the neurodevelopmental and neurodegenerative pathologies associated with FASD. METHODS: A neonatal mouse FASD model was used to examine the role of the neuroimmune system in ethanol (EtOH)-induced neuropathology. Neonatal C57BL/6 mice were treated with EtOH, with or without pioglitazone, on postnatal days 4 through 9, and tissue was harvested 1 day post treatment. Pioglitazone is a peroxisome proliferator-activated receptor (PPAR)-γ agonist that exhibits anti-inflammatory activity and is neuroprotective. We compared the effects of EtOH with or without pioglitazone on cytokine and chemokine expression and microglial morphology in the hippocampus, cerebellum, and cerebral cortex. RESULTS: In EtOH-treated animals compared with controls, cytokines interleukin-1ß and tumor necrosis factor-α mRNA levels were increased significantly in the hippocampus, cerebellum, and cerebral cortex. Chemokine CCL2 mRNA was increased significantly in the hippocampus and cerebellum. Pioglitazone effectively blocked the EtOH-induced increase in the cytokines and chemokine in all tissues to the level expressed in handled-only and vehicle-treated control animals. EtOH also produced a change in microglial morphology in all brain regions that was indicative of microglial activation, and pioglitazone blocked this EtOH-induced morphological change. CONCLUSIONS: These studies indicate that EtOH activates microglia to a pro-inflammatory stage and also increases the expression of neuroinflammatory cytokines and chemokines in diverse regions of the developing brain. Further, the anti-inflammatory and neuroprotective PPAR-γ agonist pioglitazone blocked these effects. It is proposed that microglial activation and inflammatory molecules expressed as a result of EtOH treatment during brain development contribute to the sequelae associated with FASD. Thus, pioglitazone and anti-inflammatory pharmaceuticals more broadly have potential as novel therapeutics for FASD.


Assuntos
Modelos Animais de Doenças , Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/prevenção & controle , Imunidade Celular/efeitos dos fármacos , Microglia/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Animais , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Etanol/antagonistas & inibidores , Feminino , Transtornos do Espectro Alcoólico Fetal/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Imunidade Celular/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Pioglitazona , Gravidez , Tiazolidinedionas/uso terapêutico
8.
Alcohol Clin Exp Res ; 38(2): 384-91, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24033454

RESUMO

BACKGROUND: Alcohol use occurs across the life span beginning in adolescence and continuing through adulthood. Ethanol (EtOH)-induced pathology varies with age and includes changes in neurogenesis, neurodegeneration, and glial cell activation. EtOH-induced changes in glial activation and immune activity are believed to contribute to EtOH-induced neuropathology. Recent studies indicate an emerging role of glial-derived neuroimmune molecules in alcohol abuse and addiction. METHODS: Adolescent and adult C57BL/6 mice were treated via gavage with 6 g/kg EtOH for 10 days, and tissue was harvested 1 day post treatment. We compared the effects of EtOH on chemokine and cytokine expression and astrocyte glial fibrillary acidic protein (GFAP) immunostaining and morphology in the hippocampus, cerebellum, and cerebral cortex. RESULTS: EtOH increased mRNA levels of the chemokine CCL2/MCP-1 in all 3 regions of adult mice relative to controls. The cytokine interleukin-6 (IL-6) was selectively increased only in the adult cerebellum. EtOH did not affect mRNA levels of the cytokine tumor necrosis factor-alpha (TNF-α) in any of these brain regions in adult animals. Interestingly, CCL2, IL-6, and TNF-α mRNA levels were not increased in the hippocampus, cerebellum, or cortex of adolescent mice. EtOH treatment of adult and adolescent mice resulted in increased GFAP immunostaining. CONCLUSIONS: Collectively, these data indicate an age- and region-specific susceptibility to EtOH regulation of neuroinflammatory and addiction-related molecules as well as astrocyte phenotype. These studies may have important implications concerning differential alcohol-induced neuropathology and alcohol addiction across the life span.


Assuntos
Envelhecimento/imunologia , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Imunidade/efeitos dos fármacos , Envelhecimento/fisiologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/imunologia , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Cerebelo/imunologia , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/imunologia , Quimiocina CCL2/biossíntese , DNA Complementar/biossíntese , DNA Complementar/isolamento & purificação , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/imunologia , Imunidade Celular/efeitos dos fármacos , Imuno-Histoquímica , Interleucina-6/biossíntese , Camundongos Endogâmicos C57BL , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Fator de Necrose Tumoral alfa/biossíntese
9.
J Neuroinflammation ; 10: 66, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23701841

RESUMO

BACKGROUND: Alcohol abuse has dramatic effects on the health of the elderly. Recent studies indicate that ethanol increases immune activity in younger animals and that some of these proinflammatory molecules alter alcohol consumption and addiction. However, the effects of alcohol on immune activation in aged animals have not been thoroughly investigated. FINDINGS: We compared the effects of ethanol on chemokine and cytokine expression in the hippocampus, cerebellum, and cerebral cortex of aged C57BL/6 mice. Mice were treated via gavage with 6 g/kg ethanol for 10 days and tissue was harvested 1 day post-treatment. Ethanol selectively increased mRNA levels of the chemokine (C-C motif) ligand 2/monocyte chemotactic protein-1 in the hippocampus and cerebellum, but not in the cortex of aged mice relative to control animals. In this paradigm, ethanol did not affect mRNA levels of the cytokines IL-6 or TNF-α in any of these brain regions in aged animals. CONCLUSIONS: Collectively, these data indicate a region-specific susceptibility to ethanol regulation of neuroinflammatory and addiction-related molecules in aged mice. These studies could have important implications concerning alcohol-induced neuropathology and alcohol addiction in the elderly.


Assuntos
Envelhecimento/imunologia , Envelhecimento/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Imunidade/efeitos dos fármacos , Animais , Depressores do Sistema Nervoso Central/sangue , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Quimiocina CCL2/biossíntese , DNA Complementar/biossíntese , Etanol/sangue , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real
10.
Cells ; 12(16)2023 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-37626919

RESUMO

Ethanol induces neuroinflammation, which is believed to contribute to the pathogenesis of alcohol use disorder (AUD). Toll-like receptors (TLRs) are a group of pattern recognition receptors (PRRs) expressed on both immune cells, including microglia and astrocytes, and non-immune cells in the central nervous system (CNS). Studies have shown that alcohol activates TLR4 signaling, resulting in the induction of pro-inflammatory cytokines and chemokines in the CNS. However, the effect of alcohol on signaling pathways downstream of TLR4, such as MyD88 and TRIF (TICAM) signaling, has not been evaluated extensively. In the current study, we treated male wild-type, TLR4-, MyD88-, and TRIF-deficient mice using a chronic plus binge mouse model of AUD. Evaluation of mRNA expression by qRT-PCR revealed that ethanol increased IL-1ß, TNF-α, CCL2, COX2, FosB, and JunB in the cerebellum in wild-type and TRIF-deficient mice, while ethanol generally did not increase the expression of these molecules in TLR4- and MyD88-deficient mice. Furthermore, IRF3, IRF7, and IFN-ß1, which are associated with the TRIF-dependent signaling cascade, were largely unaffected by alcohol. Collectively, these results suggest that the TLR4 and downstream MyD88-dependent signaling pathways are essential in ethanol-induced neuroinflammation in this mouse model of AUD.


Assuntos
Alcoolismo , Masculino , Animais , Camundongos , Etanol , Fator 88 de Diferenciação Mieloide , Receptor 4 Toll-Like , Doenças Neuroinflamatórias , Proteínas Adaptadoras de Transdução de Sinal , Modelos Animais de Doenças , Proteínas Adaptadoras de Transporte Vesicular
11.
Cells ; 12(5)2023 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-36899881

RESUMO

Alcohol use disorder (AUD) is one of the most common preventable mental health disorders and can result in pathology within the CNS, including the cerebellum. Cerebellar alcohol exposure during adulthood has been associated with disruptions in proper cerebellar function. However, the mechanisms regulating ethanol-induced cerebellar neuropathology are not well understood. High-throughput next generation sequencing was performed to compare control versus ethanol-treated adult C57BL/6J mice in a chronic plus binge model of AUD. Mice were euthanized, cerebella were microdissected, and RNA was isolated and submitted for RNA-sequencing. Down-stream transcriptomic analyses revealed significant changes in gene expression and global biological pathways in control versus ethanol-treated mice that included pathogen-influenced signaling pathways and cellular immune response pathways. Microglial-associated genes showed a decrease in homeostasis-associated transcripts and an increase in transcripts associated with chronic neurodegenerative diseases, while astrocyte-associated genes showed an increase in transcripts associated with acute injury. Oligodendrocyte lineage cell genes showed a decrease in transcripts associated with both immature progenitors as well as myelinating oligodendrocytes. These data provide new insight into the mechanisms by which ethanol induces cerebellar neuropathology and alterations to the immune response in AUD.


Assuntos
Alcoolismo , Etanol , Camundongos , Animais , Etanol/metabolismo , Alcoolismo/patologia , Doenças Neuroinflamatórias , Transcriptoma , Camundongos Endogâmicos C57BL , Cerebelo/metabolismo , Doença Crônica , RNA/metabolismo
12.
Front Neurosci ; 17: 1154637, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37008214

RESUMO

Fetal alcohol spectrum disorders (FASD) are a group of neurodevelopmental disorders caused by ethanol exposure in utero, which can result in neurocognitive and behavioral impairments, growth defects, and craniofacial anomalies. FASD affects up to 1-5% of school-aged children in the United States, and there is currently no cure. The underlying mechanisms involved in ethanol teratogenesis remain elusive and need greater understanding to develop and implement effective therapies. Using a third trimester human equivalent postnatal mouse model of FASD, we evaluate the transcriptomic changes induced by ethanol exposure in the cerebellum on P5 and P6, after only 1 or 2 days of ethanol exposure, with the goal of shedding light on the transcriptomic changes induced early during the onset and development of FASD. We have highlighted key pathways and cellular functions altered by ethanol exposure, which include pathways related to immune function and cytokine signaling as well as the cell cycle. Additionally, we found that ethanol exposure resulted in an increase in transcripts associated with a neurodegenerative microglia phenotype, and acute- and pan-injury reactive astrocyte phenotypes. Mixed effects on oligodendrocyte lineage cell associated transcripts and cell cycle associated transcripts were observed. These studies help to elucidate the underlying mechanisms that may be involved with the onset of FASD and provide further insights that may aid in identifying novel targets for interventions and therapeutics.

13.
Brain Behav Immun ; 25 Suppl 1: S137-45, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21376806

RESUMO

Fetal alcohol spectrum disorders (FASD) result from ethanol exposure to the developing fetus and are the most common cause of mental retardation in the United States. These disorders are characterized by a variety of neurodevelopmental and neurodegenerative anomalies which result in significant lifetime disabilities. Thus, novel therapies are required to limit the devastating consequences of FASD. Neuropathology associated with FASD can occur throughout the central nervous system (CNS), but is particularly well characterized in the developing cerebellum. Rodent models of FASD have previously demonstrated that both Purkinje cells and granule cells, which are the two major types of neurons in the cerebellum, are highly susceptible to the toxic effects of ethanol. The current studies demonstrate that ethanol decreases the viability of cultured cerebellar granule cells and microglial cells. Interestingly, microglia have dual functionality in the CNS. They provide trophic and protective support to neurons. However, they may also become pathologically activated and produce inflammatory molecules toxic to parenchymal cells including neurons. The findings in this study demonstrate that the peroxisome proliferator-activated receptor-γ agonists 15-deoxy-Δ12,15 prostaglandin J2 and pioglitazone protect cultured granule cells and microglia from the toxic effects of ethanol. Furthermore, investigations using a newly developed mouse model of FASD and stereological cell counting methods in the cerebellum elucidate that ethanol administration to neonates is toxic to both Purkinje cell neurons as well as microglia, and that in vivo administration of PPAR-γ agonists protects these cells. In composite, these studies suggest that PPAR-γ agonists may be effective in limiting ethanol-induced toxicity to the developing CNS.


Assuntos
Encéfalo/efeitos dos fármacos , Etanol/farmacologia , Transtornos do Espectro Alcoólico Fetal/tratamento farmacológico , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , PPAR gama/agonistas , Análise de Variância , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Contagem de Células , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Transtornos do Espectro Alcoólico Fetal/metabolismo , Transtornos do Espectro Alcoólico Fetal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , Neurônios/metabolismo , Neurônios/patologia , PPAR gama/farmacologia , PPAR gama/uso terapêutico , Pioglitazona , Gravidez , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacologia , Prostaglandina D2/uso terapêutico , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico
14.
Alcohol ; 96: 43-53, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34358666

RESUMO

Fetal alcohol spectrum disorders (FASD) are alarmingly common, result in significant personal and societal loss, and there are no effective treatments for these disorders. Cerebellar neuropathology is common in FASD and can cause impaired cognitive and motor function. The current study evaluates the effects of ethanol on oligodendrocyte-lineage cells, as well as molecules that modulate oligodendrocyte differentiation and function in the cerebellum in a postnatal mouse model of FASD. Neonatal mice were treated with ethanol from P4-P9 (postnatal day), the cerebellum was isolated at P10, and mRNAs encoding oligodendrocyte-associated molecules were quantitated by qRT-PCR. Our studies demonstrated that ethanol significantly reduced the expression of markers for multiple stages of oligodendrocyte maturation, including oligodendrocyte precursor cells, pre-myelinating oligodendrocytes, and mature myelinating oligodendrocytes. Additionally, we determined that ethanol significantly decreased the expression of molecules that play critical roles in oligodendrocyte differentiation. Interestingly, we also observed that ethanol significantly reduced the expression of myelin-associated inhibitors, which may act as a compensatory mechanism to ethanol toxicity. Furthermore, we demonstrate that ethanol alters the expression of a variety of molecules important in oligodendrocyte function and myelination. Collectively, our studies increase our understanding of specific mechanisms by which ethanol modulates myelination in the developing cerebellum, and potentially identify novel targets for FASD therapy.


Assuntos
Transtornos do Espectro Alcoólico Fetal , Animais , Cerebelo , Etanol/toxicidade , Feminino , Transtornos do Espectro Alcoólico Fetal/genética , Camundongos , Bainha de Mielina , Oligodendroglia , Gravidez
15.
Neurotoxicol Teratol ; 87: 107015, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34256161

RESUMO

Fetal alcohol spectrum disorders (FASD) are alarmingly common and result in significant personal and societal loss. Neuropathology of the hippocampus is common in FASD leading to aberrant cognitive function. In the current study, we evaluated the effects of ethanol on the expression of a targeted set of molecules involved in neuroinflammation, myelination, neurotransmission, and neuron function in the developing hippocampus in a postnatal model of FASD. Mice were treated with ethanol from P4-P9, hippocampi were isolated 24 h after the final treatment at P10, and mRNA levels were quantitated by qRT-PCR. We evaluated the effects of ethanol on both pro-inflammatory and anti-inflammatory molecules in the hippocampus and identified novel mechanisms by which ethanol induces neuroinflammation. We further demonstrated that ethanol decreased expression of molecules associated with mature oligodendrocytes and greatly diminished expression of a lacZ reporter driven by the first half of the myelin proteolipid protein (PLP) gene (PLP1). In addition, ethanol caused a decrease in genes expressed in oligodendrocyte progenitor cells (OPCs). Together, these studies suggest ethanol may modulate pathogenesis in the developing hippocampus through effects on cells of the oligodendrocyte lineage, resulting in altered oligodendrogenesis and myelination. We also observed differential expression of molecules important in synaptic plasticity, neurogenesis, and neurotransmission. Collectively, the molecules evaluated in these studies may play a role in ethanol-induced pathology in the developing hippocampus and contribute to cognitive impairment associated with FASD. A better understanding of these molecules and their effects on the developing hippocampus may lead to novel treatment strategies for FASD.


Assuntos
Etanol/farmacologia , Transtornos do Espectro Alcoólico Fetal/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Doenças Neuroinflamatórias/tratamento farmacológico , Animais , Modelos Animais de Doenças , Transtornos do Espectro Alcoólico Fetal/fisiopatologia , Hipocampo/metabolismo , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Neurogênese/fisiologia , Oligodendroglia/patologia
16.
Alcohol ; 42(1): 29-36, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18249267

RESUMO

Oxidative stress is a candidate mechanism for ethanol neuropathology in fetal alcohol spectrum disorders. Oxidative stress often involves production of reactive oxygen species (ROS), deterioration of the mitochondrial membrane potential (MMP), and cell death. Previous studies have produced conflicting results regarding the role of oxidative stress and the benefit of antioxidants in ethanol neuropathology in the developing brain. This study investigated the hypothesis that ethanol neurotoxicity involves production of ROS with negative downstream consequences for MMP and neuron survival. This was modeled in neonatal rats at postnatal day 4 (P4) and P14. It is well established that granule neurons in the rat cerebellar cortex are more vulnerable to ethanol neurotoxicity on P4 than at later ages. Thus, it was hypothesized that ethanol produces more oxidative stress and its negative consequences on P4 than on P14. A novel experimental approach was used in which ethanol was administered to animals in vivo (gavage 6g/kg), granule neurons were isolated 2-24h post-treatment, and ROS production and relative MMP were immediately assessed in the viable cells. Cells were also placed in culture and survival was measured 24h later. The results revealed that ethanol did not induce granule cells to produce ROS, cause deterioration of neuronal MMP, or cause neuron death when compared to vehicle controls. Further, granule neurons from neither P4 nor P14 animals mounted an oxidative response to ethanol. These findings do not support the hypothesis that oxidative stress is obligate to granule neuron death after ethanol exposure in the neonatal rat brain. Other investigators have reached a similar conclusion using either brain homogenates or cell cultures. In this context, it is likely that oxidative stress is not the sole and perhaps not the principal mechanism of ethanol neurotoxicity for cerebellar granule neurons during this stage of brain development.


Assuntos
Cerebelo/efeitos dos fármacos , Etanol/toxicidade , Estresse Oxidativo , Animais , Animais Recém-Nascidos , Biomarcadores , Metaloproteinases da Matriz/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
17.
J Clin Cell Immunol ; 7(6)2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28203487

RESUMO

Fetal alcohol spectrum disorders (FASD) result from fetal exposure to alcohol during pregnancy. These disorders present a variety of sequelae including involvement of the central nervous system (CNS) with lasting impact on cognitive function and behavior. FASD occur at an alarming rate and have significant personal and societal impact. There are currently no effective treatments for FASD. Recent studies demonstrate that ethanol induces potent neuroinflammation in many regions of the developing brain. Furthermore, anti-inflammatory agents such as peroxisome proliferator-activated receptor (PPAR)-γ agonists suppress ethanol-induced neuroinflammation and neurodegeneration. This suggests that anti-inflammatory agents may be effective in treatment of FASD. Future studies designed to determine the specific mechanisms by which alcohol induces neuroinflammation in the developing CNS may lead to targeted therapies for FASD.

18.
J Leukoc Biol ; 100(5): 951-959, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27462100

RESUMO

Fetal alcohol spectrum disorder (FASD), which results from ethanol exposure during pregnancy, and alcohol use disorder (AUD), which includes both binge and chronic alcohol abuse, are strikingly common and costly at personal and societal levels. These disorders are associated with significant pathology, including that observed in the CNS. It is now appreciated in both humans and animal models that ethanol can induce inflammation in the CNS. Neuroinflammation is hypothesized to contribute to the neuropathologic and behavioral consequences in FASD and AUD. In this review, we: 1) summarize the evidence of alcohol-induced CNS inflammation, 2) outline cellular and molecular mechanisms that may underlie alcohol induction of CNS inflammation, and 3) discuss the potential of nuclear receptor agonists for prevention or treatment of neuropathologies associated with FASD and AUD.


Assuntos
Neuropatia Alcoólica/imunologia , Sistema Nervoso Central/imunologia , Encefalomielite/induzido quimicamente , Adolescente , Adulto , Fatores Etários , Neuropatia Alcoólica/terapia , Alcoolismo/complicações , Alcoolismo/epidemiologia , Animais , Receptor 1 de Quimiocina CX3C , Quimiocina CX3CL1/imunologia , Criança , Transtornos Cognitivos/induzido quimicamente , Modelos Animais de Doenças , Encefalomielite/imunologia , Feminino , Transtornos do Espectro Alcoólico Fetal/imunologia , Transtornos do Espectro Alcoólico Fetal/prevenção & controle , Humanos , Recém-Nascido , Inflamassomos/efeitos dos fármacos , Masculino , Transtornos Mentais/induzido quimicamente , Microglia/efeitos dos fármacos , Microglia/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Receptores de Citocinas/imunologia , Receptores de HIV/imunologia , Receptor 4 Toll-Like/imunologia
19.
Curr Med Chem ; 11(21): 2883-902, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15544481

RESUMO

Protein kinase C (PKC) is a family of serine/threonine kinases that regulates a variety of cell functions including proliferation, gene expression, cell cycle, differentiation, cytoskeletal organization, cell migration, and apoptosis. The PKC signal transduction cascade coordinates complex physiological events including normal tissue function and repair. Disruption of the cellular environment through genetic mutation, disease, injury, or exposure to pro-oxidants, alcohol, or other insults can induce pathological PKC activation. Aberrant PKC activation can lead to diseases of cellular dysregulation such as cancer and diabetes. Can aberrant activation of PKC be reversed? Even 25 years after the identification of PKC, therapeutic regulation of PKC activity remains an emerging field. Because the function of each isoform remains to be elucidated, isoform specific control of gene expression is a current challenge. Natural compounds are important regulators of PKC activity, with both preventive and therapeutic efficacy. Antioxidants including vitamin A (retinoids), vitamin C (ascorbic acid) and vitamin E (tocopherols) show promise for reversal of PKC activation. beta-carotene and retinoids function as anticarcinogenic agents and antagonize the biological effects of pro-oxidants on PKC. Vitamin E reverses the deleterious effects of hyperglycemia and diabetes by down-regulating PKC activity. Antioxidants in red wine provide cardioprotective effects. However, alcohol consumption also induces oxidative stress and disrupts PKC and retinoid function in the fetus and the adult. This review examines modulation of PKC activity by natural compounds and pharmacologic analogues which can be used effectively to prevent or treat common diseases associated with aberrant activation of PKC.


Assuntos
Fatores Biológicos/farmacologia , Fatores Biológicos/uso terapêutico , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Animais , Doença , Humanos , Proteína Quinase C/classificação , Transdução de Sinais/efeitos dos fármacos , Vitaminas/farmacologia
20.
Brain Res ; 924(1): 71-81, 2002 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-11743997

RESUMO

Ethanol causes loss of Purkinje cells in the cerebellum during the early stages of differentiation and maturation by a presently unknown mechanism. Neuronal vulnerability in the cerebellum parallels the prominent temporal and anatomical gradients of development (i.e. early to late interlobular and posterior to anterior, respectively). Development of Purkinje cells is known to require binding of the neurotrophins, including brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT3), to the tyrosine-kinase (Trk) receptors TrkB and TrkC, respectively. In addition, Purkinje cells are reported to experience a critical switch between BDNF dependence and NT3 dependence during the period of highest ethanol sensitivity between postnatal days (PN) 4-6. To test the hypothesis that ethanol alters neurotrophin signaling leading to Purkinje neuronal death, the immunohistochemical expression of TrkB and TrkC receptors on Purkinje cells of rat pups following a moderate dose of ethanol was determined at various times surrounding the period of postnatal ethanol vulnerability. Ethanol selectively decreased Purkinje cell expression of TrkB and TrkC receptors following exposures within the vulnerable period (PN4-6). These results suggest that ethanol may induce loss of Purkinje cells by alteration of neurotrophic regulation at this critical stage.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Córtex Cerebelar/efeitos dos fármacos , Etanol/farmacologia , Degeneração Neural/induzido quimicamente , Fatores de Crescimento Neural/metabolismo , Células de Purkinje/efeitos dos fármacos , Receptores de Fator de Crescimento Neural/efeitos dos fármacos , Transtornos do Sistema Nervoso Induzidos por Álcool/metabolismo , Transtornos do Sistema Nervoso Induzidos por Álcool/fisiopatologia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Diferenciação Celular/fisiologia , Córtex Cerebelar/crescimento & desenvolvimento , Córtex Cerebelar/patologia , Feminino , Transtornos do Espectro Alcoólico Fetal/metabolismo , Transtornos do Espectro Alcoólico Fetal/fisiopatologia , Imuno-Histoquímica , Masculino , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Neurotrofina 3/metabolismo , Gravidez , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Ratos , Ratos Sprague-Dawley , Receptor trkB/metabolismo , Receptor trkC/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA