Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.227
Filtrar
1.
Alcohol Alcohol ; 59(4)2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38742547

RESUMO

AIMS: Continued alcohol consumption despite negative consequences is a core symptom of alcohol use disorder. This is modeled in mice by pairing negative stimuli with alcohol, such as adulterating alcohol solution with quinine. Mice consuming alcohol under these conditions are considered to be engaging in aversion-resistant intake. Previously, we have observed sex differences in this behavior, with females more readily expressing aversion-resistant consumption. We also identified three brain regions that exhibited sex differences in neuronal activation during quinine-alcohol drinking: ventromedial prefrontal cortex (vmPFC), posterior insular cortex (PIC), and ventral tegmental area (VTA). Specifically, male mice showed increased activation in vmPFC and PIC, while females exhibited increased activation in VTA. In this study, we aimed to identify what specific type of neurons are activated in these regions during quinine-alcohol drinking. METHOD: We assessed quinine-adulterated alcohol intake using the two-bottle choice procedure. We also utilized RNAscope in situ hybridization in the three brain regions that previously exhibited a sex difference to examine colocalization of Fos, glutamate, GABA, and dopamine. RESULT: Females showed increased aversion-resistant alcohol consumption compared to males. We also found that males had higher colocalization of glutamate and Fos in vmPFC and PIC, while females had greater dopamine and Fos colocalization in the VTA. CONCLUSIONS: Collectively, these experiments suggest that glutamatergic output from the vmPFC and PIC may have a role in suppressing, and dopaminergic activity in the VTA may promote, aversion-resistant alcohol consumption. Future experiments will examine neuronal circuits that contribute to sex differences in aversion resistant consumption.


Assuntos
Consumo de Bebidas Alcoólicas , Neurônios , Quinina , Caracteres Sexuais , Animais , Quinina/farmacologia , Feminino , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/efeitos dos fármacos , Mesencéfalo/metabolismo , Mesencéfalo/efeitos dos fármacos , Córtex Insular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Etanol/farmacologia , Ácido Glutâmico/metabolismo
2.
J Neurosci ; 43(3): 472-483, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36639890

RESUMO

Social deficits and dysregulations in dopaminergic midbrain-striato-frontal circuits represent transdiagnostic symptoms across psychiatric disorders. Animal models suggest that interactions between the dopamine (DA) and renin-angiotensin system (RAS) may modulate learning and reward-related processes. The present study therefore examined the behavioral and neural effects of the Angiotensin II type 1 receptor (AT1R) antagonist losartan on social reward and punishment processing in humans. A preregistered randomized double-blind placebo-controlled between-subject pharmacological design was combined with a social incentive delay (SID) functional MRI (fMRI) paradigm during which subjects could avoid social punishment or gain social reward. Healthy volunteers received a single-dose of losartan (50 mg, n = 43, female = 17) or placebo (n = 44, female = 20). We evaluated reaction times (RTs) and emotional ratings as behavioral and activation and functional connectivity as neural outcomes. Relative to placebo, losartan modulated the reaction time and arousal differences between social punishment and social reward. On the neural level the losartan-enhanced motivational salience of social rewards was accompanied by stronger ventral striatum-prefrontal connectivity during reward anticipation. Losartan increased the reward-neutral difference in the ventral tegmental area (VTA) and attenuated VTA associated connectivity with the bilateral insula in response to punishment during the outcome phase. Thus, losartan modulated approach-avoidance motivation and emotional salience during social punishment versus social reward via modulating distinct core nodes of the midbrain-striato-frontal circuits. The findings document a modulatory role of the renin-angiotensin system in these circuits and associated social processes, suggesting a promising treatment target to alleviate social dysregulations.SIGNIFICANCE STATEMENT Social deficits and anhedonia characterize several mental disorders and have been linked to the midbrain-striato-frontal circuits of the brain. Based on initial findings from animal models we here combine the pharmacological blockade of the Angiotensin II type 1 receptor (AT1R) via losartan with functional MRI (fMRI) to demonstrate that AT1R blockade enhances the motivational salience of social rewards and attenuates the negative impact of social punishment via modulating the communication in the midbrain-striato-frontal circuits in humans. The findings demonstrate for the first time an important role of the AT1R in social reward processing in humans and render the AT1R as promising novel treatment target for social and motivational deficits in mental disorders.


Assuntos
Losartan , Mesencéfalo , Motivação , Animais , Feminino , Humanos , Angiotensinas/antagonistas & inibidores , Dopamina/farmacologia , Losartan/farmacologia , Imageamento por Ressonância Magnética , Mesencéfalo/diagnóstico por imagem , Mesencéfalo/efeitos dos fármacos , Motivação/efeitos dos fármacos , Punição/psicologia , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Recompensa
3.
Nat Commun ; 13(1): 504, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-35082287

RESUMO

The pedunculopontine nucleus (PPN) is a locomotor command area containing glutamatergic neurons that control locomotor initiation and maintenance. These motor actions are deficient in Parkinson's disease (PD), where dopaminergic neurodegeneration alters basal ganglia activity. Being downstream of the basal ganglia, the PPN may be a suitable target for ameliorating parkinsonian motor symptoms. Here, we use in vivo cell-type specific PPN activation to restore motor function in two mouse models of parkinsonism made by acute pharmacological blockage of dopamine transmission. With a combination of chemo- and opto-genetics, we show that excitation of caudal glutamatergic PPN neurons can normalize the otherwise severe locomotor deficit in PD, whereas targeting the local GABAergic population only leads to recovery of slow locomotion. The motor rescue driven by glutamatergic PPN activation is independent of activity in nearby locomotor promoting glutamatergic Cuneiform neurons. Our observations point to caudal glutamatergic PPN neurons as a potential target for neuromodulatory restoration of locomotor function in PD.


Assuntos
Mesencéfalo/metabolismo , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Transtornos Parkinsonianos/metabolismo , Animais , Modelos Animais de Doenças , Dopamina/metabolismo , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Feminino , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Doença de Parkinson/fisiopatologia , Transtornos Parkinsonianos/fisiopatologia , Núcleo Tegmental Pedunculopontino/metabolismo , Núcleo Tegmental Pedunculopontino/fisiopatologia
4.
Clin Transl Med ; 12(1): e692, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35090094

RESUMO

BACKGROUND: Parkinson's disease (PD) is the second most common neurodegenerative disorder whose prevalence is rapidly increasing worldwide. The molecular mechanisms underpinning the pathophysiology of sporadic PD remain incompletely understood. Therefore, causative therapies are still elusive. To obtain a more integrative view of disease-mediated alterations, we investigated the molecular landscape of PD in human post-mortem midbrains, a region that is highly affected during the disease process. METHODS: Tissue from 19 PD patients and 12 controls were obtained from the Parkinson's UK Brain Bank and subjected to multi-omic analyses: small and total RNA sequencing was performed on an Illumina's HiSeq4000, while proteomics experiments were performed in a hybrid triple quadrupole-time of flight mass spectrometer (TripleTOF5600+) following quantitative sequential window acquisition of all theoretical mass spectra. Differential expression analyses were performed with customized frameworks based on DESeq2 (for RNA sequencing) and with Perseus v.1.5.6.0 (for proteomics). Custom pipelines in R were used for integrative studies. RESULTS: Our analyses revealed multiple deregulated molecular targets linked to known disease mechanisms in PD as well as to novel processes. We have identified and experimentally validated (quantitative real-time polymerase chain reaction/western blotting) several PD-deregulated molecular candidates, including miR-539-3p, miR-376a-5p, miR-218-5p and miR-369-3p, the valid miRNA-mRNA interacting pairs miR-218-5p/RAB6C and miR-369-3p/GTF2H3, as well as multiple proteins, such as CHI3L1, HSPA1B, FNIP2 and TH. Vertical integration of multi-omic analyses allowed validating disease-mediated alterations across different molecular layers. Next to the identification of individual molecular targets in all explored omics layers, functional annotation of differentially expressed molecules showed an enrichment of pathways related to neuroinflammation, mitochondrial dysfunction and defects in synaptic function. CONCLUSIONS: This comprehensive assessment of PD-affected and control human midbrains revealed multiple molecular targets and networks that are relevant to the disease mechanism of advanced PD. The integrative analyses of multiple omics layers underscore the importance of neuroinflammation, immune response activation, mitochondrial and synaptic dysfunction as putative therapeutic targets for advanced PD.


Assuntos
Mesencéfalo/patologia , Terapia de Alvo Molecular/métodos , Doença de Parkinson/terapia , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Mesencéfalo/anatomia & histologia , Mesencéfalo/efeitos dos fármacos , Pessoa de Meia-Idade , Terapia de Alvo Molecular/estatística & dados numéricos , Doença de Parkinson/genética , Doença de Parkinson/mortalidade , Reação em Cadeia da Polimerase em Tempo Real/métodos , Reação em Cadeia da Polimerase em Tempo Real/estatística & dados numéricos , Reino Unido
5.
Nat Commun ; 12(1): 6945, 2021 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-34836948

RESUMO

Long-term exposure to nicotine alters brain circuits and induces profound changes in decision-making strategies, affecting behaviors both related and unrelated to drug seeking and consumption. Using an intracranial self-stimulation reward-based foraging task, we investigated in mice the impact of chronic nicotine on midbrain dopamine neuron activity and its consequence on the trade-off between exploitation and exploration. Model-based and archetypal analysis revealed substantial inter-individual variability in decision-making strategies, with mice passively exposed to nicotine shifting toward a more exploitative profile compared to non-exposed animals. We then mimicked the effect of chronic nicotine on the tonic activity of dopamine neurons using optogenetics, and found that photo-stimulated mice adopted a behavioral phenotype similar to that of mice exposed to chronic nicotine. Our results reveal a key role of tonic midbrain dopamine in the exploration/exploitation trade-off and highlight a potential mechanism by which nicotine affects the exploration/exploitation balance and decision-making.


Assuntos
Comportamento Exploratório/efeitos dos fármacos , Mesencéfalo/efeitos dos fármacos , Nicotina/efeitos adversos , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Comportamento Exploratório/fisiologia , Masculino , Mesencéfalo/citologia , Mesencéfalo/metabolismo , Camundongos , Modelos Animais , Nicotina/administração & dosagem , Optogenética , Preconceito , Recompensa , Autoadministração , Técnicas Estereotáxicas
6.
Pak J Pharm Sci ; 34(3): 861-867, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34602407

RESUMO

This experiment proposed to study the efficiency omega 3 fatty acid on behavioural phenotype of Parkinson's disease (PD) in mice. Totally 7 groups (each group 6 mice) were used in this assessment, each groups were treated with saline (control), MPP+, L-DOPA, Omega 3 oil, Omega 3 oil (three different concentrations) +MPP+ separately. The behavioral assessments such as bar test, open field test, maze test, hang test were noted on 7th, 14th, 21st and 28th day. After the examination period, the tested animals' midbrains and frontal cortex were dissected to analyze TBARS, GSH, Catalase, Superoxide Dismutase and Glutathione Peroxidase assay. In the bar test, 500mg omega 3 fatty acid administrated mice showed a high cataleptic scores. In open field Test, significant reductions in behavior analysis were observed from the tested mice group. Maze test and hang test doesn't show much difference. In biochemical test, tested groups showed promising results compared to control group. The result strongly proved that the omega 3 fatty acid has remarkable abilities to control the neurodegenerative diseases.


Assuntos
Antiparkinsonianos/farmacologia , Comportamento Animal/efeitos dos fármacos , Ácidos Graxos Ômega-3/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Transtornos Parkinsonianos/fisiopatologia , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Catalase/efeitos dos fármacos , Catalase/metabolismo , Reação de Congelamento Cataléptica , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Glutationa/efeitos dos fármacos , Glutationa/metabolismo , Glutationa Peroxidase/efeitos dos fármacos , Glutationa Peroxidase/metabolismo , Levodopa/farmacologia , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Camundongos , Teste de Campo Aberto , Transtornos Parkinsonianos/induzido quimicamente , Superóxido Dismutase/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
7.
Behav Brain Res ; 414: 113513, 2021 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-34358575

RESUMO

Male Japanese quail produce high-frequency crow vocalizations to attract females during the breeding season. The nucleus of intercollicularis (ICo) is the midbrain vocal center in birds and electrical stimulation of the ICo produces calls that include crowing. Noradrenaline plays a significant role in sexual behavior but the contribution of noradrenaline in the control of courtship vocalizations in quail has not been well established. Using dose-dependent intracerebroventricular injection of clonidine, an α2-adrenergic receptor-specific agonist, crowing vocalization was immediately suppressed. At the same time as crow suppression by clonidine there was a reduction of immediate early gene, zenk mRNA, in the ICo; no zenk mRNA expression was detected in the dorsomedial division of the nucleus. Using histochemistry, we determined that the ICo receives noradrenergic innervation and expresses α2A-adrenergic receptor mRNA. Taken together, these data suggest that noradrenaline regulates courtship vocalization in quail, possibly via the α2A-adrenergic receptor expressed on ICo neurons.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Corte , Mesencéfalo/metabolismo , Norepinefrina/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Comportamento Sexual Animal/fisiologia , Vocalização Animal/fisiologia , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Animais , Clonidina/farmacologia , Coturnix , Masculino , Mesencéfalo/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Vocalização Animal/efeitos dos fármacos
8.
PLoS One ; 16(8): e0256207, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34403440

RESUMO

Thyroid hormones are messengers that bind to specific nuclear receptors and regulate a wide range of physiological processes in the early stages of vertebrate embryonic development, including neurodevelopment and myelogenesis. We here tested the effects of reduced T3 availability upon the myelination process by treating zebrafish embryos with low concentrations of iopanoic acid (IOP) to block T4 to T3 conversion. Black Gold II staining showed that T3 deficiency reduced the myelin density in the forebrain, midbrain, hindbrain and the spinal cord at 3 and 7 dpf. These observations were confirmed in 3 dpf mbp:egfp transgenic zebrafish, showing that the administration of IOP reduced the fluorescent signal in the brain. T3 rescue treatment restored brain myelination and reversed the changes in myelin-related gene expression induced by IOP exposure. NG2 immunostaining revealed that T3 deficiency reduced the amount of oligodendrocyte precursor cells in 3 dpf IOP-treated larvae. Altogether, the present results show that inhibition of T4 to T3 conversion results in hypomyelination, suggesting that THs are part of the key signaling molecules that control the timing of oligodendrocyte differentiation and myelin synthesis from very early stages of brain development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Larva/genética , Bainha de Mielina/genética , Tiroxina/deficiência , Tri-Iodotironina/deficiência , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Antígenos/genética , Antígenos/metabolismo , Embrião não Mamífero , Desenvolvimento Embrionário , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Ácido Iopanoico/farmacologia , Larva/citologia , Larva/efeitos dos fármacos , Larva/crescimento & desenvolvimento , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/crescimento & desenvolvimento , Mesencéfalo/metabolismo , Proteína Proteolipídica de Mielina/genética , Proteína Proteolipídica de Mielina/metabolismo , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/metabolismo , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Fator de Transcrição 2 de Oligodendrócitos/genética , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Oligodendroglia/citologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Prosencéfalo/citologia , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Proteoglicanas/genética , Proteoglicanas/metabolismo , Rombencéfalo/citologia , Rombencéfalo/efeitos dos fármacos , Rombencéfalo/crescimento & desenvolvimento , Rombencéfalo/metabolismo , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/crescimento & desenvolvimento , Medula Espinal/metabolismo , Tri-Iodotironina/farmacologia , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
9.
Neuropharmacology ; 197: 108722, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34273387

RESUMO

Although several ionic mechanisms are known to control rate and regularity of the slow pacemaker in dopamine (DA) neurons, the core mechanism of pacing is controversial. Here we tested the hypothesis that pacemaking of SNc DA neurons is enabled by an unconventional conductance. We found that 1-(2,4-xylyl)guanidinium (XG), an established blocker of gating pore currents, selectively inhibits pacemaking of DA neurons. The compound inhibited all slow pacemaking DA neurons that were tested, both in the substantia nigra pars compacta, and in the ventral tegmental area. Interestingly, bursting behavior was not affected by XG. Furthermore, the drug did not affect fast pacemaking of GABAergic neurons from substantia nigra pars reticulata neurons or slow pacemaking of noradrenergic neurons. In DA neurons, current-clamp analysis revealed that XG did not appear to affect ion channels involved in the action potential. Its inhibitory effect persisted during blockade of all ion channels previously suggested to contribute to pacemaking. RNA sequencing and voltage-clamp recordings yielded no evidence for a gating pore current to underlie the conductance. However, we could isolate a small subthreshold XG-sensitive current, which was carried by both Na+ and Cl- ions. Although the molecular target of XG remains to be defined, these observations represent a step towards understanding pacemaking in DA neurons.


Assuntos
Relógios Biológicos/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Guanidina/análogos & derivados , Guanidina/farmacologia , Mesencéfalo/efeitos dos fármacos , Animais , Neurônios GABAérgicos/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Norepinefrina/fisiologia , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Substância Negra/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos
10.
Cells ; 10(5)2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-34063571

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra. The inflammatory activation of microglia participates in dopaminergic neurodegeneration in PD. Therefore, chemicals that inhibit microglial activation are considered to have therapeutic potential for PD. Aromatic (ar)-turmerone is a main component of turmeric oil extracted from Curcuma longa and has anti-inflammatory activity in cultured microglia. The aims of the present study are (1) to investigate whether naturally occurring S-enantiomer of ar-turmerone (S-Tur) protects dopaminergic neurons in midbrain slice cultures and (2) to examine ar-turmerone analogs that have higher activities than S-Tur in inhibiting microglial activation and protecting dopaminergic neurons. R-enantiomer (R-Tur) and two analogs showed slightly higher anti-inflammatory effects in microglial BV2 cells. S- and R-Tur and these two analogs reversed dopaminergic neurodegeneration triggered by microglial activation in midbrain slice cultures. Unexpectedly, this neuroprotection was independent of the inhibition of microglial activation. Additionally, two analogs more potently inhibited dopaminergic neurodegeneration triggered by a neurotoxin, 1-methyl-4-phenylpyridinium, than S-Tur. Taken together, we identified two ar-turmerone analogs that directly and potently protected dopaminergic neurons. An investigation using dopaminergic neuronal precursor cells suggested the possible involvement of nuclear factor erythroid 2-related factor 2 in this neuroprotection.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Cetonas/farmacologia , Mesencéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Sesquiterpenos/farmacologia , Animais , Linhagem Celular , Células Cultivadas , Cetonas/química , Mesencéfalo/citologia , Camundongos , Microglia/efeitos dos fármacos , Fármacos Neuroprotetores/química , Ratos , Ratos Wistar , Sesquiterpenos/química
11.
Eur J Pharmacol ; 906: 174269, 2021 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-34147477

RESUMO

Icariin and icaritin, the major active components of Epimedii Genus, are considered as promising drugs with anti-inflammatory, anti-aging and neuroprotective effects. Our previous studies have demonstrated that icariin and icaritin can protect against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/1-methyl-4-phenylpyridinium (MPP+)-induced neurotoxicity on dopaminergic neurons via insulin-like growth factor-1 receptor (IGF-1 receptor) signaling. In the present study, we aimed to evaluate the role of IGF-1 receptor signaling in mediating the anti-inflammatory effects of icariin and icaritin against lipopolysaccharide (LPS)-induced neuroinflammation as well as their biological regulation effects in midbrain primary astrocytes. Our results showed that both icariin and icaritin significantly inhibited LPS-induced mRNA expressions of tumor necrosis factor (TNF-α) and interleukin-1ß (IL-1ß). Pre-treatment with IGF-1 receptor antagonist JB-1 could significantly block the anti-inflammatory effects of icariin and icaritin on LPS-induced up-regulations of TNF-α, IL-1ß, cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS). Under basal conditions of astrocytes, icariin and icaritin treatment alone increased the phosphorylation of ERK1/2 and AKT, which could be blocked by JB-1. Moreover, the mRNA expressions of glutamate transptor-1 (GLT-1) and glutamate-aspartate transporter (GLAST) could be up-regulated by icariin and icaritin in a time-dependent manner via IGF-1 receptor. Taken together, our results suggest for the first time that both icariin and icaritin exert regulatory effects in astrocytes under basal conditions and after an inflammatory challenge via IGF-1 receptor signaling pathway.


Assuntos
Astrócitos/patologia , Flavonoides/farmacologia , Doenças Neuroinflamatórias/tratamento farmacológico , Receptor IGF Tipo 1/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Flavonoides/isolamento & purificação , Flavonoides/uso terapêutico , Humanos , Lipopolissacarídeos/imunologia , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/patologia , Camundongos , Doenças Neuroinflamatórias/imunologia , Doenças Neuroinflamatórias/patologia , Cultura Primária de Células , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
12.
Biomed Pharmacother ; 141: 111832, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34153844

RESUMO

The pathological characteristics of Parkinson's disease (PD) include dopaminergic neuron damage, specifically disorders caused by dopamine synthesis, in vivo. Plastrum testudinis extract (PTE) and its bioactive ingredient ethyl stearate (PubChem CID: 8122) were reported to be correlated with tyrosine hydroxylase (TH), which is a biomarker of dopaminergic neurons. This suggests that PTE and its small-molecule active ingredient ethyl stearate have potential for development as a therapeutic drug for PD. In this study, we treated 6-hydroxydopamine (6-OHDA)-induced model rats and PC12 cells with PTE. The mechanism of action of PTE and ethyl stearate was investigated by western blotting, bisulfite sequencing PCR (BSP), real-time PCR, immunofluorescence and siRNA transfection. PTE effectively upregulated the TH expression and downregulated the alpha-synuclein expression in both the substantia nigra and the striatum of the midbrain in a PD model rat. The PC12 cell model showed that both PTE and its active monomer ethyl stearate significantly promoted TH expression and blocked alpha-synuclein, agreeing with the in vivo results. BSP showed that PTE and ethyl stearate increased the methylation level of the Snca intron 1 region. These findings suggest that some of the protective effects of PTE on dopaminergic neurons are mediated by ethyl stearate. The mechanism of ethyl stearate may involve disrupting the abnormal aggregation of DNA (cytosine-5)-methyltransferase 1 (DNMT1) with alpha-synuclein by releasing DNMT1, upregulating Snca intron 1 CpG island methylation, and ultimately, reducing the expression of alpha-synuclein.


Assuntos
Antiparkinsonianos/farmacologia , Antiparkinsonianos/uso terapêutico , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson Secundária/tratamento farmacológico , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Extratos de Tecidos/química , alfa-Sinucleína/metabolismo , Animais , DNA (Citosina-5-)-Metiltransferase 1/efeitos dos fármacos , Hidroxidopaminas , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Células PC12 , Doença de Parkinson Secundária/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Estearatos/farmacologia , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , alfa-Sinucleína/efeitos dos fármacos
13.
J Chem Neuroanat ; 116: 101992, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34166778

RESUMO

We present a robust, fresh-frozen approach to immunohistochemistry (IHC), without committing the tissue to IHC via fixation and cryopreservation while maintaining long-term storage, using LiCor-based infrared (IR) quantification for sensitive assessment of TH in immunoreacted midbrain sections for quantitative comparison across studies. In fresh-frozen tissue stored up to 1 year prior to IHC reaction, we found our method to be highly sensitive to rotenone treatment in 3-month-old Sprague-Dawley rats, and correlated with a significant decline in rotarod latency-to-fall measurement by approximately 2.5 fold. The measured midbrain region revealed a 31 % lower TH signal when compared to control (p < 0.01 by t test, n = 5). Bivariate analysis of integrated TH counts versus rotarod latency-to-fall indicates a positive slope and modest but significant correlation of R2 = 0.68 (p < 0.05, n = 10). These results indicate this rapid, instrument-based quantification method by IR detection successfully quantifies TH levels in rat brain tissue, while taking only 5 days from euthanasia to data output. This approach also allows for the identification of multiple targets by IHC with the simultaneous performance of downstream molecular analysis within the same animal tissue, allowing for the use of fewer animals per study.


Assuntos
Mesencéfalo/química , Mesencéfalo/enzimologia , Desempenho Psicomotor/fisiologia , Tirosina 3-Mono-Oxigenase/análise , Tirosina 3-Mono-Oxigenase/metabolismo , Animais , Inseticidas/toxicidade , Masculino , Mesencéfalo/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Rotenona/toxicidade , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Fatores de Tempo
14.
Nat Commun ; 12(1): 3495, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34108486

RESUMO

Lysosomal storage disorders characterized by altered metabolism of heparan sulfate, including Mucopolysaccharidosis (MPS) III and MPS-II, exhibit lysosomal dysfunctions leading to neurodegeneration and dementia in children. In lysosomal storage disorders, dementia is preceded by severe and therapy-resistant autistic-like symptoms of unknown cause. Using mouse and cellular models of MPS-IIIA, we discovered that autistic-like behaviours are due to increased proliferation of mesencephalic dopamine neurons originating during embryogenesis, which is not due to lysosomal dysfunction, but to altered HS function. Hyperdopaminergia and autistic-like behaviours are corrected by the dopamine D1-like receptor antagonist SCH-23390, providing a potential alternative strategy to the D2-like antagonist haloperidol that has only minimal therapeutic effects in MPS-IIIA. These findings identify embryonic dopaminergic neurodevelopmental defects due to altered function of HS leading to autistic-like behaviours in MPS-II and MPS-IIIA and support evidence showing that altered HS-related gene function is causative of autism.


Assuntos
Transtorno do Espectro Autista/metabolismo , Dopamina/metabolismo , Heparitina Sulfato/metabolismo , Doenças por Armazenamento dos Lisossomos/metabolismo , Animais , Transtorno do Espectro Autista/tratamento farmacológico , Transtorno do Espectro Autista/patologia , Benzazepinas/uso terapêutico , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Antagonistas de Dopamina/uso terapêutico , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Heparitina Sulfato/farmacologia , Doenças por Armazenamento dos Lisossomos/tratamento farmacológico , Doenças por Armazenamento dos Lisossomos/patologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/embriologia , Mesencéfalo/patologia , Camundongos , Mucopolissacaridose III/tratamento farmacológico , Mucopolissacaridose III/metabolismo , Mucopolissacaridose III/patologia , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D1/metabolismo
15.
Epilepsia ; 62(7): 1689-1700, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33997963

RESUMO

OBJECTIVE: Fetal exposure to the anticonvulsant drug valproic acid (VPA), used to treat certain types of epilepsy, increases the risk for birth defects, including neural tube defects, as well as learning difficulties and behavioral problems. Here, we investigated neurotoxic effects of VPA exposure using zebrafish as a model organism. The capacity of folic acid (FA) supplementation to rescue the VPA-induced neuronal and behavioral perturbations was also examined. METHODS: Zebrafish embryos of different transgenic lines with neuronal green fluorescent protein expression were exposed to increasing concentrations of VPA with or without FA supplementation. Fluorescence microscopy was used to visualize alterations in brain structures and neural progenitor cells, as well as motor neurons and neurite sprouting. A twitching behavioral assay was used to examine the functional consequences of VPA and FA treatment. RESULTS: In zebrafish embryos, VPA exposure caused a decrease in the midbrain size, an increase in the midline gap of the hindbrain, and perturbed neurite sprouting of secondary motor neurons, in a concentration-dependent manner. VPA exposure also decreased the fluorescence intensity of neuronal progenitor cells in early developmental stages, indicating fewer cells. Furthermore, VPA exposure significantly altered embryonic twitching activity, causing hyperactivity in dark and hypoactivity in light. Supplementation of FA rescued the VPA-induced smaller midbrain size and hindbrain midline gap defects. FA treatment also increased the number of neuronal progenitor cells in VPA-treated embryos and salvaged neurite sprouting of the secondary motor neurons. FA rescued the VPA-induced alterations in twitching activity in light but not in dark. SIGNIFICANCE: We conclude that VPA exposure induces specific neurotoxic perturbations in developing zebrafish embryos, and that FA reversed most of the identified defects. The results demonstrate that zebrafish is a promising model to study VPA-induced teratogenesis and to screen for countermeasures.


Assuntos
Anticonvulsivantes/toxicidade , Comportamento Animal/efeitos dos fármacos , Ácido Fólico/uso terapêutico , Síndromes Neurotóxicas/prevenção & controle , Síndromes Neurotóxicas/psicologia , Ácido Valproico/toxicidade , Vitaminas/uso terapêutico , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Suplementos Nutricionais , Desenvolvimento Embrionário/efeitos dos fármacos , Larva , Iluminação , Mesencéfalo/anatomia & histologia , Mesencéfalo/efeitos dos fármacos , Neurônios Motores/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Defeitos do Tubo Neural/induzido quimicamente , Neuritos/efeitos dos fármacos , Rombencéfalo/anatomia & histologia , Rombencéfalo/efeitos dos fármacos , Ácido Valproico/antagonistas & inibidores
16.
Eur J Pharmacol ; 903: 174112, 2021 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-33901458

RESUMO

In the present study, we investigated the anti-Parkinson's effect of vanillic acid (VA) (12 mg/kg, 25 mg/kg, 50 mg/kg p.o.) against rotenone (2 mg/kg s.c.) induced Parkinson's disease (PD) in rats. The continuous administration of rotenone for 35 days resulted in rigidity in muscles, catalepsy, and decrease in locomotor activity, body weight, and rearing behaviour along with the generation of oxidative stress in the brain (rise in the TBARS, and SAG level and reduced CAT, and GSH levels). Co-treatment of VA and levodopa-carbidopa (100 mg/kg + 25 mg/kg p.o.) lead to a significant (P < 0.001) reduction in the muscle rigidity and catalepsy along with a significant (P < 0.001) increase in body weight, rearing behaviour, locomotion and muscle activity as compared to the rotenone-treated group in the dose dependent manner, showing maximum effect at the 50 mg/kg. It also showed reversal of levels of oxidative stress parameters thus, reducing the neuronal oxidative stress. The level of DA was also estimated which showed an increase in the level of DA in the VA plus standard drug treated animals as compared to rotenone treated group. Histopathological evaluation showed a high number of eosinophilic lesions in the rotenone group which were found to be very less in the VA co-treated group. The study thus proved that co-treatment of VA and levodopa-carbidopa, significantly protected the brain from neuronal damage due to oxidative stress and attenuated the motor defects indicating the possible therapeutic potential of VA as a neuroprotective in PD.


Assuntos
Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Ácido Vanílico/farmacologia , Ácido Vanílico/uso terapêutico , Animais , Antiparkinsonianos/farmacologia , Antiparkinsonianos/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Carbidopa/farmacologia , Carbidopa/uso terapêutico , Catalase/metabolismo , Catalepsia/induzido quimicamente , Catalepsia/tratamento farmacológico , Modelos Animais de Doenças , Dopamina/metabolismo , Combinação de Medicamentos , Feminino , Glutationa/metabolismo , Levodopa/farmacologia , Levodopa/uso terapêutico , Locomoção/efeitos dos fármacos , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Mesencéfalo/patologia , Doenças Musculares/induzido quimicamente , Doenças Musculares/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Doença de Parkinson/etiologia , Equilíbrio Postural/efeitos dos fármacos , Ratos Sprague-Dawley , Rotenona/toxicidade , Superóxidos/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
17.
Food Chem Toxicol ; 152: 112164, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33819549

RESUMO

Some epidemiological studies with different levels of evidence have pointed to a higher risk of Parkinson's disease (PD) after exposure to environmental toxicants. A practically unexplored potential etiological factor is a group of naturally-occurring fungal secondary metabolites called mycotoxins. The mycotoxin ochratoxin A (OTA) has been reported to be neurotoxic in mice. To further identify if OTA exposure could have a role in PD pathology, Balb/c mice were orally treated with OTA (0.21, 0.5 mg/kg bw) four weeks and left for six months under normal diet. Effects of OTA on the onset, progression of alpha-synuclein pathology and development of motor deficits were evaluated. Immunohistochemical and biochemical analyses showed that oral subchronic OTA treatment induced loss of striatal dopaminergic innervation and dopaminergic cell dysfunction responsible for motor impairments. Phosphorylated alpha-synuclein levels were increased in gut and brain. LAMP-2A protein was decreased in tissues showing alpha-synuclein pathology. Cell cultures exposed to OTA exhibited decreased LAMP-2A protein, impairment of chaperone-mediated autophagy and decreased alpha-synuclein turnover which was linked to miRNAs deregulation, all reminiscent of PD. These results support the hypothesis that oral exposure to low OTA doses in mice can lead to biochemical and pathological changes reported in PD.


Assuntos
Micotoxinas/toxicidade , Ocratoxinas/toxicidade , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Administração Oral , Animais , Neurônios Dopaminérgicos/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Mesencéfalo/patologia , Camundongos Endogâmicos BALB C , MicroRNAs/metabolismo , Micotoxinas/administração & dosagem , Ocratoxinas/administração & dosagem , Doença de Parkinson/patologia , Parte Compacta da Substância Negra/efeitos dos fármacos , Parte Compacta da Substância Negra/metabolismo , Parte Compacta da Substância Negra/patologia , Fosforilação/efeitos dos fármacos , Fatores de Tempo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo
18.
Mol Neurobiol ; 58(7): 3405-3416, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33713017

RESUMO

Parkinson's disease is a neurodegenerative disorder characterised by nigrostriatal dopaminergic degeneration, and intracellular α-synuclein aggregation. Current pharmacological treatments are solely symptomatic so there is a need to identify agents that can slow or stop dopaminergic degeneration. One proposed class of therapeutics are neurotrophic factors which promote the survival of nigrostriatal dopaminergic neurons. However, neurotrophic factors need to be delivered directly to the brain. An alternative approach may be to identify pharmacological agents which can reach the brain to stimulate neurotrophic factor expression and/or their signalling pathways in dopaminergic neurons. BMP2 is a neurotrophic factor that is expressed in the human substantia nigra; exogenous BMP2 administration protects against dopaminergic degeneration in in vitro models of PD. In this study, we investigated the neurotrophic potential of two FDA-approved drugs, quinacrine and niclosamide, that are modulators of BMP2 signalling. We report that quinacrine and niclosamide, like BMP2, significantly increased neurite length, as a readout of neurotrophic action, in SH-SY5Y cells and dopaminergic neurons in primary cultures of rat ventral mesencephalon. We also show that these effects of quinacrine and niclosamide require the activation of BMP-Smad signalling. Finally, we demonstrate that quinacrine and niclosamide are neuroprotective against degeneration induced by the neurotoxins, MPP+ and 6-OHDA, and by viral-mediated overexpression of α-synuclein in vitro. Collectively, this study identifies two drugs, that are safe for use in patients' to 'are approved for human use, that exert neurotrophic effects on dopaminergic neurons through modulation of BMP-Smad signalling. This rationalises the further study of drugs that target the BMP-Smad pathway as potential neuroprotective pharmacotherapy for Parkinson's disease.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Niclosamida/farmacologia , Quinacrina/farmacologia , alfa-Sinucleína/toxicidade , Animais , Proteína Morfogenética Óssea 2/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Neurônios Dopaminérgicos/metabolismo , Relação Dose-Resposta a Droga , Humanos , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Degeneração Neural/induzido quimicamente , Degeneração Neural/metabolismo , Degeneração Neural/prevenção & controle , Neuritos/metabolismo , Neuroproteção/fisiologia , Neurotoxinas/toxicidade , Niclosamida/uso terapêutico , Quinacrina/uso terapêutico , Ratos , Proteínas Smad/metabolismo
19.
Int Immunopharmacol ; 94: 107431, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33578261

RESUMO

Parkinson's disease (PD) is a disabling progressive neurodegenerative disease. So far, PD's treatment remains symptomatic with no curative effects. Aside from its blatant analgesic and antipyretic efficacy, recent studies highlighted the endowed neuroprotective potentials of paracetamol (PCM). To this end: the present study investigated: (1) Possible protective role of PCM against rotenone-induced PD-like neurotoxicity in rats, and (2) the mechanisms underlying its neuroprotective actions including cannabinoid receptors' modulation. A dose-response study was conducted using three doses of PCM (25, 50, and 100 mg/kg/day, i.p.) and their effects on body weight changes, spontaneous locomotor activity, rotarod test, tyrosine hydroxylase (TH) and α-synuclein expression, and striatal dopamine (DA) content were evaluated. Results revealed that PCM (100 mg/kg/day, i.p.) halted PD motor impairment, prevented rotenone-induced weight loss, restored normal histological tissue structure, reversed rotenone-induced reduction in TH expression and striatal DA content, and markedly decreased midbrain and striatal α-synuclein expression in rotenone-treated rats. Accordingly, PCM (100 mg/kg/day, i.p.) was selected for further mechanistic investigations, where it ameliorated rotenone-induced oxidative stress, neuro-inflammation, apoptosis, and disturbed cannabinoid receptors' expression. In conclusion, our findings imply a multi-target neuroprotective effect of PCM in PD which could be attributed to its antioxidant, anti-inflammatory and anti-apoptotic activities, in addition to cannabinoid receptors' modulation.


Assuntos
Acetaminofen/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Transtornos Parkinsonianos/tratamento farmacológico , Acetaminofen/farmacologia , Animais , Apoptose/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Dopamina/metabolismo , Endocanabinoides , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/patologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/metabolismo , Ratos Wistar , Receptor CB1 de Canabinoide/genética , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/genética , Receptor CB2 de Canabinoide/metabolismo , Rotenona , alfa-Sinucleína/metabolismo
20.
Physiol Rep ; 9(2): e14709, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33484235

RESUMO

Midbrain dopamine neurons communicate signals of reward anticipation and attribution of salience. This capacity is distorted in heroin or cocaine abuse or in conditions such as human mania. A shared characteristic among rodent models of these behavioral disorders is that dopamine neurons in these animals acquired a small size and manifest an augmented spontaneous and burst activity. The biophysical mechanism underlying this increased excitation is currently unknown, but is believed to primarily follow from a substantial drop in K+ conductance secondary to morphology reduction. This work uses a dopamine neuron mathematical model to show, surprisingly, that under size diminution a reduction in K+ conductance is an adaptation that attempts to decrease cell excitability. The homeostatic response that preserves the intrinsic activity is the conservation of the ion channel density for each conductance; a result that is analytically demonstrated and challenges the experimentalist tendency to reduce intrinsic excitation to K+ conductance expression level. Another unexpected mechanism that buffers the raise in intrinsic activity is the presence of the ether-a-go-go-related gen K+ channel since its activation is illustrated to increase with size reduction. Computational experiments finally demonstrate that size attenuation results in the paradoxical enhancement of afferent-driven bursting as a reduced temporal summation indexed correlates with improved depolarization. This work illustrates, on the whole, that experimentation in the absence of mathematical models may lead to the erroneous interpretation of the counterintuitive aspects of empirical data.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/patologia , Modelos Neurológicos , Morfina/toxicidade , Potenciais de Ação , Animais , Tamanho Celular/efeitos dos fármacos , Biologia Computacional/métodos , Neurônios Dopaminérgicos/metabolismo , Fenômenos Eletrofisiológicos , Homeostase , Mesencéfalo/metabolismo , Camundongos , Entorpecentes/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA