Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Neurobiol Dis ; 193: 106452, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38401650

RESUMO

A common adverse effect of Parkinson's disease (PD) treatment is L-dopa-induced dyskinesia (LID). This condition results from both dopamine (DA)-dependent and DA-independent mechanisms, as glutamate inputs from corticostriatal projection neurons impact DA-responsive medium spiny neurons in the striatum to cause the dyskinetic behaviors. In this study, we explored whether suppression of presynaptic corticostriatal glutamate inputs might affect the behavioral and biochemical outcomes associated with LID. We first established an animal model in which 6-hydroxydopamine (6-OHDA)-lesioned mice were treated daily with L-dopa (10 mg/kg, i.p.) for 2 weeks; these mice developed stereotypical abnormal involuntary movements (AIMs). When the mice were pretreated with the NMDA antagonist, amantadine, we observed suppression of AIMs and reductions of phosphorylated ERK1/2 and NR2B in the striatum. We then took an optogenetic approach to manipulate glutamatergic activity. Slc17a6 (vGluT2)-Cre mice were injected with pAAV5-Ef1a-DIO-eNpHR3.0-mCherry and received optic fiber implants in either the M1 motor cortex or dorsolateral striatum. Optogenetic inactivation at either optic fiber implant location could successfully reduce the intensity of AIMs after 6-OHDA lesioning and L-dopa treatment. Both optical manipulation strategies also suppressed phospho-ERK1/2 and phospho-NR2B signals in the striatum. Finally, we performed intrastriatal injections of LDN 212320 in the dyskenesic mice to enhance expression of glutamate uptake transporter GLT-1. Sixteen hours after the LDN 212320 treatment, L-dopa-induced AIMs were reduced along with the levels of striatal phospho-ERK1/2 and phospho-NR2B. Together, our results affirm a critical role of corticostriatal glutamate neurons in LID and strongly suggest that diminishing synaptic glutamate, either by suppression of neuronal activity or by upregulation of GLT-1, could be an effective approach for managing LID.


Assuntos
Discinesias , Doença de Parkinson , Ratos , Camundongos , Animais , Levodopa/farmacologia , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Oxidopamina/toxicidade , Ácido Glutâmico/metabolismo , Ratos Sprague-Dawley , Dopamina/metabolismo , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Antiparkinsonianos/efeitos adversos
2.
Hum Mol Genet ; 29(14): 2408-2419, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32588892

RESUMO

Cyclin-dependent kinase-like 5 (CDKL5), a serine-threonine kinase encoded by an X-linked gene, is highly expressed in the mammalian forebrain. Mutations in this gene cause CDKL5 deficiency disorder, a neurodevelopmental encephalopathy characterized by early-onset seizures, motor dysfunction, and intellectual disability. We previously found that mice lacking CDKL5 exhibit hyperlocomotion and increased impulsivity, resembling the core symptoms in attention-deficit hyperactivity disorder (ADHD). Here, we report the potential neural mechanisms and treatment for hyperlocomotion induced by CDKL5 deficiency. Our results showed that loss of CDKL5 decreases the proportion of phosphorylated dopamine transporter (DAT) in the rostral striatum, leading to increased levels of extracellular dopamine and hyperlocomotion. Administration of methylphenidate (MPH), a DAT inhibitor clinically effective to improve symptoms in ADHD, significantly alleviated the hyperlocomotion phenotype in Cdkl5 null mice. In addition, the improved behavioral effects of MPH were accompanied by a region-specific restoration of phosphorylated dopamine- and cAMP-regulated phosphoprotein Mr 32 kDa, a key signaling protein for striatal motor output. Finally, mice carrying a Cdkl5 deletion selectively in DAT-expressing dopaminergic neurons, but not dopamine receptive neurons, recapitulated the hyperlocomotion phenotype found in Cdkl5 null mice. Our findings suggest that CDKL5 is essential to control locomotor behavior by regulating region-specific dopamine content and phosphorylation of dopamine signaling proteins in the striatum. The direct, as well as indirect, target proteins regulated by CDKL5 may play a key role in movement control and the therapeutic development for hyperactivity disorders.


Assuntos
Síndromes Epilépticas/genética , Hipercinese/genética , Proteínas Serina-Treonina Quinases/genética , Espasmos Infantis/genética , Animais , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Síndromes Epilépticas/patologia , Transtornos Neurológicos da Marcha/genética , Transtornos Neurológicos da Marcha/metabolismo , Transtornos Neurológicos da Marcha/patologia , Humanos , Hipercinese/metabolismo , Hipercinese/patologia , Metilfenidato/metabolismo , Camundongos , Camundongos Knockout , Espasmos Infantis/patologia
3.
Int J Mol Sci ; 22(14)2021 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-34299230

RESUMO

The precise neural mechanisms underlying the pathogenesis of depression are largely unknown, though stress-induced brain inflammation and serotonergic plasticity are thought to be centrally involved. Moreover, we previously demonstrated that neuropeptide FF receptor 2 (NPFFR2) overexpression provokes depressive-like behaviors in mice. Here, we assess whether NPFFR2 is involved in priming of depressive-like behaviors and downregulation of serotonergic 1A receptor (5HT1AR) after lipopolysaccharide (LPS) treatment. The forced swimming test (FST) and sucrose preference test (SPT) were used to quantify depressive-like phenotypes in wild-type (WT) and NPFFR2-knockout (KO) mice. A single dose of LPS (i.p. 1 mg/kg) readily caused increases in toll-like receptor 4 and tumor necrosis factor-α along with decreases in 5-HT1AR mRNA in the ventral hippocampus of WT mice. Furthermore, LPS treatment of WT mice increased immobility time in FST and decreased sucrose preference in SPT. In contrast, none of these effects were observed in NPFFR2-KO mice. While WT mice injected with lentiviral 5-HT1AR shRNA in the ventral hippocampus displayed an unaltered response after LPS challenge, LPS-challenged NPFFR2-KO mice displayed a profound decrease in sucrose preference when pretreated with 5-HT1AR shRNA. Taken together, these results suggest that NPFFR2 modulates LPS-induced depressive-like behavioral phenotypes by downregulating 5HT1AR in the ventral hippocampus.


Assuntos
Depressão/genética , Receptor 5-HT1A de Serotonina/metabolismo , Receptores de Neuropeptídeos/genética , Animais , Comportamento Animal/fisiologia , Depressão/metabolismo , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Lipopolissacarídeos/efeitos adversos , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Receptor 5-HT1A de Serotonina/genética , Receptores de Neuropeptídeos/metabolismo , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
4.
J Biomed Sci ; 27(1): 16, 2020 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-31900153

RESUMO

BACKGROUND: Over-stimulation of dopamine signaling is thought to underlie the pathophysiology of a list of mental disorders, such as psychosis, mania and attention-deficit/hyperactivity disorder. These disorders are frequently associated with cognitive deficits in attention or learning and memory, suggesting that persistent activation of dopamine signaling may change neural plasticity to induce cognitive or emotional malfunction. METHODS: Dopamine transporter knockdown (DAT-KD) mice were used to mimic a hyper-dopamine state. Novel object recognition (NOR) task was performed to assess the recognition memory. To test the role of dopamine D3 receptor (D3R) on NOR, DAT-KD mice were treated with either a D3R antagonist, FAUC365 or by deletion of D3R. Total or phospho-GSK3 and -ERK1/2 signals in various brain regions were measured by Western blot analyses. To examine the impact of GSK3 signal on NOR, wild-type mice were systemically treated with GSK3 inhibitor SB216763 or, micro-injected with lentiviral shRNA of GSK3ß or GSK3α in the medial prefrontal cortex (mPFC). RESULTS: We confirmed our previous findings that DAT-KD mice displayed a deficit in NOR memory, which could be prevented by deletion of D3R or exposure to FAUC365. In WT mice, p-GSK3α and p-GSK3ß were significantly decreased in the mPFC after exposure to novel objects; however, the DAT-KD mice exhibited no such change in mPFC p-GSK3α/ß levels. DAT-KD mice treated with FAUC365 or with D3R deletion exhibited restored novelty-induced GSK3 dephosphorylation in the mPFC. Moreover, inhibition of GSK3 in WT mice diminished NOR performance and impaired recognition memory. Lentiviral shRNA knockdown of GSK3ß, but not GSK3α, in the mPFC of WT mice also impaired NOR. CONCLUSION: These findings suggest that D3R acts via GSK3ß signaling in the mPFC to play a functional role in NOR memory. In addition, treatment with D3R antagonists may be a reasonable approach for ameliorating cognitive impairments or episodic memory deficits in bipolar disorder patients.


Assuntos
Transtorno Bipolar/genética , Disfunção Cognitiva/genética , Glicogênio Sintase Quinase 3 beta/genética , Quinase 3 da Glicogênio Sintase/genética , Receptores de Dopamina D3/genética , Animais , Transtorno Bipolar/patologia , Disfunção Cognitiva/patologia , Dopamina/genética , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases/genética , Memória Episódica , Camundongos , Córtex Pré-Frontal/metabolismo
5.
Int J Mol Sci ; 21(3)2020 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-32041361

RESUMO

In this study, we explored the release characteristics of analgesics, namely levobupivacaine, lidocaine, and acemetacin, from electrosprayed poly(D,L-lactide-co-glycolide) (PLGA) microparticles. The drug-loaded particles were prepared using electrospraying techniques and evaluated for their morphology, drug release kinetics, and pain relief activity. The morphology of the produced microparticles elucidated by scanning electron microscopy revealed that the optimal parameters for electrospraying were 9 kV, 1 mL/h, and 10 cm for voltage, flow rate, and travel distance, respectively. Fourier-transform infrared spectrometry indicated that the analgesics had been successfully incorporated into the PLGA microparticles. The analgesic-loaded microparticles possessed low toxicity against human fibroblasts and were able to sustainably elute levobupivacaine, lidocaine, and acemetacin in vitro. Furthermore, electrosprayed microparticles were found to release high levels of lidocaine and acemetacin (well over the minimum therapeutic concentrations) and levobupivacaine at the fracture site of rats for more than 28 days and 12 days, respectively. Analgesic-loaded microparticles demonstrated their effectiveness and sustained performance for pain relief in fracture injuries.


Assuntos
Analgésicos/administração & dosagem , Fraturas do Fêmur/complicações , Indometacina/análogos & derivados , Levobupivacaína/administração & dosagem , Lidocaína/administração & dosagem , Dor/tratamento farmacológico , Células 3T3 , Analgésicos/química , Analgésicos/farmacologia , Animais , Preparações de Ação Retardada , Modelos Animais de Doenças , Composição de Medicamentos , Fraturas Ósseas , Indometacina/administração & dosagem , Indometacina/química , Indometacina/farmacologia , Levobupivacaína/química , Levobupivacaína/farmacologia , Lidocaína/química , Lidocaína/farmacologia , Camundongos , Microtecnologia , Estrutura Molecular , Dor/etiologia , Tamanho da Partícula , Ratos , Espectrometria de Massas por Ionização por Electrospray
6.
J Headache Pain ; 21(1): 140, 2020 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-33287697

RESUMO

An amendment to this paper has been published and can be accessed via the original article.

7.
J Headache Pain ; 21(1): 87, 2020 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-32640973

RESUMO

BACKGROUND: Stimulation of trigeminovascular pathway is widely used to establish the headache animal model. Headache is a common neurological disorder, in which symptomatic attacks are mediated by calcitonin-gene-related peptide (CGRP). CGRP is synthesized and released from the trigeminal ganglion to transmit pain signals under stimulation. On the other hand, Neuropeptide FF (NPFF) is a candidate transmitter/modulator for migraine, and stimulation of its receptor, NPFFR2, increases the expression and release of CGRP in mice sensory neurons. Here, we investigate the impact of NPFFR2 on trigeminal CGRP level in a capsaicin-induced headache mouse model. METHODS: Mice were intracisternally injected with capsaicin into the cisterna magna to activate the trigeminovascular pathway and induce headache symptoms. Mice pretreated with Npffr2-shRNA or NPFFR2 knockouts were adopted to test the impact of NPFFR2 on capsaicin-induced CGRP upregulation in trigeminal ganglion. The gene silencing effect of Npffr2-shRNA in trigeminal ganglion was confirmed by real-time PCR. Trigeminal CGRP level was determined by immunofluorescence staining, and the percentage of CGRP-positive cell was calculated after setting the signal intensity threshold by Image J software. Amount of trigeminal CGRP in NPFFR2 overexpressed mice was also measured by CGRP ELISA. FINDINGS: Infusion of capsaicin into the cisterna magna upregulated the CGRP in trigeminal ganglion and induced spontaneous pain behaviors including the reduction of locomotor activity and the increase of freezing behavior. Intracisternal injection of Npffr2-shRNA reduced the mRNA of Npffr2 in trigeminal ganglion. Mice pretreatment with Npffr2-shRNA prevented capsaicin-induced CGRP upregulation in trigeminal ganglion. Similarly, CGRP upregulation was also reduced in NPFFR2 knockout mice. On the contrary, trigeminal CGRP was increased in NPFFR2 overexpressed mice. CONCLUSIONS: Reducing the level of NPFFR2 leads to the downregulation of capsaicin-induced CGRP in trigeminal ganglion, which would consequently attenuate the activation of trigeminovascular pathway. Thus, NPFFR2 could serve as a potential target for neuromodulation of cephalic pain.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Capsaicina/farmacologia , Receptores de Neuropeptídeos/metabolismo , Gânglio Trigeminal/metabolismo , Animais , Calcitonina/genética , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos de Enxaqueca/metabolismo , Neurônios Aferentes/metabolismo , Ratos , Regulação para Cima/efeitos dos fármacos
8.
Biochem Biophys Res Commun ; 517(1): 77-83, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31327496

RESUMO

Intra-neuronal α-synuclein (αSNCA) aggregation are the leading cause of dopaminergic neuron degeneration in Parkinson's disease (PD). Most PD patients is linked with αSNCA gene mutations. Gene therapy shows therapeutic potential by packing gene into viral vectors to improve gene expression through stereotactic brain injections. However, through intracranial injection, the gene expression is typically limited with tissue distribution tightly adjacent to the injection track, when expressing therapeutic genes for a wider CNS region is preferable. We use microbubble-facilitated ultrasound pulsations (MB-USP) as a new gene delivering tool to enhance the limit gene delivery of local injection in brain and evaluate the feasibility using αSNCA as model gene. We demonstrate that MB-USP can transfect naked constructs DNA of αSNCA gene into two types of neuron cells and enhance the gene expression. We confirm α-synuclein fusion protein functionality, showing that α-synuclein fusion protein significantly reduce the mitochondrial activity. We show MB-USP improves in vivo gene transfer in the brain with naked construct local injection, significantly enhances α-synuclein expression level to 1.68-fold, and broaden its distribution to 25-fold. In vivo fused α-synuclein protein aggregation is also found in gene-injected mice brains by MB-USP. MB-USP provides an alternative to α-synuclein over expression in vitro and in vivo model for investigation of α-synuclein related PD therapeutic strategies.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos/genética , Doença de Parkinson/genética , alfa-Sinucleína/genética , Animais , Linhagem Celular , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbolhas , Doença de Parkinson/terapia , Ondas Ultrassônicas , Regulação para Cima
9.
Chin J Physiol ; 62(2): 47-52, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31243174

RESUMO

Neuropeptide FF (NPFF) is known as a morphine-modulating peptide and was first isolated in 1985. It has been characterized as an RF-amide peptide. The traditional role of NPFF is mediation of the pain response, and it displays both anti-opioid and pro-opioid actions through central nervous system. In the recent decade, additional evidence has revealed some untraditional features of NPFF, such as regulation of the neuroendocrine system, energy homeostasis, anti-inflammation, pain transmission, and peripheral modulation of adipose tissue macrophages. Neuropeptide FF receptor 2 (NPFFR2) is a physiological receptor of NPFF, and the actions of NPFF may occur through downstream NPFFR2 signaling. NPFF and NPFFR2 increase the neuronal activity in various areas of the hypothalamus to modulate the hypothalamic-pituitary-adrenal axis, the autonomic nervous system, food intake, and energy balance. These underlying cellular mechanisms have been explored in the past few years. Here, we review the impact of NPFF and related RF-amide peptides on hypothalamic function. The interaction of NPFF with NPFFR2 in the hypothalamus is emphasized, and NPFF-NPFFR2 system may represent an important therapeutic target in hypothalamic-related disorders in the future.


Assuntos
Homeostase , Sistema Hipotálamo-Hipofisário , Hipotálamo , Sistemas Neurossecretores , Oligopeptídeos , Sistema Hipófise-Suprarrenal , Receptores de Neuropeptídeos
10.
Int J Mol Sci ; 18(8)2017 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-28825666

RESUMO

Neuropeptide FF (NPFF) belongs to the RFamide family and is known as a morphine-modulating peptide. NPFF regulates various hypothalamic functions through two receptors, NPFFR1 and NPFFR2. The hypothalamic-pituitary-adrenal (HPA) axis participates in physiological stress response by increasing circulating glucocorticoid levels and modulating emotional responses. Other RFamide peptides, including neuropeptide AF, neuropeptide SF and RFamide related peptide also target NPFFR1 or NPFFR2, and have been reported to activate the HPA axis and induce anxiety- or depression-like behaviors. However, little is known about the action of NPFF on HPA axis activity and anxiety-like behaviors, and the role of the individual receptors remains unclear. In this study, NPFFR2 agonists were used to examine the role of NPFFR2 in activating the HPA axis in rodents. Administration of NPFFR2 agonists, dNPA (intracerebroventricular, ICV) and AC-263093 (intraperitoneal, IP), time-dependently (in rats) and dose-dependently (in mice) increased serum corticosteroid levels and the effects were counteracted by the NPFF receptor antagonist, RF9 (ICV), as well as corticotropin-releasing factor (CRF) antagonist, α-helical CRF(9-41) (intravenous, IV). Treatment with NPFFR2 agonist (AC-263093, IP) increased c-Fos protein expression in the hypothalamic paraventricular nucleus and induced an anxiogenic effect, which was evaluated in mice using an elevated plus maze. These findings reveal, for the first time, that the direct action of hypothalamic NPFFR2 stimulates the HPA axis and triggers anxiety-like behaviors.


Assuntos
Transtorno Depressivo/metabolismo , Oligopeptídeos/metabolismo , Receptores de Neuropeptídeos/metabolismo , Corticosteroides/sangue , Animais , Ansiedade , Hormônio Liberador da Corticotropina/administração & dosagem , Hormônio Liberador da Corticotropina/antagonistas & inibidores , Transtorno Depressivo/sangue , Transtorno Depressivo/fisiopatologia , Hidrazinas/administração & dosagem , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiopatologia , Hipotálamo/metabolismo , Hipotálamo/patologia , Camundongos , Oligopeptídeos/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Ratos , Receptores de Neuropeptídeos/agonistas
11.
Learn Mem ; 22(3): 149-58, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25691515

RESUMO

The molecular mechanisms underlying drug extinction remain largely unknown, although a role for medial prefrontal cortex (mPFC) glutamate neurons has been suggested. Considering that the mPFC sends glutamate efferents to the ventral tegmental area (VTA), we tested whether the VTA is involved in methamphetamine (METH) extinction via conditioned place preference (CPP). Among various METH-CPP stages, we found that the amount of phospho-GluR1/Ser845 increased in the VTA at behavioral extinction, but not the acquisition or withdrawal stage. Via surface biotinylation, we found that levels of membrane GluR1 were significantly increased during METH-CPP extinction, while no change was observed at the acquisition stage. Specifically, the number of dendritic spines in the VTA was increased at behavioral extinction, but not during acquisition. To validate the role of the mPFC in METH-CPP extinction, we lesioned the mPFC. Ibotenic acid lesioning of the mPFC did not affect METH-CPP acquisition, however, it abolished the extinction stage and reversed the enhanced phospho-GluR1/Ser845 levels as well as increases in VTA dendritic spines during METH-CPP extinction. Overall, this study demonstrates that the mPFC plays a critical role in METH-CPP extinction and identifies the VTA as an alternative target in mediating the extinction of drug conditioning.


Assuntos
Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Metanfetamina/farmacologia , Córtex Pré-Frontal/fisiologia , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiologia , Animais , Condicionamento Psicológico/efeitos dos fármacos , Comportamento de Procura de Droga/efeitos dos fármacos , Comportamento de Procura de Droga/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/efeitos dos fármacos , Fosforilação , Receptores de AMPA/metabolismo , Área Tegmentar Ventral/ultraestrutura
12.
Clin Nutr ; 43(3): 603-619, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38301284

RESUMO

BACKGROUND: The hypothalamus is a crucial brain region that mediates the effects of insulin and leptin signals on peripheral metabolic functions. Previous research has shown that insulin signals in the hypothalamus act via multiple neuronal circuits and anabolic/catabolic pathways that converge on the vagus nerve and sympathetic fibers to coordinate energy metabolism in peripheral organs. Additionally, neuropeptide FF (NPFF) has been identified as a regulator of feeding behaviors and energy homeostasis in the hypothalamus, but the mechanisms underlying its involvement in metabolic control remain unclear. This study aims to explore the underlying mechanisms of NPFF in modulating metabolic disorders. METHODS: In this study, we investigated the physiological role of NPFF in insulin-related energy homeostasis and metabolic health. First, we evaluated the effects of NPFF and its receptors on central insulin signaling using mouse hypothalamic cell lines and Npffr2-overexpressing mice. To further explore the effects of NPFFR2 on insulin-related metabolic disorders, such as diabetes mellitus, we used Npffr2-deleted mice in combination with the streptozotocin (STZ)-induced type 1 diabetes and high-fat diet/STZ-induced type 2 diabetic mouse models. The impacts of central NPFFR2 were demonstrated specifically through Npffr2 overexpression in the hypothalamic arcuate nucleus, which subsequently induced type 2 diabetes. RESULTS: We found that stimulating NPFFR2 in the hypothalamus blocked hypothalamic insulin activity. Npffr2 deletion improved central and peripheral metabolic symptoms in both mouse models of diabetes mellitus, exerting effects on central and systemic insulin resistance, feeding behaviors, glucose and insulin intolerance, lipid metabolism, liver steatosis, and inflammation of white adipose tissues. The overexpression of ARC Npffr2 augmented the metabolic dysregulation in the mouse model of type 2 diabetes. CONCLUSIONS: Our findings demonstrate that hypothalamic NPFFR2 negatively regulates insulin signaling in the central nervous system and plays an important role in maintaining systemic metabolic health, thereby providing valuable insights for potential clinical interventions targeting these health challenges.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Animais , Camundongos , Insulina , Diabetes Mellitus Tipo 2/genética , Hipotálamo , Homeostase , Modelos Animais de Doenças
13.
Glia ; 61(4): 475-89, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23322492

RESUMO

The neurotransmitter dopamine acts on the subventricular zone (SVZ) to regulate both prenatal and postnatal neurogenesis, in particular through D(3) receptor (D(3) R) subtype. In this study, we explored the cellular mechanism(s) underlying D(3) R-mediated cell proliferation and tested if systemic delivery of a D(3) R agonist would induce SVZ multipotent neural stem/precursor cell (NSC/NPC) proliferation in vivo. We found that treatment with the D(3) R agonist, 7-OH-DPAT, enhances cell proliferation in a dose-dependent manner in cultured SVZ neurospheres from wild-type, but not D(3) R knock-out mice. Furthermore, D(3) R activation also stimulates S-phase and enhances mRNA and protein levels of cyclin D1 in wild-type neurospheres, a process which requires cellular Akt and ERK1/2 signaling. Moreover, chronic treatment with low dose 7-OH-DAPT in vivo increases BrdU(+) cell numbers in the adult SVZ, but this effect was not seen in D(3) R KO mice. Additionally, we probed the cell type specificity of D(3) R agonist-mediated cell proliferation. We found that in adult SVZ, GFAP(+) astrocytes, type-B GFAP(+) /nestin(+) and type-C EGF receptor (EGFR(+) )/nestin(+) cells express D(3) R mRNA, but type-A Doublecortin (Dcx)(+) neuroblasts do not. Using flow cytometry and immunofluorescence, we demonstrated that D(3) R activation increases GFAP(+) type-B and EGFR(+) type-C cell numbers, and the newly divided Dcx(+) type-A cells. However, BrdU(+) /Dcx(+) cell numbers were decreased in D(3) R KO mice compared to wildtype, suggesting that D(3) R maintains constitutive NSC/NPCs population in the adult SVZ. Overall, we demonstrate that D(3) R activation induces NSC/NPC proliferation through Akt and ERK1/2 signaling and increases the numbers of type-B and -C NSC/NPCs in the adult SVZ.


Assuntos
Ventrículos Laterais/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Neuroglia/metabolismo , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores de Dopamina D3/metabolismo , Fatores Etários , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Corpo Estriado/citologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Proteína Duplacortina , Ventrículos Laterais/citologia , Ventrículos Laterais/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Neuroglia/efeitos dos fármacos , Receptores de Dopamina D3/agonistas , Receptores de Dopamina D3/deficiência , Tetra-Hidronaftalenos/farmacologia
14.
J Neurochem ; 127(2): 163-76, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23952963

RESUMO

Previously, we found that chronic methamphetamine treatment altered cannabinoid type 1 receptor (CB1R)-dependent cAMP/PKA/dopamine and cAMP-regulated phosphoprotein of Mr 32,000 (DARPP-32)/T34/PP2B signaling and decreased levels of CB1R protein and mRNA in the nucleus accumbens. These findings suggested the existence of signaling interplay between mesolimbic dopamine and CB1R. In this study, we further investigate interactions between CB1R and dopamine D2 receptor (D2R) signaling. Activation of either CB1R or D2R increased extracellular signal-regulated kinases 1 and 2 (ERK1/2) phosphorylation, while co-stimulation of CB1R and D2 R evoked an additive effect on the phospho-ERK1/2 signal. This effect was mediated through a pertussis toxin-sensitive Gαi/o pathway in primary striatal cells. Furthermore, the mRNA level of CB1R was increased via dopamine D2 receptor short form (D(2S)R) by treatment with D2R agonist quinpirole in D(2S)R/C6 glioma cells. This effect could be suppressed by co-treatment with the ERK1/2 inhibitor U0126. To test if D(2S)R could transcriptionally regulate CB1R, the 5'-untranslated region (5'-UTR) of the cannabinoid receptor 1 (CNR1) gene was sequenced from rat brain. Results showed that the CNR1 gene includes two exons, which contain 375 bp of 5'-UTR and are separated by a 17-kb intron. A luciferase reporter assay showed that the maximal D(2S)R-responsive promoter activity is located in the -1 to -222 region of CNR1 promoter. Overall, we demonstrate previously unidentified crosstalk between D2R and CB1R via ERK1/2 signaling that enhances the expression of CB1R by modulating its promoter activity.


Assuntos
Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Receptor Cross-Talk/efeitos dos fármacos , Receptor CB1 de Canabinoide/efeitos dos fármacos , Receptores de Dopamina D2/efeitos dos fármacos , Animais , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Cicloexanóis/farmacologia , DNA/biossíntese , DNA/isolamento & purificação , Feminino , Genes Reporter/efeitos dos fármacos , Glioma/metabolismo , Dados de Sequência Molecular , Neostriado/citologia , Neostriado/metabolismo , Plasmídeos/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA/biossíntese , RNA/genética , RNA/isolamento & purificação , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/biossíntese , Receptor CB1 de Canabinoide/genética , Transfecção
15.
J Neuroinflammation ; 9: 189, 2012 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-22870919

RESUMO

BACKGROUND: Inflammation or nerve injury-induced upregulation and release of chemokine CC chemokine ligand 2 (CCL2) within the dorsal root ganglion (DRG) is believed to enhance the activity of DRG nociceptive neurons and cause hyperalgesia. Transient receptor potential vanilloid receptor 1 (TRPV1) and tetrodotoxin (TTX)-resistant Na(v)1.8 sodium channels play an essential role in regulating the excitability and pain transmission of DRG nociceptive neurons. We therefore tested the hypothesis that CCL2 causes peripheral sensitization of nociceptive DRG neurons by upregulating the function and expression of TRPV1 and Nav1.8 channels. METHODS: DRG neuronal culture was prepared from 3-week-old Sprague-Dawley rats and incubated with various concentrations of CCL2 for 24 to 36 hours. Whole-cell voltage-clamp recordings were performed to record TRPV1 agonist capsaicin-evoked inward currents or TTX-insensitive Na(+) currents from control or CCL2-treated small DRG sensory neurons. The CCL2 effect on the mRNA expression of TRPV1 or Na(v)1.8 was measured by real-time quantitative RT-PCR assay. RESULTS: Pretreatment of CCL2 for 24 to 36 hours dose-dependently (EC(50) value = 0.6 ± 0.05 nM) increased the density of capsaicin-induced currents in small putative DRG nociceptive neurons. TRPV1 mRNA expression was greatly upregulated in DRG neurons preincubated with 5 nM CCL2. Pretreating small DRG sensory neurons with CCL2 also increased the density of TTX-resistant Na(+) currents with a concentration-dependent manner (EC(50) value = 0.7 ± 0.06 nM). The Na(v)1.8 mRNA level was significantly increased in DRG neurons pretreated with CCL2. In contrast, CCL2 preincubation failed to affect the mRNA level of TTX-resistant Nav1.9. In the presence of the specific phosphatidylinositol-3 kinase (PI3K) inhibitor LY294002 or Akt inhibitor IV, CCL2 pretreatment failed to increase the current density of capsaicin-evoked inward currents or TTX-insensitive Na(+) currents and the mRNA level of TRPV1 or Na(v)1.8. CONCLUSIONS: Our results showed that CCL2 increased the function and mRNA level of TRPV1 channels and Na(v)1.8 sodium channels in small DRG sensory neurons via activating the PI3K/Akt signaling pathway. These findings suggest that following tissue inflammation or peripheral nerve injury, upregulation and release of CCL2 within the DRG could facilitate pain transmission mediated by nociceptive DRG neurons and could induce hyperalgesia by upregulating the expression and function of TRPV1 and Na(v)1.8 channels in DRG nociceptive neurons.


Assuntos
Quimiocina CCL2/fisiologia , Gânglios Espinais/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.8/biossíntese , Neurônios/metabolismo , Canais de Cátion TRPV/biossíntese , Regulação para Cima/genética , Potenciais de Ação/genética , Animais , Células Cultivadas , Gânglios Espinais/citologia , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Neurônios/citologia , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPV/genética
16.
Cell Commun Signal ; 10(1): 35, 2012 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-23176293

RESUMO

BACKGROUND: Matrix metalloproteinase-9 (MMP-9) plays a crucial role in pathological processes of brain inflammation, injury, and neurodegeneration. Moreover, bradykinin (BK) induces the expression of several inflammatory proteins in brain astrocytes. Recent studies have suggested that increased oxidative stress is implicated in the brain inflammation and injury. However, whether BK induced MMP-9 expression mediated through oxidative stress remains virtually unknown. Herein we investigated the role of redox signals in BK-induced MMP-9 expression in rat brain astrocytes (RBA-1 cells). RESULTS: In the study, we first demonstrated that reactive oxygen species (ROS) plays a crucial role in BK-induced MMP-9 expression in cultured brain astrocytes (in vitro) and animal brain tissue (in vivo) models. Next, BK-induced MMP-9 expression is mediated through a Ca2+-mediated PKC-α linking to p47phox/NADPH oxidase 2 (Nox2)/ROS signaling pathway. Nox2-dependent ROS generation led to activation and up-regulation of the downstream transcriptional factor AP-1 (i.e. c-Fos and c-Jun), which bound to MMP-9 promoter region, and thereby turned on transcription of MMP-9 gene. Functionally, BK-induced MMP-9 expression enhanced astrocytic migration. CONCLUSIONS: These results demonstrated that in RBA-1 cells, activation of AP-1 (c-Fos/c-Jun) by the PKC-α-mediated Nox2/ROS signals is essential for up-regulation of MMP-9 and cell migration enhanced by BK.

17.
Biomed J ; 45(6): 914-922, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-34974168

RESUMO

BACKGROUND: Parkinson's disease (PD) is one of the most prevalent neurodegenerative disorders, which characterized by increased pathological marker protein, α-synuclein (α-syn) and phosphorylated-Ser129-α-syn in the extracellular fluids. Current methods of measuring the p-Ser129-α-syn concentration in cerebrospinal fluid for PD are based on ELISA method, however, the amount of area under the curve (AUC) to predict PD is around 0.7-0.8. Higher confidence level of AUC in p-Ser129-α-syn quantification for the early diagnosis of PD would be essential. METHODS: Detection of p-Ser129-α-syn in diluted human serum for diagnosis of PD was investigated by a modified paired surface plasma wave biosensor (PSPWB) using a quarter wave plate for better detection performance. The method combining an immunoassay and non-labeled technique measures the p-Ser129-α-syn level with high sensitivity and specificity. Ten patients with PD at early stage (Hohn & Yahr stage I and II) and 11 age-matched healthy control participants were recruited for measurement of serum p-Ser129-α-syn. RESULTS: AUC of the p-Ser129-α-syn in diluted human serum was 0.92 and it shows that p-Ser129-α-syn in diluted human serum could be used as a sensitive biomarker for the diagnosis of PD in clinics. Results clearly show that the measured p-Ser129-α-syn concentration in diluted human serum displays a statistical significance between health control subjects and PD patients. CONCLUSIONS: P-Ser129-α-syn has low abundance in human serum, high detection sensitivity and specificity are critical to the success of the diagnosis of PD in clinics. In this study, a modified PSPWB was developed that the limit of detection at 1 ng/mL for p-Ser129-α-syn (standard) spiked into diluted human serum of a healthy control was performed. This result shows that the modified PSPWB can be used as a platform for detecting p-Ser129-α-syn in diluted human serum as a potential biomarker for PD.


Assuntos
Doenças Neurodegenerativas , Doença de Parkinson , Humanos , Doença de Parkinson/diagnóstico , Doença de Parkinson/metabolismo , alfa-Sinucleína , Fosforilação , Biomarcadores
18.
J Neuroinflammation ; 8: 126, 2011 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-21958434

RESUMO

BACKGROUND: During inflammation, immune cells accumulate in damaged areas and release pro-inflammatory cytokines and neurotrophins. Brain-derived neurotrophic factor (BDNF) plays a neuromodulatory role in spinal cord dorsal horn via the post-synaptic tyrosine protein kinase B (trkB) receptor to facilitate pain transmission. However, the precise role of BDNF and trkB receptor in the primary sensory neurons of dorsal root ganglia (DRG) during inflammation remains to be clarified. The aim of this study was to investigate whether and how BDNF-trkB signaling in the DRG is involved in the process of inflammatory pain. METHODS: We used complete Freund's adjuvant- (CFA-) induced and tumor necrosis factor-α- (TNF-α-) induced inflammation in rat hindpaw as animal models of inflammatory pain. Quantification of protein and/or mRNA levels of pain mediators was performed in separate lumbar L3-L5 DRGs. The cellular mechanism of TNF-α-induced BDNF and/or trkB receptor expression was examined in primary DRG cultures collected from pooled L1-L6 DRGs. Calcitonin gene-related peptide (CGRP), BDNF and substance P release were also evaluated by enzyme immunoassay. RESULTS: CFA injection into rat hindpaw resulted in mechanical hyperalgesia and significant increases in levels of TNF-α in the inflamed tissues, along with enhancement of BDNF and trkB receptor as well as the pain mediators CGRP and transient receptor potential vanilloid receptor subtype 1 (TRPV1) in DRG. Direct injection of TNF-α into rat hindpaw resulted in similar effects with retrograde transport of TNF-α along the saphenous nerve to DRG during CFA-induced inflammation. Primary DRG cultures chronically treated with TNF-α showed significant enhancement of mRNA and protein levels of BDNF and trkB receptor, BDNF release and trkB-induced phospho-ERK1/2 signal. Moreover, CGRP and substance P release were enhanced in DRG cultures after chronic TNF-α treatment or acute BDNF stimulation. In addition, we found that BDNF up-regulated trkB expression in DRG cultures. CONCLUSIONS: Based on our current experimental results, we conclude that inflammation and TNF-α up-regulate the BDNF-trkB system in DRG. This phenomenon suggests that up-regulation of BDNF in DRG may, in addition to its post-synaptic effect in spinal dorsal horn, act as an autocrine and/or paracrine signal to activate the pre-synaptic trkB receptor and regulate synaptic excitability in pain transmission, thereby contributing to the development of hyperalgesia.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Gânglios Espinais/fisiologia , Inflamação/imunologia , Dor/metabolismo , Receptor trkB/metabolismo , Células Receptoras Sensoriais/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Peptídeo Relacionado com Gene de Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Células Cultivadas , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Hiperalgesia/metabolismo , Inflamação/induzido quimicamente , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Dor/induzido quimicamente , Medição da Dor , Ratos , Ratos Sprague-Dawley , Receptor trkB/genética , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/efeitos dos fármacos , Substância P/metabolismo , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima
19.
Behav Brain Res ; 396: 112925, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-32971195

RESUMO

Chronic methamphetamine (METH) treatment induces behavioral sensitization in rodents. During this process, hyperactivation of the mesolimbic dopamine system plays a central role, and dopamine D2-like receptor-based antipsychotics are known to alleviate the behavioral hyperactivity. The atypical antipsychotic, clozapine (Clz), acts partially as a dopamine D4 receptor (D4R) antagonist and mitigates hyperdopaminergic drug addiction and/or comorbid psychotic symptoms; however, it remains unclear whether D4R blockade contributes to the therapeutic effects of Clz. Here, we evaluated the potential role of D4R in regulating hyperdopaminergia-induced behavioral hyperactivity in METH behavioral sensitization and dopamine transporter (DAT) knockdown (KD) mice. Clz or a D4R-selective antagonist, L-745,870, were co-administered to mice with daily METH in a METH sensitization model, and Clz or L-745,870 were administered alone in a DAT KD hyperactivity model. Locomotor activity and accumbal D4R expression were analyzed. Clz suppressed both the initiation and expression of METH behavioral sensitization, as well as DAT KD hyperactivity. However, repetitive Clz treatment induced tolerance to the suppression effect on METH sensitization initiation. In contrast, D4R inhibition by L-745,870 had no effect on METH sensitization or DAT KD hyperactivity. Accumbal D4R expression was similar between METH-sensitized mice with and without Clz co-treatment. In sum, our results suggest the mesolimbic D4R does not participate in behavioral sensitization encoded by hyperdopaminergia, a finding which likely extends to the therapeutic effects of Clz. Therefore, molecular targets other than D4R should be prioritized in the development of future therapeutics for treatment of hyperdopaminergia-dependent neuropsychiatric disorders.


Assuntos
Antipsicóticos/farmacologia , Sensibilização do Sistema Nervoso Central/efeitos dos fármacos , Clozapina/farmacologia , Dopaminérgicos/farmacologia , Locomoção/efeitos dos fármacos , Metanfetamina/farmacologia , Receptores de Dopamina D4/antagonistas & inibidores , Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Transtornos Relacionados ao Uso de Anfetaminas/fisiopatologia , Animais , Antipsicóticos/administração & dosagem , Comportamento Animal/efeitos dos fármacos , Clozapina/administração & dosagem , Modelos Animais de Doenças , Dopaminérgicos/administração & dosagem , Metanfetamina/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos Transgênicos , Transtornos Psicóticos/metabolismo , Transtornos Psicóticos/fisiopatologia , Piridinas/farmacologia , Pirróis/farmacologia
20.
Brain Res Bull ; 165: 263-271, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33049353

RESUMO

Synaptic dopamine (DA) concentrations are largely determined by the activities of presynaptic D2 and D3 autoreceptors (D2R and D3R) and DA transporter (DAT). Furthermore, the activity of DAT is regulated by phosphorylation events and protein interactions that affect its surface expression. Because DA autoreceptors and DAT coordinately maintain synaptic DA homeostasis, we hypothesized that D3R might crosstalk with DAT to fine-tune synaptic DA concentrations. To test this hypothesis, we established [3H]DA uptake and DAT surface expression assays in hD3/rDAT-double-transfected HEK-293 cells or limbic forebrain synaptosomal preparations. Ropinirole, a preferential D3R agonist, reduced [3H]DA uptake in HEK-hD3/rDAT cells in a dose-dependent manner, an effect which could be blocked by the D2R/D3R antagonist, raclopride. Furthermore, ropinirole also reduced DAT surface expression in limbic forebrain synaptosomes, and this effect could be blocked by raclopride or the internalization inhibitor, concanavalin A. To identify potential mediators of this apparent D3R-DAT crosstalk, DAT-associated proteins were co-immunoprecipitated from limbic forebrain synaptosomes after D3R activation and identified by MALDI-TOF. From this analysis, the Hsc70 chaperone was identified as a DAT-associated protein. Interestingly, ropinirole induced the association of Hsc70/Hsp70 with DAT, and the Hsc70/Hsp70 inhibitor, apoptozole, prevented the ropinirole-induced reduction of DAT surface expression. Together, these results suggest that D3R negatively regulates DAT activity by promoting the association of DAT and Hsc70/Hsp70.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Proteínas de Choque Térmico HSC70/metabolismo , Neurônios/metabolismo , Prosencéfalo/metabolismo , Receptores de Dopamina D3/metabolismo , Animais , Agonistas de Dopamina/farmacologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Células HEK293 , Proteínas de Choque Térmico HSC70/genética , Humanos , Indóis/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Receptores de Dopamina D3/agonistas , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa