RESUMO
Population aging is a global reality driven by increased life expectancy. This demographic phenomenon is intrinsically linked to the epidemic of cognitive disorders such as dementia and Alzheimer's disease, posing challenges for elderly and their families. In this context, the search for new therapeutic strategies to prevent or minimize cognitive impairments becomes urgent, as these deficits are primarily associated with oxidative damage and increased neuroinflammation. Ferulic acid (FA), a natural and potent antioxidant compound, is proposed to be nanoencapsulated to target the central nervous system effectively with lower doses and an extended duration of action. Here, we evaluated the effects of the nanoencapsulated FA on d-galactose (d-Gal)- induced memory impairments. Male Wistar adult rats were treated with ferulic acid-loaded nanocapsules (FA-Nc) or non-encapsulated ferulic acid (D-FA) for 8 weeks concurrently with d-Gal (150 mg/kg s.c.) injection. As expected, our findings showed that d-Gal injection impaired memory processes and increased anxiety behavior, whereas FA-Nc treatment ameliorated these behavioral impairments associated with the aging process induced by d-Gal. At the molecular level, nanoencapsulated ferulic acid (FA-Nc) ameliorated the decrease in ACh and glutamate induced by d-galactose (d-Gal), and also increased GABA levels in the dorsal hippocampus, indicating its therapeutic superiority. Additional studies are needed to elucidate the mechanisms underlying our current promising outcomes. Nanoscience applied to pharmacology can reduce drug dosage, thereby minimizing adverse effects while enhancing therapeutic response, particularly in neurodegenerative diseases associated with aging. Therefore, the strategy of brain-targeted drug delivery through nanoencapsulation can be effective in mitigating aging-related factors that may lead to cognitive deficits.
Assuntos
Envelhecimento , Ansiedade , Ácidos Cumáricos , Galactose , Ácido Glutâmico , Transtornos da Memória , Ratos Wistar , Ácido gama-Aminobutírico , Animais , Ácidos Cumáricos/farmacologia , Ácidos Cumáricos/uso terapêutico , Masculino , Envelhecimento/efeitos dos fármacos , Envelhecimento/metabolismo , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Ácido gama-Aminobutírico/metabolismo , Ansiedade/tratamento farmacológico , Ansiedade/metabolismo , Ácido Glutâmico/metabolismo , Ratos , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Nanoestruturas , NanocápsulasRESUMO
The master control of mammalian circadian rhythms is the suprachiasmatic nucleus (SCN), which is formed by the ventral and dorsal regions. In SCN neurons, GABA has an important function and even excitatory actions in adulthood. However, the physiological role of this neurotransmitter in the developing SCN is unknown. Here, we recorded GABAergic postsynaptic currents (in the perforated-patch configuration using gramicidin) to determine the chloride reversal potential (ECl) and also assessed the immunological expression of the Na-K-Cl cotransporter 1 (NKCC1) at early ages of the rat (postnatal days (P) 3 to 25), during the day and night, in the two SCN regions. We detected that ECl greatly varied with age and depending on the SCN region and time of day. Broadly speaking, ECl was more hyperpolarized with age, except for the oldest age studied (P20-25) in both day and night in the ventral SCN, where it was less negative. Likewise, ECl was more hyperpolarized in the dorsal SCN both during the day and at night; while ECl was more negative at night both in the ventral and the dorsal SCN. Moreover, the total NKCC1 fluorescent expression was higher during the day than at night. These results imply that NKCC1 regulates the circadian and developmental fluctuations in the [Cl-]i to fine-tune ECl, which is crucial for either excitatory or inhibitory GABAergic actions to occur in the SCN.
Assuntos
Cloretos , Ritmo Circadiano , Membro 2 da Família 12 de Carreador de Soluto , Núcleo Supraquiasmático , Animais , Núcleo Supraquiasmático/metabolismo , Ritmo Circadiano/fisiologia , Ratos , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Masculino , Cloretos/metabolismo , Ácido gama-Aminobutírico/metabolismo , Ratos Wistar , Técnicas de Patch-Clamp , Envelhecimento/fisiologiaRESUMO
In a previous study, we reported the in vitro potential probiotic and gamma-aminobutyric acid (GABA) production, of several strains from a collection of Lactiplantibacillus (Lpb) strains within the community of natural whey starters from the artisanal cheese industry. GABA is a non-protein amino acid widely distributed in nature and produced in animals, plants, and microorganisms. However, the best known role of GABA is its function as the major inhibitory neurotransmitter of the central nervous system. Preclinical and clinical evidence suggests that the GABAergic system has a relevant role in mental health disorders, such as anxiety and major depression. The modulation of the GABAergic system has been suggested as a potential strategy for treatment, one such mechanism of modulation is the influence of the microbiota-gut-brain axis through probiotic treatments. The present study was designed to investigate the in vivo probiotic potential of LPB145, a Lactiplantibacillus strain previously characterised as a GABA-producing potentially probiotic strain. Therefore, we evaluated the behavioural effects of chronic oral administration of LPB145 on rats' anxiety- and depression-like behaviours, using the elevated plus maze, open field, and the forced swimming test. The impact of LPB145 strain treatment on the gut microbiota structure and diversity was assessed to discern a possible mechanism of action of the LPB145 treatment through the microbiota-gut-brain axis. Our results showed that LPB145 administration induced an antidepressive-like behaviour without changes in locomotor activity. In contrast, the treatment did not modify the experimental anxiety. The structure and diversity of the intestinal microbiota remained unaffected by the treatment when compared to the control. However, specific clades that could be implicated in the behavioural changes did show differences in their relative abundance. These findings provide evidence regarding the potential of probiotic strains isolated from alimentary sources, to modulate the microbiota-gut-brain axis and positively impact mental health.
Assuntos
Ansiedade , Queijo , Depressão , Microbioma Gastrointestinal , Probióticos , Ácido gama-Aminobutírico , Animais , Probióticos/administração & dosagem , Probióticos/farmacologia , Ansiedade/microbiologia , Ratos , Ácido gama-Aminobutírico/metabolismo , Queijo/microbiologia , Depressão/microbiologia , Depressão/terapia , Masculino , Ratos Wistar , Comportamento Animal/efeitos dos fármacos , Modelos Animais de DoençasRESUMO
Understanding the neural, metabolic, and psychological mechanisms underlying human altruism and decision-making is a complex and important topic both for science and society. Here, we investigated whether transcranial Direct Current Stimulation (tDCS) applied to two prefrontal cortex regions, the ventromedial prefrontal cortex (vmPFC, anode) and the right dorsolateral prefrontal cortex (DLPFC, cathode) can induce changes in self-reported emotions and to modulate local metabolite concentrations. We employed in vivo quantitative MR Spectroscopy in healthy adult participants and quantified changes in GABA and Glx (glutamate + glutamine) before and after five sessions of tDCS delivered at 2 mA for 20 min (active group) and 1 min (sham group) while participants were engaged in a charitable donation task. In the active group, we observed increased levels of GABA in vmPFC. Glx levels decreased in both prefrontal regions and self-reported happiness increased significantly over time in the active group. Self-reported guiltiness in both active and sham groups tended to decrease. The results indicate that self-reported happiness can be modulated, possibly due to changes in Glx concentrations following repeated stimulation. Therefore, local changes may induce remote changes in the reward network through interactions with other metabolites, previously thought to be unreachable with noninvasive stimulation techniques.
Assuntos
Emoções , Córtex Pré-Frontal , Estimulação Transcraniana por Corrente Contínua , Ácido gama-Aminobutírico , Humanos , Masculino , Feminino , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiologia , Adulto , Emoções/fisiologia , Adulto Jovem , Ácido gama-Aminobutírico/metabolismo , Ácido Glutâmico/metabolismo , Altruísmo , Glutamina/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Córtex Pré-Frontal Dorsolateral/metabolismo , Córtex Pré-Frontal Dorsolateral/fisiologiaRESUMO
Germination is a process that enhances the content of health-promoting secondary metabolites. However, the bioaccessibility of these compounds depends on their stability and solubility throughout the gastrointestinal tract. The study aimed to explore how germination time influences the content and bioaccessibility of γ-aminobutyric acid and polyphenols and antioxidant capacity of lupin (Lupinus angustifolius L.) sprouts during simulated gastrointestinal digestion. Gamma-aminobutyric acid showed a decrease following gastrointestinal digestion (GID) whereas phenolic acids and flavonoids exhibited bioaccessibilities of up to 82.56 and 114.20%, respectively. Although the digestion process affected the profile of phenolic acids and flavonoids, certain isoflavonoids identified in 7-day sprouts (G7) showed resistance to GID. Germination not only favored antioxidant activity but also resulted in germinated samples exhibiting greater antioxidant properties than ungerminated counter parts after GID. Intestinal digests from G7 did not show cytotoxicity in RAW 264.7 macrophages, and notably, they showed an outstanding ability to inhibit the production of reactive oxygen species. This suggests potential benefit in mitigating oxidative stress. These findings contribute to understand the dynamic interplay between bioprocessing and digestion in modulating the bioaccessibility of bioactive compounds in lupin, thereby impacting health.
Assuntos
Antioxidantes , Disponibilidade Biológica , Digestão , Germinação , Lupinus , Lupinus/metabolismo , Lupinus/química , Antioxidantes/metabolismo , Germinação/efeitos dos fármacos , Camundongos , Células RAW 264.7 , Animais , Polifenóis/metabolismo , Flavonoides/análise , Flavonoides/metabolismo , Ácido gama-Aminobutírico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Hidroxibenzoatos/metabolismo , Hidroxibenzoatos/análise , Trato Gastrointestinal/metabolismoRESUMO
Gamma aminobutyric acid (GABA) is a critical inhibitory neurotransmitter in the central nervous system that plays a vital role in modulating neuronal excitability. Dysregulation of GABAergic signaling, particularly involving the cotransporters NKCC1 and KCC2, has been implicated in various pathologies, including epilepsy, schizophrenia, autism spectrum disorder, Down syndrome, and ischemia. NKCC1 facilitates chloride influx, whereas KCC2 mediates chloride efflux via potassium gradient. Altered expression and function of these cotransporters have been associated with excitotoxicity, inflammation, and cellular death in ischemic events characterized by reduced cerebral blood flow, leading to compromised tissue metabolism and subsequent cell death. NKCC1 inhibition has emerged as a potential therapeutic approach to attenuate intracellular chloride accumulation and mitigate neuronal damage during ischemic events. Similarly, targeting KCC2, which regulates chloride efflux, holds promise for improving outcomes and reducing neuronal damage under ischemic conditions. This review emphasizes the critical roles of GABA, NKCC1, and KCC2 in ischemic pathologies and their potential as therapeutic targets. Inhibiting or modulating the activity of these cotransporters represents a promising strategy for reducing neuronal damage, preventing excitotoxicity, and improving neurological outcomes following ischemic events. Furthermore, exploring the interactions between natural compounds and NKCC1/KCC2 provides additional avenues for potential therapeutic interventions for ischemic injury.
Assuntos
Isquemia Encefálica , Morte Celular , Cotransportadores de K e Cl- , Membro 2 da Família 12 de Carreador de Soluto , Simportadores , Ácido gama-Aminobutírico , Animais , Humanos , Ácido gama-Aminobutírico/metabolismo , Simportadores/metabolismo , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Morte Celular/fisiologia , Morte Celular/efeitos dos fármacos , Isquemia Encefálica/metabolismo , Isquemia Encefálica/tratamento farmacológicoRESUMO
The occurrence, inhibitory modulation, and trophic effects of GABA have been identified in the peripheral sympathetic nervous system. We have demonstrated that GABA and acetylcholine (ACh) may colocalize in the same axonal varicosities or be segregated into separate ones in the rat superior cervical ganglia (SCG). Neurotransmitter segregation varies with age and the presence of neurotrophic factors. Here, we explored age-dependent changes in the occurrence and segregation of GABA and ACh in rats ranging from 2 weeks old (wo) to 12 months old or older. Using immunohistochemistry, we characterized the expression of L-glutamic acid decarboxylase of 67 kDa (GAD67) and vesicular acetylcholine transporter (VAChT) in the rat SCG at 2, 4, 8, 12 wo and 12 months old or older. Our findings revealed that GAD67 was greater at 2 wo compared with the other ages, whereas VAChT levels were greater at 4 wo than at 12 wo and 12 months old or older. The segregation of these neurotransmitters was more pronounced at 2 and 4 wo. We observed a caudo-rostral gradient of segregation degree at 8 and 12 wo. Data point out that the occurrence and segregation of GABA and ACh exhibit developmental adaptative changes throughout the lifetime of rats. We hypothesize that during the early postnatal period, the increase in GABA and GABA-ACh segregation promotes the release of GABA alone which might play a role in trophic actions.
Assuntos
Acetilcolina , Gânglio Cervical Superior , Ratos , Animais , Acetilcolina/metabolismo , Axônios/metabolismo , Ácido gama-Aminobutírico/metabolismoRESUMO
Indole alkaloids are the main bioactive molecules of the Gelsemium genus plants. Diverse reports have shown the beneficial actions of Gelsemium alkaloids on the pathological states of the central nervous system (CNS). Nevertheless, Gelsemium alkaloids are toxic for mammals. To date, the molecular targets underlying the biological actions of Gelsemium alkaloids at the CNS remain poorly defined. Functional studies have determined that gelsemine is a modulator of glycine receptors (GlyRs) and GABAA receptors (GABAARs), which are ligand-gated ion channels of the CNS. The molecular and physicochemical determinants involved in the interactions between Gelsemium alkaloids and these channels are still undefined. We used electrophysiological recordings and bioinformatic approaches to determine the pharmacological profile and the molecular interactions between koumine, gelsemine, gelsevirine, and humantenmine and these ion channels. GlyRs composed of α1 subunits were inhibited by koumine and gelsevirine (IC50 of 31.5 ± 1.7 and 40.6 ± 8.2 µM, respectively), while humantenmine did not display any detectable activity. The examination of GlyRs composed of α2 and α3 subunits showed similar results. Likewise, GABAARs were inhibited by koumine and were insensitive to humantenmine. Further assays with chimeric and mutated GlyRs showed that the extracellular domain and residues within the orthosteric site were critical for the alkaloid effects, while the pharmacophore modeling revealed the physicochemical features of the alkaloids for the functional modulation. Our study provides novel information about the molecular determinants and functional actions of four major Gelsemium indole alkaloids on inhibitory receptors, expanding our knowledge regarding the interaction of these types of compounds with protein targets of the CNS.
Assuntos
Alcaloides , Gelsemium , Animais , Gelsemium/química , Alcaloides/química , Extratos Vegetais/química , Alcaloides Indólicos/química , Ácido gama-Aminobutírico , Mamíferos/metabolismoRESUMO
BACKGROUND: The flavonoid chrysin produces rapid and long-lasting anxiolytic- and antidepressant-like effects in rats. However, it is not known whether low and high doses of chrysin produce differential anti-immobility effects through the Gamma-Aminobutyric Acid sub-type A (GABAA) receptor. The goal of this work was therefore to compare low and high doses of chrysin for their effects on depression-like behavior in a longitudinal study. Moreover, chrysin was compared with the serotonergic fluoxetine and Gamma-Aminobutyric Acid (GABA)ergic allopregnanolone, and its involvement with the GABAA receptor after chronic treatment was also investigated. METHODS: Male Wistar rats were assigned to five groups (n = 8 each): vehicle, 1 mg/kg chrysin, 5 mg/kg chrysin, 1 mg/kg fluoxetine, and 1 mg/kg allopregnanolone. In the first experiment, treatments were injected daily and the effects on locomotor activity and the forced swim test were evaluated at 0, 1, 14, and 28 days of treatment, and 48 h after the final treatment. In the second experiment, similar groups were treated for 28 days with injection of 1 mg/kg picrotoxin to investigate the role of the GABAA receptor. Depending on the experimental design, one- and two-way analysis of variance (ANOVA) tests were used for statistical analysis, with p < 0.05 set as the criteria for significance. RESULTS: In both experiments, the treatments did not alter locomotor activity. However, low and high doses of chrysin, allopregnanolone, and fluoxetine gradually produced antidepressant-like effects in the forced swim test, and maintained this effect for 48 h post-treatment, except with low dose chrysin. Picrotoxin blocked the antidepressant-like effects produced by low dose chrysin, but did not affect those produced by high dose chrysin, allopregnanolone, or fluoxetine. CONCLUSIONS: The differential antidepressant-like effects caused by low and high doses of chrysin are time-dependent. Low dose chrysin produces a rapid antidepressant-like effect, whereas high dose chrysin produces a delayed but sustained the effect, even 48 h after withdrawal. The effect with high dose chrysin was similar to that observed with allopregnanolone and fluoxetine. The mechanism for the antidepressant-like effect of low chrysin appears to be GABAergic, whereas the effect of high dose chrysin may involve other neurotransmission and neuromodulation systems related to the serotonergic system.
Assuntos
Fluoxetina , Receptores de GABA-A , Ratos , Masculino , Animais , Fluoxetina/farmacologia , Pregnanolona , Ratos Wistar , Receptores de GABA , Picrotoxina , Estudos Longitudinais , Antidepressivos/farmacologia , Flavonoides/farmacologia , Ácido gama-AminobutíricoRESUMO
Basal electroretinogram (ERG) oscillations have shown predictive value for modifiable risk factors for type 2 diabetes. However, their origin remains unknown. Here, we seek to establish the pharmacological profile of the low delta-like (δ1) wave in the mouse because it shows light sensitivity in the form of a decreased peak frequency upon photopic exposure. Applying neuropharmacological drugs by intravitreal injection, we eliminated the δ1 wave using lidocaine or by blocking all chemical and electrical synapses. The δ1 wave was insensitive to the blockade of photoreceptor input, but was accelerated when all inhibitory or ionotropic inhibitory receptors in the retina were antagonized. The sole blockade of GABAA, GABAB, GABAC, and glycine receptors also accelerated the δ1 wave. In contrast, the gap junction blockade slowed the δ1 wave. Both GABAA receptors and gap junctions contribute to the light sensitivity of the δ1 wave. We further found that the day light-activated neuromodulators dopamine and nitric oxide donors mimicked the effect of photopic exposure on the δ1 wave. All drug effects were validated through light flash-evoked ERG responses. Our data indicate that the low δ-like intrinsic wave detected by the non-photic ERG arises from an inner retinal circuit regulated by inhibitory neurotransmission and nitric oxide/dopamine-sensitive gap junction-mediated communication.
Assuntos
Diabetes Mellitus Tipo 2 , Dopamina , Camundongos , Animais , Dopamina/farmacologia , Fotofobia , Estimulação Luminosa , Retina , Eletrorretinografia , Neurotransmissores/farmacologia , Receptores de GABA-A , Ácido gama-Aminobutírico/farmacologiaRESUMO
Gamma-aminobutyric acid (GABA) serves as a pivotal neurotransmitter implicated in the pathogenesis of stress, anxiety, sleep-related disorders, and heart rate (HR) reactions. Heart-rate variability (HRV), modulated by the sympathetic and parasympathetic branches of the autonomic nervous system (ANS), offers insights into cardiac autonomic control and cardiovascular well-being. The present study aimed to explore the impact of GABA supplementation on emotional metrics, sleep quality, and HRV in sedentary women with overweight or obesity partaking in physical exercise. A randomized, double-blind, placebo-controlled clinical trial was undertaken involving 30 sedentary women with overweight or obesity. Volunteers were assigned randomly to two groups: the intervention group receiving GABA (200 mg) once daily for a total of 90 supplementation doses, and the placebo group. Both groups engaged in physical exercise, while the supplementation regimen spanned 90 days. Assessments were conducted at three intervals: baseline (T0), midway through the study (T45), and study culmination (T90). Following 90 days of GABA supplementation, the intervention group demonstrated enhancements in habitual sleep efficiency, as indicated by reductions in Pittsburgh Sleep Quality Index (PSQI) scores. Moreover, an improved emotional response was observed, characterized by diminished negative affect. GABA supplementation yielded ameliorations in depression scores as per the Depression, Anxiety, and Stress Scale (DASS-21). Notably, an augmented HRV was noted, attributed to heightened parasympathetic autonomic nervous system predominance. GABA supplementation elicited noteworthy enhancements in heart rate variability, emotional response, depression mitigation, and sleep efficiency following a 90-day supplementation.
Assuntos
Depressão , Suplementos Nutricionais , Exercício Físico , Frequência Cardíaca , Sobrepeso , Comportamento Sedentário , Ácido gama-Aminobutírico , Humanos , Feminino , Frequência Cardíaca/efeitos dos fármacos , Método Duplo-Cego , Adulto , Sobrepeso/psicologia , Sobrepeso/terapia , Exercício Físico/fisiologia , Qualidade do Sono , Emoções , Obesidade/psicologia , Obesidade/terapia , Obesidade/fisiopatologia , Pessoa de Meia-Idade , Sistema Nervoso Autônomo/efeitos dos fármacos , Sistema Nervoso Autônomo/fisiopatologia , Sono/fisiologia , Sono/efeitos dos fármacosRESUMO
Network dynamics are crucial for action and sensation. Changes in synaptic physiology lead to the reorganization of local microcircuits. Consequently, the functional state of the network impacts the output signal depending on the firing patterns of its units. Networks exhibit steady states in which neurons show various activities, producing many networks with diverse properties. Transitions between network states determine the output signal generated and its functional results. The temporal dynamics of excitation/inhibition allow a shift between states in an operational network. Therefore, a process capable of modulating the dynamics of excitation/inhibition may be functionally important. This process is known as disinhibition. In this review, we describe the effect of GABA levels and GABAB receptors on tonic inhibition, which causes changes (due to disinhibition) in network dynamics, leading to synchronous functional oscillations.
Assuntos
Fenômenos Fisiológicos do Sistema Nervoso , Receptores de GABA-B , Receptores de GABA-B/metabolismo , Neurônios/metabolismo , Inibição Neural/fisiologia , Ácido gama-Aminobutírico , Receptores de GABA-A , Antagonistas GABAérgicosRESUMO
Dopamine D2 receptor (D2R) is expressed in striatopallidal neurons and decreases forskolin-stimulated cyclic adenine monophosphate (cAMP) accumulation and gamma-aminobutyric acid (GABA) release. Dopamine D3 receptor (D3R) mRNA is expressed in a population of striatal D2R-expressing neurons. Also, D3R protein and binding have been reported in the neuropil of globus pallidus. We explore whether D2R and D3R colocalize in striatopallidal terminals and whether D3R modulates the D2R effect on forskolin-stimulated [3H]cAMP accumulation in pallidal synaptosomes and high K+ stimulated-[3H]GABA release in pallidal slices. Previous reports in heterologous systems indicate that calmodulin (CaM) and CaMKII modulate D2R and D3R functions; thus, we study whether this system regulates its functional interaction. D2R immunoprecipitates with CaM, and pretreatment with ophiobolin A or depolarization of synaptosomes with 15 mM of K+ decreases it. Both treatments increase the D2R inhibition of forskolin-stimulated [3H]cAMP accumulation when activated with quinpirole, indicating a negative modulation of CaM on D2R function. Quinpirole also activates D3R, potentiating D2R inhibition of cAMP accumulation in the ophiobolin A-treated synaptosomes. D2R and D3R immunoprecipitate in pallidal synaptosomes and decrease after the kainic acid striatal lesion, indicating the striatal origin of the presynaptic receptors. CaM-kinase II alfa (CaMKIIα) immunoprecipitates with D3R and increases after high K+ depolarization. In the presence of KN62, a CaMKIIα blocker, D3R potentiates D2R effects on cAMP accumulation in depolarized synaptosomes and GABA release in pallidal slices, indicating D3R function regulation by CaMKIIα. Our data indicate that D3R potentiates the D2R effect on cAMP accumulation and GABA release at pallidal terminals, an interaction regulated by the CaM-CaMKIIα system.
Assuntos
Calmodulina , Receptores de Dopamina D3 , Sesterterpenos , Receptores de Dopamina D3/metabolismo , Quimpirol/farmacologia , Calmodulina/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Colforsina , Receptores de Dopamina D2/metabolismo , Ácido gama-Aminobutírico/metabolismoRESUMO
Masticatory myositis is an autoimmune neuromuscular disorder that affects the muscles of mastication. The affected individual has difficulties in opening or closing the mouth, pain, and swelling in the acute phase, and significant atrophy of the affected musculature in the chronic phase. A guinea pig (Cavia porcellus) with a history of chronic hyporexia, recurrent cheek teeth overgrowth, and loss of facial silhouette, was suspected of having masticatory myositis. The disease was confirmed by computed tomography and histopathological examination of muscle fragments. The patient was treated with a protocol involving corticosteroids and gabapentin, and occlusal correction procedures.
Assuntos
Músculos da Mastigação , Miosite , Animais , Cobaias , Miosite/veterinária , Miosite/diagnóstico , Miosite/etiologia , Músculos da Mastigação/patologia , Gabapentina/uso terapêutico , Masculino , Ácidos Cicloexanocarboxílicos/uso terapêutico , Aminas/uso terapêutico , Doenças dos Roedores/diagnóstico , Doenças dos Roedores/patologia , Ácido gama-Aminobutírico/uso terapêutico , Tomografia Computadorizada por Raios X/veterinária , Corticosteroides/uso terapêutico , FemininoRESUMO
Axons are equipped with the digital signaling capacity by which they generate and faithfully propagate action potentials (APs), and also with the analogue signaling capacity by which subthreshold activity in dendrites and soma is transmitted down the axon. Despite intense work, the extent and physiological role for subthreshold synaptic activity reaching the presynaptic boutons has remained elusive because of the technical limitation to record from them. To address this issue, we made simultaneous patch-clamp recordings from the presynaptic varicosities of cerebellar GABAergic interneurons together with their parent soma or postsynaptic target cells in young rat slices and/or primary cultures. Our tour-de-force direct functional dissection indicates that the somatodendritic spontaneous excitatory synaptic potentials are transmitted down the axon for significant distances, depolarizing presynaptic boutons. These analogously transmitted excitatory synaptic potentials augment presynaptic Ca++ influx upon arrival of an immediately following AP through a mechanism that involves a voltage-dependent priming of the Ca++ channels, leading to an increase in GABA release, without any modification in the presynaptic AP waveform or residual Ca++. Our work highlights the role of the axon in synaptic integration.
Assuntos
Axônios , Terminações Pré-Sinápticas , Ratos , Animais , Axônios/fisiologia , Terminações Pré-Sinápticas/fisiologia , Cerebelo/fisiologia , Potenciais de Ação/fisiologia , Interneurônios/fisiologia , Ácido gama-Aminobutírico , Transmissão Sináptica/fisiologiaRESUMO
This cross-sectional study analyzes spectroscopy data for long-term, never-medicated patients with schizophrenia to examine their levels of γ-aminobutyric acid (GABA) compared with those of healthy controls.
Assuntos
Esquizofrenia , Humanos , Esquizofrenia/diagnóstico , Espectroscopia de Ressonância Magnética , Ácido Glutâmico , Ácido gama-Aminobutírico , Córtex Pré-FrontalRESUMO
Besides its function as a local mediator of the immune response, histamine can play a role as a neurotransmitter and neuromodulator. Histamine actions are classically mediated through four different G protein-coupled receptor subtypes but non-classical actions were also described, including effects on many ligand-gated ion channels. Previous evidence indicated that histamine acts as a positive modulator on diverse GABAA receptor subtypes, such as GABAAα1ß2γ2, GABAAα2ß3γ2, GABAAα3ß3γ2, GABAAα4ß3γ2 and GABAAα5ß3γ2. Meanwhile, its effects on GABAAρ1 receptors, known to stand for tonic currents in retinal neurons, had not been examined before. The effects of histamine on the function of human homomeric GABAAρ1 receptors were studied here, using heterologous expression in Xenopus laevis oocytes followed by the electrophysiological recording of GABA-evoked Cl- currents. Histamine inhibited GABAAρ1 receptor-mediated responses. Effects were reversible, independent of the membrane potential, and strongly dependent on both histamine and GABA concentration. A rightward parallel shift in the concentration-response curve for GABA was observed in the presence of histamine, without substantial change in the maximal response or the Hill coefficient. Results were compatible with a competitive antagonism of histamine on the GABAAρ1 receptors. This is the first report of inhibitory actions exerted by histamine on an ionotropic GABA receptor.
Assuntos
Histamina , Receptores de GABA-A , Humanos , Animais , Receptores de GABA-A/metabolismo , Histamina/farmacologia , Histamina/metabolismo , Receptores de GABA , Fenômenos Eletrofisiológicos , Ácido gama-Aminobutírico/farmacologia , Ácido gama-Aminobutírico/metabolismo , Xenopus laevis/metabolismo , Oócitos/metabolismoRESUMO
Benzodiazepines are among the most prescribed drug class worldwide to treat disorders such as anxiety, insomnia, muscle spasticity, and convulsive disorders, and to induce presurgical sedation. Although benzodiazepines exhibit a high therapeutic index and low toxicity in short-term treatments, prolonged administration induces tolerance to most of their therapeutic actions. The mechanism of this tolerance remains unclear. The central actions of benzodiazepines are mediated by binding to GABAA receptors, which mediate most fast inhibitory transmission in the brain. The majority of GABAA receptors are composed of two α-(1-6), two ß-(1-3) and one γ-subunits (1-3). In a previous report, we demonstrated that the prolonged exposure of cerebrocortical neurons to diazepam produces a transcriptional repression of the GABAA receptor α1 subunit gene via a mechanism dependent on the activation of L-type voltage-gated calcium channels (L-VGCCs). The results reported here confirm that the diazepam-induced downregulation of the α1 subunit is contingent upon calcium influx from extracellular space. In addition, this regulatory mechanism involves the activation of protein kinase A (PKA) and is accompanied by the activation of two transcription factors, the cAMP-response element-binding protein (CREB) and the inducible cAMP early repressor (ICER). Together, our results suggest that diazepam s activation of an L-VGCC/Ca2+/PKA/CREB-ICER signaling pathway is responsible for the regulation of GABAA receptors. This elucidation of the intracellular signaling cascade activated by a prolonged benzodiazepine exposure, itself potentially involved in the development of tolerance, may contribute to locating molecular targets for future therapeutic interventions.
Assuntos
Diazepam , Receptores de GABA-A , Diazepam/farmacologia , Receptores de GABA-A/metabolismo , Regulação para Baixo , Benzodiazepinas/farmacologia , Transdução de Sinais , Canais de Cálcio/genética , Ácido gama-Aminobutírico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismoRESUMO
BACKGROUND: The tremor mutant mice present motor impairments comprised of whole-body tremors, ataxia, decreased exploratory behavior, and audiogenic seizures. OBJECTIVES: This study aims to investigate the development of motor dysfunction in this mutant mouse and the relationships with cortical, striatal, and cerebellar levels of GABA, glutamate, glycine, dopamine (DA), serotonin (5-HT), noradrenaline (NOR), and its metabolites. The serum cytokines levels, myelin content, and the astrocytic expression of the glial fibrillary acidic protein (GFAP) investigated the possible influence of inflammation in motor dysfunction. RESULTS: Relative to wild-type (WT) mice, the tremor mice presented: increased tremors and bradykinesia associated with postural instability, decreased range of motion, and difficulty in initiating voluntary movements directly proportional to age; reduced step length for right and left hindlimbs; reduced cortical GABA, glutamate and, aspartate levels, the DOPAC/DA and ratio and increased the NOR levels; in the striatum, the levels of glycine and aspartate were reduced while the HVA levels, the HVA/DA and 5HIAA/5-HT ratios increased; in the cerebellum the glycine, NOR and 5-HIAA levels increased. CONCLUSIONS: We suggest that the motor disturbances resulted mainly from the activation of the indirect striatal inhibitory pathway to the frontal cortex mediated by GABA, glutamate, and aspartate, reducing the dopaminergic activity at the prefrontal cortex, which was associated with the progressive tremor. The reduced striatal and increased cerebellar glycine levels could be partially responsible for the mutant tremor motor disturbances.
Assuntos
Transtornos Motores , Tremor , Camundongos , Animais , Tremor/metabolismo , Serotonina/metabolismo , Ácido Aspártico/metabolismo , Convulsões/metabolismo , Dopamina/metabolismo , Ácido Glutâmico/metabolismo , Corpo Estriado/metabolismo , Norepinefrina/metabolismo , Neurotransmissores/metabolismo , Ácido gama-Aminobutírico/metabolismo , Glicina/metabolismoRESUMO
Nicotinic receptors are present in the retina of different vertebrates, and in the chick retina, it is present during early development throughout to post-hatching. These receptors are activated by nicotine, an alkaloid with addictive and neurotransmitter release modulation properties, such as GABA signaling. Here we evaluated the mechanisms of nicotine signaling in the avian retina during the development of neuron-glia cells at a stage where synapses are peaking. Nicotine almost halved [3H]-GABA uptake, reducing it by 45% whilst increasing more than two-fold [3H]-GABA release in E12 embryonic chick retinas. Additionally, nicotine mediated a 33% increase in [3H]-D-aspartate release. MK-801 50 µM blocked 66% of nicotine-induced [3H]-GABA release and Gö 6983 100 nM prevented the nicotine-induced reduction in [3H]-GABA uptake by rescuing 40% of this neurotransmitter uptake, implicating NMDAR and PKC (respectively) in the nicotinic responses. In addition, NO-711 prevented [3H]-GABA uptake and release induced by nicotine. Furthermore, the relevance of calcium influx for PKC activation was evidenced through fura-2 imaging. We conclude that the shift of GABA transport mediated by nicotine promotes GABA release by inducing transporter reversal via nicotine-induced EAA release through EAATs, or by a direct effect of nicotine in activating nicotinic receptors permeable to calcium and promoting PKC pathway activation and shifting GAT-1 activity, both prompting calcium influx, and activation of the PKC pathway and shifting GAT-1 activity.