Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.122
Filtrar
1.
Int Rev Neurobiol ; 160: 223-250, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34696874

RESUMO

Adolescence is a critical period for brain development and behavioral maturation, marked by increased risk-taking behavior and the initiation of drug use. There are significant changes in gray matter volume and pruning of synapses along with a shift in excitatory to inhibitory balance which marks the maturation of cognition and decision-making. Because of ongoing brain development, adolescents are particularly sensitive to the detrimental effects of drugs, including alcohol, which can cause long-lasting consequences into adulthood. The extended amygdala is a region critically implicated in withdrawal and negative affect such as anxiety and depression. As negative affective disorders develop during adolescence, the effects of adolescent alcohol exposure on extended amygdala circuitry needs further inquiry. Here we aim to provide a framework to discuss the existing literature on the extended amygdala, the neuroadaptations which result from alcohol use, and the intersection of factors which contribute to the long-lasting effects of this exposure.


Assuntos
Tonsila do Cerebelo , Etanol , Glutamatos , Transdução de Sinais , Adolescente , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiopatologia , Etanol/toxicidade , Glutamatos/efeitos dos fármacos , Glutamatos/fisiologia , Humanos , Transdução de Sinais/efeitos dos fármacos
2.
CNS Neurosci Ther ; 27(11): 1374-1384, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34402181

RESUMO

AIMS: Delayed neurocognitive recovery (dNCR) is a common postoperative complication in geriatric surgical patients for which there is no efficacious therapy. Cholecystokinin octapeptide (CCK-8), an immunomodulatory peptide, regulates memory and learning. Here, we explored the effects and mechanism of action of CCK-8 on dNCR. METHODS: We applied laparotomy to establish a model of dNCR in aged mice. Morris water maze and fear conditioning tests were used to evaluate cognition. Immunofluorescence was used to detect the density of CCK-8, A1 reactive astrocytes, glutamatergic synapses, and activation of microglia in the hippocampus. Quantitative PCR was performed to determine mRNA levels of synapse-associated factors. A1 reactive astrocytes, activated microglia, and glutamatergic synapse-associated protein levels in the hippocampus were assessed by western blotting. RESULTS: Administration of CCK-8 suppressed the activation of microglia, the induction of A1 reactive astrocytes, and the expression of tumor necrosis factor alpha, complement 1q, and interleukin 1 alpha in the hippocampus. Furthermore, it promoted glutamatergic synaptogenesis and neurocognitive recovery in aged dNCR model mice. CONCLUSION: Our findings indicated that CCK-8 alleviated cognitive impairment and promoted glutamatergic synaptogenesis by inhibiting the induction of A1 reactive astrocytes and the activation of microglia. CCK-8 is, therefore, a potential therapeutic target for dNCR.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/psicologia , Glutamatos/fisiologia , Neurogênese/efeitos dos fármacos , Complicações Pós-Operatórias/prevenção & controle , Complicações Pós-Operatórias/psicologia , Sincalida/uso terapêutico , Animais , Astrócitos/efeitos dos fármacos , Disfunção Cognitiva/etiologia , Complemento C1q/metabolismo , Medo/psicologia , Feminino , Interleucina-1/metabolismo , Laparotomia , Ativação de Macrófagos , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Sinapses , Fator de Necrose Tumoral alfa/metabolismo
3.
Cereb Cortex ; 31(10): 4554-4575, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34013343

RESUMO

Temporal specification of the neural progenitors (NPs) producing excitatory glutamatergic neurons is essential for histogenesis of the cerebral cortex. Neuroepithelial cells, the primary NPs, transit to radial glia (RG). To coincide with the transition, NPs start to differentiate into neurons, undergoing a switch from symmetric to asymmetric cell division. After the onset of neurogenesis, NPs produce layer-specific neurons in a defined order with precise timing. Here, we show that GABAA receptors (GABAARs) and taurine are involved in this regulatory mechanism. Foetal exposure to GABAAR-antagonists suppressed the transition to RG, switch to asymmetric division, and differentiation into upper-layer neurons. Foetal exposure to GABAAR-agonists caused the opposite effects. Mammalian foetuses are dependent on taurine derived from the mothers. GABA and taurine function as endogenous ligands for GABAARs. Ca2+ imaging showed that NPs principally responded to taurine but not GABA before E13. The histological phenotypes of the taurine transporter knockout mice resembled those of the mice foetally exposed to GABAAR-antagonists. Foetal exposure to GABAAR-modulators resulted in considerable alterations in offspring behavior like core symptoms of autism. These results show that taurine regulates the temporal specification of NPs and that disrupting the taurine-receptor interaction possibly leads to neurodevelopmental disorders.


Assuntos
Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/fisiologia , Glutamatos/fisiologia , Células-Tronco Neurais/fisiologia , Receptores de GABA-A/fisiologia , Taurina/fisiologia , Animais , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/fisiopatologia , Córtex Cerebral/citologia , Feminino , Feto , Antagonistas GABAérgicos/farmacologia , Moduladores GABAérgicos/farmacologia , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Técnicas de Patch-Clamp , Placenta/metabolismo , Gravidez
4.
J Neurosci Res ; 99(6): 1689-1703, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33713502

RESUMO

The lateral hypothalamus (LH) plays a key role in the maintenance of cortical activation and wakefulness. In the LH, the two main neuronal cell populations consist of excitatory glutamatergic neurons and inhibitory GABAergic neurons. Recent studies have shown that inhibitory LH GABAergic neurons are wake-promoting. However, the mechanism by which excitatory LH glutamatergic neurons contribute to sleep-wake regulation remains unclear. Using fiber photometry in male mice, we demonstrated that LH glutamatergic neurons exhibited high activities during both wakefulness and rapid eye movement sleep. Chemogenetic activation of LH glutamatergic neurons induced an increase in wakefulness that lasted for 6 hr, whereas suppression of LH glutamatergic neuronal activity caused a reduction in wakefulness. Brief optogenetic activation of LH glutamatergic neurons induced an immediate transition from slow-wave sleep to wakefulness, and long-lasting optogenetic stimulation of these neurons maintained wakefulness. Moreover, we found that LH-locus coeruleus/parabrachial nucleus and LH-basal forebrain projections mediated the wake-promoting effects of LH glutamatergic neurons. Taken together, our data indicate that LH glutamatergic neurons are essential for the induction and maintenance of wakefulness. The results presented here may advance our understanding of the role of LH in the control of wakefulness.


Assuntos
Glutamatos/fisiologia , Região Hipotalâmica Lateral/fisiologia , Neurônios/fisiologia , Vigília/fisiologia , Animais , Agonistas de Aminoácidos Excitatórios/farmacologia , Neurônios GABAérgicos , Masculino , Camundongos , Optogenética , Polissonografia , Fases do Sono , Sono REM/fisiologia
5.
J Neuroendocrinol ; 33(3): e12945, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33713519

RESUMO

Neurones in the arcuate nucleus co-expressing kisspeptin, neurokinin B (NKB) and dynorphin (KNDy) play a critical role in the control of gonadotrophin-releasing hormone (GnRH) and luteinising hormone (LH) secretion. In sheep, KNDy neurones mediate both steroid-negative- and -positive-feedback during pulsatile and preovulatory surge secretions of GnRH/LH, respectively. In addition, KNDy neurones receive glutamatergic inputs expressing vGlut2, a glutamate transporter that serves as a marker for those terminals, from both KNDy neurones and other populations of glutamatergic neurones. Previous work reported higher numbers of vGlut2-positive axonal inputs onto KNDy neurones during the LH surge than in luteal phase ewes. In the present study, we further examined the effects of the ovarian steroids progesterone (P) and oestradiol (E2 ) on glutamatergic inputs to KNDy neurones. Ovariectomised (OVX) ewes received either no further treatment (OVX) or steroid treatments that mimicked the luteal phase (low E2  + P), and early (low E2 ) or late follicular (high E2 ) phases of the oestrous cycle (n = 4 or 5 per group). Brain sections were processed for triple-label immunofluorescent detection of NKB/vGlut2/synaptophysin and analysed using confocal microscopy. We found higher numbers of vGlut2 inputs onto KNDy neurones in high E2 compared to the other three treatment groups. These results suggest that synaptic plasticity of glutamatergic inputs onto KNDy neurones during the ovine follicular phase depend on increasing levels of E2 required for the preovulatory GnRH/surge. These synaptic changes likely contribute to the positive-feedback action of oestrogen on GnRH/LH secretion and thus the generation of the preovulatory surge in the sheep.


Assuntos
Dinorfinas/fisiologia , Estradiol/fisiologia , Fase Folicular/fisiologia , Glutamatos/fisiologia , Kisspeptinas/fisiologia , Neurocinina B/fisiologia , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Animais , Estradiol/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/sangue , Fase Luteal/efeitos dos fármacos , Hormônio Luteinizante/sangue , Ovariectomia , Ovinos , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
6.
Transl Res ; 234: 31-42, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33567346

RESUMO

Nociception and opioid antinociception in females are pliable processes, varying qualitatively and quantitatively over the reproductive cycle. Spinal estrogenic signaling via membrane estrogen receptors (mERs), in combination with multiple other signaling molecules [spinal dynorphin, kappa-opioid receptors (KOR), glutamate and metabotropic glutamate receptor 1 (mGluR1)], appears to function as a master coordinator, parsing functionality between pronociception and antinociception. This provides a window into pharmacologically accessing intrinsic opioid analgesic/anti-allodynic systems. In diestrus, membrane estrogen receptor alpha (mERα) signals via mGluR1 to suppress spinal endomorphin 2 (EM2) analgesia. Strikingly, in the absence of exogenous opioids, interfering with this suppression in a chronic pain model elicits opioid anti-allodynia, revealing contributions of endogenous opioid(s). In proestrus, robust spinal EM2 analgesia is manifest but this requires spinal dynorphin/KOR and glutamate-activated mGluR1. Furthermore, spinal mGluR1 blockade in a proestrus chronic pain animal (eliminating spinal EM2 analgesia) exacerbates mechanical allodynia, revealing tempering by endogenous opioid(s). A complex containing mu-opioid receptor, KOR, aromatase, mGluRs, and mERα are foundational to eliciting endogenous opioid anti-allodynia. Aromatase-mERα oligomers are also plentiful, in a central nervous system region-specific fashion. These can be independently regulated and allow estrogens to act intracellularly within the same signaling complex in which they are synthesized, explaining asynchronous relationships between circulating estrogens and central nervous system estrogen functionalities. Observations with EM2 highlight the translational relevance of extensively characterizing exogenous responsiveness to endogenous opioids and the neuronal circuits that mediate them along with the multiplicity of estrogenic systems that concomitantly function in phase and out-of-phase with the reproductive cycle.


Assuntos
Analgesia , Estrogênios/fisiologia , Glutamatos/fisiologia , Nociceptividade/fisiologia , Peptídeos Opioides/fisiologia , Analgesia/métodos , Analgésicos/farmacologia , Animais , Sistema Nervoso Central/fisiologia , Dor Crônica/tratamento farmacológico , Dor Crônica/fisiopatologia , Feminino , Humanos , Masculino , Modelos Neurológicos , Receptores de Estrogênio/fisiologia , Receptores de Glutamato/fisiologia , Receptores Opioides/fisiologia , Pesquisa Translacional Biomédica
7.
J Neurosci ; 41(7): 1553-1565, 2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33361463

RESUMO

Psychostimulant use disorder is a major public health issue, and despite the scope of the problem there are currently no Food and Drug Administration (FDA)-approved treatments. There would be tremendous utility in development of a treatment that could help patients both achieve and maintain abstinence. Previous work from our group has identified granulocyte-colony stimulating factor (G-CSF) as a neuroactive cytokine that alters behavioral response to cocaine, increases synaptic dopamine release, and enhances cognitive flexibility. Here, we investigate the role of G-CSF in affecting extinction and reinstatement of cocaine-seeking and perform detailed characterization of its proteomic effects in multiple limbic substructures. Male Sprague Dawley rats were injected with PBS or G-CSF during (1) extinction or (2) abstinence from cocaine self-administration, and drug seeking behavior was measured. Quantitative assessment of changes in the proteomic landscape in the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC) were performed via data-independent acquisition (DIA) mass spectrometry analysis. Administration of G-CSF during extinction accelerated the rate of extinction, and administration during abstinence attenuated cue-induced cocaine-seeking. Analysis of global protein expression demonstrated that G-CSF regulated proteins primarily in mPFC that are critical to glutamate signaling and synapse maintenance. Taken together, these findings support G-CSF as a viable translational research target with the potential to reduce drug craving or seeking behaviors. Importantly, recombinant G-CSF exists as an FDA-approved medication which may facilitate rapid clinical translation. Additionally, using cutting-edge multiregion discovery proteomics analyses, these studies identify a novel mechanism underlying G-CSF effects on behavioral plasticity.SIGNIFICANCE STATEMENT Pharmacological treatments for psychostimulant use disorder are desperately needed, especially given the disease's chronic, relapsing nature. However, there are currently no Food and Drug Administration (FDA)-approved pharmacotherapies. Emerging evidence suggests that targeting the immune system may be a viable translational research strategy; preclinical studies have found that the neuroactive cytokine granulocyte-colony stimulating factor (G-CSF) alters cocaine reward and reinforcement and can enhance cognitive flexibility. Given this basis of evidence we studied the effects of G-CSF treatment on extinction and reinstatement of cocaine seeking. We find that administration of G-CSF accelerates extinction and reduces cue-induced drug seeking after cocaine self-administration. In addition, G-CSF leads to downregulation of synaptic glutamatergic proteins in medial prefrontal cortex (mPFC), suggesting that G-CSF influences drug seeking via glutamatergic mechanisms.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Comportamento de Procura de Droga/efeitos dos fármacos , Glutamatos/fisiologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Animais , Transtornos Relacionados ao Uso de Cocaína/psicologia , Fissura/efeitos dos fármacos , Sinais (Psicologia) , Extinção Psicológica/efeitos dos fármacos , Sistema Límbico/efeitos dos fármacos , Masculino , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteômica , Ratos , Ratos Sprague-Dawley , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Síndrome de Abstinência a Substâncias/psicologia
8.
J Neurochem ; 155(1): 7-9, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32681746

RESUMO

Hippocampal excitatory glutamatergic transmission is critically involved in cognitive functions such as learning and memory. A severe impairment of spatial memory is associated with the Alzheimer's disease characteristic augmentation of soluble Amyloid-beta1-42 which in turn leads to glutamatergic neurotransmission dysfunction. As the molecular basis of such correlations has not been completely understood, this Editorial highlights a study in the current issue of the Journal of Neurochemistry in which Yeung and coworkers provide an elegant anatomical study that sheds light into this problematic. Through a rigorous immunohistochemical approach, a sub-regional expression pattern of ionotropic glutamate receptors and vesicular transporters was determined in control and beta amyloid-injected mouse hippocampus. The selected areas participate in information processing and thus, in memory formation. Furthermore, the authors discuss their findings in the context of cognitive deficits present in Alzheimer's disease patients delivering an intuitive analysis of plausible molecular events that disturb proper glutamate signaling. This study takes an important step toward a better understanding of the complexity of Amyloid-beta1-42 and glutamatergic neurotransmission interactions.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Glutamatos/fisiologia , Hipocampo/efeitos dos fármacos , Animais , Humanos , Camundongos , Fragmentos de Peptídeos/farmacologia , Receptores de Glutamato/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
9.
J Neurosci ; 40(20): 3882-3895, 2020 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-32291327

RESUMO

Neonatal tissue damage induces long-term deficits in inhibitory synaptic transmission within the spinal superficial dorsal horn (SDH) that include a reduction in primary afferent-evoked, feedforward inhibition onto adult projection neurons. However, the subpopulations of mature GABAergic interneurons which are compromised by early-life injury have yet to be identified. The present research illuminates the persistent effects of neonatal surgical injury on the function of inhibitory SDH interneurons derived from the prodynorphin (DYN) lineage, a population that synapses directly onto lamina I spinoparabrachial neurons and is known to suppress mechanical pain and itch in adults. The results demonstrate that hindpaw incision at postnatal day 3 (P3) significantly decreased the strength of primary afferent-evoked glutamatergic drive onto DYN neurons within the adult mouse SDH while increasing the appearance of afferent-evoked inhibition onto the same population. Neonatal injury also dampened the intrinsic membrane excitability of mature DYN neurons, and reduced their action potential discharge in response to sensory input, compared with naive littermate controls. Furthermore, P3 incision decreased the efficacy of inhibitory DYN synapses onto adult spinoparabrachial neurons, which reflected a prolonged reduction in the probability of GABA release. Collectively, the data suggest that early-life tissue damage may persistently constrain the ability of spinal DYN interneurons to limit ascending nociceptive transmission to the adult brain. This is predicted to contribute to the loss of feedforward inhibition onto mature projection neurons, and the "priming" of nociceptive circuits in the developing spinal cord, following injuries during the neonatal period.SIGNIFICANCE STATEMENT Neonatal injury has lasting effects on pain processing in the adult CNS, including a reduction in feedforward inhibition onto ascending projection neurons in the spinal dorsal horn. While it is clear that spinal GABAergic interneurons are comprised of multiple subpopulations that play distinct roles in somatosensation, the identity of those interneurons which are compromised by tissue damage during early life remains unknown. Here we document persistent deficits in spinal inhibitory circuits involving dynorphin-lineage interneurons previously implicated in gating mechanical pain and itch. Notably, neonatal injury reduced the strength of dynorphin-lineage inhibitory synapses onto mature lamina I spinoparabrachial neurons, a major output of the spinal nociceptive network, which could contribute to the priming of pain pathways by early tissue damage.


Assuntos
Dinorfinas , Membro Posterior/lesões , Inibição Neural , Vias Neurais/fisiopatologia , Corno Dorsal da Medula Espinal/lesões , Potenciais de Ação , Animais , Animais Recém-Nascidos , Análise por Conglomerados , Feminino , Glutamatos/fisiologia , Membro Posterior/inervação , Membro Posterior/fisiopatologia , Interneurônios , Camundongos , Neurônios Aferentes , Nociceptividade , Técnicas de Patch-Clamp , Medula Espinal/fisiopatologia , Corno Dorsal da Medula Espinal/fisiopatologia
10.
J Comput Neurosci ; 48(1): 65-84, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31980990

RESUMO

Hebbian plasticity means that if the firing of two neurons is correlated, then their connection is strengthened. Conversely, uncorrelated firing causes a decrease in synaptic strength. Spike-timing-dependent plasticity (STDP) represents one instantiation of Hebbian plasticity. Under STDP, synaptic changes depend on the relative timing of the pre- and post-synaptic firing. By inducing pre- and post-synaptic firing at different relative times the STDP curves of many neurons have been determined, and it has been found that there are different curves for different neuron types or synaptic sites. Biophysically, strengthening (long-term potentiation, LTP) or weakening (long-term depression, LTD) of glutamatergic synapses depends on the post-synaptic influx of calcium (Ca2+): weak influx leads to LTD, while strong, transient influx causes LTP. The voltage-dependent NMDA receptors are the main source of Ca2+ influx, but they will only open if a post-synaptic depolarisation coincides with pre-synaptic neurotransmitter release. Here we present a computational mechanism for Ca2+-dependent plasticity in which the interplay between the pre-synaptic neurotransmitter release and the post-synaptic membrane potential leads to distinct Ca2+ time-courses, which in turn lead to the change in synaptic strength. It is shown that the model complies with classic STDP results, as well as with results obtained with triplets of spikes. Furthermore, the model is capable of displaying different shapes of STDP curves, as observed in different experimental studies.


Assuntos
Sinalização do Cálcio/fisiologia , Modelos Neurológicos , Plasticidade Neuronal/fisiologia , Algoritmos , Simulação por Computador , Fenômenos Eletrofisiológicos/fisiologia , Glutamatos/fisiologia , Humanos , Potenciação de Longa Duração/fisiologia , Potenciais da Membrana/fisiologia , Neurônios/fisiologia , Neurotransmissores/metabolismo , Receptores de AMPA/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia
11.
Nat Neurosci ; 23(3): 423-432, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31959933

RESUMO

Sodium appetite is a powerful form of motivation that can drive ingestion of high, yet aversive concentrations of sodium in animals that are depleted of sodium. However, in normal conditions, sodium appetite is suppressed to prevent homeostatic deviations. Although molecular and neural mechanisms underlying the stimulation of sodium appetite have received much attention recently, mechanisms that inhibit sodium appetite remain largely obscure. Here we report that serotonin 2c receptor (Htr2c)-expressing neurons in the lateral parabrachial nucleus (LPBNHtr2c neurons) inhibit sodium appetite. Activity of these neurons is regulated by bodily sodium content, and their activation can rapidly suppress sodium intake. Conversely, inhibition of these neurons specifically drives sodium appetite, even during euvolemic conditions. Notably, the physiological role of Htr2c expressed by LPBN neurons is to disinhibit sodium appetite. Our results suggest that LPBNHtr2c neurons act as a brake against sodium appetite and that their alleviation is required for the full manifestation of sodium appetite.


Assuntos
Apetite/fisiologia , Receptor 5-HT2C de Serotonina/fisiologia , Sódio na Dieta , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/fisiologia , Animais , Plexo Braquial/fisiologia , Comportamento Alimentar , Glutamatos/fisiologia , Homeostase , Hipovolemia/psicologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Patch-Clamp , Receptor 5-HT2C de Serotonina/genética , Neurônios Serotoninérgicos/fisiologia , Sódio/metabolismo
12.
J Neurosci ; 40(3): 509-525, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31719165

RESUMO

Medial olivocochlear (MOC) efferent neurons in the brainstem comprise the final stage of descending control of the mammalian peripheral auditory system through axon projections to the cochlea. MOC activity adjusts cochlear gain and frequency tuning, and protects the ear from acoustic trauma. The neuronal pathways that activate and modulate the MOC somata in the brainstem to drive these cochlear effects are poorly understood. Evidence suggests that MOC neurons are primarily excited by sound stimuli in a three-neuron activation loop from the auditory nerve via an intermediate neuron in the cochlear nucleus. Anatomical studies suggest that MOC neurons receive diverse synaptic inputs, but the functional effect of additional synaptic influences on MOC neuron responses is unknown. Here we use patch-clamp electrophysiological recordings from identified MOC neurons in brainstem slices from mice of either sex to demonstrate that in addition to excitatory glutamatergic synapses, MOC neurons receive inhibitory GABAergic and glycinergic synaptic inputs. These synapses are activated by electrical stimulation of axons near the medial nucleus of the trapezoid body (MNTB). Focal glutamate uncaging confirms MNTB neurons as a source of inhibitory synapses onto MOC neurons. MNTB neurons inhibit MOC action potentials, but this effect depresses with repeat activation. This work identifies a new pathway of connectivity between brainstem auditory neurons and indicates that MOC neurons are both excited and inhibited by sound stimuli received at the same ear. The pathway depression suggests that the effect of MNTB inhibition of MOC neurons diminishes over the course of a sustained sound.SIGNIFICANCE STATEMENT Medial olivocochlear (MOC) neurons are the final stage of descending control of the mammalian auditory system and exert influence on cochlear mechanics to modulate perception of acoustic stimuli. The brainstem pathways that drive MOC function are poorly understood. Here we show for the first time that MOC neurons are inhibited by neurons of the MNTB, which may suppress the effects of MOC activity on the cochlea.


Assuntos
Núcleo Coclear/fisiologia , Neurônios Eferentes/fisiologia , Núcleo Olivar/fisiologia , Corpo Trapezoide/fisiologia , Estimulação Acústica , Animais , Axônios/fisiologia , Tronco Encefálico/citologia , Tronco Encefálico/fisiologia , Nervo Coclear/fisiologia , Núcleo Coclear/citologia , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/genética , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Glutamatos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Olivar/citologia , Técnicas de Patch-Clamp , Sinapses/fisiologia , Corpo Trapezoide/citologia
13.
J Neurosci ; 39(40): 7910-7919, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31420456

RESUMO

Proper function of pharyngeal dilator muscles, including the genioglossus muscle of the tongue, is required to maintain upper airway patency. During sleep, the activity of these muscles is suppressed, and as a result individuals with obstructive sleep apnea experience repeated episodes of upper airway closure when they are asleep, in particular during rapid-eye-movement (REM) sleep. Blocking cholinergic transmission in the hypoglossal motor nucleus (MoXII) restores REM sleep genioglossus activity, highlighting the importance of cholinergic transmission in the inhibition of hypoglossal motor neurons (HMNs) during REM sleep. Glutamatergic afferent input from neurons in the parahypoglossal (PH) region to the HMNs is critical for MoXII respiratory motor output. We hypothesized that state-dependent cholinergic regulation may be mediated by this pathway. Here we studied the effects of cholinergic transmission in HMNs in adult male and female mice using patch-clamp recordings in brain slices. Using channelrhodopsin-2-assisted circuit mapping, we first demonstrated that PH glutamatergic neurons directly and robustly activate HMNs (PHGlut → HMNs). We then show that carbachol consistently depresses this input and that this effect is presynaptic. Additionally, carbachol directly affects HMNs by a variable combination of muscarinic-mediated excitatory and inhibitory responses. Altogether, our results suggest that cholinergic signaling impairs upper airway dilator muscle activity by suppressing glutamatergic input from PH premotoneurons to HMNs and by directly inhibiting HMNs. Our findings highlight the complexity of cholinergic control of HMNs at both the presynaptic and postsynaptic levels and provide a possible mechanism for REM sleep suppression of upper airway muscle activity.SIGNIFICANCE STATEMENT Individuals with obstructive sleep apnea can breathe adequately when awake but experience repeated episodes of upper airway closure when asleep, in particular during REM sleep. Similar to skeletal postural muscles, pharyngeal dilator muscles responsible for maintaining an open upper airway become hypotonic during REM sleep. Unlike spinal motoneurons controlling postural muscles that are inhibited by glycinergic transmission during REM sleep, hypoglossal motoneurons that control the upper airway muscles are inhibited in REM sleep by the combination of monoaminergic disfacilitation and cholinergic inhibition. In this study, we demonstrated how cholinergic signaling inhibits hypoglossal motoneurons through presynaptic and postsynaptic muscarinic receptors. Our results provide a potential mechanism for upper airway hypotonia during REM sleep.


Assuntos
Nervo Hipoglosso/fisiopatologia , Neurônios Motores , Hipotonia Muscular/fisiopatologia , Sistema Nervoso Parassimpático/fisiopatologia , Músculos Respiratórios/fisiopatologia , Sono REM , Animais , Carbacol/farmacologia , Channelrhodopsins , Feminino , Glutamatos/fisiologia , Nervo Hipoglosso/efeitos dos fármacos , Masculino , Camundongos , Agonistas Muscarínicos/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/fisiologia , Optogenética , Sistema Nervoso Parassimpático/efeitos dos fármacos , Técnicas de Patch-Clamp , Sinapses/efeitos dos fármacos
14.
Science ; 363(6430): 948-955, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30819957

RESUMO

We investigated the roles of components of neuronal synapses for development of the Drosophila air sac primordium (ASP). The ASP, an epithelial tube, extends specialized signaling filopodia called cytonemes that take up signals such as Dpp (Decapentaplegic, a homolog of the vertebrate bone morphogenetic protein) from the wing imaginal disc. Dpp signaling in the ASP was compromised if disc cells lacked Synaptobrevin and Synaptotagmin-1 (which function in vesicle transport at neuronal synapses), the glutamate transporter, and a voltage-gated calcium channel, or if ASP cells lacked Synaptotagmin-4 or the glutamate receptor GluRII. Transient elevations of intracellular calcium in ASP cytonemes correlate with signaling activity. Calcium transients in ASP cells depend on GluRII, are activated by l-glutamate and by stimulation of an optogenetic ion channel expressed in the wing disc, and are inhibited by EGTA and by the GluR inhibitor NASPM (1-naphthylacetyl spermine trihydrochloride). Activation of GluRII is essential but not sufficient for signaling. Cytoneme-mediated signaling is glutamatergic.


Assuntos
Sinalização do Cálcio , Proteínas de Drosophila/fisiologia , Glutamatos/fisiologia , Discos Imaginais/fisiologia , Receptores Ionotrópicos de Glutamato/fisiologia , Sinapses/fisiologia , Animais , Animais Geneticamente Modificados , Canais de Cálcio/fisiologia , Drosophila melanogaster/fisiologia , Imagem Óptica , Pseudópodes/fisiologia , Proteínas R-SNARE/fisiologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/fisiologia , Sinaptotagmina I/fisiologia , Técnicas de Cultura de Tecidos
15.
Neuropharmacology ; 161: 107550, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30822498

RESUMO

Glutamate is the major excitatory neurotransmitter in the vertebrate central nervous system. During synaptic activity, glutamate is released and binds to specific membrane receptors and transporters activating, in the one hand, a wide variety of signal transduction cascades, while in the other hand, its removal from the synaptic cleft. Extracellular glutamate concentrations are maintained within physiological levels mainly by glia glutamate transporters. Inefficient clearance of this amino acid is neurotoxic due to a prolonged hyperactivation of its postsynaptic receptors, exacerbating a wide array of intracellular events linked to an ionic imbalance, that results in neuronal cell death. This process is known as excitotoxicity and is the underlying mechanisms of an important number of neurodegenerative diseases. Therefore, it is important to understand the regulation of glutamate transporters function. The transporter activity can be regulated at different levels: gene expression, transporter protein targeting and trafficking, and post-translational modifications of the transporter protein. The identification of these mechanisms has paved the way to our current understanding the role of glutamate transporters in brain physiology and will certainly provide the needed biochemical information for the development of therapeutic strategies towards the establishment of novel therapeutic approaches for the treatment and/or prevention of pathologies associated with excitotoxicity insults. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/genética , Sistema X-AG de Transporte de Aminoácidos/fisiologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Sistema X-AG de Transporte de Aminoácidos/biossíntese , Animais , Glutamatos/fisiologia , Humanos , Neurotransmissores/fisiologia
16.
J Neurosci ; 39(4): 651-662, 2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30504272

RESUMO

Lateral inhibition in the vertebrate retina depends on a negative feedback synapse between horizontal cells (HCs) and rod and cone photoreceptors. A change in pH is thought to be the signal for negative feedback, but its spatial profile in the synaptic cleft is unknown. Here we use three different membrane proteins, each fused to the same genetically-encoded pH-sensitive Green Fluorescent Protein (GFP) (pHluorin), to probe synaptic pH in retina from transgenic zebrafish (Danio rerio) of either sex. We used the cone transducin promoter to express SynaptopHluorin (pHluorin on vesicle-associated membrane protein (VAMP2)) or CalipHluorin (pHluorin on an L-type Ca2+ channel) and the HC-specific connexin-55.5 promoter to express AMPApHluorin (pHluorin on an AMPA receptor). Stimulus light led to increased fluorescence of all three probes, consistent with alkalinization of the synaptic cleft. The receptive field size, sensitivity to surround illumination, and response to activation of an alien receptor expressed exclusively in HCs, are consistent with lateral inhibition as the trigger for alkalinization. However, SynaptopHluorin and AMPApHluorin, which are displaced farther from cone synaptic ribbons than CalipHluorin, reported a smaller pH change. Hence, unlike feedforward glutamatergic transmission, which spills over to allow cross talk between terminals in the cone network, the pH change underlying HC feedback is compartmentalized to individual synaptic invaginations within a cone terminal, consistent with private line communication.SIGNIFICANCE STATEMENT Lateral inhibition (LI) is a fundamental feature of information processing in sensory systems, enhancing contrast sensitivity and enabling edge discrimination. Horizontal cells (HCs) are the first cellular substrate of LI in the vertebrate retina, but the synaptic mechanisms underlying LI are not completely understood, despite decades of study. This paper makes a significant contribution to our understanding of LI, by showing that each HC-cone synapse is a "private-line" that operates independently from other HC-cone connections. Using transgenic zebrafish expressing pHluorin, a pH-sensitive GFP variant spliced onto three different protein platforms expressed either in cones or HCs we show that the feedback pH signal is constrained to individual cone terminals, and more stringently, to individual synaptic contact sites within each terminal.


Assuntos
Retroalimentação Fisiológica/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Horizontais da Retina/fisiologia , Sinapses/fisiologia , Animais , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/fisiologia , Conexinas/metabolismo , Feminino , Glutamatos/fisiologia , Concentração de Íons de Hidrogênio , Masculino , Prótons , Receptores de AMPA/metabolismo , Células Fotorreceptoras Retinianas Cones/ultraestrutura , Células Horizontais da Retina/ultraestrutura , Sinapses/ultraestrutura , Transmissão Sináptica/fisiologia , Proteína 2 Associada à Membrana da Vesícula/genética , Proteína 2 Associada à Membrana da Vesícula/fisiologia , Peixe-Zebra
17.
PLoS One ; 13(9): e0203889, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30208107

RESUMO

The active form of vitamin B6, pyridoxal 5'-phosphate (PLP), plays an essential role in the catalytic mechanism of various proteins, including human glutamate-oxaloacetate transaminase (hGOT1), an important enzyme in amino acid metabolism. A recent molecular and genetic study showed that the E266K, R267H, and P300L substitutions in aspartate aminotransferase, the Arabidopsis analog of hGOT1, genetically suppress a developmentally arrested Arabidopsis RUS mutant. Furthermore, CD analyses suggested that the variants exist as apo proteins and implicated a possible role of PLP in the regulation of PLP homeostasis and metabolic pathways. In this work, we assessed the stability of PLP bound to hGOT1 for the three variant and wildtype (WT) proteins using a combined 6 µs of molecular dynamics (MD) simulation. For the variants and WT in the holo form, the MD simulations reproduced the "closed-open" transition needed for substrate binding. This conformational transition was associated with the rearrangement of the P15-R32 small domain loop providing substrate access to the R387/R293 binding motif. We also showed that formation of the dimer interface is essential for PLP affinity to the active site. The position of PLP in the WT binding site was stabilized by a unique hydrogen bond network of the phosphate binding cup, which placed the cofactor for formation of the covalent Schiff base linkage with K259 for catalysis. The amino acid substitutions at positions 266, 267, and 300 reduced the structural correlation between PLP and the protein active site and/or integrity of the dimer interface. Principal component analysis and energy decomposition clearly suggested dimer misalignment and dissociation for the three variants tested in our work. The low affinity of PLP in the hGOT1 variants observed in our computational work provided structural rationale for the possible role of vitamin B6 in regulating metabolic pathways.


Assuntos
Aspartato Aminotransferase Citoplasmática/genética , Aspartato Aminotransferase Citoplasmática/fisiologia , Fosfato de Piridoxal/metabolismo , Substituição de Aminoácidos/genética , Aspartato Aminotransferase Citoplasmática/ultraestrutura , Aspartato Aminotransferases/metabolismo , Sítios de Ligação/genética , Catálise , Domínio Catalítico , Simulação por Computador , Dimerização , Glutamatos/genética , Glutamatos/fisiologia , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Oxaloacetatos/metabolismo , Análise de Componente Principal , Domínios Proteicos/genética , Fosfato de Piridoxal/química , Fosfato de Piridoxal/fisiologia , Vitamina B 6/metabolismo
18.
Alcohol Clin Exp Res ; 42(11): 2186-2195, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30204234

RESUMO

BACKGROUND: Ventral tegmental area (VTA) GABA neurons have been heavily implicated in alcohol reinforcement and reward. In animals that self-administer alcohol, VTA GABA neurons exhibit increased excitability that may contribute to alcohol's rewarding effects. The present study investigated the effects of acute and chronic ethanol exposure on glutamate (GLU) synaptic transmission to VTA GABA neurons. METHODS: Whole-cell recordings of evoked, spontaneous, and miniature excitatory postsynaptic currents (eEPSCs, sEPSCs, and mEPSCs, respectively) were performed on identified GABA neurons in the VTA of GAD67-GFP+ transgenic mice. Three ethanol exposure paradigms were used: acute ethanol superfusion; a single ethanol injection; and chronic vapor exposure. RESULTS: Acute ethanol superfusion increased the frequency of EPSCs but inhibited mEPSC frequency and amplitude. During withdrawal from a single injection of ethanol, the frequency of sEPSCs was lower than saline controls. There was no difference in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/N-methyl-d-aspartate (NMDA) ratio between neurons following withdrawal from a single exposure to ethanol. However, following withdrawal from chronic ethanol, sEPSCs and mEPSCs had a greater frequency than air controls. There was no difference in AMPA/NMDA ratio following chronic ethanol. CONCLUSIONS: These results suggest that presynaptic mechanisms involving local circuit GLU neurons, and not GLU receptors, contribute to adaptations in VTA GABA neuron excitability that accrue to ethanol exposure, which may contribute to the rewarding properties of alcohol via their regulation of mesolimbic dopamine transmission.


Assuntos
Depressores do Sistema Nervoso Central/toxicidade , Etanol/toxicidade , Glutamatos/fisiologia , Neurônios/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Ácido gama-Aminobutírico/fisiologia , Animais , Dopamina/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Glutamato Descarboxilase/genética , Masculino , Camundongos , Técnicas de Patch-Clamp , Síndrome de Abstinência a Substâncias/fisiopatologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
19.
BMC Biol ; 16(1): 100, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30200940

RESUMO

BACKGROUND: Parkinson's disease (PD)-associated E3 ubiquitin ligase Parkin is enriched at glutamatergic synapses, where it ubiquitinates multiple substrates, suggesting that its mutation/loss-of-function could contribute to the etiology of PD by disrupting excitatory neurotransmission. Here, we evaluate the impact of four common PD-associated Parkin point mutations (T240M, R275W, R334C, G430D) on glutamatergic synaptic function in hippocampal neurons. RESULTS: We find that expression of these point mutants in cultured hippocampal neurons from Parkin-deficient and Parkin-null backgrounds alters NMDA and AMPA receptor-mediated currents and cell-surface levels and prevents the induction of long-term depression. Mechanistically, we demonstrate that Parkin regulates NMDA receptor trafficking through its ubiquitination of GluN1, and that all four mutants are impaired in this ubiquitinating activity. Furthermore, Parkin regulates synaptic AMPA receptor trafficking via its binding and retention of the postsynaptic scaffold Homer1, and all mutants are similarly impaired in this capacity. CONCLUSION: Our findings demonstrate that pathogenic Parkin mutations disrupt glutamatergic synaptic transmission in hippocampal neurons by impeding NMDA and AMPA receptor trafficking. Such effects may contribute to the pathophysiology of PD in PARK2 patients.


Assuntos
Glutamatos/fisiologia , Mutação , Neurônios/fisiologia , Doença de Parkinson/metabolismo , Transmissão Sináptica , Ubiquitina-Proteína Ligases/genética , Animais , Hipocampo/fisiologia , Ratos , Ratos Sprague-Dawley , Ubiquitina-Proteína Ligases/metabolismo
20.
Neuropsychopharmacology ; 43(10): 2154-2160, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29977074

RESUMO

The ability of ketamine administration to activate prefrontal glutamate neurotransmission is thought to be a key mechanism contributing to its transient psychotomimetic effects and its delayed and sustained antidepressant effects. Rodent studies employing carbon-13 magnetic resonance spectroscopy (13C MRS) methods have shown ketamine and other N-methyl-D-aspartate (NMDA) receptor antagonists to transiently increase measures reflecting glutamate-glutamine cycling and glutamate neurotransmission in the frontal cortex. However, there are not yet direct measures of glutamate neurotransmission in vivo in humans to support these hypotheses. The current first-level pilot study employed a novel prefrontal 13C MRS approach similar to that used in the rodent studies for direct measurement of ketamine effects on glutamate-glutamine cycling. Twenty-one participants (14 healthy and 7 depressed) completed two 13C MRS scans during infusion of normal saline or subanesthetic doses of ketamine. Compared to placebo, ketamine increased prefrontal glutamate-glutamine cycling, as indicated by a 13% increase in 13C glutamine enrichment (t = 2.4, p = 0.02). We found no evidence of ketamine effects on oxidative energy production, as reflected by 13C glutamate enrichment. During ketamine infusion, the ratio of 13C glutamate/glutamine enrichments, a putative measure of neurotransmission strength, was correlated with the Clinician-Administered Dissociative States Scale (r = -0.54, p = 0.048). These findings provide the most direct evidence in humans to date that ketamine increases glutamate release in the prefrontal cortex, a mechanism previously linked to schizophrenia pathophysiology and implicated in the induction of rapid antidepressant effects.


Assuntos
Antidepressivos/farmacologia , Transtorno Depressivo/fisiopatologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Glutamatos/fisiologia , Ketamina/farmacologia , Córtex Pré-Frontal/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Adulto , Idoso , Metabolismo Energético/efeitos dos fármacos , Feminino , Alucinógenos/farmacologia , Voluntários Saudáveis , Humanos , Espectroscopia de Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Córtex Pré-Frontal/efeitos dos fármacos , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...