Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.525
Filtrar
1.
Bioorg Med Chem Lett ; 71: 128807, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35605837

RESUMO

SST5 receptor activation potently inhibits insulin secretion from pancreatic ß-cells, and an orally available nonpeptide selective SST5 agonist may be used to effectively manage the blood glucose levels of congenital HI patients to avoid severe hypoglycemia. Our medicinal chemistry efforts have led to the discovery of 4-(3-aminopyrrolidinyl)-3-aryl-5-(benzimidazol-2-yl)-pyridine analogs as potent SST5 agonists. This class of molecules exhibits excellent human SST5 potency and selectivity against SST1, SST2, SST3 and SST4 receptors. Leading compound 3-{4-[(3S)-3-aminopyrrolidin-1-yl]-5-(4-methyl-1H-1,3-benzodiazol-2-yl)pyridin-3-yl-5-fluorobenzonitrile (28, CRN02481) showed limited off-target activity and good pharmacokinetic profiles in both male Sprague Dawley rats and Beagle dogs to advance into further preclinical evaluations.


Assuntos
Hiperinsulinismo Congênito , Somatostatina , Animais , Hiperinsulinismo Congênito/tratamento farmacológico , Cães , Humanos , Masculino , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Somatostatina/agonistas , Somatostatina/farmacologia , Somatostatina/fisiologia
2.
Biomolecules ; 12(2)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35204812

RESUMO

Despite the obvious differences in the pathophysiology of distinct neuropsychiatric diseases or neurodegenerative disorders, some of them share some general but pivotal mechanisms, one of which is the disruption of excitation/inhibition balance. Such an imbalance can be generated by changes in the inhibitory system, very often mediated by somatostatin-containing interneurons (SOM-INs). In physiology, this group of inhibitory interneurons, as well as somatostatin itself, profoundly shapes the brain activity, thus influencing the behavior and plasticity; however, the changes in the number, density and activity of SOM-INs or levels of somatostatin are found throughout many neuropsychiatric and neurological conditions, both in patients and animal models. Here, we (1) briefly describe the brain somatostatinergic system, characterizing the neuropeptide somatostatin itself, its receptors and functions, as well the physiology and circuitry of SOM-INs; and (2) summarize the effects of the activity of somatostatin and SOM-INs in both physiological brain processes and pathological brain conditions, focusing primarily on learning-induced plasticity and encompassing selected neuropsychological and neurodegenerative disorders, respectively. The presented data indicate the somatostatinergic-system-mediated inhibition as a substantial factor in the mechanisms of neuroplasticity, often disrupted in a plethora of brain pathologies.


Assuntos
Interneurônios , Somatostatina , Animais , Humanos , Aprendizagem/fisiologia , Plasticidade Neuronal/fisiologia , Somatostatina/farmacologia , Somatostatina/fisiologia
3.
Mol Brain ; 14(1): 130, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34429141

RESUMO

Somatostatin-expressing interneurons (SOM-INs) are a major subpopulation of GABAergic cells in CA1 hippocampus that receive excitation from pyramidal cells (PCs), and, in turn, provide feedback inhibition onto PC dendrites. Excitatory synapses onto SOM-INs show a Hebbian long-term potentiation (LTP) mediated by type 1a metabotropic glutamate receptors (mGluR1a) that is implicated in hippocampus-dependent learning. The neuropeptide somatostatin (SST) is also critical for hippocampal long-term synaptic plasticity, as well as learning and memory. SST effects on hippocampal PCs are well documented, but its actions on inhibitory interneurons remain largely undetermined. In the present work, we investigate the involvement of SST in long-term potentiation of CA1 SOM-IN excitatory synapses using pharmacological approaches targeting the somatostatinergic system and whole cell recordings in slices from transgenic mice expressing eYFP in SOM-INs. We report that application of exogenous SST14 induces long-term potentiation of excitatory postsynaptic potentials in SOM-INs via somatostatin type 1-5 receptors (SST1-5Rs) but does not affect synapses of PC or parvalbumin-expressing interneurons. Hebbian LTP in SOM-INs was prevented by inhibition of SSTRs and by depletion of SST by cysteamine treatment, suggesting a critical role of endogenous SST in LTP. LTP of SOM-IN excitatory synapses induced by SST14 was independent of NMDAR and mGluR1a, activity-dependent, and prevented by blocking GABAA receptor function. Our results indicate that endogenous SST may contribute to Hebbian LTP at excitatory synapses of SOM-INs by controlling GABAA inhibition, uncovering a novel role for SST in regulating long-term synaptic plasticity in somatostatinergic cells that may be important for hippocampus-dependent memory processes.


Assuntos
Região CA1 Hipocampal/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Neurônios GABAérgicos/efeitos dos fármacos , Interneurônios/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Somatostatina/fisiologia , Sinapses/efeitos dos fármacos , Animais , Proteínas de Bactérias , Cisteamina/farmacologia , Feminino , Antagonistas de Receptores de GABA-A/farmacologia , Neurônios GABAérgicos/metabolismo , Técnicas de Introdução de Genes , Genes Reporter , Humanos , Interneurônios/metabolismo , Proteínas Luminescentes , Masculino , Memória/fisiologia , Camundongos , Camundongos Transgênicos , Peptídeos Cíclicos/farmacologia , Receptores de Glutamato Metabotrópico/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/fisiologia , Somatostatina/farmacologia , Sinapses/fisiologia
4.
Eur J Endocrinol ; 185(4): R93-R101, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34370694

RESUMO

In 2008, the first evidence of a new hormone called neuronostatin was published. The hormone was discovered using a bioinformatic method and found to originate from the same preprohormone as somatostatin. This small peptide hormone of 13 amino acids and a C-terminal amidation was soon found to exert pleiotropic physiological effects. In animal studies, neuronostatin has been shown to reduce food intake and delay gastric emptying and gastrointestinal transit. Furthermore, neuronostatin has been shown to affect glucose metabolism by increasing glucagon secretion during situations when glucose concentrations are low. Additionally, neuronostatin has been shown to affect neural tissue and cardiomyocytes by suppressing cardiac contractility. The effects of neuronostatin have not yet been delineated in humans, but if the effects found in animal studies translate to humans it could position neuronostatin as a promising target in the treatment of obesity, hypertension and diabetes. In this review, we describe the discovery of neuronostatin and the current understanding of its physiological role and potential therapeutic applicability.


Assuntos
Hormônios Peptídicos/fisiologia , Animais , Regulação do Apetite/efeitos dos fármacos , Regulação do Apetite/genética , Diabetes Mellitus/genética , Diabetes Mellitus/terapia , Esvaziamento Gástrico/efeitos dos fármacos , Esvaziamento Gástrico/genética , Humanos , Hipertensão/genética , Hipertensão/terapia , Contração Muscular/efeitos dos fármacos , Contração Muscular/genética , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Obesidade/genética , Obesidade/terapia , Hormônios Peptídicos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Somatostatina/química , Somatostatina/farmacologia , Somatostatina/fisiologia
5.
Commun Biol ; 4(1): 732, 2021 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-34127787

RESUMO

The central amygdala (CE) emerges as a critical node for affective processing. However, how CE local circuitry interacts with brain wide affective states is yet uncharted. Using basic nociception as proxy, we find that gene expression suggests diverging roles of the two major CE neuronal populations, protein kinase C δ-expressing (PKCδ+) and somatostatin-expressing (SST+) cells. Optogenetic (o)fMRI demonstrates that PKCδ+/SST+ circuits engage specific separable functional subnetworks to modulate global brain dynamics by a differential bottom-up vs. top-down hierarchical mesoscale mechanism. This diverging modulation impacts on nocifensive behavior and may underly CE control of affective processing.


Assuntos
Afeto/fisiologia , Tonsila do Cerebelo/fisiologia , Rede Nervosa/fisiologia , Nociceptividade/fisiologia , Tonsila do Cerebelo/citologia , Animais , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Optogenética/métodos , Proteína Quinase C-delta/metabolismo , Proteína Quinase C-delta/fisiologia , Somatostatina/metabolismo , Somatostatina/fisiologia
6.
Front Endocrinol (Lausanne) ; 12: 652363, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33796080

RESUMO

Somatostatin (SST) and somatostatin receptors (SSTRs) play an important role in the brain and gastrointestinal (GI) system. SST is produced in various organs and cells, and the inhibitory function of somatostatin-containing cells is involved in a range of physiological functions and pathological modifications. The GI system is the largest endocrine organ for digestion and absorption, SST-endocrine cells and neurons in the GI system are a critical effecter to maintain homeostasis via SSTRs 1-5 and co-receptors, while SST-SSTRs are involved in chemo-sensory, mucus, and hormone secretion, motility, inflammation response, itch, and pain via the autocrine, paracrine, endocrine, and exoendocrine pathways. It is also a power inhibitor for tumor cell proliferation, severe inflammation, and post-operation complications, and is a first-line anti-cancer drug in clinical practice. This mini review focuses on the current function of producing SST endocrine cells and local neurons SST-SSTRs in the GI system, discusses new development prognostic markers, phosphate-specific antibodies, and molecular imaging emerging in diagnostics and therapy, and summarizes the mechanism of the SST family in basic research and clinical practice. Understanding of endocrines and neuroendocrines in SST-SSTRs in GI will provide an insight into advanced medicine in basic and clinical research.


Assuntos
Trato Gastrointestinal/fisiologia , Receptores de Somatostatina/fisiologia , Somatostatina/fisiologia , Animais , Antineoplásicos/farmacologia , Comunicação Celular/efeitos dos fármacos , Proliferação de Células , Modelos Animais de Doenças , Sistema Nervoso Entérico/fisiologia , Homeostase , Humanos , Inflamação , Ligantes , Neurônios/metabolismo , Sistema Nervoso Parassimpático/fisiologia , Prognóstico , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Células Secretoras de Somatostatina/metabolismo , Sistema Nervoso Simpático/fisiologia
7.
J Neurosci ; 40(48): 9224-9235, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33097639

RESUMO

Cortical responses to sensory stimuli are strongly modulated by temporal context. One of the best studied examples of such modulation is sensory adaptation. We first show that in response to repeated tones pyramidal (Pyr) neurons in male mouse auditory cortex (A1) exhibit facilitating and stable responses, in addition to adapting responses. To examine the potential mechanisms underlying these distinct temporal profiles, we developed a reduced spiking model of sensory cortical circuits that incorporated the signature short-term synaptic plasticity (STP) profiles of the inhibitory parvalbumin (PV) and somatostatin (SST) interneurons. The model accounted for all three temporal response profiles as the result of dynamic changes in excitatory/inhibitory balance produced by STP, primarily through shifts in the relative latency of Pyr and inhibitory neurons. Transition between the three response profiles was possible by changing the strength of the inhibitory PV→Pyr and SST→Pyr synapses. The model predicted that a unit's latency would be related to its temporal profile. Consistent with this prediction, the latency of stable units was significantly shorter than that of adapting and facilitating units. Furthermore, because of the history-dependence of STP the model generated a paradoxical prediction: that inactivation of inhibitory neurons during one tone would decrease the response of A1 neurons to a subsequent tone. Indeed, we observed that optogenetic inactivation of PV neurons during one tone counterintuitively decreased the spiking of Pyr neurons to a subsequent tone 400 ms later. These results provide evidence that STP is critical to temporal context-dependent responses in the sensory cortex.SIGNIFICANCE STATEMENT Our perception of speech and music depends strongly on temporal context, i.e., the significance of a stimulus depends on the preceding stimuli. Complementary neural mechanisms are needed to sometimes ignore repetitive stimuli (e.g., the tic of a clock) or detect meaningful repetition (e.g., consecutive tones in Morse code). We modeled a neural circuit that accounts for diverse experimentally-observed response profiles in auditory cortex (A1) neurons, based on known forms of short-term synaptic plasticity (STP). Whether the simulated circuit reduced, maintained, or enhanced its response to repeated tones depended on the relative dominance of two different types of inhibitory cells. The model made novel predictions that were experimentally validated. Results define an important role for STP in temporal context-dependent perception.


Assuntos
Estimulação Acústica , Córtex Auditivo/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Parvalbuminas/fisiologia , Somatostatina/fisiologia , Algoritmos , Animais , Córtex Auditivo/citologia , Simulação por Computador , Masculino , Camundongos , Optogenética , Células Piramidais/fisiologia
8.
Elife ; 92020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-33034285

RESUMO

Connections from the ventral hippocampus (vHPC) to the prefrontal cortex (PFC) regulate cognition, emotion, and memory. These functions are also tightly controlled by inhibitory networks in the PFC, whose disruption is thought to contribute to mental health disorders. However, relatively little is known about how the vHPC engages different populations of interneurons in the PFC. Here we use slice physiology and optogenetics to study vHPC-evoked feed-forward inhibition in the mouse PFC. We first show that cholecystokinin (CCK+), parvalbumin (PV+), and somatostatin (SOM+) expressing interneurons are prominent in layer 5 (L5) of infralimbic PFC. We then show that vHPC inputs primarily activate CCK+ and PV+ interneurons, with weaker connections onto SOM+ interneurons. CCK+ interneurons make stronger synapses onto pyramidal tract (PT) cells over nearby intratelencephalic (IT) cells. However, CCK+ inputs undergo depolarization-induced suppression of inhibition (DSI) and CB1 receptor modulation only at IT cells. Moreover, vHPC-evoked feed-forward inhibition undergoes DSI only at IT cells, confirming a central role for CCK+ interneurons. Together, our findings show how vHPC directly engages multiple populations of inhibitory cells in deep layers of the infralimbic PFC, highlighting unexpected roles for both CCK+ interneurons and endocannabinoid modulation in hippocampal-prefrontal communication.


Assuntos
Colecistocinina/fisiologia , Endocanabinoides/fisiologia , Hipocampo/fisiologia , Interneurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Animais , Colecistocinina/metabolismo , Endocanabinoides/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/fisiologia , Parvalbuminas/metabolismo , Células Piramidais/fisiologia , Somatostatina/metabolismo , Somatostatina/fisiologia
9.
Sci Rep ; 10(1): 15235, 2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32943676

RESUMO

Pharmacological experiments indicate that neuropeptides can effectively tune neuronal activity and modulate locomotor output patterns. However, their functions in shaping innate locomotion often remain elusive. For example, somatostatin has been previously shown to induce locomotion when injected in the brain ventricles but to inhibit fictive locomotion when bath-applied in the spinal cord in vitro. Here, we investigated the role of somatostatin in innate locomotion through a genetic approach by knocking out somatostatin 1.1 (sst1.1) in zebrafish. We automated and carefully analyzed the kinematics of locomotion over a hundred of thousand bouts from hundreds of mutant and control sibling larvae. We found that the deletion of sst1.1 did not impact acousto-vestibular escape responses but led to abnormal exploration. sst1.1 mutant larvae swam over larger distance, at higher speed and performed larger tail bends, indicating that Somatostatin 1.1 inhibits spontaneous locomotion. Altogether our study demonstrates that Somatostatin 1.1 innately contributes to slowing down spontaneous locomotion.


Assuntos
Somatostatina/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Fenômenos Biomecânicos , Comportamento Exploratório/fisiologia , Feminino , Mutação da Fase de Leitura , Técnicas de Inativação de Genes , Larva/fisiologia , Locomoção/fisiologia , Masculino , Deleção de Sequência , Somatostatina/deficiência , Somatostatina/genética , Natação/fisiologia , Gravação em Vídeo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
10.
J Neurosci ; 40(27): 5214-5227, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32467358

RESUMO

The limitation of plasticity in the adult brain impedes functional recovery later in life from brain injury or disease. This pressing clinical issue may be resolved by enhancing plasticity in the adult brain. One strategy for triggering robust plasticity in adulthood is to reproduce one of the hallmark physiological events of experience-dependent plasticity observed during the juvenile critical period: to rapidly reduce the activity of parvalbumin (PV)-expressing interneurons and disinhibit local excitatory neurons. This may be achieved through the enhancement of local inhibitory inputs, particularly those of somatostatin (SST)-expressing interneurons. However, to date the means for manipulating SST interneurons for enhancing cortical plasticity in the adult brain are not known. We show that SST interneuron-selective overexpression of Lypd6, an endogenous nicotinic signaling modulator, enhances ocular dominance plasticity in the adult primary visual cortex (V1). Lypd6 overexpression mediates a rapid experience-dependent increase in the visually evoked activity of SST interneurons as well as a simultaneous reduction in PV interneuron activity and disinhibition of excitatory neurons. Recapitulating this transient activation of SST interneurons using chemogenetics similarly enhanced V1 plasticity. Notably, we show that SST-selective Lypd6 overexpression restores visual acuity in amblyopic mice that underwent early long-term monocular deprivation. Our data in both male and female mice reveal selective modulation of SST interneurons and a putative downstream circuit mechanism as an effective method for enhancing experience-dependent cortical plasticity as well as functional recovery in adulthood.SIGNIFICANCE STATEMENT The decline of cortical plasticity after closure of juvenile critical period consolidates neural circuits and behavior, but this limits functional recovery from brain diseases and dysfunctions in later life. Here we show that activation of cortical somatostatin (SST) interneurons by Lypd6, an endogenous modulator of nicotinic acetylcholine receptors, enhances experience-dependent plasticity and recovery from amblyopia in adulthood. This manipulation triggers rapid reduction of PV interneuron activity and disinhibition of excitatory neurons, which are known hallmarks of cortical plasticity during juvenile critical periods. Our study demonstrates modulation of SST interneurons by Lypd6 to achieve robust levels of cortical plasticity in the adult brain and may provide promising targets for restoring brain function in the event of brain trauma or disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas Ligadas por GPI/fisiologia , Interneurônios/fisiologia , Plasticidade Neuronal/fisiologia , Somatostatina/fisiologia , Córtex Visual/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Dominância Ocular/genética , Potenciais Evocados Visuais/genética , Potenciais Evocados Visuais/fisiologia , Feminino , Proteínas Ligadas por GPI/genética , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Plasticidade Neuronal/genética , Fosfatidilinositóis/farmacologia , Receptores Nicotínicos/genética , Recuperação de Função Fisiológica/genética , Visão Monocular/genética , Visão Monocular/fisiologia , Acuidade Visual/genética
11.
Acta Physiol (Oxf) ; 229(3): e13464, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32145704

RESUMO

AIM: It is debated whether the inhibition of glucagon secretion by glucose results from direct effects of glucose on the α-cell (intrinsic regulation) or by paracrine effects exerted by beta- or delta-cell products. METHODS: To study this in a more physiological model than isolated islets, we perfused isolated rat pancreases and measured glucagon, insulin and somatostatin secretion in response to graded increases in perfusate glucose concentration (from 3.5 to 4, 5, 6, 7, 8, 10, 12 mmol/L) as well as glucagon responses to blockage/activation of insulin/GABA/somatostatin signalling with or without addition of glucose. RESULTS: Glucagon secretion was reduced by about 50% (compared to baseline secretion at 3.5 mmol/L) within minutes after increasing glucose from 4 to 5 mmol/L (P < .01, n = 13). Insulin secretion was increased minimally, but significantly, compared to baseline (3.5 mmol/L) at 4 mmol/L, whereas somatostatin secretion was not significantly increased from baseline until 7 mmol/L. Hereafter secretion of both increased gradually up to 12 mmol/L glucose. Neither recombinant insulin (1 µmol/L), GABA (300 µmol/L) or the insulin-receptor antagonist S961 (at 1 µmol/L) affected basal (3.5 mmol/L) or glucose-induced (5.0 mmol/L) attenuation of glucagon secretion (n = 7-8). Somatostatin-14 attenuated glucagon secretion by ~ 95%, and blockage of somatostatin-receptor (SSTR)-2 or combined blockage of SSTR-2, -3 and -5 by specific antagonists increased glucagon output (at 3.5 mmol/L glucose) and prevented glucose-induced (from 3.5 to 5.0 mmol/L) suppression of secretion. CONCLUSION: Somatostatin is a powerful and tonic inhibitor of glucagon secretion from the rat pancreas and is required for glucose to inhibit glucagon secretion.


Assuntos
Glucagon/sangue , Glucose/administração & dosagem , Pâncreas/fisiologia , Somatostatina/fisiologia , Animais , Insulina/sangue , Perfusão , Ratos
12.
J Smooth Muscle Res ; 56(0): 69-81, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33473062

RESUMO

Gastric contractions show two specific patterns in many species, migrating motor contractions (MMC) and postprandial contractions (PPCs), that occur in the fasted and fed states, respectively. In this study, we examined the role of somatostatin (SST) in gastric motility both in vivo and in vitro using the Asian house shrew (Suncus murinus). We performed in vivo recordings of gastric motility and in vitro organ bath experiments using S. murinus, which was recently established as a small laboratory animal for use in tests of gastrointestinal motility. SST (1.65 µg kg-1 min-1) was intravenously administered during phase II of MMC and PPCs. Next, the effect of SST on motilin-induced gastric contractions at phase I of MMC was measured. Cyclosomatostatin (CSST), an SST receptor antagonist, was administered at the peak of phase III of MMC. In addition, the effect of SST (10-11-10-9 M) on motilin-induced gastric contractions was evaluated using an organ bath experiment in vitro. In conscious, free-moving S. murinus, the administration of SST decreased the occurrence of the spontaneous phase II of MMC and PPCs. Pretreatment with SST and octreotide suppressed the induction of motilin-induced gastric contractions both in vivo and in vitro. Administration of CSST before the peak of spontaneous phase III contractions had no effect on gastric contractions. Endogenous SST is not involved in the regulation of gastric MMC and PPCs, but exogenous SST suppresses spontaneous gastric contractions. Thus, SST would be good for treating abnormal gastrointestinal motility disorders.


Assuntos
Motilidade Gastrointestinal/efeitos dos fármacos , Somatostatina/farmacologia , Animais , Depressão Química , Feminino , Técnicas In Vitro , Masculino , Motilina/farmacologia , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Complexo Mioelétrico Migratório/efeitos dos fármacos , Período Pós-Prandial , Musaranhos , Somatostatina/fisiologia , Somatostatina/uso terapêutico
13.
J Neurosci ; 39(41): 8038-8050, 2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31471471

RESUMO

Integration and modulation of primary afferent sensory information begins at the first terminating sites within the CNS, where central inhibitory circuits play an integral role. Viscerosensory information is conveyed to the nucleus of the solitary tract (NTS) where it initiates neuroendocrine, behavioral, and autonomic reflex responses that ensure optimal internal organ function. This excitatory input is modulated by diverse, local inhibitory interneurons, whose functions are not clearly understood. Here we show that, in male rats, 65% of somatostatin-expressing (SST) NTS neurons also express GAD67, supporting their likely role as inhibitory interneurons. Using whole-cell recordings of NTS neurons, from horizontal brainstem slices of male and female SST-yellow fluorescent protein (YFP) and SST-channelrhodopsin 2 (ChR2)-YFP mice, we quantified the impact of SST-NTS neurons on viscerosensory processing. Light-evoked excitatory photocurrents were reliably obtained from SST-ChR2-YFP neurons (n = 16) and the stimulation-response characteristics determined. Most SST neurons (57%) received direct input from solitary tract (ST) afferents, indicating that they form part of a feedforward circuit. All recorded SST-negative NTS neurons (n = 72) received SST-ChR2 input. ChR2-evoked PSCs were largely inhibitory and, in contrast to previous reports, were mediated by both GABA and glycine. When timed to coincide, the ChR2-activated SST input suppressed ST-evoked action potentials at second-order NTS neurons, demonstrating strong modulation of primary viscerosensory input. These data indicate that the SST inhibitory network innervates broadly within the NTS, with the potential to gate viscerosensory input to powerfully alter autonomic reflex function and other behaviors.SIGNIFICANCE STATEMENT Sensory afferent input is modulated according to state. For example the baroreflex is altered during a stress response or exercise, but the basic mechanisms underpinning this sensory modulation are not fully understood in any sensory system. Here we demonstrate that the neuronal processing of viscerosensory information begins with synaptic gating at the first central synapse with second-order neurons in the NTS. These data reveal that the somatostatin subclass of inhibitory interneurons are driven by visceral sensory input to play a major role in gating viscerosensory signals, placing them within a feedforward circuit within the NTS.


Assuntos
Rede Nervosa/fisiologia , Neurônios/fisiologia , Sensação/fisiologia , Filtro Sensorial/fisiologia , Somatostatina/fisiologia , Animais , Retroalimentação Fisiológica , Feminino , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/fisiologia , Glicina/fisiologia , Interneurônios/fisiologia , Masculino , Camundongos , Rede Nervosa/citologia , Estimulação Luminosa , Ratos , Ratos Sprague-Dawley , Núcleo Solitário/citologia , Núcleo Solitário/fisiologia , Fibras Aferentes Viscerais/fisiologia , Ácido gama-Aminobutírico/fisiologia
14.
Science ; 364(6442)2019 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-31123108

RESUMO

Hippocampal pyramidal cells encode memory engrams, which guide adaptive behavior. Selection of engram-forming cells is regulated by somatostatin-positive dendrite-targeting interneurons, which inhibit pyramidal cells that are not required for memory formation. Here, we found that γ-aminobutyric acid (GABA)-releasing neurons of the mouse nucleus incertus (NI) selectively inhibit somatostatin-positive interneurons in the hippocampus, both monosynaptically and indirectly through the inhibition of their subcortical excitatory inputs. We demonstrated that NI GABAergic neurons receive monosynaptic inputs from brain areas processing important environmental information, and their hippocampal projections are strongly activated by salient environmental inputs in vivo. Optogenetic manipulations of NI GABAergic neurons can shift hippocampal network state and bidirectionally modify the strength of contextual fear memory formation. Our results indicate that brainstem NI GABAergic cells are essential for controlling contextual memories.


Assuntos
Aprendizagem por Associação/fisiologia , Neurônios GABAérgicos/fisiologia , Núcleos da Rafe/fisiologia , Animais , Feminino , Interneurônios/química , Interneurônios/fisiologia , Masculino , Testes de Memória e Aprendizagem , Camundongos , Camundongos Endogâmicos C57BL , Inibição Neural/fisiologia , Células Piramidais/química , Células Piramidais/fisiologia , Somatostatina/análise , Somatostatina/fisiologia , Ritmo Teta
15.
J Physiol ; 597(8): 2297-2314, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30784081

RESUMO

KEY POINTS: There is a rapid interneuronal response to focal activity in cortex, which restrains laterally propagating activity, including spreading epileptiform activity. The interneuronal response involves intense activation of both parvalbumin- and somatostatin-expressing interneurons. Interneuronal bursting is time-locked to glutamatergic barrages in the pre-ictal period. Ca2+ imaging using conditional expression of GCaMP6f provides an accurate readout of the evolving firing patterns in both types of interneuron. The activation profiles of the two interneuronal classes are temporally offset, with the parvalbumin population being activated first, and typically, at higher rates. ABSTRACT: Previous work has described powerful restraints on laterally spreading activity in cortical networks, arising from a rapid feedforward interneuronal response to focal activity. This response is particularly prominent ahead of an ictal wavefront. Parvalbumin-positive interneurons are considered to be critically involved in this feedforward inhibition, but it is not known what role, if any, is provided by somatostatin-expressing interneurons, which target the distal dendrites of pyramidal cells. We used a combination of electrophysiology and cell class-specific Ca2+ imaging in mouse brain slices bathed in 0 Mg2+ medium to characterize the activity profiles of pyramidal cells and parvalbumin- and somatostatin-expressing interneurons during epileptiform activation. The GCaMP6f signal strongly correlates with the level of activity for both interneuronal classes. Both interneuronal classes participate in the feedfoward inhibition. This contrasts starkly with the pattern of pyramidal recruitment, which is greatly delayed. During these barrages, both sets of interneurons show intense bursting, at rates up to 300Hz, which is time-locked to the glutamatergic barrages. The activity of parvalbumin-expressing interneurons appears to peak early in the pre-ictal period, and can display depolarizing block during the ictal event. In contrast, somatostatin-expressing interneuronal activity peaks significantly later, and firing persists throughout the ictal events. Interictal events appear to be very similar to the pre-ictal period, albeit with slightly lower firing rates. Thus, the inhibitory restraint arises from a coordinated pattern of activity in the two main classes of cortical interneurons.


Assuntos
Interneurônios/fisiologia , Parvalbuminas/fisiologia , Somatostatina/fisiologia , Animais , Encéfalo/fisiologia , Feminino , Masculino , Camundongos Transgênicos
16.
J Neurosci ; 39(5): 788-801, 2019 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-30523065

RESUMO

Hippocampus-dependent learning processes are coordinated via a large diversity of GABAergic inhibitory mechanisms. The α5 subunit-containing GABAA receptor (α5-GABAAR) is abundantly expressed in the hippocampus populating primarily the extrasynaptic domain of CA1 pyramidal cells, where it mediates tonic inhibitory conductance and may cause functional deficits in synaptic plasticity and hippocampus-dependent memory. However, little is known about synaptic expression of the α5-GABAAR and, accordingly, its location site-specific function. We examined the cell- and synapse-specific distribution of the α5-GABAAR in the CA1 stratum oriens/alveus (O/A) using a combination of immunohistochemistry, whole-cell patch-clamp recordings and optogenetic stimulation in hippocampal slices obtained from mice of either sex. In addition, the input-specific role of the α5-GABAAR in spatial learning and anxiety-related behavior was studied using behavioral testing and chemogenetic manipulations. We demonstrate that α5-GABAAR is preferentially targeted to the inhibitory synapses made by the vasoactive intestinal peptide (VIP)- and calretinin-positive terminals onto dendrites of somatostatin-expressing interneurons. In contrast, synapses made by the parvalbumin-positive inhibitory inputs to O/A interneurons showed no or little α5-GABAAR. Inhibiting the α5-GABAAR in control mice in vivo improved spatial learning but also induced anxiety-like behavior. Inhibiting the α5-GABAAR in mice with inactivated CA1 VIP input could still improve spatial learning and was not associated with anxiety. Together, these data indicate that the α5-GABAAR-mediated phasic inhibition via VIP input to interneurons plays a predominant role in the regulation of anxiety while the α5-GABAAR tonic inhibition via this subunit may control spatial learning.SIGNIFICANCE STATEMENT The α5-GABAAR subunit exhibits high expression in the hippocampus, and regulates the induction of synaptic plasticity and the hippocampus-dependent mnemonic processes. In CA1 principal cells, this subunit occupies mostly extrasynaptic sites and mediates tonic inhibition. Here, we provide evidence that, in CA1 somatostatin-expressing interneurons, the α5-GABAAR subunit is targeted to synapses formed by the VIP- and calretinin-expressing inputs, and plays a specific role in the regulation of anxiety-like behavior.


Assuntos
Região CA1 Hipocampal/metabolismo , Neurônios/metabolismo , Receptores de GABA-A/metabolismo , Sinapses/metabolismo , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/efeitos dos fármacos , Calbindina 2/fisiologia , Feminino , Antagonistas de Receptores de GABA-A/farmacologia , Interneurônios/efeitos dos fármacos , Interneurônios/fisiologia , Interneurônios/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/ultraestrutura , Optogenética , Técnicas de Patch-Clamp , Somatostatina/fisiologia , Sinapses/efeitos dos fármacos , Sinapses/ultraestrutura , Peptídeo Intestinal Vasoativo/fisiologia
17.
Cereb Cortex ; 29(1): 42-53, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29161383

RESUMO

The basal forebrain provides modulatory input to the cortex regulating brain states and cognitive processing. Somatostatin-expressing neurons constitute a heterogeneous GABAergic population known to functionally inhibit basal forebrain cortically projecting cells thus favoring sleep and cortical synchronization. However, it remains unclear if somatostatin cells can regulate population activity patterns in the basal forebrain and modulate cortical dynamics. Here, we demonstrate that somatostatin neurons regulate the corticopetal synaptic output of the basal forebrain impinging on cortical activity and behavior. Optogenetic inactivation of somatostatin neurons in vivo rapidly modified neural activity in the basal forebrain, with the consequent enhancement and desynchronization of activity in the prefrontal cortex, reflected in both neuronal spiking and network oscillations. Cortical activation was partially dependent on cholinergic transmission, suppressing slow waves and potentiating gamma oscillations. In addition, recruitment dynamics was cell type-specific, with interneurons showing similar temporal profiles, but stronger responses than pyramidal cells. Finally, optogenetic stimulation of quiescent animals during resting periods prompted locomotor activity, suggesting generalized cortical activation and increased arousal. Altogether, we provide physiological and behavioral evidence indicating that somatostatin neurons are pivotal in gating the synaptic output of the basal forebrain, thus indirectly controlling cortical operations via both cholinergic and non-cholinergic mechanisms.


Assuntos
Potenciais de Ação/fisiologia , Prosencéfalo Basal/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Somatostatina/fisiologia , Animais , Prosencéfalo Basal/química , Prosencéfalo Basal/citologia , Feminino , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/química , Optogenética/métodos , Técnicas de Cultura de Órgãos , Córtex Pré-Frontal/química , Córtex Pré-Frontal/citologia , Somatostatina/análise
18.
Science ; 361(6397): 76-81, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29976824

RESUMO

The tuberal nucleus (TN) is a surprisingly understudied brain region. We found that somatostatin (SST) neurons in the TN, which is known to exhibit pathological or cytological changes in human neurodegenerative diseases, play a crucial role in regulating feeding in mice. GABAergic tuberal SST (TNSST) neurons were activated by hunger and by the hunger hormone, ghrelin. Activation of TNSST neurons promoted feeding, whereas inhibition reduced it via projections to the paraventricular nucleus and bed nucleus of the stria terminalis. Ablation of TNSST neurons reduced body weight gain and food intake. These findings reveal a previously unknown mechanism of feeding regulation that operates through orexigenic TNSST neurons, providing a new perspective for understanding appetite changes.


Assuntos
Regulação do Apetite/fisiologia , Neurônios GABAérgicos/fisiologia , Somatostatina/fisiologia , Núcleos Ventrais do Tálamo/fisiologia , Animais , Grelina/fisiologia , Camundongos , Camundongos Mutantes , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Núcleos Ventrais do Tálamo/citologia
19.
J Neurosci ; 38(22): 5168-5181, 2018 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-29735555

RESUMO

Recent studies have identified an especially important role for basal forebrain GABAergic (BFVGAT) neurons in the regulation of behavioral waking and fast cortical rhythms associated with cognition. However, BFVGAT neurons comprise several neurochemically and anatomically distinct subpopulations, including parvalbumin-containing BFVGAT neurons and somatostatin-containing BFVGAT neurons (BFSOM neurons), and it was recently reported that optogenetic activation of BFSOM neurons increases the probability of a wakefulness to non-rapid-eye movement (NREM) sleep transition when stimulated during the rest period of the animal. This finding was unexpected given that most BFSOM neurons are not NREM sleep active and that central administration of the synthetic somatostatin analog, octreotide, suppresses NREM sleep or increases REM sleep. Here we used a combination of genetically driven chemogenetic and optogenetic activation, chemogenetic inhibition, and ablation approaches to further explore the in vivo role of BFSOM neurons in arousal control. Our findings indicate that acute activation or inhibition of BFSOM neurons is neither wakefulness nor NREM sleep promoting and is without significant effect on the EEG, and that chronic loss of these neurons is without effect on total 24 h sleep amounts, although a small but significant increase in waking was observed in the lesioned mice during the early active period. Our in vitro cell recordings further reveal electrophysiological heterogeneity in BFSOM neurons, specifically suggesting at least two distinct subpopulations. Together, our data support the more nuanced view that BFSOM neurons are electrically heterogeneous and are not NREM sleep or wake promoting per se, but may exert, in particular during the early active period, a modest inhibitory influence on arousal circuitry.SIGNIFICANCE STATEMENT The cellular basal forebrain (BF) is a highly complex area of the brain that is implicated in a wide range of higher-level neurobiological processes, including regulating and maintaining normal levels of electrocortical and behavioral arousal. The respective in vivo roles of BF cell populations and their neurotransmitter systems in the regulation of electrocortical and behavioral arousal remains incompletely understood. Here we seek to define the neurobiological contribution of GABAergic somatostatin-containing BF neurons to arousal control. Understanding the respective contribution of BF cell populations to arousal control may provide critical insight into the pathogenesis of a host of neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, schizophrenia, and the cognitive impairments of normal aging.


Assuntos
Prosencéfalo Basal/fisiologia , Comportamento Animal/fisiologia , Neurônios/fisiologia , Somatostatina/fisiologia , Animais , Prosencéfalo Basal/citologia , Eletroencefalografia , Fenômenos Eletrofisiológicos/fisiologia , Feminino , Deleção de Genes , Genótipo , Masculino , Camundongos , Optogenética , Sono de Ondas Lentas/fisiologia , Somatostatina/metabolismo , Ativação Transcricional , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/fisiologia , Vigília/fisiologia
20.
Nat Neurosci ; 21(6): 851-859, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29786081

RESUMO

How learning enhances neural representations for behaviorally relevant stimuli via activity changes of cortical cell types remains unclear. We simultaneously imaged responses of pyramidal cells (PYR) along with parvalbumin (PV), somatostatin (SOM), and vasoactive intestinal peptide (VIP) inhibitory interneurons in primary visual cortex while mice learned to discriminate visual patterns. Learning increased selectivity for task-relevant stimuli of PYR, PV and SOM subsets but not VIP cells. Strikingly, PV neurons became as selective as PYR cells, and their functional interactions reorganized, leading to the emergence of stimulus-selective PYR-PV ensembles. Conversely, SOM activity became strongly decorrelated from the network, and PYR-SOM coupling before learning predicted selectivity increases in individual PYR cells. Thus, learning differentially shapes the activity and interactions of multiple cell classes: while SOM inhibition may gate selectivity changes, PV interneurons become recruited into stimulus-specific ensembles and provide more selective inhibition as the network becomes better at discriminating behaviorally relevant stimuli.


Assuntos
Interneurônios/fisiologia , Aprendizagem/fisiologia , Córtex Visual/fisiologia , Ácido gama-Aminobutírico/fisiologia , Animais , Aprendizagem por Discriminação/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Rede Nervosa/citologia , Rede Nervosa/fisiologia , Parvalbuminas/fisiologia , Técnicas de Patch-Clamp , Reconhecimento Fisiológico de Modelo/fisiologia , Células Piramidais/metabolismo , Células Piramidais/fisiologia , Filtro Sensorial/fisiologia , Somatostatina/fisiologia , Peptídeo Intestinal Vasoativo/fisiologia , Córtex Visual/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...