Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Neurosci ; 43(6): 965-978, 2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-36623875

RESUMO

Recent findings from our laboratory demonstrated that the rostral nucleus of the solitary tract (rNST) retains some responsiveness to sugars in double-knock-out mice lacking either the T1R1+T1R3 (KO1+3) or T1R2+T1R3 (KO2+3) taste receptor heterodimers. Here, we extended these findings in the parabrachial nucleus (PBN) of male and female KO1+3 mice using warm stimuli to optimize sugar responses and employing additional concentrations and pharmacological agents to probe mechanisms. PBN T1R-independent sugar responses, including those to concentrated glucose, were more evident than in rNST. Similar to the NST, there were no "sugar-best" neurons in KO1+3 mice. Nevertheless, 1000 mm glucose activated nearly 55% of PBN neurons, with responses usually occurring in neurons that also displayed acid and amiloride-insensitive NaCl responses. In wild-type (WT) mice, concentrated sugars activated the same electrolyte-sensitive neurons but also "sugar-best" cells. Regardless of genotype, phlorizin, an inhibitor of the sodium-glucose co-transporter (SGLT), a component of a hypothesized alternate glucose-sensing mechanism, did not diminish responses to 1000 mm glucose. The efficacy of concentrated sugars for driving neurons broadly responsive to electrolytes implied an origin from Type III taste bud cells. To test this, we used the carbonic anhydrase (CA) inhibitor dorzolamide (DRZ), previously shown to inhibit amiloride-insensitive sodium responses arising from Type III taste bud cells. Dorzolamide had no effect on sugar-elicited responses in WT sugar-best PBN neurons but strongly suppressed them in WT and KO1+3 electrolyte-generalist neurons. These findings suggest a novel T1R-independent mechanism for hyperosmotic sugars, involving a CA-dependent mechanism in Type III taste bud cells.SIGNIFICANCE STATEMENT Since the discovery of Tas1r receptors for sugars and artificial sweeteners, evidence has accrued that mice lacking these receptors maintain some behavioral, physiological, and neural responsiveness to sugars. But the substrate(s) has remained elusive. Here, we recorded from parabrachial nucleus (PBN) taste neurons and identified T1R-independent responses to hyperosmotic sugars dependent on carbonic anhydrase (CA) and occurring primarily in neurons broadly responsive to NaCl and acid, implying an origin from Type III taste bud cells. The effectiveness of different sugars in driving these T1R-independent responses did not correlate with their efficacy in driving licking, suggesting they evoke a nonsweet sensation. Nevertheless, these salient responses are likely to comprise an adequate cue for learned preferences that occur in the absence of T1R receptors.


Assuntos
Papilas Gustativas , Paladar , Animais , Feminino , Masculino , Camundongos , Amilorida/farmacologia , Glucose , Camundongos Knockout , Cloreto de Sódio/farmacologia , Açúcares/farmacologia , Paladar/fisiologia , Papilas Gustativas/fisiologia
2.
eNeuro ; 9(5)2022.
Artigo em Inglês | MEDLINE | ID: mdl-36104278

RESUMO

The rostral nucleus of the solitary tract (rNST), the initial CNS site for processing gustatory information, is comprised of two major cell types, glutamatergic excitatory and GABAergic inhibitory neurons. Although many investigators have described taste responses of rNST neurons, the phenotypes of these cells were unknown. To directly compare the response characteristics of both inhibitory and noninhibitory neurons, we recorded from mice expressing Channelrhodopsin-2 (ChR2) under the control of GAD65, a synthetic enzyme for GABA. We observed that chemosensitive profiles of GABAergic taste neurons (G+TASTE) were similar to non-GABA taste neurons (G-TASTE) but had much lower response rates. We further observed a novel subpopulation of GABA cells located more ventrally in the nucleus that were unresponsive to taste stimulation (G+UNR), suggesting pathways for inhibition initiated by centrifugal sources. This preparation also allowed us to determine how optogenetic activation of the rNST GABA network impacted the taste responses of G-TASTE neurons. Activating rNST inhibitory circuitry suppressed gustatory responses of G-TASTE neurons across all qualities and chemosensitive types of neurons. Although the tuning curves of identified G-TASTE were modestly sharpened, the overall shape of response profiles and the ensemble pattern remained highly stable. These neurophysiological effects were consistent with the behavioral consequences of activating GAD65-expressing inhibitory neurons using DREADDs. In a brief-access licking task, concentration-response curves to both palatable (sucrose, maltrin) and unpalatable (quinine) stimuli were shifted to the right when GABA neurons were activated. Thus, the rNST GABAergic network is poised to modulate taste intensity across the qualitative and hedonic spectrum.


Assuntos
Núcleo Solitário , Paladar , Animais , Channelrhodopsins/genética , Neurônios GABAérgicos/fisiologia , Camundongos , Quinina/farmacologia , Sacarose/farmacologia , Paladar/fisiologia
4.
J Neurophysiol ; 123(2): 843-859, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31913749

RESUMO

Strong evidence supports a major role for heterodimers of the type 1 taste receptor (T1R) family in the taste transduction of sugars (T1R2+T1R3) and amino acids (T1R1+T1R3), but there are also neural and behavioral data supporting T1R-independent mechanisms. Most neural evidence for alternate mechanisms comes from whole nerve recordings in mice with deletion of a single T1R family member, limiting conclusions about the functional significance and T1R independence of the remaining responses. To clarify these issues, we recorded single-unit taste responses from the nucleus of the solitary tract in T1R double-knockout (double-KO) mice lacking functional T1R1+T1R3 [KO1+3] or T1R2+T1R3 [KO2+3] receptors and their wild-type background strains [WT; C57BL/6J (B6), 129X1/SvJ (S129)]. In both double-KO strains, responses to sugars and a moderate concentration of an monosodium glutamate + amiloride + inosine 5'-monophosphate cocktail (0.1 M, i.e., umami) were profoundly depressed, whereas a panel of 0.6 M amino acids were mostly unaffected. Strikingly, in contrast to WT mice, no double-KO neurons responded selectively to sugars and umami, precluding segregation of this group of stimuli from those representing other taste qualities in a multidimensional scaling analysis. Nevertheless, residual sugar responses, mainly elicited by monosaccharides, persisted as small "sideband" responses in double-KOs. Thus other receptors may convey limited information about sugars to the central nervous system, but T1Rs appear critical for coding the distinct perceptual features of sugar and umami stimuli. The persistence of amino acid responses supports previous proposals of alternate receptors, but because these stimuli affected multiple neuron types, further investigations are necessary.NEW & NOTEWORTHY The type 1 taste receptor (T1R) family is crucial for transducing sugars and amino acids, but there is evidence for T1R-independent mechanisms. In this study, single-unit recordings from the nucleus of the solitary tract in T1R double-knockout mice lacking T1R1+T1R3 or T1R2+T1R3 receptors revealed greatly reduced umami synergism and sugar responses. Nevertheless, residual sugar responses persisted, mainly elicited by monosaccharides and evident as "sidebands" in neurons activated more vigorously by other qualities.


Assuntos
Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais/fisiologia , Núcleo Solitário/fisiologia , Papilas Gustativas/fisiologia , Percepção Gustatória/fisiologia , Aminoácidos/farmacologia , Animais , Açúcares da Dieta/farmacologia , Feminino , Aromatizantes/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética
5.
Am J Physiol Regul Integr Comp Physiol ; 314(6): R802-R810, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29443544

RESUMO

The taste of l-glutamate and its synergism with 5'-ribonucleotides is thought to be primarily mediated through the T1R1+T1R3 heterodimer in some mammals, including rodents and humans. While knockout (KO) mice lacking either receptor subunit show impaired sensitivity to a range of monosodium glutamate (MSG) concentrations mixed with 2.5 mM inosine 5'-monophosphate (IMP) in amiloride, wild-type (WT) controls can detect this IMP concentration, hindering direct comparison between genotypes. Moreover, some residual sensitivity persists in the KO group, suggesting that the remaining subunit could maintain a limited degree of function. Here, C57BL/6J, 129X1/SvJ, and T1R1+T1R3 double KO mice ( n = 16 each to start the experiment) were trained in a two-response operant task in gustometers and then tested for their ability to discriminate 100 µM amiloride from MSG (starting with 0.6 M) and IMP (starting with 2.5 mM) in amiloride (MSG+I+A). Testing continued with successive dilutions of both MSG and IMP (in amiloride). The two WT strains were similarly sensitive to MSG+I+A ( P > 0.8). KO mice, however, were significantly impaired relative to either WT strain ( P < 0.01), although they were able to detect the highest concentrations. Thus, normal detectability of MSG+I+A requires an intact T1R1+T1R3 receptor, without regard for allelic variation in the T1R3 gene between the WT strains. Nevertheless, residual sensitivity by the T1R1+T1R3 KO mice demonstrates that a T1R-independent mechanism can contribute to the detectability of high concentrations of this prototypical umami compound stimulus.


Assuntos
Inosina Monofosfato/farmacologia , Receptores Acoplados a Proteínas G/genética , Glutamato de Sódio/farmacologia , Paladar/efeitos dos fármacos , Amilorida/farmacologia , Sistemas de Transporte de Aminoácidos , Animais , Proteínas de Arabidopsis , Condicionamento Operante/efeitos dos fármacos , Discriminação Psicológica/efeitos dos fármacos , Diuréticos/farmacologia , Feminino , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cloreto de Sódio/farmacologia
6.
JPEN J Parenter Enteral Nutr ; 41(1): 113-124, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-26598504

RESUMO

This article summarizes research findings from 6 experts in the field of taste and feeding that were presented at the 2015 American Society for Parenteral and Enteral Nutrition Research Workshop. The theme was focused on the interaction of taste signals with those of a postingestive origin and how this contributes to regulation of food intake through both physiological and learning processes. Gastric bypass results in exceptional loss of fat mass and increases in circulating levels of key gut peptides, some of which are also expressed along with their cognate receptors in taste buds. Changes in taste preference and food selection in both bariatric surgery patients and rodent models have been reported. Accordingly, the effects of this surgery on taste-related behavior were examined. The conservation of receptor and peptide signaling mechanisms in gustatory and extraoral tissues was discussed in the context of taste responsiveness and the regulation of metabolism. New findings detailing the features of neural circuits between the caudal nucleus of the solitary tract (NST), receiving visceral input from the vagus nerve, and the rostral NST, receiving taste input, were discussed, as was how early life experience with taste stimuli and learned associations between flavor and postoral consequences of nutrients can exert potent and long-lasting effects on feeding.


Assuntos
Papilas Gustativas/fisiologia , Animais , Cirurgia Bariátrica , Glicemia/metabolismo , Congressos como Assunto , Modelos Animais de Doenças , Preferências Alimentares , Hormônios Gastrointestinais/sangue , Humanos , Ratos , Receptores Acoplados a Proteínas G/metabolismo , Paladar
7.
Chem Senses ; 41(6): 515-24, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27131102

RESUMO

Peripheral gustatory neurons express P2X2 purinergic receptors and terminate in the rostral portion of the nucleus tractus solitarius (rNTS), but a relationship between the P2X2 terminal field and taste evoked activity has not been established. Additionally, a portion of somatosensory neurons from the trigeminal nerve, which are devoid of P2X2 expression, also terminate in the lateral rNTS. We hypothesized that P2X2 receptor expression on afferent nerve endings could be used as an anatomical tool for segregating gustatory from mechanosensory responsive regions in the mouse rNTS. C57BL/6 mice were used to record extracellular activity from neurons within the rNTS and the laterally adjacent reticular formation and trigeminal nucleus. Histological reconstruction of electrolytic lesions indicated that gustatory activity coincided with electrode tracks that traversed through P2X2 terminal fields. Gustatory recordings made more rostral in the rNTS had receptive fields located in the anterior oral cavity (AO), whereas gustatory recordings made more caudal in the rNTS had receptive fields located in the posterior oral cavity (PO). Mechanosensory neurons with AO receptive fields were recorded near the lateral border of the P2X2 terminal field and became numerous on electrode tracks made lateral to the P2X2 terminal field. In contrast, mechanosensory responses with PO receptive fields were recorded within the P2X2 terminal field along with gustatory activity and transitioned to mechanosensory only outside the P2X2 terminal field. Collectively, our results indicate that the lateral border of the P2X2 terminal field, demarcates a faithful "transition zone," where AO responses transition from gustatory to mechanosensory.


Assuntos
Mecanorreceptores/metabolismo , Receptores Purinérgicos P2X2/metabolismo , Núcleo Solitário/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL
8.
Chem Senses ; 40(4): 213-5, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25740303

RESUMO

The intriguing observation that leptin decreases sweet-evoked peripheral gustatory responses has aroused much interest (Kawai K, Sugimoto K, Nakashima K, Miura H, Ninomiya Y. 2000. Leptin as a modulator of sweet taste sensitivities in mice. Proc Natl Acad Sci U S A. 97(20):11044-11049.) due to its implied importance in controlling appetite. The effects of this anorexic hormone, however, appear more conditional than originally believed. In this issue of Chemical Senses, a careful study by Glendinning and colleagues, find no effects of leptin on sweet-evoked chorda tympani responses, whereas an equally careful study by Meredith and colleagues, find decreased release of ATP and increased release of 5-HT from taste buds in response to sweet stimuli.


Assuntos
Cálcio/metabolismo , Leptina/sangue , Leptina/farmacologia , Neurotransmissores/metabolismo , Edulcorantes/farmacologia , Papilas Gustativas/efeitos dos fármacos , Papilas Gustativas/metabolismo , Paladar/efeitos dos fármacos , Paladar/fisiologia , Língua/metabolismo , Animais , Feminino , Masculino
9.
J Neurophysiol ; 109(11): 2815-26, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23486207

RESUMO

Taste processing in the rostral nucleus of the solitary tract (rNST) is subject to modulatory influences including opioid peptides. Behavioral pharmacological studies suggest an influence of µ-opioid receptors in rNST, but the underlying mechanism is unknown. To determine the cellular site of action, we tested the effects of the µ-opioid receptor agonist DAMGO in vitro. Whole cell patch-clamp recordings were made in brain stem slices from GAD67-GFP knockin mice expressing enhanced green fluorescent protein (EGFP) under the control of the endogenous promoter for GAD67, a synthetic enzyme for GABA. Neuron counts showed that ∼36% of rNST neurons express GABA. We recorded monosynaptic solitary tract (ST)-evoked currents (jitter ≤ 300 µs) in both GAD67-EGFP-positive (GAD67+) and GAD67-EGFP-negative (GAD67-) neurons with equal frequency (25/31; 22/28), but the inputs to the GAD67+ neurons had significantly smaller paired-pulse ratios compared with GAD67- neurons. DAMGO (0.3 µM) significantly suppressed ST-evoked currents in both cell types (mean suppression = 46 ± 3.3% SE), significantly increased the paired-pulse ratio of these currents, and reduced the frequency of spontaneous miniature excitatory postsynaptic currents but did not diminish their amplitude, indicating a presynaptic site of action. Under inhibitory amino acid receptor blockade, DAMGO was significantly more suppressive in GAD67+ neurons (59% reduction) compared with GAD67- neurons (35% reduction), while the reverse was true in normal artificial cerebrospinal fluid (GAD67+: 35% reduction; GAD67-: 57% reduction). These findings suggest that DAMGO suppresses activity in rNST neurons predominantly via a presynaptic mechanism, and that this effect may interact significantly with tonic or evoked inhibitory activity.


Assuntos
Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Potenciais Evocados , Neurônios GABAérgicos/efeitos dos fármacos , Inibição Neural , Neurotransmissores/farmacologia , Receptores Opioides mu/agonistas , Núcleo Solitário/fisiologia , Animais , Neurônios GABAérgicos/fisiologia , Camundongos , Potenciais Pós-Sinápticos em Miniatura , Núcleo Solitário/citologia , Núcleo Solitário/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/fisiologia
10.
Am J Physiol Regul Integr Comp Physiol ; 302(12): R1401-10, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22513746

RESUMO

Consummatory responses to taste stimuli are modulated by visceral signals processed in the caudal nucleus of the solitary tract (cNST) and ventrolateral medulla. On the basis of decerebrate preparations, this modulation can occur through local brain stem pathways. Among the large number of neuropeptides and neuromodulators implicated in these visceral pathways is neuropeptide Y (NPY), which is oftentimes colocalized in catecholaminergic neurons themselves implicated in glucoprivic-induced feeding and satiety. In addition to the cNST and ventrolateral medulla, noradrenergic and NPY receptors are found in circumscribed regions of the medullary reticular formation rich in preoromotor neurons. To test the hypothesis that NPY may act as a neuromodulator on preoromotor neurons, we recorded the effects of bath application of NPY and specific Y1 and Y2 agonists on currents elicited from electrical stimulation of the rostral (taste) NST in prehypoglossal neurons in a brain stem slice preparation. A high proportion of NST-driven responses were suppressed by NPY, as well as Y1 and Y2 agonists. On the basis of paired pulse ratios and changes in membrane resistance, we concluded that Y1 receptors influence these neurons both presynaptically and postsynaptically and that Y2 receptors have a presynaptic locus. To test the hypothesis that NPY may act in concert with norepinephrine (NE), we examined neurons showing suppressed responses in the presence of a Y2 agonist and demonstrated a greater degree of suppression to a Y2 agonist/NE cocktail. These suppressive effects on preoromotoneurons may reflect a satiety pathway originating from A2 neurons in the caudal brain stem.


Assuntos
Tronco Encefálico/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptores de Neuropeptídeo Y/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Paladar/efeitos dos fármacos , Animais , Neuropeptídeo Y/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeo Y/agonistas , Receptores de Neuropeptídeo Y/antagonistas & inibidores
11.
Am J Physiol Regul Integr Comp Physiol ; 301(3): R690-700, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21697523

RESUMO

The neural control of feeding involves many neuromodulators, including the endogenous opioids that bind µ-opioid receptors (MORs). Injections of the MOR agonist, Damgo, into limbic and hypothalamic forebrain sites increase intake, particularly of palatable foods. Indeed, forebrain Damgo injections increase sucrose-elicited licking but reduce aversive responding (gaping) to quinine, suggesting that MOR activation may enhance taste palatability. A µ-opioid influence on taste reactivity has not been assessed in the brain stem. However, MORs are present in the first-order taste relay, the rostral nucleus of the solitary tract (rNST), and in the immediately subjacent reticular formation (RF), a region known to be essential for consummatory responses. Thus, to evaluate the consequences of rNST/dorsal RF Damgo in this region, we implanted rats with intraoral cannulas, electromyographic electrodes, and brain cannulas aimed at the ventral border of the rNST. Licking and gaping elicited with sucrose, water, and quinine were assessed before and after intramedullary Damgo and saline infusions. Damgo slowed the rate, increased the amplitude, and decreased the size of fluid-induced lick and gape bouts. In addition, the neutral stimulus water, which typically elicits licks, began to evoke gapes. Thus, the current results demonstrate that µ-opioid activation in the rNST/dorsal RF exerts complex effects on oromotor responding that contrast with forebrain effects and are more indicative of a suppressive, rather than a facilitatory effect on ingestion.


Assuntos
Analgésicos Opioides/farmacologia , Comportamento Consumatório/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Receptores Opioides mu/agonistas , Formação Reticular/efeitos dos fármacos , Núcleo Solitário/efeitos dos fármacos , Paladar/efeitos dos fármacos , Analgésicos Opioides/administração & dosagem , Análise de Variância , Animais , Ingestão de Alimentos/efeitos dos fármacos , Eletromiografia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Infusões Parenterais , Injeções , Masculino , Atividade Motora/efeitos dos fármacos , Quinina/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/metabolismo , Formação Reticular/metabolismo , Núcleo Solitário/metabolismo , Sacarose/administração & dosagem , Fatores de Tempo
12.
Behav Neurosci ; 125(4): 519-28, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21604835

RESUMO

There is growing evidence of heterogeneity among responses to bitter stimuli at the peripheral, central and behavioral levels. For instance, the glossopharyngeal (GL) nerve and neurons receiving its projections are more responsive to bitter stimuli than the chorda tympani (CT) nerve, and this is particularly true for some bitter stimuli like PROP & cycloheximide that stimulate the GL to a far greater extent. Given this information, we hypothesized that cutting the GL would have a greater effect on behavioral avoidance of cycloheximide and PROP than quinine and denatonium, which also stimulate the CT, albeit to a lesser degree than salts and acids. Forty male SD rats were divided into four surgery groups: bilateral GL transection (GLX), chorda tympani transection (CTX), SHAM surgery, and combined transection (CTX + GLX). Postsurgical avoidance functions were generated for the four bitter stimuli using a brief-access test. GLX significantly compromised avoidance compared to both CTX and SHAM groups for all stimuli (p < .02), while CTX and SHAM groups did not differ. Contrary to our hypothesis, GLX had a greater effect on quinine than cycloheximide (mean shift of 1.02 vs. 0.27 log10 units). Moreover, combined CTX + GLX transection shifted the concentration-response function further than GLX alone for every stimulus except cycloheximide (ps < .03), suggesting that the GSP nerve is capable of maintaining avoidance of this stimulus to a large degree. This hypothesis is supported by reports of cycloheximide-responsive cells with GSP-innervated receptive fields in the NST and PBN.


Assuntos
Aprendizagem da Esquiva/fisiologia , Traumatismos do Nervo Glossofaríngeo/complicações , Deficiências da Aprendizagem/etiologia , Limiar Gustativo/fisiologia , Paladar/fisiologia , Análise de Variância , Animais , Nervo da Corda do Tímpano/lesões , Nervo da Corda do Tímpano/fisiologia , Cicloeximida/farmacologia , Relação Dose-Resposta a Droga , Masculino , Inibidores da Síntese de Proteínas/farmacologia , Quinina/farmacologia , Ratos , Ratos Sprague-Dawley , Estimulação Química , Limiar Gustativo/efeitos dos fármacos
13.
Behav Neurosci ; 124(2): 225-33, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20364882

RESUMO

The appetitive component of feeding is controlled by forebrain substrates, but the consummatory behaviors of licking, mastication, and swallowing are organized in the brainstem. The target of forebrain appetitive signals is unclear but likely includes regions of the medullary reticular formation (RF). This study was undertaken to determine the necessity of different RF regions for mastication induced by a descending appetitive signal. We measured solid food intake in response to third ventricular (3V) infusions of the orexigenic peptide neuropeptide Y 3-36 in awake, freely moving rats and determined whether focal RF infusions of the GABAA agonist muscimol suppressed eating. RF infusions were centered in either the lateral tegmental field, comprising the intermediate (IRt) and parvocellular (PCRt) RF, or in the nucleus gigantocellularis (Gi). Infusions of NPY 3-36 (5 microg/5 microl) into 3V significantly increased feeding of solid food over a 90-min period compared with the noninfused condition (4.3 g +/- 0.56 vs. 0.57 g +/- 0.57, p < .001). NPY 3-36-induced food intake was suppressed (1.7 g +/- 0.48) by simultaneous infusions of muscimol (0.6 mM/100 nl) into the IRt/PCRt (p < .01). Coincident with the decrease in feeding was a decrease in the amplitude of anterior digastric muscle contractions in response to intraoral sucrose infusions. In contrast, infusions of muscimol into Gi had no discernible effect on food intake or EMG amplitude. These data suggest that the IRt/PCRt is essential for forebrain-initiated mastication, but that the Gi is not a necessary link in this pathway.


Assuntos
Estimulantes do Apetite/antagonistas & inibidores , Ingestão de Alimentos/efeitos dos fármacos , Agonistas GABAérgicos/farmacologia , Bulbo/efeitos dos fármacos , Muscimol/farmacologia , Neuropeptídeo Y/antagonistas & inibidores , Fragmentos de Peptídeos/antagonistas & inibidores , Formação Reticular/efeitos dos fármacos , Animais , Estimulantes do Apetite/administração & dosagem , Estimulantes do Apetite/farmacologia , Interações Medicamentosas , Ingestão de Alimentos/fisiologia , Agonistas GABAérgicos/administração & dosagem , Injeções Intraventriculares , Mastigação/efeitos dos fármacos , Mastigação/fisiologia , Bulbo/fisiologia , Muscimol/administração & dosagem , Neuropeptídeo Y/administração & dosagem , Neuropeptídeo Y/farmacologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Formação Reticular/fisiologia , Estômago/efeitos dos fármacos , Estômago/fisiologia , Sacarose/farmacologia
14.
Physiol Behav ; 97(5): 592-603, 2009 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-19303890

RESUMO

The sensation that humans describe as "bitter" is evoked by a large group of chemically diverse ligands. Bitter stimuli are avoided by a range of species and elicit reflex rejection, behaviors considered adaptations to the toxicity of many of these compounds. We review novel evidence for neurons that are narrowly tuned to bitter ligands at the initial stages of central processing. These "B-best" neurons in the nucleus of the solitary tract (NST) and parabrachial nucleus (PBN) respond to multiple types of bitter stimuli and exhibit average responses to bitter tastants that are 6-8 times larger than to moderate concentrations of compounds representing other qualities. However, in the PBN B-best units are appreciably activated by intense salt and acid. Neurons broadly sensitive to salts and acids ("AN" neurons) also responded to bitter stimuli. This sensitivity appeared restricted to stronger intensities of ionic bitters, as cycloheximide remained ineffective across concentrations. In addition to chemosensitive profile, B-best neurons were also distinctive with regard to their posterior receptive fields, long latencies, slow firing rates and projection status. Compared to B-best NST cells, those in the PBN received increased convergence from anterior and posterior receptive fields and responded to a greater number of bitter stimuli. We conclude that B-best neurons likely contribute to pathways underlying gaping, aversive hedonic quality and taste coding. The differential responsiveness of B-best and AN neurons to ionic and nonionic bitter ligands also suggests a potential substrate for discrimination within this quality.


Assuntos
Neurônios/fisiologia , Núcleo Solitário/citologia , Papilas Gustativas/fisiologia , Potenciais de Ação/fisiologia , Animais , Células Quimiorreceptoras , Estimulação Elétrica/métodos , Tempo de Reação/fisiologia , Estimulação Química , Paladar/fisiologia , Papilas Gustativas/citologia , Limiar Gustativo/fisiologia
15.
J Neurophysiol ; 101(3): 1598-612, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19129294

RESUMO

Bitterness is a distinctive taste sensation, but central coding for this quality remains enigmatic. Although some receptor cells and peripheral fibers are selectively responsive to bitter ligands, central bitter responses are most typical in broadly tuned neurons. Recently we reported more specifically tuned bitter-best cells (B-best) in the nucleus of the solitary tract (NST). Most had glossopharyngeal receptive fields and few projected to the parabrachial nucleus (PBN), suggesting a role in reflexes. To determine their potential contribution to other functions, the present study investigated whether B-best neurons occur further centrally. Responses from 90 PBN neurons were recorded from anesthetized rats. Stimulation with four bitter tastants (quinine, denatonium, propylthiouracil, cycloheximide) and sweet, umami, salty, and sour ligands revealed a substantial proportion of B-best cells (22%). Receptive fields for B-best NST neurons were overwhelmingly foliate in origin, but in PBN, about half received foliate and nasoincisor duct input. Despite convergence, most B-best PBN neurons were as selectively tuned as their medullary counterparts and response profiles were reliable. Regardless of intensity, cycloheximide did not activate broadly tuned acid/sodium (AN) neurons but did elicit robust responses in B-best cells. However, stronger quinine activated AN neurons and concentrated electrolytes stimulated B-best cells, suggesting that B-best neurons might contribute to higher-order functions such as taste quality coding but work in conjunction with other cell types to unambiguously signal bitter-tasting ligands. In this ensemble, B-best neurons would help discriminate sour from bitter stimuli, whereas AN neurons might be more important in differentiating ionic from nonionic bitter stimuli.


Assuntos
Ponte/citologia , Células Receptoras Sensoriais/fisiologia , Paladar/fisiologia , Potenciais de Ação/fisiologia , Análise de Variância , Animais , Mapeamento Encefálico , Masculino , Vias Neurais/fisiologia , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/classificação , Núcleo Solitário/citologia , Estimulação Química , Papilas Gustativas/fisiologia , Limiar Gustativo/fisiologia
16.
Am J Physiol Regul Integr Comp Physiol ; 295(2): R436-48, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18495833

RESUMO

Intraoral infusions of bitter tastants activate expression of the immediate-early gene c-Fos in neurons located in the medial third of the rostral nucleus of the solitary tract (rNST). The distribution of these neurons is distinct from that activated by sour or sweet stimuli. Bitter stimuli are also distinctive because of their potency for eliciting gaping, an oral reflex that functions to actively reject potentially toxic substances. Glossopharyngeal nerve transection profoundly reduces, whereas decerebration spares, the bitter-evoked Fos-like immunoreactivity (FLI) pattern and gaping, implicating the medial rNST as a substrate for the sensory limb of oral rejection. The present experiment tested this hypothesis using microstimulation (100 Hz, 0.2 ms, 5-40 microA) to activate the rNST in awake rats. NST microstimulation elicited licking and gaping, and gaping was evoked from a restricted rNST region. The results indicated some topographic organization in sites effective for evoking gaping, but, in direct conflict with the hypothesis, lateral sites farther from bitter-evoked FLI were more effective than medial sites centered closer to FLI-expressing neurons. The gape-effective sites resemble locations of bitter-responsive neurons recently observed in neurophysiological recordings. These results indicate that bitter-responsive rNST neurons critical for triggering gaping may not express FLI and imply an alternate function for bitter-responsive neurons that do.


Assuntos
Comportamento Animal , Mapeamento Encefálico , Atividade Motora , Reflexo , Núcleo Solitário/fisiologia , Papilas Gustativas/fisiologia , Paladar , Língua/inervação , Animais , Mapeamento Encefálico/métodos , Estimulação Elétrica , Masculino , Ratos , Ratos Sprague-Dawley
17.
J Comp Neurol ; 500(4): 746-60, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17154256

RESUMO

The current investigation used double labeling for NADPHd and Fos-like immunoreactivity to define the relationship between nitric oxide synthase-containing neural elements and taste-activated neurons in the nucleus of the solitary tract (NST) and subjacent reticular formation (RF). Stimulation of awake rats with citric acid and quinine resulted in significant increases in the numbers of double-labeled neurons in both the NST and RF, suggesting that some medullary gustatory neurons utilize nitric oxide (NO) as a transmitter. Overall, double-labeled neurons were most numerous in the caudal reaches of the gustatory zone of the NST, where taste neurons receive inputs from the IXth nerve, suggesting a preferential role for NO neurons in processing gustatory inputs from the posterior oral cavity. However, double-labeled neurons also exhibited a preferential distribution depending on the gustatory stimulus. In the NST, double-labeled neurons were most numerous in the rostral central subnucleus after either stimulus but had a medial bias after quinine stimulation. In the RF, after citric acid stimulation, there was a cluster of double-labeled neurons with distinctive large soma in the parvicellular division of the lateral RF, subjacent to the rostral tip of NST. In contrast, in response to quinine, there was a cluster of double-labeled neurons with much smaller soma in the intermediate zone of the medial RF, a few hundred micrometers caudal to the citric acid cluster. These differential distributions of double-labeled neurons in the NST and RF suggest a role for NO in stimulus-specific gustatory autonomic and oromotor reflex circuits.


Assuntos
Neurônios/enzimologia , Óxido Nítrico Sintase/metabolismo , Formação Reticular/enzimologia , Transdução de Sinais/fisiologia , Núcleo Solitário/enzimologia , Paladar/fisiologia , Animais , Imuno-Histoquímica , Masculino , NADH Desidrogenase/metabolismo , Vias Neurais/enzimologia , Óxido Nítrico/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Formação Reticular/citologia , Núcleo Solitário/citologia
18.
J Neurophysiol ; 96(5): 2513-27, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16899635

RESUMO

Molecular data suggest that receptors for all bitter ligands are coexpressed in the same taste receptor cells (TRCs), whereas physiological results indicate that individual TRCs respond to only a subset of bitter stimuli. It is also unclear to what extent bitter-responsive neurons are stimulated by nonbitter stimuli. To explore these issues, single neuron responses were recorded from the rat nucleus of the solitary tract (NST) during whole mouth stimulation with a variety of bitter compounds: 10 microM cycloheximide, 7 mM propylthiouracil, 10 mM denatonium benzoate, and 3 mM quinine hydrochloride at intensities matched for behavioral effectiveness. Stimuli representing the remaining putative taste qualities were also tested. Particular emphasis was given to activating taste receptors in the foliate papillae innervated by the quinine-sensitive glossopharyngeal nerve. This method revealed a novel population of bitter-best (B-best) cells with foliate receptive fields and significant selectivity for bitter tastants. Across all neurons, multidimensional scaling depicted bitter stimuli as loosely clustered yet clearly distinct from nonbitter tastants. When neurons with posterior receptive fields were analyzed alone, bitter stimuli formed a tighter cluster. Nevertheless, responses to bitter stimuli were variable across B-best neurons, with cycloheximide the most, and quinine the least frequent optimal stimulus. These results indicate heterogeneity for the processing of ionic and nonionic bitter tastants, which is dependent on receptive field. Further, they suggest that neurons selective for bitter substances could contribute to taste coding.


Assuntos
Neurônios/fisiologia , Núcleo Solitário/citologia , Núcleo Solitário/fisiologia , Paladar/fisiologia , Algoritmos , Animais , Comportamento Animal/fisiologia , Plexo Braquial/citologia , Plexo Braquial/fisiologia , Células Quimiorreceptoras/efeitos dos fármacos , Células Quimiorreceptoras/fisiologia , Interpretação Estatística de Dados , Estimulação Elétrica , Eletrofisiologia , Masculino , Vias Neurais/citologia , Vias Neurais/fisiologia , Neurônios/classificação , Neurônios/efeitos dos fármacos , Quinina/farmacologia , Ratos , Ratos Sprague-Dawley , Núcleo Solitário/efeitos dos fármacos , Estimulação Química , Paladar/efeitos dos fármacos
19.
J Comp Neurol ; 488(1): 28-47, 2005 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-15912497

RESUMO

Numerous studies suggest an essential role for the intermediate (IRt) and parvocellular (PCRt) reticular formation (RF) in consummatory ingestive responses. Although the IRt and PCRt contain a large proportion of neurons with projections to the oromotor nuclei, these areas of the RF are heterogeneous with respect to neurotransmitter phenotypes. Glutamatergic, GABAergic, cholinergic, and nitrergic neurons are all found in the PCRt and IRt, but the projections of neurons with these phenotypes to the motor trigeminal (mV) and hypoglossal nucleus (mXII) has not been fully evaluated. In the present study, after small injections of Fluorogold (FG) into mV and mXII, sections were processed immunohistochemically to detect retrogradely labeled FG neurons in combination with the synthetic enzymes for nitric oxide (nitric oxide synthase) or acetylcholine (choline acetyltransferase) or in situ hybridization for the synthetic enzyme for GABA (GAD65/67) or the brainstem vesicular transporter for glutamate (VGLUT2). In three additional cases, FG injections were made into one motor nucleus and cholera toxin (subunit b) injected in the other to determine the presence of dual projection neurons. Premotor neurons to mXII (pre-mXII) were highly concentrated in the IRt. In contrast, there were nearly equal proportions of premotor-trigeminal neurons (pre-mV) in the IRt and PCRt. A high proportion of pre-oromotor neurons were positive for VGLUT2 (pre-mXII: 68%; pre-mV: 53%) but GABAergic projections were differentially distributed with a greater projection to mV (25%) compared to mXII (8%). Significant populations of cholinergic and nitrergic neurons overlapped pre-oromotor neurons, but there was sparse double-labeling (<10%). The IRt also contained a high proportion of neurons that projected to both mV and MXII. These different classes of premotor neurons in the IRt and PCRt provide a substrate for the rhythmic activation of lingual and masticatory muscles.


Assuntos
Mapeamento Encefálico , Vias Neurais/metabolismo , Neurônios/metabolismo , Neurotransmissores/metabolismo , Formação Reticular/metabolismo , Núcleos do Trigêmeo/metabolismo , Acetilcolina/metabolismo , Animais , Comportamento Consumatório/fisiologia , Comportamento Alimentar/fisiologia , Ácido Glutâmico/metabolismo , Nervo Hipoglosso/citologia , Nervo Hipoglosso/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Masculino , Vias Neurais/citologia , Neurônios/citologia , Neurotransmissores/classificação , Ácido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley , Formação Reticular/citologia , Distribuição Tecidual , Núcleos do Trigêmeo/citologia , Ácido gama-Aminobutírico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA