Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Gastrointest Liver Physiol ; 326(4): G345-G359, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38261717

RESUMO

Multiple studies describe prodromal, nonmotor dysfunctions that affect the quality of life of patients who subsequently develop Parkinson's disease (PD). These prodromal dysfunctions comprise a wide array of autonomic issues, including severe gastrointestinal (GI) motility disorders such as dysphagia, delayed gastric emptying, and chronic constipation. Indeed, strong evidence from studies in humans and animal models suggests that the GI tract and its neural, mainly vagal, connection to the central nervous system (CNS) could have a major role in the etiology of PD. In fact, misfolded α-synuclein aggregates that form Lewy bodies and neurites, i.e., the histological hallmarks of PD, are detected in the enteric nervous system (ENS) before clinical diagnosis of PD. The aim of the present review is to provide novel insights into the pathogenesis of GI dysmotility in PD, focusing our attention on functional, neurochemical, and molecular alterations in animal models.


Assuntos
Sistema Nervoso Entérico , Gastroenteropatias , Doença de Parkinson , Animais , Humanos , Roedores , Qualidade de Vida , Gastroenteropatias/etiologia
2.
J Physiol ; 601(21): 4751-4766, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37772988

RESUMO

A monosynaptic pathway connects the substantia nigra pars compacta (SNpc) to neurons of the dorsal motor nucleus of the vagus (DMV). This monosynaptic pathway modulates the vagal control of gastric motility. It is not known, however, whether this nigro-vagal pathway also modulates the tone and motility of the proximal colon. In rats, microinjection of retrograde tracers in the proximal colon and of anterograde tracers in SNpc showed that bilaterally labelled colonic-projecting neurons in the DMV received inputs from SNpc neurons. Microinjections of the ionotropic glutamate receptor agonist, NMDA, in the SNpc increased proximal colonic motility and tone, as measured via a strain gauge aligned with the colonic circular smooth muscle; the motility increase was inhibited by acute subdiaphragmatic vagotomy. Upon transfection of SNpc with pAAV-hSyn-hM3D(Gq)-mCherry, chemogenetic activation of nigro-vagal nerve terminals by brainstem application of clozapine-N-oxide increased the firing rate of DMV neurons and proximal colon motility; both responses were abolished by brainstem pretreatment with the dopaminergic D1-like antagonist SCH23390. Chemogenetic inhibition of nigro-vagal nerve terminals following SNpc transfection with pAAV-hSyn-hM4D(Gi)-mCherry decreased the firing rate of DMV neurons and inhibited proximal colon motility. These data suggest that a nigro-vagal pathway modulates activity of the proximal colon motility tonically via a discrete dopaminergic synapse in a manner dependent on vagal efferent nerve activity. Impairment of this nigro-vagal pathway may contribute to the severely reduced colonic transit and prominent constipation observed in both patients and animal models of parkinsonism. KEY POINTS: Substantia nigra pars compacta (SNpc) neurons are connected to the dorsal motor nucleus of the vagus (DMV) neurons via a presumed direct pathway. Brainstem neurons in the lateral DMV innervate the proximal colon. Colonic-projecting DMV neurons receive inputs from neurons of the SNpc. The nigro-vagal pathway modulates tone and motility of the proximal colon via D1-like receptors in the DMV. The present study provides the mechanistic basis for explaining how SNpc alterations may lead to a high rate of constipation in patients with Parkinson's Disease.


Assuntos
Estômago , Substância Negra , Humanos , Ratos , Animais , Estômago/fisiologia , Ratos Sprague-Dawley , Substância Negra/metabolismo , Nervo Vago/fisiologia , Motilidade Gastrointestinal/fisiologia , Colo , Constipação Intestinal/metabolismo
3.
J Physiol ; 601(4): 801-829, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36696965

RESUMO

Prolonged high-fat diet (HFD) exposure is associated with hyperphagia, excess caloric intake and weight gain. After initial exposure to a HFD, a brief (24-48 h) period of hyperphagia is followed by the regulation of caloric intake and restoration of energy balance within an acute (3-5 day) period. Previous studies have demonstrated this occurs via a vagally mediated signalling cascade that increases glutamatergic transmission via activation of NMDA receptors located on gastric-projecting neurons of the dorsal motor nucleus of the vagus (DMV). The present study used electrophysiological recordings from thin brainstem slice preparations, in vivo recordings of gastric motility and tone, measurement of gastric emptying rates, and food intake studies to investigate the hypothesis that activation of brainstem astrocytes in response to acute HFD exposure is responsible for the increased glutamatergic drive to DMV neurons and the restoration of caloric balance. Pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV excitability, dysregulated gastric tone and motility, attenuated the homeostatic delay in gastric emptying, and prevented the decrease in food intake that is observed during the period of energy regulation following initial exposure to HFD. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome. KEY POINTS: Initial exposure to a high fat diet is associated with a brief period of hyperphagia before caloric intake and energy balance is restored. This period of homeostatic regulation is associated with a vagally mediated signalling cascade that increases glutamatergic transmission to dorsal motor nucleus of the vagus (DMV) neurons via activation of synaptic NMDA receptors. The present study demonstrates that pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV neuronal excitability, dysregulated gastric motility and tone and emptying, and prevented the regulation of food intake following high-fat diet exposure. Astrocyte regulation of glutamatergic transmission to DMV neurons appears to involve release of the gliotransmitters glutamate and ATP. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome.


Assuntos
Astrócitos , Ingestão de Energia , Hiperfagia , Animais , Ratos , Astrócitos/fisiologia , Tronco Encefálico/citologia , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato , Nervo Vago/fisiologia , Dieta Hiperlipídica
4.
J Physiol ; 601(14): 2853-2875, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37154244

RESUMO

Perinatal high-fat diet (pHFD) exposure alters the development of vagal neurocircuits that control gastrointestinal (GI) motility and reduce stress resiliency in offspring. Descending oxytocin (OXT; prototypical anti-stress peptide) and corticotropin releasing factor (CRF; prototypical stress peptide) inputs from the paraventricular nucleus (PVN) of the hypothalamus to the dorsal motor nucleus of the vagus (DMV) modulate the GI stress response. How these descending inputs, and their associated changes to GI motility and stress responses, are altered following pHFD exposure are, however, unknown. The present study used retrograde neuronal tracing experiments, cerebrospinal fluid extraction, in vivo recordings of gastric tone, motility and gastric emptying rates, and in vitro electrophysiological recordings from brainstem slice preparations to investigate the hypothesis that pHFD alters descending PVN-DMV inputs and dysregulates vagal brain-gut responses to stress. Compared to controls, rats exposed to pHFD had slower gastric emptying rates and did not respond to acute stress with the expected delay in gastric emptying. Neuronal tracing experiments demonstrated that pHFD reduced the number of PVNOXT neurons that project to the DMV, but increased PVNCRF neurons. Both in vitro electrophysiology recordings of DMV neurons and in vivo recordings of gastric motility and tone demonstrated that, following pHFD, PVNCRF -DMV projections were tonically active, and that pharmacological antagonism of brainstem CRF1 receptors restored the appropriate gastric response to brainstem OXT application. These results suggest that pHFD exposure disrupts descending PVN-DMV inputs, leading to a dysregulated vagal brain-gut response to stress. KEY POINTS: Maternal high-fat diet exposure is associated with gastric dysregulation and stress sensitivity in offspring. The present study demonstrates that perinatal high-fat diet exposure downregulates hypothalamic-vagal oxytocin (OXT) inputs but upregulates hypothalamic-vagal corticotropin releasing factor (CRF) inputs. Both in vitro and in vivo studies demonstrated that, following perinatal high-fat diet, CRF receptors were tonically active at NTS-DMV synapses, and that pharmacological antagonism of these receptors restored the appropriate gastric response to OXT. The current study suggests that perinatal high-fat diet exposure disrupts descending PVN-DMV inputs, leading to a dysregulated vagal brain-gut response to stress.


Assuntos
Hormônio Liberador da Corticotropina , Ocitocina , Gravidez , Feminino , Ratos , Animais , Ratos Sprague-Dawley , Dieta Hiperlipídica/efeitos adversos , Estômago/fisiologia , Motilidade Gastrointestinal , Nervo Vago/fisiologia
5.
Am J Physiol Gastrointest Liver Physiol ; 322(5): G513-G522, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35170350

RESUMO

Previous studies have shown that pharmacological manipulations with stress-related hormones such as corticotropin-releasing factor and thyrotropin-releasing hormone induce neuroplasticity in brainstem vagal neurocircuits, which modulate gastric tone and motility. The prototypical antistress hormone oxytocin (OXT) has been shown to modulate gastric tone and motility via vagal pathways, and descending hypothalamic oxytocinergic inputs play a major role in the vagally dependent gastric-related adaptations to stress. The aim of this study was to investigate the possible cellular mechanisms through which OXT modulates central vagal brainstem and peripheral enteric neurocircuits of male Sprague-Dawley rats in response to chronic repetitive stress. After chronic (5 consecutive days) of homotypic or heterotypic stress load, the response to exogenous brainstem administration of OXT was examined using whole cell patch-clamp recordings from gastric-projecting vagal motoneurons and in vivo recordings of gastric tone and motility. GABAergic currents onto vagal motoneurons were decreased by OXT in stressed, but not in naïve rats. In naïve rats, microinjections of OXT in vagal brainstem nuclei-induced gastroinhibition via peripheral release of nitric oxide (NO). In stressed rats, however, the OXT-induced gastroinhibition was determined by the release of both NO and vasoactive intestinal peptide (VIP). Taken together, our data indicate that stress induces neuroplasticity in the response to OXT in the neurocircuits, which modulate gastric tone and motility. In particular, stress uncovers the OXT-mediated modulation of brainstem GABAergic currents and alters the peripheral gastric response to vagal stimulation.NEW & NOTEWORTHY The prototypical antistress hormone, oxytocin (OXT), modulates gastric tone and motility via vagal pathways, and descending hypothalamic-brainstem OXT neurocircuits play a major role in the vagally dependent adaptation of gastric motility and tone to stress. The current study suggests that in the neurocircuits, which modulate gastric tone and motility, stress induces neuroplasticity in the response to OXT and may reflect the dysregulation observed in stress-exacerbated functional motility disorders.


Assuntos
Tronco Encefálico , Ocitocina , Estômago , Animais , Tronco Encefálico/fisiologia , Masculino , Plasticidade Neuronal , Ocitocina/farmacologia , Ratos , Ratos Sprague-Dawley , Estômago/fisiologia , Nervo Vago/fisiologia
6.
Adv Neonatal Care ; 21(6): 452-461, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34847103

RESUMO

BACKGROUND: Necrotizing enterocolitis (NEC) is the leading cause of death due to gastrointestinal disease in preterm neonates; yet, clinicians lack reliable and noninvasive predictive tools. PURPOSE: We aimed to test that diminished high-frequency heart rate variability (HF-HRV) and elevated levels of proinflammatory cytokines would have utility in NEC prediction. METHODS: In this multisite prospective study, we enrolled 250 preterm (26-34 weeks' postmenstrual age [PMA]) neonates with physiological stability at 72 hours of life. HRV was measured noninvasively using electrocardiograhic data from standardized cardiorespiratory monitors at postnatal week 1 of life and weekly thereafter until 35 weeks' PMA or discharge; blood was collected for cytokines at postnatal weeks 1 and 3. NEC was diagnosed via Modified Bell's Staging Criteria. RESULTS: HF-HRV was decreased at weeks 1 and 2 in neonates (47% females) who developed feeding intolerance or stage 2+ NEC. In addition, these neonates displayed elevated levels of IL-8 at week 1 and increased levels of IL-1ß, IL-6, TNF-α, and IL-8 at week 3 of life. Low HF-HRV was associated with elevated IL-6 or IL-8 levels at weeks 1 and 3 of life. Logistic regression indicated that only HF-HRV was a significant predictor of feeding intolerance or NEC development. IMPLICATIONS FOR PRACTICE AND RESEARCH: HRV is a promising noninvasive modality for NEC risk detection. The association of low HF-HRV with elevated proinflammatory cytokines provides evidence for a putative role of the vagal cholinergic pathway in NEC pathogenesis. Future studies should focus on application of these techniques to test clinical therapeutics.Video Abstract available at https://journals.lww.com/advancesinneonatalcare/Pages/videogallery.aspx?autoPlay=false&videoId=54.


Assuntos
Enterocolite Necrosante , Doenças do Recém-Nascido , Citocinas , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Estudos Prospectivos
7.
J Physiol ; 598(21): 4941-4955, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32864736

RESUMO

KEY POINTS: Stress triggers and exacerbates the symptoms of functional gastrointestinal disorders, such as delayed gastric emptying and impaired gastric motility. Understanding the mechanisms by which the neural circuits, impaired by stress, are restored may help to identify potential targets for more effective therapeutic interventions. Oxytocin administration or release ameliorates the stress-induced delayed gastric emptying and motility. However, is it unclear whether the effects are mediated via the hypothalamic-pituitary-adrenocortical axis or the oxytocinergic projections from the paraventricular nucleus of the hypothalamus to brainstem neurones of the dorsal vagal complex. We used Cre-inducible designer receptors exclusively activated by designer drugs to demonstrate the fundamental role of the oxytocinergic hypothalamic-vagal projections in the gastric adaptation to stress. ABSTRACT: Stress triggers and exacerbates the symptoms of functional gastrointestinal (GI) disorders, such as delayed gastric emptying and impaired gastric motility. The prototypical anti-stress hormone, oxytocin (OXT), plays a major role in the modulation of gastric emptying and motility following stress. It is not clear, however, whether the amelioration of dysregulated GI functions by OXT is mediated via an effect on the hypothalamic-pituitary-adrenocortical axis or the oxytocinergic projections from the paraventricular nucleus of the hypothalamus (PVN) to neurones of the dorsal vagal complex (DVC). In the present study we tested the hypothesis that the activity of hypothalamic-vagal oxytocinergic neurocircuits plays a major role in the gastric adaptation to stress. Cre-inducible designer receptors exclusively activated by designer drugs (DREADDs) were injected into the DVC of rats and retrogradely transported to allow selective expression in OXT neurones in the PVN. Following acute stress and either chronic heterotypic (CHe) or chronic homotypic (CHo) stress, gastric emptying was assessed via the [13 C]-octanoic acid breath test, and gastric tone and motility were assessed via strain gauges sewn on the surface of the stomach. Activation of the hypothalamic-vagal oxytocinergic neurocircuitry, by DREADD agonist clozapine-N-oxide (CNO), prevented the delayed gastric emptying observed following acute or CHe stress, and 4th ventricular administration of CNO increased gastric tone and motility. Conversely, CNO-mediated inhibition of the hypothalamic-vagal oxytocinergic neurocircuitry prevented the CHo-induced adaptation in gastric emptying, and an increase in gastric tone and motility. Taken together, the data support the hypothesis that hypothalamic-vagal oxytocinergic neurocircuits play a major role in the modulation of gastric emptying and motility following stress.


Assuntos
Esvaziamento Gástrico , Nervo Vago , Animais , Motilidade Gastrointestinal , Hipotálamo , Ocitocina , Núcleo Hipotalâmico Paraventricular , Ratos
8.
J Neurophysiol ; 121(5): 1856-1864, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30917059

RESUMO

Parkinson's disease (PD) is predominantly idiopathic in origin, and a large body of evidence indicates that gastrointestinal (GI) dysfunctions are a significant comorbid clinical feature; these dysfunctions include dysphagia, nausea, delayed gastric emptying, and severe constipation, all of which occur commonly before the onset of the well-known motor symptoms of PD. Based on a distinct distribution pattern of Lewy bodies (LB) in the enteric nervous system (ENS) and in the preganglionic neurons of the dorsal motor nucleus of the vagus (DMV), and together with the early onset of GI symptoms, it was suggested that idiopathic PD begins in the ENS and spreads to the central nervous system (CNS), reaching the DMV and the substantia nigra pars compacta (SNpc). These two areas are connected by a recently discovered monosynaptic nigro-vagal pathway, which is dysfunctional in rodent models of PD. An alternative hypothesis downplays the role of LB transport through the vagus nerve and proposes that PD pathology is governed by regional or cell-restricted factors as the leading cause of nigral neuronal degeneration. The purpose of this brief review is to summarize the neuronal electrophysiological findings in the SNpc and DMV in PD.


Assuntos
Doença de Parkinson/fisiopatologia , Substância Negra/fisiopatologia , Transmissão Sináptica , Animais , Dopamina/metabolismo , Humanos , Doença de Parkinson/metabolismo , Substância Negra/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 317(4): G476-G483, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31393788

RESUMO

Functional gastrointestinal disorders, including delayed gastric emptying and decreased gastric motility, are more prevalent in women, suggesting a potential role for circulating gonadal hormones, including estrogen. Gastric motility is tuned by the vagal inputs arising from the dorsal motor nucleus of the vagus (DMV), which is itself controlled by tonic GABAergic inputs. Estrogen increases GABA functions in various central nervous system areas; however, the effect of the estrus cycle in modulating GABAergic inputs onto DMV neurons, hence vagal control of gastric motility, has not been investigated. The aim of the present study was to test the hypothesis that GABAergic tone to DMV neurons, hence the vagal output to the stomach, varies according to sex and the estrus cycle. Experiments were performed on age-matched Sprague-Dawley male and virgin female rats; females were subdivided according to the high-estrogen (HE) or low-estrogen (LE) period of their cycle. Whole-cell patch-clamp recordings were made from gastric-projecting DMV neurons, and the response to perfusion with the GABAA receptor antagonist bicuculline was examined. The response of corpus and antrum tone and motility to bicuculline microinjected in the dorsal vagal complex, recorded via strain gauges sewn to the anterior gastric surface, was also assessed. Bicuculline increased the firing rate of DMV neurons, as well as gastric tone and motility, to a larger extent in HE compared with LE or male rats, suggesting a higher GABAergic tone in HE female rats. Taken together, the data support the hypothesis that GABAergic tone to DMV neurons varies according to sex and estrus cycle.NEW & NOTEWORTHY GABAergic neurotransmission to the dorsal motor nucleus of the vagus (DMV) plays a pivotal role in the modulation of gastric tone and motility. Gastric motility is reduced in women and may contribute to the higher incidence of functional gastrointestinal disorders. In the present study, we report that GABAergic tone to rat DMV neurons, hence vagal output to the stomach, varies according to sex and estrus cycle, and the GABAergic tone is increased during the high-estrogen period of the estrus cycle.


Assuntos
Transmissão Sináptica/fisiologia , Nervo Vago/fisiologia , Ácido gama-Aminobutírico/fisiologia , Animais , Bicuculina/farmacologia , Estrogênios/metabolismo , Ciclo Estral , Feminino , Antagonistas GABAérgicos/farmacologia , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/fisiologia , Masculino , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Caracteres Sexuais , Estômago/inervação
10.
Am J Physiol Gastrointest Liver Physiol ; 317(1): G1-G7, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31042398

RESUMO

The majority of patients with Parkinson's disease (PD) experience gastrointestinal dysfunction. Recently, we described a nigro-vagal pathway that uses dopaminergic (DA) inputs to the dorsal motor nucleus of the vagus (DMV) and A2 area neurons to modulate gastric motility and tone. This pathway is disrupted in a rodent model of PD. The aim of the present study was to test the hypothesis that brain-stem DA modulation of gastric tone and motility is altered in a rodent model of PD. Male Sprague-Dawley rats received three weekly intraperitoneal injections of paraquat (10 mg/kg) or saline (control). In naive conditions, microinjection of DA into the DMV induced a gastroinhibitory response in 100% of animals. In 19 of 28 PQ-treated animals, however, microinjection of DA into the DVC induced a biphasic response, with an initial increase in gastric tone and motility followed by a profound gastroinhibition. The excitatory response to DA microinjection was attenuated by a combination of DA type 1 (DA1)- and DA2-like receptor antagonists. Conversely, the inhibitory response was reduced by the DA2-like receptor antagonist only. Pretreatment with the α2-adrenoceptor antagonist yohimbine did not modulate the response to DA, thus excluding involvement of the A2 area. At the end of the experiments, induction of the Parkinson phenotype was confirmed by the presence of α-synuclein immunoreactivity in the DMV and substantia nigra pars compacta. These data suggest a maladaptive neural plasticity in brain-stem vagal circuits regulating gastric motility in PQ-treated rats that may be responsible for the gastric dysfunction observed in PD models. NEW & NOTEWORTHY After paraquat treatment and induction of Parkinson's disease, brain-stem dopamine (DA) application induces a biphasic gastric response in the majority of rats, with an initial increase in tone and motility followed by gastroinhibition. The initial increase in gastric tone and motility is mediated via a combined activation of DA type 1 (DA1)- and DA2-like receptors. The inhibitory effects of DA are mediated by DA2-like receptors and are not affected by blockade of adrenergic inputs mediated by α2-adrenoceptors.


Assuntos
Tronco Encefálico/metabolismo , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Motilidade Gastrointestinal , Plasticidade Neuronal , Transtornos Parkinsonianos/metabolismo , Estômago/inervação , Nervo Vago/metabolismo , Animais , Tronco Encefálico/fisiopatologia , Modelos Animais de Doenças , Masculino , Inibição Neural , Paraquat , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/fisiopatologia , Ratos Sprague-Dawley , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Nervo Vago/fisiopatologia
11.
Am J Physiol Gastrointest Liver Physiol ; 317(2): G203-G209, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31241977

RESUMO

Symptoms of functional gastrointestinal disorders (FGIDs), including fullness, bloating, abdominal pain, and altered gastrointestinal (GI) motility, present a significant clinical problem, with a reported prevalence of 25%-40% within the general population. More than 60% of those affected seek and require healthcare, and affected individuals report a significantly decreased quality of life. FGIDs are highly correlated with episodes of acute and chronic stress and are increased in prevalence and reported severity in women compared with men. Although there is evidence that sex and stress interact to exacerbate FGID symptoms, the physiological mechanisms that mediate these sex-dependent disparities are incompletely understood, although hormonal-related differences in GI motility and visceral sensitivity have been purported to play a significant role in the etiology. In this mini review, we will discuss brain-gut axis control of GI motility and sensitivity, the influence of estrogen on GI motility and sensitivity, and stress modulation of the brain-gut axis.


Assuntos
Encéfalo/metabolismo , Estrogênios/metabolismo , Gastroenteropatias , Trato Gastrointestinal , Estresse Psicológico/fisiopatologia , Gastroenteropatias/metabolismo , Gastroenteropatias/psicologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/fisiopatologia , Humanos
12.
Am J Physiol Gastrointest Liver Physiol ; 317(1): G40-G50, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31042399

RESUMO

Perinatal high-fat diet (pHFD) exposure increases the inhibition of dorsal motor nucleus of the vagus (DMV) neurons, potentially contributing to the dysregulation of gastric functions. The aim of this study was to test the hypothesis that pHFD increases the inhibition of DMV neurons by disrupting GABAA receptor subunit development. In vivo gastric recordings were made from adult anesthetized Sprague-Dawley rats fed a control or pHFD (14 or 60% kcal from fat, respectively) from embryonic day 13 (E13) to postnatal day 42 (P42), and response to brainstem microinjection of benzodiazepines was assessed. Whole cell patch clamp recordings from DMV neurons assessed the functional expression of GABAA α subunits, whereas mRNA and protein expression were measured via qPCR and Western blotting, respectively. pHFD decreased basal antrum and corpus motility, whereas brainstem microinjection of L838,417 (positive allosteric modulator of α2/3 subunit-containing GABAA receptors) produced a larger decrease in gastric tone and motility. GABAergic miniature inhibitory postsynaptic currents in pHFD DMV neurons were responsive to L838,417 throughout development, unlike control DMV neurons, which were responsive only at early postnatal timepoints. Brainstem mRNA and protein expression of the GABAA α1,2, and 3 subunits, however, did not differ between control and pHFD rats. This study suggests that pHFD exposure arrests the development of synaptic GABAA α2/3 receptor subunits on DMV neurons and that functional synaptic expression is maintained into adulthood, although cellular localization may differ. The tonic activation of slower GABAA α2/3 subunit-containing receptors implies that such developmental changes may contribute to the observed decreased gastric motility. NEW & NOTEWORTHY Vagal neurocircuits involved in the control of gastric functions, satiation, and food intake are subject to significant developmental regulation postnatally, with immature GABAA receptors expressing slower α2/3-subunits, whereas mature GABAA receptor express faster α1-subunits. After perinatal high-fat diet exposure, this developmental regulation of dorsal motor nucleus of the vagus (DMV) neurons is disrupted, increasing their tonic GABAergic inhibition, decreasing efferent output, and potentially decreasing gastric motility.


Assuntos
Tronco Encefálico/metabolismo , Dieta Hiperlipídica , Motilidade Gastrointestinal , Efeitos Tardios da Exposição Pré-Natal , Receptores de GABA-A/metabolismo , Estômago/inervação , Nervo Vago/metabolismo , Fatores Etários , Fenômenos Fisiológicos da Nutrição Animal , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Potenciais Pós-Sinápticos Inibidores , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Potenciais Pós-Sinápticos em Miniatura , Inibição Neural , Gravidez , Ratos Sprague-Dawley , Receptores de GABA-A/genética
13.
Am J Perinatol ; 36(5): 505-510, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30193382

RESUMO

OBJECTIVE: An estimation of the individual's ability to cope with environmental adversity, that is, stress resiliency, can be extrapolated by measuring cardiac vagal tone, that is, high-frequency heart rate variability (HF-HRV); indeed, higher HF-HRV is associated with health and developmental advantages for preterm neonates. Previous studies show skin-to-skin contact (SSC) improves stress resiliency; however, linkages between SSC and HF-HRV on outcomes have not been assessed. We aimed to test the hypothesis that increased SSC frequency would enhance HF-HRV, reduce neonatal morbidity, and improve developmental outcomes. STUDY DESIGN: Weekly electrocardiograms and clinical data were obtained from 101 preterm neonates. SSC frequency was determined from the electronic medical record. RESULTS: At postnatal week 1, frequency of SSC and HF-HRV were positively correlated (p =.02); further, multiple stepwise regressions showed higher HF-HRV and SSC predicted reduced days on ventilation and oxygen, and shorter hospital stay (p < 0.001). Higher HF-HRV predicted lower postmenstrual age (PMA) at discharge (p < 0.01). CONCLUSION: Higher SSC frequency was associated with increased HF-HRV during the first postnatal week. SSC and HF-HRV uniquely predicted diminished neonatal morbidity throughout hospitalization. Additionally, HF-HRV uniquely predicted earlier PMA at discharge. Augmenting SSC early in life enhances stress resiliency and improves health outcomes.


Assuntos
Frequência Cardíaca/fisiologia , Recém-Nascido Prematuro/fisiologia , Método Canguru , Nervo Vago/fisiologia , Eletrocardiografia , Feminino , Humanos , Recém-Nascido , Masculino , Estudos Prospectivos , Análise de Regressão
14.
Am J Physiol Gastrointest Liver Physiol ; 315(1): G20-G26, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29597355

RESUMO

The last few decades have seen a major increase in the number of neurotransmitters and neuropeptides recognized as playing a role in brain stem neurocircuits, including those involved in homeostatic functions such as stress responsiveness, gastrointestinal motility, feeding, and/or arousal/wakefulness. This minireview will focus on the known physiological role of three of these novel neuropeptides, i.e., apelin, nesfatin-1, and neuropeptide-S, with a special emphasis on their hypothetical roles in vagal signaling related to gastrointestinal motor functions.


Assuntos
Tronco Encefálico/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação a DNA/metabolismo , Motilidade Gastrointestinal/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Nervo Vago/fisiologia , Humanos , Nucleobindinas
15.
Am J Physiol Gastrointest Liver Physiol ; 314(4): G504-G516, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351390

RESUMO

Chronic stress exerts vagally dependent effects to disrupt gastric motility; previous studies have shown that, among other nuclei, A2 neurons are involved in mediating these effects. Several studies have also shown robust in vitro and in vivo effects of α2-adrenoceptor agonists on vagal motoneurons. We have demonstrated previously that brainstem vagal neurocircuits undergo remodeling following acute stress; however, the effects following brief periods of chronic stress have not been investigated. Our aim, therefore, was to test the hypothesis that different types of chronic stress influence gastric tone and motility by inducing plasticity in the response of vagal neurocircuits to α2-adrenoreceptor agonists. In rats that underwent 5 days of either homotypic or heterotypic stress loading, we applied the α2-adrenoceptor agonist, UK14304, either by in vitro brainstem perfusion to examine its ability to modulate GABAergic synaptic inputs to vagal motoneurons or in vivo brainstem microinjection to observe actions to modulate antral tone and motility. In neurons from naïve rats, GABAergic currents were unresponsive to exogenous application of UK14304. In contrast, GABAergic currents were inhibited by UK14304 in all neurons from homotypic and, in a subpopulation of neurons, heterotypic stressed rats. In control rats, UK14304 microinjection inhibited gastric tone and motility via withdrawal of vagal cholinergic tone; in heterotypic stressed rats, the larger inhibition of antrum tone was due to a concomitant activation of peripheral nonadrenergic, noncholinergic pathways. These data suggest that stress induces plasticity in brainstem vagal neurocircuits, leading to an upregulation of α2-mediated responses. NEW & NOTEWORTHY Catecholaminergic neurons of the A2 area play a relevant role in stress-related dysfunction of the gastric antrum. Brief periods of chronic stress load induce plastic changes in the actions of adrenoceptors on vagal brainstem neurocircuits.


Assuntos
Tronco Encefálico/metabolismo , Motilidade Gastrointestinal , Neurônios Motores , Estômago , Nervo Vago/efeitos dos fármacos , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Catecolaminas/metabolismo , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/fisiologia , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa/metabolismo , Estômago/inervação , Estômago/fisiologia , Estresse Fisiológico/fisiologia
16.
Am J Physiol Gastrointest Liver Physiol ; 314(5): G623-G634, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29368945

RESUMO

Obesity is associated with dysregulation of vagal neurocircuits controlling gastric functions, including food intake and energy balance. In the short term, however, caloric intake is regulated homeostatically although the precise mechanisms responsible are unknown. The present study examined the effects of acute high-fat diet (HFD) on glutamatergic neurotransmission within central vagal neurocircuits and its effects on gastric motility. Sprague-Dawley rats were fed a control or HFD diet (14% or 60% kcal from fat, respectively) for 3-5 days. Whole cell patch-clamp recordings and brainstem application of antagonists were used to assess the effects of acute HFD on glutamatergic transmission to dorsal motor nucleus of the vagus (DMV) neurons and subsequent alterations in gastric tone and motility. After becoming hyperphagic initially, caloric balance was restored after 3 days following HFD exposure. In control rats, the non- N-methyl-d-aspartate (NMDA) receptor antagonist, 6,7-dinitroquinoxaline-2,3-dione (DNQX), but not the NMDA receptor antagonist, amino-5-phosphonopentanoate (AP5), significantly decreased excitatory synaptic currents and action potential firing rate in gastric-projecting DMV neurons. In contrast, both AP5 and DNQX decreased excitatory synaptic transmission and action potential firing in acute HFD neurons. When microinjected into the brainstem, AP5, but not DNQX, decreased gastric motility and tone in acute HFD rats only. These results suggest that acute HFD upregulates NMDA receptor-mediated currents, increasing DMV neuronal excitability and activating the vagal efferent cholinergic pathway, thus increasing gastric tone and motility. Although such neuroplasticity may be a persistent adaptation to the initial exposure to HFD, it may also be an important mechanism in homeostatic regulation of energy balance. NEW & NOTEWORTHY Vagal neurocircuits are critical to the regulation of gastric functions, including satiation and food intake. Acute high-fat diet upregulates glutamatergic signaling within central vagal neurocircuits via activation of N-methyl-d-aspartate receptors, increasing vagal efferent drive to the stomach. Although it is possible that such neuroplasticity is a persistent adaptation to initial exposure to the high-fat diet, it may also play a role in the homeostatic control of feeding.


Assuntos
Dieta Hiperlipídica/métodos , Esvaziamento Gástrico , Ácido Glutâmico/metabolismo , Quinoxalinas , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Estômago , Nervo Vago , Potenciais de Ação , Animais , Tronco Encefálico/metabolismo , Tronco Encefálico/fisiologia , Antagonistas de Aminoácidos Excitatórios/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Esvaziamento Gástrico/efeitos dos fármacos , Esvaziamento Gástrico/fisiologia , Plasticidade Neuronal/fisiologia , Quinoxalinas/metabolismo , Quinoxalinas/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Estômago/inervação , Estômago/fisiologia , Transmissão Sináptica/fisiologia , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiologia
17.
Am J Physiol Gastrointest Liver Physiol ; 314(2): G201-G210, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29025730

RESUMO

The expression of apelin and its receptors (APJ) in central autonomic networks suggests that apelin may regulate gastrointestinal motor functions. In rodents, central administration of apelin-13 has been shown to inhibit gastric emptying; however, the mechanisms involved remain to be determined. Using male adult Sprague-Dawley rats, the aims of the present study were 1) to determine the expression of APJ receptor in the dorsal vagal complex (DVC), 2) to assess the effects of central application of apelin-13 into the DVC on gastric tone and motility, and 3) to investigate the neuronal pathways responsible for apelin-induced alterations. APJ receptor immunoreactivity was detected in gastric-projecting and choline acetyltransferase-positive neurons of the DVC. Microinjection of apelin-13 into the DVC significantly decreased gastric tone and motility in both corpus and antrum. The apelin-induced reduction in gastric tone and motility was prevented by surgical vagotomy or fourth ventricular application of the APJ receptor antagonist, [Ala13]apelin-13 (F13A). Systemic administration of the muscarinic receptor antagonist atropine, but not the nitric oxide synthase inhibitor nitro-l-arginine methyl ester (l-NAME), abolished the apelin-induced inhibitory responses. The present results indicate a central modulatory role of apelin in the vagal neurocircuitry that controls gastric motor functions via withdrawal of the tonically active cholinergic pathway. NEW & NOTEWORTHY This is the first study investigating the effects induced by brain stem application of apelin-13 while monitoring gastric tone and motility in rats. We have found that gastric-projecting neurons of the dorsal vagal complex express apelin receptors (APJ), which mediate the inhibitory actions of apelin-13. The inhibitory effects of apelin were abolished by systemic preadministration of atropine, but not nitro-l-arginine methyl ester (l-NAME). Apelin seems to modulate gastric motility via withdrawal of the tonically active vagal cholinergic pathway.


Assuntos
Acetilcolina/metabolismo , Tronco Encefálico/efeitos dos fármacos , Fibras Colinérgicas/efeitos dos fármacos , Motilidade Gastrointestinal/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Estômago/inervação , Nervo Vago/efeitos dos fármacos , Animais , Receptores de Apelina/agonistas , Receptores de Apelina/metabolismo , Tronco Encefálico/metabolismo , Colina O-Acetiltransferase/metabolismo , Fibras Colinérgicas/metabolismo , Relação Dose-Resposta a Droga , Masculino , Microinjeções , Antagonistas Muscarínicos/farmacologia , Ratos Sprague-Dawley , Vagotomia , Nervo Vago/metabolismo
18.
Gastroenterology ; 153(6): 1581-1593, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28912019

RESUMO

BACKGROUND & AIMS: In most patients with Parkinson's disease, gastrointestinal (GI) dysfunctions, such as gastroparesis and constipation, are prodromal to the cardinal motor symptoms of the disease. Sporadic Parkinson's disease has been proposed to develop after ingestion of neurotoxicants that affect the brain-gut axis via the vagus nerve, and then travel to higher centers, compromising the substantia nigra pars compacta (SNpc) and, later, the cerebral cortex. We aimed to identify the pathway that connects the brainstem vagal nuclei and the SNpc, and to determine whether this pathway is compromised in a rat model of Parkinsonism. METHODS: To study this neural pathway in rats, we placed tracers in the dorsal vagal complex or SNpc; brainstem and midbrain were examined for tracer distribution and neuronal neurochemical phenotype. Rats were given injections of paraquat once weekly for 3 weeks to induce features of Parkinsonism, or vehicle (control). Gastric tone and motility were recorded after N-methyl-d-aspartate microinjection in the SNpc and/or optogenetic stimulation of nigro-vagal terminals in the dorsal vagal complex. RESULTS: Stimulation of the SNpc increased gastric tone and motility via activation of dopamine 1 receptors in the dorsal vagal complex. In the paraquat-induced model of Parkinsonism, this nigro-vagal pathway was compromised during the early stages of motor deficit development. CONCLUSIONS: We identified and characterized a nigro-vagal monosynaptic pathway in rats that controls gastric tone and motility. This pathway might be involved in the prodromal gastric dysmotility observed in patients with early-stage Parkinson's disease.


Assuntos
Tronco Encefálico/fisiopatologia , Esvaziamento Gástrico , Doença de Parkinson Secundária/fisiopatologia , Parte Compacta da Substância Negra/fisiopatologia , Estômago/inervação , Nervo Vago/fisiopatologia , Animais , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/metabolismo , Modelos Animais de Doenças , Esvaziamento Gástrico/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Atividade Motora , Vias Neurais/metabolismo , Vias Neurais/fisiopatologia , Técnicas de Rastreamento Neuroanatômico , Neurotransmissores/farmacologia , Optogenética , Paraquat , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/metabolismo , Parte Compacta da Substância Negra/efeitos dos fármacos , Parte Compacta da Substância Negra/metabolismo , Ratos Sprague-Dawley , Receptores de Dopamina D1/metabolismo , Fatores de Tempo , Nervo Vago/efeitos dos fármacos , Nervo Vago/metabolismo
19.
Adv Neonatal Care ; 18(1): 49-57, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29261561

RESUMO

BACKGROUND: While biological and behavioral stress response systems are intact in early gestation, preterm infants' behaviors are often more subtle and difficult to interpret compared with full-term infants. They are also more vulnerable for regulatory issues (ie, colic) that are known to impact caregiver-infant interactions. Biobehavioral measures such as behavioral responsivity and heart rate variability (HRV), particularly cardiac vagal tone, may help elucidate preterm infants' stress/regulatory systems. PURPOSE: To test the hypotheses that preterm infants' consoling behaviors and high-frequency (HF) HRV in the first week of life are significantly associated and they are inverse correlates of future colic risk. METHODS/SEARCH STRATEGY: Thirty preterm (mean ± SE = 32.7 ± 0.3 weeks postmenstrual age [PMA]) infants underwent direct NIDCAP (Newborn Individualized Development and Assessment Program) observation during routine care and had HRV measurements during their first week postbirth. Sixty-three percent of mothers completed the Infant Colic Scale at 6 to 8 weeks adjusted postnatal age. Nonparametric tests were used to determine associations among behaviors, HRV, and maternal perceptions of infant colic. FINDINGS/RESULTS: Self-consoling behaviors were positively associated with HF-HRV (vagal tone). In addition, stress behaviors were positively associated with low-frequency/high-frequency HRV (sympathetic dominance). Infants who displayed more stress behaviors also demonstrated more self-consoling behaviors. No significant associations were found with colic. IMPLICATIONS FOR PRACTICE: HF-HRV provides information on the infant's capacity to modulate stress and is a useful, noninvasive measure when behaviors are more difficult to discern. IMPLICATIONS FOR RESEARCH: Further study in a larger sample is needed to determine whether behavioral stress measures and HF-HRV may be useful to determine colic risk.


Assuntos
Sistema Nervoso Autônomo/fisiopatologia , Cólica , Frequência Cardíaca/fisiologia , Comportamento do Lactente/fisiologia , Comportamento Materno , Cólica/diagnóstico , Cólica/fisiopatologia , Cólica/psicologia , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro/fisiologia , Recém-Nascido Prematuro/psicologia , Masculino , Relações Mãe-Filho , Exame Físico/métodos , Prognóstico , Reprodutibilidade dos Testes , Medição de Risco , Índice de Gravidade de Doença
20.
J Neurophysiol ; 116(4): 1705-1714, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27440241

RESUMO

Prior immunohistochemical studies have demonstrated that at early postnatal time points, central vagal neurons receive both glycinergic and GABAergic inhibitory inputs. Functional studies have demonstrated, however, that adult vagal efferent motoneurons receive only inhibitory GABAergic synaptic inputs, suggesting loss of glycinergic inhibitory neurotransmission during postnatal development. The purpose of the present study was to test the hypothesis that the loss of glycinergic inhibitory synapses occurs in the immediate postnatal period. Whole cell patch-clamp recordings were made from dorsal motor nucleus of the vagus (DMV) neurons from postnatal days 1-30, and the effects of the GABAA receptor antagonist bicuculline (1-10 µM) and the glycine receptor antagonist strychnine (1 µM) on miniature inhibitory postsynaptic current (mIPSC) properties were examined. While the baseline frequency of mIPSCs was not altered by maturation, perfusion with bicuculline either abolished mIPSCs altogether or decreased mIPSC frequency and decay constant in the majority of neurons at all time points. In contrast, while strychnine had no effect on mIPSC frequency, its actions to increase current decay time declined during postnatal maturation. These data suggest that in early postnatal development, DMV neurons receive both GABAergic and glycinergic synaptic inputs. Glycinergic neurotransmission appears to decline by the second postnatal week, and adult neurons receive principally GABAergic inhibitory inputs. Disruption of this developmental switch from GABA-glycine to purely GABAergic transmission in response to early life events may, therefore, lead to adverse consequences in vagal efferent control of visceral functions.


Assuntos
Potenciais Pós-Sinápticos Inibidores/fisiologia , Bulbo/crescimento & desenvolvimento , Bulbo/metabolismo , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Neurônios/metabolismo , Animais , Animais Recém-Nascidos , Bicuculina/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Glicinérgicos/farmacologia , Imuno-Histoquímica , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Bulbo/citologia , Potenciais Pós-Sinápticos em Miniatura/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Ratos Sprague-Dawley , Receptores de GABA-A/metabolismo , Receptores de Glicina/antagonistas & inibidores , Receptores de Glicina/metabolismo , Estricnina/farmacologia , Técnicas de Cultura de Tecidos , Nervo Vago/citologia , Nervo Vago/crescimento & desenvolvimento , Nervo Vago/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa