Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Gastrointest Liver Physiol ; 320(2): G183-G192, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33206550

RESUMO

Gastric vagal afferents (GVAs) sense food-related mechanical stimuli and signal to the central nervous system, to integrate control of meal termination. Pregnancy is characterized by increased maternal food intake, which is essential for normal fetal growth and to maximize progeny survival and health. However, it is unknown whether GVA function is altered during pregnancy to promote food intake. This study aimed to determine the mechanosensitivity of GVAs and food intake during early, mid-, and late stages of pregnancy in mice. Pregnant mice consumed more food compared with nonpregnant mice, notably in the light phase during mid- and late pregnancy. The increased food intake was predominantly due to light-phase increases in meal size across all stages of pregnancy. The sensitivity of GVA tension receptors to gastric distension was significantly attenuated in mid- and late pregnancy, whereas the sensitivity of GVA mucosal receptors to mucosal stroking was unchanged during pregnancy. To determine whether pregnancy-associated hormonal changes drive these adaptations, the effects of estradiol, progesterone, prolactin, and growth hormone on GVA tension receptor mechanosensitivity were determined in nonpregnant female mice. The sensitivity of GVA tension receptors to gastric distension was augmented by estradiol, attenuated by growth hormone, and unaffected by progesterone or prolactin. Together, the data indicate that the sensitivity of GVA tension receptors to tension is reduced during pregnancy, which may attenuate the perception of gastric fullness and explain increased food intake. Further, these adaptations may be driven by increases in maternal circulating growth hormone levels during pregnancy.NEW & NOTEWORTHY This study provides first evidence that gastric vagal afferent signaling is attenuated during pregnancy and inversely associated with meal size. Growth hormone attenuated mechanosensitivity of gastric vagal afferents, adding support that increases in maternal growth hormone may mediate adaptations in gastric vagal afferent signaling during pregnancy. These findings have important implications for the peripheral control of food intake during pregnancy.


Assuntos
Vias Aferentes/fisiologia , Plasticidade Neuronal/fisiologia , Estômago/inervação , Nervo Vago/fisiologia , Animais , Feminino , Camundongos , Gravidez
2.
Neurogastroenterol Motil ; 31(12): e13711, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31509314

RESUMO

BACKGROUND: Gastric vagal afferents (GVAs) respond to mechanical stimulation, initiating satiety. These afferents exhibit diurnal fluctuations in mechanosensitivity, facilitating food intake during the dark phase in rodents. In humans, desynchrony of diurnal rhythms (eg, shift work) is associated with a higher risk of obesity. To test the hypothesis that shift work disrupts satiety signaling, the effect of a rotating light cycles on diurnal rhythms in GVA mechanosensitivity in lean and high-fat diet (HDF)-induced obese mice was determined. METHODS: Male C57BL/6 mice were fed standard laboratory diet (SLD) or HFD for 12 weeks. After 4 weeks, mice were randomly allocated to a normal light (NL; 12 hour light: 12 hour dark; lights on at zeitgeber time [ZT] 0) or rotating light (RL; 3-day NL cycle, 4-day reversed light cycle [lights on: ZT12] repeated) cycle for 8 weeks. At week 12, eight mice from each group were housed in metabolic cages. After 12 weeks, ex vivo GVA recordings were taken at 3 hour intervals starting at ZT0. KEY RESULTS: SLD-RL and HFD-RL gained more weight compared to SLD-NL and HFD-NL mice, respectively. Gonadal fat pad mass was higher in SLD-RL compared to SLD-NL mice. In SLD-NL mice, tension and mucosal receptor mechanosensitivity exhibited diurnal rhythms with a peak at ZT9. These rhythms were lost in SLD-RL, HFD-NL, and HFD-RL mice and associated with dampened diurnal rhythms in food intake. CONCLUSIONS & INFERENCES: GVA diurnal rhythms are susceptible to disturbances in the light cycle and/or the obese state. This may underpin the observed changes in feeding behavior.


Assuntos
Ritmo Circadiano/fisiologia , Comportamento Alimentar/fisiologia , Mecanorreceptores/fisiologia , Resposta de Saciedade/fisiologia , Estômago/inervação , Nervo Vago/fisiopatologia , Adiposidade , Vias Aferentes/fisiopatologia , Animais , Peso Corporal , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Ingestão de Energia , Mucosa Gástrica/inervação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/fisiopatologia , Fotoperíodo , Distribuição Aleatória , Jornada de Trabalho em Turnos , Tolerância ao Trabalho Programado
3.
J Physiol ; 597(6): 1503-1515, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30605228

RESUMO

KEY POINTS: Tenascin X (TNX) functions in the extracellular matrix of skin and joints where it maintains correct intercellular connections and tissue architecture TNX is associated exclusively with vagal-afferent endings and some myenteric neurones in mouse and human stomach, respectively. TNX-deficient mice have accelerated gastric emptying and hypersensitivity of gastric vagal mechanoreceptors that can be normalized by an inhibitor of vagal-afferent sensitivity. Cultured nodose ganglion neurones showed no changes in response to capsaicin, cholecystokinin and potassium chloride in TNX-deficient mice. TNX-deficient patients have upper gastric dysfunction consistent with those in a mouse model. Our translational studies suggest that abnormal gastric sensory function may explain the upper gut symptoms present in TNX deficient patients, thus making it important to study gastric physiology. TNX deficiency should be evaluated routinely in patients with connective tissue abnormalities, which will enable a better understanding of its role and allow targeted treatment. For example, inhibitors of vagal afferents-baclofen could be beneficial in patients. These hypotheses need confirmation via targeted clinical trials. ABSTRACT: Tenascin-X (TNX) is a glycoprotein that regulates tissue structure via anti-adhesive interactions with collagen in the extracellular matrix. TNX deficiency causes a phenotype similar to hypermobility Ehlers-Danlos syndrome involving joint hypermobility, skin hyperelasticity, pain and gastrointestinal dysfunction. Previously, we have shown that TNX is required for neural control of the bowel by a specific subtype of mainly cholinergic enteric neurones and regulates sprouting and sensitivity of nociceptive sensory endings in mouse colon. These findings correlate with symptoms shown by TNX-deficient patients and mice. We aimed to identify whether TNX is similarly present in neural structures found in mouse and human gastric tissue. We then determined whether TNX has a functional role, specifically in gastric motor and sensory function and nodose ganglia neurones. We report that TNX was present in calretinin-immunoreactive extrinsic nerve endings in mouse and human stomach. TNX deficient mice had accelerated gastric emptying and markedly increased vagal afferent responses to gastric distension that could be rescued with GABAB receptor agonist. There were no changes in nodose ganglia excitability in TNX deficient mice, suggesting that vagal afferent responses are probably the result of altered peripheral mechanosensitivity. In TNXB-deficient patients, significantly greater symptoms of reflux, indigestion and abdominal pain were reported. In the present study, we report the first role for TNX in gastric function. Further studies are required in TNX deficient patients to determine whether symptoms can be relieved using GABAB agonists.


Assuntos
Síndrome de Ehlers-Danlos/genética , Esvaziamento Gástrico , Estômago/fisiologia , Tenascina/genética , Animais , Células Cultivadas , Síndrome de Ehlers-Danlos/fisiopatologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Neurônios Aferentes/metabolismo , Neurônios Aferentes/fisiologia , Gânglio Nodoso/citologia , Gânglio Nodoso/metabolismo , Gânglio Nodoso/fisiologia , Estômago/fisiopatologia , Tenascina/metabolismo , Nervo Vago/metabolismo , Nervo Vago/fisiologia
4.
Am J Physiol Gastrointest Liver Physiol ; 315(6): G932-G942, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30188750

RESUMO

Processes involved in regulation of energy balance and intermediary metabolism are aligned to the light-dark cycle. Shift-work and high-fat diet (HFD)-induced obesity disrupt circadian rhythmicity and are associated with increased risk of nonalcoholic fatty liver disease. This study aimed to determine the effect of simulating shift work on hepatic lipid accumulation in lean and HFD mice. C57BL/6 mice fed a standard laboratory diet (SLD) or HFD for 4 wk were further allocated to a normal light (NL) cycle (lights on: 0600-1800) or rotating light (RL) cycle [3 days NL and 4 days reversed (lights on: 1800-0600) repeated] for 8 wk. Tissue was collected every 3 h beginning at 0600. HFD mice gained more weight than SLD mice, and RL mice gained more weight than NL mice. SLD-NL and HFD-NL mice, but not RL mice, were more active, had higher respiratory quotients, and consumed/expended more energy during the dark phase compared with the light phase. Blood glucose and plasma cholesterol and triglyceride concentrations were elevated in HFD and SLD-RL compared with SLD-NL mice. Hepatic glycogen was elevated in HFD compared with SLD mice. Hepatic triglycerides were elevated in SLD-RL and HFD mice compared with SLD-NL. Circadian rhythmicity of hepatic acetyl-CoA carboxylase (ACACA) mRNA was phase shifted in SLD-RL and HFD-NL and lost in HFD-RL mice. Hepatic ACACA protein was reduced in SLD-RL and HFD mice compared with SLD-NL mice. Hepatic adipose triglyceride lipase was elevated in HFD-NL compared with SLD-NL but lower in RL mice compared with NL mice irrespective of diet. In conclusion, an RL cycle model of shift work promotes weight gain and hepatic lipid storage even in lean conditions. NEW & NOTEWORTHY In this publication we describe the effects of a rotating light cycle model of shift work in lean and high-fat diet-induced obese mice on body mass, diurnal patterns of energy intake and expenditure, and hepatic lipid storage. The data indicate that modeling shift work, via a rotating light cycle, promotes weight gain and hepatic lipid accumulation even in mice on a standard laboratory diet.


Assuntos
Ritmo Circadiano , Glicogênio/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Aumento de Peso , Acetil-CoA Carboxilase/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fotoperíodo , Triglicerídeos/sangue , Triglicerídeos/metabolismo
5.
Physiol Behav ; 194: 466-473, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29964068

RESUMO

Gastric vagal afferents play an important role in the peripheral control of food intake. Apelin, a central appetite regulating hormone, is also abundantly released from the stomach. Whether apelin modulates gastric vagal afferent signalling is unknown. We aimed to determine whether apelin modulates gastric vagal afferent signalling under different states of nutrition. Female C57BL/6 mice were fed either a standard laboratory diet (SLD) or a high fat diet (HFD) for 12 weeks. An in vitro gastric vagal afferent preparation was used to determine the effect of apelin on gastric vagal afferent mechanosensitivity in SLD mice, fed ad libitum or fasted overnight, and HFD mice. To determine the signalling pathway of apelin via gastric vagal afferents, we determined the expression of apelin receptor (APJ receptor) in the gastric mucosa, the whole nodose ganglion and in gastric vagal afferent neurons innervating the stomach using retrograde tracing and real-time quantitative reverse-transcription polymerase chain reaction (qRT-PCR). The location of apelin and APJ receptor within the gastric mucosa was determined by immunohistochemistry. Expression of apelin and APJ receptor mRNA in gastric mucosa was determined using qRT-PCR. Apelin inhibited the response of gastric mucosal vagal afferents to mucosal stroking in fasted SLD mice, but not in mice fed ad libitum a SLD or HFD. Apelin inhibited the response of gastric tension sensitive afferents to circular stretch in SLD mice fed ad libitum or fasted, an effect not observed in HFD mice. APJ receptor mRNA was detected in the gastric mucosa and whole nodose ganglion, but not specifically in gastric vagal afferents neurons. In the gastric mucosa, APJ receptor immunoreactive cells were co-localised or closely associated with apelin containing cells and co-localised with serotonin, gastrin, histamine and gastric intrinsic factor containing cells. In conclusion, apelin modulates gastric vagal afferent signalling in a nutritional status dependent manner. Further, apelin modulates gastric vagal afferents through an indirect pathway, possibly through the release of hormones/peptides from the gastric mucosa.


Assuntos
Apelina/fisiologia , Mecanotransdução Celular/fisiologia , Nervo Vago/fisiologia , Animais , Apelina/metabolismo , Receptores de Apelina/biossíntese , Dieta Hiperlipídica , Feminino , Polipeptídeo Inibidor Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Gastrinas/metabolismo , Histamina/metabolismo , Camundongos , Neurônios Aferentes/metabolismo , Gânglio Nodoso/metabolismo , Serotonina/metabolismo , Nervo Vago/metabolismo
6.
J Neurosci ; 38(22): 5088-5095, 2018 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-29760179

RESUMO

Mechanosensitive gastric vagal afferents (GVAs) are involved in the regulation of food intake. GVAs exhibit diurnal rhythmicity in their response to food-related stimuli, allowing time of day-specific satiety signaling. This diurnal rhythmicity is ablated in high-fat-diet (HFD)-induced obesity. Time-restricted feeding (TRF) has a strong influence on peripheral clocks. This study aimed to determine whether diurnal patterns in GVA mechanosensitivity are entrained by TRF. Eight-week-old male C57BL/6 mice (N = 256) were fed a standard laboratory diet (SLD) or HFD for 12 weeks. After 4 weeks of diet acclimatization, the mice were fed either ad libitum or only during the light phase [Zeitgeber time (ZT) 0-12] or dark phase (ZT12-24) for 8 weeks. A subgroup of mice from all conditions (n = 8/condition) were placed in metabolic cages. After 12 weeks, ex vivo GVA recordings were taken at 3 h intervals starting at ZT0. HFD mice gained more weight than SLD mice. TRF did not affect weight gain in the SLD mice, but decreased weight gain in the HFD mice regardless of the TRF period. In SLD mice, diurnal rhythms in food intake were inversely associated with diurnal rhythmicity of GVA mechanosensitivity. These diurnal rhythms were entrained by the timing of food intake. In HFD mice, diurnal rhythms in food intake and diurnal rhythmicity of GVA mechanosensitivity were dampened. Loss of diurnal rhythmicity in HFD mice was abrogated by TRF. In conclusion, diurnal rhythmicity in GVA responses to food-related stimuli can be entrained by food intake. TRF prevents the loss of diurnal rhythmicity that occurs in HFD-induced obesity.SIGNIFICANCE STATEMENT Diurnal control of food intake is vital for maintaining metabolic health. Diet-induced obesity is associated with strong diurnal changes in food intake. Vagal afferents are involved in regulation of feeding behavior, particularly meal size, and exhibit diurnal fluctuations in mechanosensitivity. These diurnal fluctuations in vagal afferent mechanosensitivity are lost in diet-induced obesity. This study provides evidence that time-restricted feeding entrains diurnal rhythmicity in vagal afferent mechanosensitivity in lean and high-fat-diet (HFD)-induced obese mice and, more importantly, prevents the loss of rhythmicity in HFD-induced obesity. These data have important implications for the development of strategies to treat obesity.


Assuntos
Vias Aferentes/fisiopatologia , Ritmo Circadiano , Dieta Hiperlipídica , Jejum , Mecanorreceptores , Obesidade/fisiopatologia , Estômago/inervação , Estômago/fisiopatologia , Nervo Vago/fisiopatologia , Animais , Escuridão , Ingestão de Alimentos , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Aumento de Peso
7.
Cell Death Dis ; 8(10): e3136, 2017 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-29072701

RESUMO

Caspase-2 has been shown to be involved in metabolic homeostasis. Here, we show that caspase-2 deficiency alters basal energy metabolism by shifting the balance in fuel choice from fatty acid to carbohydrate usage. At 4 weeks of age, whole-body carbohydrate utilisation was increased in Casp2-/- mice and was maintained into adulthood. By 17 weeks of age, Casp2-/- mice had reduced white adipose mass, smaller white adipocytes decreased fasting blood glucose and plasma triglycerides but maintained normal insulin levels. When placed on a 12-week high-fat diet (HFD), Casp2-/- mice resisted the development of obesity, fatty liver, hyperinsulinemia and insulin resistance. In addition, HFD-fed Casp2-/- mice had reduced white adipocyte hypertrophy, apoptosis and expansion of both subcutaneous and visceral adipose depots. Increased expression of UCP1 and the maintenance of adiponectin levels in white adipose tissue of HFD-fed Casp2-/- mice indicated increased browning and adipocyte hyperplasia. We found that while the preference for whole-body carbohydrate utilisation was maintained, HFD-fed Casp2-/- mice were not impaired in their ability to switch to utilising fats as a fuel source. Our findings suggest that caspase-2 impacts basal energy metabolism by regulating adipocyte biology and fat expansion, most likely via a non-apoptotic function. Furthermore, we show that caspase-2 deficiency shifts the balance in fuel choice towards increased carbohydrate utilisation and propose that this is due to mild energy stress. As a consequence, Casp2-/- mice show an adaptive remodelling of adipose tissue that protects from HFD-induced obesity and improves glucose homeostasis while paradoxically increasing their susceptibility to oxidative stress induced damage and premature ageing.


Assuntos
Caspase 2/deficiência , Obesidade/enzimologia , Animais , Dieta Hiperlipídica , Metabolismo Energético , Expressão Gênica , Humanos , Masculino , Camundongos , Obesidade/metabolismo , Obesidade/patologia , Ratos
8.
Peptides ; 89: 35-41, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28087413

RESUMO

Food intake is regulated by vagal afferent signals from the stomach. Nesfatin-1 is an anorexigenic peptide produced within the gastrointestinal tract and has well defined central effects. We aimed to determine if nesfatin-1 can modulate gastric vagal afferent signals in the periphery and further whether this is altered in different nutritional states. Female C57BL/6J mice were fed either a standard laboratory diet (SLD) or a high fat diet (HFD) for 12 weeks or fasted overnight. Plasma nucleobindin-2 (NUCB2; nesfatin-1 precursor)/nesfatin-1 levels were assayed, the expression of NUCB2 in the gastric mucosa and adipose tissue was assessed using real-time quantitative reverse-transcription polymerase chain reaction. An in vitro preparation was used to determine the effect of nesfatin-1 on gastric vagal afferent mechanosensitivity. HFD mice exhibited an increased body weight and adiposity. Plasma NUCB2/nesfatin-1 levels were unchanged between any of the groups of mice. NUCB2 mRNA was detected in the gastric mucosa and gonadal fat of SLD, HFD and fasted mice with no difference in mRNA abundance between groups in either tissue. In SLD and fasted mice nesfatin-1 potentiated mucosal receptor mechanosensitivity, an effect not observed in HFD mice. Tension receptor mechanosensitivity was unaffected by nesfatin-1 in SLD and fasted mice, but was inhibited in HFD mice. In conclusion, Nesfatin-1 modulates gastric vagal afferent mechanosensitivity in a nutritional state dependent manner.


Assuntos
Vias Aferentes/metabolismo , Proteínas de Ligação ao Cálcio/sangue , Proteínas de Ligação a DNA/sangue , Ingestão de Alimentos/genética , Proteínas do Tecido Nervoso/sangue , Obesidade/genética , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação a DNA/genética , Dieta Hiperlipídica , Comportamento Alimentar , Mucosa Gástrica/metabolismo , Regulação da Expressão Gênica/genética , Camundongos , Proteínas do Tecido Nervoso/genética , Nucleobindinas , Obesidade/metabolismo , Vagotomia Gástrica Proximal , Nervo Vago/metabolismo , Nervo Vago/patologia , Aumento de Peso/genética
9.
J Neurosci ; 36(11): 3199-207, 2016 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-26985030

RESUMO

Rats with high-fat diet (HFD)-induced obesity increase daytime eating, suggesting an alteration in circadian food intake mechanisms. Gastric vagal afferents (GVAs) respond to mechanical stimuli to initiate satiety. These signals are dampened in HFD mice and exhibit circadian variations inversely with food intake in lean mice. Furthermore, leptin shows circadian variation in its circulating level and is able to modulate GVA mechanosensitivity. However, whether leptin's ability to modulate GVAs occurs in a circadian manner is unknown. Therefore, we investigated whether changes in the circadian intake of food in HFD-induced obesity is associated with a disruption in GVA circadian rhythms. Eight-week-old male C57BL/6 mice were fed a standard laboratory diet (SLD) or a HFD for 12 weeks. A subgroup of SLD and HFD mice were housed in metabolic cages. After 12 weeks, ex vivo GVA recordings were taken at 3 h intervals starting at zeitgeber time 0 (ZT0) and stomach content was measured. After 12 weeks, HFD mice consumed more food during the light phase through larger and more frequent meals compared with SLD mice. SLD mice exhibited circadian fluctuation in stomach content, which peaked at ZT18 and reached a nadir at ZT9. At these time points, both tension and mucosal receptor mechanosensitivity were the lowest and highest, respectively. HFD mice exhibited little circadian variation in stomach content or GVA mechanosensitivity. Leptin potentiated mucosal receptor mechanosensitivity only in SLD mice and with reduced potency during the dark phase. In conclusion, loss of circadian variation in GVA signaling may underpin changes in eating behavior in HFD-induced obesity. SIGNIFICANCE STATEMENT: Appropriate circadian control of food intake is vital for maintaining metabolic health. Diet-induced obesity is associated with strong circadian changes in food intake, but the contributing mechanisms have yet to be determined. Vagal afferents are involved in regulation of feeding behavior, particularly meal size, and have been shown to exhibit circadian fluctuation in mechanosensitivity, potentially allowing for time of day-specific levels of satiety signaling. Our study indicates that, in diet-induced obesity, these circadian fluctuations in gastric vagal afferent mechanosensitivity are lost. This was accompanied by increased light phase eating, particularly increased meal size. This is the first evidence that diet-induced disruption to vagal afferent signaling may cause a perturbation in circadian eating patterns.


Assuntos
Ritmo Circadiano/fisiologia , Dieta Hiperlipídica/efeitos adversos , Obesidade/etiologia , Estômago/inervação , Nervo Vago/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Vias Aferentes/fisiologia , Animais , Peso Corporal/fisiologia , Proteínas CLOCK/metabolismo , Modelos Animais de Doenças , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Grelina/farmacologia , Leptina/farmacologia , Masculino , Mecanorreceptores/efeitos dos fármacos , Mecanorreceptores/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Estimulação Física , Ratos , Nervo Vago/efeitos dos fármacos , Nervo Vago/patologia
11.
PLoS One ; 10(8): e0135892, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26285043

RESUMO

AIM: Within the gastrointestinal tract vagal afferents play a role in control of food intake and satiety signalling. Activation of mechanosensitive gastric vagal afferents induces satiety. However, gastric vagal afferent responses to mechanical stretch are reduced in high fat diet mice. Transient receptor potential vanilloid 1 channels (TRPV1) are expressed in vagal afferents and knockout of TRPV1 reduces gastro-oesophageal vagal afferent responses to stretch. We aimed to determine the role of TRPV1 on gastric vagal afferent mechanosensitivity and food intake in lean and HFD-induced obese mice. METHODS: TRPV1+/+ and -/- mice were fed either a standard laboratory diet or high fat diet for 20wks. Gastric emptying of a solid meal and gastric vagal afferent mechanosensitivity was determined. RESULTS: Gastric emptying was delayed in high fat diet mice but there was no difference between TRPV1+/+ and -/- mice on either diet. TRPV1 mRNA expression in whole nodose ganglia of TRPV1+/+ mice was similar in both dietary groups. The TRPV1 agonist N-oleoyldopamine potentiated the response of tension receptors in standard laboratory diet but not high fat diet mice. Food intake was greater in the standard laboratory diet TRPV1-/- compared to TRPV1+/+ mice. This was associated with reduced response of tension receptors to stretch in standard laboratory diet TRPV1-/- mice. Tension receptor responses to stretch were decreased in high fat diet compared to standard laboratory diet TRPV1+/+ mice; an effect not observed in TRPV1-/- mice. Disruption of TRPV1 had no effect on the response of mucosal receptors to mucosal stroking in mice on either diet. CONCLUSION: TRPV1 channels selectively modulate gastric vagal afferent tension receptor mechanosensitivity and may mediate the reduction in gastric vagal afferent mechanosensitivity in high fat diet-induced obesity.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Obesidade/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estômago/inervação , Canais de Cátion TRPV/metabolismo , Nervo Vago/efeitos dos fármacos , Nervo Vago/patologia , Tecido Adiposo/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Esôfago/efeitos dos fármacos , Esôfago/inervação , Técnicas de Inativação de Genes , Masculino , Mecanotransdução Celular/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/genética , Obesidade/patologia , Obesidade/fisiopatologia , Estômago/efeitos dos fármacos , Canais de Cátion TRPV/deficiência , Canais de Cátion TRPV/genética
12.
Peptides ; 71: 141-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26209028

RESUMO

AIM: Gastric vagal afferents are activated in response to mechanical stimulation, an effect attenuated by neuropeptide W (NPW) in 20-week-old female mice. In this study we aimed to determine whether there were age and sex dependent effects of NPW on gastric vagal afferent mechanosensitivity. METHODS: An in vitro gastro-oesophageal preparation was used to determine the effect of NPW on gastric vagal afferent mechanosensitivity from 8 and 20-week-old male and female C57BL/6 mice. Retrograde tracing and laser capture microdissection were used to selectively collect gastric vagal afferent cell bodies. Expression of NPW in the gastric mucosa and its receptor, GPR7, in gastric vagal afferent cell bodies was determined using quantitative RT-PCR. RESULTS: NPW inhibited gastric tension sensitive vagal afferents from 20-week-old male and female mice, but not 8-week-old mice. In contrast, NPW inhibited the mechanosensitivity of gastric mucosal vagal afferents in 8-week-old male and female mice, but not 20-week-old mice. NPW mRNA expression in the gastric mucosa was higher in 20-week-old male mice compared to 8-week-old male mice. GPR7 mRNA expression in vagal afferent neurons innervating the gastric muscular layers was higher in 20-week-old mice compared to 8-week-old mice in both sexes. CONCLUSION: The inhibitory effect of NPW on gastric tension sensitive and mucosal vagal afferents is age but not sex-dependent. These findings suggest that the physiological role of NPW varies depending on the age of the mice.


Assuntos
Envelhecimento/metabolismo , Mucosa Gástrica/inervação , Mecanotransdução Celular/efeitos dos fármacos , Neuropeptídeos/farmacologia , Caracteres Sexuais , Nervo Vago/metabolismo , Animais , Feminino , Mucosa Gástrica/metabolismo , Masculino , Camundongos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/metabolismo
13.
Physiol Behav ; 152(Pt B): 354-62, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26074203

RESUMO

Food intake is regulated by vagal afferent signals from the stomach. Adiponectin, secreted primarily from adipocytes, also has a role in regulating food intake. However, the involvement of vagal afferents in this effect remains to be established. We aimed to determine if adiponectin can modulate gastric vagal afferent (GVA) satiety signals and further whether this is altered in high fat diet (HFD)-induced obesity. Female C57BL/6J mice were fed either a standard laboratory diet (SLD) or a HFD for 12weeks. Plasma adiponectin levels were assayed, and the expression of adiponectin in the gastric mucosa was assessed using real-time quantitative reverse-transcription polymerase chain reaction (qRT-PCR). The location of adiponectin protein within the gastric mucosa was determined by immunohistochemistry. To evaluate the direct effect of adiponectin on vagal afferent endings we determined adiponectin receptor expression in whole nodose ganglia (NDG) and also specifically in GVA neurons using retrograde tracing and qRT-PCR. An in vitro preparation was used to determine the effect of adiponectin on GVA response to mechanical stimulation. HFD mice exhibited an increased body weight and adiposity and showed delayed gastric emptying relative to SLD mice. Plasma adiponectin levels were not significantly different in HFD compared to SLD mice. Adiponectin mRNA was detected in the gastric mucosa of both SLD and HFD mice and presence of protein was confirmed immunohistochemically by the detection of adiponectin immunoreactive cells in the mucosal layer of the stomach. Adiponectin receptor 1 (ADIPOR1) and 2 (ADIPOR2) mRNA was present in both the SLD and HFD whole NDG and also specifically traced gastric mucosal and muscular neurons. There was a reduction in ADIPOR1 mRNA in the mucosal afferents of the HFD mice relative to the SLD mice. In HFD mice adiponectin potentiated gastric mucosal afferent responses to mucosal stroking, an effect not observed in SLD mice. Adiponectin reduced the responses of tension receptors to circular stretch to a similar extent in both SLD and HFD mice. In conclusion, adiponectin modulates GVA satiety signals. This modulatory effect is altered in HFD-induced obesity. It remains to be conclusively determined whether this modulation is involved in the regulation of food intake and what the whole animal phenotypic consequence is.


Assuntos
Adiponectina/metabolismo , Dieta Hiperlipídica/efeitos adversos , Mucosa Gástrica/metabolismo , Neurônios/metabolismo , Gânglio Nodoso/metabolismo , Nervo Vago/metabolismo , Tecido Adiposo/patologia , Vias Aferentes/metabolismo , Vias Aferentes/patologia , Animais , Feminino , Esvaziamento Gástrico/fisiologia , Mucosa Gástrica/patologia , Camundongos Endogâmicos C57BL , Músculo Liso/metabolismo , Músculo Liso/patologia , Técnicas de Rastreamento Neuroanatômico , Neurônios/patologia , Gânglio Nodoso/patologia , RNA Mensageiro/metabolismo , Distribuição Aleatória , Receptores de Adiponectina/metabolismo , Nervo Vago/patologia , Aumento de Peso/fisiologia
14.
J Physiol ; 593(4): 775-86, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25433079

RESUMO

Gastrointestinal (GI) vagal afferents are a key mediatory of food intake. Through a balance of responses to chemical and mechanical stimuli food intake can be tightly controlled via the ascending satiety signals initiated in the GI tract. However, vagal responses to both mechanical and chemical stimuli are modified in diet-induced obesity (DIO). Much of the research to date whilst in relatively isolated/controlled circumstances indicates a shift between a balance of orexigenic and anorexigenic vagal signals to blunted anorexigenic and potentiated orexigenic capacity. Although the mechanism responsible for the DIO shift in GI vagal afferent signalling is unknown, one possible contributing factor is the gut microbiota. Nevertheless, whatever the mechanism, the observed changes in gastrointestinal vagal afferent signalling may underlie the pathophysiological changes in food consumption that are pivotal for the development and maintenance of obesity.


Assuntos
Trato Gastrointestinal/inervação , Obesidade/fisiopatologia , Nervo Vago/fisiologia , Animais , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiologia , Humanos , Neurônios Aferentes/fisiologia
15.
Peptides ; 62: 6-14, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25270269

RESUMO

Neuropeptide W (NPW) is secreted from gastrin (G) cells in the stomach in response to food intake. The mechanisms underlying food intake-induced regulation of gastric NPW is largely unknown. We hypothesized that specific macronutrients were responsible for food-induced NPW secretion. We evaluated the acute effects of fat, carbohydrate and protein on plasma NPW concentrations in humans and mice. The effect of different nutrients on expression of NPW in the antral stomach was also determined in mice. Primary cell cultures of mouse gastric antral mucosal cells were used to investigate the signaling pathway of NPW expression. Plasma NPW concentrations did not change after nutrient ingestion in either humans or mice. NPW mRNA expression and the number of NPW positive cells in the mouse antrum were increased in mice gavage fed with protein or glucose, but not lipid. In primary antral mucosal cell culture, NPW mRNA expression was stimulated by l-phenylalanine, but not glucose. Calcium-sensing receptor (CaSR) positive cells were largely co-localized with NPW in mouse gastric antral mucosal cells, and NPW mRNA expression was inhibited by a selective antagonist of CaSR NPS2143. However, the l-phenylalanine-induced increase in NPW expression was not affected by NPS2143. In conclusion, these studies indicated an inconsistency between plasma and gastric NPW expression in response to nutrient ingestion, suggesting food induced gastric NPW expression may play a more important role locally. Moreover, glucose and especially protein are potent regulators of gastric NPW, via distinct mechanisms.


Assuntos
Ingestão de Alimentos , Mucosa Gástrica/metabolismo , Neuropeptídeos/sangue , Receptores de Detecção de Cálcio/metabolismo , Animais , Carboidratos/administração & dosagem , Alimentos , Regulação da Expressão Gênica , Glucose/administração & dosagem , Humanos , Camundongos , Naftalenos/administração & dosagem , Neuropeptídeos/biossíntese , Proteínas/administração & dosagem , Antro Pilórico/efeitos dos fármacos , Antro Pilórico/metabolismo , RNA Mensageiro/biossíntese , Receptores de Detecção de Cálcio/antagonistas & inibidores , Estômago/fisiologia
16.
Mol Metab ; 3(6): 593-4, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25161882
17.
J Physiol ; 592(15): 3287-301, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24879868

RESUMO

Neuronal nitric oxide (NO) plays an important role in gastric motor activity and modulates the mechanosensitivity of gastro-oesophageal vagal afferents. Effects of NO on food intake are dependent on feeding status. We sought to determine the effect of NO on gastro-oesophageal vagal afferent activity in the normally fed and food-restricted states and the second messenger pathways mediating these effects. Eight week old female C56BL/6 mice were fed ad libitum or food restricted for 14 h. An in vitro preparation was used to determine the functional effects of NO and the second messenger pathways involved. Expression of NO signal transduction molecules in vagal afferents was determined by reverse-transcription polymerase chain reaction (RT-PCR). Endogenous NO and the NO donor S-nitroso-N-acetylpenicillamine (SNAP) inhibited vagal mucosal afferent responses to tactile stimuli in mice fed ad libitum. After a 14 h fast endogenous NO and SNAP potentiated tension and mucosal afferent responses to mechanical stimulation. The excitatory effect of NO was blocked by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor apocynin. After a 14 h fast expression of NADPH oxidase 2 (NOX2) mRNA in whole nodose ganglia was significantly reduced and the excitatory effect of NO on gastro-oesophageal vagal afferents was lost. Under fasting conditions the inhibitory effect of NO was blocked with the hyperpolarisation-activated cyclic nucleotide-gated (HCN) channel blocker ivabradine and mRNA expression of HCN3 in the nodose ganglia was elevated. In conclusion, the role of NO in the peripheral modulation of gastro-oesophageal vagal afferents is dynamic and dependent on feeding status.


Assuntos
Dieta , Esôfago/inervação , Mecanotransdução Celular , Neurônios Aferentes/fisiologia , Óxido Nítrico/metabolismo , Gânglio Nodoso/fisiologia , Acetofenonas/farmacologia , Animais , Benzazepinas/farmacologia , Esôfago/fisiologia , Feminino , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/antagonistas & inibidores , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Ivabradina , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidase 2 , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Neurônios Aferentes/metabolismo , Gânglio Nodoso/efeitos dos fármacos , Gânglio Nodoso/metabolismo , Canais de Potássio/genética , Canais de Potássio/metabolismo
18.
Physiol Behav ; 136: 170-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24657740

RESUMO

Vagal afferents are a vital link between the peripheral tissue and central nervous system (CNS). There is an abundance of vagal afferents present within the proximal gastrointestinal tract which are responsible for monitoring and controlling gastrointestinal function. Whilst essential for maintaining homeostasis there is a vast amount of literature emerging which describes remarkable plasticity of vagal afferents in response to endogenous as well as exogenous stimuli. This plasticity for the most part is vital in maintaining healthy processes; however, there are increased reports of vagal plasticity being disrupted in pathological states, such as obesity. Many of the disruptions, observed in obesity, have the potential to reduce vagal afferent satiety signalling which could ultimately perpetuate the obese state. Understanding how plasticity occurs within vagal afferents will open a whole new understanding of gut function as well as identify new treatment options for obesity.


Assuntos
Trato Gastrointestinal/fisiologia , Plasticidade Neuronal/fisiologia , Nervo Vago/fisiologia , Animais , Tronco Encefálico/citologia , Humanos
19.
J Neurosci ; 33(49): 19238-42, 2013 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-24305819

RESUMO

Food intake is coordinated to cellular metabolism by clock gene expression with a master clock in the suprachiasmatic nucleus synchronized by light exposure. Gastric vagal afferents play a role in regulating food intake, but it is unknown whether they exhibit circadian variation in their mechanosensitivity. We aimed to determine whether gastric vagal afferents express clock genes and whether their response to mechanical stimuli oscillates throughout the light/dark cycle. Nodose ganglia were collected from 8-week-old female C57BL/6 mice every 3 h starting at lights off (1800 h) to quantify Bmal1, Per1, Per2, and Nr1d1 mRNA by qRT-PCR. Additionally in vitro single-fiber recordings of gastric vagal mechanoreceptors were taken at all time points. Per1, Per2, Bmal1, and Nr1d1 mRNA is expressed in the nodose ganglia and levels oscillated over a 24 h period. In mice fed ad libitum, gastric content was 3 times higher at 0000 h and 0300 h than 1200 h. The response of tension receptors to 3 g stretch was reduced by up to 70% at 2100 h, 0000 h, and 0300 h compared with 1200 h. Gastric mucosal receptor response to stroking with a 50 mg von Frey hair was 3 times greater at 1200 h and 1500 h than the response at 0000 h. Similar findings were obtained in mice fasted for 6 h or maintained in darkness for 3 d before study. Therefore, these changes do not result from food intake or the light/dark cycle. Thus, gastric vagal mechanoreceptors display circadian rhythm, which may act to control food intake differentially at different times of the day.


Assuntos
Ritmo Circadiano/fisiologia , Mecanorreceptores/fisiologia , Neurônios Aferentes/fisiologia , Estômago/inervação , Nervo Vago/fisiologia , Animais , Proteínas CLOCK/genética , Escuridão , Ingestão de Alimentos/fisiologia , Esôfago/inervação , Esôfago/fisiologia , Feminino , Mucosa Gástrica/inervação , Mucosa Gástrica/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular/fisiologia , Gânglio Nodoso/citologia , Gânglio Nodoso/fisiologia , Estimulação Física , Reação em Cadeia da Polimerase em Tempo Real
20.
Peptides ; 46: 150-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23792934

RESUMO

Leptin, ghrelin and neuropeptide W (NPW) modulate vagal afferent activity, which may underlie their appetite regulatory actions. High fat diet (HFD)-induced obesity induces changes in the plasma levels of these peptides and alters the expression of receptors on vagal afferents. We investigated homologous and heterologous receptor regulation by leptin, ghrelin and NPW. Mice were fed (12 weeks) a standard laboratory diet (SLD) or HFD. Nodose ganglia were cultured overnight in the presence or absence of each peptide. Leptin (LepR), ghrelin (GHS-R), NPW (GPR7) and cholecystokinin type-1 (CCK1R) receptor mRNA, and the plasma leptin, ghrelin and NPW levels were measured. SLD: leptin reduced LepR, GPR7, increased GHS-R and CCK1R mRNA; ghrelin increased LepR, GPR7, CCK1R, and decreased GHS-R. HFD: leptin decreased GHS-R and GPR7, ghrelin increased GHS-R and GPR7. NPW decreased all receptors except GPR7 which increased with HFD. Plasma leptin was higher and NPW lower in HFD. Thus, HFD-induced obesity disrupts inter-regulation of appetite regulatory receptors in vagal afferents.


Assuntos
Regulação do Apetite/fisiologia , Apetite/fisiologia , Grelina/sangue , Leptina/sangue , Neuropeptídeos/sangue , Animais , Células Cultivadas , Dieta Hiperlipídica , Feminino , Grelina/metabolismo , Leptina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeos/metabolismo , Gânglio Nodoso/citologia , Gânglio Nodoso/efeitos dos fármacos , Obesidade/sangue , RNA Mensageiro , Receptor de Colecistocinina A/genética , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Grelina/metabolismo , Receptores para Leptina/metabolismo , Receptores de Neuropeptídeos/biossíntese , Receptores de Neuropeptídeos/metabolismo , Nervo Vago/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...