Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Animal ; 13(11): 2714-2726, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31387651

RESUMO

The gastrointestinal tract (GIT) is an interface between the external and internal milieus that requires continuous monitoring for nutrients or pathogens and toxic chemicals. The study of the physiological/molecular mechanisms, mediating the responses to the monitoring of the GIT contents, has been referred to as chemosensory science. While most of the progress in this area of research has been obtained in laboratory rodents and humans, significant steps forward have also been reported in pigs. The objective of this review was to update the current knowledge on nutrient chemosensing in pigs in light of recent advances in humans and laboratory rodents. A second objective relates to informing the existence of nutrient sensors with their functionality, particularly linked to the gut peptides relevant to the onset/offset of appetite. Several cell types of the intestinal epithelium such as Paneth, goblet, tuft and enteroendocrine cells (EECs) contain subsets of chemosensory receptors also found on the tongue as part of the taste system. In particular, EECs show specific co-expression patterns between nutrient sensors and/or transceptors (transport proteins with sensing functions) and anorexigenic hormones such as cholecystokinin (CCK), peptide tyrosine tyrosine (PYY) or glucagon-like peptide-1 (GLP-1), amongst others. In addition, the administration of bitter compounds has an inhibitory effect on GIT motility and on appetite through GLP-1-, CCK-, ghrelin- and PYY-labelled EECs in the human small intestine and colon. Furthermore, the mammalian chemosensory system is the target of some bacterial metabolites. Recent studies on the human microbiome have discovered that commensal bacteria have developed strategies to stimulate chemosensory receptors and trigger host cellular functions. Finally, the study of gene polymorphisms related to nutrient sensors explains differences in food choices, food intake and appetite between individuals.


Assuntos
Preferências Alimentares , Microbioma Gastrointestinal , Suínos/fisiologia , Animais , Apetite , Metabolismo dos Carboidratos , Colecistocinina/metabolismo , Células Enteroendócrinas/metabolismo , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiologia , Grelina/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Nutrientes , Peptídeo YY/metabolismo , Suínos/microbiologia , Paladar
2.
Annu Rev Physiol ; 80: 117-141, 2018 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-29029594

RESUMO

The gastrointestinal tract represents the largest interface between the human body and the external environment. It must continuously monitor and discriminate between nutrients that need to be assimilated and harmful substances that need to be expelled. The different cells of the gut epithelium are therefore equipped with a subtle chemosensory system that communicates the sensory information to several effector systems involved in the regulation of appetite, immune responses, and gastrointestinal motility. Disturbances or adaptations in the communication of this sensory information may contribute to the development or maintenance of disease. This is a new emerging research field in which perception of taste can be considered as a novel key player participating in the regulation of gut function. Specific diets or agonists that target these chemosensory signaling pathways may be considered as new therapeutic targets to tune adequate physiological processes in the gut in health and disease.


Assuntos
Células Quimiorreceptoras/metabolismo , Trato Gastrointestinal/metabolismo , Transdução de Sinais/fisiologia , Animais , Hormônios Gastrointestinais/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Paladar/fisiologia
3.
Artigo em Inglês | MEDLINE | ID: mdl-28776826

RESUMO

BACKGROUND: Intragastric administration of the bitter tastant denatonium benzoate inhibits the increase of motilin plasma levels and antral contractility. While these findings suggest that gastrointestinal bitter taste receptors could be new targets to modulate gastrointestinal motility and hormone release, they need confirmation with other bitter receptor agonists. The primary aim was to evaluate the effect of intragastric administration of the bitter tastant quinine-hydrochloride (QHCl) on motilin and ghrelin plasma levels. Secondly, we studied the effect on interdigestive motility. METHODS: Ten healthy female volunteers were recruited (33±4 y; 22±0.5 kg/m²). Placebo or QHCl (10 µmol/kg) was administered intragastrically through a nasogastric feeding tube after an overnight fast in a single-blind randomized fashion. Administration started 20 min after the first phase III of the migrating motor complex. The measurement continued for another 2 h after the administration. Blood samples were collected every 10 min with the baseline sample taken 10 min prior to administration. KEY RESULTS: The increase in plasma levels of motilin (administration; P=.04) and total ghrelin (administration; P=.02) was significantly lower after QHCl. The fluctuation of octanoylated ghrelin was reduced after QHCl (time by administration; P=.03). Duodenal motility did not differ. The fluctuation of antral activity differed over time between placebo and QHCl (time by administration; P=.03). CONCLUSIONS AND INFERENCES: QHCl suppresses the increase of both motilin and ghrelin plasma levels. Moreover, QHCl reduced the fluctuation of antral motility. These findings confirm the potential of bitter taste receptors as targets for modifying interdigestive motility in man.


Assuntos
Jejum , Motilidade Gastrointestinal , Grelina/sangue , Motilina/sangue , Quinina/administração & dosagem , Adulto , Duodeno/efeitos dos fármacos , Duodeno/fisiologia , Feminino , Humanos , Antro Pilórico/efeitos dos fármacos , Antro Pilórico/fisiologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-28782145

RESUMO

BACKGROUND: The gut hormone motilin stimulates gastrointestinal motility by inducing gastric phase III of the migrating motor complex (MMC) and enhancing the rate of gastric emptying. Camicinal (GSK962040), a small molecule motilin receptor agonist, has been shown to increase gastrointestinal motility. METHODS: In this proof of concept study the effects of camicinal on MMC activity, esophageal and gastric pH was evaluated in eight healthy volunteers as a secondary endpoint. Doses of 50 and 150 mg were compared to placebo for a period of 24 hours in a double-blinded randomized crossover trial. KEY RESULTS: The 50 mg dose (n=4) of camicinal had no significant impact on gastroduodenal manometry or pH parameters. A single dose of 150 mg (n=4) induced a gastric phase III after 0:34 h (0:25-0:58), which was significantly faster compared to placebo (18:15 h (4:32-22:16); P=.03). Moreover, the high dose significantly increased the occurrence of gastric phase III contractions compared to placebo (12% vs 39%; P=.0003). This increase in gastric phase III contractions during a period of 24 hour was due to an increased occurrence of gastric phases III during the daytime (5% vs 50%; P=.0001). The same dose however did not affect small bowel manometry parameters or esophageal and gastric pH. CONCLUSIONS AND INFERENCES: Considering its stimulating effect on the MMC and gastric emptying, camicinal is an attractive candidate for the treatment of gastroparesis and gastroesophageal reflux disease. This trial was registered at clinicaltrials.gov as NCT00562848.


Assuntos
Esvaziamento Gástrico/efeitos dos fármacos , Fármacos Gastrointestinais/administração & dosagem , Complexo Mioelétrico Migratório/efeitos dos fármacos , Piperazinas/administração & dosagem , Piperidinas/administração & dosagem , Receptores dos Hormônios Gastrointestinais/agonistas , Receptores de Neuropeptídeos/agonistas , Adolescente , Adulto , Estudos Cross-Over , Método Duplo-Cego , Feminino , Humanos , Masculino , Manometria , Pessoa de Meia-Idade , Adulto Jovem
5.
Artigo em Inglês | MEDLINE | ID: mdl-28429863

RESUMO

BACKGROUND: Electrical stimulation of the cervical vagus nerve (VNS) prevents postoperative ileus (POI) in mice. As this approach requires an additional cervical procedure, we explored the possibility of peroperative abdominal VNS in mice and human. METHODS: The effect of cervical and abdominal VNS was studied in a murine model of POI and lipopolysaccharide (LPS)-induced sepsis. Postoperative ileus was quantified by assessment of intestinal transit of fluorescent dextran expressed as geometric center (GC). Next, the effect of cervical and abdominal VNS on heart rate was determined in eight Landrace pigs to select the optimal electrode for VNS in human. Finally, the effect of sham or abdominal VNS on LPS-induced cytokine production of whole blood was studied in patients undergoing colorectal surgery. KEY RESULTS: Similar to cervical VNS, abdominal VNS significantly decreased LPS-induced serum tumor necrosis factor-α (TNFα) levels (abdominal VNS: 366±33 pg/mL vs sham: 822±105 pg/mL; P<.01). In line, in a murine model of POI, abdominal VNS significantly improved intestinal transit (GC: sham 5.1±0.2 vs abdominal VNS: 7.8±0.6; P<.01) and reduced intestinal inflammation (abdominal VNS: 35±7 vs sham: 80±8 myeloperoxidase positive cells/field; P<.05). In pigs, heart rate was reduced by cervical VNS but not by abdominal VNS. In humans, abdominal VNS significantly reduced LPS-induced IL8 and IL6 production by whole blood. CONCLUSIONS & INFERENCES: Abdominal VNS is feasible and safe in humans and has anti-inflammatory properties. As abdominal VNS improves POI similar to cervical VNS in mice, our data indicate that peroperative abdominal VNS may represent a novel approach to shorten POI in man.


Assuntos
Íleus/prevenção & controle , Complicações Pós-Operatórias/prevenção & controle , Estimulação do Nervo Vago/métodos , Animais , Citocinas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Polipeptídeo Pancreático/sangue , Projetos Piloto , Suínos
6.
Neurogastroenterol Motil ; 28(6): 934-47, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26891411

RESUMO

BACKGROUND: Postoperative ileus (POI) is characterized by a transient inhibition of gastrointestinal (GI) motility after abdominal surgery mediated by the inflammation of the muscularis externa (ME). The aim of this study was to identify alterations in the enteric nervous system that may contribute to the pathogenesis of POI. METHODS: Gastrointestinal transit, contractility of isolated smooth muscle strips and inflammatory parameters were evaluated at different time points (1.5 h to 10 days) after intestinal manipulation (IM) in mice. Immune-labeling was used to visualize changes in myenteric neurons. KEY RESULTS: Intestinal manipulation resulted in an immediate inhibition of GI transit recovering between 24 h and 5 days. In vitro contractility to K(+) (60 mM) or carbachol (10(-9) to 10(-4) M) was biphasically suppressed over 24 h after IM (with transient recovery at 6 h). The first phase of impaired myogenic contractility was associated with increased expression of TNF-α, IL-6 and IL-1α. After 24 h, we identified a significant reduction in electrical field stimulation-evoked contractions and relaxations, lasting up to 10 days after IM. This was associated with a reduced expression of chat and nos1 genes. CONCLUSIONS & INFERENCES: Intestinal manipulation induces two waves of smooth muscle inhibition, most likely mediated by inflammatory cytokines, lasting up to 3 days after IM. Further, we here identify a late third phase (>24 h) characterized by impaired cholinergic and nitrergic neurotransmission persisting after recovery of muscle contractility. These findings illustrate that POI results from inflammation-mediated impaired smooth muscle contraction, but also involves a long-lasting impact of IM on the enteric nervous system.


Assuntos
Sistema Nervoso Entérico/fisiopatologia , Íleus/fisiopatologia , Mediadores da Inflamação , Músculo Liso/fisiopatologia , Complicações Pós-Operatórias/fisiopatologia , Animais , Sistema Nervoso Entérico/metabolismo , Feminino , Motilidade Gastrointestinal/fisiologia , Íleus/metabolismo , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso/metabolismo , Técnicas de Cultura de Órgãos , Complicações Pós-Operatórias/metabolismo
7.
Gut ; 65(2): 214-24, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25539673

RESUMO

RATIONALE: Hunger is controlled by the brain, which receives input from signals of the GI tract (GIT). During fasting, GIT displays a cyclical motor pattern, the migrating motor complex (MMC), regulated by motilin. OBJECTIVES: To study the relationship between hunger and MMC phases (I-III), focusing on spontaneous and pharmacologically induced phase III and the correlation with plasma motilin and ghrelin levels. The role of phase III was also studied in the return of hunger after a meal in healthy individuals and in patients with loss of appetite. FINDINGS: In fasting healthy volunteers, mean hunger ratings during a gastric (62.5±7.5) but not a duodenal (40.4±5.4) phase III were higher (p<0.0005) than during phase I (27.4±4.7) and phase II (37±4.5). The motilin agonist erythromycin, but not the cholinesterase inhibitor neostigmine, induced a premature gastric phase III, which coincided with an increase in hunger scores from 29.2±7 to 61.7±8. The somatostatin analogue octreotide induced a premature intestinal phase III without a rise in hunger scores. Hunger ratings significantly correlated (ß=0.05; p=0.01) with motilin plasma levels, and this relationship was lost after erythromycin administration. Motilin, but not ghrelin administration, induced a premature gastric phase III and a rise in hunger scores. In contrast to octreotide, postprandial administration of erythromycin induced a premature gastric phase III accompanied by an early rise in hunger ratings. In patients with unexplained loss of appetite, gastric phase III was absent and hunger ratings were lower. CONCLUSIONS: Motilin-induced gastric phase III is a hunger signal from GIT in man.


Assuntos
Fome/fisiologia , Motilina/fisiologia , Contração Muscular/fisiologia , Complexo Mioelétrico Migratório/fisiologia , Estômago/fisiologia , Apetite/fisiologia , Inibidores da Colinesterase/farmacologia , Duodeno/fisiologia , Ingestão de Alimentos/fisiologia , Eritromicina/farmacologia , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/fisiologia , Grelina/fisiologia , Humanos , Fome/efeitos dos fármacos , Manometria , Motilina/agonistas , Motilina/sangue , Neostigmina/farmacologia , Octreotida/farmacologia , Fragmentos de Peptídeos/farmacologia , Somatostatina/farmacologia
8.
Gut ; 65(7): 1110-8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-25986945

RESUMO

OBJECTIVE: Motilin-induced phase III contractions of the migrating motor complex (MMC) signal hunger in healthy volunteers. The current aim was to study the role of motilin as a hunger-inducing factor in obese patients and to evaluate the effect of Roux-en-Y gastric bypass (RYGB) surgery on plasma motilin levels and hunger scores. DESIGN: Motilin and ghrelin plasma levels were determined during a complete MMC cycle in controls and obese patients selected for RYGB before, 6 months and 1 year after surgery. 20 min after the end of the second phase III, obese patients received an intravenous infusion of 40 mg erythromycin. Hunger was scored every 5 min. Hedonic hunger was assessed in obese patients with the Power of Food Scale questionnaire. RESULTS: Obesity caused a switch in the origin of phase III from antrum to duodenum. Obese patients had significantly higher motilin levels compared with controls during the MMC but tended to lack the motilin peak prior to phase III necessary to trigger hunger. Hunger scores during phase III were significantly lower in obese patients, but could be restored to control levels through the administration of a low dose of the motilin agonist, erythromycin. After RYGB surgery motilin, but not ghrelin, levels decreased in parallel with hedonic hunger scores. CONCLUSIONS: Motilin may be an important regulator involved in the pathogenesis of obesity.


Assuntos
Fome/fisiologia , Motilina/sangue , Complexo Mioelétrico Migratório , Obesidade/sangue , Obesidade/cirurgia , Adulto , Estudos de Casos e Controles , Duodeno/fisiopatologia , Eritromicina/farmacologia , Feminino , Derivação Gástrica , Fármacos Gastrointestinais/farmacologia , Grelina/sangue , Humanos , Fome/efeitos dos fármacos , Masculino , Período Pós-Operatório , Período Pré-Operatório , Antro Pilórico/fisiopatologia , Inquéritos e Questionários
9.
Acta Physiol (Oxf) ; 216(4): 407-20, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26493384

RESUMO

The bitter taste receptor (TAS2R)-family of G-protein-coupled receptors has been identified on the tongue as detectors of bitter taste over a decade ago. In the last few years, they have been discovered in an ever growing number of extra-oral tissues, including the airways, the gut, the brain and even the testis. In tissues that contact the exterior, protective functions for TAS2Rs have been proposed, in analogy to their function on the tongue as toxicity detector. However, TAS2Rs have also been found in internal organs, suggesting other roles for these receptors, perhaps involving as yet unidentified endogenous ligands. The current review gives an overview of the different proposed functions for TAS2Rs in tissues other than the oral cavity; from appetite regulation to the treatment of asthma, regulation of gastrointestinal motility and control of airway innate immunity.


Assuntos
Receptores Acoplados a Proteínas G , Humanos
10.
Obes Rev ; 17(2): 108-25, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26693661

RESUMO

Although obesity is considered to result from an imbalance between energy uptake and energy expenditure, the strategy of dietary changes and physical exercise has failed to tackle the global obesity epidemic. In search of alternative and more adequate treatment options, research has aimed at further unravelling the mechanisms underlying this excessive weight gain. While numerous studies are focusing on the neuroendocrine alterations that occur after bariatric Roux-en-Y gastric bypass surgery, an increasing amount of chronobiological studies have started to raise awareness concerning the pivotal role of the circadian system in the development and exacerbation of obesity. This internal timekeeping mechanism rhythmically regulates metabolic and physiological processes in order to meet the fluctuating demands in energy use and supply throughout the 24-h day. This review elaborates on the extensive bidirectional interaction between the circadian system and metabolism and explains how disruption of body clocks by means of shift work, frequent time zone travelling or non-stop consumption of calorie-dense foods can evoke detrimental metabolic alterations that contribute to obesity. Altering the body's circadian rhythms by means of time-related dietary approaches (chrononutrition) or pharmacological substances (chronobiotics) may therefore represent a novel and interesting way to prevent or treat obesity and associated comorbidities.


Assuntos
Transtornos Cronobiológicos/fisiopatologia , Obesidade/fisiopatologia , Transtornos Cronobiológicos/etiologia , Transtornos Cronobiológicos/metabolismo , Ritmo Circadiano , Metabolismo Energético , Humanos , Obesidade/complicações , Obesidade/metabolismo , Aumento de Peso , Redução de Peso
11.
Sci Rep ; 5: 16748, 2015 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-26576661

RESUMO

As adequate food intake is crucial to survival, organisms have evolved endogenous circadian clocks to generate optimal temporal patterns of food-related behavior and physiology. The gastric ghrelin-secreting cell is thought to be part of this network of peripheral food-entrainable oscillators (FEOs), regulating the circadian release of this orexigenic peptide. This study aimed to determine the role of the core clock gene Bmal1 and the gastric ghrelin-secreting cell as an FEO in the circadian rhythmicity of ghrelin expression and secretion in vivo and in vitro. Bmal1-deficient mice not only lacked circadian rhythmicity in plasma ghrelin levels and food intake, but also showed decreased gastric mRNA expression of ghrelin and ghrelin O-acyltransferase (GOAT), the ghrelin activating enzyme. Furthermore, in the absence of the hypothalamic master clock, food-related stimuli entrained the molecular clock of gastric ghrelinoma cells to regulate the rhythmic release of ghrelin. Divergent responses in octanoyl and total ghrelin release towards different food cues were observed, suggesting that the FEO also regulates the circadian rhythmicity of GOAT. Collectively, these findings indicate that circadian rhythmicity of ghrelin signaling requires Bmal1 and is driven by a food-responsive clock in the gastric ghrelin-secreting cell that not only regulates ghrelin, but also GOAT activity.


Assuntos
Fatores de Transcrição ARNTL/genética , Aciltransferases/metabolismo , Relógios Circadianos/genética , Ritmo Circadiano/fisiologia , Mucosa Gástrica/metabolismo , Grelina/metabolismo , Estômago/citologia , Animais , Linhagem Celular Tumoral , Sinais (Psicologia) , Ingestão de Alimentos , Feminino , Deleção de Genes , Expressão Gênica , Grelina/genética , Masculino , Proteínas de Membrana , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
12.
Sci Rep ; 5: 15725, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26510380

RESUMO

Taste receptors on enteroendocrine cells sense nutrients and transmit signals that control gut hormone release. This study aimed to investigate the amino acid (AA) sensing mechanisms of the ghrelin cell in a gastric ghrelinoma cell line, tissue segments and mice. Peptone and specific classes of amino acids stimulate ghrelin secretion in the ghrelinoma cell line. Sensing of L-Phe occurs via the CaSR, monosodium glutamate via the TAS1R1-TAS1R3 while L-Ala and peptone act via 2 different amino acid taste receptors: CaSR &TAS1R1-TAS1R3 and CaSR &GPRC6A, respectively. The stimulatory effect of peptone on ghrelin release was mimicked ex vivo in gastric but not in jejunal tissue segments, where peptone inhibited ghrelin release. The latter effect could not be blocked by receptor antagonists for CCK, GLP-1 or somatostatin. In vivo, plasma ghrelin levels were reduced both upon intragastric (peptone or L-Phe) or intravenous (L-Phe) administration, indicating that AA- sensing is not polarized and is due to inhibition of ghrelin release from the stomach or duodenum respectively. In conclusion, functional AA taste receptors regulate AA-induced ghrelin release in vitro. The effects differ between stomach and jejunum but these local nutrient sensing mechanisms are overruled in vivo by indirect mechanisms inhibiting ghrelin release.


Assuntos
Aminoácidos/metabolismo , Grelina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular Tumoral , Grelina/genética , Peptídeo 1 Semelhante ao Glucagon/genética , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Camundongos , Proteína Serina-Treonina Quinase 2 de Interação com Receptor , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Receptores Acoplados a Proteínas G/genética
13.
Neurogastroenterol Motil ; 27(11): 1542-52, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26227790

RESUMO

BACKGROUND: The orexigenic peptide ghrelin has anti-inflammatory properties in colitis, however, the mechanism of action and the immune cells targeted remain still to be elucidated. Here, we assessed the possible effect of ghrelin on T helper (Th) cells in a T cell transfer model of chronic colitis. METHODS: Disease was induced in the recombination activating gene 1 knockout mice (Rag1(-/-) ) by adoptive transfer of naïve Th cells from ghrelin receptor knockout mice (GRLN-R(-/-) ) or littermate wild-type (WT) mice. The course and severity of colitis was assessed by monitoring body weight, diarrhea score, histological analysis, gene expression, and flow cytometry analysis. The possible effects of ghrelin on Th cell proliferation, polarization, and apoptosis was examined in vitro. KEY RESULTS: Our data showed that Rag1(-/-) mice injected with GRLN-R(-/-) Th cells displayed increased severity of colitis compared to mice injected with WT Th cells. In addition, Rag1(-/-) mice injected with GRLN-R(-/-) Th cells had significantly higher intestinal inflammation and increased accumulation of Th1 and Th17 cells in the colon. In vitro, ghrelin directly affected proliferation of Th cells and induced apoptosis whereas it did not influence Th cell polarization. CONCLUSION & INFERENCES: Our observations suggest that ghrelin modulates Th effector cells in the gut controlling proliferation and inducing apoptosis. Our findings further support the use of ghrelin as a novel therapeutic option to treat intestinal inflammatory diseases.


Assuntos
Colite/imunologia , Receptores de Grelina/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Transferência Adotiva , Animais , Apoptose/imunologia , Proliferação de Células , Modelos Animais de Doenças , Citometria de Fluxo , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real
14.
Mol Metab ; 4(6): 437-60, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26042199

RESUMO

BACKGROUND: The gastrointestinal peptide hormone ghrelin was discovered in 1999 as the endogenous ligand of the growth hormone secretagogue receptor. Increasing evidence supports more complicated and nuanced roles for the hormone, which go beyond the regulation of systemic energy metabolism. SCOPE OF REVIEW: In this review, we discuss the diverse biological functions of ghrelin, the regulation of its secretion, and address questions that still remain 15 years after its discovery. MAJOR CONCLUSIONS: In recent years, ghrelin has been found to have a plethora of central and peripheral actions in distinct areas including learning and memory, gut motility and gastric acid secretion, sleep/wake rhythm, reward seeking behavior, taste sensation and glucose metabolism.

15.
Neurogastroenterol Motil ; 27(1): 63-71, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25393165

RESUMO

BACKGROUND: Fluctuations in motilin plasma levels have been implicated in the control of the migrating motor complex (MMC). A plasma peak of motilin is present before a gastric phase III. Furthermore, not only exogenous administration of motilin but also ghrelin induces a gastric phase III in man. Aim of this study was to investigate the role of endogenous ghrelin in the regulation of the MMC. METHODS: Plasma samples for motilin and ghrelin were taken in between two consecutive phases III of either origin measured using high-resolution manometry. KEY RESULTS: The duration of 1 complete MMC cycle was on average 95 ± 12 min. Sixty percent of the first phases III and 40% of the second phases III had a gastric origin (p = 0.0574). Motilin (p < 0.05) plasma levels differed significantly between the phases of the MMC but total and octanoylated ghrelin did not. The percentage change in motilin during the MMC was dependent on the origin of phase III (p < 0.05). Motilin levels increased on average with 35 ± 10% right before a gastric phase III and with 3 ± 4% before a duodenal phase III (p < 0.05). The percentage change in total and octanoylated ghrelin plasma levels was not affected by the origin of phase III. CONCLUSIONS & INFERENCES: These results confirm the role of motilin but not of ghrelin as an endogenous physiological regulator of the MMC with a gastric phase III.


Assuntos
Grelina/sangue , Motilina/sangue , Complexo Mioelétrico Migratório , Adulto , Feminino , Humanos , Masculino , Manometria
16.
Neurogastroenterol Motil ; 25(9): 720-32, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23910374

RESUMO

BACKGROUND: Ghrelin, an orexigenic hormone secreted from the stomach, was soon after its discovery hypothesized to be a prokinetic agent, due to its homology to motilin. Studies in animals and humans, using ghrelin and ghrelin receptor agonists, confirmed this hypothesis, suggesting a therapeutic potential for the ghrelin receptor in the treatment of gastrointestinal motility disorders. Precilinical studies demonstrated that ghrelin can act directly on ghrelin receptors on the enteric nervous system, but the predominant route of action under physiological circumstances is signaling via the vagus nerve in the upper gastrointestinal tract and the pelvic nerves in the colon. Different pharmaceutical companies have designed stable ghrelin mimetics that revealed promising results in trials for the treatment of diabetic gastroparesis and post-operative ileus. Nevertheless, no drug was able to reach the market so far, facing problems proving superiority over placebo treatment in larger trials. PURPOSE: This review aims to summarize the road that led to the current knowledge concerning the prokinetic properties of ghrelin with a focus on the therapeutic potential of ghrelin receptor agonists in the treatment of hypomotility disorders. In addition, we outline some of the problems that could be at the basis of the negative outcome of the trials with ghrelin agonists and question whether the right target groups were selected. It is clear that a new approach is needed to develop marketable drugs with this class of gastroprokinetic agents.


Assuntos
Motilidade Gastrointestinal/fisiologia , Grelina/metabolismo , Mucosa Intestinal/metabolismo , Transdução de Sinais/fisiologia , Animais , Fármacos Gastrointestinais/farmacologia , Gastroenteropatias/tratamento farmacológico , Motilidade Gastrointestinal/efeitos dos fármacos , Grelina/farmacologia , Humanos , Receptores de Grelina/agonistas , Transdução de Sinais/efeitos dos fármacos
17.
Neurogastroenterol Motil ; 25(9): e599-608, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23781841

RESUMO

BACKGROUND: Ghrelin is the only known peripherally active orexigenic hormone produced by the stomach that activates vagal afferents to stimulate food intake and to accelerate gastric emptying. Vagal sensory neurons within the nodose ganglia are surrounded by glial cells, which are able to receive and transmit chemical signals. We aimed to investigate whether ghrelin activates or influences the interaction between both types of cells. The effect of ghrelin was compared with that of leptin and cholecystokinin (CCK). METHODS: Cultures of rat nodose ganglia were characterized by immunohistochemistry and the functional effects of peptides, neurotransmitters, and pharmacological blockers were measured by Ca(2+) imaging using Fluo-4-AM as an indicator. KEY RESULTS: Neurons responded to KCl and were immunoreactive for PGP-9.5 whereas glial cells responded to lysophosphatidic acid and had the typical SOX-10-positive nuclear staining. Neurons were only responsive to CCK (31 ± 5%) whereas glial cells responded equally to the applied stimuli: ghrelin (27 ± 2%), leptin (21 ± 2%), and CCK (30 ± 2%). In contrast, neurons stained more intensively for the ghrelin receptor than glial cells. ATP induced [Ca(2+) ]i rises in 90% of the neurons whereas ACh and the NO donor, SIN-1, mainly induced [Ca(2+) ]i changes in glial cells (41 and 51%, respectively). The percentage of ghrelin-responsive glial cells was not affected by pretreatment with suramin, atropine, hexamethonium or 1400 W, but was reduced by l-NAME and by tetrodotoxin. Neurons were shown to be immunoreactive for neuronal NO-synthase (nNOS). CONCLUSIONS & INFERENCES: Our data show that ghrelin induces Ca(2+) signaling in glial cells of the nodose ganglion via the release of NO originating from the neurons.


Assuntos
Sinalização do Cálcio/fisiologia , Grelina/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Comunicação Parácrina/fisiologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Imunofluorescência , Grelina/farmacologia , Imuno-Histoquímica , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Gânglio Nodoso , Comunicação Parácrina/efeitos dos fármacos , Ratos , Ratos Wistar
18.
Neurogastroenterol Motil ; 25(8): e540-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23711101

RESUMO

BACKGROUND: The severity of postoperative ileus (POI) has been reported to result from decreased contractility of the muscularis inversely related to the number of infiltrating leukocytes. However, we previously observed that the severity of POI is independent of the number of infiltrating leukocytes, indicating that different mechanisms must be involved. Here, we hypothesize that the degree of tissue damage in response to intestinal handling determines the upregulation of local cytokine production and correlates with the severity of POI. METHODS: Intestinal transit, the inflammatory response, I-FABP (marker for tissue damage) levels and brain activation were determined after different intensities of intestinal handling. KEY RESULTS: Intense handling induced a more pronounced ileus compared with gentle intestinal manipulation (IM). No difference in leukocytic infiltrates in the handled and non-handled parts of the gut was observed between the two intensities of intestinal handling. However, intense handling resulted in significantly more tissue damage and was accompanied by a systemic inflammation with increased plasma levels of pro-inflammatory cytokines. In addition, intense but not gentle handling triggered enhanced c-Fos expression in the nucleus of the solitary tract (NTS) and area postrema (AP). In patients, plasma levels of I-FABP and inflammatory cytokines were significantly higher after open compared with laparoscopic surgery, and were associated with more severe POI. CONCLUSIONS & INFERENCES: Not the influx of leukocytes, rather the manipulation-induced damage and subsequent inflammatory response determine the severity of POI. The release of tissue damage mediators and pro-inflammatory cytokines into the systemic circulation most likely contribute to the impaired motility of non-manipulated intestine.


Assuntos
Encéfalo/metabolismo , Íleus/metabolismo , Mediadores da Inflamação/fisiologia , Complicações Pós-Operatórias/metabolismo , Índice de Gravidade de Doença , Animais , Trânsito Gastrointestinal/fisiologia , Humanos , Íleus/patologia , Camundongos Endogâmicos C57BL , Complicações Pós-Operatórias/patologia , Fatores de Tempo
19.
Neurogastroenterol Motil ; 25(2): e151-60, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23279281

RESUMO

BACKGROUND: The intimate association between glial cells and neurons within the enteric nervous system has confounded careful examination of the direct responsiveness of enteric glia to different neuroligands. Therefore, we aimed to investigate whether neurotransmitters known to elicit fast excitatory potentials in enteric nerves also activate enteric glia directly. METHODS: We studied the effect of acetylcholine (ACh), serotonin (5-HT), and adenosine triphosphate (ATP) on intracellular Ca(2+) signaling using aequorin-expressing and Fluo-4 AM-loaded CRL-2690 rat and human enteric glial cell cultures devoid of neurons. The influence of these neurotransmitters on the proliferation of glia was measured and their effect on the expression of c-Fos as well as glial fibrillary acidic protein (GFAP), Sox10, and S100 was examined by immunohistochemistry and quantitative RT-PCR. KEY RESULTS: Apart from ATP, also ACh and 5-HT induced a dose-dependent increase in intracellular Ca(2+) concentration in CRL-2690 cells. Similarly, these neurotransmitters also evoked Ca(2+) transients in human primary enteric glial cells obtained from mucosal biopsies. In contrast with ATP, stimulation with ACh and 5-HT induced early gene expression in CRL-2690 cells. The proliferation of enteric glia and their expression of GFAP, Sox10, and S100 were not affected following stimulation with these neurotransmitters. CONCLUSIONS & INFERENCES: We provide evidence that enteric glial cells respond to fast excitatory neurotransmitters by changes in intracellular Ca(2+). On the basis of our experimental in vitro setting, we show that enteric glia are not only directly responsive to purinergic but also to serotonergic and cholinergic signaling mechanisms.


Assuntos
Sistema Nervoso Entérico/fisiologia , Neuroglia/metabolismo , Neurotransmissores/metabolismo , Transmissão Sináptica/fisiologia , Animais , Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Sistema Nervoso Entérico/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Neuroglia/efeitos dos fármacos , Neurotransmissores/farmacologia , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transmissão Sináptica/efeitos dos fármacos
20.
Oral Dis ; 18(5): 501-5, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22273265

RESUMO

OBJECTIVE: To investigate the expression and distribution of AQP5 in submandibular acinar cells from sham- and streptozotocin (STZ)-treated mice in relation to the salivary flow. METHODS: Mice were sham or STZ injected. Distribution of AQP5 subcellular expression in submandibular glands was determined by immunohistochemistry. AQP5 labelling indices (LI), reflecting AQP5 subcellular distribution, were determined in acinar cells. Western blotting was performed to determine the expression of AQP5 in submandibular glands. Blood glycaemia and osmolality and saliva flow rates were also determined. RESULTS: AQP5 immunoreactivity was primarily located at the apical and apical-basolateral membranes of submandibular gland acinar cells from sham- and STZ-treated mice. No significant differences in AQP5 protein levels were observed between sham- and STZ-treated mice. Compared to sham-treated mice, STZ-treated mice had significant increased glycaemia, while no significant differences in blood osmolality were observed. Saliva flow rate was significantly decreased in STZ-treated mice as compared to sham-treated mice. CONCLUSIONS: In STZ-treated mice, significant reduction in salivary flow rate was observed without any concomitant modification in AQP5 expression and localization.


Assuntos
Aquaporina 5/genética , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/fisiopatologia , Saliva/metabolismo , Células Acinares/metabolismo , Animais , Aquaporina 5/biossíntese , Membrana Celular/química , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/genética , Modelos Animais de Doenças , Hiperglicemia/fisiopatologia , Masculino , Camundongos , Concentração Osmolar , Taxa Secretória , Estreptozocina , Glândula Submandibular/fisiologia , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...