Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Acta Biochim Pol ; 69(3): 579-586, 2022 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-36057985

RESUMEN

OBJECTIVE: To explore the mechanism by which miR-129-3p affected the autophagy of interstitial cells of Cajal (ICCs) in slow transit constipation tissues through the SCF C-kit signaling pathway. METHODS: Colon samples from 20 Slow transit constipation (STC) patients who underwent total colectomy plus ileorectal anastomosis or subtotal colon resection plus anti-peristaltic rectal anastomosis were collected in our hospital. The colon of 20 non-STC patients was used as control. The control of this study was 20 patients undergoing radical surgery for colon cancer (left colon cancer) in our hospital. Fifty healthy SPF Kunming mice were purchased from Liaoning Changsheng Biotechnology Co., Ltd. RESULTS: The mRNA expression of miR-129-3p in the STC group was lower than that in the control group (CTLR) group (P<0.05). The mRNA expression of miR-129-3p in STC group was lower than that in the NC group (P<0.05), and mRNA expression in STC+miR-129-3p group was higher than that in STC+miR-NC group (P<0.05). In the first week, the weight of dry and wet feces of the STC group was lower than that of the NC mice (P<0.05), and the weight of dry feces and wet feces of the STC group was lower than that of the NC group at the 2, 3, and 4 weeks, STC+miR-129 -3p was higher than that in the STC group (P<0.05). CONCLUSION: The increased expression of C-kit and SCF regulated by miR-129-3p contributed to the protection of interstitial cells. Knockdown of miR-129-3p expression could inhibit the activation of AKT/mTOR signaling pathway, reduce cell proliferation activity.


Asunto(s)
Neoplasias del Colon , Células Intersticiales de Cajal , MicroARNs , Animales , Autofagia , Neoplasias del Colon/metabolismo , Estreñimiento/genética , Estreñimiento/metabolismo , Tránsito Gastrointestinal/genética , Humanos , Células Intersticiales de Cajal/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit , ARN Mensajero/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
2.
Sci Rep ; 11(1): 22462, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789790

RESUMEN

We examined the relationship between zonulin and gastric motility in critical care patients and a translational mouse model of systemic inflammation. Gastric motility and haptoglobin (HP) 2 isoform quantification, proxy for zonulin, were examined in patients. Inflammation was triggered by lipopolysaccharide (LPS) injection in C57Bl/6 zonulin transgenic mouse (Ztm) and wildtype (WT) mice as controls, and gastro-duodenal transit was examined by fluorescein-isothiocyanate, 6 and 12 h after LPS-injection. Serum cytokines and zonulin protein levels, and zonulin gastric-duodenal mRNA expression were examined. Eight of 20 patients [14 years, IQR (12.25, 18)] developed gastric dysmotility and were HP2 isoform-producing. HP2 correlated with gastric dysmotility (r = - 0.51, CI - 0.81 to 0.003, p = 0.048). LPS injection induced a time-dependent increase in IL-6 and KC-Gro levels in all mice (p < 0.0001). Gastric dysmotility was reduced similarly in Ztm and WT mice in a time-dependent manner. Ztm had 16% faster duodenal motility than WT mice 6H post-LPS, p = 0.01. Zonulin mRNA expression by delta cycle threshold (dCT) was higher in the stomach (9.7, SD 1.4) than the duodenum (13.9, SD 1.4) 6H post-LPS, p = 0.04. Serum zonulin protein levels were higher in LPS-injected mice compared to vehicle-injected animals in a time-dependent manner. Zonulin correlated with gastric dysmotility in patients. A mouse model had time-dependent gastro-duodenal dysmotility after LPS-injection that paralleled zonulin mRNA expression and protein levels.


Asunto(s)
Vaciamiento Gástrico/genética , Tránsito Gastrointestinal/genética , Haptoglobinas/metabolismo , Precursores de Proteínas/sangre , Sepsis/sangre , Adolescente , Animales , Niño , Estudios de Cohortes , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Mucosa Gástrica/metabolismo , Haptoglobinas/genética , Humanos , Mucosa Intestinal/metabolismo , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Precursores de Proteínas/genética , Sepsis/inducido químicamente
3.
Neurogastroenterol Motil ; 33(12): e14190, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34190380

RESUMEN

BACKGROUND: The breakpoint cluster region (BCR) is a protein that originally forms a fusion protein with c-Abl tyrosine kinase and induces leukemia. Researchers have shown that BCR is enriched in the central nervous system and may contribute to neurological disorders. We aimed to investigate the physiological function of BCR in neural development in the gastrointestinal (GI) tract and brain. METHODS: Whole-exome sequencing was used to screen for mutations in the BCR. Bcr knockout mice (Bcr-/- , ΔExon 2-22) were generated using the CRISPR/Cas9 system. Transit of carmine red dye and glass bead expulsion assays were used to record total and proximal GI transit and distal colonic transit. KEY RESULTS: In an infant with pediatric intestinal pseudo-obstruction, we found a heterozygous de novo mutation (NM_004327.3:c.3072+1G>A) in BCR. Bcr deficiency mice (Bcr-/- ) exhibited growth retardation and impaired gastrointestinal motility. Bcr-/- mice had a prolonged average total GI transit time with increased distal colonic transit and proximal GI transit in isolation. Morphology analysis indicated that Bcr-/- mice had a less number of neurons in the submucosal plexus and myenteric plexus. Bcr-/- mice exhibited apparent structural defects in the brain, particularly in the cortex. Additionally, Bcr- depletion in the mouse cortex altered the expression of Ras homologous (Rho) family small GTPases. CONCLUSIONS AND INFERENCES: BCR mutations are associated with intestinal obstruction in children. Loss of Bcr can cause intestinal dysmotility and brain developmental defects may via regulation of Rho GTPases.


Asunto(s)
Encéfalo/metabolismo , Enfermedades Gastrointestinales/genética , Motilidad Gastrointestinal/genética , Seudoobstrucción Intestinal/genética , Proteínas Proto-Oncogénicas c-bcr/genética , Animales , Femenino , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/fisiopatología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/fisiopatología , Tránsito Gastrointestinal/genética , Humanos , Seudoobstrucción Intestinal/metabolismo , Seudoobstrucción Intestinal/patología , Ratones , Ratones Noqueados , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-bcr/metabolismo
4.
Gut ; 70(9): 1665-1674, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33722860

RESUMEN

BACKGROUND AND AIMS: Gut transit time is a key modulator of host-microbiome interactions, yet this is often overlooked, partly because reliable methods are typically expensive or burdensome. The aim of this single-arm, single-blinded intervention study is to assess (1) the relationship between gut transit time and the human gut microbiome, and (2) the utility of the 'blue dye' method as an inexpensive and scalable technique to measure transit time. METHODS: We assessed interactions between the taxonomic and functional potential profiles of the gut microbiome (profiled via shotgun metagenomic sequencing), gut transit time (measured via the blue dye method), cardiometabolic health and diet in 863 healthy individuals from the PREDICT 1 study. RESULTS: We found that gut microbiome taxonomic composition can accurately discriminate between gut transit time classes (0.82 area under the receiver operating characteristic curve) and longer gut transit time is linked with specific microbial species such as Akkermansia muciniphila, Bacteroides spp and Alistipes spp (false discovery rate-adjusted p values <0.01). The blue dye measure of gut transit time had the strongest association with the gut microbiome over typical transit time proxies such as stool consistency and frequency. CONCLUSIONS: Gut transit time, measured via the blue dye method, is a more informative marker of gut microbiome function than traditional measures of stool consistency and frequency. The blue dye method can be applied in large-scale epidemiological studies to advance diet-microbiome-health research. Clinical trial registry website https://clinicaltrials.gov/ct2/show/NCT03479866 and trial number NCT03479866.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Tránsito Gastrointestinal , Adulto , Akkermansia , Bacteroides , Bacteroidetes , Biomarcadores , Colorantes , Heces/microbiología , Femenino , Tránsito Gastrointestinal/genética , Tránsito Gastrointestinal/fisiología , Humanos , Masculino , Metagenómica , Persona de Mediana Edad
5.
Gastroenterology ; 160(7): 2451-2466.e19, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33662386

RESUMEN

BACKGROUND & AIMS: Gastrointestinal (GI) motility is regulated by serotonin (5-hydroxytryptamine [5-HT]), which is primarily produced by enterochromaffin (EC) cells in the GI tract. However, the precise roles of EC cell-derived 5-HT in regulating gastric motility remain a major point of conjecture. Using a novel transgenic mouse line, we investigated the distribution of EC cells and the pathophysiologic roles of 5-HT deficiency in gastric motility in mice and humans. METHODS: We developed an inducible, EC cell-specific Tph1CreERT2/+ mouse, which was used to generate a reporter mouse line, Tph1-tdTom, and an EC cell-depleted line, Tph1-DTA. We examined EC cell distribution, morphology, and subpopulations in reporter mice. GI motility was measured in vivo and ex vivo in EC cell-depleted mice. Additionally, we evaluated 5-HT content in biopsy and plasma specimens from patients with idiopathic gastroparesis (IG). RESULTS: Tph1-tdTom mice showed EC cells that were heterogeneously distributed throughout the GI tract with the greatest abundance in the antrum and proximal colon. Two subpopulations of EC cells were identified in the gut: self-renewal cells located at the base of the crypt and mature cells observed in the villi. Tph1-DTA mice displayed delayed gastric emptying, total GI transit, and colonic transit. These gut motility alterations were reversed by exogenous provision of 5-HT. Patients with IG had a significant reduction of antral EC cell numbers and 5-HT content, which negatively correlated with gastric emptying rate. CONCLUSIONS: The Tph1CreERT2/+ mouse provides a powerful tool to study the functional roles of EC cells in the GI tract. Our findings suggest a new pathophysiologic mechanism of 5-HT deficiency in IG.


Asunto(s)
Vaciamiento Gástrico/genética , Tránsito Gastrointestinal/genética , Serotonina/deficiencia , Animales , Línea Celular , Células Enterocromafines/fisiología , Humanos , Ratones , Ratones Transgénicos , Triptófano Hidroxilasa/metabolismo
6.
Sci Rep ; 10(1): 11521, 2020 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-32661417

RESUMEN

Intestinal ischemia/reperfusion (I/R) injury has severe consequences on myenteric neurons, which can be irreversibly compromised resulting in slowing of transit and hindered food digestion. Myenteric neurons synthesize hyaluronan (HA) to form a well-structured perineuronal net, which undergoes derangement when myenteric ganglia homeostasis is perturbed, i.e. during inflammation. In this study we evaluated HA involvement in rat small intestine myenteric plexus after in vivo I/R injury induced by clamping a branch of the superior mesenteric artery for 60 min, followed by 24 h of reperfusion. In some experiments, 4-methylumbelliferone (4-MU, 25 mg/kg), a HA synthesis inhibitor, was intraperitoneally administered to normal (CTR), sham-operated (SH) and I/R animals for 24 h. In longitudinal muscle myenteric plexus (LMMP) whole-mount preparations, HA binding protein staining as well as HA levels were significantly higher in the I/R group, and were reduced after 4-MU treatment. HA synthase 1 and 2 (HAS1 and HAS2) labelled myenteric neurons and mRNA levels in LMMPs increased in the I/R group with respect to CTR, and were reduced by 4-MU. The efficiency of the gastrointestinal transit was significantly reduced in I/R and 4-MU-treated I/R groups with respect to CTR and SH groups. In the 4-MU-treated I/R group gastric emptying was reduced with respect to the CTR, SH and I/R groups. Carbachol (CCh) and electrical field (EFS, 0.1-40 Hz) stimulated contractions and EFS-induced (10 Hz) NANC relaxations were reduced in the I/R group with respect to both CTR and SH groups. After I/R, 4-MU treatment increased EFS contractions towards control values, but did not affect CCh-induced contractions. NANC on-relaxations after I/R were not influenced by 4-MU treatment. Main alterations in the neurochemical coding of both excitatory (tachykinergic) and inhibitory pathways (iNOS, VIPergic) were also observed after I/R, and were influenced by 4-MU administration. Overall, our data suggest that, after an intestinal I/R damage, changes of HA homeostasis in specific myenteric neuron populations may influence the efficiency of the gastrointestinal transit. We cannot exclude that modulation of HA synthesis in these conditions may ameliorate derangement of the enteric motor function preventing, at least in part, the development of dysmotility.


Asunto(s)
Tránsito Gastrointestinal/fisiología , Ácido Hialurónico/metabolismo , Intestino Delgado/metabolismo , Daño por Reperfusión/metabolismo , Animales , Modelos Animales de Enfermedad , Ganglios/metabolismo , Motilidad Gastrointestinal/genética , Motilidad Gastrointestinal/fisiología , Tránsito Gastrointestinal/genética , Humanos , Hialuronano Sintasas/genética , Íleon/metabolismo , Íleon/fisiología , Intestino Delgado/patología , Plexo Mientérico/metabolismo , Fenómenos Fisiológicos del Sistema Nervioso , Neuronas/metabolismo , Neuronas/patología , Ratas , Daño por Reperfusión/genética , Daño por Reperfusión/patología
7.
World J Gastroenterol ; 26(7): 717-724, 2020 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-32116419

RESUMEN

BACKGROUND: Chronic constipation is a gastrointestinal functional disease that seriously harms physical and mental health and impacts the quality of life of patients. Its incidence rate is 2%-27%. Slow transit constipation (STC) is a common type of chronic functional constipation, accounting for 10.3%-45.5% of such cases. Scholars have performed many studies on the pathogenesis of STC. These studies have indicated that the occurrence of STC may be related to multiple factors, such as dysfunction of the enteric nervous system, interstitial cells of Cajal (ICC) damage, and changes in neurotransmitters regulating intestinal peristalsis. AIM: To investigate the role of Tenascin-X (TNX) in regulating the TGF-ß/Smad signaling pathway in the pathogenesis of STC. METHODS: This study included an experimental group and a control group. The experimental group included 28 patients with severe colonic STC, and the control group included 18 patients with normal colon tissues. Immunohistochemistry (IHC) was used to detect c-Kit, a specific marker of the ICC. Western blot, immunofluorescence, and IHC were used to detect the localization and expression of TNX and TGF-ß/Smad. RESULTS: IHC showed that the number of ICC with positive c-Kit expression was significantly reduced in the colon of STC patients (22.17 ± 3.28 vs 28.69 ± 3.53, P < 0.05) and that the distribution was abnormal. Western blot results showed that c-Kit and Smad7 levels were significantly decreased in the colon of STC patients (c-kit: 0.462 ± 0.099 vs 0.783 ± 0.178, P < 0.01; Smad7: 0.626 ± 0.058 vs 0.799 ± 0.03, P < 0.01) and that TNX and Smad2/3 levels were higher in the STC group (TNX: 0.868 ± 0.028 vs 0.482 ± 0.032, P < 0.01). There was no significant difference in TGF-ß between the two groups (0.476 ± 0.028 vs 0.511 ± 0.044, P = 0.272). Pearson correlation analysis showed that the TNX protein exhibited a strong correlation with Smad2/3 and Smad7 (P < 0.05, |R| > 0.8) and TGF-ß (P < 0.05, |R| = 0.7). CONCLUSION: The extracellular matrix protein TNX may activate the TGF-ß/Smad signaling pathway by upregulating the Smad 2/3 signaling protein and thereby induce slight or complete epithelial stromal cell transformation, leading to an abnormal distribution and dysfunction of ICC in the diseased colon, which promotes the occurrence and development of STC.


Asunto(s)
Estreñimiento/genética , Transducción de Señal/genética , Proteínas Smad/metabolismo , Tenascina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Anciano , Western Blotting , Estudios de Casos y Controles , Colon/metabolismo , Matriz Extracelular/metabolismo , Femenino , Tránsito Gastrointestinal/genética , Humanos , Células Intersticiales de Cajal/metabolismo , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-kit/metabolismo
8.
Autism Res ; 12(7): 1043-1056, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31119867

RESUMEN

Gastrointestinal (GI) problems constitute an important comorbidity in many patients with autism. Multiple mutations in the neuroligin family of synaptic adhesion molecules are implicated in autism, however whether they are expressed and impact GI function via changes in the enteric nervous system is unknown. We report the GI symptoms of two brothers with autism and an R451C mutation in Nlgn3 encoding the synaptic adhesion protein, neuroligin-3. We confirm the presence of an array of synaptic genes in the murine GI tract and investigate the impact of impaired synaptic protein expression in mice carrying the human neuroligin-3 R451C missense mutation (NL3R451C ). Assessing in vivo gut dysfunction, we report faster small intestinal transit in NL3R451C compared to wild-type mice. Using an ex vivo colonic motility assay, we show increased sensitivity to GABAA receptor modulation in NL3R451C mice, a well-established Central Nervous System (CNS) feature associated with this mutation. We further show increased numbers of small intestine myenteric neurons in NL3R451C mice. Although we observed altered sensitivity to GABAA receptor modulators in the colon, there was no change in colonic neuronal numbers including the number of GABA-immunoreactive myenteric neurons. We further identified altered fecal microbial communities in NL3R451C mice. These results suggest that the R451C mutation affects small intestinal and colonic function and alter neuronal numbers in the small intestine as well as impact fecal microbes. Our findings identify a novel GI phenotype associated with the R451C mutation and highlight NL3R451C mice as a useful preclinical model of GI dysfunction in autism. Autism Res 2019, 12: 1043-1056. © 2019 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: People with autism commonly experience gastrointestinal problems, however the cause is unknown. We report gut symptoms in patients with the autism-associated R451C mutation encoding the neuroligin-3 protein. We show that many of the genes implicated in autism are expressed in mouse gut. The neuroligin-3 R451C mutation alters the enteric nervous system, causes gastrointestinal dysfunction, and disrupts gut microbe populations in mice. Gut dysfunction in autism could be due to mutations that affect neuronal communication.


Asunto(s)
Trastorno Autístico/genética , Moléculas de Adhesión Celular Neuronal/genética , Análisis Mutacional de ADN , Enfermedades Gastrointestinales/genética , Expresión Génica/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Animales , Comorbilidad , Enfermedades Gastrointestinales/fisiopatología , Microbioma Gastrointestinal/genética , Tránsito Gastrointestinal/genética , Humanos , Masculino , Ratones , Plexo Mientérico/fisiopatología , Neuronas/fisiología , Fenotipo
9.
J Gastroenterol ; 54(5): 407-418, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30382364

RESUMEN

BACKGROUND: Inositol 1,4,5-trisphosphate receptors (IP3Rs) are a family of intracellular Ca2+ release channels located on the membrane of endoplasmic reticulum, which have been shown to play critical roles in various cellular and physiological functions. However, their function in regulating gastrointestinal (GI) tract motility in vivo remains unknown. Here, we investigated the physiological function of IP3R1 in the GI tract using genetically engineered mouse models. METHODS: Pdgfrb-Cre mice were bred with homozygous Itpr1 floxed (Itpr1f/f) mice to generate conditional IP3R1 knockout (pcR1KO) mice. Cell lineage tracing was used to determine where Pdgfrb-Cre-mediated gene deletion occurred in the GI tract. Isometric tension recording was used to measure the effects of IP3R1 deletion on muscle contraction. RESULTS: In the mouse GI tract, Itpr1 gene deletion by Pdgfrb-Cre occurred in smooth muscle cells, enteric neurons, and interstitial cells of Cajal. pcR1KO mice developed impaired GI motility, with prolonged whole-gut transit time and abdominal distention. pcR1KO mice also exhibited lethality as early as 8 weeks of age and 50% of pcR1KO mice were dead by 40 weeks after birth. The frequency of spontaneous contractions in colonic circular muscles was dramatically decreased and the amplitude of spontaneous contractions was increased in pcR1KO mice. Deletion of IP3R1 in the GI tract also reduced the contractile response to the muscarinic agonist, carbachol, as well as to electrical field stimulation. However, KCl-induced contraction and expression of smooth muscle-specific contractile genes were not significantly altered in pcR1KO mice. CONCLUSIONS: Here, we provided a novel mouse model for impaired GI motility and demonstrated that IP3R1 plays a critical role in regulating physiological function of GI tract in vivo.


Asunto(s)
Tránsito Gastrointestinal/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Seudoobstrucción Intestinal/genética , Contracción Muscular/genética , Animales , Carbacol/farmacología , Agonistas Colinérgicos/farmacología , Colon/efectos de los fármacos , Colon/patología , Colon/fisiopatología , Modelos Animales de Enfermedad , Integrasas/genética , Células Intersticiales de Cajal , Seudoobstrucción Intestinal/fisiopatología , Ratones , Ratones Noqueados , Contracción Muscular/efectos de los fármacos , Músculo Liso/fisiopatología , Miocitos del Músculo Liso , Neuronas , Cloruro de Potasio/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Tasa de Supervivencia
10.
Gastroenterology ; 155(2): 514-528.e6, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29782847

RESUMEN

BACKGROUND & AIMS: Strategies are needed to increase gastrointestinal transit without systemic pharmacologic agents. We investigated whether optogenetics, focal application of light to control enteric nervous system excitability, could be used to evoke propagating contractions and increase colonic transit in mice. METHODS: We generated transgenic mice with Cre-mediated expression of light-sensitive channelrhodopsin-2 (ChR2) in calretinin neurons (CAL-ChR2 Cre+ mice); Cre- littermates served as controls. Colonic myenteric neurons were analyzed by immunohistochemistry, patch-clamp, and calcium imaging studies. Motility was assessed by mechanical, electrophysiological, and video recording in vitro and by fecal output in vivo. RESULTS: In isolated colons, focal light stimulation of calretinin enteric neurons evoked classic polarized motor reflexes (50/58 stimulations), followed by premature anterograde propagating contractions (39/58 stimulations). Light stimulation could evoke motility from sites along the entire colon. These effects were prevented by neural blockade with tetrodotoxin (n = 2), and did not occur in control mice (n = 5). Light stimulation of proximal colon increased the proportion of natural fecal pellets expelled over 15 minutes in vitro (75% ± 17% vs 32% ± 8% for controls) (P < .05). In vivo, activation of wireless light-emitting diodes implanted onto the colon wall significantly increased hourly fecal pellet output in conscious, freely moving mice (4.2 ± 0.4 vs 1.3 ± 0.3 in controls) (P < .001). CONCLUSIONS: In studies of mice, we found that focal activation of a subset of enteric neurons can increase motility of the entire colon in vitro, and fecal output in vivo. Optogenetic control of enteric neurons might therefore be used to modify gut motility.


Asunto(s)
Colon/fisiología , Sistema Nervioso Entérico/fisiología , Tránsito Gastrointestinal/efectos de la radiación , Luz , Optogenética/métodos , Animales , Calbindina 2/genética , Calbindina 2/metabolismo , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Channelrhodopsins/efectos de la radiación , Colon/inervación , Colon/efectos de la radiación , Sistema Nervioso Entérico/citología , Tránsito Gastrointestinal/genética , Ratones , Ratones Transgénicos , Modelos Animales , Neuronas/metabolismo , Neuronas/efectos de la radiación
11.
Fish Physiol Biochem ; 44(1): 119-135, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28894993

RESUMEN

Growth hormone (GH) transgenic (T) coho salmon consistently show remarkably enhanced growth associated with increased appetite and food consumption compared to non-transgenic wild-type (NT) coho salmon. To improve understanding of the mechanism by which GH overexpression mediates food intake and digestion in T fish, feed intake and gastric evacuation rate (over 7 days) were measured in size-matched T and NT coho salmon. T fish displayed greatly enhanced feed intake levels (~ 2.5-fold), and more than 3-fold increase in gastric evacuation rates relative to NT coho salmon. Despite the differences in feed intake, no differences were noted in the time taken from first ingestion of food to stomach evacuation between genotypes. These results indicate that enhanced feed intake is coupled with an overall increased processing rate to enhance energy intake by T fish. To further investigate the molecular basis of these responses, we examined the messenger RNA (mRNA) levels of several genes in appetite- and gastric-regulation pathways (Agrp1, Bbs, Cart, Cck, Glp, Ghrelin, Grp, Leptin, Mc4r, Npy, and Pomc) by qPCR analyses in the brain (hypothalamus, preoptic area) and pituitary, and in peripheral tissues associated with digestion (liver, stomach, intestine, and adipose tissue). Significant increases in mRNA levels were found for Agrp1 in the preoptic area (POA) of the brain, and Grp and Pomc in pituitary for T coho salmon relative to NT. Mch and Npy showed significantly lower mRNA levels than NT fish in all brain tissues examined across all time-points after feeding. Mc4r and Cart for T showed significantly lower mRNA levels than NT in the POA and hypothalamus, respectively. In the case of peripheral tissues, T fish had lower mRNA levels of Glp and Leptin than NT fish in the intestine and adipose tissue, respectively. Grp, Cck, Bbs, Glp, and Leptin in stomach, adipose tissue, and/or intestine showed significant differences across the time-points after feeding, but Ghrelin showed no significant difference between T and NT fish in all tested tissues.


Asunto(s)
Animales Modificados Genéticamente , Tránsito Gastrointestinal/genética , Hormona del Crecimiento/genética , Oncorhynchus kisutch/genética , Animales , Digestión/fisiología , Conducta Alimentaria/fisiología , Tránsito Gastrointestinal/fisiología , Genotipo , Hormona del Crecimiento/metabolismo , Oncorhynchus kisutch/fisiología
12.
Radiología (Madr., Ed. impr.) ; 57(1): 9-21, ene.-feb. 2015. tab, ilus
Artículo en Español | IBECS | ID: ibc-136631

RESUMEN

El radiólogo debe ser capaz de reconocer los signos de la malrotación intestinal en la imagen al tratarse de una entidad patológica con complicaciones potencialmente letales, como el vólvulo de intestino medio. Para diagnosticarla correctamente, es tan importante que exista un índice de sospecha clínica elevado como que el radiólogo sepa reconocer los signos específicos de malrotación y las variantes de la normalidad que pueden conducir a un diagnóstico erróneo. Aunque la posición no retroperitoneal de la tercera porción duodenal en ecografía, TC o RM parece ser un signo fiable para el diagnóstico, el tránsito gastrointestinal continúa siendo el estándar de referencia para ver la unión duodeno-yeyunal en una posición anómala. Nuestro objetivo es revisar los principales signos radiológicos de esta enfermedad y hacer hincapié en el papel de la ecografía para diagnosticar el vólvulo de intestino medio (AU)


Radiologists must be able to recognize the imaging signs of intestinal malrotation because this condition can lead to potentially lethal complications such as midgut volvulus. The correct diagnosis depends on both high clinical suspicion and the radiologist's ability to recognize the specific signs of malrotation and the normal variants that can lead to the wrong diagnosis. Although the location of the third portion of the duodenum outside the retroperitoneal area on ultrasonography, CT, or MRI seems to be a reliable sign of malrotation, the gold standard for determining whether the duodenojejunal flexure is in an abnormal location continues to be the upper gastrointestinal series. In this article, we review the most important imaging signs of malrotation and emphasize the role of ultrasonography in diagnosing midgut volvulus (AU)


Asunto(s)
Femenino , Humanos , Masculino , Obstrucción Duodenal/complicaciones , Obstrucción Duodenal , Vólvulo Intestinal/complicaciones , Intestino Delgado/patología , Ciego/patología , Ciego , Vólvulo Intestinal/patología , Vólvulo Intestinal , Tránsito Gastrointestinal/genética , Enema
13.
Am J Physiol Gastrointest Liver Physiol ; 306(1): G13-26, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24200957

RESUMEN

The study objectives were: to mine the complete exome to identify putative rare single nucleotide variants (SNVs) associated with irritable bowel syndrome (IBS)-diarrhea (IBS-D) phenotype, to assess genes that regulate bile acids in IBS-D, and to explore univariate associations of SNVs with symptom phenotype and quantitative traits in an independent IBS cohort. Using principal components analysis, we identified two groups of IBS-D (n = 16) with increased fecal bile acids: rapid colonic transit or high bile acids synthesis. DNA was sequenced in depth, analyzing SNVs in bile acid genes (ASBT, FXR, OSTα/ß, FGF19, FGFR4, KLB, SHP, CYP7A1, LRH-1, and FABP6). Exome findings were compared with those of 50 similar ethnicity controls. We assessed univariate associations of each SNV with quantitative traits and a principal components analysis and associations between SNVs in KLB and FGFR4 and symptom phenotype in 405 IBS, 228 controls and colonic transit in 70 IBS-D, 71 IBS-constipation. Mining the complete exome did not reveal significant associations with IBS-D over controls. There were 54 SNVs in 10 of 11 bile acid-regulating genes, with no SNVs in FGF19; 15 nonsynonymous SNVs were identified in similar proportions of IBS-D and controls. Variations in KLB (rs1015450, downstream) and FGFR4 [rs434434 (intronic), rs1966265, and rs351855 (nonsynonymous)] were associated with colonic transit (rs1966265; P = 0.043), fecal bile acids (rs1015450; P = 0.064), and principal components analysis groups (all 3 FGFR4 SNVs; P < 0.05). In the 633-person cohort, FGFR4 rs434434 was associated with symptom phenotype (P = 0.027) and rs1966265 with 24-h colonic transit (P = 0.066). Thus exome sequencing identified additional variants in KLB and FGFR4 associated with bile acids or colonic transit in IBS-D.


Asunto(s)
Ácidos y Sales Biliares , Diarrea/genética , Tránsito Gastrointestinal/genética , Síndrome del Colon Irritable/genética , Proteínas de la Membrana/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Adulto , Secuencia de Bases , Ácidos y Sales Biliares/biosíntesis , Ácidos y Sales Biliares/genética , Ácidos y Sales Biliares/metabolismo , Transporte Biológico/genética , Diarrea/etiología , Diarrea/fisiopatología , Exoma , Heces/química , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Síndrome del Colon Irritable/complicaciones , Síndrome del Colon Irritable/fisiopatología , Proteínas Klotho , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
14.
J Gastroenterol Hepatol ; 29(2): 276-83, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23926887

RESUMEN

BACKGROUND: Constipation is one of the most common chronic digestive complaints. Gastrointestinal transit studies have divided it into three patterns: normal transit, slow transit constipation (STC), and outlet obstruction. It has been demonstrated that STC patients respond poorly to standard therapies, and the etiology of STC remains poorly understood. Animal studies have also shown that fatty acid amide hydrolase (FAAH) controls intestinal motility through its putative receptors or non-receptor-mediated pathways. However, the role of FAAH in STC has not been elaborated. METHODS: A case series was carried out on thirty-two STC patients fulfilling the Rome II criteria and on 24 controls. All of the subjects underwent a laparotomy in Shengjing Hospital. Colonic specimens were obtained and used for FAAH expression analysis, enzyme activity assay, and cannabinoid detection. RESULTS: FAAH immunoreactivity occurred in the enteric neurons and in the surface epithelial and glands. The expression level and enzyme activity of FAAH in the STC group were both significantly lower than those in the control group (P < 0.05). The amounts of anandamide, 2-arachidonylglycerol, and palmitoylethanolamide, which are negatively correlated with enzyme activity, were significantly higher in the constipation group than that in the control group. In the STC group, cannabinoid receptor type 1 immunoreactivity occurred predominantly in the submucosal and myenteric fibers that were obviously strong and wave-like in their appearance. Enteric ganglions decreased or disappeared. CONCLUSIONS: The tone of the enteric cannabinoids system is disturbed in STC, and the decreased enteric FAAH activity contributes to colonic inertia in STC.


Asunto(s)
Amidohidrolasas/metabolismo , Amidohidrolasas/fisiología , Estreñimiento/enzimología , Estreñimiento/genética , Estreñimiento/fisiopatología , Tránsito Gastrointestinal/genética , Adolescente , Adulto , Niño , Estreñimiento/clasificación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB1/fisiología , Adulto Joven
15.
Am J Physiol Gastrointest Liver Physiol ; 304(10): G897-907, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23494121

RESUMEN

The intestinal transporter PEPT1 mediates the absorption of di- and tripeptides originating from breakdown of dietary proteins. Whereas mice lacking PEPT1 did not display any obvious changes in phenotype on a high-carbohydrate control diet (HCD), Pept1(-/-) mice fed a high-fat diet (HFD) showed a markedly reduced weight gain and reduced body fat stores. They were additionally protected from hyperglycemia and hyperinsulinemia. Energy balance studies revealed that Pept1(-/-) mice on HFD have a reduced caloric intake, no changes in energy expenditure, but increased energy content in feces. Cecal biomass in Pept1(-/-) mice was as well increased twofold on both diets, suggesting a limited capacity in digesting and/or absorbing the dietary constituents in the small intestine. GC-MS-based metabolite profiling of cecal contents revealed high levels and a broad spectrum of sugars in PEPT1-deficient mice on HCD, whereas animals fed HFD were characterized by high levels of free fatty acids and absence of sugars. In search of the origin of the impaired digestion/absorption, we observed that Pept1(-/-) mice lack the adaptation of the upper small intestinal mucosa to the trophic effects of the diet. Whereas wild-type mice on HFD adapt to diet with increased villus length and surface area, Pept1(-/-) mice failed to show this response. In search for the origin of this, we recorded markedly reduced systemic IL-6 levels in all Pept1(-/-) mice, suggesting that IL-6 could contribute to the lack of adaptation of the mucosal architecture to the diets.


Asunto(s)
Digestión/genética , Ingestión de Energía/genética , Síndromes de Malabsorción/genética , Obesidad/genética , Simportadores/fisiología , Animales , Composición Corporal/genética , Temperatura Corporal/fisiología , Peso Corporal/genética , Peso Corporal/fisiología , Dieta , Ingestión de Líquidos/genética , Ingestión de Alimentos/genética , Ingestión de Alimentos/psicología , Heces/química , Cromatografía de Gases y Espectrometría de Masas , Tránsito Gastrointestinal/genética , Tránsito Gastrointestinal/fisiología , Metabolismo de los Lípidos/genética , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/genética , Tamaño de los Órganos/genética , Tamaño de los Órganos/fisiología , Transportador de Péptidos 1 , ARN/biosíntesis , ARN/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Recto/fisiología , Simportadores/genética
16.
Gastroenterology ; 144(1): 145-54, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23041323

RESUMEN

BACKGROUND & AIMS: Abnormal delivery of bile acids (BAs) to the colon as a result of disease or therapy causes constipation or diarrhea by unknown mechanisms. The G protein-coupled BA receptor TGR5 (or GPBAR1) is expressed by enteric neurons and endocrine cells, which regulate motility and secretion. METHODS: We analyzed gastrointestinal and colon transit, as well as defecation frequency and water content, in wild-type, knockout, and transgenic mice (trg5-wt, tgr5-ko, and tgr5-tg, respectively). We analyzed colon tissues for contractility, peristalsis, and transmitter release. RESULTS: Deoxycholic acid inhibited contractility of colonic longitudinal muscle from tgr5-wt but not tgr5-ko mice. Application of deoxycholic acid, lithocholic acid, or oleanolic acid (a selective agonist of TGR5) to the mucosa of tgr5-wt mice caused oral contraction and caudal relaxation, indicating peristalsis. BAs stimulated release of the peristaltic transmitters 5-hydroxytryptamine and calcitonin gene-related peptide; antagonists of these transmitters suppressed BA-induced peristalsis, consistent with localization of TGR5 to enterochromaffin cells and intrinsic primary afferent neurons. tgr5-ko mice did not undergo peristalsis or transmitter release in response to BAs. Mechanically induced peristalsis and transmitter release were not affected by deletion of tgr5. Whole-gut transit was 1.4-fold slower in tgr5-ko than tgr5-wt or tgr5-tg mice, whereas colonic transit was 2.2-fold faster in tgr5-tg mice. Defecation frequency was reduced 2.6-fold in tgr5-ko and increased 1.4-fold in tgr5-tg mice compared with tgr5-wt mice. Water content in stool was lower (37%) in tgr5-ko than tgr5-tg (58%) or tgr5-wt mice (62%). CONCLUSIONS: The receptor TGR5 mediates the effects of BAs on colonic motility, and deficiency of TGR5 causes constipation in mice. These findings might mediate the long-known laxative properties of BAs, and TGR5 might be a therapeutic target for digestive diseases.


Asunto(s)
Colon/efectos de los fármacos , Colon/fisiología , Defecación/efectos de los fármacos , Tránsito Gastrointestinal/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Colon/metabolismo , Defecación/genética , Ácido Desoxicólico/farmacología , Células Enterocromafines/efectos de los fármacos , Células Enterocromafines/metabolismo , Heces/química , Tránsito Gastrointestinal/genética , Mucosa Intestinal/efectos de los fármacos , Ácido Litocólico/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Contracción Muscular/efectos de los fármacos , Relajación Muscular/efectos de los fármacos , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/metabolismo , Ácido Oleanólico/farmacología , Peristaltismo , Receptores Acoplados a Proteínas G/genética , Serotonina/metabolismo , Agua/análisis
17.
J Pediatr Gastroenterol Nutr ; 55(6): 689-94, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22699839

RESUMEN

OBJECTIVES: Intestinal dysmotility is one of the effects of cystic fibrosis (CF), but when and how this develops is not well understood. The goal of the present study was to use the Cftr knockout mouse to determine when in development circular smooth muscle of the small intestine becomes dysfunctional. METHODS: Wild-type (WT) and CF mice were used at postnatal day 5 (P5) through adult. Pieces of small intestine were used to measure contractile activity of the circular muscle. Bacterial overgrowth was measured by quantitative polymerase chain reaction (PCR) of the bacterial 16S gene. Intestinal gene expression was determined by quantitative reverse transcription polymerase chain reaction (RT-PCR). Prostaglandin E2 (PGE2) and its metabolites were measured by enzyme immunoassay. RESULTS: CF circular muscle response to cholinergic stimulation was similar to WT at P5, became somewhat impaired at P7, and was severely impaired by P14. In the CF intestine, bacterial overgrowth occurred by P4 and was maintained into adulthood. Eicosanoid metabolic gene expression in the CF intestine did not differ from WT shortly after birth. The phospholipase A2 genes, Pla2g4c and Pla2g5 exhibited increased expression in CF mice at P24. Prostaglandin degradative genes, Hpgd and Ptgr1, showed lower expression in CF as compared with WT at P16 and P24, respectively. PGE2 levels were significantly greater in CF mice at most ages from P7 through adulthood. CONCLUSIONS: The results clearly demonstrate that lack of CFTR itself does not cause smooth muscle dysfunction, because the circular muscle from P5 CF mice had normal activity and dysfunction developed between P7 and P14.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/fisiopatología , Tránsito Gastrointestinal/fisiología , Enfermedades Intestinales/fisiopatología , Intestino Delgado/fisiopatología , Contracción Muscular , Músculo Liso/fisiopatología , Animales , Bacterias/crecimiento & desarrollo , Colinérgicos , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Dinoprostona/metabolismo , Tránsito Gastrointestinal/genética , Expresión Génica , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/microbiología , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CFTR , Ratones Noqueados , Contracción Muscular/genética , Fosfolipasas A2/genética , Fosfolipasas A2/metabolismo
18.
Neurogastroenterol Motil ; 24(4): 358-e169, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22288893

RESUMEN

BACKGROUND: Genetic variation in endocannabinoid metabolism is associated with colonic transit in irritable bowel syndrome (IBS) with diarrhea (IBS-D). The nonselective cannabinoid (CB) receptor agonist, dronabinol (DRO), reduced fasting colonic motility in nonconstipated IBS. FAAH and CNR1 variants influenced DRO's effects on colonic motility. Our aims were: (i) to compare dose-related effects of DRO to placebo (PLA) on gut transit in IBS-D, and (ii) to examine influence of genetic variations in CB mechanisms on DRO's transit effects. METHODS: Thirty-six IBS-D volunteers were randomized (double-blind, concealed allocation) to twice per day PLA (n = 13), DRO 2.5 mg (n = 10), or DRO 5 mg (n = 13) for 2 days. We assessed gastric, small bowel, and colonic transit by validated radioscintigraphy and genotyped the single nucleotide polymorphisms CNR1 rs806378 and FAAH rs324420. Data analysis utilized a dominant genetic model. KEY RESULTS: Overall treatment effects of DRO on gastric, small bowel, or colonic transit were not detected. CNR1 rs806378 CT/TT was associated with a modest delay in colonic transit at 24 h compared with CC (P = 0.13 for differential treatment effects on postminus pretreatment changes in colonic transit by genotype). No significant interaction of treatment with FAAH rs324420 was detected. CONCLUSIONS & INFERENCES: Overall, DRO 2.5 or 5 mg twice per day for 2 days had no effect on gut transit in IBS-D. There appears to be a treatment-by-genotype effect, whereby DRO preferentially delays colonic transit in those with the CNR1 rs806378 CT/TT genotypes. Further study of CB pharmacogenetics may help identify a subset of IBS-D patients most likely to benefit from CB agonist therapy.


Asunto(s)
Analgésicos no Narcóticos/administración & dosificación , Diarrea/tratamiento farmacológico , Dronabinol/administración & dosificación , Tránsito Gastrointestinal/efectos de los fármacos , Síndrome del Colon Irritable/tratamiento farmacológico , Síndrome del Colon Irritable/genética , Amidohidrolasas/genética , Analgésicos no Narcóticos/farmacocinética , Diarrea/etiología , Diarrea/genética , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Dronabinol/farmacocinética , Tránsito Gastrointestinal/genética , Genómica , Genotipo , Humanos , Síndrome del Colon Irritable/complicaciones , Polimorfismo de Nucleótido Simple , Receptor Cannabinoide CB1/genética
19.
Dig Dis Sci ; 57(5): 1222-6, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22271411

RESUMEN

BACKGROUND: Protein products of klothoß (KLB) and fibroblast growth factor receptor 4 (FGFR4) impact fibroblast growth factor 19-mediated feedback inhibition of hepatic bile acid (BA) synthesis. Variants of KLB and FGFR4 influence colonic transit (CT) in diarrhea-predominant irritable bowel syndrome (IBS-D). AIM: The purpose of this study was to test the hypothesis that colesevelam's slowing effects on CT in IBS-D patients is influenced by genetic variants in KLB and FGFR4. METHODS: We examined pharmacogenetic effects of KLB and FGFR4 coding variants (SNPs) on scintigraphic CT response to the BA sequestrant, colesevelam 1.875 g b.i.d. versus placebo (PLA) for 14 days in 24 female IBS-D patients. RESULTS: FGFR4 rs351855 and KLB rs497501 were associated with differential colesevelam effects on ascending colon (AC) half-emptying time (t(1/2), P = 0.046 and P = 0.085 respectively) and on overall CT at 24 h (geometric center, GC24: P = 0.073 and P = 0.042, respectively), with slower transit for rs351855 GA/AA (but not for GG) and rs497501 CA/AA (but not CC) genotypes. CONCLUSION: FGFR4 rs351855 and KLB rs4975017 SNPs may identify a subset of IBS-D patients with beneficial response to colesevelam.


Asunto(s)
Alilamina/análogos & derivados , Ácidos y Sales Biliares , Diarrea , Tránsito Gastrointestinal/genética , Síndrome del Colon Irritable , Proteínas de la Membrana/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Adulto , Alilamina/administración & dosificación , Alilamina/farmacocinética , Anticolesterolemiantes/administración & dosificación , Anticolesterolemiantes/farmacocinética , Ácidos y Sales Biliares/biosíntesis , Ácidos y Sales Biliares/genética , Biotransformación , Clorhidrato de Colesevelam , Colon/metabolismo , Colon/fisiopatología , Diarrea/tratamiento farmacológico , Diarrea/etiología , Diarrea/genética , Diarrea/fisiopatología , Retroalimentación Fisiológica/efectos de los fármacos , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Síndrome del Colon Irritable/complicaciones , Síndrome del Colon Irritable/tratamiento farmacológico , Síndrome del Colon Irritable/genética , Síndrome del Colon Irritable/fisiopatología , Proteínas Klotho , Farmacogenética , Polimorfismo de Nucleótido Simple
20.
Neurogastroenterol Motil ; 23(11): 995-9, e458, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21883702

RESUMEN

BACKGROUND: The membrane bound bile acid (BA) receptor, TGR5, is located on myenteric, cholinergic and nitrergic neurons in colon and proximal small intestine. Our aim was to assess the association of genetic variation in TGR5 and small bowel transit (SBT) and colonic transit. METHODS: In 230 healthy controls and 414 patients with lower functional GI disorders [FGID: irritable bowel syndrome (IBS)-alternators (Alt) 84, IBS-constipation (IBS-C) 157, IBS-diarrhea (IBS-D) 173], we tested the association between TGR5 SNP rs11554825 (minor allele frequency 41%) with symptom phenotype (total cohort) and intermediate phenotype (SBT or colonic transit by radioscintigraphy) which was available in 213 people in this cohort. The association with symptom phenotype was assessed using logistic regression, while the association with colonic filling at 6 h (CF6), and colonic transit [geometric center (GC) at 24 h] was assessed using ancova, in each instance assuming a dominant genetic model. KEY RESULTS: There was no significant association with symptom phenotype. We observed a potential association of SNP rs11554825 with overall transit: CF6 (P = 0.061) and GC24 (P = 0.083). The association of the SNP with CF6 in the IBS-D subgroup (P = 0.017) indicated the TC/CC subgroup had an average 50% faster SBT compared with the TT subgroup. In IBS-D patients, GC24 was not significantly associated with rs11554825 (TC/CC vs TT). CONCLUSIONS & INFERENCES: Variation in TGR5 may contribute to altered SBT and colonic transit in lower FGID. Further studies are required to characterize the potential role of BA receptor, TGR5, in the mechanism and treatment of bowel dysfunction in lower FGID.


Asunto(s)
Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/fisiopatología , Tránsito Gastrointestinal/genética , Tránsito Gastrointestinal/fisiología , Variación Genética , Receptores Acoplados a Proteínas G/genética , Adulto , Ácidos y Sales Biliares/metabolismo , Estreñimiento/genética , Estreñimiento/fisiopatología , Diarrea/genética , Diarrea/fisiopatología , Frecuencia de los Genes , Humanos , Síndrome del Colon Irritable/genética , Síndrome del Colon Irritable/fisiopatología , Persona de Mediana Edad , Receptores Acoplados a Proteínas G/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...