Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gastroenterology ; 162(2): 535-547.e13, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34688712

RESUMEN

BACKGROUND AND AIMS: The gastrointestinal (GI) tract extracts nutrients from ingested meals while protecting the organism from infectious agents frequently present in meals. Consequently, most animals conduct the entire digestive process within the GI tract while keeping the luminal contents entirely outside the body, separated by the tightly sealed GI epithelium. Therefore, like the skin and oral cavity, the GI tract must sense the chemical and physical properties of the its external interface to optimize its function. Specialized sensory enteroendocrine cells (EECs) in GI epithelium interact intimately with luminal contents. A subpopulation of EECs express the mechanically gated ion channel Piezo2 and are developmentally and functionally like the skin's touch sensor- the Merkel cell. We hypothesized that Piezo2+ EECs endow the gut with intrinsic tactile sensitivity. METHODS: We generated transgenic mouse models with optogenetic activators in EECs and Piezo2 conditional knockouts. We used a range of reference standard and novel techniques from single cells to living animals, including single-cell RNA sequencing and opto-electrophysiology, opto-organ baths with luminal shear forces, and in vivo studies that assayed GI transit while manipulating the physical properties of luminal contents. RESULTS: Piezo2+ EECs have transcriptomic features of synaptically connected, mechanosensory epithelial cells. EEC activation by optogenetics and forces led to Piezo2-dependent alterations in colonic propagating contractions driven by intrinsic circuitry, with Piezo2+ EECs detecting the small luminal forces and physical properties of the luminal contents to regulate transit times in the small and large bowel. CONCLUSIONS: The GI tract has intrinsic tactile sensitivity that depends on Piezo2+ EECs and allows it to detect luminal forces and physical properties of luminal contents to modulate physiology.


Asunto(s)
Células Enteroendocrinas/metabolismo , Mucosa Intestinal/metabolismo , Canales Iónicos/genética , Tacto/fisiología , Animales , Células Enteroendocrinas/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Técnicas de Inactivación de Genes , Mucosa Intestinal/citología , Mucosa Intestinal/fisiología , Canales Iónicos/metabolismo , Mecanorreceptores , Ratones , Ratones Transgénicos , Optogenética , Peristaltismo/fisiología
3.
Proc Natl Acad Sci U S A ; 115(32): E7632-E7641, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30037999

RESUMEN

Enterochromaffin (EC) cells constitute the largest population of intestinal epithelial enteroendocrine (EE) cells. EC cells are proposed to be specialized mechanosensory cells that release serotonin in response to epithelial forces, and thereby regulate intestinal fluid secretion. However, it is unknown whether EE and EC cells are directly mechanosensitive, and if so, what the molecular mechanism of their mechanosensitivity is. Consequently, the role of EE and EC cells in gastrointestinal mechanobiology is unclear. Piezo2 mechanosensitive ion channels are important for some specialized epithelial mechanosensors, and they are expressed in mouse and human EC cells. Here, we use EC and EE cell lineage tracing in multiple mouse models to show that Piezo2 is expressed in a subset of murine EE and EC cells, and it is distributed near serotonin vesicles by superresolution microscopy. Mechanical stimulation of a subset of isolated EE cells leads to a rapid inward ionic current, which is diminished by Piezo2 knockdown and channel inhibitors. In these mechanosensitive EE cells force leads to Piezo2-dependent intracellular Ca2+ increase in isolated cells as well as in EE cells within intestinal organoids, and Piezo2-dependent mechanosensitive serotonin release in EC cells. Conditional knockout of intestinal epithelial Piezo2 results in a significant decrease in mechanically stimulated epithelial secretion. This study shows that a subset of primary EE and EC cells is mechanosensitive, uncovers Piezo2 as their primary mechanotransducer, defines the molecular mechanism of their mechanotransduction and mechanosensitive serotonin release, and establishes the role of epithelial Piezo2 mechanosensitive ion channels in regulation of intestinal physiology.


Asunto(s)
Células Enterocromafines/fisiología , Canales Iónicos/metabolismo , Yeyuno/fisiología , Mecanotransducción Celular/fisiología , Serotonina/metabolismo , Animales , Células Cultivadas , Canales Iónicos/genética , Yeyuno/citología , Ratones , Ratones Transgénicos , Organoides/fisiología , Cultivo Primario de Células , ARN Interferente Pequeño/metabolismo , Análisis de la Célula Individual
4.
Gastroenterology ; 154(8): 2122-2136.e12, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29501441

RESUMEN

BACKGROUND & AIMS: Muscularis propria macrophages lie close to cells that regulate gastrointestinal motor function, including interstitial cells of Cajal (ICC) and myenteric neurons. In animal models of diabetic gastroparesis, development of delayed gastric emptying has been associated with loss of macrophages that express cytoprotective markers and reduced networks of ICC. Mice with long-term diabetes and normal gastric emptying have macrophages that express anti-inflammatory markers and have normal gastric ICC. Mice homozygous for the osteopetrosis spontaneous mutation in the colony-stimulating factor 1 gene (Csf1op/op) do not have macrophages; when they are given streptozotocin to induce diabetes, they do not develop delayed gastric emptying. We investigated whether population of the gastric muscularis propria of diabetic Csf1op/op mice with macrophages is necessary to change gastric emptying, ICC, and myenteric neurons and investigated the macrophage-derived factors that determine whether diabetic mice do or do not develop delayed gastric emptying. METHODS: Wild-type and Csf1op/op mice were given streptozotocin to induce diabetes. Some Csf1op/op mice were given daily intraperitoneal injections of CSF1 for 7 weeks; gastric tissues were collected and cellular distributions were analyzed by immunohistochemistry. CD45+, CD11b+, F4/80+ macrophages were dissociated from gastric muscularis propria, isolated by flow cytometry and analyzed by quantitative real-time polymerase chain reaction. Cultured gastric muscularis propria from Csf1op/op mice was exposed to medium that was conditioned by culture with bone marrow-derived macrophages from wild-type mice. RESULTS: Gastric muscularis propria from Csf1op/op mice given CSF1 contained macrophages; 11 of 15 diabetic mice given CSF1 developed delayed gastric emptying and had damaged ICC. In non-diabetic Csf1op/op mice, administration of CSF1 reduced numbers of gastric myenteric neurons but did not affect the proportion of nitrergic neurons or ICC. In diabetic Csf1op/op mice given CSF1 that developed delayed gastric emptying, the proportion of nitrergic neurons was the same as in non-diabetic wild-type controls. Medium conditioned by macrophages previously exposed to oxidative injury caused damage to ICC in cultured gastric muscularis propria from Csf1op/op mice; neutralizing antibodies against IL6R or TNF prevented this damage to ICC. CD45+, CD11b+, and F4/80+ macrophages isolated from diabetic wild-type mice with delayed gastric emptying expressed higher levels of messenger RNAs encoding inflammatory markers (IL6 and inducible nitric oxide synthase) and lower levels of messenger RNAs encoding markers of anti-inflammatory cells (heme oxygenase 1, arginase 1, and FIZZ1) than macrophages isolated from diabetic mice with normal gastric emptying. CONCLUSIONS: In studies of Csf1op/op and wild-type mice with diabetes, we found delayed gastric emptying to be associated with increased production of inflammatory factors, and reduced production of anti-inflammatory factors, by macrophages, leading to loss of ICC.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Vaciamiento Gástrico/fisiología , Gastroparesia/fisiopatología , Macrófagos/fisiología , Estómago/fisiopatología , Animales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Gastroparesia/etiología , Humanos , Inmunohistoquímica , Células Intersticiales de Cajal/fisiología , Factor Estimulante de Colonias de Macrófagos/genética , Ratones , Músculo Liso/citología , Músculo Liso/patología , Músculo Liso/fisiopatología , Mutación , Estómago/citología , Estómago/patología , Estreptozocina/toxicidad
5.
Exp Physiol ; 103(1): 40-57, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28971566

RESUMEN

NEW FINDINGS: What is the central question of this study? The aim was to investigate the roles of extracellular chloride in electrical slow waves and resting membrane potential of mouse jejunal smooth muscle by replacing chloride with the impermeant anions gluconate and isethionate. What is the main finding and its importance? The main finding was that in smooth muscle cells, the resting Cl- conductance is low, whereas transmembrane Cl- movement in interstitial cells of Cajal (ICCs) is a major contributor to the shape of electrical slow waves. Furthermore, the data confirm that ICCs set the smooth muscle membrane potential and that altering Cl- homeostasis in ICCs can alter the smooth muscle membrane potential. Intracellular Cl- homeostasis is regulated by anion-permeable channels and transporters and contributes to excitability of many cell types, including smooth muscle and interstitial cells of Cajal (ICCs). Our aims were to investigate the effects on electrical activity in mouse jejunal muscle strips of replacing extracellular Cl- (Cl-o ) with the impermeant anions gluconate and isethionate. On reducing Cl-o , effects were observed on electrical slow waves, with small effects on smooth muscle membrane voltage (Em ). Restoration of Cl- hyperpolarized smooth muscle Em proportional to the change in Cl-o concentration. Replacement of 90% of Cl-o with gluconate reversibly abolished slow waves in five of nine preparations. Slow waves were maintained in isethionate. Gluconate and isethionate substitution had similar concentration-dependent effects on peak amplitude, frequency, width at half peak amplitude, rise time and decay time of residual slow waves. Gluconate reduced free ionized Ca2+ in Krebs solutions to 0.13 mm. In Krebs solutions containing normal Cl- and 0.13 mm free Ca2+ , slow wave frequency was lower, width at half peak amplitude was smaller, and decay time was faster. The transient hyperpolarization following restoration of Cl-o was not observed in W/Wv mice, which lack pacemaker ICCs in the small intestine. We conclude that in smooth muscle cells, the resting Cl- conductance is low, whereas transmembrane Cl- movement in ICCs plays a major role in generation or propagation of slow waves. Furthermore, these data support a role for ICCs in setting smooth muscle Em and that altering Cl- homeostasis in ICCs can alter smooth muscle Em .


Asunto(s)
Cloruros/fisiología , Líquido Extracelular/fisiología , Células Intersticiales de Cajal/fisiología , Yeyuno/fisiología , Potenciales de la Membrana/fisiología , Músculo Liso/fisiología , Animales , Cloruros/farmacología , Líquido Extracelular/efectos de los fármacos , Femenino , Células Intersticiales de Cajal/efectos de los fármacos , Yeyuno/citología , Yeyuno/efectos de los fármacos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Músculo Liso/efectos de los fármacos , Técnicas de Cultivo de Órganos
6.
Sci Rep ; 7(1): 15650, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-29142310

RESUMEN

In the gastrointestinal (GI) epithelium, enterochromaffin (EC) cells are enteroendocrine cells responsible for producing >90% of the body's serotonin (5-hydroxytryptamine, 5-HT). However, the molecular mechanisms of EC cell function are poorly understood. Here, we found that EC cells in mouse primary cultures fired spontaneous bursts of action potentials. We examined the repertoire of voltage-gated sodium channels (NaV) in fluorescence-sorted mouse EC cells and found that Scn3a was highly expressed. Scn3a-encoded NaV1.3 was specifically and densely expressed at the basal side of both human and mouse EC cells. Using electrophysiology, we found that EC cells expressed robust NaV1.3 currents, as determined by their biophysical and pharmacologic properties. NaV1.3 was not only critical for generating action potentials in EC cells, but it was also important for regulating 5-HT release by these cells. Therefore, EC cells use Scn3a-encoded voltage-gated sodium channel NaV1.3 for electrical excitability and 5-HT release. NaV1.3-dependent electrical excitability and its contribution to 5-HT release is a novel mechanism of EC cell function.


Asunto(s)
Células Enterocromafines/metabolismo , Canal de Sodio Activado por Voltaje NAV1.3/genética , Serotonina/metabolismo , Canales de Sodio Activados por Voltaje/genética , Potenciales de Acción , Animales , Fenómenos Biofísicos , Electrofisiología , Células Enterocromafines/efectos de los fármacos , Células Enteroendocrinas/efectos de los fármacos , Células Enteroendocrinas/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Ratones , Canal de Sodio Activado por Voltaje NAV1.3/metabolismo , Cultivo Primario de Células , Serotonina/biosíntesis , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio Activados por Voltaje/metabolismo
7.
Cell Mol Gastroenterol Hepatol ; 2(4): 454-467, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27795979

RESUMEN

BACKGROUND & AIMS: Gastroparesis is a complication of diabetes characterized by delayed emptying of stomach contents and accompanied by early satiety, nausea, vomiting, and pain. No safe and reliable treatments are available. Interleukin 10 (IL10) activates the M2 cytoprotective phenotype of macrophages and induces expression of heme oxygenase 1 (HO1) protein. We investigated whether IL10 administration could improve gastric emptying and reverse the associated cellular and electrical abnormalities in diabetic mice. METHODS: Nonobese diabetic mice with delayed gastric emptying were given either IL10 (0.1-1 µg, twice/day) or vehicle (controls). Stomach tissues were isolated, and sharp microelectrode recordings were made of the electrical activity in the gastric muscle layers. Changes to interstitial cells of Cajal (ICC), reduced nicotinamide adenine dinucleotide phosphate diaphorase, and levels and distribution of HO1 protein were determined by histochemical and imaging analyses of the same tissues. RESULTS: Gastric emptying remained delayed in vehicle-treated diabetic mice but returned to normal in mice given IL10 (n = 10 mice; P < .05). In mice given IL10, normalization of gastric emptying was associated with a membrane potential difference between the proximal and distal stomach, and lower irregularity and higher frequency of slow-wave activity, particularly in the distal stomach. Levels of HO1 protein were higher in stomach tissues from mice given IL10, and ICC networks were more organized, better connected, and more evenly distributed compared with controls. CONCLUSIONS: IL10 increases gastric emptying in diabetic mice and has therapeutic potential for patients with diabetic gastroparesis. This response is associated with up-regulation of HO1 and repair of connectivity of ICC networks.

8.
Genome Med ; 8(1): 103, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27716401

RESUMEN

BACKGROUND: Nutritional interventions often fail to prevent growth failure in childhood and adolescent malnutrition and the mechanisms remain unclear. Recent studies revealed altered microbiota in malnourished children and anorexia nervosa. To facilitate mechanistic studies under physiologically relevant conditions, we established a mouse model of growth failure following chronic dietary restriction and examined microbiota in relation to age, diet, body weight, and anabolic treatment. METHODS: Four-week-old female BALB/c mice (n = 12/group) were fed ad libitum (AL) or offered limited food to abolish weight gain (LF). A subset of restricted mice was treated with an insulin-like growth factor 1 (IGF1) analog. Food access was restored in a subset of untreated LF (LF-RF) and IGF1-treated LF mice (TLF-RF) on day 97. Gut microbiota were determined on days 69, 96-99 and 120 by next generation sequencing of the V3-5 region of the 16S rRNA gene. Microbiota-host factor associations were analyzed by distance-based PERMANOVA and quantified by the coefficient of determination R2 for age, diet, and normalized body weight change (Δbwt). Microbial taxa on day 120 were compared following fitting with an overdispersed Poisson regression model. The machine learning algorithm Random Forests was used to predict age based on the microbiota. RESULTS: On day 120, Δbwt in AL, LF, LF-RF, and TLF-RF mice was 52 ± 3, -6 ± 1*, 40 ± 3*, and 46 ± 2 % (*, P < 0.05 versus AL). Age and diet, but not Δbwt, were associated with gut microbiota composition. Age explained a larger proportion of the microbiota variability than diet or Δbwt. Random Forests predicted chronological age based on the microbiota and indicated microbiota immaturity in the LF mice before, but not after, refeeding. However, on day 120, the microbiota community structure of LF-RF mice was significantly different from that of both AL and LF mice. IGF1 mitigated the difference from the AL group. Refed groups had a higher abundance of Bacteroidetes and Proteobacteria and a lower abundance of Firmicutes than AL mice. CONCLUSIONS: Persistent growth failure can be induced by 97-day dietary restriction in young female mice and is associated with microbiota changes seen in lean mice and individuals and anorexia nervosa. IGF1 facilitates recovery of body weights and microbiota.


Asunto(s)
Anorexia Nerviosa/microbiología , Restricción Calórica , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Pérdida de Peso/genética , Animales , Anorexia Nerviosa/genética , Anorexia Nerviosa/fisiopatología , Conducta Alimentaria , Femenino , Ratones , Ratones Endogámicos BALB C , Filogenia , Reacción en Cadena de la Polimerasa , ARN Ribosómico 16S/genética , Destete
9.
Cell Mol Gastroenterol Hepatol ; 2(1): 40-47, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26771000

RESUMEN

BACKGROUND & AIMS: Diabetic gastroparesis is associated with changes in interstitial cells of Cajal (ICC), neurons and smooth muscle cells in both animal models and humans. Macrophages appear to be critical to the development of cellular damage that leads to delayed gastric emptying but the mechanisms involved are not well understood. Csf1op/op (Op/Op) mice lack biologically active Csf1, resulting in the absence of Csf1-dependent tissue macrophages. The aim of this study was to use Csf1op/op mice to determine the role of macrophages in the development of delayed gastric emptying. METHODS: Animals were injected with streptozotocin to make them diabetic. Gastric emptying was determined weekly. Immunohistochemistry was used to identify macrophages and ICC networks in the gastric muscular layers. Oxidative stress was measured by serum malondialdehyde (MDA) levels. Quantitative, reverse transcription PCR was used to measure levels of mRNA. RESULTS: Csf1op/op mice had normal ICC. With onset of diabetes both Csf1op/op and wild type Csf1+/+ mice developed increased levels of oxidative stress (75.8 ± 9.1 and 41.2±13.6 nmol/mL MDA respectively). Wild type Csf1+/+ mice developed delayed gastric emptying after onset of diabetes (4/13) whereas no diabetic Csf1op/op mouse developed delayed gastric emptying (0/15, P=0.035). ICC were disrupted in diabetic wild type Csf1+/+ mice with delayed gastric emptying but remained normal in diabetic Csf1op/op mice. CONCLUSIONS: Cellular injury and development of delayed gastric emptying in diabetes requires the presence of muscle layer macrophages. Targeting macrophages may be an effective therapeutic option to prevent cellular damage and development of delayed gastric emptying in diabetes.

10.
FASEB J ; 29(4): 1395-403, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25550456

RESUMEN

Gut microbiota alterations have been described in several diseases with altered gastrointestinal (GI) motility, and awareness is increasing regarding the role of the gut microbiome in modulating GI function. Serotonin [5-hydroxytryptamine (5-HT)] is a key regulator of GI motility and secretion. To determine the relationship among gut microbes, colonic contractility, and host serotonergic gene expression, we evaluated mice that were germ-free (GF) or humanized (HM; ex-GF colonized with human gut microbiota). 5-HT reduced contractile duration in both GF and HM colons. Microbiota from HM and conventionally raised (CR) mice significantly increased colonic mRNAs Tph1 [(tryptophan hydroxylase) 1, rate limiting for mucosal 5-HT synthesis; P < 0.01] and chromogranin A (neuroendocrine secretion; P < 0.01), with no effect on monoamine oxidase A (serotonin catabolism), serotonin receptor 5-HT4, or mouse serotonin transporter. HM and CR mice also had increased colonic Tph1 protein (P < 0.05) and 5-HT concentrations (GF, 17 ± 3 ng/mg; HM, 25 ± 2 ng/mg; and CR, 35 ± 3 ng/mg; P < 0.05). Enterochromaffin (EC) cell numbers (cells producing 5-HT) were unchanged. Short-chain fatty acids (SCFAs) promoted TPH1 transcription in BON cells (human EC cell model). Thus, gut microbiota acting through SCFAs are important determinants of enteric 5-HT production and homeostasis.


Asunto(s)
Colon/metabolismo , Sistema Digestivo/microbiología , Células Enterocromafines/metabolismo , Ácidos Grasos Volátiles/metabolismo , Serotonina/biosíntesis , Animales , Recuento de Células , Línea Celular , Cromogranina A/genética , Colon/citología , Colon/microbiología , Células Enterocromafines/citología , Células Enterocromafines/microbiología , Femenino , Motilidad Gastrointestinal , Vida Libre de Gérmenes , Humanos , Masculino , Ratones , Microbiota , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
11.
J Physiol ; 592(18): 4051-68, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25063822

RESUMEN

Interstitial cells of Cajal (ICC) are pacemaker cells that generate electrical activity to drive contractility in the gastrointestinal tract via ion channels. Ano1 (Tmem16a), a Ca(2+)-activated Cl(-) channel, is an ion channel expressed in ICC. Genetic deletion of Ano1 in mice resulted in loss of slow waves in smooth muscle of small intestine. In this study, we show that Ano1 is required to maintain coordinated Ca(2+) transients between myenteric ICC (ICC-MY) of small intestine. First, we found spontaneous Ca(2+) transients in ICC-MY in both Ano1 WT and knockout (KO) mice. However, Ca(2+) transients within the ICC-MY network in Ano1 KO mice were uncoordinated, while ICC-MY Ca(2+) transients in Ano1 WT mice were rhythmic and coordinated. To confirm the role of Ano1 in the loss of Ca(2+) transient coordination, we used pharmacological inhibitors of Ano1 activity and shRNA-mediated knock down of Ano1 expression in organotypic cultures of Ano1 WT small intestine. Coordinated Ca(2+) transients became uncoordinated using both these approaches, supporting the conclusion that Ano1 is required to maintain coordination/rhythmicity of Ca(2+) transients. We next determined the effect on smooth muscle contractility using spatiotemporal maps of contractile activity in Ano1 KO and WT tissues. Significantly decreased contractility that appeared to be non-rhythmic and uncoordinated was observed in Ano1 KO jejunum. In conclusion, Ano1 has a previously unidentified role in the regulation of coordinated gastrointestinal smooth muscle function through coordination of Ca(2+) transients in ICC-MY.


Asunto(s)
Señalización del Calcio , Canales de Cloruro/metabolismo , Células Intersticiales de Cajal/metabolismo , Yeyuno/metabolismo , Contracción Muscular , Animales , Anoctamina-1 , Calcio/metabolismo , Canales de Cloruro/genética , Células Intersticiales de Cajal/fisiología , Yeyuno/fisiología , Ratones
12.
Gastroenterology ; 147(2): 303-13, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24798417

RESUMEN

Carbon monoxide (CO) and hydrogen sulfide (H2S) used to be thought of simply as lethal and (for H2S) smelly gaseous molecules; now they are known to have important signaling functions in the gastrointestinal tract. CO and H2S, which are produced in the gastrointestinal tract by different enzymes, regulate smooth muscle membrane potential and tone, transmit signals from enteric nerves, and can regulate the immune system. The pathways that produce nitric oxide, H2S, and CO interact; each can inhibit and potentiate the level and activity of the other. However, there are significant differences between these molecules, such as in half-lives; CO is more stable and therefore able to have effects distal to the site of production, whereas nitric oxide and H2S are short lived and act only close to sites of production. We review their signaling functions in the luminal gastrointestinal tract and discuss how their pathways interact. We also describe other physiological functions of CO and H2S and how they might be used as therapeutic agents.


Asunto(s)
Monóxido de Carbono/metabolismo , Tracto Gastrointestinal/metabolismo , Sulfuro de Hidrógeno/metabolismo , Músculo Liso/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal , Animales , Monóxido de Carbono/uso terapéutico , Sistema Nervioso Entérico/metabolismo , Fármacos Gastrointestinales/uso terapéutico , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/inervación , Humanos , Sulfuro de Hidrógeno/uso terapéutico , Músculo Liso/efectos de los fármacos , Músculo Liso/inmunología , Músculo Liso/inervación , Transducción de Señal/efectos de los fármacos
13.
J Neurosci ; 33(31): 12638-46, 2013 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-23904600

RESUMEN

Hydrogen sulfide (H2S) plays important roles in the enteric system in the wall of the gastrointestinal tract. There have been no studies on whether H2S is endogenously generated in peripheral sympathetic ganglia and, if so, its effect on synaptic transmission. In this study, we examined the effect of H2S on cholinergic excitatory fast synaptic transmission in the mouse superior mesenteric ganglion (SMG). Our study revealed that NaHS and endogenously generated H2S selectively potentiated cholinergic fast EPSPs (F-EPSPs) evoked by splanchnic nerve stimulation but not F-EPSPs evoked by colonic nerve stimulation. The H2S-producing enzyme cystathionine-γ-lyase (CSE) was expressed in both neurons and glial cells. The CSE blocker PAG (dl-propargylglycine) significantly reduced the amplitude of F-EPSPs evoked by splanchnic nerve stimulation but not F-EPSPs evoked by colonic nerve stimulation. Inhibiting the breakdown of endogenously generated H2S with stigmatellin potentiated the amplitude of F-EPSPs evoked by splanchnic nerve stimulation but not F-EPSPs evoked by colonic nerve stimulation. Splanchnic F-EPSPs but not colonic F-EPSPs were reduced in CSE knock-out (KO) mice. Functional studies showed that NaHS enhanced the inhibitory effect of splanchnic nerve stimulation on colonic motility. Colonic motility in CSE-KO mice was significantly higher than colonic motility in wild-type mice. We conclude that endogenously generated H2S acted selectively on presynaptic terminals of splanchnic nerves to modulate fast cholinergic synaptic input and that this effect of H2S modulates CNS control of gastrointestinal motility. Our results show for the first time that the facilitatory effect of endogenous H2S in the mouse SMG is pathway specific.


Asunto(s)
Acetilcolina/farmacología , Fibras Autónomas Preganglionares/fisiología , Agonistas Colinérgicos/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ganglios Simpáticos/efectos de los fármacos , Sulfuro de Hidrógeno/farmacología , Animales , Colon Descendente/inervación , Cistationina gamma-Liasa/deficiencia , Interacciones Farmacológicas , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Potenciales Postsinápticos Excitadores/genética , Antagonistas de Receptores de GABA-A/farmacología , Ganglios Simpáticos/fisiología , Motilidad Gastrointestinal/efectos de los fármacos , Motilidad Gastrointestinal/genética , Cobayas , Sulfuro de Hidrógeno/metabolismo , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nervios Esplácnicos/fisiología , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
14.
J Vis Exp ; (73): e50301, 2013 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-23542813

RESUMEN

Gastric emptying studies in mice have been limited by the inability to follow gastric emptying changes in the same animal since the most commonly used techniques require killing of the animals and postmortem recovery of the meal(1,2). This approach prevents longitudinal studies to determine changes in gastric emptying with age and progression of disease. The commonly used [(13)C]-octanoic acid breath test for humans(3) has been modified for use in mice(4-6) and rats(7) and we previously showed that this test is reliable and responsive to changes in gastric emptying in response to drugs and during diabetic disease progression(8). In this video presentation the principle and practical implementation of this modified test is explained. As in the previous study, NOD LtJ mice are used, a model of type 1 diabetes(9). A proportion of these mice develop the symptoms of gastroparesis, a complication of diabetes characterized by delayed gastric emptying without mechanical obstruction of the stomach(10). This paper demonstrates how to train the mice for testing, how to prepare the test meal and obtain 4 hr gastric emptying data and how to analyze the obtained data. The carbon isotope analyzer used in the present study is suitable for the automatic sampling of the air samples from up to 12 mice at the same time. This technique allows the longitudinal follow-up of gastric emptying from larger groups of mice with diabetes or other long-standing diseases.


Asunto(s)
Pruebas Respiratorias/instrumentación , Pruebas Respiratorias/métodos , Caprilatos , Radioisótopos de Carbono , Vaciamiento Gástrico/fisiología , Radiofármacos , Animales , Caprilatos/administración & dosificación , Ratones , Radiofármacos/administración & dosificación
15.
Am J Physiol Gastrointest Liver Physiol ; 300(6): G1105-14, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21393430

RESUMEN

Hydrogen sulfide (H(2)S) is produced endogenously by L-cysteine metabolism. H(2)S modulates several ion channels with an unclear mechanism of action. A possible mechanism is through reduction-oxidation reactions attributable to the redox potential of the sulfur moiety. The aims of this study were to determine the effects of the H(2)S donor NaHS on Na(V)1.5, a voltage-dependent sodium channel expressed in the gastrointestinal tract in human jejunum smooth muscle cells and interstitial cells of Cajal, and to elucidate whether H(2)S acts on Na(V)1.5 by redox reactions. Whole cell Na(+) currents were recorded in freshly dissociated human jejunum circular myocytes and Na(V)1.5-transfected human embryonic kidney-293 cells. RT-PCR amplified mRNA for H(2)S enzymes cystathionine ß-synthase and cystathionine γ-lyase from the human jejunum. NaHS increased native Na(+) peak currents and shifted the half-point (V(1/2)) of steady-state activation and inactivation by +21 ± 2 mV and +15 ± 3 mV, respectively. Similar effects were seen on the heterologously expressed Na(V)1.5 α subunit with EC(50)s in the 10(-4) to 10(-3) M range. The reducing agent dithiothreitol (DTT) mimicked in part the effects of NaHS by increasing peak current and positively shifting steady-state activation. DTT together with NaHS had an additive effect on steady-state activation but not on peak current, suggesting that the latter may be altered via reduction. Pretreatment with the Hg(2+)-conjugated oxidizer thimerosal or the alkylating agent N-ethylmaleimide inhibited or decreased NaHS induction of Na(V)1.5 peak current. These studies show that H(2)S activates the gastrointestinal Na(+) channel, and the mechanism of action of H(2)S is partially redox independent.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Yeyuno/metabolismo , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Canales de Sodio/metabolismo , Sodio/metabolismo , Alquilantes/farmacología , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Ditiotreitol/farmacología , Relación Dosis-Respuesta a Droga , Etilmaleimida/farmacología , Células HEK293 , Humanos , Yeyuno/efectos de los fármacos , Potenciales de la Membrana , Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5 , Oxidantes/farmacología , Oxidación-Reducción , Técnicas de Placa-Clamp , Sustancias Reductoras/farmacología , Canales de Sodio/efectos de los fármacos , Canales de Sodio/genética , Sulfuros/farmacología , Timerosal/farmacología , Transfección
16.
J Biol Chem ; 286(15): 13393-403, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21349842

RESUMEN

Diabetes affects many organs including the stomach. Altered number and function of interstitial cells of Cajal (ICC), the gastrointestinal pacemaker cells, underlie a number of gastrointestinal motility disorders, including diabetic gastroparesis. In the muscle layers, ICC selectively express Ano1, thought to underlie classical Ca(2+)-activated Cl(-) currents. Mice homozygous for Ano1 knock-out exhibit abnormal ICC function and motility. Several transcripts for Ano1 are generated by alternative splicing of four exons. Here, we report expression levels of transcripts encoded by alternative splicing of Ano1 gene in gastric muscles of patients with diabetic gastroparesis and nondiabetic control tissues. Expression of mRNA from two alternatively transcribed exons are significantly different between patients and controls. Furthermore, patients with diabetic gastroparesis express mRNA for a previously unknown variant of Ano1. The 5' end of this novel variant lacks exons 1 and 2 and part of exon 3. Expression of this variant in HEK cells produces a decreased density of Ca(2+)-activated Cl(-) currents that exhibit slower kinetics compared with the full-length Ano1. These results identify important changes in expression and splicing of Ano1 in patients with diabetic gastroparesis that alter the electrophysiological properties of the channel. Changes in Ano1 expression in ICC may directly contribute to diabetic gastroparesis.


Asunto(s)
Empalme Alternativo , Complicaciones de la Diabetes/metabolismo , Gastroparesia/metabolismo , Regulación de la Expresión Génica , Células Intersticiales de Cajal/metabolismo , Proteínas de la Membrana/biosíntesis , Músculo Liso/metabolismo , Proteínas de Neoplasias/biosíntesis , Animales , Anoctamina-1 , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Complicaciones de la Diabetes/genética , Complicaciones de la Diabetes/patología , Femenino , Gastroparesia/genética , Gastroparesia/patología , Células HEK293 , Humanos , Células Intersticiales de Cajal/patología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Músculo Liso/patología , Proteínas de Neoplasias/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
17.
Am J Physiol Gastrointest Liver Physiol ; 298(6): G1013-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20378827

RESUMEN

Diabetic gastroparesis is associated with increased oxidative stress attributable to loss of upregulation of heme oxygenase-1 (HO1), with resultant damage to interstitial cells of Cajal and delayed gastric emptying. These changes can be reversed by induction of HO1. HO1 catalyzes the breakdown of heme into iron, biliverdin and, carbon monoxide (CO). The aim of this study was to determine whether inhalation of CO can mimic the protective effects of HO1. Nonobese diabetic (NOD) mice with delayed gastric emptying were treated with CO inhalation. Serum malondialdehyde was measured as a marker of oxidative stress. Gastric emptying of solids was measured using a [(13)C]octanoic acid breath test. Kit expression levels were determined in immunoblots of protein extracted from the external muscle layers of the gastric body and antrum. The effect of CO on oxidative stress and gastric emptying was also determined in the presence of HO activity inhibitor chromium mesoporphyrin. CO inhalation reduced oxidative stress, restored Kit expression and reversed delayed gastric emptying in diabetic NOD mice with delayed gastric emptying. CO inhalation maintained this effect in the presence of the HO activity inhibitor, chromium mesoporphyrin, also resulting in restoration of the delay in gastric emptying. CO inhalation mimics the protective effect of upregulation of HO1 and decreased oxidative stress, increased Kit expression, and restored delay in gastric emptying. This effect of CO was independent of HO activity, suggesting that its effects were downstream of HO1. CO represents a potential therapeutic option for treatment of diabetic gastroparesis.


Asunto(s)
Monóxido de Carbono/farmacología , Complicaciones de la Diabetes/tratamiento farmacológico , Gastroparesia/tratamiento farmacológico , Animales , Femenino , Vaciamiento Gástrico/efectos de los fármacos , Ratones , Ratones Endogámicos NOD , Estrés Oxidativo
18.
Am J Physiol Gastrointest Liver Physiol ; 298(5): G706-13, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20203058

RESUMEN

Antispasmodics are used clinically to treat a variety of gastrointestinal disorders by inhibition of smooth muscle contraction. The main pathway for smooth muscle Ca(2+) entry is through L-type channels; however, there is increasing evidence that T-type Ca(2+) channels also play a role in regulating contractility. Otilonium bromide, an antispasmodic, has previously been shown to inhibit L-type Ca(2+) channels and colonic contractile activity. The objective of this study was to determine whether otilonium bromide also inhibits T-type Ca(2+) channels. Whole cell currents were recorded by patch-clamp technique from HEK293 cells transfected with cDNAs encoding the T-type Ca(2+) channels, Ca(V)3.1 (alpha1G), Ca(V)3.2 (alpha1H), or Ca(V)3.3 (alpha1I) alpha subunits. Extracellular solution was exchanged with otilonium bromide (10(-8) to 10(-5) M). Otilonium bromide reversibly blocked all T-type Ca(2+) channels with a significantly greater affinity for Ca(V)3.3 than Ca(V)3.1 or Ca(V)3.2. Additionally, the drug slowed inactivation in Ca(V)3.1 and Ca(V)3.3. Inhibition of T-type Ca(2+) channels may contribute to inhibition of contractility by otilonium bromide. This may represent a new mechanism of action for antispasmodics and may contribute to the observed increased clinical effectiveness of antispasmodics compared with selective L-type Ca(2+) channel blockers.


Asunto(s)
Canales de Calcio Tipo T/efectos de los fármacos , Parasimpatolíticos/farmacología , Compuestos de Amonio Cuaternario/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Línea Celular , Colon/efectos de los fármacos , Colon/fisiología , Humanos , Cinética , Mibefradil/farmacología , Contracción Muscular/efectos de los fármacos , Técnicas de Placa-Clamp
19.
Gastroenterology ; 138(7): 2399-409, 2409.e1, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20178793

RESUMEN

BACKGROUND & AIMS: Gastroparesis is a well-recognized complication of diabetes. In diabetics, up-regulation of heme oxygenase-1 (HO1) in gastric macrophages protects against oxidative stress-induced damage. Loss of up-regulation of HO1, the subsequent increase in oxidative stress, and loss of Kit delays gastric emptying; this effect is reversed by induction of HO1. Macrophages have pro- and anti-inflammatory activities, depending on their phenotype. We investigated the number and phenotype of gastric macrophages in NOD/ShiLtJ (nonobese diabetic [NOD]) mice after onset of diabetes, when delayed gastric emptying develops, and after induction of HO1 to reverse delay. METHODS: Four groups of NOD and db/db mice were studied: nondiabetic, diabetic with normal emptying, diabetic with delayed gastric emptying, and diabetic with delayed gastric emptying reversed by the HO1 inducer hemin. Whole mount samples from stomach were labeled in triplicate with antisera against F4/80, HO1, and CD206, and macrophages were quantified in stacked confocal images. Markers for macrophage subtypes were measured by quantitative polymerase chain reaction. RESULTS: Development of diabetes was associated with an increased number of macrophages and up-regulation of HO1 in CD206(+) M2 macrophages. Onset of delayed gastric emptying did not alter the total number of macrophages, but there was a selective loss of CD206(+)/HO1(+) M2 macrophages. Normalization of gastric emptying was associated with repopulation of CD206(+)/HO1(+) M2 macrophages. CONCLUSIONS: CD206(+) M2 macrophages that express HO1 appear to be required for prevention of diabetes-induced delayed gastric emptying. Induction of HO1 in macrophages might be a therapeutic option for patients with diabetic gastroparesis.


Asunto(s)
Complicaciones de la Diabetes/prevención & control , Gastroparesia/prevención & control , Hemo-Oxigenasa 1/fisiología , Lectinas Tipo C/análisis , Macrófagos/fisiología , Lectinas de Unión a Manosa/análisis , Proteínas de la Membrana/fisiología , Receptores de Superficie Celular/análisis , Animales , Arginasa/genética , Glucemia/análisis , Femenino , Vaciamiento Gástrico , Hemo-Oxigenasa 1/análisis , Interleucina-10/genética , Macrófagos/enzimología , Receptor de Manosa , Proteínas de la Membrana/análisis , Ratones , Ratones Endogámicos NOD
20.
Antioxid Redox Signal ; 12(9): 1135-46, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19769466

RESUMEN

Hydrogen sulfide (H(2)S) has long been associated with the gastrointestinal tract, especially the bacteria-derived H(2)S present in flatus. Along with evidence from other organ systems, the finding that gastrointestinal tissues are capable of endogenous production of H(2)S has led to the hypothesis that H(2)S is an endogenous gaseous signaling molecule. In this review, the criteria of gasotransmitters are reexamined, and evidence from the literature regarding H(2)S as a gaseous signaling molecule is discussed. H(2)S is produced enzymatically by gastrointestinal tissues, but evidence is lacking on whether H(2)S production is regulated. H(2)S causes well-defined physiologic effects in gastrointestinal tissues, but evidence for a receptor for H(2)S is lacking. H(2)S is inactivated through enzymatic oxidation, but evidence is lacking on whether manipulating H(2)S oxidation alters endogenous cell signaling. Remaining questions regarding the role of H(2)S as a gaseous signaling molecule in the gastrointestinal tract suggest that H(2)S currently remains a molecule in search of a physiologic function.


Asunto(s)
Tracto Gastrointestinal/metabolismo , Sulfuro de Hidrógeno/metabolismo , Animales , Bacterias/metabolismo , Cristalografía por Rayos X/métodos , Gases , Tracto Gastrointestinal/microbiología , Humanos , Modelos Biológicos , Modelos Moleculares , Conformación Molecular , Oxígeno/química , Oxígeno/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...