Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Toxicology ; 458: 152831, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34097992

RESUMEN

Aryl hydrocarbon receptor (AHR) activation via 2,3,7,8-tetrachlorodibenzofuran (TCDF) induces the accumulation of hepatic lipids. Here we report that AHR activation by TCDF (24  µg/kg body weight given orally for five days) induced significant elevation of hepatic lipids including ceramides in mice, was associated with increased expression of key ceramide biosynthetic genes, and increased activity of their respective enzymes. Results from chromatin immunoprecipitation (ChIP), electrophoretic mobility shift assay (EMSA) and cell-based reporter luciferase assays indicated that AHR directly activated the serine palmitoyltransferase long chain base subunit 2 (Sptlc2, encodes serine palmitoyltransferase 2 (SPT2)) gene whose product catalyzes the initial rate-limiting step in de novo sphingolipid biosynthesis. Hepatic ceramide accumulation was further confirmed by mass spectrometry-based lipidomics. Taken together, our results revealed that AHR activation results in the up-regulation of Sptlc2, leading to ceramide accumulation, thus promoting lipogenesis, which can induce hepatic lipid accumulation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ceramidas/biosíntesis , Lipogénesis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Activación Metabólica/efectos de los fármacos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Benzofuranos/farmacología , Ceramidas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Lipidómica , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Hidrocarburo de Aril/genética , Serina C-Palmitoiltransferasa/genética , Serina C-Palmitoiltransferasa/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Triglicéridos/metabolismo
2.
mBio ; 11(4)2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32665268

RESUMEN

Beneficial microorganisms associated with animals derive their nutritional requirements entirely from the animal host, but the impact of these microorganisms on host metabolism is largely unknown. The focus of this study was the experimentally tractable tripartite symbiosis between the pea aphid Acyrthosiphon pisum, its obligate intracellular bacterial symbiont Buchnera, and the facultative bacterium Hamiltonella which is localized primarily to the aphid hemolymph (blood). Metabolome experiments on, first, multiple aphid genotypes that naturally bear or lack Hamiltonella and, second, one aphid genotype from which Hamiltonella was experimentally eliminated revealed no significant effects of Hamiltonella on aphid metabolite profiles, indicating that Hamiltonella does not cause major reconfiguration of host metabolism. However, the titer of just one metabolite, 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), displayed near-significant enrichment in Hamiltonella-positive aphids in both metabolome experiments. AICAR is a by-product of biosynthesis of the essential amino acid histidine in Buchnera and, hence, an index of histidine biosynthetic rates, suggesting that Buchnera-mediated histidine production is elevated in Hamiltonella-bearing aphids. Consistent with this prediction, aphids fed on [13C]histidine yielded a significantly elevated 12C/13C ratio of histidine in Hamiltonella-bearing aphids, indicative of increased (∼25%) histidine synthesized de novo by Buchnera However, in silico analysis predicted an increase of only 0.8% in Buchnera histidine synthesis in Hamiltonella-bearing aphids. We hypothesize that Hamiltonella imposes increased host demand for histidine, possibly for heightened immune-related functions. These results demonstrate that facultative bacteria can alter the dynamics of host metabolic interactions with co-occurring microorganisms, even when the overall metabolic homeostasis of the host is not substantially perturbed.IMPORTANCE Although microbial colonization of the internal tissues of animals generally causes septicemia and death, various animals are persistently associated with benign or beneficial microorganisms in their blood or internal organs. The metabolic consequences of these persistent associations for the animal host are largely unknown. Our research on the facultative bacterium Hamiltonella, localized primarily to the hemolymph of pea aphids, demonstrated that although Hamiltonella imposed no major reconfiguration of the aphid metabolome, it did alter the metabolic relations between the aphid and its obligate intracellular symbiont, Buchnera Specifically, Buchnera produced more histidine in Hamiltonella-positive aphids to support both Hamiltonella demand for histidine and Hamiltonella-induced increase in host demand. This study demonstrates how microorganisms associated with internal tissues of animals can influence specific aspects of metabolic interactions between the animal host and co-occurring microorganisms.


Asunto(s)
Áfidos/metabolismo , Áfidos/microbiología , Bacterias/metabolismo , Interacciones Microbiota-Huesped , Simbiosis , Animales , Buchnera/metabolismo , Femenino , Genotipo , Hemolinfa/microbiología , Histidina/metabolismo , Metabolómica
3.
Drug Metab Dispos ; 47(2): 86-93, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30409838

RESUMEN

Intestinal bacteria play an important role in bile acid metabolism and in the regulation of multiple host metabolic pathways (e.g., lipid and glucose homeostasis) through modulation of intestinal farnesoid X receptor (FXR) activity. Here, we examined the effect of berberine (BBR), a natural plant alkaloid, on intestinal bacteria using in vitro and in vivo models. In vivo, the metabolomic response and changes in mouse intestinal bacterial communities treated with BBR (100 mg/kg) for 5 days were assessed using NMR- and mass spectrometry-based metabolomics coupled with multivariate data analysis. Short-term BBR exposure altered intestinal bacteria by reducing Clostridium cluster XIVa and IV and their bile salt hydrolase (BSH) activity, which resulted in the accumulation of taurocholic acid (TCA). The accumulation of TCA was associated with activation of intestinal FXR, which can mediate bile acid, lipid, and glucose metabolism. In vitro, isolated mouse cecal bacteria were incubated with three doses of BBR (0.1, 1, and 10 mg/ml) for 4 hours in an anaerobic chamber. NMR-based metabolomics combined with flow cytometry was used to evaluate the direct physiologic and metabolic effect of BBR on the bacteria. In vitro, BBR exposure not only altered bacterial physiology but also changed bacterial community composition and function, especially reducing BSH-expressing bacteria like Clostridium spp. These data suggest that BBR directly affects bacteria to alter bile acid metabolism and activate FXR signaling. These data provide new insights into the link between intestinal bacteria, nuclear receptor signaling, and xenobiotics.


Asunto(s)
Berberina/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Amidohidrolasas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Ciego/efectos de los fármacos , Ciego/metabolismo , Ciego/microbiología , Clostridium/efectos de los fármacos , Clostridium/aislamiento & purificación , Clostridium/fisiología , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Metabolómica/métodos , Ratones , Ratones Endogámicos C57BL , Ácido Taurocólico/metabolismo
4.
J Proteome Res ; 17(4): 1375-1382, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29521512

RESUMEN

The liver and the mammary gland have complementary metabolic roles during lactation. Substrates synthesized by the liver are released into the circulation and are taken up by the mammary gland for milk production. The aryl hydrocarbon receptor (AHR) has been identified as a lactation regulator in mice, and its activation has been associated with myriad morphological, molecular, and functional defects such as stunted gland development, decreased milk production, and changes in gene expression. In this study, we identified adverse metabolic changes in the lactation network (mammary, liver, and serum) associated with AHR activation using 1H nuclear magnetic resonance (NMR)-based metabolomics. Pregnant mice expressing Ahr d (low affinity) or Ahr b (high affinity) were fed diets containing beta naphthoflavone (BNF), a potent AHR agonist. Mammary, serum, and liver metabolomics analysis identified significant changes in lipid and TCA cycle intermediates in the Ahr b mice. We observed decreased amino acid and glucose levels in the mammary gland extracts of Ahr b mice fed BNF. The serum of BNF fed Ahr b mice had significant changes in LDL/VLDL (increased) and HDL, PC, and GPC (decreased). Quantitative PCR analysis revealed ∼50% reduction in the expression of key lactogenesis mammary genes including whey acid protein, α-lactalbumin, and ß-casein. We also observed morphologic and developmental disruptions in the mammary gland that are consistent with previous reports. Our observations support that AHR activity contributes to metabolism regulation in the lactation network.


Asunto(s)
Metabolómica , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Caseínas/genética , Femenino , Expresión Génica , Lactalbúmina/genética , Lactancia/genética , Lactancia/metabolismo , Hígado/metabolismo , Glándulas Mamarias Animales/metabolismo , Ratones , Embarazo , Receptores de Hidrocarburo de Aril/agonistas , Proteína de Suero de Leche/genética
5.
Diabetes ; 66(3): 613-626, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28223344

RESUMEN

Increasing evidence supports the view that intestinal farnesoid X receptor (FXR) is involved in glucose tolerance and that FXR signaling can be profoundly impacted by the gut microbiota. Selective manipulation of the gut microbiota-FXR signaling axis was reported to significantly impact glucose intolerance, but the precise molecular mechanism remains largely unknown. Here, caffeic acid phenethyl ester (CAPE), an over-the-counter dietary supplement and an inhibitor of bacterial bile salt hydrolase, increased levels of intestinal tauro-ß-muricholic acid, which selectively suppresses intestinal FXR signaling. Intestinal FXR inhibition decreased ceramide levels by suppressing expression of genes involved in ceramide synthesis specifically in the intestinal ileum epithelial cells. The lower serum ceramides mediated decreased hepatic mitochondrial acetyl-CoA levels and pyruvate carboxylase (PC) activities and attenuated hepatic gluconeogenesis, independent of body weight change and hepatic insulin signaling in vivo; this was reversed by treatment of mice with ceramides or the FXR agonist GW4064. Ceramides substantially attenuated mitochondrial citrate synthase activities primarily through the induction of endoplasmic reticulum stress, which triggers increased hepatic mitochondrial acetyl-CoA levels and PC activities. These results reveal a mechanism by which the dietary supplement CAPE and intestinal FXR regulates hepatic gluconeogenesis and suggest that inhibiting intestinal FXR is a strategy for treating hyperglycemia.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Ácidos Cafeicos/farmacología , Ceramidas/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Gluconeogénesis/efectos de los fármacos , Íleon/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Alcohol Feniletílico/análogos & derivados , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Acetilcoenzima A/efectos de los fármacos , Acetilcoenzima A/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Dieta Alta en Grasa , Íleon/metabolismo , Insulina/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Isoxazoles/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Hepáticas/metabolismo , Alcohol Feniletílico/farmacología , Piruvato Carboxilasa/efectos de los fármacos , Piruvato Carboxilasa/metabolismo , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/efectos de los fármacos , Ácido Taurocólico/análogos & derivados , Ácido Taurocólico/metabolismo
6.
Cell Mol Gastroenterol Hepatol ; 2(3): 328-339, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27446985

RESUMEN

BACKGROUND & AIMS: High-fat diet (HFD) feeding is associated with gastrointestinal motility disorders. We recently reported delayed colonic motility in mice fed a HFD mice for 11 weeks. In this study, we investigated the contributing role of gut microbiota in HFD-induced gut dysmotility. METHODS: Male C57BL/6 mice were fed a HFD (60% kcal fat) or a regular/control diet (RD) (18% kcal fat) for 13 weeks. Serum and fecal endotoxin levels were measured, and relative amounts of specific gut bacteria in the feces assessed by real time PCR. Intestinal transit was measured by fluorescent-labeled marker and bead expulsion test. Enteric neurons were assessed by immunostaining. Oligofructose (OFS) supplementation with RD or HFD for 5 weeks was also studied. In vitro studies were performed using primary enteric neurons and an enteric neuronal cell line. RESULTS: HFD-fed mice had reduced numbers of enteric nitrergic neurons and exhibited delayed gastrointestinal transit compared to RD-fed mice. HFD-fed mice had higher fecal Firmicutes and Escherichia coli and lower Bacteroidetes compared to RD-fed mice. OFS supplementation protected against enteric nitrergic neurons loss in HFD-fed mice, and improved intestinal transit time. OFS supplementation resulted in a reductions in fecal Firmicutes and Escherichia coli and serum endotoxin levels. In vitro, palmitate activation of TLR4 induced enteric neuronal apoptosis in a p-JNK1 dependent pathway. This apoptosis was prevented by a JNK inhibitor and in neurons from TLR4-/- mice. CONCLUSIONS: Together our data suggest that intestinal dysbiosis in HFD fed mice contribute to the delayed intestinal motility by inducing a TLR4-dependant neuronal loss. Manipulation of gut microbiota with OFS improved intestinal motility in HFD mice.

7.
J Proteome Res ; 15(8): 2626-33, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27246581

RESUMEN

Endometriosis is a common chronic estrogen-dependent gynecological disease affecting 10% of women in their reproductive age. It is characterized by proliferation of functional endometrial glands and stroma outside the uterine cavity. In the present study, we used mass spectrometry-based lipidomics to investigate the alterations in serum lipid profiles of mice induced with endometriosis. We identified several dysregulated lipids such as phosphatidylcholines, sphingomyelins, phosphatidylethanolamines, and triglycerides and show that triglycerides may be due to a general inflammatory condition in the peritoneum. We also show that in addition to phosphatidylcholine alteration, there is also an effect in the ratio of phosphatidylcholine/phosphatidylethanolamine in serum of mice induced with the disease and that this change may be due to increased expression of the phosphatidylethanolamine N-methyltransferase gene. The study provides new insight into the etiology of endometriosis.


Asunto(s)
Endometriosis/metabolismo , Metabolismo de los Lípidos , Metabolómica , Animales , Endometriosis/etiología , Femenino , Ratones , Fosfatidilcolinas/sangre , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/sangre , Fosfatidiletanolaminas/metabolismo , Esfingomielinas/metabolismo , Triglicéridos/metabolismo
8.
Mol Ther Methods Clin Dev ; 2: 15028, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26366426

RESUMEN

Depending on the population examined, from 6 to 83% of people with diabetes mellitus exhibit symptoms of altered gut motility, manifesting as dysphagia, reflux, early satiety, nausea, abdominal pain, diarrhea, or constipation. Hyperglycemia-induced cell loss within the enteric nervous system has been demonstrated in both diabetic rodents and patients with diabetes. Glycemic control is recommended to prevent diabetic gastroenteropathy but is often difficult to achieve with current treatment modalities. We asked if hepatic insulin gene therapy (HIGT) could inhibit the development of diabetic gastroenteropathy in mice. Bowel length, bowel transit, colonic muscle relaxation, and the numbers of both stimulatory and inhibitory neurons in the colonic myenteric plexus were compared in groups of diabetic mice (DM), control nondiabetic mice (Con), and diabetic mice treated with HIGT (HIGT). Delivery of a metabolically responsive insulin transgene to the liver of STZ-diabetic mice with an adeno-associated virus, sero-type 8 (AAV8) produced near-normal blood sugars for over 1 month and prevented anatomic, functional, and neurohistologic changes observed in diabetic mice. We conclude that in addition to normalizing oxidative metabolism in diabetic rodents, HIGT is sufficient to prevent the development of diabetic gastroenteropathy.

9.
Gastroenterology ; 146(2): 473-83.e3, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24507550

RESUMEN

BACKGROUND & AIMS: A high-fat diet (HFD) can cause serious health problems, including alteration of gastrointestinal transit, the exact mechanism of which is not clear. Several microRNAs (miRNAs) are involved in energy homeostasis, lipid metabolism, and HFD-induced weight gain. We investigated the role of miRNAs in HFD-induced damage to the enteric nervous system. METHODS: Male mice were fed a HFD (60% calories from fat) or regular diets (18% calories from fat) for 11 weeks. Mice on regular diets and HFDs were given intraperitoneal injections of Mir375 inhibitor or a negative control. Body weights, food intake, stool indices, and gastrointestinal transit (following Evans blue gavage) were measured. An enteric neuronal cell line (immorto-fetal enteric neuronal) and primary enteric neurons were used for in vitro studies. RESULTS: HFD delayed intestinal transit, which was associated with increased apoptosis and loss of colonic myenteric neurons. Mice fed a low-palmitate HFD did not develop a similar phenotype. Palmitate caused apoptosis of enteric neuronal cells associated with mitochondrial dysfunction and endoplasmic reticulum stress. Palmitate significantly increased the expression of Mir375 in vitro; transfection of cells with a Mir375 inhibitor prevented the palmitate-induced enteric neuronal cell apoptosis. Mir375 expression was increased in myenteric ganglia of mice fed HFD and associated with decreased levels of Mir375 target messenger RNAs, including Pdk1. Systemic injection of a Mir375 inhibitor for 5 weeks prevented HFD-induced delay in intestinal transit and morphologic changes. CONCLUSIONS: HFDs delay colonic transit, partly by inducing apoptosis in enteric neuronal cells. This effect is mediated by Mir375 and is associated with reduced levels of Pdk1. Mir375 might be targeted to increase survival of enteric neurons and gastrointestinal motility.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/efectos adversos , Sistema Nervioso Entérico/patología , Tránsito Gastrointestinal/fisiología , MicroARNs/metabolismo , Neuronas/patología , Palmitatos/efectos adversos , Animales , Apoptosis/fisiología , Biomarcadores/metabolismo , Línea Celular , Colon/inervación , Colon/patología , Colon/fisiopatología , Sistema Nervioso Entérico/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/administración & dosificación , MicroARNs/antagonistas & inhibidores , Neuronas/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Distribución Aleatoria , Estrés Fisiológico
10.
Am J Physiol Gastrointest Liver Physiol ; 306(6): G515-25, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24458024

RESUMEN

Obesity is a growing epidemic with limited effective treatments. The neurotrophic factor glial cell line-derived neurotrophic factor (GDNF) was recently shown to enhance ß-cell mass and improve glucose control in rodents. Its role in obesity is, however, not well characterized. In this study, we investigated the ability of GDNF to protect against high-fat diet (HFD)-induced obesity. GDNF transgenic (Tg) mice that overexpress GDNF under the control of the glial fibrillary acidic protein promoter and wild-type (WT) littermates were maintained on a HFD or regular rodent diet for 11 wk, and weight gain, energy expenditure, and insulin sensitivity were monitored. Differentiated mouse brown adipocytes and 3T3-L1 white adipocytes were used to study the effects of GDNF in vitro. Tg mice resisted the HFD-induced weight gain, insulin resistance, dyslipidemia, hyperleptinemia, and hepatic steatosis seen in WT mice despite similar food intake and activity levels. They exhibited significantly (P<0.001) higher energy expenditure than WT mice and increased expression in skeletal muscle and brown adipose tissue of peroxisome proliferator activated receptor-α and ß1- and ß3-adrenergic receptor genes, which are associated with increased lipolysis and enhanced lipid ß-oxidation. In vitro, GDNF enhanced ß-adrenergic-mediated cAMP release in brown adipocytes and suppressed lipid accumulation in differentiated 3T3L-1 cells through a p38MAPK signaling pathway. Our studies demonstrate a novel role for GDNF in the regulation of high-fat diet-induced obesity through increased energy expenditure. They show that GDNF and its receptor agonists may be potential targets for the treatment or prevention of obesity.


Asunto(s)
Dieta Alta en Grasa , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Obesidad/prevención & control , Células 3T3-L1 , Animales , Metabolismo Energético , Hígado Graso/prevención & control , Resistencia a la Insulina , Masculino , Ratones , Ratones Transgénicos , Triglicéridos/metabolismo
11.
Dig Dis Sci ; 58(6): 1516-27, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23371009

RESUMEN

BACKGROUND: Recently, two enteric neuronal cell lines, one fetal and the other post-natal (IM-PEN), have been developed from the H-2K(b)-tsA58 transgenic mouse (immortomouse). However, their electrophysiological properties are not known. The goal of this study was to determine the electrical excitability and ionic conductance of the immortalized postnatal enteric neuronal (IM-PEN) cell line. METHODS: Whole cell patch clamp studies, immunohistochemistry and RT-PCR were performed on differentiated IM-PEN cells following propagation at 33 °C and differentiation at 37 °C. RESULTS: Differentiated IM-PEN cells stained positively for the neuron specific markers ßIII-tubulin and PGP9.5. The mRNA for several ion channels expressed in enteric neurons were detected by RT-PCR. In current clamp, the resting membrane potential was -24.6 ± 2.1 mV (n = 6) for IM-FEN and -29.8 ± 0.9 mV (n = 30) for IM-PEN. Current injections from Vh -80 mV resulted in passive responses but not action potentials. Depolarizing pulses in the whole cell voltage clamp configuration from Vh -80 mV elicited small nifedipine-sensitive inward currents. Additionally, outward currents with slow deactivating tail currents were blocked by niflumic acid and low chloride solution. A volume-regulated anion current was elicited by hypo-osmotic solution and inhibited by 10 µM DCPIB. Growth with rabbit gastrointestinal smooth muscle did not yield significant differences in the active properties of the IM-PEN cell line. Transient expression of L-type Ca(2+) channels produced large inward currents demonstrating a working mechanism for protein folding and transport. CONCLUSION: The electrophysiological characteristics of IM-PEN cells suggest that chloride channels in IM-PEN cells play an important role in their resting state, and membrane trafficking of some of the ion channels may preclude their electrical excitability.


Asunto(s)
Sistema Nervioso Entérico/citología , Activación del Canal Iónico , Potenciales de la Membrana , Neuronas/fisiología , Animales , Biomarcadores/metabolismo , Línea Celular , Inmunohistoquímica , Canales Iónicos/fisiología , Ratones , Ratones Transgénicos , Técnicas de Placa-Clamp , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Gastroenterology ; 143(4): 1006-16.e4, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22732731

RESUMEN

BACKGROUND & AIMS: Altered gastrointestinal motility is associated with significant morbidity and health care costs. Toll-like receptors (TLR) regulate intestinal homeostasis. We examined the roles of TLR4 signaling in survival of enteric neurons and gastrointestinal motility. METHODS: We assessed changes in intestinal motility by assessing stool frequency, bead expulsion, and isometric muscle recordings of colonic longitudinal muscle strips from mice that do not express TLR4 (Tlr4(Lps-d) or TLR4(-/-)) or Myd88 (Myd88(-/-)), in wild-type germ-free mice or wild-type mice depleted of the microbiota, and in mice with neural crest-specific deletion of Myd88 (Wnt1Cre(+/-)/Myd88(fl/fl)). We studied the effects of the TLR4 agonist lipopolysaccharide (LPS) on survival of cultured, immortalized fetal enteric neurons and enteric neuronal cells isolated from wild-type and Tlr4(Lps-d) mice at embryonic day 13.5. RESULTS: There was a significant delay in gastrointestinal motility and reduced numbers of nitrergic neurons in TLR4(Lps-d), TLR4(-/-), and Myd88(-/-) mice compared with wild-type mice. A similar phenotype was observed in germ-free mice, mice depleted of intestinal microbiota, and Wnt1Cre(+/-)/Myd88(fl/fl) mice. Incubation of enteric neuronal cells with LPS led to activation of the transcription factor nuclear factor (NF)-κB and increased cell survival. CONCLUSIONS: Interactions between enteric neurons and microbes increases neuron survival and gastrointestinal motility in mice. LPS activation of TLR4 and NF-κB appears to promote survival of enteric neurons. Factors that regulate TLR4 signaling in neurons might be developed to alter gastrointestinal motility.


Asunto(s)
Sistema Nervioso Entérico/metabolismo , Motilidad Gastrointestinal , Metagenoma , Factor 88 de Diferenciación Mieloide/metabolismo , Neuronas Nitrérgicas/metabolismo , Receptor Toll-Like 4/metabolismo , Análisis de Varianza , Animales , Antibacterianos/farmacología , Apoptosis , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Neuronas Colinérgicas/fisiología , Colon/fisiología , Defecación , Ingestión de Alimentos , Endotoxinas/sangre , Sistema Nervioso Entérico/microbiología , Heces/microbiología , Femenino , Motilidad Gastrointestinal/efectos de los fármacos , Lipopolisacáridos/sangre , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Contracción Muscular , Músculo Liso/fisiología , Factor 88 de Diferenciación Mieloide/genética , Neuronas Nitrérgicas/microbiología , Neuronas Nitrérgicas/fisiología , Fenotipo , Transducción de Señal , Receptor Toll-Like 4/genética
13.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 36(6): 546-53, 2011 Jun.
Artículo en Chino | MEDLINE | ID: mdl-21743147

RESUMEN

OBJECTIVE: To evaluate the effect of hepatic insulin gene therapy on diabetic enteric neuropathy. METHODS: Mice were randomly allocated into 3 groups: a normal control group, a diabetic group, and a diabetic gene therapy group. Diabetes were induced by penial vein injection of streptozocin (STZ). The gene therapy group received hepatic insulin gene therapy while the other 2 groups only received an empty virus expressing green fluorescent protein. Random blood glucose, body weight growth, gastric emptying, total bowel length, absolute and relative bowel transit, electric field stimulation of colon smooth muscle, colon nuclei staining and counting were measured. RESULTS: We successully established a mouse model of diabetic enteric neuropathy which manifests as: 8 weeks of continuous hyperglycemia,increased total bowel length, decreased relative bowel transit, impaired colon smooth muscle relaxation and loss of inhibitory neurons in colon. Through gene therapy, the above indexes were normalized or ameliorated, suggesting hepatic insulin gene therapy is capable of preventing diabetic enteric neuropathy. CONCLUSION: Hepatic insulin gene therapy can prevent STZ induced diabetic enteric neuropathy.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Neuropatías Diabéticas/terapia , Sistema Nervioso Entérico/patología , Terapia Genética , Insulina/genética , Adenoviridae , Animales , Diabetes Mellitus Experimental/complicaciones , Sistema Nervioso Entérico/metabolismo , Enfermedades Gastrointestinales/etiología , Enfermedades Gastrointestinales/terapia , Técnicas de Transferencia de Gen , Vectores Genéticos , Hepatocitos/metabolismo , Insulina/metabolismo , Ratones , Proinsulina/genética
14.
Am J Physiol Gastrointest Liver Physiol ; 299(1): G283-92, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20448145

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) is a factor produced by glial cells that is required for the development of the enteric nervous system. In transgenic mice that overexpress GDNF in the pancreas, GDNF has been shown to enhance beta-cell mass and improve glucose control, but the transcriptional and cellular processes involved are not known. In this study we examined the influence of GDNF on the expression of neurogenin3 (Ngn3) and other transcription factors implicated in early beta-cell development, as well as on beta-cell proliferation during embryonic and early postnatal mouse pancreas development. Embryonic day 15.5 (E15.5) mouse pancreatic tissue when exposed to GDNF for 24 h showed higher Ngn3, pancreatic and duodenal homeobox gene 1 (Pdx1), neuroD1/beta(2), paired homeobox gene 4 (Pax4), and insulin mRNA expression than tissue exposed to vehicle only. Transgenic expression of GDNF in mouse pancreata was associated with increased numbers of Ngn3-expressing pancreatic cells and higher beta-cell mass at embryonic day 18 (E18), as well as higher beta-cell proliferation and Pdx1 expression in beta-cells at E18 and postnatal day 1. In the HIT-T15 beta-cell line, GDNF enhanced the expression of Pax6. This response was, however, blocked in the presence of Pdx1 small interfering RNA (siRNA). Chromatin immunoprecipitation studies using the HIT-T15 beta-cell line demonstrated that GDNF can influence Pdx1 gene expression by enhancing the binding of Sox9 and neuroD1/beta(2) to the Pdx1 promoter. Our data provide evidence of a mechanism by which GDNF influences beta-cell development. GDNF could be a potential therapeutic target for the treatment and prevention of diabetes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proliferación Celular , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Páncreas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sitios de Unión , Línea Celular , Inmunoprecipitación de Cromatina , Cricetinae , Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Organogénesis , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Páncreas/embriología , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Mensajero/metabolismo , Ratas , Proteínas Recombinantes/metabolismo , Proteínas Represoras/metabolismo , Factor de Transcripción SOX9/metabolismo , Transactivadores/metabolismo , Factor de Transcripción HES-1 , Activación Transcripcional , Transfección , Regulación hacia Arriba
15.
Am J Physiol Gastrointest Liver Physiol ; 298(3): G375-83, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20007850

RESUMEN

The bone morphogenetic protein (BMP) family is a class of transforming growth factor (TGF-beta) superfamily molecules that have been implicated in neuronal differentiation. We studied the effects of BMP2 and glial cell line-derived neurotrophic factor (GDNF) on inducing differentiation of enteric neurons and the signal transduction pathways involved. Studies were performed using a novel murine fetal enteric neuronal cell line (IM-FEN) and primary enteric neurons. Enteric neurons were cultured in the presence of vehicle, GDNF (100 ng/ml), BMP2 (10 ng/ml), or both (GDNF + BMP2), and differentiation was assessed by neurite length, markers of neuronal differentiation (neurofilament medium polypeptide and beta-III-tubulin), and neurotransmitter expression [neuropeptide Y (NPY), neuronal nitric oxide synthase (nNOS), tyrosine hydroxylase (TH), choline acetyltransferase (ChAT) and Substance P]. BMP2 increased the differentiation of enteric neurons compared with vehicle and GDNF-treated neurons (P < 0.001). BMP2 increased the expression of the mature neuronal markers (P < 0.05). BMP2 promoted differentiation of NPY-, nNOS-, and TH-expressing neurons (P < 0.001) but had no effect on the expression of cholinergic neurons (ChAT, Substance P). Neurons cultured in the presence of BMP2 have higher numbers of TH-expressing neurons after exposure to 1-methyl 4-phenylpyridinium (MPP(+)) compared with those cultured with MPP(+) alone (P < 0.01). The Smad signal transduction pathway has been implicated in TGF-beta signaling. BMP2 induced phosphorylation of Smad1, and the effects of BMP2 on differentiation of enteric neurons were significantly reduced in the presence of Smad1 siRNA, implicating the role of Smad1 in BMP2-induced differentiation. The effects of BMP2 on catecholaminergic neurons may have therapeutic implications in gastrointestinal motility disturbances.


Asunto(s)
Proteína Morfogenética Ósea 2/farmacología , Catecolaminas/metabolismo , Diferenciación Celular/fisiología , Sistema Nervioso Entérico/citología , Neuronas/citología , Neuronas Nitrérgicas/citología , Proteína Smad1/metabolismo , 1-Metil-4-fenilpiridinio/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Ratones , Proteínas de Neurofilamentos/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Neuronas Nitrérgicas/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Proteína Smad1/genética , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , Ubiquitina Tiolesterasa/metabolismo
16.
Gastroenterology ; 134(5): 1424-35, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18471518

RESUMEN

BACKGROUND & AIMS: The isolation and culture of primary enteric neurons is a difficult process and yields a small number of neurons. We developed fetal and postnatal enteric neuronal cell lines using H-2K(b)-tsA58 transgenic mice (immortomice) that have a temperature-sensitive mutation of the SV40 large tumor antigen gene under the control of an interferon gamma-inducible H-2K(b) promoter element. METHODS: Enteric neuronal precursors were isolated from the intestines of E13-mouse fetuses and second day postnatal mice using magnetic immunoselection with a p75NTR antibody. The cells were maintained at the permissive temperature, 33 degrees C, and interferon-gamma for 24 or 48 hours, and then transferred to 39 degrees C in the presence of glial cell line-derived neurotrophic factor for 7 days for further differentiation. Neuronal markers were assessed by reverse-transcription polymerase chain reaction, Western blot, and immunocytochemistry. Neuronal function was assessed by transplanting these cells into the colons of Piebald or nNOS(-/-) mice. RESULTS: Expression analysis of cells showed the presence of neuronal markers peripherin, PGP9.5, HuD, tau, synaptic marker synaptophysin, characteristic receptors of enteric neurons, Ret, and 5-hydroxytryptamine-receptor subtypes at 33 degrees C and 39 degrees C. Nestin, S-100beta, and alpha-smooth muscle actin were expressed minimally at 39 degrees C. Glial cell line-derived neurotrophic factor resulted in increased phosphorylation of Akt in these cells, similar to primary enteric neurons. Transplantation of cells into the piebald or nNOS(-/-) mice colon improved colonic motility. CONCLUSIONS: We have developed novel enteric neuronal cell lines that have neuronal characteristics similar to primary enteric neurons. These cells can help us in understanding newer therapeutic options for Hirschsprung's disease.


Asunto(s)
Colon/inervación , Sistema Nervioso Entérico/embriología , Motilidad Gastrointestinal/fisiología , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , ARN/genética , Actinas/biosíntesis , Actinas/genética , Animales , Western Blotting , Línea Celular , Colon/embriología , Colon/cirugía , Proteínas ELAV/biosíntesis , Proteínas ELAV/genética , Proteína 4 Similar a ELAV , Sistema Nervioso Entérico/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Inmunohistoquímica , Proteínas de Filamentos Intermediarios/biosíntesis , Proteínas de Filamentos Intermediarios/genética , Contracción Isométrica/fisiología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso/inervación , Músculo Liso/fisiología , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/biosíntesis , Nestina , Neuroglía/citología , Neuroglía/metabolismo , Neuroglía/trasplante , Neuronas/citología , Periferinas , Embarazo , Proteínas Proto-Oncogénicas c-ret/biosíntesis , Proteínas Proto-Oncogénicas c-ret/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subunidad beta de la Proteína de Unión al Calcio S100 , Proteínas S100/biosíntesis , Proteínas S100/genética , Serotonina/biosíntesis , Serotonina/genética , Sinaptofisina/biosíntesis , Sinaptofisina/genética , Ubiquitina Tiolesterasa/biosíntesis , Ubiquitina Tiolesterasa/genética , Proteínas de Xenopus , Proteínas tau/biosíntesis
17.
Gastroenterology ; 134(3): 727-37, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18241861

RESUMEN

BACKGROUND & AIMS: Pancreatic beta-cell mass increases in response to increased demand for insulin, but the factors involved are largely unknown. Glial cell line-derived neurotrophic factor (GDNF) is a growth factor that plays a role in the development and survival of the enteric nervous system. We investigated the role of GDNF in regulating beta-cell survival. METHODS: Studies were performed using the beta-TC-6 pancreatic beta-cell line, isolated mouse pancreatic beta cells, and in vivo in transgenic mice that overexpress GDNF in pancreatic glia. GDNF receptor family alpha1 and c-Ret receptor expression were assessed by reverse-transcription polymerase chain reaction and immunofluorescence microscopy. Apoptosis was evaluated by assessing caspase-3 cleavage. Phosphoinositol-3-kinase signaling pathway was analyzed by Akt phosphorylation. Glucose homeostasis was assessed by performing intraperitoneal glucose tolerance tests. Insulin sensitivity was assessed using intraperitoneal injection of insulin. RESULTS: We demonstrate the presence of receptors for GDNF, GFRalpha1, and c-Ret on beta cells. GDNF promoted beta-cell survival and proliferation and protected them from thapsigargin-induced apoptosis (P<.0001) in vitro. Exposure of beta-cells to GDNF also resulted in phosphorylation of Akt and GSK3beta. Transgenic mice that overexpress GDNF in glia exhibit increased beta-cell mass, proliferation, and insulin content. No differences in insulin sensitivity and c-peptide levels were noted. Compared with wild-type mice, GDNF-transgenic mice have significantly lower blood glucose levels and improved glucose tolerance (P<.01). GDNF-transgenic mice are resistant to streptozotocin-induced beta-cell loss (P<.001) and subsequent hyperglycemia. CONCLUSIONS: We demonstrate that over expression of GDNF in pancreatic glia improves glucose tolerance and that GDNF may be a therapeutic target for improving beta-cell mass.


Asunto(s)
Glucemia/metabolismo , Proliferación Celular , Diabetes Mellitus Experimental/prevención & control , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Intolerancia a la Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Apoptosis , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Intolerancia a la Glucosa/patología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Insulina/sangre , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Ratones , Ratones Transgénicos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Transducción de Señal , Tapsigargina/farmacología , Factores de Tiempo , Regulación hacia Arriba
18.
Exp Neurol ; 207(1): 4-12, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17586496

RESUMEN

Gastrointestinal (GI) dysfunction is the most common non-motor symptom of Parkinson's disease (PD). Symptoms of GI dysmotility include early satiety and nausea from delayed gastric emptying, bloating from poor small bowel coordination, and constipation and defecatory dysfunction from impaired colonic transit. Understanding the pathophysiology and treatment of these symptoms in PD patients has been hampered by the lack of investigation into GI symptoms and pathology in PD animal models. We report that the prototypical parkinsonian neurotoxin, MPTP (1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine), is a selective dopamine neuron toxin in the enteric nervous system (ENS). When examined 10 days after treatment, there was a 40% reduction of dopamine neurons in the ENS of C57Bl/6 mice administered MPTP (60 mg/kg). There were no differences in the density of cholinergic or nitric oxide neurons. Electrophysiological recording of neural-mediated muscle contraction in isolated colon from MPTP-treated animals confirmed a relaxation defect associated with dopaminergic degeneration. Behaviorally, MPTP induced a transient increase in colon motility, but no changes in gastric emptying or small intestine transit. These results provide the first comprehensive assessment of gastrointestinal pathophysiology in an animal model of PD. They provide insight into the impact of dopaminergic dysfunction on gastrointestinal motility and a benchmark for assessment of other PD model systems.


Asunto(s)
Colon/fisiopatología , Dopamina/metabolismo , Sistema Nervioso Entérico/patología , Motilidad Gastrointestinal , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson Secundaria/patología , Enfermedad de Parkinson Secundaria/fisiopatología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Catecolaminas/metabolismo , Recuento de Células , Dopaminérgicos , Sistema Nervioso Entérico/metabolismo , Vaciamiento Gástrico/efectos de los fármacos , Tránsito Gastrointestinal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibición Neural , Enfermedad de Parkinson Secundaria/inducido químicamente
19.
Gastroenterology ; 131(4): 1164-78, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17030186

RESUMEN

BACKGROUND & AIMS: Glial-derived neurotrophic factor (GDNF) promotes the survival and proliferation of enteric neurons. Neuropeptide Y (NPY) is an important peptide regulating gastrointestinal motility. The role of NPY on the survival and proliferation of enteric neurons is not known. We examined the effects of GDNF on the expression and release of NPY from enteric neurons and the role of NPY in promoting enteric neuronal proliferation and survival. METHODS: Studies were performed in primary enteric neuronal cultures and NPY knockout mice (NPY(-/-)). GDNF-induced expression of NPY was assessed by reverse-transcription polymerase chain reaction (RT-PCR), immunocytochemistry, and enzyme-linked immunosorbent assay. Using NPY-siRNA and NPY-Y1 receptor antagonist, we examined the role of NPY in mediating the survival and proliferation effects of GDNF. Gastrointestinal motility was assessed by measuring gastric emptying, intestinal transit, and isometric muscle recording from intestinal muscle strips. RESULTS: GDNF induced a significant increase in NPY messenger RNA and protein expression in primary enteric neurons and the release of NPY into the culture medium. NPY (1 mumol/L) significantly increased proliferation of neurons and reduced apoptosis. In the presence of NPY-siRNA and NPY-Y1 receptor antagonist or in enteric neurons cultured from NPY(-/-) mice, GDNF-mediated neuronal proliferation and survival was reduced. NPY increased the phosphorylation of Akt, a downstream target of the PI-3-kinase pathway. In NPY(-/-) mice, there were significantly fewer nNOS-containing enteric neurons compared with wild-type (WT) mice. NPY(-/-) mice had accelerated gastric emptying and delayed intestinal transit compared with WT mice. CONCLUSIONS: We demonstrate that NPY acts as an autocrine neurotrophic factor for enteric neurons.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Plexo Mientérico/citología , Plexo Mientérico/fisiología , Neuronas/citología , Neuropéptido Y/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Cromonas/farmacología , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Vaciamiento Gástrico/fisiología , Motilidad Gastrointestinal/fisiología , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Intestinos/inervación , Intestinos/fisiología , Ratones , Ratones Noqueados , Morfolinas/farmacología , Relajación Muscular/fisiología , Músculo Liso/inervación , Músculo Liso/fisiología , Neuropéptido Y/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/metabolismo
20.
J Clin Invest ; 116(2): 344-56, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16453021

RESUMEN

Diabetes can result in loss of enteric neurons and subsequent gastrointestinal complications. The mechanism of enteric neuronal loss in diabetes is not known. We examined the effects of hyperglycemia on enteric neuronal survival and the effects of glial cell line-derived neurotrophic factor (GDNF) on modulating this survival. Exposure of primary enteric neurons to 20 mM glucose (hyperglycemia) for 24 hours resulted in a significant increase in apoptosis compared with 5 mM glucose (normoglycemia). Exposure to 20 mM glucose resulted in decreased Akt phosphorylation and enhanced nuclear translocation of forkhead box O3a (FOXO3a). Treatment of enteric neurons with GDNF ameliorated these changes. In streptozotocin-induced diabetic mice, there was evidence of myenteric neuronal apoptosis and reduced Akt phosphorylation. Diabetic mice had loss of NADPH diaphorase-stained myenteric neurons, delayed gastric emptying, and increased intestinal transit time. The pathophysiological effects of hyperglycemia (apoptosis, reduced Akt phosphorylation, loss of inhibitory neurons, motility changes) were reversed in diabetic glial fibrillary acidic protein-GDNF (GFAP-GDNF) Tg mice. In conclusion, we demonstrate that hyperglycemia induces neuronal loss through a reduction in Akt-mediated survival signaling and that these effects are reversed by GDNF. GDNF may be a potential therapeutic target for the gastrointestinal motility disorders related to diabetes.


Asunto(s)
Neuropatías Diabéticas/metabolismo , Sistema Nervioso Entérico , Enfermedades Gastrointestinales , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Hiperglucemia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apoptosis/fisiología , Caspasa 3 , Caspasas/metabolismo , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Sistema Nervioso Entérico/metabolismo , Sistema Nervioso Entérico/patología , Activación Enzimática , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Enfermedades Gastrointestinales/etiología , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/patología , Tracto Gastrointestinal/anatomía & histología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/patología , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Glucosa/metabolismo , Humanos , Ratones , Neuronas/metabolismo , Neuronas/ultraestructura , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA