Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Gastro Hep Adv ; 3(1): 38-47, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38390283

RESUMEN

BACKGROUND AND AIMS: The overexpression of glial cell-derived neurotrophic factor (GDNF) in the liver and adipose tissues offers strong protection against high-fat diet (HFD)-induced obesity in mice. We hypothesize that sustainably enhancing GDNF expression in the liver may provide a therapeutic effect that can prevent the progression of HFD-induced obesity in mice. METHODS: Expression lentivector encoding mouse GDNF (GDNF(pDNA) or empty vector (pDNA, control) were encapsulated in lipid nanoparticles (LNPs) using the thin-film hydration method. Mice were fed with regular diet (RD) or HFD for 20 weeks prior to injection and the GDNF and control vector-loaded LNPs were administered by intravenous (IV) injection to mice once weekly for 5 weeks. Changes in body weight were monitored and mice tissues were collected and imaged for fluorescence using an IVIS in vivo imaging system. Post-treatment abdominal fat weight, colon length, and spleen weight were obtained. GDNF protein levels in the liver and serum were quantified by enzyme-linked immunosorbent assay, while liver AKT serine/threonine kinase and AMP-activated protein kinase phosphorylation levels were evaluated by Western blotting. RESULTS: IV-injected GDNF(pDNA)-loaded LNPs targeted the liver and remained in there for up to 15 days postinjection. A single injection of GDNF(pDNA)-loaded LNPs significantly increased GDNF expression for 7 days and consequently increased the levels of phosphorylated AKT serine/threonine kinase and AMP-activated protein kinase. Once weekly injections of GDNF(pDNA)-loaded LNPs for 5 weeks slowed increase in body weight, reduced abdominal fat, and modulated the gut microbiota toward a healthier composition in HFD-fed mice. CONCLUSION: GDNF(pDNA)-loaded LNPs could potentially be developed as a therapeutic strategy to reverse weight gain in obese patients.

2.
Biomolecules ; 13(2)2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36830577

RESUMEN

BACKGROUND: Diarrhea is present in up to 30-50% of patients with COVID-19. The mechanism of SARS-CoV-2-induced diarrhea remains unclear. We hypothesized that enterocyte-enteric neuron interactions were important in SARS-CoV-2-induced diarrhea. SARS-CoV-2 induces endoplasmic reticulum (ER) stress in enterocytes causing the release of damage associated molecular patterns (DAMPs). The DAMPs then stimulate the release of enteric neurotransmitters that disrupt gut electrolyte homeostasis. METHODS: Primary mouse enteric neurons (EN) were exposed to a conditioned medium from ACE2-expressing Caco-2 colonic epithelial cells infected with SARS-CoV-2 or treated with tunicamycin (ER stress inducer). Vasoactive intestinal peptides (VIP) expression and secretion by EN were assessed by RT-PCR and ELISA, respectively. Membrane expression of NHE3 was determined by surface biotinylation. RESULTS: SARS-CoV-2 infection led to increased expression of BiP/GRP78, a marker and key regulator for ER stress in Caco-2 cells. Infected cells secreted the DAMP protein, heat shock protein 70 (HSP70), into the culture media, as revealed by proteomic and Western analyses. The expression of VIP mRNA in EN was up-regulated after treatment with a conditioned medium of SARS-CoV-2-infected Caco-2 cells. CD91, a receptor for HSP70, is abundantly expressed in the cultured mouse EN. Tunicamycin, an inducer of ER stress, also induced the release of HSP70 and Xbp1s, mimicking SARS-CoV-2 infection. Co-treatment of Caco-2 with tunicamycin (apical) and VIP (basolateral) induced a synergistic decrease in membrane expression of Na+/H+ exchanger (NHE3), an important transporter that mediates intestinal Na+/fluid absorption. CONCLUSIONS: Our findings demonstrate that SARS-CoV-2 enterocyte infection leads to ER stress and the release of DAMPs that up-regulates the expression and release of VIP by EN. VIP in turn inhibits fluid absorption through the downregulation of brush-border membrane expression of NHE3 in enterocytes. These data highlight the role of epithelial-enteric neuronal crosstalk in COVID-19-related diarrhea.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Ratones , Animales , SARS-CoV-2/metabolismo , Intercambiador 3 de Sodio-Hidrógeno , Tunicamicina , Células CACO-2 , Medios de Cultivo Condicionados , Proteómica , Intercambiadores de Sodio-Hidrógeno/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo , Diarrea , Chaperón BiP del Retículo Endoplásmico , Neuronas/metabolismo
3.
Sci Rep ; 12(1): 22076, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36543902

RESUMEN

Enteric neuron degeneration has been observed during aging, and in individuals with metabolic dysfunction including obesity and diabetes. Honokiol, a naturally occurring compound, is an activator of Sirtuin-3 (SIRT3) that has antioxidant activity. Its role in modulating enteric neuron-specific neurodegeneration is unknown. We studied the effects of honokiol and its fluorinated analog, hexafluoro-honokiol, on enteric neuronal differentiation and survival. We used a previously established model of mouse primary enteric neuronal cells and an enteric neuronal cell line treated with palmitate (PA) and lipopolysaccharide (LPS) to induce mitochondrial dysfunction and enteric neuronal cell death. The effect of honokiol and hexafluoro-honokiol was assessed on neuronal phenotype, fiber density, differentiation, and pyroptosis. Honokiol and hexafluoro-honokiol significantly increased neuronal networks and fiber density in enteric neurons and increased levels of neuronal nitric oxide synthase and Choline acetyltransferase mRNA. Hexafluoro-honokiol and honokiol also significantly increased SIRT3 mRNA levels and suppressed palmitate and LPS-induced neuronal pyroptosis. SIRT3 knock-down prevented the hexafluoro-honokiol mediated suppression of mitochondrial superoxide release. Our data supports a neuroprotective effect of honokiol and its derivative and these could be used as prophylactic or therapeutic agents for treating enteric neurodegeneration and associated motility disorders.


Asunto(s)
Sistema Nervioso Entérico , Sirtuina 3 , Animales , Ratones , Compuestos de Bifenilo/farmacología , Compuestos de Bifenilo/uso terapéutico , Diferenciación Celular/genética , Sistema Nervioso Entérico/efectos de los fármacos , Sistema Nervioso Entérico/metabolismo , Lipopolisacáridos/farmacología , Neuronas/metabolismo , Palmitatos/farmacología , Sirtuina 3/genética , Sirtuina 3/metabolismo
4.
Sci Rep ; 12(1): 15838, 2022 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-36151131

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is associated with increased oxidative stress that leads to hepatocyte and mitochondrial damage. In this study we investigated the mechanisms involved in the induction of oxidative stress and impairment of mitochondrial quality control and mitophagy in hepatocytes by the saturated fatty acid palmitate and Western diet feeding in mice and if their harmful effects could be reversed by the neurotrophic factor glial cell derived neurotrophic factor (GDNF). Western diet (WD)-feeding increased hepatic lipid peroxidation in control mice and, in vitro palmitate induced oxidative stress and impaired the mitophagic clearance of damaged mitochondria in hepatocytes. This was accompanied by reductions in hepatocyte sirtuin 3 (SIRT3) deacetylase activity, gene expression and protein levels as well as in superoxide dismutase enzyme activity. These reductions were reversed in the liver of Western diet fed GDNF transgenic mice and in hepatocytes exposed to palmitate in the presence of GDNF. We demonstrate an important role for Western diet and palmitate in inducing oxidative stress and impairing mitophagy in hepatocytes and an ability of GDNF to prevent this. These findings suggest that GDNF or its agonists may be a potential therapy for the prevention or treatment of NAFLD.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial , Enfermedad del Hígado Graso no Alcohólico , Estrés Oxidativo , Sirtuina 3 , Animales , Dieta Alta en Grasa , Dieta Occidental/efectos adversos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Hepatocitos/metabolismo , Ratones , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Palmitatos/efectos adversos , Sirtuina 3/genética , Sirtuina 3/metabolismo , Superóxido Dismutasa/metabolismo
5.
J Clin Invest ; 130(7): 3621-3636, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32484462

RESUMEN

Enteric neuronal degeneration, as seen in inflammatory bowel disease, obesity, and diabetes, can lead to gastrointestinal dysmotility. Pyroptosis is a novel form of programmed cell death but little is known about its role in enteric neuronal degeneration. We observed higher levels of cleaved caspase-1, a marker of pyroptosis, in myenteric ganglia of overweight and obese human subjects compared with normal-weight subjects. Western diet-fed (WD-fed) mice exhibited increased myenteric neuronal pyroptosis, delayed colonic transit, and impaired electric field stimulation-induced colonic relaxation responses. WD increased TLR4 expression and cleaved caspase-1 in myenteric nitrergic neurons. Overactivation of nitrergic neuronal NF-κB signaling resulted in increased pyroptosis and delayed colonic motility. In caspase-11-deficient mice, WD did not induce nitrergic myenteric neuronal pyroptosis and colonic dysmotility. To understand the contributions of saturated fatty acids and bacterial products to the steps leading to enteric neurodegeneration, we performed in vitro experiments using mouse enteric neurons. Palmitate and lipopolysaccharide (LPS) increased nitrergic, but not cholinergic, enteric neuronal pyroptosis. LPS gained entry to the cytosol in the presence of palmitate, activating caspase-11 and gasdermin D, leading to pyroptosis. These results support a role of the caspase-11-mediated pyroptotic pathway in WD-induced myenteric nitrergic neuronal degeneration and colonic dysmotility, providing important therapeutic targets for enteric neuropathy.


Asunto(s)
Caspasas Iniciadoras/metabolismo , Caspasas/metabolismo , Colon , Dieta Occidental/efectos adversos , Sistema Nervioso Entérico , Motilidad Gastrointestinal , Neuronas , Piroptosis , Animales , Caspasas/genética , Caspasas Iniciadoras/genética , Colon/enzimología , Colon/inervación , Colon/patología , Sistema Nervioso Entérico/enzimología , Sistema Nervioso Entérico/patología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Neuronas/enzimología , Neuronas/patología
6.
Front Neurosci ; 14: 614331, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33414704

RESUMEN

Neurodegeneration of the central and enteric nervous systems is a common feature of aging and aging-related diseases, and is accelerated in individuals with metabolic dysfunction including obesity and diabetes. The molecular mechanisms of neurodegeneration in both the CNS and ENS are overlapping. Sirtuins are an important family of histone deacetylases that are important for genome stability, cellular response to stress, and nutrient and hormone sensing. They are activated by calorie restriction (CR) and by the coenzyme, nicotinamide adenine dinucleotide (NAD+). Sirtuins, specifically the nuclear SIRT1 and mitochondrial SIRT3, have been shown to have predominantly neuroprotective roles in the CNS while the cytoplasmic sirtuin, SIRT2 is largely associated with neurodegeneration. A systematic study of sirtuins in the ENS and their effect on enteric neuronal growth and survival has not been conducted. Recent studies, however, also link sirtuins with important hormones such as leptin, ghrelin, melatonin, and serotonin which influence many important processes including satiety, mood, circadian rhythm, and gut homeostasis. In this review, we address emerging roles of sirtuins in modulating the metabolic challenges from aging, obesity, and diabetes that lead to neurodegeneration in the ENS and CNS. We also highlight a novel role for sirtuins along the microbiota-gut-brain axis in modulating neurodegeneration.

7.
Hepatology ; 69(6): 2455-2470, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30715741

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) is a protein that is required for the development and survival of enteric, sympathetic, and catecholaminergic neurons. We previously reported that GDNF is protective against high fat diet (HFD)-induced hepatic steatosis in mice through suppression of hepatic expression of peroxisome proliferator activated receptor-γ and genes encoding enzymes involved in de novo lipogenesis. We also reported that transgenic overexpression of GDNF in mice prevented the HFD-induced liver accumulation of the autophagy cargo-associated protein p62/sequestosome 1 characteristic of impaired autophagy. Here we investigated the effects of GDNF on hepatic autophagy in response to increased fat load, and on hepatocyte mitochondrial fatty acid ß-oxidation and cell survival. GDNF not only prevented the reductions in the liver levels of some key autophagy-related proteins, including Atg5, Atg7, Beclin-1 and LC3A/B-II, seen in HFD-fed control mice, but enhanced their levels after 12 weeks of HFD feeding. In vitro, GDNF accelerated autophagic cargo clearance in primary mouse hepatocytes and a rat hepatocyte cell line, and reduced the phosphorylation of the mechanistic target of rapamycin complex downstream-target p70S6 kinase similar to the autophagy activator rapamycin. GDNF also enhanced mitochondrial fatty acid ß-oxidation in primary mouse and rat hepatocytes, and protected against palmitate-induced lipotoxicity. Conclusion: We demonstrate a role for GDNF in enhancing hepatic autophagy and in potentiating mitochondrial function and fatty acid oxidation. Our studies show that GDNF and its receptor agonists could be useful for enhancing hepatocyte survival and protecting against fatty acid-induced hepatic lipotoxicity.


Asunto(s)
Autofagia/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Hepatocitos/metabolismo , Lipogénesis/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Palmitatos/metabolismo , Animales , Muerte Celular , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Células Hep G2/citología , Células Hep G2/metabolismo , Hepatocitos/citología , Humanos , Lipólisis/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Consumo de Oxígeno/fisiología , Distribución Aleatoria , Ratas , Sensibilidad y Especificidad , Transducción de Señal , Sirolimus/farmacología
8.
J Physiol ; 595(5): 1831-1846, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28000223

RESUMEN

KEY POINTS: A high-fat diet (60% kcal from fat) is associated with motility disorders inducing constipation and loss of nitrergic myenteric neurons in the proximal colon. Gut microbiota dysbiosis, which occurs in response to HFD, contributes to endotoxaemia. High levels of lipopolysaccharide lead to apoptosis in cultured myenteric neurons that express Toll-like receptor 4 (TLR4). Consumption of a Western diet (WD) (35% kcal from fat) for 6 weeks leads to gut microbiota dysbiosis associated with altered bacterial metabolites and increased levels of plasma free fatty acids. These disorders precede the nitrergic myenteric cell loss observed in the proximal colon. Mice lacking TLR4 did not exhibit WD-induced myenteric cell loss and dysmotility. Lipopolysaccharide-induced in vitro enteric neurodegeneration requires the presence of palmitate and may be a result of enhanced NO production. The present study highlights the critical role of plasma saturated free fatty acids that are abundant in the WD with respect to driving enteric neuropathy and colonic dysmotility. ABSTRACT: The consumption of a high-fat diet (HFD) is associated with myenteric neurodegeneration, which in turn is associated with delayed colonic transit and constipation. We examined the hypothesis that an inherent increase in plasma free fatty acids (FFA) in the HFD together with an HFD-induced alteration in gut microbiota contributes to the pathophysiology of these disorders. C57BL/6 mice were fed a Western diet (WD) (35% kcal from fat enriched in palmitate) or a purified regular diet (16.9% kcal from fat) for 3, 6, 9 and 12 weeks. Gut microbiota dysbiosis was investigated by fecal lipopolysaccharide (LPS) measurement and metabolomics (linear trap quadrupole-Fourier transform mass spectrometer) analysis. Plasma FFA and LPS levels were assessed, in addition to colonic and ileal nitrergic myenteric neuron quantifications and motility. Compared to regular diet-fed control mice, WD-fed mice gained significantly more weight without blood glucose alteration. Dysbiosis was exhibited after 6 weeks of feeding, as reflected by increased fecal LPS and bacterial metabolites and concomitant higher plasma FFA. The numbers of nitrergic myenteric neurons were reduced in the proximal colon after 9 and 12 weeks of WD and this was also associated with delayed colonic transit. WD-fed Toll-like receptor 4 (TLR4)-/- mice did not exhibit myenteric cell loss or dysmotility. Finally, LPS (0.5-2 ng·ml-1 ) and palmitate (20 and 30 µm) acted synergistically to induce neuronal cell death in vitro, which was prevented by the nitric oxide synthase inhibitor NG-nitro-l-arginine methyl ester. In conclusion, WD-feeding results in increased levels of FFA and microbiota that, even in absence of hyperglycaemia or overt endotoxaemia, synergistically induce TLR4-mediated neurodegeneration and dysmotility.


Asunto(s)
Colon/fisiología , Dieta Occidental , Receptor Toll-Like 4/fisiología , Tejido Adiposo/metabolismo , Animales , Colon/metabolismo , Colon/microbiología , Citocinas/metabolismo , Ácidos Grasos no Esterificados/sangre , Ácidos Grasos no Esterificados/metabolismo , Heces/química , Femenino , Flagelina/metabolismo , Microbioma Gastrointestinal , Células HEK293 , Humanos , Lipocalina 2/metabolismo , Lipopolisacáridos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/fisiología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
9.
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.

10.
Am J Physiol Gastrointest Liver Physiol ; 310(2): G103-16, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26564715

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) protects against high-fat diet (HFD)-induced hepatic steatosis in mice, however, the mechanisms involved are not known. In this study we investigated the effects of GDNF overexpression and nanoparticle delivery of GDNF in mice on hepatic steatosis and fibrosis and the expression of genes involved in the regulation of hepatic lipid uptake and de novo lipogenesis. Transgenic overexpression of GDNF in liver and other metabolically active tissues was protective against HFD-induced hepatic steatosis. Mice overexpressing GDNF had significantly reduced P62/sequestosome 1 protein levels suggestive of accelerated autophagic clearance. They also had significantly reduced peroxisome proliferator-activated receptor-γ (PPAR-γ) and CD36 gene expression and protein levels, and lower expression of mRNA coding for enzymes involved in de novo lipogenesis. GDNF-loaded nanoparticles were protective against short-term HFD-induced hepatic steatosis and attenuated liver fibrosis in mice with long-standing HFD-induced hepatic steatosis. They also suppressed the liver expression of steatosis-associated genes. In vitro, GDNF suppressed triglyceride accumulation in Hep G2 cells through enhanced p38 mitogen-activated protein kinase-dependent signaling and inhibition of PPAR-γ gene promoter activity. These results show that GDNF acts directly in the liver to protect against HFD-induced cellular stress and that GDNF may have a role in the treatment of nonalcoholic fatty liver disease.


Asunto(s)
Dieta Alta en Grasa , Hígado Graso/metabolismo , Hígado Graso/prevención & control , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Hígado/metabolismo , PPAR gamma/metabolismo , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Hígado Graso/etiología , Hígado Graso/patología , Factor Neurotrófico Derivado de la Línea Celular Glial/administración & dosificación , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Células Hep G2 , Humanos , Hígado/patología , Ratones , Ratones Transgénicos , Nanopartículas/administración & dosificación , Nanopartículas/uso terapéutico , PPAR gamma/genética , Transducción de Señal/fisiología , Triglicéridos/metabolismo
11.
Liver Transpl ; 22(4): 459-67, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26714616

RESUMEN

Moderate macrovesicular steatosis (>30%), which is present in almost 50% of livers considered for transplantation, increases the risk of primary graft dysfunction. Our previously published data showed that glial cell line-derived neurotrophic factor (GDNF) is protective against high-fat diet (HFD)-induced hepatic steatosis in mice. Hence, we hypothesized that perfusion of steatotic livers with GDNF may reduce liver fat content before transplantation. Livers from 8 weeks of regular diet (RD) and of HFD-fed mice were perfused ex vivo for 4 hours with either vehicle, GDNF, or a previously described defatting cocktail. The liver's residual fat was quantified colorimetrically using a triglyceride (TG) assay kit and by Oil Red O (ORO) and Nile red/Hoechst staining. Liver tissue injury was assessed by using a lactate dehydrogenase (LDH) activity assay. In vitro induction of lipolysis in HepG2 cells was assessed by measuring glycerol and free fatty acid release. ORO staining showed significantly more steatosis in livers from HFD-fed mice compared with RD-fed mice (P < 0.001). HFD livers perfused with GDNF had significantly less steatosis than those not perfused (P = 0.001) or perfused with vehicle (P < 0.05). GDNF is equally effective in steatotic liver defatting compared to the defatting cocktail; however, GDNF induces less liver damage than the defatting cocktail. These observations were consistent with data obtained from assessment of liver TG content. Assessment of liver injury revealed significant hepatocyte injury in livers perfused with the control defatting cocktail but no evidence of injury in livers perfused with either GDNF or vehicle. In vitro, GDNF reduced TG accumulation in HepG2 cells and stimulated increased TG lipolysis. In conclusion, GDNF can decrease mice liver fat content to an acceptable range and could be a potential defatting agent before liver transplantation.


Asunto(s)
Hígado Graso/terapia , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Trasplante de Hígado/métodos , Disfunción Primaria del Injerto/prevención & control , Triglicéridos/metabolismo , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Colorimetría , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Hígado Graso/etiología , Factor Neurotrófico Derivado de la Línea Celular Glial/efectos adversos , Supervivencia de Injerto/efectos de los fármacos , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Lipólisis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Perfusión , Ratas , Triglicéridos/análisis
12.
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
13.
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
14.
Transplantation ; 92(7): 745-51, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21869742

RESUMEN

BACKGROUND: Development of pretransplantation islet culture strategies that preserve or enhance ß-cell viability would eliminate the requirement for the large numbers of islets needed to restore insulin independence in type 1 diabetes patients. We investigated whether glial cell line-derived neurotrophic factor (GDNF) could improve human islet survival and posttransplantation function in diabetic mice. METHODS: Human islets were cultured in medium supplemented with or without GDNF (100 ng/mL) and in vitro islet survival and function assessed by analyzing ß-cell apoptosis and glucose stimulated insulin release. In vivo effects of GDNF were assessed in streptozotocin-induced diabetic nude mice transplanted under the kidney capsule with 2000 islet equivalents of human islets precultured in medium supplemented with or without GDNF. RESULTS: In vitro, human islets cultured for 2 to 10 days in medium supplemented with GDNF showed lower ß-cell death, increased Akt phosphorylation, and higher glucose-induced insulin secretion than islets cultured in vehicle. Human islets precultured in medium supplemented with GDNF restored more diabetic mice to normoglycemia and for a longer period after transplantation than islets cultured in vehicle. CONCLUSIONS: Our study shows that GDNF has beneficial effects on human islet survival and could be used to improve islet posttransplantation survival.


Asunto(s)
Diabetes Mellitus Experimental/cirugía , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Supervivencia de Injerto/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Glucosa/farmacología , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Ratones , Ratones Desnudos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estreptozocina/efectos adversos , Trasplante Heterólogo
16.
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
17.
Otolaryngol Head Neck Surg ; 142(6): 879-85, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20493362

RESUMEN

OBJECTIVE: Age-related changes in the larynx lead to significant voice impairment and reduced quality of life. There is a need for aged animal models that have practical generation times to study the fundamental changes and new therapeutics for the aging voice. The senescence accelerated prone mouse strain (SAMP) animals experience rapid aging without any experimental manipulation. The main objective of this study was to demonstrate the use of senescence accelerated mice to study aging in the larynx. STUDY DESIGN: Murine model. SETTING: Department of Animal Resources, Emory University. SUBJECTS AND METHODS: Larynges from five senescence accelerated prone mice, five normal aging senescence resistant mice, and five C57BL/6 mice were harvested and processed for paraffin sections. Histomorphometry was performed for assessment of collagen and hyaluronic acid distribution. In addition, frozen laryngeal tissue was harvested for transcriptional and translational assessment of collagen-1, using real-time polymerase chain reaction with specific primers and Western blots. Myofibroblast assessment was performed by immunostaining for the presence of alpha-smooth muscle actin. RESULTS: The deposition of collagen increased at six months of age in the SAMP vocal fold, and the level of collagen-1 mRNA increased with age. The myofibroblast protein alpha-smooth muscle actin was also found at a higher concentration in the SAMP vocal tissue. In contrast, the levels of hyaluronic acid in the vocal folds of SAMP mice decreased with age when compared to age-matched C57BL/6 mice. CONCLUSION: SAMP mice show accelerated, age-related changes in the vocal fold that were evident at as early as six months of age. The use of senescence accelerated mice offers promise as a model to study age-related laryngeal changes.


Asunto(s)
Envejecimiento/fisiología , Laringe/fisiología , Modelos Animales , Animales , Colágeno/metabolismo , Ácido Hialurónico/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , ARN Mensajero/análisis
18.
Science ; 328(5975): 228-31, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20203013

RESUMEN

Metabolic syndrome is a group of obesity-related metabolic abnormalities that increase an individual's risk of developing type 2 diabetes and cardiovascular disease. Here, we show that mice genetically deficient in Toll-like receptor 5 (TLR5), a component of the innate immune system that is expressed in the gut mucosa and that helps defend against infection, exhibit hyperphagia and develop hallmark features of metabolic syndrome, including hyperlipidemia, hypertension, insulin resistance, and increased adiposity. These metabolic changes correlated with changes in the composition of the gut microbiota, and transfer of the gut microbiota from TLR5-deficient mice to wild-type germ-free mice conferred many features of metabolic syndrome to the recipients. Food restriction prevented obesity, but not insulin resistance, in the TLR5-deficient mice. These results support the emerging view that the gut microbiota contributes to metabolic disease and suggest that malfunction of the innate immune system may promote the development of metabolic syndrome.


Asunto(s)
Fenómenos Fisiológicos Bacterianos , Inmunidad Innata , Intestinos/microbiología , Síndrome Metabólico/etiología , Receptor Toll-Like 5/metabolismo , Animales , Glucemia/análisis , Distribución de la Grasa Corporal , Peso Corporal , Restricción Calórica , Grasas de la Dieta/administración & dosificación , Femenino , Vida Libre de Gérmenes , Hiperfagia/etiología , Resistencia a la Insulina , Mucosa Intestinal/inmunología , Masculino , Síndrome Metabólico/inmunología , Síndrome Metabólico/microbiología , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/inmunología , Obesidad/microbiología , Obesidad/prevención & control , Receptor Toll-Like 5/deficiencia , Receptor Toll-Like 5/genética
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...