Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38861671

RESUMEN

Ferroportin (Fpn) is the only iron exporter, playing a crucial role in systemic iron homeostasis. Fpn is negatively regulated by its ligand hepcidin, but other potential regulators in physiological and disease conditions remain poorly understood. Diabetes is a metabolic disorder that develops body iron loading with unknown mechanisms. By utilizing diabetic mouse models and human duodenal specimens, we demonstrated that intestinal Fpn expression was increased in diabetes in a hepcidin-independent manner. Protein kinase C (PKC) is hyperactivated in diabetes. We showed that PKC was required to sustain baseline Fpn expression and diabetes induced Fpn upregulation in the enterocytes and macrophages. Knockout of PKC abolished diabetes associated iron overload. Mechanistically, activation of PKC increased the exocytotic while decreased the endocytic trafficking of Fpn in the resting state. Hyperactive PKC also suppressed hepcidin-induced ubiquitination, internalization, and degradation of Fpn. We further observed that iron loading in the enterocytes and macrophages activated PKC, acting as a novel mechanism to enhance Fpn-dependent iron efflux. Finally, we demonstrated that the loss-of-function of PKC and pharmacological inhibition of PKC significantly alleviated hereditary hemochromatosis associated iron overload. Our study has highlighted, for the first time, that PKC is an important positive regulator of Fpn and a new target in the control of iron homeostasis.

2.
Am J Physiol Endocrinol Metab ; 325(3): E214-E226, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37467022

RESUMEN

Gastrointestinal (GI) complications, including diarrhea, constipation, and gastroparesis, are common in patients with diabetes. Dysregulation of the Na+/H+ exchanger NHE3 in the intestine is linked to diarrhea and constipation, and recent studies showed that NHE3 expression is reduced in type 1 diabetes and metformin causes diarrhea in the db/db mouse model of type 2 diabetes (T2D) via inhibition of NHE3. In this study, we investigated whether NHE3 expression is altered in type 2 diabetic intestine and the underlying mechanism that dysregulates NHE3. NHE3 expression in the brush border membrane (BBM) of the intestine of diabetic mice and humans was decreased. Protein kinase C (PKC) activation is associated with pathologies of diabetes, and immunofluorescence (IF) analysis revealed increased BBM PKCα abundance. Inhibition of PKCα increased NHE3 BBM abundance and NHE3-mediated intestinal fluid absorption in db/db mice. Previous studies have shown that Lactobacillus acidophilus (LA) stimulates intestinal ion transporters. LA increased NHE3 BBM expression and mitigated metformin-mediated inhibition of NHE3 in vitro and in vivo. To understand the underlying mechanism of LA-mediated stimulation of NHE3, we used Caco-2bbe cells overexpressing PKCα that mimic the elevated state of PKCα in T2D. LA diminished PKCα BBM expression, increased phosphorylation of ezrin, and the interaction of NHE3 with NHE regulatory factor 2 (NHERF2). In addition, inhibition of PKCι blocked phosphorylation of ezrin and activation of NHE3 by LA. These findings demonstrate that NHE3 is downregulated in T2D, and LA restores NHE3 expression via regulation of PKCα, PKCι, and ezrin.NEW & NOTEWORTHY We used mouse models of type 2 diabetes (T2D) and human patient-derived samples to show that Na+/H+ exchanger 3 (NHE3) expression is decreased in T2D. We show that protein kinase C-α (PKCα) is activated in diabetes and inhibition of PKCα increased NHE3 expression and mitigates diarrhea. We show that Lactobacillus acidophilus (LA) stimulates NHE3 via inhibition of PKCα and phosphorylation of ezrin.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Metformina , Animales , Humanos , Ratones , Estreñimiento , Diarrea/metabolismo , Lactobacillus acidophilus/metabolismo , Metformina/farmacología , Proteína Quinasa C-alfa/metabolismo , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/metabolismo
3.
Am J Physiol Gastrointest Liver Physiol ; 322(3): G368-G382, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35084215

RESUMEN

Enteric neuronal cells play a vital role in gut motility in humans and experimental rodent models. Patients with diabetes are more vulnerable to gastrointestinal dysfunction due to enteric neuronal degeneration. In this study, we examined the mechanistic role and regulation of nuclear factor-erythroid 2-related factor 2 (Nrf2) in hyperglycemia-induced enteric neuronal cell apoptosis in vitro by using adult mouse primary enteric neuronal crest cells (pENCs). Our data show that hyperglycemia (HG) or inhibition of Nrf2 induces apoptosis by elevating proinflammatory cytokines, reactive oxygen species (ROS) and suppresses neuronal nitric oxide synthase (nNOS-α) via PI3K/Nrf2-mediated signaling. Conversely, treating pENCs with cinnamaldehyde (CNM), a naturally occurring Nrf2 activator, prevented HG-induced apoptosis. These novel data reveal a negative feedback mechanism for GSK-3 activation. To further demonstrate that loss of Nrf2 leads to inflammation, oxidative stress, and reduces nNOS-mediated gastric function, we have used streptozotocin (STZ)-induced diabetic and Nrf2 null female mice. In vivo activation of Nrf2 with CNM (50 mg/kg, 3 days a week, ip) attenuated impaired nitrergic relaxation and delayed gastric emptying (GE) in conventional type 1 diabetic but not in Nrf2 null female mice. Supplementation of CNM normalized diabetes-induced altered gastric antrum protein expression of 1) p-AKT/p-p38MAPK/p-GSK-3ß, 2) BH4 (cofactor of nNOS) biosynthesis enzyme GCH-1, 3) nNOSα, 4) TLR4, NF-κB, and 5) inflammatory cytokines (TNF-α, IL-1ß, IL-6). We conclude that activation of Nrf2 prevents hyperglycemia-induced apoptosis in pENCs and restores nitrergic-mediated gastric motility and GE in STZ-induced diabetes female mice.NEW & NOTEWORTHY Primary neuronal cell crust (pENCs) in the intestine habitats nNOS and Nrf2, which was suppressed in diabetic gastroparesis. Activation of Nrf2 restored nNOS by suppressing inflammatory markers in pENCs cells. Inhibition of Nrf2 reveals a negative feedback mechanism for the activation of GSK-3. Activation of Nrf2 alleviates STZ-induced delayed gastric emptying and nitrergic relaxation in female mice. Activation of Nrf2 restored impaired gastric BH4 biosynthesis enzyme GCH-1, nNOSα expression thus regulating nitric oxide levels.


Asunto(s)
Diabetes Mellitus Experimental , Gastroparesia , Animales , Citocinas , Diabetes Mellitus Experimental/complicaciones , Femenino , Glucógeno Sintasa Quinasa 3 , Glucógeno Sintasa Quinasa 3 beta , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Antro Pilórico
4.
Nature ; 519(7541): 92-6, 2015 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-25731162

RESUMEN

The intestinal tract is inhabited by a large and diverse community of microbes collectively referred to as the gut microbiota. While the gut microbiota provides important benefits to its host, especially in metabolism and immune development, disturbance of the microbiota-host relationship is associated with numerous chronic inflammatory diseases, including inflammatory bowel disease and the group of obesity-associated diseases collectively referred to as metabolic syndrome. A primary means by which the intestine is protected from its microbiota is via multi-layered mucus structures that cover the intestinal surface, thereby allowing the vast majority of gut bacteria to be kept at a safe distance from epithelial cells that line the intestine. Thus, agents that disrupt mucus-bacterial interactions might have the potential to promote diseases associated with gut inflammation. Consequently, it has been hypothesized that emulsifiers, detergent-like molecules that are a ubiquitous component of processed foods and that can increase bacterial translocation across epithelia in vitro, might be promoting the increase in inflammatory bowel disease observed since the mid-twentieth century. Here we report that, in mice, relatively low concentrations of two commonly used emulsifiers, namely carboxymethylcellulose and polysorbate-80, induced low-grade inflammation and obesity/metabolic syndrome in wild-type hosts and promoted robust colitis in mice predisposed to this disorder. Emulsifier-induced metabolic syndrome was associated with microbiota encroachment, altered species composition and increased pro-inflammatory potential. Use of germ-free mice and faecal transplants indicated that such changes in microbiota were necessary and sufficient for both low-grade inflammation and metabolic syndrome. These results support the emerging concept that perturbed host-microbiota interactions resulting in low-grade inflammation can promote adiposity and its associated metabolic effects. Moreover, they suggest that the broad use of emulsifying agents might be contributing to an increased societal incidence of obesity/metabolic syndrome and other chronic inflammatory diseases.


Asunto(s)
Colitis/inducido químicamente , Colitis/microbiología , Dieta/efectos adversos , Emulsionantes/efectos adversos , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/microbiología , Adiposidad/efectos de los fármacos , Animales , Carboximetilcelulosa de Sodio/administración & dosificación , Carboximetilcelulosa de Sodio/efectos adversos , Colitis/patología , Emulsionantes/administración & dosificación , Heces/microbiología , Femenino , Tracto Gastrointestinal/patología , Vida Libre de Gérmenes , Inflamación/inducido químicamente , Inflamación/microbiología , Inflamación/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Síndrome Metabólico/patología , Ratones , Microbiota/efectos de los fármacos , Obesidad/inducido químicamente , Obesidad/microbiología , Obesidad/patología , Polisorbatos/administración & dosificación , Polisorbatos/efectos adversos
5.
Surg Endosc ; 35(8): 4418-4426, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32880014

RESUMEN

BACKGROUND: Esophagogastric junction obstruction (EGJO) post-fundoplication (PF) is difficult to identify with currently available tests. We aimed to assess the diagnostic accuracy of EGJ opening on functional lumen imaging probe (FLIP) and dilation outcome in FLIP-detected EGJO in PF dysphagia. METHODS: We prospectively collected data on PF patients referred to Esophageal Clinic over 18 months. EGJO diagnosis was made by (a) endoscopist's description of a narrow EGJ/wrap area, (b) appearance of wrap obstruction or contrast/tablet retention on esophagram, or (c) EGJ-distensibility index (DI) < 2.8 mm2/mmHg on real-time FLIP. In patients with EGJO and dysphagia, EGJ dilation was performed to 20 mm, 30 mm, or 35 mm in a stepwise fashion. Outcome was assessed as % dysphagia improvement during phone call or on brief esophageal dysphagia questionnaire (BEDQ) score. RESULTS: Twenty-six patients were included, of whom 17 (65%) had a low EGJ-DI. No patients had a hiatal hernia greater than 3 cm. Dysphagia was the primary symptom in 17/26 (65%). In 85% (κ = 0.677) of cases, EGJ assessment (tight vs. open) was congruent between the combination of endoscopy (n = 26) and esophagram (n = 21) vs. EGJ-DI (n = 26) on FLIP. Follow-up data were available in 11 patients who had dilation based on a low EGJ-DI (4 with 20 mm balloon and 7 with ≥ 30 mm balloon). Overall, the mean % improvement in dysphagia was 60% (95% CI 37.7-82.3%, p = 0.0001). Nine out of 11 patients, including 6 out of 7 undergoing pneumatic dilation, had improvement ≥ 50% in dysphagia (mean % improvement 72.2%; 95% CI 56.1-88.4%, p = 0.0001). CONCLUSIONS AND INFERENCES: Functional lumen imaging probe is an accurate modality for evaluating for EGJ obstruction PF. FLIP may be used to select patients who may benefit from larger diameter dilation.


Asunto(s)
Trastornos de Deglución , Acalasia del Esófago , Trastornos de Deglución/etiología , Unión Esofagogástrica/diagnóstico por imagen , Fundoplicación , Humanos , Manometría
6.
Am J Physiol Gastrointest Liver Physiol ; 319(4): G481-G493, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32812777

RESUMEN

Diabetic gastroparesis (DG) is a clinical syndrome characterized by delayed gastric emptying (DGE). Loss of nuclear factor erythroid 2-related factor 2 (Nrf2) is associated with reduced neuronal nitric oxide synthase-α (nNOSα)-mediated gastric motility and DGE. Previous studies have shown that nuclear exclusion and inactivation of Nrf2 is partly regulated by glycogen synthase kinase 3ß (GSK-3ß). In the current study, the molecular signaling of GSK-3ß-mediated Nrf2 activation and its mechanistic role on DG were investigated in high-fat diet (HFD)-induced obese/Type 2 diabetes (T2D) female mice. Adult female C57BL/6J mice were fed with HFD or normal diet (ND) with or without GSK-3ß inhibitor (SB 216763, 10 mg/kg body wt ip) start from the 14th wk and continued feeding mice for an additional 3-wk time period. Our results show that treatment with GSK-3ß inhibitor SB attenuated DGE in obese/T2D mice. Treatment with SB restored impaired gastric 1) Nrf2 and phase II antioxidant enzymes through PI3K/ERK/AKT-mediated pathway, 2) tetrahydrobiopterin (BH4, cofactor of nNOS) biosynthesis enzyme dihydrofolate reductase, and 3) nNOSα dimerization in obese/T2 diabetic female mice. SB treatment normalized caspase 3 activity and downstream GSK-3ß signaling in the gastric tissues of the obese/T2 diabetic female mice. In addition, GSK-3ß inhibitor restored impaired nitrergic relaxation in hyperglycemic conditions. Finally, SB treatment reduced GSK3 marker, pTau in adult primary enteric neuronal cells. These findings emphasize the importance of GSK-3ß on regulating gastric Nrf2 and nitrergic mediated gastric emptying in obese/diabetic rodents.NEW & NOTEWORTHY Inhibition of glycogen synthase kinase 3ß (GSK-3ß) with SB 216763 attenuates delayed gastric emptying through gastric nuclear factor erythroid 2-related factor 2 (Nrf2)-phase II enzymes in high-fat diet-fed female mice. SB 216763 restored impaired gastric PI3K/AKT/ ß-catenin/caspase 3 expression. Inhibition of GSK-3ß normalized gastric dihydrofolate reductase, neuronal nitric oxide synthase-α expression, dimerization and nitrergic relaxation. SB 216763 normalized both serum estrogen and nitrate levels in female obese/Type 2 diabetes mice. SB 216763 reduced downstream signaling of GSK-3ß in enteric neuronal cells in vitro.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Vaciamiento Gástrico/efectos de los fármacos , Gastroparesia/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Indoles/farmacología , Maleimidas/farmacología , Obesidad/complicaciones , Animales , Antioxidantes/fisiología , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Tipo 2/complicaciones , Dieta Alta en Grasa , Femenino , Vaciamiento Gástrico/fisiología , Gastroparesia/etiología , Glucosa/metabolismo , Glucógeno Sintasa Quinasa 3 beta/fisiología , Resistencia a la Insulina/fisiología , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/efectos de los fármacos , Obesidad/etiología
7.
Gastroenterology ; 157(1): 179-192.e2, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30930024

RESUMEN

BACKGROUND & AIMS: Reduced gastrointestinal (GI) motility is a feature of disorders associated with intestinal dysbiosis and loss of beneficial microbes. It is not clear how consumption of beneficial commensal microbes, marketed as probiotics, affects the enteric nervous system (ENS). We studied the effects of the widely used probiotic and the commensal Lactobacillus rhamnosus GG (LGG) on ENS and GI motility in mice. METHODS: Conventional and germ free C57B6 mice were gavaged with LGG and intestinal tissues were collected; changes in the enteric neuronal subtypes were assessed by real-time polymerase chain reaction, immunoblots, and immunostaining. Production of reactive oxygen species (ROS) in the jejunal myenteric plexi and phosphorylation (p) of mitogen-activated protein kinase 1 (MAPK1) in the enteric ganglia were assessed by immunoblots and immunostaining. Fluorescence in situ hybridization was performed on jejunal cryosections with probes to detect formyl peptide receptor 1 (FPR1). GI motility in conventional mice was assessed after daily gavage of LGG for 1 week. RESULTS: Feeding of LGG to mice stimulated myenteric production of ROS, increased levels of phosphorylated MAPK1, and increased expression of choline acetyl transferase by neurons (P < .001). These effects were not observed in mice given N-acetyl cysteine (a ROS inhibitor) or LGGΩSpaC (an adhesion-mutant strain of LGG) or FPR1-knockout mice. Gavage of mice with LGG for 1 week significantly increased stool frequency, reduced total GI transit time, and increased contractions of ileal circular muscle strips in ex vivo experiments (P < .05). CONCLUSIONS: Using mouse models, we found that LGG-mediated signaling in the ENS requires bacterial adhesion, redox mechanisms, and FPR1. This pathway might be activated to increase GI motility in patients.


Asunto(s)
Motilidad Gastrointestinal/fisiología , Tránsito Gastrointestinal/fisiología , Íleon/metabolismo , Yeyuno/metabolismo , Lacticaseibacillus rhamnosus , Plexo Mientérico/metabolismo , Neuronas/metabolismo , Probióticos , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Colina O-Acetiltransferasa/metabolismo , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/metabolismo , Motilidad Gastrointestinal/efectos de los fármacos , Tránsito Gastrointestinal/efectos de los fármacos , Vida Libre de Gérmenes , Íleon/efectos de los fármacos , Íleon/inervación , Hibridación Fluorescente in Situ , Yeyuno/efectos de los fármacos , Yeyuno/inervación , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Contracción Muscular/efectos de los fármacos , Plexo Mientérico/citología , Neuronas/efectos de los fármacos , Fosforilación , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Formil Péptido/genética
8.
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
9.
FASEB J ; 33(3): 3549-3561, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30423260

RESUMEN

Excessive iron increases the incidence of diabetes and worsens diabetic complications. Reciprocally, diabetes induces iron loading, partially attributable to elevated intestinal iron export according to a recent report. Herein, we show that iron uptake and the mRNA expression of iron importer divalent metal transporter 1 (DMT1) were significantly increased in the duodenum of streptozotocin-induced diabetic mice. Immunofluorescence staining of human intestinal biopsies revealed increased brush border membrane (BBM) and decreased cytoplasmic DMT1 expression in patients with diabetes, suggesting translocation of DMT1. This pattern of DMT1 regulation was corroborated by immunoblotting results in diabetic mice showing that BBM DMT1 expression was increased by 210%, in contrast to a 60% increase in total DMT1. PKC mediates many diabetic complications, and PKCα activity was increased in diabetic mouse intestine. Intriguingly, diabetic mice with PKCα deficiency did not show increases in iron uptake and BBM DMT1 expression. High-glucose treatment increased plasma membrane DMT1 expression via the activation of PKCα in cultured IECs. Inhibition of PKCα potentiated the ubiquitination and degradation of DMT1 protein. We further showed that high glucose suppressed membrane DMT1 internalization. These findings demonstrate that PKCα promotes microvillus membrane DMT1 expression and intestinal iron uptake, contributing to diabetic iron loading.-Zhao, L., Bartnikas, T., Chu, X., Klein, J., Yun, C., Srinivasan, S., He, P. Hyperglycemia promotes microvillus membrane expression of DMT1 in intestinal epithelial cells in a PKCα-dependent manner.


Asunto(s)
Duodeno/metabolismo , Células Epiteliales/metabolismo , Hiperglucemia/metabolismo , Microvellosidades/metabolismo , Proteína Quinasa C-alfa/metabolismo , Factores de Transcripción/metabolismo , Adulto , Anciano , Animales , Transporte Biológico/fisiología , Diabetes Mellitus Experimental/metabolismo , Glucosa/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Hierro/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos/metabolismo , Persona de Mediana Edad , Ubiquitinación/fisiología
10.
Reprod Biomed Online ; 40(1): 7-11, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31836436

RESUMEN

Rather than consider endometriosis as an enigmatic disease, reading John Sampson's two theories/mechanisms explains virtually all cases affecting the female. It is true that Sampson's most recent publication, in 1940, which talks about retrograde menstruation via the fallopian tubes, clearly fails to explain many types of endometriosis, particularly that located in extra-pelvic sites. However, his earlier publications of 1911 and 1912, on radiographic studies of hysterectomy specimens that had been injected with various gelatin/bismuth/pigment mixtures examining the unique uterine vasculature, were more important. These studies enabled him to describe 'the escape of foreign material from the uterine cavity into the uterine veins' in 1918 and subsequently to demonstrate metastatic or embolic endometriosis in the first of his two important publications in 1927. Later in that same year, in response to 'academic banter' from other historic gynaecologists, he published a second article that indicated his studies had been redirected to explore the retrograde tubal menstruation idea; this required undertaking his hysterectomies during menses. That work led to his 1940 presentation at the invitation of The American College of Obstetricians and Gynecologists to focus on the second theory/mechanism of endometriosis. This appears to have caused his more important first theory/mechanism to have been forgotten.


Asunto(s)
Endometriosis/etiología , Útero/patología , Endometriosis/patología , Femenino , Humanos
11.
Reprod Biomed Online ; 39(2): 183-186, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31204258

RESUMEN

A recent article supports our longstanding view that all intramural fibroids can cause disturbance of uterine function. This may be reflected in the symptom of menorrhagia or fertility-related issues, as well as pregnancy losses at all gestational stages. However, it was disappointing that there was no reference to either the mechanism by which fibroids disturb uterine function nor to the gynaecologist who described this more than 100 years ago, namely John Sampson. In fact, Sampson's findings about the unique venous drainage mechanism from the endometrium explains how menstrual loss is contained in normal physiology, but which can be excessive when the protective 'anaemic' zone is disturbed. Two more recent and pertinent observations include the hysteroscopic findings of Osamu Sugimoto, who showed in the 1970s that the endometrium overlying submucous fibroids is actually atrophic, hence the oft-cited reason of hyperplastic or excessive endometrium cannot be the cause of the associated menorrhagia. Furthermore, recent imaging techniques describe an additional 'junctional zone' adjacent to the endometrium in cases of fibroids and adenomyosis. We believe this all adds up to disturbed venous drainage as described by Sampson and needs to immediately enter the educational training of medical students, doctors and gynaecologists worldwide.


Asunto(s)
Ginecología/historia , Leiomioma/diagnóstico , Femenino , Historia del Siglo XX , Humanos , Histeroscopía , Leiomioma/historia , Leiomioma/terapia , Enfermedades Uterinas/historia , Útero/irrigación sanguínea , Útero/patología
12.
Curr Gastroenterol Rep ; 21(4): 15, 2019 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-30887257

RESUMEN

PURPOSE OF REVIEW: The goal of the present review is to explore the relationship between dietary changes and alterations in gut microbiota that contribute to disorders of gut motility and obesity. RECENT FINDINGS: We review the microbiota changes that are seen in obesity, diarrhea, and constipation and look at potential mechanisms of how dysbiosis can predispose to these. We find that microbial metabolites, particularly short chain fatty acids, can lead to signaling changes in the host enterocytes. Microbial alteration leading to both motility disorders and obesity may be mediated by the release of hormones including glucagon-like peptides 1 and 2 (GLP-1, GLP-2) and polypeptide YY (PYY). These pathways provide avenues for microbiota-targeted interventions that can treat both disorders of motility and obesity. In summary, multiple mechanisms contribute to the interplay between the microbial dysbiosis, obesity, and dysmotility.


Asunto(s)
Estreñimiento/microbiología , Diarrea/microbiología , Dieta , Microbioma Gastrointestinal , Obesidad/microbiología , Dieta/efectos adversos , Disbiosis/complicaciones , Motilidad Gastrointestinal , Humanos
13.
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
14.
Am J Physiol Gastrointest Liver Physiol ; 312(2): G103-G111, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27856419

RESUMEN

We have demonstrated that neuropeptide Y (NPY), abundantly produced by enteric neurons, is an important regulator of intestinal inflammation. However, the role of NPY in the progression of chronic inflammation to tumorigenesis is unknown. We investigated whether NPY could modulate epithelial cell proliferation and apoptosis, and thus regulate tumorigenesis. Repeated cycles of dextran sodium sulfate (DSS) were used to model inflammation-induced tumorigenesis in wild-type (WT) and NPY knockout (NPY-/-) mice. Intestinal epithelial cell lines (T84) were used to assess the effects of NPY (0.1 µM) on epithelial proliferation and apoptosis in vitro. DSS-WT mice exhibited enhanced intestinal inflammation, polyp size, and polyp number (7.5 ± 0.8) compared with DSS-NPY-/- mice (4 ± 0.5, P < 0.01). Accordingly, DSS-WT mice also showed increased colonic epithelial proliferation (PCNA, Ki67) and reduced apoptosis (TUNEL) compared with DSS-NPY-/- mice. The apoptosis regulating microRNA, miR-375, was significantly downregulated in the colon of DSS-WT (2-fold, P < 0.01) compared with DSS-NPY-/--mice. In vitro studies indicated that NPY promotes cell proliferation (increase in PCNA and ß-catenin, P < 0.05) via phosphatidyl-inositol-3-kinase (PI3-K)-ß-catenin signaling, suppressed miR-375 expression, and reduced apoptosis (increase in phospho-Bad). NPY-treated cells also displayed increased c-Myc and cyclin D1, and reduction in p21 (P < 0.05). Addition of miR-375 inhibitor to cells already treated with NPY did not further enhance the effects induced by NPY alone. Our findings demonstrate a novel regulation of inflammation-induced tumorigenesis by NPY-epithelial cross talk as mediated by activation of PI3-K signaling and downregulation of miR-375. NEW & NOTEWORTHY: Our work exemplifies a novel role of neuropeptide Y (NPY) in regulating inflammation-induced tumorigenesis via two modalities: first by enhanced proliferation (PI3-K/pAkt), and second by downregulation of microRNA-375 (miR-375)-dependent apoptosis in intestinal epithelial cells. Our data establish the existence of a microRNA-mediated cross talk between enteric neurons producing NPY and intestinal epithelial cells, and the potential of neuropeptide-regulated miRNAs as potential therapeutic molecules for the management of inflammation-associated tumors in the gut.


Asunto(s)
Carcinogénesis/metabolismo , Proliferación Celular/fisiología , Neoplasias del Colon/etiología , Células Epiteliales/fisiología , Inflamación/metabolismo , Neuropéptido Y/metabolismo , Animales , Neoplasias del Colon/patología , Células Epiteliales/citología , Femenino , Regulación Neoplásica de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Neuropéptido Y/genética
15.
Am J Gastroenterol ; 112(2): 356-364, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27922028

RESUMEN

OBJECTIVES: Constipation is the most common GI symptom in patients with diabetes mellitus (DM). Importantly, patients with constipation have lower health-related quality of life than those without constipation. Effective therapies for constipation are limited and there is a paucity of data evaluating the treatment of constipation in diabetics. METHODS: Diabetic patients with chronic idiopathic constipation (CIC) as defined by Rome III criteria were recruited from outpatient clinics at a tertiary-care center and a Veterans Administration Hospital. Demographic data, baseline stool patterns, and a constipation-specific quality of life survey (Patient Assessment of Constipation Quality of Life (PAC-QOL)) were obtained. Baseline colonic transit time (CTT) was evaluated utilizing the wireless motility capsule. Patients were randomized in a double-blind fashion to 48 mcg per day lubiprostone or placebo for 8 weeks. The primary end point measured was the difference in number of spontaneous bowel movements (SBMs) per week vs. baseline for each group at each week after initiation of therapy. Secondary end points included changes in CTT after 4 weeks of therapy, PAC-QOL after 8 weeks of therapy, and changes from baseline in associated gastrointestinal (GI) symptoms as well as need for rescue medication at 2, 4, and 8 weeks. RESULTS: Seventy-six patients (mean age, 56.9±9.1 years, 62% females) were randomized. There were no significant differences between the two groups' baseline data or demographics. During the 8-week treatment period, patients in the lubiprostone group experienced an average of 1.83±0.80 (P=0.02) more SBMs per week than those in the placebo group as compared with baseline. The duration of CTT at Week 4 was shorter by an average of 13 h compared with baseline in the lubiprostone group, and was prolonged by an average of 7 h compared with baseline in the placebo group, leading to a treatment effect of 20.3±7.3 h (P=0.006). PAC-QOL improved in both the groups; however, there was no significant difference between the groups. There was no difference in associated GI symptoms and need for rescue medication between the two groups after 8 weeks. There were no serious adverse events reported during the study. CONCLUSIONS: This study suggests that lubiprostone is a safe and effective treatment for increasing weekly SBMs and decreasing CTT in patients with DM and CIC.


Asunto(s)
Agonistas de los Canales de Cloruro/uso terapéutico , Estreñimiento/tratamiento farmacológico , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Tránsito Gastrointestinal , Lubiprostona/uso terapéutico , Anciano , Colon/fisiopatología , Estreñimiento/complicaciones , Estreñimiento/fisiopatología , Defecación , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Calidad de Vida , Encuestas y Cuestionarios , Resultado del Tratamiento
16.
Acta Neuropathol ; 134(2): 281-295, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28620692

RESUMEN

Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) in young adults that has serious negative socioeconomic effects. In addition to symptoms caused by CNS pathology, the majority of MS patients frequently exhibit gastrointestinal dysfunction, which was previously either explained by the presence of spinal cord lesions or not directly linked to the autoimmune etiology of the disease. Here, we studied the enteric nervous system (ENS) in a B cell- and antibody-dependent mouse model of MS by immunohistochemistry and electron microscopy at different stages of the disease. ENS degeneration was evident prior to the development of CNS lesions and the onset of neurological deficits in mice. The pathology was antibody mediated and caused a significant decrease in gastrointestinal motility, which was associated with ENS gliosis and neuronal loss. We identified autoantibodies against four potential target antigens derived from enteric glia and/or neurons by immunoprecipitation and mass spectrometry. Antibodies against three of the target antigens were also present in the plasma of MS patients as confirmed by ELISA. The analysis of human colon resectates provided evidence of gliosis and ENS degeneration in MS patients compared to non-MS controls. For the first time, this study establishes a pathomechanistic link between the well-established autoimmune attack on the CNS and ENS pathology in MS, which might provide a paradigm shift in our current understanding of the immunopathogenesis of the disease with broad diagnostic and therapeutic implications.


Asunto(s)
Autoanticuerpos/sangre , Enfermedades Gastrointestinales/etiología , Esclerosis Múltiple , Animales , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Sistema Nervioso Entérico/metabolismo , Sistema Nervioso Entérico/patología , Sistema Nervioso Entérico/ultraestructura , Femenino , Adyuvante de Freund/toxicidad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Músculo Liso/patología , Músculo Liso/ultraestructura , Proteína Básica de Mielina/inmunología , Proteína Básica de Mielina/metabolismo , Proteína Básica de Mielina/toxicidad , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Plexo Mientérico/patología , Plexo Mientérico/ultraestructura , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/toxicidad , Tubulina (Proteína)/metabolismo
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA