Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 151(2): 414-26, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-23063129

RESUMEN

Diabetes, obesity, and cancer affect upward of 15% of the world's population. Interestingly, all three diseases juxtapose dysregulated intracellular signaling with altered metabolic state. Exactly which genetic factors define stable metabolic set points in vivo remains poorly understood. Here, we show that hedgehog signaling rewires cellular metabolism. We identify a cilium-dependent Smo-Ca(2+)-Ampk axis that triggers rapid Warburg-like metabolic reprogramming within minutes of activation and is required for proper metabolic selectivity and flexibility. We show that Smo modulators can uncouple the Smo-Ampk axis from canonical signaling and identify cyclopamine as one of a new class of "selective partial agonists," capable of concomitant inhibition of canonical and activation of noncanonical hedgehog signaling. Intriguingly, activation of the Smo-Ampk axis in vivo drives robust insulin-independent glucose uptake in muscle and brown adipose tissue. These data identify multiple noncanonical endpoints that are pivotal for rational design of hedgehog modulators and provide a new therapeutic avenue for obesity and diabetes.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Glucólisis , Proteínas Hedgehog/metabolismo , Células Musculares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Quinasas de la Proteína-Quinasa Activada por el AMP , Adipocitos/metabolismo , Animales , Línea Celular , Células Cultivadas , Cilios/metabolismo , Diabetes Mellitus/metabolismo , Humanos , Ratones , Neoplasias/metabolismo , Obesidad/metabolismo , Proteínas Quinasas/metabolismo , Receptor Smoothened
2.
Clin Microbiol Rev ; : e0004523, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38940505

RESUMEN

SUMMARYThe gut microbiota is a major factor contributing to the regulation of energy homeostasis and has been linked to both excessive body weight and accumulation of fat mass (i.e., overweight, obesity) or body weight loss, weakness, muscle atrophy, and fat depletion (i.e., cachexia). These syndromes are characterized by multiple metabolic dysfunctions including abnormal regulation of food reward and intake, energy storage, and low-grade inflammation. Given the increasing worldwide prevalence of obesity, cachexia, and associated metabolic disorders, novel therapeutic strategies are needed. Among the different mechanisms explaining how the gut microbiota is capable of influencing host metabolism and energy balance, numerous studies have investigated the complex interactions existing between nutrition, gut microbes, and their metabolites. In this review, we discuss how gut microbes and different microbiota-derived metabolites regulate host metabolism. We describe the role of the gut barrier function in the onset of inflammation in this context. We explore the importance of the gut-to-brain axis in the regulation of energy homeostasis and glucose metabolism but also the key role played by the liver. Finally, we present specific key examples of how using targeted approaches such as prebiotics and probiotics might affect specific metabolites, their signaling pathways, and their interactions with the host and reflect on the challenges to move from bench to bedside.

3.
Cell ; 140(1): 148-60, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-20074523

RESUMEN

Over 1 billion people are estimated to be overweight, placing them at risk for diabetes, cardiovascular disease, and cancer. We performed a systems-level genetic dissection of adiposity regulation using genome-wide RNAi screening in adult Drosophila. As a follow-up, the resulting approximately 500 candidate obesity genes were functionally classified using muscle-, oenocyte-, fat-body-, and neuronal-specific knockdown in vivo and revealed hedgehog signaling as the top-scoring fat-body-specific pathway. To extrapolate these findings into mammals, we generated fat-specific hedgehog-activation mutant mice. Intriguingly, these mice displayed near total loss of white, but not brown, fat compartments. Mechanistically, activation of hedgehog signaling irreversibly blocked differentiation of white adipocytes through direct, coordinate modulation of early adipogenic factors. These findings identify a role for hedgehog signaling in white/brown adipocyte determination and link in vivo RNAi-based scanning of the Drosophila genome to regulation of adipocyte cell fate in mammals.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Obesidad/genética , Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Adipogénesis , Animales , AMP Cíclico/metabolismo , Glucocorticoides/metabolismo , Humanos , Ratones , Ratones Noqueados , Células Musculares/metabolismo , Proteínas Represoras/genética
4.
Int J Food Sci Nutr ; 75(1): 58-69, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37921224

RESUMEN

Gut microbiota is implicated in the control of host physiology by releasing bioactive actors that could exert a direct or indirect effect on tissue. A dysfunction of the gut microbiota to tissue axis could participate in the development of pathological states such as obesity and diabetes. The aim of this study was to identify the metabolic effect of Limosilactobacillus reuteri (known as Lactobacillus reuteri) BIO7251 (L. reuteri BIO7251) isolated from Corsican clementine orange. Body weight gain, adiposity, glucose tolerance, glucose absorption and food intake were measured in mice fed a high-fat diet in response to a preventive oral administration of L. reuteri BIO7251. This strain of bacteria exerts a beneficial effect on body weight gain by decreasing the subcutaneous adipose tissue mass. The treatment with L. reuteri BIO7251 decreases glucose absorption and food intake in obese/diabetic mice. L. reuteri BIO7251 could be tested as new probiotic strain that could manage body weight during obesity.


Asunto(s)
Diabetes Mellitus Experimental , Resistencia a la Insulina , Limosilactobacillus reuteri , Probióticos , Ratones , Animales , Dieta Alta en Grasa/efectos adversos , Ratones Obesos , Obesidad/metabolismo , Peso Corporal , Aumento de Peso , Glucosa/metabolismo , Fenotipo , Tejido Adiposo/metabolismo , Sistema Nervioso/metabolismo
5.
Gut ; 70(6): 1078-1087, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33020209

RESUMEN

OBJECTIVE: The enteric nervous system (ENS) plays a key role in controlling the gut-brain axis under normal and pathological conditions, such as type 2 diabetes. The discovery of intestinal actors, such as enterosynes, able to modulate the ENS-induced duodenal contraction is considered an innovative approach. Among all the intestinal factors, the understanding of the role of gut microbes in controlling glycaemia is still developed. We studied whether the modulation of gut microbiota by prebiotics could permit the identification of novel enterosynes. DESIGN: We measured the effects of prebiotics on the production of bioactive lipids in the intestine and tested the identified lipid on ENS-induced contraction and glucose metabolism. Then, we studied the signalling pathways involved and compared the results obtained in mice to human. RESULTS: We found that modulating the gut microbiota with prebiotics modifies the actions of enteric neurons, thereby controlling duodenal contraction and subsequently attenuating hyperglycaemia in diabetic mice. We discovered that the signalling pathway involved in these effects depends on the synthesis of a bioactive lipid 12-hydroxyeicosatetraenoic acid (12-HETE) and the presence of mu-opioid receptors (MOR) on enteric neurons. Using pharmacological approaches, we demonstrated the key role of the MOR receptors and proliferator-activated receptor γ for the effects of 12-HETE. These findings are supported by human data showing a decreased expression of the proenkephalin and MOR messanger RNAs in the duodenum of patients with diabetic. CONCLUSIONS: Using a prebiotic approach, we identified enkephalin and 12-HETE as new enterosynes with potential real beneficial and safety impact in diabetic human.


Asunto(s)
Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/biosíntesis , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Duodeno/fisiología , Sistema Nervioso Entérico/fisiología , Prebióticos , Receptores Opioides mu/metabolismo , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/farmacología , Adulto , Anciano , Animales , Eje Cerebro-Intestino , Diabetes Mellitus Experimental/fisiopatología , Duodeno/inervación , Encefalinas/genética , Encefalinas/metabolismo , Sistema Nervioso Entérico/efectos de los fármacos , Microbioma Gastrointestinal , Prueba de Tolerancia a la Glucosa , Humanos , Contracción Isotónica/efectos de los fármacos , Masculino , Ratones , Persona de Mediana Edad , Músculo Liso/fisiología , Neuronas/fisiología , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Oligosacáridos/farmacología , PPAR gamma/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores Opioides mu/genética , Transducción de Señal
6.
Gut ; 70(6): 1088-1097, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32978245

RESUMEN

OBJECTIVE: Data from clinical research suggest that certain probiotic bacterial strains have the potential to modulate colonic inflammation. Nonetheless, these data differ between studies due to the probiotic bacterial strains used and the poor knowledge of their mechanisms of action. DESIGN: By mass-spectrometry, we identified and quantified free long chain fatty acids (LCFAs) in probiotics and assessed the effect of one of them in mouse colitis. RESULTS: Among all the LCFAs quantified by mass spectrometry in Escherichia coli Nissle 1917 (EcN), a probiotic used for the treatment of multiple intestinal disorders, the concentration of 3-hydroxyoctadecaenoic acid (C18-3OH) was increased in EcN compared with other E. coli strains tested. Oral administration of C18-3OH decreased colitis induced by dextran sulfate sodium in mice. To determine whether other bacteria composing the microbiota are able to produce C18-3OH, we targeted the gut microbiota of mice with prebiotic fructooligosaccharides (FOS). The anti-inflammatory properties of FOS were associated with an increase in colonic C18-3OH concentration. Microbiota analyses revealed that the concentration of C18-3OH was correlated with an increase in the abundance in Allobaculum, Holdemanella and Parabacteroides. In culture, Holdemanella biformis produced high concentration of C18-3OH. Finally, using TR-FRET binding assay and gene expression analysis, we demonstrated that the C18-3OH is an agonist of peroxisome proliferator activated receptor gamma. CONCLUSION: The production of C18-3OH by bacteria could be one of the mechanisms implicated in the anti-inflammatory properties of probiotics. The production of LCFA-3OH by bacteria could be implicated in the microbiota/host interactions.


Asunto(s)
Colitis/tratamiento farmacológico , Mucosa Intestinal/metabolismo , PPAR gamma/metabolismo , Estearatos/metabolismo , Estearatos/uso terapéutico , Animales , Bacteroidetes , Células CACO-2 , Permeabilidad de la Membrana Celular , Quimiocina CXCL1/genética , Colitis/inducido químicamente , Colitis/metabolismo , Sulfato de Dextran , Células Epiteliales/fisiología , Escherichia coli/metabolismo , Firmicutes/metabolismo , Microbioma Gastrointestinal/fisiología , Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1beta/genética , Espectrometría de Masas , Ratones , Oligosacáridos/farmacología , PPAR gamma/genética , Proteínas Asociadas a Pancreatitis/genética , Permeabilidad , Ganglios Linfáticos Agregados , Prebióticos , Probióticos/química , Estearatos/análisis , Proteína de la Zonula Occludens-1/genética
7.
Neuroendocrinology ; 110(1-2): 139-146, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31280267

RESUMEN

The gut-brain axis is of crucial importance for controlling glucose homeostasis. Alteration of this axis promotes the type 2 diabetes (T2D) phenotype (hyperglycaemia, insulin resistance). Recently, a new concept has emerged to demonstrate the crucial role of the enteric nervous system in the control of glycaemia via the hypothalamus. In diabetic patients and mice, modification of enteric neurons activity in the proximal part of the intestine generates a duodenal hyper-contractility that generates an aberrant message from the gut to the brain. In turn, the hypothalamus sends an aberrant efferent message that provokes a state of insulin resistance, which is characteristic of a T2D state. Targeting the enteric nervous system of the duodenum is now recognized as an innovative strategy for treatment of diabetes. By acting in the intestine, bioactive gut molecules that we called "enterosynes" can modulate the function of a specific type of neurons of the enteric nervous system to decrease the contraction of intestinal smooth muscle cells. Here, we focus on the origins of enterosynes (hormones, neurotransmitters, nutrients, microbiota, and immune factors), which could be considered therapeutic factors, and we describe their modes of action on enteric neurons. This unsuspected action of enterosynes is proposed for the treatment of T2D, but it could be applied for other therapeutic solutions that implicate communication between the gut and brain.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Sistema Nervioso Entérico/efectos de los fármacos , Sistema Nervioso Entérico/fisiología , Obesidad/tratamiento farmacológico , Animales , Humanos
9.
Mol Syst Biol ; 13(3): 921, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28302863

RESUMEN

Gut microbiota dysbiosis has been implicated in a variety of systemic disorders, notably metabolic diseases including obesity and impaired liver function, but the underlying mechanisms are uncertain. To investigate this question, we transferred caecal microbiota from either obese or lean mice to antibiotic-free, conventional wild-type mice. We found that transferring obese-mouse gut microbiota to mice on normal chow (NC) acutely reduces markers of hepatic gluconeogenesis with decreased hepatic PEPCK activity, compared to non-inoculated mice, a phenotypic trait blunted in conventional NOD2 KO mice. Furthermore, transferring of obese-mouse microbiota changes both the gut microbiota and the microbiome of recipient mice. We also found that transferring obese gut microbiota to NC-fed mice then fed with a high-fat diet (HFD) acutely impacts hepatic metabolism and prevents HFD-increased hepatic gluconeogenesis compared to non-inoculated mice. Moreover, the recipient mice exhibit reduced hepatic PEPCK and G6Pase activity, fed glycaemia and adiposity. Conversely, transfer of lean-mouse microbiota does not affect markers of hepatic gluconeogenesis. Our findings provide a new perspective on gut microbiota dysbiosis, potentially useful to better understand the aetiology of metabolic diseases.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/fisiología , Hígado/metabolismo , Obesidad/microbiología , Animales , Disbiosis , Gluconeogénesis , Glucosa-6-Fosfatasa/genética , Ratones , Ratones Endogámicos C57BL , Obesidad/inducido químicamente , Obesidad/genética , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética
10.
Ecotoxicol Environ Saf ; 161: 459-466, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29909315

RESUMEN

Since 2010, Bisphenol A (BPA), an endocrine disruptor has been restricted and replaced by analogues like Bisphenol S (BPS). However, little is known about BPS effects and growing concern have suspected the "BPA-free" Label. Several recent studies suggest that BPS is associated with increased risk of diabetes and obesity. However, the underlying mechanisms remain unidentified. The current study investigates investigate BPS effects on hypothalamic neuropeptides regulating feeding behavior, either orexigenic or anorexigenic in Swiss Albino mice. We also studied the effect of BPS on the apelinergic system (apelin/apelin receptor (APJ)) as an original physiological system with pleiotropic actions. Bisphenol S at 25, 50, 100 µg/kg was administered to mice in water drink for 10 weeks started after weaning. Our results showed that BPS exposure alters orexigenic hypothalamic neuropeptide (AgRP) regulating feeding behavior but not anorexigenic neuropeptides (POMC, CART). Such orexigenic alterations may underlay appetite disorders leading to a concomitant food intake and body weight gain increase. In addition, data show that BPS affects the hypothalamic apelinergic system. We found a significant decrease in APJ mRNA but not in apelin expression. Based on hypothalamic APJ distribution, we suggested a potent specific physiological alteration of this receptor in mediating neuroendocrine responses in hypothalamus. Thus, our findings provide that BPS exposure could contribute to the development of obesity and metabolic disorders.


Asunto(s)
Apelina/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Neuropéptidos/metabolismo , Fenoles/toxicidad , Sulfonas/toxicidad , Animales , Receptores de Apelina/metabolismo , Compuestos de Bencidrilo , Peso Corporal , Conducta Alimentaria/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/genética , Obesidad/inducido químicamente , Proopiomelanocortina/metabolismo , ARN Mensajero/metabolismo , Aumento de Peso
11.
J Am Soc Nephrol ; 28(11): 3205-3217, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28739648

RESUMEN

AKI is a frequent condition that involves renal microcirculation impairment, infiltration of inflammatory cells with local production of proinflammatory cytokines, and subsequent epithelial disorders and mitochondrial dysfunction. Peroxisome proliferator-activated receptor γ coactivator 1-α (PPARGC1A), a coactivator of the transcription factor PPAR-γ that controls mitochondrial biogenesis and function, has a pivotal role in the early dysfunction of the proximal tubule and the subsequent renal repair. Here, we evaluated the potential role of hepatocyte nuclear factor-1ß (HNF-1ß) in regulating PPARGC1A expression in AKI. In mice, endotoxin injection to induce AKI also induced early and transient inflammation and PPARGC1A inhibition, which overlapped with downregulation of the HNF-1ß transcriptional network. In vitro, exposure of proximal tubule cells to the inflammatory cytokines IFN-γ and TNF-α led to inhibition of HNF-1ß transcriptional activity. Moreover, inhibition of HNF-1ß significantly reduced PPARGC1A expression and altered mitochondrial morphology and respiration in proximal tubule cells. Chromatin immunoprecipitation assays and PCR analysis confirmed HNF-1ß binding to the Ppargc1a promoter in mouse kidneys. We also demonstrated downregulation of renal PPARGC1A expression in a patient with an HNF1B germinal mutation. Thus, we propose that HNF-1ß links extracellular inflammatory signals to mitochondrial dysfunction during AKI partly via PPARGC1A signaling. Our findings further strengthen the view of HNF1B-related nephropathy as a mitochondrial disorder in adulthood.


Asunto(s)
Lesión Renal Aguda/metabolismo , Factor Nuclear 1-beta del Hepatocito/fisiología , Túbulos Renales Proximales/metabolismo , Mitocondrias/metabolismo , Lesión Renal Aguda/etiología , Adulto , Animales , Factor Nuclear 1-beta del Hepatocito/antagonistas & inhibidores , Factor Nuclear 1-beta del Hepatocito/genética , Humanos , Ratones Endogámicos C57BL , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/fisiología
12.
Gut ; 66(2): 258-269, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-26565000

RESUMEN

OBJECTIVE: The gut-brain axis is considered as a major regulatory checkpoint in the control of glucose homeostasis. The detection of nutrients and/or hormones in the duodenum informs the hypothalamus of the host's nutritional state. This process may occur via hypothalamic neurons modulating central release of nitric oxide (NO), which in turn controls glucose entry into tissues. The enteric nervous system (ENS) modulates intestinal contractions in response to various stimuli, but the importance of this interaction in the control of glucose homeostasis via the brain is unknown. We studied whether apelin, a bioactive peptide present in the gut, regulates ENS-evoked contractions, thereby identifying a new physiological partner in the control of glucose utilisation via the hypothalamus. DESIGN: We measured the effect of apelin on electrical and mechanical duodenal responses via telemetry probes and isotonic sensors in normal and obese/diabetic mice. Changes in hypothalamic NO release, in response to duodenal contraction modulated by apelin, were evaluated in real time with specific amperometric probes. Glucose utilisation in tissues was measured with orally administrated radiolabeled glucose. RESULTS: In normal and obese/diabetic mice, glucose utilisation is improved by the decrease of ENS/contraction activities in response to apelin, which generates an increase in hypothalamic NO release. As a consequence, glucose entry is significantly increased in the muscle. CONCLUSIONS: Here, we identify a novel mode of communication between the intestine and the hypothalamus that controls glucose utilisation. Moreover, our data identified oral apelin administration as a novel potential target to treat metabolic disorders.


Asunto(s)
Adipoquinas/farmacología , Sistema Nervioso Entérico/efectos de los fármacos , Glucosa/metabolismo , Hipotálamo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Contracción Muscular/efectos de los fármacos , Animales , Apelina , Técnicas Biosensibles , Diabetes Mellitus/fisiopatología , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Sistema Nervioso Entérico/fisiología , Motilidad Gastrointestinal/efectos de los fármacos , Homeostasis , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso/fisiología , Óxido Nítrico/metabolismo , Obesidad/fisiopatología , Telemetría
13.
Am J Physiol Gastrointest Liver Physiol ; 310(9): G645-58, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26939867

RESUMEN

The gastrointestinal tract is an important interface of exchange between ingested food and the body. Glucose is one of the major dietary sources of energy. All along the gastrointestinal tube, e.g., the oral cavity, small intestine, pancreas, and portal vein, specialized cells referred to as glucosensors detect variations in glucose levels. In response to this glucose detection, these cells send hormonal and neuronal messages to tissues involved in glucose metabolism to regulate glycemia. The gastrointestinal tract continuously communicates with the brain, especially with the hypothalamus, via the gut-brain axis. It is now well established that the cross talk between the gut and the brain is of crucial importance in the control of glucose homeostasis. In addition to receiving glucosensing information from the gut, the hypothalamus may also directly sense glucose. Indeed, the hypothalamus contains glucose-sensitive cells that regulate glucose homeostasis by sending signals to peripheral tissues via the autonomous nervous system. This review summarizes the mechanisms by which glucosensors along the gastrointestinal tract detect glucose, as well as the results of such detection in the whole body, including the hypothalamus. We also highlight how disturbances in the glucosensing process may lead to metabolic disorders such as type 2 diabetes. A better understanding of the pathways regulating glucose homeostasis will further facilitate the development of novel therapeutic strategies for the treatment of metabolic diseases.


Asunto(s)
Tracto Gastrointestinal/metabolismo , Glucosa/metabolismo , Homeostasis , Hiperglucemia/metabolismo , Animales , Tracto Gastrointestinal/fisiología , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiología
14.
Med Sci (Paris) ; 31(3): 275-81, 2015 Mar.
Artículo en Francés | MEDLINE | ID: mdl-25855281

RESUMEN

The APJ receptor cloned in 1993 found its ligand in 1998 with the discovery of apelin. The presence of APJ in the central nervous system (more particularly in the hypothalamus) and in various tissues (heart, blood vessels, stomach, etc.) makes it a potential pharmacological target. Interest in APJ has allowed the development of peptidic molecules able to stimulate and/or inhibit the receptor and, more recently, to discover another endogenous ligand: apela. Among the functions regulated by the APJ/apelin system, the control of energy metabolism appears today in the forefront. A better understanding of the pharmacology of APJ receptor should allow innovative therapeutic approaches in the treatment of metabolic diseases.


Asunto(s)
Diabetes Mellitus/terapia , Terapia Molecular Dirigida , Receptores Acoplados a Proteínas G/fisiología , Animales , Apelina , Receptores de Apelina , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Ratones , Obesidad/genética , Obesidad/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal/fisiología
15.
Heliyon ; 10(7): e28320, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38586362

RESUMEN

Background and objective: The leaky gut syndrome is characterized by an intestinal hyperpermeability observed in multiple chronic disorders. Alterations of the gut barrier are associated with translocation of bacterial components increasing inflammation, oxidative stress and eventually dysfunctions of cellular interactions at the origin pathologies. Therapeutic and/or preventive approaches have to focus on the identification of novel targets to improve gut homeostasis. In this context, this study aims to identify the role of PERMEAPROTECT + TOLERANE©, known as PERMEA, a food complement composed of a combination of factors (including l-Glutamine) known to improve gut physiology. Methods: We tested the effects of PERMEA or l-Glutamine alone (as reference) on gut permeability (FITC dextran method, expression of tight junctions) and its inflammatory/oxidative consequences (cytokines and redox assays, RT-qPCR) in a co-culture of human cells (peripheral blood mononuclear cells and intestinal epithelial cells) challenged with TNFα. Results: PERMEA prevented intestinal hyperpermeability induced by inflammation. This was linked with its antioxidant and immunomodulatory properties showing a better efficacity than l-Glutamine alone on several parameters including permeability, global antioxidant charge and production of cytokines. Conclusion: PERMEA is more efficient to restore intestinal physiology, reinforcing the concept that combination of food constituents could be used to prevent the development of numerous diseases.

16.
Life Sci ; 351: 122793, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38848938

RESUMEN

The enteric nervous system (ENS) regulates numerous functional and immunological attributes of the gastrointestinal tract. Alterations in ENS cell function have been linked to intestinal outcomes in various metabolic, intestinal, and neurological disorders. Chronic kidney disease (CKD) is associated with a challenging intestinal environment due to gut dysbiosis, which further affects patient quality of life. Although the gut-related repercussions of CKD have been thoroughly investigated, the involvement of the ENS in this puzzle remains unclear. ENS cell dysfunction, such as glial reactivity and alterations in cholinergic signaling in the small intestine and colon, in CKD are associated with a wide range of intestinal pathways and responses in affected patients. This review discusses how the ENS is affected in CKD and how it is involved in gut-related outcomes, including intestinal permeability, inflammation, oxidative stress, and dysmotility.


Asunto(s)
Sistema Nervioso Entérico , Insuficiencia Renal Crónica , Humanos , Sistema Nervioso Entérico/fisiopatología , Insuficiencia Renal Crónica/fisiopatología , Insuficiencia Renal Crónica/metabolismo , Animales , Riñón/fisiopatología , Microbioma Gastrointestinal , Estrés Oxidativo , Disbiosis/complicaciones , Tracto Gastrointestinal/fisiopatología , Tracto Gastrointestinal/metabolismo , Inflamación
17.
Nat Aging ; 4(1): 80-94, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38238601

RESUMEN

Skeletal muscle plays a central role in the regulation of systemic metabolism during lifespan. With aging, this function is perturbed, initiating multiple chronic diseases. Our knowledge of mechanisms responsible for this decline is limited. Glycerophosphocholine phosphodiesterase 1 (Gpcpd1) is a highly abundant muscle enzyme that hydrolyzes glycerophosphocholine (GPC). The physiological functions of Gpcpd1 remain largely unknown. Here we show, in mice, that the Gpcpd1-GPC metabolic pathway is perturbed in aged muscles. Further, muscle-specific, but not liver- or fat-specific, inactivation of Gpcpd1 resulted in severely impaired glucose metabolism. Western-type diets markedly worsened this condition. Mechanistically, Gpcpd1 muscle deficiency resulted in accumulation of GPC, causing an 'aged-like' transcriptomic signature and impaired insulin signaling in young Gpcpd1-deficient muscles. Finally, we report that the muscle GPC levels are markedly altered in both aged humans and patients with type 2 diabetes, displaying a high positive correlation between GPC levels and chronological age. Our findings reveal that the muscle GPCPD1-GPC metabolic pathway has an important role in the regulation of glucose homeostasis and that it is impaired during aging, which may contribute to glucose intolerance in aging.


Asunto(s)
Diabetes Mellitus Tipo 2 , Glucosa , Glicerilfosforilcolina , Fosfolipasas , Anciano , Animales , Humanos , Ratones , Envejecimiento/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Redes y Vías Metabólicas , Músculo Esquelético/metabolismo , Fosfolipasas/metabolismo , Glicerilfosforilcolina/metabolismo
18.
J Neurosci ; 32(35): 11970-9, 2012 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-22933782

RESUMEN

Hormones such as leptin and ghrelin can rapidly rewire hypothalamic feeding circuits when injected into rodent brains. These experimental manipulations suggest that the hypothalamus might reorganize continually in adulthood to integrate the metabolic status of the whole body. In this study, we examined whether hypothalamic plasticity occurs in naive animals according to their nutritional conditions. For this purpose, we fed mice with a short-term high-fat diet (HFD) and assessed brain remodeling through its molecular and functional signature. We found that HFD for 3 d rewired the hypothalamic arcuate nucleus, increasing the anorexigenic tone due to activated pro-opiomelanocortin (POMC) neurons. We identified the polysialic acid molecule (PSA) as a mediator of the diet-induced rewiring of arcuate POMC. Moreover, local pharmacological inhibition and genetic disruption of the PSA signaling limits the behavioral and metabolic adaptation to HFD, as treated mice failed to normalize energy intake and showed increased body weight gain after the HFD challenge. Altogether, these findings reveal the existence of physiological hypothalamic rewiring involved in the homeostatic response to dietary fat. Furthermore, defects in the hypothalamic plasticity-driven adaptive response to HFD are obesogenic and could be involved in the development of metabolic diseases.


Asunto(s)
Adaptación Fisiológica/fisiología , Núcleo Arqueado del Hipotálamo/fisiología , Grasas de la Dieta/administración & dosificación , Proopiomelanocortina/fisiología , Ácidos Siálicos/fisiología , Animales , Ingestión de Energía/genética , Metabolismo Energético/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Red Nerviosa/fisiología , Plasticidad Neuronal/genética , Técnicas de Cultivo de Órganos , Proopiomelanocortina/metabolismo , Sialiltransferasas/deficiencia , Sialiltransferasas/genética , Transducción de Señal/genética , Aumento de Peso/genética
19.
Heliyon ; 9(7): e18196, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37501991

RESUMEN

Background and objective: Pasteurized Akkermansia muciniphila cells have shown anti-diabetic effects in rodents and human. Although, its primary site of action consists in maintaining the gut barrier function, there are no study exploring if A. muciniphila controls glycemia via a gut to brain axis. Targeting the gut motility represents an alternative pathway to treat hyperglycemia. Here, we tested the impact of pasteurized A. muciniphila on gut motility, gut-brain axis and glucose metabolism. Methods: We used mice fed a 45% high-fat (HFD) treated or not with pasteurized A. muciniphila MucT during 12 weeks. We measured the effects of the treatment on body weight gain, glucose metabolism (insulin, glycemia, glucose tolerance), gut contraction and enteric neurotransmitter release, and hypothalamic nitric oxide (NO) release. Results: We show that pasteurized A. muciniphila exerts positive effects on different metabolic parameters such as body weight, fat mass, insulin, glycemia and glucose tolerance. This could be explained by the ability of pasteurized A. muciniphila supplementation to decrease duodenal contraction and to increase hypothalamic NO release in HFD mice. Conclusion: We demonstrate a novel mode of action of pasteurized A. muciniphila explaining its beneficial impact on the control of glycemia in a preclinical model of type 2 diabetes via gut-brain axis signaling.

20.
Nutrients ; 15(18)2023 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-37764826

RESUMEN

BACKGROUND: Human milk banks (HMBs) provide sterilized donor milk (DM) for the feeding of preterm infants. Most HMBs use the standard method of Holder pasteurization (HoP) performed by heating DM at 62.5 °C for 30 min. High hydrostatic pressure (HHP) processing has been proposed as an alternative to HoP. This study aims to evaluate intestinal barrier integrity and microbiota composition in adult mice subjected to a chronic oral administration of HoP- or HHP-DM. METHODS: Mice were treated by daily gavages with HoP- or HHP-DM over seven days. Intestinal barrier integrity was assessed through in vivo 4 kDa FITC-dextran permeability assay and mRNA expression of several tight junctions and mucins in ileum and colon. Cecal short chain fatty acids (SCFAs) and microbiota were analyzed. RESULTS: HHP-DM mice displayed decreased intestinal permeability to FITC-dextran and increased ileal mRNA expression levels of two tight junctions (Ocln and Cdh1) and Muc2. In the colon, mRNA expression levels of two tight junctions (Cdh1 and Tjp1) and of two mucins (Muc2 and Muc4) were decreased in HHP-DM mice. Cecal SCFAs and microbiota were not different between groups. CONCLUSIONS: HHP processing of DM reinforces intestinal barrier integrity in vivo without affecting gut microbiota and SCFAs production. This study reinforces previous findings showing that DM sterilization through HHP might be beneficial for the intestinal maturation of preterm infants compared with the use of HoP for the treatment of DM.


Asunto(s)
Pasteurización , Recién Nacido , Adulto , Lactante , Humanos , Animales , Ratones , Leche Humana , Presión Hidrostática , Recien Nacido Prematuro , Esterilización , ARN Mensajero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA