39 results on '"Medina-Gómez G"'
Search Results
2. Cardiotrophin-1 contributes to metabolic adaptations through the regulation of lipid metabolism and to the fasting-induced fatty acid mobilization
- Author
-
Carneros D, Medina-Gómez G, Giralt M, León-Camacho M, Campbell M, Moreno-Aliaga MJ, Villarroya F, and Bustos M
- Subjects
adipose tissue ,lipids ,fatty acid mobilization ,food restriction ,peroxisome proliferator-activated receptors - Abstract
It is becoming clear that several human pathologies are caused by altered metabolic adaptations. During liver development, there are physiological changes, from the predominant utilization of glucose (fetal life) to the use of lipids (postnatal life). Fasting is another physiological stress that elicits well-known metabolic adjustments. We have reported the metabolic properties of cardiotrophin-1 (CT-1), a member of the interleukin-6 family of cytokines. Here, we aimed at analyzing the role of CT-1 in response to these metabolic changes. We used different in vivo models. Furthermore, a differential study was carried out with wild-type and CT-1 null mice in fed (ad libitum) and food-restricted conditions. We demonstrated that Ct-1 is a metabolic gene induced in the liver via PPARa in response to lipids in mice (neonates- and food-restricted adults). We found that Ct-1 mRNA expression in white adipose tissue directly involved PPARa and PPAR?. Finally, the physiological role of CT-1 in fasting is confirmed by the impaired food restriction-induced adipose tissue lipid mobilization in CT-1 null mice. Our findings support a previously unrecognized physiological role of CT-1 in metabolic adaptations, through the regulation of lipid metabolism and contributes to fasting-induced free fatty acid mobilization.
- Published
- 2020
3. Insulin action is severely impaired in adipocytes of apparently healthy overweight and obese subjects
- Author
-
Rydén, M., primary, Petrus, P., additional, Andersson, D. P., additional, Medina‐Gómez, G., additional, Escasany, E., additional, Corrales Cordón, P., additional, Dahlman, I., additional, Kulyté, A., additional, and Arner, P., additional
- Published
- 2019
- Full Text
- View/download PDF
4. Early peroxisome proliferator-activated receptor gamma regulated genes involved in expansion of pancreatic beta cell mass
- Author
-
Vivas Yurena, Martínez-García Cristina, Izquierdo Adriana, Garcia-Garcia Francisco, Callejas Sergio, Velasco Ismael, Campbell Mark, Ros Manuel, Dopazo Ana, Dopazo Joaquin, Vidal-Puig Antonio, and Medina-Gomez Gema
- Subjects
Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background The progression towards type 2 diabetes depends on the allostatic response of pancreatic beta cells to synthesise and secrete enough insulin to compensate for insulin resistance. The endocrine pancreas is a plastic tissue able to expand or regress in response to the requirements imposed by physiological and pathophysiological states associated to insulin resistance such as pregnancy, obesity or ageing, but the mechanisms mediating beta cell mass expansion in these scenarios are not well defined. We have recently shown that ob/ob mice with genetic ablation of PPARγ2, a mouse model known as the POKO mouse failed to expand its beta cell mass. This phenotype contrasted with the appropriate expansion of the beta cell mass observed in their obese littermate ob/ob mice. Thus, comparison of these models islets particularly at early ages could provide some new insights on early PPARγ dependent transcriptional responses involved in the process of beta cell mass expansion Results Here we have investigated PPARγ dependent transcriptional responses occurring during the early stages of beta cell adaptation to insulin resistance in wild type, ob/ob, PPARγ2 KO and POKO mice. We have identified genes known to regulate both the rate of proliferation and the survival signals of beta cells. Moreover we have also identified new pathways induced in ob/ob islets that remained unchanged in POKO islets, suggesting an important role for PPARγ in maintenance/activation of mechanisms essential for the continued function of the beta cell. Conclusions Our data suggest that the expansion of beta cell mass observed in ob/ob islets is associated with the activation of an immune response that fails to occur in POKO islets. We have also indentified other PPARγ dependent differentially regulated pathways including cholesterol biosynthesis, apoptosis through TGF-β signaling and decreased oxidative phosphorylation.
- Published
- 2011
- Full Text
- View/download PDF
5. Constitutive Pleiotrophin Deletion Results in a Phenotype with an Altered Pancreatic Morphology and Function in Old Mice.
- Author
-
Ballesteros-Pla C, Sevillano J, Sánchez-Alonso MG, Limones M, Pita J, Zapatería B, Sanz-Cuadrado MI, Pizarro-Delgado J, Izquierdo-Lahuerta A, Medina-Gómez G, Herradón G, and Ramos-Álvarez MDP
- Subjects
- Animals, Mice, Female, Phenotype, Glucose Transporter Type 2 genetics, Glucose Transporter Type 2 metabolism, Insulin Secretion genetics, Vesicle-Associated Membrane Protein 2 metabolism, Vesicle-Associated Membrane Protein 2 genetics, Synaptosomal-Associated Protein 25 genetics, Synaptosomal-Associated Protein 25 metabolism, Pancreas metabolism, Pancreas pathology, Insulin Resistance genetics, Somatostatin metabolism, Somatostatin genetics, Glucagon metabolism, Glucose metabolism, Aging genetics, Aging metabolism, Gene Deletion, Mice, Inbred C57BL, Cytokines metabolism, Carrier Proteins genetics, Carrier Proteins metabolism, Islets of Langerhans metabolism, Islets of Langerhans pathology, Mice, Knockout, Insulin metabolism, Insulin blood
- Abstract
Pleiotrophin (PTN) is crucial for embryonic development and pancreas organogenesis as it regulates metainflammation, metabolic homeostasis, thermogenesis, and glucose tolerance. Pleiotrophin deletion is associated with a lipodystrophic phenotype in which adipose tissue plasticity is altered in late life. This study explored the impact of pleiotrophin deletion on pancreatic morphology and function in later life. We analyzed glucose tolerance and circulating parameters on female wild-type ( Ptn
+/+ ) and knock-out ( Ptn-/- ) mice. At 9 and 15 months, we conducted morphometric analyses of pancreatic islets and evaluated the levels of insulin, glucagon, somatostatin, glucose transporter 2 (GLUT2), vesicle-associated membrane protein 2 (VAMP2), and synaptosome-associated protein 25 (SNAP25) via immunofluorescence. The effect of PTN on glucose-stimulated insulin secretion (GSIS) was evaluated in INS1E cells and isolated islets. Ptn-/- mice showed hyperinsulinemia, impaired glucose tolerance, and increased homeostatic model assessment for insulin resistance (HOMA-IR) with age. While Ptn+/+ islets enlarge with age, in Ptn-/- mice, the median size decreased, and insulin content increased. Vesicle transport and exocytosis proteins were significantly increased in 9-month-old Ptn-/- islets. Islets from Ptn-/- mice showed impaired GSIS and decreased cell membrane localization of GLUT2 whereas, PTN increased GSIS in INS1E cells. Ptn deletion accelerated age-related changes in the endocrine pancreas, affecting islet number and size , and altering VAMP2 and SNAP25 levels and GLUT2 localization leading to impaired GSIS and insulin accumulation in islets.- Published
- 2024
- Full Text
- View/download PDF
6. Interleukin-16 is increased in obesity and alters adipogenesis and inflammation in vitro .
- Author
-
Reyes-Farias M, Fernández-García P, Corrales P, González L, Soria-Gondek A, Martínez E, Pellitero S, Tarascó J, Moreno P, Sumoy L, Medina-Gómez G, Sánchez-Infantes D, and Herrero L
- Subjects
- Humans, Fibrosis, Inflammation metabolism, Lipids, Obesity metabolism, Adipogenesis, Interleukin-16
- Abstract
Introduction: Obesity is a chronic condition associated with low-grade inflammation mainly due to immune cell infiltration of white adipose tissue (WAT). WAT is distributed into two main depots: subcutaneous WAT (sWAT) and visceral WAT (vWAT), each with different biochemical features and metabolic roles. Proinflammatory cytokines including interleukin (IL)-16 are secreted by both adipocytes and infiltrated immune cells to upregulate inflammation. IL-16 has been widely studied in the peripheral proinflammatory immune response; however, little is known about its role in adipocytes in the context of obesity., Aim & Methods: We aimed to study the levels of IL-16 in WAT derived from sWAT and vWAT depots of humans with obesity and the role of this cytokine in palmitate-exposed 3T3-L1 adipocytes., Results: The results demonstrated that IL-16 expression was higher in vWAT compared with sWAT in individuals with obesity. In addition, IL-16 serum levels were higher in patients with obesity compared with normal-weight individuals, increased at 6 months after bariatric surgery, and at 12 months after surgery decreased to levels similar to before the intervention. Our in vitro models showed that IL-16 could modulate markers of adipogenesis (Pref1), lipid metabolism (Plin1, Cd36, and Glut4), fibrosis (Hif1a, Col4a, Col6a, and Vegf), and inflammatory signaling (IL6) during adipogenesis and in mature adipocytes. In addition, lipid accumulation and glycerol release assays suggested lipolysis alteration., Discussion: Our results suggest a potential role of IL-16 in adipogenesis, lipid and glucose homeostasis, fibrosis, and inflammation in an obesity context., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Reyes-Farias, Fernández-García, Corrales, González, Soria-Gondek, Martínez, Pellitero, Tarascó, Moreno, Sumoy, Medina-Gómez, Sánchez-Infantes and Herrero.)
- Published
- 2024
- Full Text
- View/download PDF
7. Adults with excess weight or obesity, but not with overweight, report greater pain intensities than individuals with normal weight: a systematic review and meta-analysis.
- Author
-
Garcia MM, Corrales P, Huerta MÁ, Czachorowski MJ, López-Miranda V, Medina-Gómez G, Cobos EJ, Goicoechea C, and Molina-Álvarez M
- Subjects
- Humans, Adult, Body Mass Index, Chronic Pain epidemiology, Pain epidemiology, Pain etiology, Pain Measurement methods, Overweight complications, Overweight epidemiology, Obesity complications, Obesity epidemiology
- Abstract
Context: Over 1.9 billion adult people have overweight or obesity. Considered as a chronic disease itself, obesity is associated with several comorbidities. Chronic pain affects approximately 60 million people and its connection with obesity has been displayed in several studies. However, controversial results showing both lower and higher pain thresholds in subjects with obesity compared to individuals with normal weight and the different parameters used to define such association (e.g., pain severity, frequency or duration) make it hard to draw straight forward conclusions in the matter. The objective of this article is to examine the relationship between overweight and obesity (classified with BMI as recommended by WHO) and self-perceived pain intensity in adults., Methods: A literature search was conducted following PRISMA guidelines using the databases CINAHL, Cochrane Library, EMBASE, PEDro, PubMed, Scopus and Web of Science to identify original studies that provide BMI values and their associated pain intensity assessed by self-report scales. Self-report pain scores were normalized and pooled within meta-analyses. The Cochrane's Q test and I
2 index were used to clarify the amount of heterogeneity; meta-regression was performed to explore the relationship between each outcome and the risk of bias., Results: Of 2194 studies, 31 eligible studies were identified and appraised, 22 of which provided data for a quantitative analysis. The results herein suggested that adults with excess weight (BMI ≥ 25.0) or obesity (BMI ≥ 30.0) but not with overweight (pre-obesity) alone (BMI 25.0-29.9), are more likely to report greater intensities of pain than individuals of normal weight (BMI 18.5-24.9). Subgroup analyses regarding the pathology of the patients showed no statistically significant differences between groups. Also, influence of age in the effect size, evaluated by meta-regression, was only observed in one of the four analyses. Furthermore, the robustness of the findings was supported by two different sensitivity analyses., Conclusion: Subjects with obesity and excess weight, but not overweight, reported greater pain intensities than individuals with normal weight. This finding encourages treatment of obesity as a component of pain management. More research is required to better understand the mechanisms of these differences and the clinical utility of the findings., Systematic Review Registration: https://doi.org/10.17605/OSF.IO/RF2G3, identifier OSF.IO/RF2G3., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Garcia, Corrales, Huerta, Czachorowski, López-Miranda, Medina-Gómez, Cobos, Goicoechea and Molina-Álvarez.)- Published
- 2024
- Full Text
- View/download PDF
8. Claudin-1 as a novel target gene induced in obesity and associated to inflammation, fibrosis, and cell differentiation.
- Author
-
Fernández-García P, Taxerås SD, Reyes-Farias M, González L, Soria-Gondek A, Pellitero S, Tarascó J, Moreno P, Sumoy L, Stephens JM, Yoo LG, Galán M, Izquierdo A, Medina-Gómez G, Herrero L, Corrales P, Villarroya F, Cereijo R, and Sánchez-Infantes D
- Subjects
- Humans, Cell Differentiation, Fibrosis, Inflammation metabolism, T-Lymphocytes metabolism, Adipose Tissue, White metabolism, Claudin-1 metabolism, Obesity complications
- Abstract
Objective: T lymphocytes from visceral and subcutaneous white adipose tissues (vWAT and sWAT, respectively) can have opposing roles in the systemic metabolic changes associated with obesity. However, few studies have focused on this subject. Claudin-1 (CLDN1) is a protein involved canonically in tight junctions and tissue paracellular permeability. We evaluated T-lymphocyte gene expression in vWAT and sWAT and in the whole adipose depots in human samples., Methods: A Clariom D-based transcriptomic analysis was performed on T lymphocytes magnetically separated from vWAT and sWAT from patients with obesity (Cohort 1; N = 11). Expression of candidate genes resulting from that analysis was determined in whole WAT from individuals with and without obesity (Cohort 2; patients with obesity: N = 13; patients without obesity: N = 14)., Results: We observed transcriptional differences between T lymphocytes from sWAT compared with vWAT. Specifically, CLDN1 expression was found to be dramatically induced in vWAT T cells relative to those isolated from sWAT in patients with obesity. CLDN1 was also induced in obesity in vWAT and its expression correlates with genes involved in inflammation, fibrosis, and adipogenesis., Conclusion: These results suggest that CLDN1 is a novel marker induced in obesity and differentially expressed in T lymphocytes infiltrated in human vWAT as compared with sWAT. This protein may have a crucial role in the crosstalk between T lymphocytes and other adipose tissue cells and may contribute to inflammation, fibrosis, and alter homeostasis and promote metabolic disease in obesity., Competing Interests: Conflict of interest: None declared., (© The Author(s) 2024. Published by Oxford University Press on behalf of European Society of Endocrinology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
9. DIDO is necessary for the adipogenesis that promotes diet-induced obesity.
- Author
-
García-López MÁ, Mora A, Corrales P, Pons T, Sánchez de Diego A, Talavera Gutiérrez A, van Wely KHM, Medina-Gómez G, Sabio G, Martínez-A C, and Fischer T
- Subjects
- Animals, Mice, Cell Differentiation, Diet, Obesity genetics, Overweight, Adipogenesis genetics, Lipodystrophy
- Abstract
The prevalence of overweight and obesity continues to rise in the population worldwide. Because it is an important predisposing factor for cancer, cardiovascular diseases, diabetes mellitus, and COVID-19, obesity reduces life expectancy. Adipose tissue (AT), the main fat storage organ with endocrine capacity, plays fundamental roles in systemic metabolism and obesity-related diseases. Dysfunctional AT can induce excess or reduced body fat (lipodystrophy). Dido1 is a marker gene for stemness; gene-targeting experiments compromised several functions ranging from cell division to embryonic stem cell differentiation, both in vivo and in vitro. We report that mutant mice lacking the DIDO N terminus show a lean phenotype. This consists of reduced AT and hypolipidemia, even when mice are fed a high-nutrient diet. DIDO mutation caused hypothermia due to lipoatrophy of white adipose tissue (WAT) and dermal fat thinning. Deep sequencing of the epididymal white fat (Epi WAT) transcriptome supported Dido1 control of the cellular lipid metabolic process. We found that, by controlling the expression of transcription factors such as C/EBPα or PPARγ, Dido1 is necessary for adipocyte differentiation, and that restoring their expression reestablished adipogenesis capacity in Dido1 mutants. Our model differs from other lipodystrophic mice and could constitute a new system for the development of therapeutic intervention in obesity., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
10. Detection of binucleated nephrin-marked podocytes by flow cytometry in the urine of patients with obesity.
- Author
-
Carrasco AG, Izquierdo-Lahuerta A, de Pablos IG, Vila-Bedmar R, Martin-Taboada M, Porrini E, Morales E, and Medina-Gómez G
- Subjects
- Humans, Flow Cytometry, Membrane Proteins, Obesity, Podocytes, Kidney Diseases
- Published
- 2024
- Full Text
- View/download PDF
11. The protective role of peroxisome proliferator-activated receptor gamma in lipotoxic podocytes.
- Author
-
Carrasco AG, Izquierdo-Lahuerta A, Valverde ÁM, Ni L, Flores-Salguero E, Coward RJ, and Medina-Gómez G
- Subjects
- Humans, PPAR gamma metabolism, Pioglitazone pharmacology, Bexarotene pharmacology, Podocytes, Thiazolidinediones metabolism, Thiazolidinediones pharmacology, Thiazolidinediones therapeutic use, Kidney Diseases drug therapy
- Abstract
Podocytes are specialized epithelial cells that maintain the glomerular filtration barrier. These cells are susceptible to lipotoxicity in the obese state and irreversibly lost during kidney disease leading to proteinuria and renal injury. PPARγ is a nuclear receptor whose activation can be renoprotective. This study examined the role of PPARγ in the lipotoxic podocyte using a PPARγ knockout (PPARγKO) cell line and since the activation of PPARγ by Thiazolidinediones (TZD) is limited by their side effects, it explored other alternative therapies to prevent podocyte lipotoxic damage. Wild-type and PPARγKO podocytes were exposed to the fatty acid palmitic acid (PA) and treated with the TZD (Pioglitazone) and/or the Retinoid X receptor (RXR) agonist Bexarotene (BX). It revealed that podocyte PPARγ is essential for podocyte function. PPARγ deletion reduced key podocyte proteins including podocin and nephrin while increasing basal levels of oxidative and ER stress causing apoptosis and cell death. A combination therapy of low-dose TZD and BX activated both the PPARγ and RXR receptors reducing PA-induced podocyte damage. This study confirms the crucial role of PPARγ in podocyte biology and that their activation in combination therapy of TZD and BX may be beneficial in the treatment of obesity-related kidney disease., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Adriana Izquierdo-Lahuerta reports travel was provided by Ministerio de Educación Cultura y Deporte. Gema Medina-Gomez reports financial support was provided by Ministerio de Economía y Competitividad de España. Gema Medina-Gomez reports financial support was provided by Community of Madrid Health Service. Richard J. Coward reports financial support was provided by British Medical Research Council., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Myeloid p38 activation maintains macrophage-liver crosstalk and BAT thermogenesis through IL-12-FGF21 axis.
- Author
-
Crespo M, Nikolic I, Mora A, Rodríguez E, Leiva-Vega L, Pintor-Chocano A, Horrillo D, Hernández-Cosido L, Torres JL, Novoa E, Nogueiras R, Medina-Gómez G, Marcos M, Leiva M, and Sabio G
- Subjects
- Humans, Animals, Mice, Interleukin-12, Obesity metabolism, Adipose Tissue, Brown metabolism, Energy Metabolism, Inflammation metabolism, Diet, High-Fat, Macrophages metabolism, Thermogenesis, Mice, Inbred C57BL, Fatty Liver metabolism, Insulin Resistance
- Abstract
Obesity features excessive fat accumulation in several body tissues and induces a state of chronic low-grade inflammation that contributes to the development of diabetes, steatosis, and insulin resistance. Recent research has shown that this chronic inflammation is crucially dependent on p38 pathway activity in macrophages, suggesting p38 inhibition as a possible treatment for obesity comorbidities. Nevertheless, we report here that lack of p38 activation in myeloid cells worsens high-fat diet-induced obesity, diabetes, and steatosis. Deficient p38 activation increases macrophage IL-12 production, leading to inhibition of hepatic FGF21 and reduction of thermogenesis in the brown fat. The implication of FGF21 in the phenotype was confirmed by its specific deletion in hepatocytes. We also found that IL-12 correlates with liver damage in human biopsies, indicating the translational potential of our results. Our findings suggest that myeloid p38 has a dual role in inflammation and that drugs targeting IL-12 might improve the homeostatic regulation of energy balance in response to metabolic stress., (Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of American Association for the Study of Liver Diseases.)
- Published
- 2023
- Full Text
- View/download PDF
13. p27 Kip1 Deficiency Impairs Brown Adipose Tissue Function Favouring Fat Accumulation in Mice.
- Author
-
Colon-Mesa I, Sainz N, Corrales P, Collantes M, Kaldis P, Martinez JA, Medina-Gómez G, Moreno-Aliaga MJ, and Escoté X
- Subjects
- Animals, Mice, Diet, High-Fat adverse effects, Fluorodeoxyglucose F18 metabolism, Insulin metabolism, Mice, Inbred C57BL, Mice, Knockout, Obesity genetics, Obesity metabolism, Thermogenesis, Adipose Tissue, Brown metabolism, Insulin Resistance genetics
- Abstract
The aim of this work was to investigate the effect of the whole-body deletion of p27 on the activity of brown adipose tissue and the susceptibility to develop obesity and glucose homeostasis disturbances in mice, especially when subjected to a high fat diet. p27 knockout ( p27
-/- ) and wild type (WT) mice were fed a normal chow diet or a high fat diet (HFD) for 10-weeks. Body weight and composition were assessed. Insulin and glucose tolerance tests and indirect calorimetry assays were performed. Histological analysis of interscapular BAT (iBAT) was carried out, and expression of key genes/proteins involved in BAT function were characterized by qPCR and Western blot. iBAT activity was estimated by18 F-fluorodeoxyglucose (18 FDG) uptake with microPET. p27-/- mice were more prone to develop obesity and insulin resistance, exhibiting increased size of all fat depots. p27-/- mice displayed a higher respiratory exchange ratio. iBAT presented larger adipocytes in p27-/- HFD mice, accompanied by downregulation of both Glut1 and uncoupling protein 1 (UCP1) in parallel with defective insulin signalling. Moreover, p27-/- HFD mice exhibited impaired response to cold exposure, characterized by a reduced iBAT18 FDG uptake and difficulty to maintain body temperature when exposed to cold compared to WT HFD mice, suggesting reduced thermogenic capacity. These data suggest that p27 could play a role in BAT activation and in the susceptibility to develop obesity and insulin resistance.- Published
- 2023
- Full Text
- View/download PDF
14. Novel Insights in the Physiopathology and Management of Obesity-Related Kidney Disease.
- Author
-
Sandino J, Martín-Taboada M, Medina-Gómez G, Vila-Bedmar R, and Morales E
- Subjects
- Glucagon-Like Peptide 1 agonists, Humans, Hypertrophy complications, Lipids, Sodium-Glucose Transporter 2, Diabetic Nephropathies etiology, Obesity complications, Obesity therapy
- Abstract
Obesity is recognized as an independent risk factor for the development of kidney disease, which has led to the designation of obesity-related glomerulopathy (ORG). Common renal features observed in this condition include glomerular hypertrophy, glomerulosclerosis, haemodynamic changes and glomerular filtration barrier defects. Additionally, and although less studied, obesity-related kidney disease also involves alterations in renal tubules, including tubule hypertrophy, lipid deposition and tubulointerstitial fibrosis. Although not completely understood, the harmful effects of obesity on the kidney may be mediated by different mechanisms, with alterations in adipose tissue probably playing an important role. An increase in visceral adipose tissue has classically been associated with the development of kidney damage, however, recent studies point to adipose tissue surrounding the kidney, and specifically to the fat within the renal sinus, as potentially involved in the development of ORG. In addition, new strategies for the treatment of patients with obesity-related kidney disease are focusing on the management of obesity. In this regard, some non-invasive options, such as glucagon-like peptide-1 (GLP-1) receptor agonists or sodium-glucose cotransporter-2 (SGLT2) inhibitors, are being considered for application in the clinic, not only for patients with diabetic kidney disease but as a novel pharmacological strategy for patients with ORG. In addition, bariatric surgery stands as one of the most effective options, not only for weight loss but also for the improvement of kidney outcomes in obese patients with chronic kidney disease.
- Published
- 2022
- Full Text
- View/download PDF
15. Potential benefits of egg white hydrolysate in the prevention of Hg-induced dysfunction in adipose tissue.
- Author
-
Rizzetti DA, Corrales P, Uranga-Ocio JA, Medina-Gómez G, Peçanha FM, Vassallo DV, Miguel M, and Wiggers GA
- Subjects
- Adiponectin genetics, Adiponectin metabolism, Adipose Tissue, Adipose Tissue, White metabolism, Animals, Antioxidants pharmacology, Egg White, Glucose metabolism, Leptin metabolism, Lipids pharmacology, PPAR alpha metabolism, PPAR gamma genetics, PPAR gamma metabolism, RNA, Messenger metabolism, Rats, Rats, Wistar, Insulins metabolism, Insulins pharmacology, Mercury metabolism, Mercury pharmacology
- Abstract
Aim : To investigate the effects of egg white hydrolysate (EWH) on the lipid and glycemic metabolism disruption in the white adipose tissue (WAT) dysfunction induced by mercury (Hg). Experimental : Wistar rats were treated for 60 days: control (saline, intramuscular - i.m.); hydrolysate (EWH, gavage, 1 g kg
-1 day-1 ); mercury (HgCl2 , i.m., 1st dose 4.6 μg kg-1 , subsequent doses 0.07 μg kg-1 day-1 ) and hydrolysate-mercury (EWH-HgCl2 ). Hg level and histological analyses were performed in epididymal WAT (eWAT), pancreas and liver. GRP78, CHOP, PPARα, PPARγ, leptin, adiponectin, and CD11 mRNA expressions were analyzed in eWAT. The plasma lipid profile, glucose, and insulin levels were measured. Antioxidant status was also evaluated in the plasma and liver. Results : EWH intake prevented the reduced eWAT weight, adipocyte size, insulin levels, and antioxidant defenses and the increased glucose and triglyceride levels induced by Hg exposure; hepatic glutathione levels were higher in rats co-treated with EWH. The increased mRNA expression of CHOP, PPARα, and leptin induced by Hg was reduced in co-treated rats. EWH did not modify the elevated mRNA expression of GRP78, PPARγ and adiponectin in Hg-treated rats. Increased levels of Hg were found in the liver; the co-treatment did not alter this parameter. EWH prevented the morphological and metabolic disorder induced by Hg, by improving antioxidant defenses, inactivating pro-apoptotic pathways and normalizing the mRNA expression of PPARs and adipokines. Its effects enabled an increase in insulin levels and a normal balance between the fat storage and expenditure mechanisms in WAT. Conclusions : EWH may have potential benefits in the prevention and management of Hg-related metabolic disorders.- Published
- 2022
- Full Text
- View/download PDF
16. Tackling the effects of extracellular vesicles in fibrosis.
- Author
-
Martín-Taboada M, Corrales P, Medina-Gómez G, and Vila-Bedmar R
- Subjects
- Adipocytes, Adipose Tissue, Female, Fibrosis, Humans, Male, Obesity, Extracellular Vesicles, Kidney Diseases
- Abstract
Fibrosis is a physiological process of tissue repair that turns into pathological when becomes chronic, damaging the functional structure of the tissue. In this review we outline the current status of extracellular vesicles as modulators of the fibrotic process at different levels. In adipose tissue, extracellular vesicles mediate the intercellular communication not only between adipocytes, but also between adipocytes and other cells of the stromal vascular fraction. Thus, they could be altering essential processes for the functionality of adipose tissue, such as adipocyte hypertrophy/hyperplasia, tissue plasticity, adipogenesis and/or inflammation, and ultimately trigger fibrosis. This process is particularly important in obesity, and may eventually, influence the development of obesity-associated alterations. In this regard, obesity is now recognized as an independent risk factor for the development of chronic kidney disease, although the role of extracellular vesicles in this connection has not been explored so far. Nonetheless, the role of extracellular vesicles in the onset and progression of renal fibrosis has been highlighted due to the critical role of fibrosis as a common feature of kidney diseases. In fact, the content of extracellular vesicles disturbs cellular signaling cascades involved in fibrosis in virtually all types of renal cells. What is certain is that the study of extracellular vesicles is complex, as their isolation and manipulation is still difficult to reproduce, which complicates the overview of their physiopathological effects. Nevertheless, new strategies have been developed to exploit the potential of extracellular vesicles and their cargo, both as biomarkers and as therapeutic tools to prevent the progression of fibrosis towards an irreversible event., (Copyright © 2022 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
17. Obesity and pregnancy, the perfect metabolic storm.
- Author
-
Corrales P, Vidal-Puig A, and Medina-Gómez G
- Subjects
- Epigenesis, Genetic, Female, Humans, Obesity, Placenta metabolism, Pregnancy, Insulin Resistance, Overnutrition, Pregnancy Complications
- Abstract
Pregnancy is a physiological stress that requires dynamic, regulated changes affecting maternal and fetal adiposity. Excessive accumulation of dysfunctional adipose tissue defined by metabolic and molecular alterations cause severe health consequences for mother and fetus. When subjected to sustained overnutrition, the cellular and lipid composition of the adipose tissue changes predisposing to insulin resistance, diabetes, and other metabolic disorders compromising the outcome of the pregnancy. Moreover, excessive maternal weight gain, usually in the context of obesity, predisposes to an increased flux of nutrients from mother to fetus throughout the placenta. The fetus of an obese mother will accumulate more adiposity and may increase the risk of future metabolic disorder later in life. Thus, further understanding of the interaction between maternal metabolism, epigenetic regulation of the adipose tissue, and their transgenerational transfer are required to mitigate the adverse health outcomes for the mother and the fetus associated with maternal obesity., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
18. Deletion of pleiotrophin impairs glucose tolerance and liver metabolism in pregnant mice: Moonlighting role of glycerol kinase.
- Author
-
Zapatería B, Sevillano J, Sánchez-Alonso MG, Limones M, Pizarro-Delgado J, Zuccaro A, Herradón G, Medina-Gómez G, and Ramos-Álvarez MP
- Subjects
- Animals, Cholesterol metabolism, Fatty Acids metabolism, Female, Glucose biosynthesis, Glucose metabolism, Lipoproteins metabolism, Membrane Transport Proteins metabolism, Mice, Pregnancy, Transcription Factors metabolism, Triglycerides metabolism, Weight Gain genetics, Carrier Proteins genetics, Cytokines deficiency, Cytokines genetics, Gene Deletion, Glucose Intolerance genetics, Glycerol Kinase metabolism, Lipid Metabolism, Liver metabolism
- Abstract
Pleiotrophin is a pleiotropic cytokine that has been demonstrated to have a critical role in regulating energy metabolism, lipid turnover and plasticity of adipose tissue. Here, we hypothesize that this cytokine can be involved in regulatory processes of glucose and lipid homeostasis in the liver during pregnancy. Using 18-days pregnant Ptn-deficient mice, we evaluated the biochemical profile (circulating variables), tissue mRNA expression (qPCR) and protein levels of key enzymes and transcription factors involved in main metabolic pathways. Ptn deletion was associated with a reduction in body weight gain, hyperglycemia and glucose intolerance. Moreover, we observed an impairment in glucose synthesis and degradation during late pregnancy in Ptn
-/- mice. Hepatic lipid content was significantly lower (73.6%) in Ptn-/- mice and was associated with a clear reduction in fatty acid, triacylglycerides and cholesterol synthesis. Ptn deletion was accompanying with a diabetogenic state in the mother and a decreased expression of key proteins involved in glucose and lipid uptake and metabolism. Moreover, Ptn-/- pregnant mice have a decreased expression of transcription factors, such as PPAR-α, regulating lipid uptake and glucose and lipid utilization. Furthermore, the augmented expression and nuclear translocation of glycerol kinase, and the decrease in NUR77 protein levels in the knock-out animals can further explain the alterations observed in hepatic glucose metabolism. Our results point out for the first time that pleiotrophin is an important player in maintaining hepatic metabolic homeostasis during late gestation, and further highlighted the moonlighting role of glycerol kinase in the regulation of maternal glucose homeostasis during pregnancy., (© 2021 The Authors. The FASEB Journal published by Wiley Periodicals LLC on behalf of Federation of American Societies for Experimental Biology.)- Published
- 2021
- Full Text
- View/download PDF
19. Transforming growth factor β3 deficiency promotes defective lipid metabolism and fibrosis in murine kidney.
- Author
-
Escasany E, Lanzón B, García-Carrasco A, Izquierdo-Lahuerta A, Torres L, Corrales P, Rodríguez Rodríguez AE, Luis-Lima S, Martínez Álvarez C, Javier Ruperez F, Ros M, Porrini E, Rydén M, and Medina-Gómez G
- Subjects
- Animals, Fibrosis, Kidney metabolism, Mice, Mice, Knockout, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta1 metabolism, Lipid Metabolism, Transforming Growth Factor beta3 metabolism
- Abstract
Glomerulosclerosis and tubulointerstitial fibrosis are pathological features of chronic kidney disease. Transforming growth factor β (TGFβ) is a key player in the development of fibrosis. However, of the three known TGFβ isoforms, only TGFβ1 has an established role in fibrosis, and the pathophysiological relevance of TGFβ2 and TGFβ3 is unknown. Because Tgfb3 deficiency in mice results in early postnatal lethality, we analyzed the kidney phenotype of heterozygous Tgfb3-knockout mice (Tgfb3+/-) and compared it with that of matched wild-type mice. Four-month-old Tgfb3+/- mice exhibited incipient renal fibrosis with epithelial-mesenchymal transition, in addition to glomerular basement membrane thickening and podocyte foot process effacement associated with albuminuria. Also evident was insulin resistance and oxidative stress at the renal level, together with aberrant renal lipid metabolism and mitochondrial function. Omics analysis revealed toxic species, such as diacylglycerides and ceramides, and dysregulated mitochondrial metabolism in Tgfb3+/- mice. Kidneys of Tgfb3+/- mice showed morphological alterations of mitochondria and overactivation of non-canonical MAPK ERK1/2 and JNK cascades. Our study indicates that renal TGFβ3 might have antifibrotic and renoprotective properties, opposing or counteracting the activity of TGFβ1. This article has an associated First Person interview with the first author of the paper., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2021. Published by The Company of Biologists Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
20. Pleiotrophin Deficiency Induces Browning of Periovarian Adipose Tissue and Protects against High-Fat Diet-Induced Hepatic Steatosis.
- Author
-
Zuccaro A, Zapatería B, Sánchez-Alonso MG, Haro M, Limones M, Terrados G, Izquierdo A, Corrales P, Medina-Gómez G, Herradón G, Sevillano J, and Ramos-Álvarez MDP
- Subjects
- Adipose Tissue, White metabolism, Animals, Biomarkers, Carrier Proteins, Disease Models, Animal, Energy Metabolism, Fatty Liver pathology, Gene Expression, Liver metabolism, Liver pathology, Mice, Mice, Knockout, Organ Size, Uncoupling Protein 1 genetics, Uncoupling Protein 1 metabolism, Adipose Tissue, Brown metabolism, Cytokines deficiency, Diet, High-Fat adverse effects, Disease Susceptibility, Fatty Liver etiology, Fatty Liver metabolism
- Abstract
(1) Background: Pleiotrophin preserves insulin sensitivity, regulates adipose tissue lipid turnover and plasticity, energy metabolism and thermogenesis. The aim of this study was to determine the role of pleiotrophin in hepatic lipid metabolism and in the metabolic crosstalk between the liver and brown and white adipose tissue (AT) in a high-fat diet-induced (HFD) obesity mice model. (2) Methods: We analyzed circulating variables, lipid metabolism (hepatic lipid content and mRNA expression), brown AT thermogenesis (UCP-1 expression) and periovarian AT browning (brown adipocyte markers mRNA and immunodetection) in Ptn
-/- mice either fed with standard-chow diet or with HFD and in their corresponding Ptn+/+ counterparts. (3) Results: HFD- Ptn-/- mice are protected against the development of HFD-induced insulin resistance, had lower liver lipid content and lower expression of the key enzymes involved in triacylglycerides and fatty acid synthesis in liver. HFD- Ptn-/- mice showed higher UCP-1 expression in brown AT. Moreover, Ptn deletion increased the expression of specific markers of brown/beige adipocytes and was associated with the immunodetection of UCP-1 enriched multilocular adipocytes in periovarian AT. (4) Conclusions: Ptn deletion protects against the development of HFD-induced insulin resistance and liver steatosis, by increasing UCP-1 expression in brown AT and promoting periovarian AT browning.- Published
- 2021
- Full Text
- View/download PDF
21. Autocrine IGF2 programmes β-cell plasticity under conditions of increased metabolic demand.
- Author
-
Sandovici I, Hammerle CM, Virtue S, Vivas-Garcia Y, Izquierdo-Lahuerta A, Ozanne SE, Vidal-Puig A, Medina-Gómez G, and Constância M
- Subjects
- Animals, Female, Glucose metabolism, Homeostasis, Insulin blood, Insulin Resistance, Insulin-Secreting Cells metabolism, Male, Mice, Mice, Knockout, Pregnancy, Cell Plasticity physiology, Insulin-Like Growth Factor II physiology, Insulin-Secreting Cells cytology
- Abstract
When exposed to nutrient excess and insulin resistance, pancreatic β-cells undergo adaptive changes in order to maintain glucose homeostasis. The role that growth control genes, highly expressed in early pancreas development, might exert in programming β-cell plasticity in later life is a poorly studied area. The imprinted Igf2 (insulin-like growth factor 2) gene is highly transcribed during early life and has been identified in recent genome-wide association studies as a type 2 diabetes susceptibility gene in humans. Hence, here we investigate the long-term phenotypic metabolic consequences of conditional Igf2 deletion in pancreatic β-cells (Igf2
βKO ) in mice. We show that autocrine actions of IGF2 are not critical for β-cell development, or for the early post-natal wave of β-cell remodelling. Additionally, adult Igf2βKO mice maintain glucose homeostasis when fed a chow diet. However, pregnant Igf2βKO females become hyperglycemic and hyperinsulinemic, and their conceptuses exhibit hyperinsulinemia and placentomegalia. Insulin resistance induced by congenital leptin deficiency also renders Igf2βKO females more hyperglycaemic compared to leptin-deficient controls. Upon high-fat diet feeding, Igf2βKO females are less susceptible to develop insulin resistance. Based on these findings, we conclude that in female mice, autocrine actions of β-cell IGF2 during early development determine their adaptive capacity in adult life.- Published
- 2021
- Full Text
- View/download PDF
22. The Kidney-Heart Connection in Obesity.
- Author
-
García-Carrasco A, Izquierdo-Lahuerta A, and Medina-Gómez G
- Subjects
- Cardio-Renal Syndrome physiopathology, Humans, Cardio-Renal Syndrome complications, Heart physiopathology, Kidney physiopathology, Obesity physiopathology
- Abstract
There is a strong relationship between the kidney and the heart, where if one of these organs fails, so does the other, in the so-called cardiorenal syndrome (CRS). Besides, there are also interactions with the rest of the body leading to a metabolic state that establishes a feedback loop that is perpetuated. The CRS is characterized by hemodynamic changes, activation of neuro-humoral systems, natriuretic peptides, and changes in mineral metabolism. In this scenario, the kidney and heart, connected by a dysfunctional endothelium, inevitably fail. In obesity, this syndrome is exacerbated due to the complications of adipose tissue dysfunction, in the so-called cardiorenal metabolic syndrome (CRMetS). Obesity promotes adipose tissue dysfunction because it exceeds lipid storage capacity and leads to a lipotoxic state, characterized by inflammation, hypertension, insulin resistance and dyslipidemia, oxidative stress, and hyperuricemia, among others, that affect different organs other than the adipose tissue. In addition, the pro-inflammatory gut microbiota present in obese patients releases uremic toxins, contributing to oxidative stress and inflammation, perpetuating and accelerating the progression of this pathology. In this article, we describe the contribution of obesity, the factors and mechanisms implicated in the development of the CRMetS. Despite the great knowledge about the CRS, more research is needed to characterize the CRMetS given the global obesity epidemic., (© 2021 S. Karger AG, Basel.)
- Published
- 2021
- Full Text
- View/download PDF
23. From Obesity to Chronic Kidney Disease: How Can Adipose Tissue Affect Renal Function?
- Author
-
Martin-Taboada M, Vila-Bedmar R, and Medina-Gómez G
- Subjects
- Biomarkers metabolism, Humans, Kidney Failure, Chronic metabolism, Kidney Failure, Chronic physiopathology, Kidney Failure, Chronic therapy, MicroRNAs metabolism, Obesity physiopathology, Adipose Tissue physiopathology, Kidney Failure, Chronic etiology, Obesity complications
- Abstract
Obesity is directly associated with an increased risk of developing CKD, regardless of other comorbid conditions. Although the molecular mechanisms that link both diseases are not well established, the role of adipose tissue (AT) is becoming increasingly important in obesity-associated kidney damage. In the context of obesity, lipotoxicity and the alteration of AT secretion profile promote inflammation, oxidative stress, and fibrosis in the kidney, which ultimately leads to impaired renal function. Different studies have highlighted the importance of body weight loss in the improvement of renal function markers. In this regard, bariatric surgery, rather than low-calorie diets, has been accepted as the most effective option to lose weight. In fact, a significant reduction in proteinuria and hyperfiltration has been observed in association with surgically induced weight loss. Detection of early signs of kidney dysfunction in patients with obesity has not been accomplished yet, though. Therefore, understanding the harmful effects within the adipo-renal axis is essential to prevent the progression to the irreversible renal insufficiency. MicroRNAs have recently been described as important modulators of normal kidney function. Some of these microRNAs could be potential early markers of kidney damage, which would help with the diagnosis and the prevention of CKD., (© 2021 S. Karger AG, Basel.)
- Published
- 2021
- Full Text
- View/download PDF
24. Renoprotective role of bariatric surgery in patients with established chronic kidney disease.
- Author
-
Morales E, Porrini E, Martin-Taboada M, Luis-Lima S, Vila-Bedmar R, González de Pablos I, Gómez P, Rodríguez E, Torres L, Lanzón B, Rodríguez AE, Maíz M, Medina-Gómez G, and Praga M
- Abstract
Background: Bariatric surgery (BS) has been postulated as the most effective measure for weight reduction. Weight loss improves metabolic parameters and exerts changes in renal function that lead to the amelioration of absolute or relative glomerular hyperfiltration, a condition that may be renoprotective in the long term. However, few studies have demonstrated the influence of BS in patients with severe obesity and chronic kidney disease (CKD). Our objective was to analyse the evolution of renal function, adipose tissue-derived molecules and inflammatory parameters in patients with CKD after BS., Methods: This is an observational and prospective study. Thirty patients were screened and 12 were included between January 2016 and January 2018 with a 24-month follow-up. Glomerular filtration rate (GFR) was determined by plasma iohexol clearance. Adipokines, cytokines, circulating hormones and fibrotic parameters were evaluated before and 12 months after BS using the Bioplex system., Results: The mean age was 50.6 years and 58.3% were males. Seven patients had a body mass index >40 kg/m
2 and 66.7% were diabetic. Twenty-four months following BS there was a significant decrease in body weight (36.4%). Proteinuria decreased by 63.7 ± 28.2%. Measured GFR significantly diminished from before surgery to Month 24 after surgery (94 ± 44 to 79 ± 44 mL/min, P = 0.03). There was a significant decrease in adipocyte-derived molecules (leptin and vifastin) as well as in pro-inflammatory cytokines [interleukin (IL)-1β, tumour necrosis factor α, IL-6 and monocyte chemoattractant protein-1] and other circulating factors (vascular endothelial growth factor and transforming growth factor β isoforms)., Conclusions: BS is an effective option to prevent kidney damage in obese subjects with CKD due to the improvement of glomerular hyperfiltration, adipocyte cytokines metabolic and inflammatory parameters., (© The Author(s) 2020. Published by Oxford University Press on behalf of ERA-EDTA.)- Published
- 2020
- Full Text
- View/download PDF
25. Mesenchyme-derived IGF2 is a major paracrine regulator of pancreatic growth and function.
- Author
-
Hammerle CM, Sandovici I, Brierley GV, Smith NM, Zimmer WE, Zvetkova I, Prosser HM, Sekita Y, Lam BYH, Ma M, Cooper WN, Vidal-Puig A, Ozanne SE, Medina-Gómez G, and Constância M
- Subjects
- Acinar Cells metabolism, Acinar Cells pathology, Amino Acids genetics, Animals, Cell Lineage genetics, Chromium, DNA Methylation genetics, Female, Flow Cytometry, Gene Expression Regulation, Developmental genetics, Genomic Imprinting genetics, Insulin-Secreting Cells metabolism, Insulin-Secreting Cells pathology, Mice, Nicotinic Acids genetics, Pancreas cytology, Pancreas metabolism, Pregnancy, RNA, Long Noncoding genetics, Insulin-Like Growth Factor II genetics, Mesoderm growth & development, Pancreas growth & development, Paracrine Communication genetics
- Abstract
The genetic mechanisms that determine the size of the adult pancreas are poorly understood. Imprinted genes, which are expressed in a parent-of-origin-specific manner, are known to have important roles in development, growth and metabolism. However, our knowledge regarding their roles in the control of pancreatic growth and function remains limited. Here we show that many imprinted genes are highly expressed in pancreatic mesenchyme-derived cells and explore the role of the paternally-expressed insulin-like growth factor 2 (Igf2) gene in mesenchymal and epithelial pancreatic lineages using a newly developed conditional Igf2 mouse model. Mesenchyme-specific Igf2 deletion results in acinar and beta-cell hypoplasia, postnatal whole-body growth restriction and maternal glucose intolerance during pregnancy, suggesting that the mesenchyme is a developmental reservoir of IGF2 used for paracrine signalling. The unique actions of mesenchymal IGF2 are demonstrated by the absence of any discernible growth or functional phenotypes upon Igf2 deletion in the developing pancreatic epithelium. Additionally, increased IGF2 levels specifically in the mesenchyme, through conditional Igf2 loss-of-imprinting or Igf2r deletion, leads to pancreatic acinar overgrowth. Furthermore, ex-vivo exposure of primary acinar cells to exogenous IGF2 activates AKT, a key signalling node, and increases their number and amylase production. Based on these findings, we propose that mesenchymal Igf2, and perhaps other imprinted genes, are key developmental regulators of adult pancreas size and function., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
26. Factors Accounting for Obesity and Its Perception among the Adult Spanish Population: Data from 1,000 Computer-Assisted Telephone Interviews.
- Author
-
Lecube A, Sánchez E, Monereo S, Medina-Gómez G, Bellido D, García-Almeida JM, Martínez de Icaya P, Malagón MM, Goday A, and Tinahones FJ
- Subjects
- Adult, Body Mass Index, Body Weight, Computers, Family, Female, Humans, Male, Middle Aged, Self Report, Spain epidemiology, Telephone, Obesity epidemiology, Self Concept
- Abstract
Objective: Our aim was to go deeper in the self-perception of weight and health status among the Spanish population, together with the connections of familiar relationships, physical activity practice, nutritional habits, and sleep patterns with the presence of obesity., Methods: A total of 1,000 subjects were enrolled in April 2017 in a representative adult Spanish population sample. Computer-assisted telephone interviewing was used and self-reported anthropometric data was obtained., Results: The population was composed of 51.3% women, with a mean age of 48 (36-63) years and a BMI of 23.2 (20.3-26.6). Although only 17.7% of subjects with self-reported obesity exhibited the self-perception to suffer from obesity, they referred a bad (16%) or regular (47%) self-perceived health status. Subjects who considered themselves as people with overweight and obesity displayed a BMI of 30.5 (28.7-32.2) and 37.1 (34.8-41.5), respectively. The obesity group displayed the highest percentage (71.9%) of participants with some first-degree relative with overweight or obesity (p < 0.001) in comparison with the other groups. The main reason put forward of preventing healthy eating among subjects with obesity was that they dislike healthy food. The multivariable logistic regression model for presence of obesity showed that there was a significant association with older age, presence of a first-degree relative with weight excess, a positive snacking habit, and daily alcohol consumption (p ≤ 0.019)., Conclusion: The Spanish population has a low self-perception of obesity. Our data also reinforces the strong association between obesity and age, family interactions, usual snacking, and daily consumption of wine or beer., (© 2020 The Author(s) Published by S. Karger AG, Basel.)
- Published
- 2020
- Full Text
- View/download PDF
27. SUCNR1 controls an anti-inflammatory program in macrophages to regulate the metabolic response to obesity.
- Author
-
Keiran N, Ceperuelo-Mallafré V, Calvo E, Hernández-Alvarez MI, Ejarque M, Núñez-Roa C, Horrillo D, Maymó-Masip E, Rodríguez MM, Fradera R, de la Rosa JV, Jorba R, Megia A, Zorzano A, Medina-Gómez G, Serena C, Castrillo A, Vendrell J, and Fernández-Veledo S
- Subjects
- Adipose Tissue drug effects, Adipose Tissue immunology, Adipose Tissue metabolism, Animals, Cells, Cultured, Cytokines genetics, Cytokines immunology, Cytokines metabolism, Gene Expression Profiling methods, Humans, Inflammation genetics, Inflammation metabolism, Macrophages metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Obesity genetics, Obesity metabolism, Receptors, G-Protein-Coupled deficiency, Receptors, G-Protein-Coupled genetics, Succinic Acid immunology, Succinic Acid metabolism, Succinic Acid pharmacology, THP-1 Cells, Inflammation immunology, Macrophages immunology, Obesity immunology, Receptors, G-Protein-Coupled immunology
- Abstract
Succinate is a signaling metabolite sensed extracellularly by succinate receptor 1 (SUNCR1). The accumulation of succinate in macrophages is known to activate a pro-inflammatory program; however, the contribution of SUCNR1 to macrophage phenotype and function has remained unclear. Here we found that activation of SUCNR1 had a critical role in the anti-inflammatory responses in macrophages. Myeloid-specific deficiency in SUCNR1 promoted a local pro-inflammatory phenotype, disrupted glucose homeostasis in mice fed a normal chow diet, exacerbated the metabolic consequences of diet-induced obesity and impaired adipose-tissue browning in response to cold exposure. Activation of SUCNR1 promoted an anti-inflammatory phenotype in macrophages and boosted the response of these cells to type 2 cytokines, including interleukin-4. Succinate decreased the expression of inflammatory markers in adipose tissue from lean human subjects but not that from obese subjects, who had lower expression of SUCNR1 in adipose-tissue-resident macrophages. Our findings highlight the importance of succinate-SUCNR1 signaling in determining macrophage polarization and assign a role to succinate in limiting inflammation.
- Published
- 2019
- Full Text
- View/download PDF
28. Pleiotrophin deletion alters glucose homeostasis, energy metabolism and brown fat thermogenic function in mice.
- Author
-
Sevillano J, Sánchez-Alonso MG, Zapatería B, Calderón M, Alcalá M, Limones M, Pita J, Gramage E, Vicente-Rodríguez M, Horrillo D, Medina-Gómez G, Obregón MJ, Viana M, Valladolid-Acebes I, Herradón G, and Ramos-Álvarez MP
- Subjects
- Animals, Carrier Proteins genetics, Cytokines genetics, Energy Metabolism genetics, Female, Insulin Resistance genetics, Insulin Resistance physiology, Longitudinal Studies, Mice, Mice, Knockout, Thermogenesis genetics, Adipose Tissue, Brown metabolism, Carrier Proteins metabolism, Cytokines metabolism, Energy Metabolism physiology, Thermogenesis physiology
- Abstract
Aims/hypothesis: Pleiotrophin, a developmentally regulated and highly conserved cytokine, exerts different functions including regulation of cell growth and survival. Here, we hypothesise that this cytokine can play a regulatory role in glucose and lipid homeostasis., Methods: To test this hypothesis, we performed a longitudinal study characterising the metabolic profile (circulating variables and tissue mRNA expression) of gene-targeted Ptn-deficient female mice and their corresponding wild-type counterparts at different ages from young adulthood (3 months) to older age (15 months). Metabolic cages were used to investigate the respiratory exchange ratio and energy expenditure, at both 24°C and 30°C. Undifferentiated immortalised mouse brown adipocytes (mBAs) were treated with 0.1 μg/ml pleiotrophin until day 6 of differentiation, and markers of mBA differentiation were analysed by quantitative real-time PCR (qPCR)., Results: Ptn deletion was associated with a reduction in total body fat (20.2% in Ptn
+/+ vs 13.9% in Ptn-/- mice) and an enhanced lipolytic response to isoprenaline in isolated adipocytes from 15-month-old mice (189% in Ptn+/+ vs 273% in Ptn-/- mice). We found that Ptn-/- mice exhibited a significantly lower QUICKI value and an altered lipid profile; plasma triacylglycerols and NEFA did not increase with age, as happens in Ptn+/+ mice. Furthermore, the contribution of cold-induced thermogenesis to energy expenditure was greater in Ptn-/- than Ptn+/+ mice (42.6% and 33.6%, respectively). Body temperature and the activity and expression of deiodinase, T3 and mitochondrial uncoupling protein-1 in the brown adipose tissue of Ptn-/- mice were higher than in wild-type controls. Finally, supplementing brown pre-adipocytes with pleiotrophin decreased the expression of the brown adipocyte markers Cidea (20% reduction), Prdm16 (21% reduction), and Pgc1-α (also known as Ppargc1a, 11% reduction)., Conclusions/interpretation: Our results reveal for the first time that pleiotrophin is a key player in preserving insulin sensitivity, driving the dynamics of adipose tissue lipid turnover and plasticity, and regulating energy metabolism and thermogenesis. These findings open therapeutic avenues for the treatment of metabolic disorders by targeting pleiotrophin in the crosstalk between white and brown adipose tissue.- Published
- 2019
- Full Text
- View/download PDF
29. PPARs and Metabolic Disorders Associated with Challenged Adipose Tissue Plasticity.
- Author
-
Corrales P, Vidal-Puig A, and Medina-Gómez G
- Subjects
- Animals, Caloric Restriction, Humans, Metabolic Diseases genetics, Obesity genetics, Obesity metabolism, Peroxisome Proliferator-Activated Receptors genetics, Adipose Tissue metabolism, Metabolic Diseases metabolism, Peroxisome Proliferator-Activated Receptors metabolism
- Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of a family of nuclear hormone receptors that exert their transcriptional control on genes harboring PPAR-responsive regulatory elements (PPRE) in partnership with retinoid X receptors (RXR). The activation of PPARs coordinated by specific coactivators/repressors regulate networks of genes controlling diverse homeostatic processes involving inflammation, adipogenesis, lipid metabolism, glucose homeostasis, and insulin resistance. Defects in PPARs have been linked to lipodystrophy, obesity, and insulin resistance as a result of the impairment of adipose tissue expandability and functionality. PPARs can act as lipid sensors, and when optimally activated, can rewire many of the metabolic pathways typically disrupted in obesity leading to an improvement of metabolic homeostasis. PPARs also contribute to the homeostasis of adipose tissue under challenging physiological circumstances, such as pregnancy and aging. Given their potential pathogenic role and their therapeutic potential, the benefits of PPARs activation should not only be considered relevant in the context of energy balance-associated pathologies and insulin resistance but also as potential relevant targets in the context of diabetic pregnancy and changes in body composition and metabolic stress associated with aging. Here, we review the rationale for the optimization of PPAR activation under these conditions.
- Published
- 2018
- Full Text
- View/download PDF
30. Maintenance of Kidney Metabolic Homeostasis by PPAR Gamma.
- Author
-
Corrales P, Izquierdo-Lahuerta A, and Medina-Gómez G
- Subjects
- Animals, Humans, Insulin metabolism, Kidney drug effects, Kidney physiopathology, PPAR gamma agonists, Renin-Angiotensin System, Thiazolidinediones pharmacology, Glucose metabolism, Homeostasis, Kidney metabolism, Lipid Metabolism, PPAR gamma metabolism
- Abstract
Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear hormone receptors that control the transcription of specific genes by binding to regulatory DNA sequences. Among the three subtypes of PPARs, PPARγ modulates a broad range of physiopathological processes, including lipid metabolism, insulin sensitization, cellular differentiation, and cancer. Although predominantly expressed in adipose tissue, PPARγ expression is also found in different regions of the kidney and, upon activation, can redirect metabolism. Recent studies have highlighted important roles for PPARγ in kidney metabolism, such as lipid and glucose metabolism and renal mineral control. PPARγ is also implicated in the renin-angiotensin-aldosterone system and, consequently, in the control of systemic blood pressure. Accordingly, synthetic agonists of PPARγ have reno-protective effects both in diabetic and nondiabetic patients. This review focuses on the role of PPARγ in renal metabolism as a likely key factor in the maintenance of systemic homeostasis.
- Published
- 2018
- Full Text
- View/download PDF
31. Hypothalamic AMPK-ER Stress-JNK1 Axis Mediates the Central Actions of Thyroid Hormones on Energy Balance.
- Author
-
Martínez-Sánchez N, Seoane-Collazo P, Contreras C, Varela L, Villarroya J, Rial-Pensado E, Buqué X, Aurrekoetxea I, Delgado TC, Vázquez-Martínez R, González-García I, Roa J, Whittle AJ, Gomez-Santos B, Velagapudi V, Tung YCL, Morgan DA, Voshol PJ, Martínez de Morentin PB, López-González T, Liñares-Pose L, Gonzalez F, Chatterjee K, Sobrino T, Medina-Gómez G, Davis RJ, Casals N, Orešič M, Coll AP, Vidal-Puig A, Mittag J, Tena-Sempere M, Malagón MM, Diéguez C, Martínez-Chantar ML, Aspichueta P, Rahmouni K, Nogueiras R, Sabio G, Villarroya F, and López M
- Subjects
- Adipose Tissue, Brown metabolism, Animals, Lipid Metabolism, Liver metabolism, Male, Mice, Inbred C57BL, Rats, Rats, Sprague-Dawley, Thermogenesis, Triiodothyronine metabolism, Energy Metabolism, Hypothalamus metabolism, Mitogen-Activated Protein Kinase 8 metabolism, Signal Transduction, Thyroid Hormones metabolism
- Abstract
Thyroid hormones (THs) act in the brain to modulate energy balance. We show that central triiodothyronine (T3) regulates de novo lipogenesis in liver and lipid oxidation in brown adipose tissue (BAT) through the parasympathetic (PSNS) and sympathetic nervous system (SNS), respectively. Central T3 promotes hepatic lipogenesis with parallel stimulation of the thermogenic program in BAT. The action of T3 depends on AMP-activated protein kinase (AMPK)-induced regulation of two signaling pathways in the ventromedial nucleus of the hypothalamus (VMH): decreased ceramide-induced endoplasmic reticulum (ER) stress, which promotes BAT thermogenesis, and increased c-Jun N-terminal kinase (JNK) activation, which controls hepatic lipid metabolism. Of note, ablation of AMPKα1 in steroidogenic factor 1 (SF1) neurons of the VMH fully recapitulated the effect of central T3, pointing to this population in mediating the effect of central THs on metabolism. Overall, these findings uncover the underlying pathways through which central T3 modulates peripheral metabolism., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
32. Hypothalamus and thermogenesis: Heating the BAT, browning the WAT.
- Author
-
Contreras C, Nogueiras R, Diéguez C, Medina-Gómez G, and López M
- Subjects
- Animals, Humans, Models, Biological, Adipose Tissue, Brown physiology, Adipose Tissue, White physiology, Hot Temperature, Hypothalamus physiology, Thermogenesis physiology
- Abstract
Brown adipose tissue (BAT) has been also considered as the main thermogenic organ responsible of maintenance body temperature through heat production. However, a new type of thermogenic fat has been characterized during the last years, the beige or brite fat, that is developed from white adipose tissue (WAT) in response to different stimuli by a process known as browning. The activities of brown and beige adipocytes ameliorate metabolic disease, including obesity in mice and correlate with leanness in humans. Many genes and pathways that regulate brown and beige adipocyte biology have now been identified, providing a variety of promising therapeutic targets for metabolic disease. The hypothalamus is the main central place orchestrating the outflow signals that drive the sympathetic nerve activity to BAT and WAT, controlling heat production and energy homeostasis. Recent data have revealed new hypothalamic molecular mechanisms, such as hypothalamic AMP-activated protein kinase (AMPK), that control both thermogenesis and browning. This review provides an overview of the factors influencing BAT and WAT thermogenesis, with special focus on the integration of peripheral information on hypothalamic circuits controlling thermoregulation., (Copyright © 2016 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
33. Maternal Exposure to Bisphenol-A During Pregnancy Increases Pancreatic β-Cell Growth During Early Life in Male Mice Offspring.
- Author
-
García-Arévalo M, Alonso-Magdalena P, Servitja JM, Boronat-Belda T, Merino B, Villar-Pazos S, Medina-Gómez G, Novials A, Quesada I, and Nadal A
- Subjects
- Animals, Apoptosis drug effects, C-Peptide blood, Cell Proliferation drug effects, Fasting blood, Female, Glucose Tolerance Test, Insulin blood, Insulin-Secreting Cells cytology, Insulin-Secreting Cells metabolism, Leptin blood, Male, Mice, Pregnancy, Prenatal Exposure Delayed Effects, Uterus drug effects, Benzhydryl Compounds toxicity, Insulin-Secreting Cells physiology, Maternal Exposure adverse effects, Phenols toxicity
- Abstract
Alterations during development of metabolic key organs such as the endocrine pancreas affect the phenotype later in life. There is evidence that in utero or perinatal exposure to bisphenol-A (BPA) leads to impaired glucose metabolism during adulthood. However, how BPA exposure during pregnancy affects pancreatic β-cell growth and function in offspring during early life has not been explored. We exposed pregnant mice to either vehicle (control) or BPA (10 and 100 μg/kg·d, BPA10 and BPA100) and examined offspring on postnatal days (P) P0, P21, P30, and P120. BPA10 and BPA100 mice presented lower birth weight than control and subsequently gained weight until day 30. At that age, concentration of plasma insulin, C-peptide, and leptin were increased in BPA-exposed animals in the nonfasting state. Insulin secretion and content were diminished in BPA10 and maintained in BPA100 compared with control. A global gene expression analysis indicated that genes related with cell division were increased in islets from BPA-treated animals. This was associated with an increase in pancreatic β-cell mass at P0, P21, and P30 together with increased β-cell proliferation and decreased apoptosis. On the contrary, at P120, BPA-treated animals presented either equal or decreased β-cell mass compared with control and altered fasting glucose levels. These data suggest that in utero exposure to environmentally relevant doses of BPA alters the expression of genes involved in β-cell growth regulation, incrementing β-cell mass/area, and β-cell proliferation during early life. An excess of insulin signaling during early life may contribute to impaired glucose tolerance during adulthood.
- Published
- 2016
- Full Text
- View/download PDF
34. Lipotoxicity as a trigger factor of renal disease.
- Author
-
Izquierdo-Lahuerta A, Martínez-García C, and Medina-Gómez G
- Subjects
- Adipokines metabolism, Adipose Tissue physiopathology, Animals, Dietary Fats administration & dosage, Dietary Fats metabolism, Dyslipidemias epidemiology, Dyslipidemias genetics, Dyslipidemias physiopathology, Endoplasmic Reticulum Stress, Energy Intake, Genetic Markers, Glomerular Filtration Rate, Humans, Insulin Resistance, Kidney physiopathology, MicroRNAs genetics, Obesity epidemiology, Obesity genetics, Obesity physiopathology, RNA, Long Noncoding genetics, Reactive Oxygen Species metabolism, Renal Insufficiency epidemiology, Renal Insufficiency genetics, Renal Insufficiency physiopathology, Risk Assessment, Risk Factors, Signal Transduction, Adipose Tissue metabolism, Dietary Fats adverse effects, Dyslipidemias metabolism, Energy Metabolism, Kidney metabolism, Obesity metabolism, Renal Insufficiency metabolism
- Abstract
In the last few decades, rapid changes in lifestyle have led to an alarming increase in the prevalence of obesity and obesity-associated complications. Obese patients are at increased risk of developing hypertension, heart disease, insulin resistance, dyslipidemia, type 2 diabetes and kidney disease. The surplus of calories is normally stored as triglycerides in adipose tissue. However, excess lipids can also accumulate ectopically in other organs, including the kidney, contributing to their damage through toxic processes named lipotoxicity. The kidney is negatively affected by dyslipidemia, lipid accumulation and changes in circulating adipokines that bring about alterations in renal lipid metabolism and promote insulin resistance, generation of reactive oxygen species and endoplasmic reticulum stress, ultimately leading to alterations in the glomerular filtration barrier and renal failure. This review focuses on the pathogenic molecular mechanisms associated with renal lipotoxicity, and presents new insights about potential new therapeutic targets and biomarkers such as microRNAs and long non-coding RNAs, of relevance for the early detection of lipid-associated kidney disease.
- Published
- 2016
- Full Text
- View/download PDF
35. Obesity and type 2 diabetes in renal pathology.
- Author
-
Medina-Gómez G
- Subjects
- Humans, Diabetes Mellitus, Type 2 complications, Kidney Diseases etiology, Obesity complications
- Published
- 2013
- Full Text
- View/download PDF
36. Mitochondria and endocrine function of adipose tissue.
- Author
-
Medina-Gómez G
- Subjects
- Adipocytes metabolism, Adipose Tissue, Brown cytology, Adipose Tissue, Brown metabolism, Adipose Tissue, White cytology, Adipose Tissue, White metabolism, Animals, Diabetes Mellitus, Type 2 etiology, Humans, Lipid Metabolism, Mitochondria metabolism, Obesity etiology, Adipocytes physiology, Adipose Tissue, Brown physiology, Adipose Tissue, White physiology, Mitochondria physiology
- Abstract
Excess of adipose tissue is accompanied by an increase in the risk of developing insulin resistance, type 2 diabetes (T2D) and other complications. Nevertheless, total or partial absence of fat or its accumulation in other tissues (lipotoxicity) is also associated to these complications. White adipose tissue (WAT) was traditionally considered a metabolically active storage tissue for lipids while brown adipose tissue (BAT) was considered as a thermogenic adipose tissue with higher oxidative capacity. Nowadays, WAT is also considered an endocrine organ that contributes to energy homeostasis. Experimental evidence tends to link the malfunction of adipose mitochondria with the development of obesity and T2D. This review discusses the importance of mitochondrial function in adipocyte biology and the increased evidences of mitochondria dysfunction in these epidemics. New strategies targeting adipocyte mitochondria from WAT and BAT are also discussed as therapies against obesity and its complications in the near future., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
37. [Obesity, adipogenesis and insulin resistance].
- Author
-
Ros Pérez M and Medina-Gómez G
- Subjects
- Endoplasmic Reticulum metabolism, Humans, Inflammation etiology, Lipodystrophy metabolism, Obesity complications, Oxidative Stress, PPAR gamma physiology, Adipogenesis, Insulin Resistance, Obesity metabolism
- Abstract
Insulin resistance precedes the development of type 2 diabetes mellitus and is also a common denominator in the so-called metabolic syndrome. Although the cause of insulin resistance has not been fully elucidated, it seems clear that lifestyle changes, including little physical exercise and constant access to food, particularly in developed and economically emergent countries, as well as genetic factors, appear to have triggered the escalating incidence of diseases related to insulin resistance, including type 2 diabetes and metabolic syndrome. Obesity is considered as a risk factor for developing insulin resistance. Increased adipose tissue has been related to an increased production of pro-inflammatory cytokines which, together with fatty acids, appear to be responsible for the development of insulin resistance. Thus, a greater or lesser expansibility or ability of adipose tissue to store lipids also appears to play a significant role in the development of insulin resistance because overcoming of this capacity, which is variable in each case, would result in leaking of lipids to other tissues where they could interfere with insulin signaling. This article reviews various molecular mechanisms related to the development of insulin resistance and its relationship to expansibility of adipose tissue and obesity., (Copyright © 2011 SEEN. Published by Elsevier Espana. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
38. [Adipose tissue as a therapeutic target in obesity].
- Author
-
Medina-Gómez G and Vidal-Puig A
- Subjects
- Adipocytes drug effects, Adipocytes metabolism, Adipocytes physiology, Adipogenesis, Adipokines metabolism, Adipose Tissue blood supply, Adipose Tissue metabolism, Adipose Tissue physiopathology, Animals, Apoptosis, Autocrine Communication, Cell Size, Cytokines metabolism, Energy Metabolism, Humans, Inflammation etiology, Inflammation physiopathology, Insulin Resistance, Lipid Metabolism, Macrophages physiology, Mice, Mice, Knockout, Obesity physiopathology, PPAR gamma physiology, Paracrine Communication, Adipose Tissue drug effects, Anti-Obesity Agents pharmacology, Drug Delivery Systems, Obesity drug therapy
- Abstract
Obesity is characterized by an increase of adipose tissue as a result of a positive imbalance between food intake and energy expenditure. Recent studies have indicated that adipocyte function is more complex than expected, since these cells have multiple functions and are integrated in a homeostatic network to optimize energy resources. As metabolic sensors in the body, adipocytes and the surrounding stromal vascular cells produce and secrete autocrine, paracrine and endocrine factors, able to regulate aspects involved in the development of adipocytes, as well as effects in peripheral organs important for metabolism. Regulation of these endocrine factors could lead to new therapeutic approaches targeted at aspects related to adipogenesis, preadipocyte proliferation and differentiation, inflammatory cytokine release and secretion, adipose tissue vascularization, and regulation of lipid metabolism or, alternatively, regulation of energy dissipation in mitochondria. In the study of the mechanisms of adipogenesis and remodulation of adipose tissue with respect to adipocyte size and function, an alternative and unorthodox strategy to improve obesity-associated metabolic complications could consist of increasing the storage capacity of adipose tissue to prevent a toxic response known as lipotoxicity.
- Published
- 2009
- Full Text
- View/download PDF
39. Mitochondrial fusion is increased by the nuclear coactivator PGC-1beta.
- Author
-
Liesa M, Borda-d'Agua B, Medina-Gómez G, Lelliott CJ, Paz JC, Rojo M, Palacín M, Vidal-Puig A, and Zorzano A
- Subjects
- Animals, Cell Fusion, Cells, Cultured, GTP Phosphohydrolases genetics, Gene Expression Regulation, HeLa Cells, Humans, Liver metabolism, Mice, Mice, Knockout, Muscle, Skeletal metabolism, Myocardium metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, Receptors, Estrogen metabolism, Receptors, Estrogen physiology, Trans-Activators genetics, Transcription Factors, Transcription, Genetic, ERRalpha Estrogen-Related Receptor, Mitochondria physiology, Mitochondrial Size genetics, Trans-Activators physiology
- Abstract
Background: There is no evidence to date on whether transcriptional regulators are able to shift the balance between mitochondrial fusion and fission events through selective control of gene expression., Methodology/principal Findings: Here, we demonstrate that reduced mitochondrial size observed in knock-out mice for the transcriptional regulator PGC-1beta is associated with a selective reduction in Mitofusin 2 (Mfn2) expression, a mitochondrial fusion protein. This decrease in Mfn2 is specific since expression of the remaining components of mitochondrial fusion and fission machinery were not affected. Furthermore, PGC-1beta increases mitochondrial fusion and elongates mitochondrial tubules. This PGC-1beta-induced elongation specifically requires Mfn2 as this process is absent in Mfn2-ablated cells. Finally, we show that PGC-1beta increases Mfn2 promoter activity and transcription by coactivating the nuclear receptor Estrogen Related Receptor alpha (ERRalpha)., Conclusions/significance: Taken together, our data reveal a novel mechanism by which mammalian cells control mitochondrial fusion. In addition, we describe a novel role of PGC-1beta in mitochondrial physiology, namely the control of mitochondrial fusion mainly through Mfn2.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.