37 results on '"Porteiro B"'
Search Results
2. Parabrachial Interleukin-6 Reduces Body Weight and Food Intake and Increases Thermogenesis to Regulate Energy Metabolism
- Author
-
Mishra, D., Richard, J.E., Maric, I., Porteiro, B., Haring, M., Kooijman, S., Musovic, S., Eerola, K., Lopez-Ferreras, L., Peris, E., Grycel, K., Shevchouk, O.T., Micallef, P., Olofsson, C.S., Asterholm, I.W., Grill, H.J., Nogueiras, R., Skibicka, K.P., Universidade de Santiago de Compostela. Centro de Investigación en Medicina Molecular e Enfermidades Crónicas, and Universidade de Santiago de Compostela. Departamento de Fisioloxía
- Subjects
Leptin ,Male ,sex differences ,Thyroid Hormones ,obesity ,Sympathetic Nervous System ,diet-induced obesity ,Cold exposure ,cold exposure ,Energy balance ,Brown adipose tissue ,Article ,thyroid ,Rats, Sprague-Dawley ,Eating ,Mice ,Adipose Tissue, Brown ,Sex differences ,Animals ,Obesity ,lcsh:QH301-705.5 ,Neurons ,Thyroid ,Interleukin-6 ,Body Weight ,lateral PBN ,Diet-induced obesity ,Thermogenesis ,brown adipose tissue ,Parabrachial Nucleus ,Hindbrain ,energy balance ,Rats ,Mice, Inbred C57BL ,lcsh:Biology (General) ,Astrocytes ,Female ,PVN ,Energy Metabolism ,Lateral PBN ,hindbrain - Abstract
Summary Chronic low-grade inflammation and increased serum levels of the cytokine IL-6 accompany obesity. For brain-produced IL-6, the mechanisms by which it controls energy balance and its role in obesity remain unclear. Here, we show that brain-produced IL-6 is decreased in obese mice and rats in a neuroanatomically and sex-specific manner. Reduced IL-6 mRNA localized to lateral parabrachial nucleus (lPBN) astrocytes, microglia, and neurons, including paraventricular hypothalamus-innervating lPBN neurons. IL-6 microinjection into lPBN reduced food intake and increased brown adipose tissue (BAT) thermogenesis in male lean and obese rats by increasing thyroid and sympathetic outflow to BAT. Parabrachial IL-6 interacted with leptin to reduce feeding. siRNA-mediated reduction of lPBN IL-6 leads to increased weight gain and adiposity, reduced BAT thermogenesis, and increased food intake. Ambient cold exposure partly normalizes the obesity-induced suppression of lPBN IL-6. These results indicate that lPBN-produced IL-6 regulates feeding and metabolism and pinpoints (patho)physiological contexts interacting with lPBN IL-6., Graphical Abstract, Highlights • Obesogenic diet robustly reduces, and 4°C cold exposure increases, rodent lPBN IL-6 • IL-6 interacts with leptin at the level of the lPBN to reduce food intake • LPBN IL-6 leads to increased BAT thermogenesis by thyroid and sympathetic mechanisms • Reduction in lPBN IL-6 increases weight gain and adiposity, Mishra et al. demonstrate that IL-6 is produced by lPBN neurons, astrocytes, and microglia. They show that IL-6 acts in the lPBN to reduce body weight by increasing thermogenesis and reducing food intake. They also identify two pathophysiological/physiological contexts, obesity and cold exposure, that divergently affect lPBN IL-6 production.
- Published
- 2019
3. Atualização e protocolo de atuação na Drepanocitose no Adulto
- Author
-
Major, M, Porteiro, B, Costa, M, Martins, J, and Silva, R
- Subjects
Terapêutica ,Adulto ,Drepanocitose ,Anemia falciforme - Abstract
N/A
- Published
- 2019
4. Tuberculomas cerebrais e tuberculose disseminada.
- Author
-
Costa, M., Porteiro, B., Vitorino, M., Nunes, F., Alexys, B., Major, M., and Machado, J.
- Abstract
Copyright of RPDI - Revista Portuguesa de Doenças Infecciosas is the property of Sociedade Portuguesa de Doencas Infecciosas e Microbiologia Clinica and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2019
5. Remodeling p38 signaling in muscle controls locomotor activity via IL-15.
- Author
-
Folgueira C, Herrera-Melle L, López JA, Galvan-Alvarez V, Martin-Rincon M, Cuartero MI, García-Culebras A, Dumesic PA, Rodríguez E, Leiva-Vega L, León M, Porteiro B, Iglesias C, Torres JL, Hernández-Cosido L, Bonacasa C, Marcos M, Moro MÁ, Vázquez J, Calbet JAL, Spiegelman BM, Mora A, and Sabio G
- Subjects
- Humans, Animals, Locomotion, Mice, p38 Mitogen-Activated Protein Kinases metabolism, Signal Transduction, Male, MAP Kinase Signaling System, Obesity metabolism, Interleukin-15 metabolism, Muscle, Skeletal metabolism, Exercise physiology
- Abstract
Skeletal muscle has gained recognition as an endocrine organ releasing myokines upon contraction during physical exercise. These myokines exert both local and pleiotropic health benefits, underscoring the crucial role of muscle function in countering obesity and contributing to the overall positive effects of exercise on health. Here, we found that exercise activates muscle p38γ, increasing locomotor activity through the secretion of interleukin-15 (IL-15). IL-15 signals in the motor cortex, stimulating locomotor activity. This activation of muscle p38γ, leading to an increase locomotor activity, plays a crucial role in reducing the risk of diabetes and liver steatosis, unveiling a vital muscle-brain communication pathway with profound clinical implications. The correlation between p38γ activation in human muscle during acute exercise and increased blood IL-15 levels highlights the potential therapeutic relevance of this pathway in treating obesity and metabolic diseases. These findings provide valuable insights into the molecular basis of exercise-induced myokine responses promoting physical activity.
- Published
- 2024
- Full Text
- View/download PDF
6. Inhibition of hepatic bile salt uptake by Bulevirtide reduces atherosclerosis in Oatp1a1 -/- Ldlr -/- mice.
- Author
-
Porteiro B, Roscam Abbing RLP, In Het Panhuis W, de Waart DR, Duijst S, Bolt I, Vogels EW, Levels JHM, Bosmans LA, Vos WG, Oude Elferink RPJ, Lutgens E, and van de Graaf SFJ
- Subjects
- Animals, Female, Mice, Mice, Knockout, Organic Anion Transporters metabolism, Organic Anion Transporters genetics, Organic Anion Transporters antagonists & inhibitors, Organic Anion Transporters, Sodium-Dependent antagonists & inhibitors, Organic Anion Transporters, Sodium-Dependent metabolism, Organic Anion Transporters, Sodium-Dependent genetics, Symporters metabolism, Symporters genetics, Symporters antagonists & inhibitors, Organic Cation Transport Proteins genetics, Organic Cation Transport Proteins metabolism, Atherosclerosis drug therapy, Atherosclerosis metabolism, Atherosclerosis genetics, Bile Acids and Salts metabolism, Liver metabolism, Liver drug effects, Receptors, LDL deficiency, Receptors, LDL genetics, Receptors, LDL metabolism
- Abstract
Bile salts can strongly influence energy metabolism through systemic signaling, which can be enhanced by inhibiting the hepatic bile salt transporter Na
+ taurocholate cotransporting polypeptide (NTCP), thereby delaying hepatic reuptake of bile salts to increase systemic bile salt levels. Bulevirtide is an NTCP inhibitor and was originally developed to prevent NTCP-mediated entry of Hepatitis B and D into hepatocytes. We previously demonstrated that NTCP inhibition lowers body weight, induces glucagon-like peptide-1 (GLP1) secretion, and lowers plasma cholesterol levels in murine obesity models. In humans, a genetic loss-of-function variant of NTCP has been associated with reduced plasma cholesterol levels. Here, we aimed to assess if Bulevirtide treatment attenuates atherosclerosis development by treating female Ldlr-/- mice with Bulevirtide or vehicle for 11 weeks. Since this did not result in the expected increase in plasma bile salt levels, we generated Oatp1a1-/- Ldlr-/- mice, an atherosclerosis-prone model with human-like hepatic bile salt uptake characteristics. These mice showed delayed plasma clearance of bile salts and elevated bile salt levels upon Bulevirtide treatment. At the study endpoint, Bulevirtide-treated female Oatp1a1-/- Ldlr-/- mice had reduced atherosclerotic lesion area in the aortic root that coincided with lowered plasma LDL-c levels, independent of intestinal cholesterol absorption. In conclusion, Bulevirtide, which is considered safe and is EMA-approved for the treatment of Hepatitis D, reduces atherosclerotic lesion area by reducing plasma LDL-c levels. We anticipate that its application may extend to atherosclerotic cardiovascular diseases, which warrants clinical trials., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
7. Inhibition of hepatic p63 ameliorates steatohepatitis with fibrosis in mice.
- Author
-
Fondevila MF, Novoa E, Fernandez U, Dorta V, Parracho T, Kreimeyer H, Garcia-Vence M, Chantada-Vazquez MP, Bravo SB, Porteiro B, Cabaleiro A, Koning M, Senra A, Souto Y, Verheij J, Guallar D, Fidalgo M, Meijnikman AS, da Silva Lima N, Dieguez C, Gonzalez-Rellan MJ, and Nogueiras R
- Subjects
- Animals, Mice, Male, Disease Models, Animal, Diet, High-Fat adverse effects, Trans-Activators metabolism, Trans-Activators genetics, Proteomics, Methionine deficiency, Methionine metabolism, Liver metabolism, Liver pathology, Liver Cirrhosis metabolism, Liver Cirrhosis pathology, Mice, Inbred C57BL, Fatty Liver metabolism, Fatty Liver pathology
- Abstract
Objective: p63 is a transcription factor involved in multiple biological functions. In the liver, the TAp63 isoform induces lipid accumulation in hepatocytes. However, the role of liver TAp63 in the progression of metabolic dysfunction-associated steatohepatitis (MASH) with fibrosis is unknown., Methods: We evaluated the hepatic p63 levels in different mouse models of steatohepatitis with fibrosis induced by diet. Next, we used virogenetic approaches to manipulate the expression of TAp63 in adult mice under diet-induced steatohepatitis with fibrosis and characterized the disease condition. Finally, we performed proteomics analysis in mice with overexpression and knockdown of hepatic TAp63., Results: Levels of TAp63, but not of ΔN isoform, are increased in the liver of mice with diet-induced steatohepatitis with fibrosis. Both preventive and interventional strategies for the knockdown of hepatic TAp63 significantly ameliorated diet-induced steatohepatitis with fibrosis in mice fed a methionine- and choline-deficient diet (MCDD) and choline deficient and high fat diet (CDHFD). The overexpression of hepatic TAp63 in mice aggravated the liver condition in mice fed a CDHFD. Proteomic analysis in the liver of these mice revealed alteration in multiple proteins and pathways, such as oxidative phosphorylation, antioxidant activity, peroxisome function and LDL clearance., Conclusions: These results indicate that liver TAp63 plays a critical role in the progression of diet-induced steatohepatitis with fibrosis, and its inhibition ameliorates the disease., Competing Interests: Declaration of competing interest The authors have nothing to declare., (Copyright © 2024 The Author(s). Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Mitochondrial antiviral signaling protein enhances MASLD progression through the ERK/TNFα/NFκβ pathway.
- Author
-
Nóvoa E, da Silva Lima N, Gonzalez-Rellan MJ, Chantada-Vazquez MDP, Verheij J, Rodriguez A, Esquinas-Roman EM, Fondevila MF, Koning M, Fernandez U, Cabaleiro A, Parracho T, Iglesias-Moure J, Seoane S, Porteiro B, Escudero A, Senra A, Perez-Fernandez R, López M, Fidalgo M, Guallar D, Martinez-Chantar ML, Dieguez C, Varela-Rey M, Prevot V, Schwaninger M, Meijnikman A, Bravo SB, Frühbeck G, and Nogueiras R
- Abstract
Background and Aims: Mitochondrial antiviral signaling protein (MAVS) is a critical regulator that activates the host's innate immunity against RNA viruses, and its signaling pathway has been linked to the secretion of proinflammatory cytokines. However, the actions of MAVS on inflammatory pathways during the development of metabolic dysfunction-associated steatotic liver disease (MASLD) have been little studied., Approach and Results: Liver proteomic analysis of mice with genetically manipulated hepatic p63, a transcription factor that induces liver steatosis, revealed MAVS as a target downstream of p63. MAVS was thus further evaluated in liver samples from patients and in animal models with MASLD. Genetic inhibition of MAVS was performed in hepatocyte cell lines, primary hepatocytes, spheroids, and mice. MAVS expression is induced in the liver of both animal models and people with MASLD as compared with those without liver disease. Using genetic knockdown of MAVS in adult mice ameliorates diet-induced MASLD. In vitro, silencing MAVS blunts oleic and palmitic acid-induced lipid content, while its overexpression increases the lipid load in hepatocytes. Inhibiting hepatic MAVS reduces circulating levels of the proinflammatory cytokine TNFα and the hepatic expression of both TNFα and NFκβ. Moreover, the inhibition of ERK abolished the activation of TNFα induced by MAVS. The posttranslational modification O -GlcNAcylation of MAVS is required to activate inflammation and to promote the high lipid content in hepatocytes., Conclusions: MAVS is involved in the development of steatosis, and its inhibition in previously damaged hepatocytes can ameliorate MASLD., (Copyright © 2024 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
9. p63 controls metabolic activation of hepatic stellate cells and fibrosis via an HER2-ACC1 pathway.
- Author
-
Fondevila MF, Novoa E, Gonzalez-Rellan MJ, Fernandez U, Heras V, Porteiro B, Parracho T, Dorta V, Riobello C, da Silva Lima N, Seoane S, Garcia-Vence M, Chantada-Vazquez MP, Bravo SB, Senra A, Leiva M, Marcos M, Sabio G, Perez-Fernandez R, Dieguez C, Prevot V, Schwaninger M, Woodhoo A, Martinez-Chantar ML, Schwabe R, Cubero FJ, Varela-Rey M, Crespo J, Iruzubieta P, and Nogueiras R
- Subjects
- Humans, Mice, Animals, Activation, Metabolic, Liver Cirrhosis genetics, Liver Cirrhosis chemically induced, Liver Cirrhosis metabolism, Fibrosis, Acetyl-CoA Carboxylase genetics, Acetyl-CoA Carboxylase metabolism, Hepatic Stellate Cells metabolism, Hepatic Stellate Cells pathology, Non-alcoholic Fatty Liver Disease pathology
- Abstract
The p63 protein has pleiotropic functions and, in the liver, participates in the progression of nonalcoholic fatty liver disease (NAFLD). However, its functions in hepatic stellate cells (HSCs) have not yet been explored. TAp63 is induced in HSCs from animal models and patients with liver fibrosis and its levels positively correlate with NAFLD activity score and fibrosis stage. In mice, genetic depletion of TAp63 in HSCs reduces the diet-induced liver fibrosis. In vitro silencing of p63 blunts TGF-β1-induced HSCs activation by reducing mitochondrial respiration and glycolysis, as well as decreasing acetyl CoA carboxylase 1 (ACC1). Ectopic expression of TAp63 induces the activation of HSCs and increases the expression and activity of ACC1 by promoting the transcriptional activity of HER2. Genetic inhibition of both HER2 and ACC1 blunt TAp63-induced activation of HSCs. Thus, TAp63 induces HSC activation by stimulating the HER2-ACC1 axis and participates in the development of liver fibrosis., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Whipple's Disease: A Challenging Diagnosis.
- Author
-
Porteiro B, Freitas AR, Nunes F, Major M, and Batista F
- Abstract
Whipple's disease (WD) is a chronic multisystemic infection caused by Tropheryma whipplei . It is a rare condition with a wide spectrum of clinical presentations, necessitating a high clinical suspicion to arrive at the diagnosis. We present the case of a 65-year-old woman who experienced chronic, intermittent, and migratory polyarthralgia, weight loss, anorexia, and pyrosis. She was admitted due to bilateral deep vein thrombosis (DVT). She exhibited lymphadenopathy without hepatosplenomegaly, and lymph node biopsy revealed reactive lymphadenitis with intrahistiocytic bacilli that reacted positively to periodic acid-Schiff staining. This led to the suspicion of WD, which was subsequently confirmed through small bowel biopsies. She initiated treatment with ceftriaxone and experienced rapid clinical improvement. WD poses a diagnostic challenge. The signs and symptoms are often nonspecific and can result in misdiagnosis as a rheumatic or neoplastic disease. The presentation with DVT, while unusual, has been reported as a manifestation of WD., Competing Interests: The authors have declared that no competing interests exist., (Copyright © 2024, Porteiro et al.)
- Published
- 2024
- Full Text
- View/download PDF
11. Hepatic p63 regulates glucose metabolism by repressing SIRT1.
- Author
-
Gonzalez-Rellan MJ, Novoa E, da Silva Lima N, Rodriguez A, Veyrat-Durebex C, Seoane S, Porteiro B, Fondevila MF, Fernandez U, Varela-Rey M, Senra A, Iglesias C, Escudero A, Fidalgo M, Guallar D, Perez-Fernandez R, Prevot V, Schwaninger M, López M, Dieguez C, Coppari R, Frühbeck G, and Nogueiras R
- Subjects
- Animals, Mice, Glucose metabolism, Liver metabolism, Pyruvates metabolism, Diabetes Mellitus, Type 2, Sirtuin 1 metabolism, Trans-Activators metabolism
- Abstract
Objective: p63 is a transcription factor within the p53 protein family that has key roles in development, differentiation and prevention of senescence, but its metabolic actions remain largely unknown. Herein, we investigated the physiological role of p63 in glucose metabolism., Design: We used cell lines and mouse models to genetically manipulate p63 in hepatocytes. We also measured p63 in the liver of patients with obesity with or without type 2 diabetes (T2D)., Results: We show that hepatic p63 expression is reduced on fasting. Mice lacking the specific isoform TAp63 in the liver (p63LKO) display higher postprandial and pyruvate-induced glucose excursions. These mice have elevated SIRT1 levels, while SIRT1 knockdown in p63LKO mice normalises glycaemia. Overexpression of TAp63 in wild-type mice reduces postprandial, pyruvate-induced blood glucose and SIRT1 levels. Studies carried out in hepatocyte cell lines show that TAp63 regulates SIRT1 promoter by repressing its transcriptional activation. TAp63 also mediates the inhibitory effect of insulin on hepatic glucose production, as silencing TAp63 impairs insulin sensitivity. Finally, protein levels of TAp63 are reduced in obese persons with T2D and are negatively correlated with fasting glucose and homeostasis model assessment index., Conclusions: These results demonstrate that p63 physiologically regulates glucose homeostasis., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
12. Inhibition of carnitine palmitoyltransferase 1A in hepatic stellate cells protects against fibrosis.
- Author
-
Fondevila MF, Fernandez U, Heras V, Parracho T, Gonzalez-Rellan MJ, Novoa E, Porteiro B, Alonso C, Mayo R, da Silva Lima N, Iglesias C, Filliol AA, Senra A, Delgado TC, Woodhoo A, Herrero L, Serra D, Prevot V, Schwaninger M, López M, Dieguez C, Millet O, Mato JM, Cubero FJ, Varela-Rey M, Iruzubieta P, Crespo J, Martinez-Chantar ML, Schwabe RF, and Nogueiras R
- Subjects
- Animals, Choline, Fatty Acids metabolism, Fibrosis, Humans, Liver pathology, Liver Cirrhosis metabolism, Liver Cirrhosis prevention & control, Mice, Carnitine O-Palmitoyltransferase genetics, Carnitine O-Palmitoyltransferase metabolism, Hepatic Stellate Cells metabolism
- Abstract
Background & Aims: The pathogenesis of liver fibrosis requires activation of hepatic stellate cells (HSCs); once activated, HSCs lose intracellular fatty acids but the role of fatty acid oxidation and carnitine palmitoyltransferase 1A (CPT1A) in this process remains largely unexplored., Methods: CPT1A was found in HSCs of patients with fibrosis. Pharmacological and genetic manipulation of CPT1A were performed in human HSC cell lines and primary HCSs. Finally, we induced fibrosis in mice lacking CPT1A specifically in HSCs., Results: Herein, we show that CPT1A expression is elevated in HSCs of patients with non-alcoholic steatohepatitis, showing a positive correlation with the fibrosis score. This was corroborated in rodents with fibrosis, as well as in primary human HSCs and LX-2 cells activated by transforming growth factor β1 (TGFβ1) and fetal bovine serum (FBS). Furthermore, both pharmacological and genetic silencing of CPT1A prevent TGFβ1- and FBS-induced HSC activation by reducing mitochondrial activity. The overexpression of CPT1A, induced by saturated fatty acids and reactive oxygen species, triggers mitochondrial activity and the expression of fibrogenic markers. Finally, mice lacking CPT1A specifically in HSCs are protected against fibrosis induced by a choline-deficient high-fat diet, a methionine- and choline-deficient diet, or treatment with carbon tetrachloride., Conclusions: These results indicate that CPT1A plays a critical role in the activation of HSCs and is implicated in the development of liver fibrosis, making it a potentially actionable target for fibrosis treatment., Lay Summary: We show that the enzyme carnitine palmitoyltransferase 1A (CPT1A) is elevated in hepatic stellate cells (HSCs) in patients with fibrosis and mouse models of fibrosis, and that CPT1A induces the activation of these cells. Inhibition of CPT1A ameliorates fibrosis by preventing the activation of HSCs., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest related to the study. Please refer to the accompanying ICMJE disclosure forms for further details., (Copyright © 2022 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
13. Differential and organ-specific functions of organic solute transporter α and β in experimental cholestasis.
- Author
-
van de Wiel SMW, Porteiro B, Belt SC, Vogels EWM, Bolt I, Vermeulen JLM, de Waart DR, Verheij J, Muncan V, Oude Elferink RPJ, and van de Graaf SFJ
- Abstract
Background & Aims: Organic solute transporter (OST) subunits OSTα and OSTβ facilitate bile acid efflux from the enterocyte into the portal circulation. Patients with deficiency of OSTα or OSTβ display considerable variation in the level of bile acid malabsorption, chronic diarrhea, and signs of cholestasis. Herein, we generated and characterized a mouse model of OSTβ deficiency., Methods: Ostβ
-/- mice were generated using CRISR/Cas9 and compared to wild-type and Ostα-/- mice. OSTβ was re-expressed in livers of Ostβ-/- mice using adeno-associated virus serotype 8 vectors. Cholestasis was induced in both models by bile duct ligation (BDL) or 3.5-diethoxycarbonyl-1.4-dihydrocollidine (DDC) feeding., Results: Similar to Ostα-/- mice, Ostβ-/- mice exhibited elongated small intestines with blunted villi and increased crypt depth. Increased expression levels of ileal Fgf15, and decreased Asbt expression in Ostβ-/- mice indicate the accumulation of bile acids in the enterocyte. In contrast to Ostα-/- mice, induction of cholestasis in Ostβ-/- mice by BDL or DDC diet led to lower survival rates and severe body weight loss, but an improved liver phenotype. Restoration of hepatic Ostβ expression via adeno-associated virus-mediated overexpression did not rescue the phenotype of Ostβ-/- mice., Conclusions: OSTβ is pivotal for bile acid transport in the ileum and its deficiency leads to an intestinal phenotype similar to Ostα-/- mice, but it exerts distinct effects on survival and the liver phenotype, independent of its expression in the liver. Our findings provide insights into the variable clinical presentation of patients with OSTα and OSTβ deficiencies., Lay Summary: Organic solute transporter (OST) subunits OSTα and OSTβ together facilitate the efflux of conjugated bile acids into the portal circulation. Ostα knockout mice have longer and thicker small intestines and are largely protected against experimental cholestatic liver injury. Herein, we generated and characterized Ostβ knockout mice for the first time. Ostα and Ostβ knockout mice shared a similar phenotype under normal conditions. However, in cholestasis, Ostβ knockout mice had a worsened overall phenotype which indicates a separate and specific role of OSTβ, possibly as an interacting partner of other intestinal proteins., Competing Interests: The authors declare no conflicts of interest that pertain to this work. Please refer to the accompanying ICMJE disclosure forms for further details., (© 2022 The Authors.)- Published
- 2022
- Full Text
- View/download PDF
14. Methionine adenosyltransferase 1a antisense oligonucleotides activate the liver-brown adipose tissue axis preventing obesity and associated hepatosteatosis.
- Author
-
Sáenz de Urturi D, Buqué X, Porteiro B, Folgueira C, Mora A, Delgado TC, Prieto-Fernández E, Olaizola P, Gómez-Santos B, Apodaka-Biguri M, González-Romero F, Nieva-Zuluaga A, Ruiz de Gauna M, Goikoetxea-Usandizaga N, García-Rodríguez JL, Gutierrez de Juan V, Aurrekoetxea I, Montalvo-Romeral V, Novoa EM, Martín-Guerrero I, Varela-Rey M, Bhanot S, Lee R, Banales JM, Syn WK, Sabio G, Martínez-Chantar ML, Nogueiras R, and Aspichueta P
- Subjects
- Animals, Energy Metabolism, Liver metabolism, Mice, NF-E2-Related Factor 2 genetics, NF-E2-Related Factor 2 metabolism, Adipose Tissue, Brown metabolism, Insulin Resistance, Methionine Adenosyltransferase genetics, Methionine Adenosyltransferase metabolism, Obesity genetics, Obesity metabolism, Obesity prevention & control, Oligonucleotides, Antisense metabolism, Oligonucleotides, Antisense pharmacology
- Abstract
Altered methionine metabolism is associated with weight gain in obesity. The methionine adenosyltransferase (MAT), catalyzing the first reaction of the methionine cycle, plays an important role regulating lipid metabolism. However, its role in obesity, when a plethora of metabolic diseases occurs, is still unknown. By using antisense oligonucleotides (ASO) and genetic depletion of Mat1a, here, we demonstrate that Mat1a deficiency in diet-induce obese or genetically obese mice prevented and reversed obesity and obesity-associated insulin resistance and hepatosteatosis by increasing energy expenditure in a hepatocyte FGF21 dependent fashion. The increased NRF2-mediated FGF21 secretion induced by targeting Mat1a, mobilized plasma lipids towards the BAT to be catabolized, induced thermogenesis and reduced body weight, inhibiting hepatic de novo lipogenesis. The beneficial effects of Mat1a ASO were abolished following FGF21 depletion in hepatocytes. Thus, targeting Mat1a activates the liver-BAT axis by increasing NRF2-mediated FGF21 secretion, which prevents obesity, insulin resistance and hepatosteatosis., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
15. Eagle's Syndrome Presenting as Peripheral Facial Palsy.
- Author
-
Nunes F, Fernandes MJ, Silva M, Porteiro B, and Dutschmann R
- Abstract
Eagle's syndrome (ES) is the elongation of the ossified styloid process that causes symptoms such as foreign body sensation, neck pain, and odynophagia. A styloid process greater than 25 mm in length should be considered abnormal. Facial palsy is a condition that affects the facial nerve and results in weakness or total paralysis of the facial muscles that control expression. Here, we describe a rare presentation of ES presenting as facial palsy. We present the case of a 62-year-old female who was admitted to the emergency department with right peripheral facial palsy. A computed tomography (CT) scan of the neck confirmed the diagnosis. The patient underwent conservative management and physical therapy, which resulted in good evolution with an improvement of symptoms. She was referred to the otorhinolaryngologist for surgical evaluation., Competing Interests: The authors have declared that no competing interests exist., (Copyright © 2022, Nunes et al.)
- Published
- 2022
- Full Text
- View/download PDF
16. Small extracellular vesicle-mediated targeting of hypothalamic AMPKα1 corrects obesity through BAT activation.
- Author
-
Milbank E, Dragano NRV, González-García I, Garcia MR, Rivas-Limeres V, Perdomo L, Hilairet G, Ruiz-Pino F, Mallegol P, Morgan DA, Iglesias-Rey R, Contreras C, Vergori L, Cuñarro J, Porteiro B, Gavaldà-Navarro A, Oelkrug R, Vidal A, Roa J, Sobrino T, Villarroya F, Diéguez C, Nogueiras R, García-Cáceres C, Tena-Sempere M, Mittag J, Carmen Martínez M, Rahmouni K, Andriantsitohaina R, and López M
- Subjects
- Animals, Energy Metabolism, Mice, Thermogenesis, Weight Loss, AMP-Activated Protein Kinases metabolism, Adipose Tissue, Brown enzymology, Extracellular Vesicles metabolism, Hypothalamus enzymology, Obesity metabolism
- Abstract
Current pharmacological therapies for treating obesity are of limited efficacy. Genetic ablation or loss of function of AMP-activated protein kinase alpha 1 (AMPKα1) in steroidogenic factor 1 (SF1) neurons of the ventromedial nucleus of the hypothalamus (VMH) induces feeding-independent resistance to obesity due to sympathetic activation of brown adipose tissue (BAT) thermogenesis. Here, we show that body weight of obese mice can be reduced by intravenous injection of small extracellular vesicles (sEVs) delivering a plasmid encoding an AMPKα1 dominant negative mutant (AMPKα1-DN) targeted to VMH-SF1 neurons. The beneficial effect of SF1-AMPKα1-DN-loaded sEVs is feeding-independent and involves sympathetic nerve activation and increased UCP1-dependent thermogenesis in BAT. Our results underscore the potential of sEVs to specifically target AMPK in hypothalamic neurons and introduce a broader strategy to manipulate body weight and reduce obesity., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
17. O-GlcNAcylated p53 in the liver modulates hepatic glucose production.
- Author
-
Gonzalez-Rellan MJ, Fondevila MF, Fernandez U, Rodríguez A, Varela-Rey M, Veyrat-Durebex C, Seoane S, Bernardo G, Lopitz-Otsoa F, Fernández-Ramos D, Bilbao J, Iglesias C, Novoa E, Ameneiro C, Senra A, Beiroa D, Cuñarro J, Dp Chantada-Vazquez M, Garcia-Vence M, Bravo SB, Da Silva Lima N, Porteiro B, Carneiro C, Vidal A, Tovar S, Müller TD, Ferno J, Guallar D, Fidalgo M, Sabio G, Herzig S, Yang WH, Cho JW, Martinez-Chantar ML, Perez-Fernandez R, López M, Dieguez C, Mato JM, Millet O, Coppari R, Woodhoo A, Fruhbeck G, and Nogueiras R
- Subjects
- Animals, Base Sequence, Caloric Restriction, Cell Line, Colforsin pharmacology, Diabetes Mellitus, Type 2 complications, Diabetes Mellitus, Type 2 metabolism, Epinephrine metabolism, Glucagon metabolism, Glucocorticoids metabolism, Gluconeogenesis drug effects, Glycosylation, Hepatocytes drug effects, Hepatocytes metabolism, Humans, Hydrocortisone metabolism, Hyperglycemia complications, Hyperglycemia metabolism, Insulin Resistance, Intracellular Signaling Peptides and Proteins metabolism, Liver drug effects, Mice, Inbred C57BL, Mice, Knockout, Obesity complications, Obesity metabolism, Phosphoenolpyruvate Carboxykinase (GTP) metabolism, Promoter Regions, Genetic genetics, Protein Binding drug effects, Protein Stability drug effects, Pyruvic Acid metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Transcription, Genetic drug effects, Tumor Suppressor Protein p53 genetics, Mice, Acetylglucosamine metabolism, Glucose metabolism, Liver metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
p53 regulates several signaling pathways to maintain the metabolic homeostasis of cells and modulates the cellular response to stress. Deficiency or excess of nutrients causes cellular metabolic stress, and we hypothesized that p53 could be linked to glucose maintenance. We show here that upon starvation hepatic p53 is stabilized by O-GlcNAcylation and plays an essential role in the physiological regulation of glucose homeostasis. More specifically, p53 binds to PCK1 promoter and regulates its transcriptional activation, thereby controlling hepatic glucose production. Mice lacking p53 in the liver show a reduced gluconeogenic response during calorie restriction. Glucagon, adrenaline and glucocorticoids augment protein levels of p53, and administration of these hormones to p53 deficient human hepatocytes and to liver-specific p53 deficient mice fails to increase glucose levels. Moreover, insulin decreases p53 levels, and over-expression of p53 impairs insulin sensitivity. Finally, protein levels of p53, as well as genes responsible of O-GlcNAcylation are elevated in the liver of type 2 diabetic patients and positively correlate with glucose and HOMA-IR. Overall these results indicate that the O-GlcNAcylation of p53 plays an unsuspected key role regulating in vivo glucose homeostasis., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
18. The L-α-Lysophosphatidylinositol/G Protein-Coupled Receptor 55 System Induces the Development of Nonalcoholic Steatosis and Steatohepatitis.
- Author
-
Fondevila MF, Fernandez U, Gonzalez-Rellan MJ, Da Silva Lima N, Buque X, Gonzalez-Rodriguez A, Alonso C, Iruarrizaga-Lejarreta M, Delgado TC, Varela-Rey M, Senra A, Garcia-Outeiral V, Novoa E, Iglesias C, Porteiro B, Beiroa D, Folgueira C, Tojo M, Torres JL, Hernández-Cosido L, Blanco Ó, Arab JP, Barrera F, Guallar D, Fidalgo M, López M, Dieguez C, Marcos M, Martinez-Chantar ML, Arrese M, Garcia-Monzon C, Mato JM, Aspichueta P, and Nogueiras R
- Subjects
- Acetyl-CoA Carboxylase antagonists & inhibitors, Acetyl-CoA Carboxylase metabolism, Adult, Aged, Animals, Biopsy, Cannabinoid Receptor Agonists pharmacology, Cell Line, Cohort Studies, Diet, High-Fat adverse effects, Disease Models, Animal, Female, Gene Knockdown Techniques, Hepatic Stellate Cells, Hepatocytes, Humans, Lipogenesis drug effects, Liver pathology, Lysophospholipids blood, Male, Mice, Middle Aged, Non-alcoholic Fatty Liver Disease blood, Non-alcoholic Fatty Liver Disease etiology, Non-alcoholic Fatty Liver Disease pathology, Obesity blood, Obesity metabolism, Receptors, Cannabinoid genetics, Up-Regulation, Lysophospholipids metabolism, Non-alcoholic Fatty Liver Disease metabolism, Obesity complications, Receptors, Cannabinoid metabolism
- Abstract
Background and Aims: G protein-coupled receptor (GPR) 55 is a putative cannabinoid receptor, and l-α-lysophosphatidylinositol (LPI) is its only known endogenous ligand. Although GPR55 has been linked to energy homeostasis in different organs, its specific role in lipid metabolism in the liver and its contribution to the pathophysiology of nonalcoholic fatty liver disease (NAFLD) remains unknown., Approach and Results: We measured (1) GPR55 expression in the liver of patients with NAFLD compared with individuals without obesity and without liver disease, as well as animal models with steatosis and nonalcoholic steatohepatitis (NASH), and (2) the effects of LPI and genetic disruption of GPR55 in mice, human hepatocytes, and human hepatic stellate cells. Notably, we found that circulating LPI and liver expression of GPR55 were up-regulated in patients with NASH. LPI induced adenosine monophosphate-activated protein kinase activation of acetyl-coenzyme A carboxylase (ACC) and increased lipid content in human hepatocytes and in the liver of treated mice by inducing de novo lipogenesis and decreasing β-oxidation. The inhibition of GPR55 and ACCα blocked the effects of LPI, and the in vivo knockdown of GPR55 was sufficient to improve liver damage in mice fed a high-fat diet and in mice fed a methionine-choline-deficient diet. Finally, LPI promoted the initiation of hepatic stellate cell activation by stimulating GPR55 and activation of ACC., Conclusions: The LPI/GPR55 system plays a role in the development of NAFLD and NASH by activating ACC., (© 2020 The Authors. Hepatology published by Wiley Periodicals LLC on behalf of American Association for the Study of Liver Diseases.)
- Published
- 2021
- Full Text
- View/download PDF
19. Vav2 catalysis-dependent pathways contribute to skeletal muscle growth and metabolic homeostasis.
- Author
-
Rodríguez-Fdez S, Lorenzo-Martín LF, Fernández-Pisonero I, Porteiro B, Veyrat-Durebex C, Beiroa D, Al-Massadi O, Abad A, Diéguez C, Coppari R, Nogueiras R, and Bustelo XR
- Subjects
- Adipocytes, White drug effects, Adipocytes, White metabolism, Adipose Tissue, Brown metabolism, Animals, Body Composition drug effects, Body Weight drug effects, Cell Line, Cell Size drug effects, Genotype, Glucose pharmacology, Insulin metabolism, Insulin-Like Growth Factor I metabolism, Mice, Inbred C57BL, Mice, Knockout, Muscle Cells cytology, Muscle Cells drug effects, Muscle Cells metabolism, Muscle, Skeletal drug effects, Organ Size drug effects, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation drug effects, Proto-Oncogene Proteins c-akt metabolism, Up-Regulation drug effects, rac1 GTP-Binding Protein metabolism, Biocatalysis drug effects, Homeostasis drug effects, Metabolism, Muscle, Skeletal growth & development, Muscle, Skeletal metabolism, Proto-Oncogene Proteins c-vav metabolism, Signal Transduction drug effects
- Abstract
Skeletal muscle promotes metabolic balance by regulating glucose uptake and the stimulation of multiple interorgan crosstalk. We show here that the catalytic activity of Vav2, a Rho GTPase activator, modulates the signaling output of the IGF1- and insulin-stimulated phosphatidylinositol 3-kinase pathway in that tissue. Consistent with this, mice bearing a Vav2 protein with decreased catalytic activity exhibit reduced muscle mass, lack of proper insulin responsiveness and, at much later times, a metabolic syndrome-like condition. Conversely, mice expressing a catalytically hyperactive Vav2 develop muscle hypertrophy and increased insulin responsiveness. Of note, while hypoactive Vav2 predisposes to, hyperactive Vav2 protects against high fat diet-induced metabolic imbalance. These data unveil a regulatory layer affecting the signaling output of insulin family factors in muscle.
- Published
- 2020
- Full Text
- View/download PDF
20. Case Report: Pure Red Cell Aplasia due to Angioimmunoblastic T-Cell Lymphoma.
- Author
-
Vitorino M, Nunes F, Costa M, Porteiro B, Borges AR, and Machado J
- Abstract
Pure red cell aplasia (PRCA) is a rare bone marrow failure characterized by a progressive normocytic anemia and reticulocytopenia without leukopenia and thrombocytopenia. It can be associated with various hematological disorders but exceedingly rarely with angioimmunoblastic T-cell lymphoma (AITL). We report the case of a 72-year-old woman with PRCA associated with AITL. The patient presented with severe anemia (hemoglobin 2.6 g/dL) and a low reticulocyte count 0.7%. Direct and indirect Coombs tests were positive. A CT scan of the chest, abdomen, and pelvis revealed multiple lymphadenopathies. A cervical lymph node biopsy was compatible with AITL. A bone marrow biopsy showed medullary involvement by AITL and a severe erythroid hypoplasia with a myeloid:erythroid ratio of 19.70. The patient was started on CHOP and after 6 cycles the PET scan confirmed complete remission., Competing Interests: The authors have no conflicts of interest to declare., (Copyright © 2020 by S. Karger AG, Basel.)
- Published
- 2020
- Full Text
- View/download PDF
21. Hypothalamic dopamine signaling regulates brown fat thermogenesis.
- Author
-
Folgueira C, Beiroa D, Porteiro B, Duquenne M, Puighermanal E, Fondevila MF, Barja-Fernández S, Gallego R, Hernández-Bautista R, Castelao C, Senra A, Seoane P, Gómez N, Aguiar P, Guallar D, Fidalgo M, Romero-Pico A, Adan R, Blouet C, Labandeira-García JL, Jeanrenaud F, Kallo I, Liposits Z, Salvador J, Prevot V, Dieguez C, Lopez M, Valjent E, Frühbeck G, Seoane LM, and Nogueiras R
- Subjects
- Animals, Bromocriptine administration & dosage, Bromocriptine pharmacology, Female, Humans, Hypothalamus drug effects, Injections, Intraventricular, Male, Rats, Adipose Tissue, Brown metabolism, Dopamine metabolism, Hypothalamus metabolism, Signal Transduction, Thermogenesis physiology
- Abstract
Dopamine signaling is a crucial part of the brain reward system and can affect feeding behavior. Dopamine receptors are also expressed in the hypothalamus, which is known to control energy metabolism in peripheral tissues. Here we show that pharmacological or chemogenetic stimulation of dopamine receptor 2 (D2R) expressing cells in the lateral hypothalamic area (LHA) and the zona incerta (ZI) decreases body weight and stimulates brown fat activity in rodents in a feeding-independent manner. LHA/ZI D2R stimulation requires an intact sympathetic nervous system and orexin system to exert its action and involves inhibition of PI3K in the LHA/ZI. We further demonstrate that, as early as 3 months after onset of treatment, patients treated with the D2R agonist cabergoline experience an increase in energy expenditure that persists for one year, leading to total body weight and fat loss through a prolactin-independent mechanism. Our results may provide a mechanistic explanation for how clinically used D2R agonists act in the CNS to regulate energy balance., Competing Interests: The authors declare no conflict of interests.
- Published
- 2019
- Full Text
- View/download PDF
22. Uroguanylin Improves Leptin Responsiveness in Diet-Induced Obese Mice.
- Author
-
Folgueira C, Beiroa D, González-Rellán MJ, Porteiro B, Milbank E, Castelao C, García-Palacios M, Casanueva FF, López M, Diéguez C, Seoane LM, and Nogueiras R
- Subjects
- Animals, Diet adverse effects, Hypothalamus metabolism, Mice, Mice, Obese, Obesity etiology, Phosphatidylinositol 3-Kinase metabolism, STAT3 Transcription Factor metabolism, Eating drug effects, Leptin metabolism, Natriuretic Peptides metabolism, Obesity metabolism, Signal Transduction drug effects
- Abstract
The gastrointestinal-brain axis is a key mediator of the body weight and energy homeostasis regulation. Uroguanylin (UGN) has been recently proposed to be a part of this gut-brain axis regulating food intake, body weight and energy expenditure. Expression of UGN is regulated by the nutritional status and dependent on leptin levels. However, the exact molecular mechanisms underlying this UGN-leptin metabolic regulation at a hypothalamic level still remains unclear. Using leptin resistant diet-induced obese (DIO) mice, we aimed to determine whether UGN could improve hypothalamic leptin sensitivity. The present work demonstrates that the central co-administration of UGN and leptin potentiates leptin's ability to decrease the food intake and body weight in DIO mice, and that UGN activates the hypothalamic signal transducer and activator of transcription 3 (STAT3) and phosphatidylinositide 3-kinases (PI3K) pathways. At a functional level, the blockade of PI3K, but not STAT3, blunted UGN-mediated leptin responsiveness in DIO mice. Overall, these findings indicate that UGN improves leptin sensitivity in DIO mice.
- Published
- 2019
- Full Text
- View/download PDF
23. Parabrachial Interleukin-6 Reduces Body Weight and Food Intake and Increases Thermogenesis to Regulate Energy Metabolism.
- Author
-
Mishra D, Richard JE, Maric I, Porteiro B, Häring M, Kooijman S, Musovic S, Eerola K, López-Ferreras L, Peris E, Grycel K, Shevchouk OT, Micallef P, Olofsson CS, Wernstedt Asterholm I, Grill HJ, Nogueiras R, and Skibicka KP
- Subjects
- Adipose Tissue, Brown metabolism, Animals, Astrocytes metabolism, Female, Interleukin-6 genetics, Leptin metabolism, Male, Mice, Mice, Inbred C57BL, Neurons metabolism, Parabrachial Nucleus physiology, Rats, Rats, Sprague-Dawley, Sympathetic Nervous System physiology, Thyroid Hormones metabolism, Body Weight, Eating, Energy Metabolism, Interleukin-6 metabolism, Parabrachial Nucleus metabolism, Thermogenesis
- Abstract
Chronic low-grade inflammation and increased serum levels of the cytokine IL-6 accompany obesity. For brain-produced IL-6, the mechanisms by which it controls energy balance and its role in obesity remain unclear. Here, we show that brain-produced IL-6 is decreased in obese mice and rats in a neuroanatomically and sex-specific manner. Reduced IL-6 mRNA localized to lateral parabrachial nucleus (lPBN) astrocytes, microglia, and neurons, including paraventricular hypothalamus-innervating lPBN neurons. IL-6 microinjection into lPBN reduced food intake and increased brown adipose tissue (BAT) thermogenesis in male lean and obese rats by increasing thyroid and sympathetic outflow to BAT. Parabrachial IL-6 interacted with leptin to reduce feeding. siRNA-mediated reduction of lPBN IL-6 leads to increased weight gain and adiposity, reduced BAT thermogenesis, and increased food intake. Ambient cold exposure partly normalizes the obesity-induced suppression of lPBN IL-6. These results indicate that lPBN-produced IL-6 regulates feeding and metabolism and pinpoints (patho)physiological contexts interacting with lPBN IL-6., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
24. Publisher Correction: Obestatin controls skeletal muscle fiber-type determination.
- Author
-
Santos-Zas I, Cid-Díaz T, González-Sánchez J, Gurriarán-Rodriguez U, Seoane-Mosteiro C, Porteiro B, Nogueiras R, Casabiell X, Relova JL, Gallego R, Mouly V, Pazos Y, and Camiña JP
- Abstract
A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.
- Published
- 2018
- Full Text
- View/download PDF
25. SerpinA3N is a novel hypothalamic gene upregulated by a high-fat diet and leptin in mice.
- Author
-
Sergi D, Campbell FM, Grant C, Morris AC, Bachmair EM, Koch C, McLean FH, Muller A, Hoggard N, de Roos B, Porteiro B, Boekschoten MV, McGillicuddy FC, Kahn D, Nicol P, Benzler J, Mayer CD, Drew JE, Roche HM, Muller M, Nogueiras R, Dieguez C, Tups A, and Williams LM
- Abstract
Background: Energy homeostasis is regulated by the hypothalamus but fails when animals are fed a high-fat diet (HFD), and leptin insensitivity and obesity develops. To elucidate the possible mechanisms underlying these effects, a microarray-based transcriptomics approach was used to identify novel genes regulated by HFD and leptin in the mouse hypothalamus., Results: Mouse global array data identified serpinA3N as a novel gene highly upregulated by both a HFD and leptin challenge. In situ hybridisation showed serpinA3N expression upregulation by HFD and leptin in all major hypothalamic nuclei in agreement with transcriptomic gene expression data. Immunohistochemistry and studies in the hypothalamic clonal neuronal cell line, mHypoE-N42 (N42), confirmed that alpha 1-antichymotrypsin (α
1 AC), the protein encoded by serpinA3 , is localised to neurons and revealed that it is secreted into the media. SerpinA3N expression in N42 neurons is upregulated by palmitic acid and by leptin, together with IL-6 and TNFα , and all three genes are downregulated by the anti-inflammatory monounsaturated fat, oleic acid. Additionally, palmitate upregulation of serpinA3 in N42 neurons is blocked by the NFκB inhibitor, BAY11, and the upregulation of serpinA3N expression in the hypothalamus by HFD is blunted in IL-1 receptor 1 knockout ( IL-1R1-/- ) mice., Conclusions: These data demonstrate that serpinA3 expression is implicated in nutritionally mediated hypothalamic inflammation., Competing Interests: The authors declare that they have no competing interests.Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.- Published
- 2018
- Full Text
- View/download PDF
26. Pharmacological stimulation of p53 with low-dose doxorubicin ameliorates diet-induced nonalcoholic steatosis and steatohepatitis.
- Author
-
Porteiro B, Fondevila MF, Buque X, Gonzalez-Rellan MJ, Fernandez U, Mora A, Beiroa D, Senra A, Gallego R, Fernø J, López M, Sabio G, Dieguez C, Aspichueta P, and Nogueiras R
- Subjects
- Animals, Cell Line, Diet, High-Fat adverse effects, Doxorubicin administration & dosage, Doxorubicin pharmacology, Hep G2 Cells, Humans, Lipid Metabolism drug effects, Liver drug effects, Liver metabolism, Mice, Mice, Inbred C57BL, Non-alcoholic Fatty Liver Disease etiology, Topoisomerase II Inhibitors administration & dosage, Topoisomerase II Inhibitors pharmacology, Tumor Suppressor Protein p53 genetics, Doxorubicin therapeutic use, Non-alcoholic Fatty Liver Disease drug therapy, Topoisomerase II Inhibitors therapeutic use, Tumor Suppressor Protein p53 metabolism
- Abstract
Objective: Recent reports have implicated the p53 tumor suppressor in the regulation of lipid metabolism. We hypothesized that the pharmacological activation of p53 with low-dose doxorubicin, which is widely used to treat several types of cancer, may have beneficial effects on nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH)., Methods: We used long-term pharmacological activation of p53 by i.p. or oral administration of low-dose doxorubicin in different animal models of NAFLD (high fat diet containing 45% and 60% kcal fat) and NASH (methionine- and choline-deficient diet and choline deficiency combined with high fat diet). We also administered doxorubicin in mice lacking p53 in the liver and in two human hepatic cells lines (HepG2 and THLE2)., Results: The attenuation of liver damage was accompanied by the stimulation of fatty acid oxidation and decrease of lipogenesis, inflammation, and ER stress. The effects of doxorubicin were abrogated in mice with liver-specific ablation of p53. Finally, the effects of doxorubicin on lipid metabolism found in animal models were also present in two human hepatic cells lines, in which the drug stimulated fatty acid oxidation and inhibited de novo lipogenesis at doses that did not cause changes in apoptosis or cell viability., Conclusion: These data provide new evidence for targeting p53 as a strategy to treat liver disease., (Copyright © 2017 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
27. The MST3/STK24 kinase mediates impaired fasting blood glucose after a high-fat diet.
- Author
-
Iglesias C, Floridia E, Sartages M, Porteiro B, Fraile M, Guerrero A, Santos D, Cuñarro J, Tovar S, Nogueiras R, Pombo CM, and Zalvide J
- Subjects
- Animals, Diabetes Mellitus, Type 2 metabolism, Diet, High-Fat adverse effects, Fasting blood, Female, Gluconeogenesis physiology, Hep G2 Cells, Humans, Insulin Resistance physiology, Male, Mice, Mice, Inbred C57BL, Protein Serine-Threonine Kinases genetics, Blood Glucose metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Aims/hypothesis: The identification of mediators in the pathogenesis of type 2 diabetes mellitus is essential for the full understanding of this disease. Protein kinases are especially important because of their potential as pharmacological targets. The goal of this study was to investigate whether mammalian sterile-20 3 (MST3/STK24), a stress-regulated kinase, is involved in metabolic alterations in obesity., Methods: Glucose regulation of Mst3 (also known as Stk24)-knockout mice was analysed both in 129;C57 mixed background mice and in C57/BL6J mice fed normally or with a high-fat diet (HFD). This work was complemented with an analysis of the insulin signalling pathway in cultured human liver cells made deficient in MST3 using RNA interference., Results: MST3 is phosphorylated in the livers of mice subject to an obesity-promoting HFD, and its deficiency lowers the hyperglycaemia, hyperinsulinaemia and insulin resistance that the animals develop with this diet, an effect that is seen even without complete inactivation of the kinase. Lack of MST3 results in activation of the insulin signalling pathway downstream of IRS1, in both cultured liver cells and the liver of animals after HFD. This effect increases the inhibition of forkhead box (FOX)O1, with subsequent downregulation of the expression of gluconeogenic enzymes., Conclusions/interpretation: MST3 inhibits the insulin signalling pathway and is important in the development of insulin resistance and impaired blood glucose levels after an HFD.
- Published
- 2017
- Full Text
- View/download PDF
28. Hypothalamic pathways regulate the anorectic action of p-chloro-diphenyl diselenide in rats.
- Author
-
Bortolatto CF, Nogueira CW, Porteiro B, Imbernón M, and Nogueiras R
- Subjects
- Adipose Tissue, Brown drug effects, Adipose Tissue, Brown metabolism, Adipose Tissue, White drug effects, Adipose Tissue, White metabolism, Animals, Anorexia blood, Anorexia psychology, Body Composition drug effects, Body Weight drug effects, Eating drug effects, Hypothalamus metabolism, Liver drug effects, Liver metabolism, Male, Rats, Rats, Sprague-Dawley, Satiety Response drug effects, Time Factors, Triglycerides blood, Anorexia chemically induced, Anorexia pathology, Hypothalamus drug effects, Hypothalamus pathology, Organoselenium Compounds adverse effects
- Abstract
Behavioral studies have suggested that (p-ClPhSe)
2 elicits an anorectic-like action in rats by inducing multiple effects such as satiety-enhancing effect, malaise and specific flavor; however, the molecular mechanisms underlying its anorexigenic action remain unclarified. Here, male Sprague-Dawley rats received acute and sub-chronic intraperitoneal treatments with (p-ClPhSe)2 ; thereafter, in vivo and ex vivo analyses were carried out. The present study reveals that the reduction of food intake resulting from a single treatment with (p-ClPhSe)2 (1mg/kg, i.p.) was associated with decreased hypothalamic levels of pro-melanin-concentrating hormone (pro-MCH) and orexin precursor. In addition, repeated administrations of (p-ClPhSe)2 (10mg/kg; i.p.) for 7 days induced sustained food intake suppression, body weight loss and white fat reduction. Measurements of brown adipose tissue content and temperature as well as data obtained from a pair-fed group indicated that the effects of (p-ClPhSe)2 on the body weight are closely related to its anorexigenic actions, ruling out the possibility of increased thermogenesis. Furthermore, (p-ClPhSe)2 reduced the hypothalamic orexin precursor levels when repeatedly administered to rats. Sub-chronic treatment with (p-ClPhSe)2 caused a decrease of serum triglyceride levels and down-regulation of hepatic cholesterol content. Therefore, the current study characterized the anorectic and reducing body weight actions of (p-ClPhSe)2 in Sprague-Dawley rats. Besides, the set of results suggests that food intake suppressant effects triggered after (p-ClPhSe)2 administration to rats are mainly related with the lower orexin levels in hypothalamus after acute and sub-chronic treatments., (Copyright © 2017 Elsevier B.V. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
29. Corrigendum: Hepatic p63 regulates steatosis via IKKβ/ER stress.
- Author
-
Porteiro B, Fondevila MF, Delgado TC, Iglesias C, Imbernon M, Iruzubieta P, Crespo J, Zabala-Letona A, Fernø J, González-Terán B, Matesanz N, Hernández-Cosido L, Marcos M, Tovar S, Vidal A, Sánchez-Ceinos J, Malagon MM, Pombo C, Zalvide J, Carracedo A, Buque X, Dieguez C, Sabio G, López M, Aspichueta P, Martínez-Chantar ML, and Nogueiras R
- Abstract
This corrects the article DOI: 10.1038/ncomms15111.
- Published
- 2017
- Full Text
- View/download PDF
30. Obestatin controls skeletal muscle fiber-type determination.
- Author
-
Santos-Zas I, Cid-Díaz T, González-Sánchez J, Gurriarán-Rodriguez U, Seoane-Mosteiro C, Porteiro B, Nogueiras R, Casabiell X, Relova JL, Gallego R, Mouly V, Pazos Y, and Camiña JP
- Subjects
- Animals, Cell Line, MEF2 Transcription Factors metabolism, Male, Mice, Muscle Fibers, Skeletal cytology, Muscle Fibers, Skeletal drug effects, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Proto-Oncogene Proteins c-akt metabolism, Rats, Receptors, G-Protein-Coupled metabolism, Signal Transduction, TOR Serine-Threonine Kinases metabolism, Ghrelin pharmacology, Muscle Development drug effects, Muscle Fibers, Skeletal metabolism
- Abstract
Obestatin/GPR39 signaling stimulates skeletal muscle growth and repair by inducing both G-protein-dependent and -independent mechanisms linking the activated GPR39 receptor with distinct sets of accessory and effector proteins. In this work, we describe a new level of activity where obestatin signaling plays a role in the formation, contractile properties and metabolic profile of skeletal muscle through determination of oxidative fiber type. Our data indicate that obestatin regulates Mef2 activity and PGC-1α expression. Both mechanisms result in a shift in muscle metabolism and function. The increase in Mef2 and PGC-1α signaling activates oxidative capacity, whereas Akt/mTOR signaling positively regulates myofiber growth. Taken together, these data indicate that the obestatin signaling acts on muscle fiber-type program in skeletal muscle.
- Published
- 2017
- Full Text
- View/download PDF
31. Hepatic p63 regulates steatosis via IKKβ/ER stress.
- Author
-
Porteiro B, Fondevila MF, Delgado TC, Iglesias C, Imbernon M, Iruzubieta P, Crespo J, Zabala-Letona A, Fernø J, González-Terán B, Matesanz N, Hernández-Cosido L, Marcos M, Tovar S, Vidal A, Sánchez-Ceinos J, Malagon MM, Pombo C, Zalvide J, Carracedo A, Buque X, Dieguez C, Sabio G, López M, Aspichueta P, Martínez-Chantar ML, and Nogueiras R
- Subjects
- Adult, Animals, Fatty Liver genetics, Fatty Liver physiopathology, Female, Hepatocytes metabolism, Humans, I-kappa B Kinase genetics, Lipid Metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Phosphoproteins genetics, Phosphoproteins metabolism, Trans-Activators genetics, Trans-Activators metabolism, Transcription Factors genetics, Tumor Suppressor Proteins genetics, X-Box Binding Protein 1 genetics, X-Box Binding Protein 1 metabolism, Endoplasmic Reticulum Stress, Fatty Liver metabolism, I-kappa B Kinase metabolism, Liver metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- Abstract
p53 family members control several metabolic and cellular functions. The p53 ortholog p63 modulates cellular adaptations to stress and has a major role in cell maintenance and proliferation. Here we show that p63 regulates hepatic lipid metabolism. Mice with liver-specific p53 deletion develop steatosis and show increased levels of p63. Down-regulation of p63 attenuates liver steatosis in p53 knockout mice and in diet-induced obese mice, whereas the activation of p63 induces lipid accumulation. Hepatic overexpression of N-terminal transactivation domain TAp63 induces liver steatosis through IKKβ activation and the induction of ER stress, the inhibition of which rescues the liver functions. Expression of TAp63, IKKβ and XBP1s is also increased in livers of obese patients with NAFLD. In cultured human hepatocytes, TAp63 inhibition protects against oleic acid-induced lipid accumulation, whereas TAp63 overexpression promotes lipid storage, an effect reversible by IKKβ silencing. Our findings indicate an unexpected role of the p63/IKKβ/ER stress pathway in lipid metabolism and liver disease.
- Published
- 2017
- Full Text
- View/download PDF
32. Obesity- and gender-dependent role of endogenous somatostatin and cortistatin in the regulation of endocrine and metabolic homeostasis in mice.
- Author
-
Luque RM, Cordoba-Chacon J, Pozo-Salas AI, Porteiro B, de Lecea L, Nogueiras R, Gahete MD, and Castaño JP
- Subjects
- Animals, Dietary Fats pharmacology, Disease Models, Animal, Female, Male, Mice, Mice, Knockout, Neuropeptides genetics, Obesity chemically induced, Obesity genetics, Obesity pathology, Pituitary Gland pathology, Somatostatin genetics, Dietary Fats adverse effects, Homeostasis, Neuropeptides metabolism, Obesity metabolism, Pituitary Gland metabolism, Sex Characteristics, Somatostatin metabolism
- Abstract
Somatostatin (SST) and cortistatin (CORT) regulate numerous endocrine secretions and their absence [knockout (KO)-models] causes important endocrine-metabolic alterations, including pituitary dysregulations. We have demonstrated that the metabolic phenotype of single or combined SST/CORT KO-models is not drastically altered under normal conditions. However, the biological actions of SST/CORT are conditioned by the metabolic-status (e.g. obesity). Therefore, we used male/female SST- and CORT-KO mice fed low-fat (LF) or high-fat (HF) diet to explore the interplay between SST/CORT and obesity in the control of relevant pituitary-axes and whole-body metabolism. Our results showed that the SST/CORT role in the control of GH/prolactin secretions is maintained under LF- and HF-diet conditions as SST-KOs presented higher GH/prolactin-levels, while CORT-KOs displayed higher GH- and lower prolactin-levels than controls under both diets. Moreover, the impact of lack of SST/CORT on the metabolic-function was gender- and diet-dependent. Particularly, SST-KOs were more sensitive to HF-diet, exhibiting altered growth and body-composition (fat/lean percentage) and impaired glucose/insulin-metabolism, especially in males. Conversely, only males CORT-KO under LF-diet conditions exhibited significant alterations, displaying higher glucose-levels and insulin-resistance. Altogether, these data demonstrate a tight interplay between SST/CORT-axis and the metabolic status in the control of endocrine/metabolic functions and unveil a clear dissociation of SST/CORT roles.
- Published
- 2016
- Full Text
- View/download PDF
33. Hypothalamic kappa opioid receptor mediates both diet-induced and melanin concentrating hormone-induced liver damage through inflammation and endoplasmic reticulum stress.
- Author
-
Imbernon M, Sanchez-Rebordelo E, Romero-Picó A, Kalló I, Chee MJ, Porteiro B, Al-Massadi O, Contreras C, Fernø J, Senra A, Gallego R, Folgueira C, Seoane LM, van Gestel M, Adan RA, Liposits Z, Dieguez C, López M, and Nogueiras R
- Subjects
- Animals, Inflammation complications, Inflammation etiology, Mice, Mice, Inbred C57BL, Rats, Rats, Sprague-Dawley, Diet, Endoplasmic Reticulum Stress, Hypothalamic Hormones physiology, Hypothalamus physiology, Liver Diseases etiology, Melanins physiology, Pituitary Hormones physiology, Receptors, Opioid, kappa physiology
- Abstract
Unlabelled: The opioid system is widely known to modulate the brain reward system and thus affect the behavior of humans and other animals, including feeding. We hypothesized that the hypothalamic opioid system might also control energy metabolism in peripheral tissues. Mice lacking the kappa opioid receptor (κOR) and adenoviral vectors overexpressing or silencing κOR were stereotaxically delivered in the lateral hypothalamic area (LHA) of rats. Vagal denervation was performed to assess its effect on liver metabolism. Endoplasmic reticulum (ER) stress was inhibited by pharmacological (tauroursodeoxycholic acid) and genetic (overexpression of the chaperone glucose-regulated protein 78 kDa) approaches. The peripheral effects on lipid metabolism were assessed by histological techniques and western blot. We show that in the LHA κOR directly controls hepatic lipid metabolism through the parasympathetic nervous system, independent of changes in food intake and body weight. κOR colocalizes with melanin concentrating hormone receptor 1 (MCH-R1) in the LHA, and genetic disruption of κOR reduced melanin concentrating hormone-induced liver steatosis. The functional relevance of these findings was given by the fact that silencing of κOR in the LHA attenuated both methionine choline-deficient, diet-induced and choline-deficient, high-fat diet-induced ER stress, inflammation, steatohepatitis, and fibrosis, whereas overexpression of κOR in this area promoted liver steatosis. Overexpression of glucose-regulated protein 78 kDa in the liver abolished hypothalamic κOR-induced steatosis by reducing hepatic ER stress., Conclusions: This study reveals a novel hypothalamic-parasympathetic circuit modulating hepatic function through inflammation and ER stress independent of changes in food intake or body weight; these findings might have implications for the clinical use of opioid receptor antagonists. (Hepatology 2016;64:1086-1104)., (© 2016 The Authors. (Hepatology published by Wiley Periodicals, Inc., on behalf of the American Association for the Study of Liver Diseases.)
- Published
- 2016
- Full Text
- View/download PDF
34. Pharmacological and Genetic Manipulation of p53 in Brown Fat at Adult But Not Embryonic Stages Regulates Thermogenesis and Body Weight in Male Mice.
- Author
-
Al-Massadi O, Porteiro B, Kuhlow D, Köhler M, Gonzalez-Rellan MJ, Garcia-Lavandeira M, Díaz-Rodríguez E, Quiñones M, Senra A, Alvarez CV, López M, Diéguez C, Schulz TJ, and Nogueiras R
- Subjects
- Adipose Tissue, Brown metabolism, Animals, Body Composition drug effects, Body Composition genetics, Body Weight genetics, Cell Line, Doxorubicin pharmacology, Male, Mice, Mice, Knockout, Obesity metabolism, Rats, Somatotrophs cytology, Somatotrophs drug effects, Somatotrophs metabolism, Thermogenesis genetics, Tumor Suppressor Protein p53 agonists, Tumor Suppressor Protein p53 metabolism, Adipose Tissue, Brown drug effects, Body Weight drug effects, Obesity genetics, Thermogenesis drug effects, Tumor Suppressor Protein p53 genetics
- Abstract
p53 is a well-known tumor suppressor that plays multiple biological roles, including the capacity to modulate metabolism at different levels. However, its metabolic role in brown adipose tissue (BAT) remains largely unknown. Herein we sought to investigate the physiological role of endogenous p53 in BAT and its implication on BAT thermogenic activity and energy balance. To this end, we generated and characterized global p53-null mice and mice lacking p53 specifically in BAT. Additionally we performed gain-and-loss-of-function experiments in the BAT of adult mice using virogenetic and pharmacological approaches. BAT was collected and analyzed by immunohistochemistry, thermography, real-time PCR, and Western blot. p53-deficient mice were resistant to diet-induced obesity due to increased energy expenditure and BAT activity. However, the deletion of p53 in BAT using a Myf5-Cre driven p53 knockout did not show any changes in body weight or the expression of thermogenic markers. The acute inhibition of p53 in the BAT of adult mice slightly increased body weight and inhibited BAT thermogenesis, whereas its overexpression in the BAT of diet-induced obese mice reduced body weight and increased thermogenesis. On the other hand, pharmacological activation of p53 improves body weight gain due to increased BAT thermogenesis by sympathetic nervous system in obese adult wild-type mice but not in p53(-/-) animals. These results reveal that p53 regulates BAT metabolism by coordinating body weight and thermogenesis, but these metabolic actions are tissue specific and also dependent on the developmental stage.
- Published
- 2016
- Full Text
- View/download PDF
35. Ghrelin requires p53 to stimulate lipid storage in fat and liver.
- Author
-
Porteiro B, Díaz-Ruíz A, Martínez G, Senra A, Vidal A, Serrano M, Gualillo O, López M, Malagón MM, Diéguez C, and Nogueiras R
- Subjects
- Adipose Tissue, White enzymology, Adipose Tissue, White pathology, Adiposity, Animals, Crosses, Genetic, Female, Gene Expression Profiling, Ghrelin administration & dosage, Injections, Intraperitoneal, Liver enzymology, Liver pathology, Male, Mice, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Obesity blood, Obesity etiology, Obesity metabolism, Obesity pathology, Tissue Culture Techniques, Triglycerides blood, Triglycerides metabolism, Tumor Suppressor Protein p53 genetics, Weight Gain, Adipogenesis, Adipose Tissue, White metabolism, Enzyme Induction, Ghrelin metabolism, Lipogenesis, Liver metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Ghrelin, a stomach-derived peptide, stimulates feeding behavior and adiposity. For its orexigenic action, ghrelin triggers a central SIRT1/p53/AMPK pathway. The tumor suppressor p53 also plays an important role in white adipose tissue (WAT), where it is up-regulated in the adipocytes of obese mice. It is not known, however, whether p53 has any role in mediating the peripheral action of ghrelin. In the present study, chronic peripheral ghrelin treatment resulted in increased body weight and fat-mass gain in wild-type mice. Correspondingly, mRNA levels of several adipogenic and fat-storage-promoting enzymes were up-regulated in WAT, whereas hepatic triglyceride content and lipogenic enzymes were also increased in wild-type mice following ghrelin treatment. In contrast, mice lacking p53 failed to respond to ghrelin treatment, with their body weight, fat mass, and adipocyte and hepatic metabolism remaining unchanged. Thus, our results show that p53 is necessary for the actions of ghrelin on WAT and liver, leading to changes in expression levels of lipogenic and adipogenic genes, and modifying body weight.
- Published
- 2013
- Full Text
- View/download PDF
36. Central melanin-concentrating hormone influences liver and adipose metabolism via specific hypothalamic nuclei and efferent autonomic/JNK1 pathways.
- Author
-
Imbernon M, Beiroa D, Vázquez MJ, Morgan DA, Veyrat-Durebex C, Porteiro B, Díaz-Arteaga A, Senra A, Busquets S, Velásquez DA, Al-Massadi O, Varela L, Gándara M, López-Soriano FJ, Gallego R, Seoane LM, Argiles JM, López M, Davis RJ, Sabio G, Rohner-Jeanrenaud F, Rahmouni K, Dieguez C, and Nogueiras R
- Subjects
- Adipocytes drug effects, Adipose Tissue drug effects, Animals, Eating, Fatty Acids metabolism, Fatty Liver metabolism, Fatty Liver physiopathology, Hypothalamic Area, Lateral drug effects, Hypothalamic Hormones administration & dosage, Lipid Metabolism drug effects, Lipid Metabolism physiology, Lipogenesis drug effects, Lipogenesis physiology, Liver drug effects, Male, Melanins administration & dosage, Mice, Non-alcoholic Fatty Liver Disease, Pituitary Hormones administration & dosage, Rats, Rats, Sprague-Dawley, Receptors, Pituitary Hormone agonists, Receptors, Pituitary Hormone physiology, Vagus Nerve drug effects, Vagus Nerve physiology, Vagus Nerve physiopathology, Adipocytes metabolism, Adipose Tissue metabolism, Adiposity physiology, Hypothalamic Area, Lateral physiology, Hypothalamic Hormones physiology, Liver metabolism, Melanins physiology, Mitogen-Activated Protein Kinase 8 metabolism, Pituitary Hormones physiology
- Abstract
Background & Aims: Specific neuronal circuits modulate autonomic outflow to liver and white adipose tissue. Melanin-concentrating hormone (MCH)-deficient mice are hypophagic, lean, and do not develop hepatosteatosis when fed a high-fat diet. Herein, we sought to investigate the role of MCH, an orexigenic neuropeptide specifically expressed in the lateral hypothalamic area, on hepatic and adipocyte metabolism., Methods: Chronic central administration of MCH and adenoviral vectors increasing MCH signaling were performed in rats and mice. Vagal denervation was performed to assess its effect on liver metabolism. The peripheral effects on lipid metabolism were assessed by real-time polymerase chain reaction and Western blot., Results: We showed that the activation of MCH receptors promotes nonalcoholic fatty liver disease through the parasympathetic nervous system, whereas it increases fat deposition in white adipose tissue via the suppression of sympathetic traffic. These metabolic actions are independent of parallel changes in food intake and energy expenditure. In the liver, MCH triggers lipid accumulation and lipid uptake, with c-Jun N-terminal kinase being an essential player, whereas in adipocytes MCH induces metabolic pathways that promote lipid storage and decreases lipid mobilization. Genetic activation of MCH receptors or infusion of MCH specifically in the lateral hypothalamic area modulated hepatic lipid metabolism, whereas the specific activation of this receptor in the arcuate nucleus affected adipocyte metabolism., Conclusions: Our findings show that central MCH directly controls hepatic and adipocyte metabolism through different pathways., (Copyright © 2013 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
37. Female Nur77-deficient mice show increased susceptibility to diet-induced obesity.
- Author
-
Perez-Sieira S, Martinez G, Porteiro B, Lopez M, Vidal A, Nogueiras R, and Dieguez C
- Subjects
- Adipose Tissue, White metabolism, Adipose Tissue, White pathology, Adiposity, Animals, Apoptosis Regulatory Proteins metabolism, Blood Glucose metabolism, Body Weight, Cholesterol blood, Disease Susceptibility, Energy Metabolism, Fatty Acids, Nonesterified blood, Female, Gene Knockout Techniques, Homeostasis, Insulin blood, Leptin blood, Lipolysis, Liver metabolism, Male, Mice, Muscles metabolism, Nuclear Receptor Subfamily 4, Group A, Member 1 genetics, Obesity metabolism, Obesity pathology, Signal Transduction, Triglycerides blood, Diet, High-Fat adverse effects, Nuclear Receptor Subfamily 4, Group A, Member 1 deficiency, Obesity etiology, Obesity genetics
- Abstract
Adipose tissue is essential in the regulation of body weight. The key process in fat catabolism and the provision of energy substrate during times of nutrient deprivation or enhanced energy demand is the hydrolysis of triglycerides and the release of fatty acids and glycerol. Nur77 is a member of the NR4A subfamily of nuclear receptors that plays an important metabolic role, modulating hepatic glucose metabolism and lipolysis in muscle. However, its endogenous role on white adipose tissue, as well as the gender dependency of these mechanisms, remains largely unknown. Male and female wild type and Nur77 deficient mice were fed with a high fat diet (45% calories from fat) for 4 months. Mice were analyzed in vivo with the indirect calorimetry system, and tissues were analyzed by real-time PCR and Western blot analysis. Female, but not male Nur77 deficient mice, gained more weight and fat mass when compared to wild type mice fed with high fat diet, which can be explained by decreased energy expenditure. The lack of Nur77 also led to a decreased pHSL/HSL ratio in white adipose tissue and increased expression of CIDEA in brown adipose tissue of female Nur77 deficient mice. Overall, these findings suggest that Nur77 is an important physiological modulator of lipid metabolism in adipose tissue and that there are gender differences in the sensitivity to deletion of the Nur77 signaling. The decreased energy expenditure and the actions of Nur77 on liver, muscle, brown and white adipose tissue contribute to the increased susceptibility to diet-induced obesity in females lacking Nur77.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.