1
|
Cicuéndez B, Mora A, López JA, Curtabbi A, Pérez-García J, Porteiro B, Jimenez-Blasco D, Latorre-Muro P, Vo P, Jerome M, Gómez-Santos B, Romero-Becerra R, Leiva M, Rodríguez E, León M, Leiva-Vega L, Gómez-Lado N, Torres JL, Hernández-Cosido L, Aguiar P, Marcos M, Jastroch M, Daiber A, Aspichueta P, Bolaños JP, Spinelli JB, Puigserver P, Enriquez JA, Vázquez J, Folgueira C, Sabio G. Absence of MCJ/DnaJC15 promotes brown adipose tissue thermogenesis. Nat Commun 2025; 16:229. [PMID: 39805849 PMCID: PMC11730624 DOI: 10.1038/s41467-024-54353-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
Obesity poses a global health challenge, demanding a deeper understanding of adipose tissue (AT) and its mitochondria. This study describes the role of the mitochondrial protein Methylation-controlled J protein (MCJ/DnaJC15) in orchestrating brown adipose tissue (BAT) thermogenesis. Here we show how MCJ expression decreases during obesity, as evident in human and mouse adipose tissue samples. MCJKO mice, even without UCP1, a fundamental thermogenic protein, exhibit elevated BAT thermogenesis. Electron microscopy unveils changes in mitochondrial morphology resembling BAT activation. Proteomic analysis confirms these findings and suggests involvement of the eIF2α mediated stress response. The pivotal role of eIF2α is scrutinized by in vivo CRISPR deletion of eIF2α in MCJKO mice, abrogating thermogenesis. These findings uncover the importance of MCJ as a regulator of BAT thermogenesis, presenting it as a promising target for obesity therapy.
Collapse
Grants
- K99 DK133502 NIDDK NIH HHS
- R01 DK136640 NIDDK NIH HHS
- This work has been supported by the following projects: PMP21/00057 funded by the Instituto de Salud Carlos III (ISCIII) - European Union (FEDER/FSE) "Una manera de hacer Europa"/ "El FSE invierte en tu futuro"/ Next Generation EU and cofunded by the European Union / Plan de Recuperación, Transformación y Resiliencia (PRTR); PID2022-138525OB-I00 de la Agencia Estatal de Investigación 10.13039/501100011033, financiado por MICIU/AEI/10.13039/501100011033 fondos FEDER and EU, PDC2021-121147-I00 and PID2019-104399RB-I00 funded by MCIN/AEI/10.13039/501100011033 and the European Union “NextGenerationEU”/Plan de Recuperación Transformación y Resiliencia -PRTR; Grant RED2022-134397-T funded by MCIN/AEI/10.13039/501100011033 and, as appropriate, by “ERDF A way of making Europe”, by the “European Union” or by the “European Union NextGenerationEU/PRTR”; Fundación Jesús Serra; EFSD/Lilly Dr Sabio; 2017 Leonardo Grant BBVA Foundation (Investigadores-BBVA-2017); Comunidad de Madrid IMMUNOTHERCAN-CM S2010/BMD-2326 and B2017/BMD-373; Fundación AECC PROYE19047SABI, PGC2018-097019-B-I00 and PT17/0019/0003- ISCIII-SGEFI /ERDF, ProteoRed. PreMed-Exp: PMP21/00057, PMP21/00113 Infraestructura de Medicina de Precisión asociada a la Ciencia y Tecnología IMPACT-2021 Instituto de Salud Carlos III (GS, JLT).. G.S is a Miembro Numerario of the RACVE. The CNIC is supported by the Instituto de Salud Carlos III (ISCIII), the Ministerio de Ciencia e Innovación (MCIN) and the Pro CNIC Foundation) and is a Severo Ochoa Center of Excellence (grant CEX2020-001041-S funded by MICIN/AEI/10.13039/501100011033).
- A.C was supported by the European Union's Horizon 2020 research and 328 innovation program under the Marie Skłodowska-Curie grant agreement n. 713,673.
- J.P-G was supported by the fellowship from” la Caixa” Foundation (ID 100010434), the fellowship code is LCF/BQ/DR24/12080018.
- M.M is supported by Instituto de Salud Carlos III (ISCIII) and the European Union project PI20/00743.
- P.A is supported by MCIU/AEI/FEDER, UE (PID2021-124425OB-I00) and Basque Government, Department of Education (IT1476-22).
- J.P.B is funded by AEI grants PID2019-105699RB-I00, PID2022-138813OB-I00 and PDC2021-121013-I00; HORIZON-MSCA-2021-DN-01grant 101072759; and La Caixa Research Health grant HR23-00793.
- C.F was funded with Sara Borrell (CD19/ 00078), NNF23SA0083952-EASO/Novo Nordisk New Investigator Award in Basic Sciences 2023, EFSD/Lilly Young Investigator Award 2022, Society for Endocrinology/Early Career Grant 2022, FSEEN/ Jóvenes endocrinólogos 2022, EFSD/Novo Nordisk Rising Star 2024, IBSA Foundation Fellowship Endocrinology 2023.
Collapse
Affiliation(s)
- Beatriz Cicuéndez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Juan Antonio López
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Andrea Curtabbi
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Pérez-García
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Begoña Porteiro
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Jimenez-Blasco
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
| | - Pedro Latorre-Muro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Paula Vo
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Madison Jerome
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Beatriz Gómez-Santos
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU. Leioa, Biobizkaia Health Research Institute, Barakaldo, Spain
| | | | - Magdalena Leiva
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Elena Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Marta León
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Noemi Gómez-Lado
- Molecular Imaging Biomarkers and Theragnosis Lab, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). University of Santiago de Compostela (USC), Santiago de Compostela, Spain
- Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Nuclear Medicine Service, University Clinical Hospital of Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | | | - Lourdes Hernández-Cosido
- Bariatric Surgery Unit. Department of General Surgery, University Hospital of Salamanca. Department of Surgery. University of Salamanca, Salamanca, Spain
| | - Pablo Aguiar
- Molecular Imaging Biomarkers and Theragnosis Lab, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). University of Santiago de Compostela (USC), Santiago de Compostela, Spain
- Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Nuclear Medicine Service, University Clinical Hospital of Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca-IBSAL, Salamanca, Spain
- Department of Medicine. University of Salamanca, Salamanca, Spain
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Andreas Daiber
- Department of Cardiology 1, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU. Leioa, Biobizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red sobre enfermedades hepáticas y digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Pedro Bolaños
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
| | - Jessica B Spinelli
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA
- UMass Chan Medical School Cancer Center, Worcester, MA, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - José Antonio Enriquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Cintia Folgueira
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain.
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain.
| |
Collapse
|
2
|
Ratko M, Crljen V, Tkalčić M, Mažuranić A, Bubalo P, Škavić P, Banovac I, Dugandžić A. Expression of guanylate cyclase C in human prefrontal cortex depends on sex and feeding status. Front Mol Neurosci 2024; 17:1361089. [PMID: 38840774 PMCID: PMC11150535 DOI: 10.3389/fnmol.2024.1361089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction Guanylate cyclase C (GC-C) has been detected in the rodent brain in neurons of the cerebral cortex, amygdala, midbrain, hypothalamus, and cerebellum. Methods In this study we determined GC-C protein expression in Brodmann areas (BA) 9, BA10, BA11, and BA32 of the human prefrontal cortex involved in regulation of feeding behavior, as well as in the cerebellar cortex, arcuate nucleus of hypothalamus and substantia nigra in brain samples of human 21 male and 13 female brains by ELISA with postmortem delay < 24 h. Results GC-C was found in all tested brain areas and it was expressed in neurons of the third cortical layer of BA9. The regulation of GC-C expression by feeding was found in male BA11 and BA10-M, where GC-C expression was in negative correlation to the volume of stomach content during autopsy. In female BA11 there was no correlation detected, while in BA10-M there was even positive correlation. This suggests sex differences in GC-C expression regulation in BA11 and BA10-M. The amount of GC-C was higher in female BA9 only when the death occurred shortly after a meal, while expression of GC-C was higher in BA10-O only when the stomach was empty. The expression of GC-C in female hypothalamus was lower when compared to male hypothalamus only when the stomach was full, suggesting possibly lower satiety effects of GC-C agonists in women. Discussion These results point toward the possible role of GC-C in regulation of feeding behavior. Since, this is first study of GC-C regulation and its possible function in prefrontal cortex, to determine exact role of GC-C in different region of prefrontal cortex, especially in humans, need further studies.
Collapse
Affiliation(s)
- Martina Ratko
- Laboratory for Cellular Neurophysiology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Vladiana Crljen
- Laboratory for Cellular Neurophysiology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Physiology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Martina Tkalčić
- Institute for Forensic Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Anton Mažuranić
- Institute for Forensic Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Pero Bubalo
- Institute for Forensic Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Petar Škavić
- Institute for Forensic Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivan Banovac
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Aleksandra Dugandžić
- Laboratory for Cellular Neurophysiology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Physiology, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
3
|
Cicuéndez B, Pérez-García J, Folgueira C. A Combination of a Dopamine Receptor 2 Agonist and a Kappa Opioid Receptor Antagonist Synergistically Reduces Weight in Diet-Induced Obese Rodents. Nutrients 2024; 16:424. [PMID: 38337707 PMCID: PMC10857008 DOI: 10.3390/nu16030424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
As the global obesity rate increases, so does the urgency to find effective anti-obesity drugs. In the search for therapeutic targets, central nervous system (CNS) mechanisms engaged in the regulation of energy expenditure and food intake, such as the opioid and dopamine systems, are crucial. In this study, we examined the effect on body weight of two drugs: bromocriptine (BC), a D2R receptor agonist, and PF-04455242, a selective κ opioid receptor (KOR) antagonist. Using diet-induced obese (DIO) rats, we aimed to ascertain whether the administration of BC and PF-04455242, independently or in combination, could enhance body weight loss. Furthermore, the present work demonstrates that the peripheral coadministration of BC and PF-04455242 enhances the reduction of weight in DIO rats and leads to a decrease in adiposity in a food-intake-independent manner. These effects were based on heightened energy expenditure, particularly through the activation of brown adipose tissue (BAT) thermogenesis. Overall, our findings indicate that the combination of BC and PF-04455242 effectively induces body weight loss through increased energy expenditure by increasing thermogenic activity and highlight the importance of the combined use of drugs to combat obesity.
Collapse
Affiliation(s)
| | | | - Cintia Folgueira
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (B.C.); (J.P.-G.)
| |
Collapse
|
4
|
Otero A, Becerril S, Martín M, Cienfuegos JA, Valentí V, Moncada R, Catalán V, Gómez-Ambrosi J, Burrell MA, Frühbeck G, Rodríguez A. Effect of guanylin peptides on pancreas steatosis and function in experimental diet-induced obesity and after bariatric surgery. Front Endocrinol (Lausanne) 2023; 14:1185456. [PMID: 37274331 PMCID: PMC10233012 DOI: 10.3389/fendo.2023.1185456] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/04/2023] [Indexed: 06/06/2023] Open
Abstract
Introduction Obesity contributes to ectopic fat deposition in non-adipose organs, including the pancreas. Pancreas steatosis associates with inflammation and β-cell dysfunction, contributing to the onset of insulin resistance and type 2 diabetes. An improvement of pancreatic steatosis and indices of insulin resistance is observed following bariatric surgery, but the underlying mechanisms remain unknown. We sought to analyze whether guanylin (GUCA2A) and uroguanylin (GUCA2B), two gut hormones involved in the regulation of satiety, food preference and adiposity, are involved in the amelioration of pancreas fat accumulation after bariatric surgery. Methods Pancreas steatosis, inflammation, islet number and area were measured in male Wistar rats with diet-induced obesity (n=125) subjected to surgical (sham operation and sleeve gastrectomy) or dietary (pair-fed to the amount of food eaten by gastrectomized animals) interventions. The tissue distribution of guanylate cyclase C (GUCY2C) and the expression of the guanylin system were evaluated in rat pancreata by real-time PCR, Western-blot and immunohistochemistry. The effect of guanylin and uroguanylin on factors involved in insulin secretion and lipogenesis was determined in vitro in RIN-m5F β-cells exposed to lipotoxic conditions. Results Sleeve gastrectomy reduced pancreas steatosis and inflammation and improved insulin sensitivity and synthesis. An upregulation of GUCA2A and GUCY2C, but not GUCA2B, was observed in pancreata from rats with diet-induced obesity one month after sleeve gastrectomy. Interestingly, both guanylin and uroguanylin diminished the lipotoxicity in palmitate-treated RIN-m5F β-cells, evidenced by lower steatosis and downregulated lipogenic factors Srebf1, Mogat2 and Dgat1. Both guanylin peptides reduced insulin synthesis (Ins1 and Ins2) and release from RIN-m5F β-cells, but only guanylin upregulated Wnt4, a factor that controls β-cell proliferation and function. Discussion Together, sleeve gastrectomy reduced pancreatic steatosis and improved β-cell function. Several mechanisms, including the modulation of inflammation and lipogenesis as well as the upregulation of GUCA2A in the pancreas, might explain this beneficial effect of bariatric surgery.
Collapse
Affiliation(s)
- Aarón Otero
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Marina Martín
- Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Javier A. Cienfuegos
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Víctor Valentí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - María A. Burrell
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
5
|
Ferreira V, Folgueira C, García-Altares M, Guillén M, Ruíz-Rosario M, DiNunzio G, Garcia-Martinez I, Alen R, Bookmeyer C, Jones JG, Cigudosa JC, López-Larrubia P, Correig-Blanchar X, Davis RJ, Sabio G, Rada P, Valverde ÁM. Hypothalamic JNK1-hepatic fatty acid synthase axis mediates a metabolic rewiring that prevents hepatic steatosis in male mice treated with olanzapine via intraperitoneal: Additional effects of PTP1B inhibition. Redox Biol 2023; 63:102741. [PMID: 37230004 DOI: 10.1016/j.redox.2023.102741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023] Open
Abstract
Olanzapine (OLA), a widely used second-generation antipsychotic (SGA), causes weight gain and metabolic alterations when administered orally to patients. Recently, we demonstrated that, contrarily to the oral treatment which induces weight gain, OLA administered via intraperitoneal (i.p.) in male mice resulted in body weight loss. This protection was due to an increase in energy expenditure (EE) through a mechanism involving the modulation of hypothalamic AMPK activation by higher OLA levels reaching this brain region compared to those of the oral treatment. Since clinical studies have shown hepatic steatosis upon chronic treatment with OLA, herein we further investigated the role of the hypothalamus-liver interactome upon OLA administration in wild-type (WT) and protein tyrosine phosphatase 1B knockout (PTP1B-KO) mice, a preclinical model protected against metabolic syndrome. WT and PTP1B-KO male mice were fed an OLA-supplemented diet or treated via i.p. Mechanistically, we found that OLA i.p. treatment induces mild oxidative stress and inflammation in the hypothalamus in a JNK1-independent and dependent manner, respectively, without features of cell dead. Hypothalamic JNK activation up-regulated lipogenic gene expression in the liver though the vagus nerve. This effect concurred with an unexpected metabolic rewiring in the liver in which ATP depletion resulted in increased AMPK/ACC phosphorylation. This starvation-like signature prevented steatosis. By contrast, intrahepatic lipid accumulation was observed in WT mice treated orally with OLA; this effect being absent in PTP1B-KO mice. We also demonstrated an additional benefit of PTP1B inhibition against hypothalamic JNK activation, oxidative stress and inflammation induced by chronic OLA i.p. treatment, thereby preventing hepatic lipogenesis. The protection conferred by PTP1B deficiency against hepatic steatosis in the oral OLA treatment or against oxidative stress and neuroinflammation in the i.p. treatment strongly suggests that targeting PTP1B might be also a therapeutic strategy to prevent metabolic comorbidities in patients under OLA treatment in a personalized manner.
Collapse
Affiliation(s)
- Vitor Ferreira
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - Cintia Folgueira
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | - María García-Altares
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain; Rovira I Virgili University, Department of Electronic Engineering, Tarragona, Spain
| | - Maria Guillén
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain
| | | | - Giada DiNunzio
- Center for Neurosciences and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | - Irma Garcia-Martinez
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - Rosa Alen
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - Christoph Bookmeyer
- Rovira I Virgili University, Department of Electronic Engineering, Tarragona, Spain
| | - John G Jones
- Center for Neurosciences and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | | | - Pilar López-Larrubia
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain
| | - Xavier Correig-Blanchar
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain; Rovira I Virgili University, Department of Electronic Engineering, Tarragona, Spain; Institut D'Investigacio Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Roger J Davis
- Program in Molecular Medicine, Chan Medical School, University of Massachusetts, Worcester, USA
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | - Patricia Rada
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain.
| | - Ángela M Valverde
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain.
| |
Collapse
|
6
|
Claret M, Nogueiras R. Human defensin 5-based compounds: a new approach to fight obesity? Gut 2023; 72:610-611. [PMID: 35961769 DOI: 10.1136/gutjnl-2022-328158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 12/08/2022]
Affiliation(s)
- Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, A Coruna, Spain
| |
Collapse
|
7
|
Wang B, Steinberg GR. Environmental toxicants, brown adipose tissue, and potential links to obesity and metabolic disease. Curr Opin Pharmacol 2022; 67:102314. [PMID: 36334331 DOI: 10.1016/j.coph.2022.102314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/12/2022] [Accepted: 10/03/2022] [Indexed: 12/15/2022]
Abstract
Rates of human obesity, type 2 diabetes, and non-alcoholic fatty liver disease (NAFLD) have risen faster than anticipated and cannot solely be explained by excessive caloric intake or physical inactivity. Importantly, this effect is also observed in many other domesticated and non-domesticated mammals, which has led to the hypothesis that synthetic environmental pollutants may be contributing to disease development. While the impact of these chemicals on appetite and adipogenesis has been extensively studied, their potential role in reducing energy expenditure is less studied. An important component of whole-body energy expenditure is adaptive and diet-induced thermogenesis in human brown adipose tissue (BAT). This review summarizes recent evidence that environmental pollutants such as the pesticide chlorpyrifos inhibit BAT function, diet-induced thermogenesis and the potential signaling pathways mediating these effects. Lastly, we discuss the importance of housing experimental mice at thermoneutrality, rather than room temperature, to maximize the translation of findings to humans.
Collapse
Affiliation(s)
- Bo Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, PR China.
| | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Canada; Division of Endocrinology and Metabolism, Department of Medicine, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Canada
| |
Collapse
|
8
|
Caspi A, Entezari AA, Crutcher M, Snook AE, Waldman SA. Guanylyl cyclase C as a diagnostic and therapeutic target in colorectal cancer. Per Med 2022; 19:457-472. [PMID: 35920071 DOI: 10.2217/pme-2022-0026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 06/16/2022] [Indexed: 11/21/2022]
Abstract
Colorectal cancer remains a major cause of mortality in the USA, despite advances in prevention and screening. Existing therapies focus primarily on generic treatment such as surgical intervention and chemotherapy, depending on disease severity. As personalized medicine and targeted molecular oncology continue to develop as promising treatment avenues, there has emerged a need for effective targets and biomarkers of colorectal cancer. The transmembrane receptor guanylyl cyclase C (GUCY2C) regulates intestinal homeostasis and has emerged as a tumor suppressor. Further, it is universally expressed in advanced metastatic colorectal tumors, as well as other cancer types that arise through intestinal metaplasia. In this context, GUCY2C satisfies many characteristics of a compelling target and biomarker for gastrointestinal malignancies.
Collapse
Affiliation(s)
- Adi Caspi
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ariana A Entezari
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Madison Crutcher
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam E Snook
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Scott A Waldman
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
9
|
Pena-Leon V, Folgueira C, Barja-Fernández S, Pérez-Lois R, Da Silva Lima N, Martin M, Heras V, Martinez-Martinez S, Valero P, Iglesias C, Duquenne M, Al-Massadi O, Beiroa D, Souto Y, Fidalgo M, Sowmyalakshmi R, Guallar D, Cunarro J, Castelao C, Senra A, González-Saenz P, Vázquez-Cobela R, Leis R, Sabio G, Mueller-Fielitz H, Schwaninger M, López M, Tovar S, Casanueva FF, Valjent E, Diéguez C, Prevot V, Nogueiras R, Seoane LM. Prolonged breastfeeding protects from obesity by hypothalamic action of hepatic FGF21. Nat Metab 2022; 4:901-917. [PMID: 35879461 PMCID: PMC9314260 DOI: 10.1038/s42255-022-00602-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/08/2022] [Indexed: 12/25/2022]
Abstract
Early-life determinants are thought to be a major factor in the rapid increase of obesity. However, while maternal nutrition has been extensively studied, the effects of breastfeeding by the infant on the reprogramming of energy balance in childhood and throughout adulthood remain largely unknown. Here we show that delayed weaning in rat pups protects them against diet-induced obesity in adulthood, through enhanced brown adipose tissue thermogenesis and energy expenditure. In-depth metabolic phenotyping in this rat model as well as in transgenic mice reveals that the effects of prolonged suckling are mediated by increased hepatic fibroblast growth factor 21 (FGF21) production and tanycyte-controlled access to the hypothalamus in adulthood. Specifically, FGF21 activates GABA-containing neurons expressing dopamine receptor 2 in the lateral hypothalamic area and zona incerta. Prolonged breastfeeding thus constitutes a protective mechanism against obesity by affecting long-lasting physiological changes in liver-to-hypothalamus communication and hypothalamic metabolic regulation.
Collapse
Affiliation(s)
- Veronica Pena-Leon
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Cintia Folgueira
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Silvia Barja-Fernández
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
| | - Raquel Pérez-Lois
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Natália Da Silva Lima
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marion Martin
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm, University of Lille, Lille, France
| | - Violeta Heras
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Sara Martinez-Martinez
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Paola Valero
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Cristina Iglesias
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Mannon Duquenne
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm, University of Lille, Lille, France
| | - Omar Al-Massadi
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Daniel Beiroa
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Yara Souto
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel Fidalgo
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Rasika Sowmyalakshmi
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm, University of Lille, Lille, France
| | - Diana Guallar
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Juan Cunarro
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Cecilia Castelao
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Ana Senra
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Patricia González-Saenz
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
| | - Rocío Vázquez-Cobela
- Pediatrics Department, GI Pediatric Nutrition, Galicia Research Unit for Development, Growth and Human Nutrition, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain
| | - Rosaura Leis
- Pediatrics Department, GI Pediatric Nutrition, Galicia Research Unit for Development, Growth and Human Nutrition, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain
| | | | - Helge Mueller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Miguel López
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Sulay Tovar
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Felipe F Casanueva
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Emmanuel Valjent
- IGF, University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Carlos Diéguez
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm, University of Lille, Lille, France
| | - Rubén Nogueiras
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain.
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain.
- Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| | - Luisa M Seoane
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain.
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain.
| |
Collapse
|
10
|
Ratko M, Habek N, Radmilović MD, Škokić S, Justić H, Barić A, Dugandžić A. Role of uroguanylin's signaling pathway in the development of ischemic stroke. Eur J Neurosci 2022; 56:3720-3737. [PMID: 35445449 PMCID: PMC9542124 DOI: 10.1111/ejn.15674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/30/2022] [Accepted: 04/17/2022] [Indexed: 11/28/2022]
Abstract
Stroke is one of the leading causes of mortality and disability worldwide. By affecting bradykinin function, activation of guanylate cyclase (GC)‐A has been shown to have a neuroprotective effect after ischaemic stroke, whereas the same has not been confirmed for GC‐B; therefore, we aimed to determine the possible role of GC‐C and its agonist, uroguanylin (UGN), in the development of stroke. In this study, middle cerebral artery occlusion (MCAO) was performed on wild‐type (WT), GC‐C KO and UGN KO mice. MR images were acquired before and 24 h after MCAO. On brain slices 48 h after MCAO, the Ca2+ response to UGN stimulation was recorded. Our results showed that the absence of GC‐C in GC‐C KO mice resulted in the development of smaller ischaemic lesions compared with WT littermates, which is an opposite effect compared with the effects of GC‐A agonists on brain lesions. WT and UGN KO animals showed a stronger Ca2+ response upon UGN stimulation in astrocytes of the peri‐ischaemic cerebral cortex compared with the same cortical region of the unaffected contralateral hemisphere. This stronger activation was not observed in GC‐C KO animals, which may be the reason for smaller lesion development in GC‐C KO mice. The reason why GC‐C might affect Ca2+ signalling in peri‐ischaemic astrocytes is that GC‐C is expressed in these cells after MCAO, whereas under normoxic conditions, it is expressed mainly in cortical neurons. Stronger activation of the Ca2+‐dependent signalling pathway could lead to the stronger activation of the Na+/H+ exchanger, tissue acidification and neuronal death.
Collapse
Affiliation(s)
- Martina Ratko
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Nikola Habek
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | | | - Siniša Škokić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Helena Justić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Anja Barić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Aleksandra Dugandžić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia.,Department of Physiology, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
11
|
Kappa-Opioid Receptor Blockade Ameliorates Obesity Caused by Estrogen Withdrawal via Promotion of Energy Expenditure through mTOR Pathway. Int J Mol Sci 2022; 23:ijms23063118. [PMID: 35328539 PMCID: PMC8953356 DOI: 10.3390/ijms23063118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/25/2022] [Accepted: 03/11/2022] [Indexed: 12/11/2022] Open
Abstract
Weight gain is a hallmark of decreased estradiol (E2) levels because of menopause or following surgical ovariectomy (OVX) at younger ages. Of note, this weight gain tends to be around the abdomen, which is frequently associated with impaired metabolic homeostasis and greater cardiovascular risk in both rodents and humans. However, the molecular underpinnings and the neuronal basis for these effects remain to be elucidated. The aim of this study is to elucidate whether the kappa-opioid receptor (k-OR) system is involved in mediating body weight changes associated with E2 withdrawal. Here, we document that body weight gain induced by OVX occurs, at least partially, in a k-OR dependent manner, by modulation of energy expenditure independently of food intake as assessed in Oprk1−/−global KO mice. These effects were also observed following central pharmacological blockade of the k-OR system using the k-OR-selective antagonist PF-04455242 in wild type mice, in which we also observed a decrease in OVX-induced weight gain associated with increased UCP1 positive immunostaining in brown adipose tissue (BAT) and browning of white adipose tissue (WAT). Remarkably, the hypothalamic mTOR pathway plays an important role in regulating weight gain and adiposity in OVX mice. These findings will help to define new therapies to manage metabolic disorders associated with low/null E2 levels based on the modulation of central k-OR signaling.
Collapse
|
12
|
Frühbeck G, Becerril S, Martín M, Ramírez B, Valentí V, Moncada R, Catalán V, Gómez-Ambrosi J, Silva C, Burrell MA, Escalada J, Rodríguez A. High plasma and lingual uroguanylin as potential contributors to changes in food preference after sleeve gastrectomy. Metabolism 2022; 128:155119. [PMID: 34990711 DOI: 10.1016/j.metabol.2021.155119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/25/2021] [Accepted: 12/27/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND The biological mediators supporting long-term weight loss and changes in dietary choice behaviour after sleeve gastrectomy remain unclear. Guanylin and uroguanylin are gut hormones involved in the regulation of satiety, food preference and adiposity. Thus, we sought to analyze whether the guanylin system is involved in changes in food preference after sleeve gastrectomy in obesity. METHODS Proguanylin (GUCA2A) and prouroguanylin (GUCA2B) were determined in patients with severe obesity (n = 41) as well as in rats with diet-induced obesity (n = 48), monogenic obesity (Zucker fa/fa) (n = 18) or in a food choice paradigm (normal diet vs high-fat diet) (n = 16) submitted to sleeve gastrectomy. Lingual distribution and expression of guanylins (GUCA2A and GUCA2B) and their receptor GUCY2C as well as the fatty acid receptor CD36 were evaluated in the preclinical models. RESULTS Circulating concentrations of GUCA2A and GUCA2B were increased after sleeve gastrectomy in patients with severe obesity as well as in rats with diet-induced and monogenic (fa/fa) obesity. Interestingly, the lower dietary fat preference observed in obese rats under the food choice paradigm as well as in patients with obesity after sleeve gastrectomy were negatively associated with post-surgical GUCA2B levels. Moreover, sleeve gastrectomy upregulated the low expression of GUCA2A and GUCA2B in taste bud cells of tongues from rats with diet-induced and monogenic (fa/fa) obesity in parallel to a downregulation of the lingual lipid sensor CD36. CONCLUSIONS The increased circulating and lingual GUCA2B after sleeve gastrectomy suggest an association between the uroguanylin-GUCY2C endocrine axis and food preference through the regulation of gustatory responses.
Collapse
Affiliation(s)
- Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain.
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Marina Martín
- Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Víctor Valentí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Camilo Silva
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - María A Burrell
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Javier Escalada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
13
|
Prasad H, Mathew JKK, Visweswariah SS. Receptor Guanylyl Cyclase C and Cyclic GMP in Health and Disease: Perspectives and Therapeutic Opportunities. Front Endocrinol (Lausanne) 2022; 13:911459. [PMID: 35846281 PMCID: PMC9276936 DOI: 10.3389/fendo.2022.911459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Receptor Guanylyl Cyclase C (GC-C) was initially characterized as an important regulator of intestinal fluid and ion homeostasis. Recent findings demonstrate that GC-C is also causally linked to intestinal inflammation, dysbiosis, and tumorigenesis. These advances have been fueled in part by identifying mutations or changes in gene expression in GC-C or its ligands, that disrupt the delicate balance of intracellular cGMP levels and are associated with a wide range of clinical phenotypes. In this review, we highlight aspects of the current knowledge of the GC-C signaling pathway in homeostasis and disease, emphasizing recent advances in the field. The review summarizes extra gastrointestinal functions for GC-C signaling, such as appetite control, energy expenditure, visceral nociception, and behavioral processes. Recent research has expanded the homeostatic role of GC-C and implicated it in regulating the ion-microbiome-immune axis, which acts as a mechanistic driver in inflammatory bowel disease. The development of transgenic and knockout mouse models allowed for in-depth studies of GC-C and its relationship to whole-animal physiology. A deeper understanding of the various aspects of GC-C biology and their relationships with pathologies such as inflammatory bowel disease, colorectal cancer, and obesity can be leveraged to devise novel therapeutics.
Collapse
Affiliation(s)
- Hari Prasad
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | | | - Sandhya S. Visweswariah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru, India
- *Correspondence: Sandhya S. Visweswariah,
| |
Collapse
|
14
|
Nogueiras R. MECHANISMS IN ENDOCRINOLOGY: The gut-brain axis: regulating energy balance independent of food intake. Eur J Endocrinol 2021; 185:R75-R91. [PMID: 34260412 PMCID: PMC8345901 DOI: 10.1530/eje-21-0277] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
Obesity is a global pandemic with a large health and economic burden worldwide. Bodyweight is regulated by the ability of the CNS, and especially the hypothalamus, to orchestrate the function of peripheral organs that play a key role in metabolism. Gut hormones play a fundamental role in the regulation of energy balance, as they modulate not only feeding behavior but also energy expenditure and nutrient partitioning. This review examines the recent discoveries about hormones produced in the stomach and gut, which have been reported to regulate food intake and energy expenditure in preclinical models. Some of these hormones act on the hypothalamus to modulate thermogenesis and adiposity in a food intake-independent fashion. Finally, the association of these gut hormones to eating, energy expenditure, and weight loss after bariatric surgery in humans is discussed.
Collapse
Affiliation(s)
- Ruben Nogueiras
- Department of Physiology, CIMUS, USC, CIBER Fisiopatología Obesidad y Nutrición (CiberOBN), Instituto Salud Carlos III, Galician Agency of Innovation, Xunta de Galicia, Santiago de Compostela, Spain
| |
Collapse
|
15
|
Barton JR, Snook AE, Waldman SA. From leptin to lasers: the past and present of mouse models of obesity. Expert Opin Drug Discov 2021; 16:777-790. [PMID: 33472452 PMCID: PMC8243785 DOI: 10.1080/17460441.2021.1877654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/14/2021] [Indexed: 10/22/2022]
Abstract
Introduction: Obesity is a prevalent condition that accounts for significant morbidity and mortality across the globe. Despite substantial effort, most obesity pharmacotherapies have proven unsafe or ineffective. The use of obese mouse models provides unique insight into the hormones and mechanisms that regulate appetite and metabolism. Paramount among these models are the 'obese' and 'diabetic' mice that revealed the powerful satiety hormone leptin, revolutionizing obesity research.Areas Covered: In this article, the authors discuss work on leptin therapy, and the clinical response to leptin in humans. The authors describe the use of modern mouse genetics to study targetable mechanisms for genetic forms of human obesity. Additionally, they describe mouse models of neuromodulation and their utility in unraveling neural circuits that govern appetite and metabolism.Expert opinion: Combining past and present models of obesity is required for the development of safe, effective, and impactful obesity therapy. Current research in obesity can benefit from repositories of genetically engineered mouse models to discover interactions between appetitive systems and circuits. Combining leptin therapy with other satiety signals comprising the gut-brain axis is a promising approach to induce significant enduring weight loss.
Collapse
Affiliation(s)
- Joshua R. Barton
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam E. Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Scott A. Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
16
|
Wei R, Zhuge X, Yue P, Liu M, Zhu L, Liu J, Xia C. Effect of hepatic sympathetic nerve removal on energy metabolism in an animal model of cognitive impairment and its relationship to Glut2 expression. Open Life Sci 2021; 15:311-317. [PMID: 33817219 PMCID: PMC7874542 DOI: 10.1515/biol-2020-0033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/25/2020] [Accepted: 03/19/2020] [Indexed: 01/14/2023] Open
Abstract
The aims of this study were to investigate the effect of hepatic sympathetic nerve removal on glucose and lipid metabolism in rats with cognitive impairment and to evaluate the relationship between these effects and liver Glut2 expression. Hippocampal injection of Aβ1–42 was used to induce cognitive impairment. Impaired rats were divided into experimental, sham, and control groups. The experimental group was injected with 6-hydroxydopamine to remove the sympathetic nerve. At 4 weeks post injection, body weight, food and water intake, blood sugar, and blood lipids were measured, and periodic acid-Schiff (PAS) staining was used to assess the liver glycogen content. Liver Glut2 mRNA and protein were also detected. The experimental group showed reduced body weight, food intake, and blood glucose levels and elevated insulin levels compared with the control group. PAS staining showed higher glycogen contents in the experimental group than in controls. The expression levels of Glut2 mRNA and protein in the experimental group were significantly lower than in the controls. Metabolism was significantly impacted in rats with cognitive impairment following removal of the hepatic sympathetic nerve. Disruption to Glut2 liver expression via sympathetic nerve disruption represents a possible underlying mechanism.
Collapse
Affiliation(s)
- Riming Wei
- College of Biotechnology, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Xiuhong Zhuge
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Pengpeng Yue
- College of Biotechnology, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Manjun Liu
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Lin Zhu
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Jianxiang Liu
- College of Biotechnology, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Chunbo Xia
- Department of Human Anatomy of Basic Medical College, Guilin Medical University, Guilin, Guangxi, 541004, China
| |
Collapse
|
17
|
Schalla MA, Taché Y, Stengel A. Neuroendocrine Peptides of the Gut and Their Role in the Regulation of Food Intake. Compr Physiol 2021; 11:1679-1730. [PMID: 33792904 DOI: 10.1002/cphy.c200007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of food intake encompasses complex interplays between the gut and the brain. Among them, the gastrointestinal tract releases different peptides that communicate the metabolic state to specific nuclei in the hindbrain and the hypothalamus. The present overview gives emphasis on seven peptides that are produced by and secreted from specialized enteroendocrine cells along the gastrointestinal tract in relation with the nutritional status. These established modulators of feeding are ghrelin and nesfatin-1 secreted from gastric X/A-like cells, cholecystokinin (CCK) secreted from duodenal I-cells, glucagon-like peptide 1 (GLP-1), oxyntomodulin, and peptide YY (PYY) secreted from intestinal L-cells and uroguanylin (UGN) released from enterochromaffin (EC) cells. © 2021 American Physiological Society. Compr Physiol 11:1679-1730, 2021.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
18
|
Quiñones M, Hernández-Bautista R, Beiroa D, Heras V, Torres-Leal FL, Lam BYH, Senra A, Fernø J, Gómez-Valadés AG, Schwaninger M, Prevot V, Yeo G, Claret M, López M, Diéguez C, Al-Massadi O, Nogueiras R. Sirt3 in POMC neurons controls energy balance in a sex- and diet-dependent manner. Redox Biol 2021; 41:101945. [PMID: 33744652 PMCID: PMC8005845 DOI: 10.1016/j.redox.2021.101945] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
Sirtuin 3 (SIRT3) is one of the seven mammalian sirtuin homologs of the yeast Sir2 gene that has emerged as an important player in the regulation of energy metabolism in peripheral tissues. However, its role in the hypothalamus has not been explored. Herein, we show that the genetic inhibition of SIRT3 in the hypothalamic arcuate nucleus (ARC) induced a negative energy balance and improvement of several metabolic parameters. These effects are specific for POMC neurons, because ablation of SIRT3 in POMC, but not in AgRP neurons, decreased body weight and adiposity, increased energy expenditure and brown adipose tissue (BAT) activity, and induced browning in white adipose tissue (WAT). Notably, the depletion of SIRT3 in POMC neurons caused these effects in male mice fed a chow diet but failed to affect energy balance in males fed a high fat diet and females under both type of diets. Overall, we provide the first evidence pointing for a key role of SIRT3 in POMC neurons in the regulation of energy balance.
Collapse
Affiliation(s)
- Mar Quiñones
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain.
| | - René Hernández-Bautista
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Daniel Beiroa
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Violeta Heras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Francisco L Torres-Leal
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; Metabolic Diseases, Exercise and Nutrition (DOMEN) Research Group, Federal University of Piauí, Teresina, Brazil
| | - Brian Y H Lam
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Ana Senra
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Alicia García Gómez-Valadés
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, European Genomic Institute for Diabetes (EGID), F-59000, Lille, France
| | - Giles Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036, Barcelona, Spain; School of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Omar Al-Massadi
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana S/n, 15706, Santiago de Compostela, Spain.
| | - Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain; Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| |
Collapse
|
19
|
Abstract
Introduction Introduction: obesity causes millions of deaths each year. Its high prevalence in children and adolescents from southern European countries, including Spain, is associated with the new food preferences and decreased physical activity. Objective: to evaluate diet quality and physical activity in Galician schoolchildren in order to assess if modifying the current intervention strategies in lifestyles is required. Methods: in the present study, 662 students from the south of Galicia were studied (9-17 years). Body mass index (BMI) was calculated as adiposity indicator. The dietary pattern was evaluated through the Kidmed test and physical activity with PAQ-C. The sample was stratified by BMI according to Cole international standards. Data were analysed with the SPPS software. Results: fifty-six per cent of children are at high risk of overnutrition or need to improve their nutrition. The average adherence to the Mediterranean diet was low (7.64 ± 2.28), worsening with age and adiposity significantly. The average level of physical activity was moderate (3.02 ± 0.74) and it was lower in girls and adolescents; 58% showed a low-moderate level of physical activity. Quality diet was positively correlated with physical activity and negatively with BMI. Conclusions: Galician students showed a diet and physical activity risky pattern according to international/national recommendations. After several years of awareness campaigns and lifestyle interventions, improving their diet and increasing physical activity is still necessary.
Collapse
|
20
|
Bose A, Banerjee S, Visweswariah SS. Mutational landscape of receptor guanylyl cyclase C: Functional analysis and disease-related mutations. IUBMB Life 2020; 72:1145-1159. [PMID: 32293781 DOI: 10.1002/iub.2283] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/18/2022]
Abstract
Guanylyl cyclase C (GC-C) is the receptor for the heat-stable enterotoxin, which causes diarrhea, and the endogenous ligands, guanylin and uroguanylin. GC-C is predominantly expressed in the intestinal epithelium and regulates fluid and ion secretion in the gut. The receptor has a complex domain organization, and in the absence of structural information, mutational analysis provides clues to mechanisms of regulation of this protein. Here, we review the mutational landscape of this receptor that reveals regulatory features critical for its activity. We also summarize the available information on mutations in GC-C that have been reported in humans and contribute to severe gastrointestinal abnormalities. Since GC-C is also expressed in extra-intestinal tissues, it is likely that mutations thus far reported in humans may also affect other organ systems, warranting a close observation of these patients in future.
Collapse
Affiliation(s)
- Avipsa Bose
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Sanghita Banerjee
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Sandhya S Visweswariah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
21
|
Activation of brown adipose tissue in diet-induced thermogenesis is GC-C dependent. Pflugers Arch 2020; 472:405-417. [PMID: 31940065 DOI: 10.1007/s00424-020-02347-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/09/2019] [Accepted: 01/02/2020] [Indexed: 10/25/2022]
Abstract
Uroguanylin (UGN) is released from the intestine after a meal. When applied in brain ventricles, UGN increases expression of markers of thermogenesis in brown adipose tissue (BAT). Therefore, we determine the effects of its receptor, guanylate cyclase C (GC-C), on mouse interscapular BAT (iBAT) activity during diet-induced thermogenesis (DIT). The activation of iBAT after a meal is diminished in GC-C KO mice, decreased in female wild type (WT) mice, and abolished in old WT animals. The activation of iBAT after a meal is the highest in male WT animals which leads to an increase in GC-C expression in the hypothalamus, an increase in iBAT volume by aging, and induction of iBAT markers of thermogenesis. In contrast to iBAT activation after a meal, iBAT activation after a cold exposure could still exist in GC-C KO mice and it is significantly higher in female WT mice. The expression of GC-C in the proopiomelanocortin neurons of the arcuate nucleus of the hypothalamus but not in iBAT suggests central regulation of iBAT function. The iBAT activity during DIT has significantly reduced in old mice but an intranasal application of UGN leads to an increase in iBAT activity in a dose-dependent manner which is in strong negative correlation to glucose concentration in blood. This activation was not present in GC-C KO mice. Our results suggest the physiological role of GC-C on the BAT regulation and its importance in the regulation of glucose homeostasis and the development of new therapy for obesity and insulin resistance.
Collapse
|
22
|
Folgueira C, Torres-Leal FL, Beiroa D, Pena-León V, Da Silva Lima N, Milbank E, Senra A, Al-Massadi O, López M, Diéguez C, Seoane LM, Nogueiras R. Oral Pharmacological Activation of Hypothalamic Guanylate Cyclase 2C Receptor Stimulates Brown Fat Thermogenesis to Reduce Body Weight. Neuroendocrinology 2020; 110:1042-1054. [PMID: 31945763 DOI: 10.1159/000505972] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/15/2020] [Indexed: 12/14/2022]
Abstract
Linaclotide is a synthetic peptide approved by the FDA for the treatment of constipation-predominant irritable bowel syndrome and chronic constipation. Linaclotide binds and activates the transmembrane receptor guanylate cyclase 2C (Gucy2c). Uroguanylin (UGN) is a 16 amino acid peptide that is mainly secreted by enterochromaffin cells in the duodenum and proximal small intestine. UGN is the endogenous ligand of Gucy2c and decreases body weight in diet-induced obese (DIO) mice via the activation of the thermogenic program in brown adipose tissue. Therefore, we wanted to evaluate whether oral linaclotide could also improve DIO mice metabolic phenotype. In this study, we have demonstrated that DIO mice orally treated with linaclotide exhibited a significant reduction of body weight without modifying food intake. Linaclotide exerts its actions through the central nervous system, and more specifically, via Gucy2c receptors located in the mediobasal hypothalamus, leading to the activation of the sympathetic nervous system to trigger the thermogenic activity of brown fat stimulating energy expenditure. These findings indicate for first time that, in addition to its effects at intestinal level to treat irritable bowel syndrome with constipation and chronic constipation, linaclotide also exerts a beneficial effect in whole body metabolism.
Collapse
Affiliation(s)
- Cintia Folgueira
- Department of Physiology, CIMUS, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria Santiago de Compostela, Complejo Hospitalario Universitario Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto Salud Carlos III, Madrid, Spain
| | - Francisco Leonardo Torres-Leal
- Department of Physiology, CIMUS, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- Department of Biophysics and Physiology, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Center for Health Sciences, Federal University of Piaui, Teresina, Brazil
| | - Daniel Beiroa
- Department of Physiology, CIMUS, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto Salud Carlos III, Madrid, Spain
| | - Verónica Pena-León
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria Santiago de Compostela, Complejo Hospitalario Universitario Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
| | - Natália Da Silva Lima
- Department of Physiology, CIMUS, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Edward Milbank
- Department of Physiology, CIMUS, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Ana Senra
- Department of Physiology, CIMUS, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Omar Al-Massadi
- Department of Physiology, CIMUS, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto Salud Carlos III, Madrid, Spain
| | - Miguel López
- Department of Physiology, CIMUS, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto Salud Carlos III, Madrid, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto Salud Carlos III, Madrid, Spain
| | - Luisa M Seoane
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria Santiago de Compostela, Complejo Hospitalario Universitario Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto Salud Carlos III, Madrid, Spain
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain,
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto Salud Carlos III, Madrid, Spain,
- Galician Agency of Innovation, Xunta de Galicia, Santiago de Compostela, Spain,
| |
Collapse
|
23
|
Patterson M, Ward H, Halvai D, Holm Nilsen HA, Reeves S. Postprandial regulation of prouroguanylin in humans of a healthy weight and those who are overweight or with obesity. Peptides 2020; 123:170179. [PMID: 31697966 DOI: 10.1016/j.peptides.2019.170179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/12/2019] [Accepted: 10/21/2019] [Indexed: 10/25/2022]
Abstract
Uroguanylin is a peptide gut hormone proposed to have a role in signalling post meal satiety. Uroguanylin circulates as its pro-hormone, prouroguanylin. There has been limited investigation of the regulation of prouroguanylin by food; therefore we investigated prouroguanylin regulation following meals. In separate experiments we investigated the effects of high calorie (1451 kcal) and medium calorie (725 kcal), high fat meals, on plasma prouroguanylin concentrations. We then examined the effect of a 722.5 kcal high carbohydrate breakfast on prouroguanylin concentrations, comparing the response in healthy weight adults versus those who are overweight/ with obesity. The 1451 kcal meal increased prouroguanylin concentrations, versus fasting at 60 (P < 0.05), 90 (P < 0.01) and 120 (P < 0.001) minutes. After the 725 kcal meal hormone concentrations rose more slowly and were significant versus fasting concentrations at 120 min (P < 0.01). The high carbohydrate breakfast 722.5 kcal, led to an initial suppression of hormone concentrations at 30 min. post meal (P < 0.05) followed by an increase in concentrations until they were significant versus fasting at 120 min. (P < 0.01). Participants overweight/ with obesity had lower fasting prouroguanylin concentrations (P < 0.05), but post meal concentrations did not differ between the groups. Our results suggest there is a delayed increase in prouroguanylin concentrations following, large and regular sized mixed macronutrient meals rich in fat or carbohydrate. Fasting levels are suppressed in people who are overweight/ with obesity, but the post meal response remains intact. There may be potential to target post meal release of prouroguanylin in obesity.
Collapse
Affiliation(s)
| | - Hannah Ward
- Department of Life Sciences, University of Roehampton, London, UK
| | - Delaram Halvai
- Department of Life Sciences, University of Roehampton, London, UK
| | | | - Sue Reeves
- Department of Life Sciences, University of Roehampton, London, UK
| |
Collapse
|
24
|
González-García I, Milbank E, Martinez-Ordoñez A, Diéguez C, López M, Contreras C. HYPOTHesizing about central comBAT against obesity. J Physiol Biochem 2019; 76:193-211. [PMID: 31845114 DOI: 10.1007/s13105-019-00719-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022]
Abstract
The hypothalamus is a brain region in charge of many vital functions. Among them, BAT thermogenesis represents an essential physiological function to maintain body temperature. In the metabolic context, it has now been established that energy expenditure attributed to BAT function can contribute to the energy balance in a substantial extent. Thus, therapeutic interest in this regard has increased in the last years and some studies have shown that BAT function in humans can make a real contribution to improve diabetes and obesity-associated diseases. Nevertheless, how the hypothalamus controls BAT activity is still not fully understood. Despite the fact that much has been known about the mechanisms that regulate BAT activity in recent years, and that the central regulation of thermogenesis offers a very promising target, many questions remain still unsolved. Among them, the possible human application of knowledge obtained from rodent studies, and drug administration strategies able to specifically target the hypothalamus. Here, we review the current knowledge of homeostatic regulation of BAT, including the molecular insights of brown adipocytes, its central control, and its implication in the development of obesity.
Collapse
Affiliation(s)
- Ismael González-García
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.
| | - Edward Milbank
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Anxo Martinez-Ordoñez
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
| | - Carlos Diéguez
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Miguel López
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Cristina Contreras
- Department of Physiology, Pharmacy School, Complutense University of Madrid, 28040, Madrid, Spain.
| |
Collapse
|
25
|
Dugandzic A, Ratko M, Habek N. Anxiety‐like behavior in female mice changes by feeding, possible effect of guanylate cyclase C. Eur J Neurosci 2019; 52:2781-2790. [DOI: 10.1111/ejn.14607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 10/14/2019] [Accepted: 10/22/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Aleksandra Dugandzic
- Laboratory of cellular neurophysiology Croatian Institute for Brain Research School of Medicine University of Zagreb Zagreb Croatia
- Centre of Excellence for Basic, Clinical and Translational Neuroscience School of Medicine University of Zagreb Zagreb Croatia
- Department of Physiology School of Medicine University of Zagreb Zagreb Croatia
| | - Martina Ratko
- Laboratory of cellular neurophysiology Croatian Institute for Brain Research School of Medicine University of Zagreb Zagreb Croatia
- Centre of Excellence for Basic, Clinical and Translational Neuroscience School of Medicine University of Zagreb Zagreb Croatia
| | - Nikola Habek
- Laboratory of cellular neurophysiology Croatian Institute for Brain Research School of Medicine University of Zagreb Zagreb Croatia
- Centre of Excellence for Basic, Clinical and Translational Neuroscience School of Medicine University of Zagreb Zagreb Croatia
- Department of Physiology School of Medicine University of Zagreb Zagreb Croatia
| |
Collapse
|
26
|
Dye FS, Larraufie P, Kay R, Darwish T, Rievaj J, Goldspink DA, Meek CL, Middleton SJ, Hardwick RH, Roberts GP, Percival-Alwyn JL, Vaughan T, Ferraro F, Challis BG, O'Rahilly S, Groves M, Gribble FM, Reimann F. Characterisation of proguanylin expressing cells in the intestine - evidence for constitutive luminal secretion. Sci Rep 2019; 9:15574. [PMID: 31666564 PMCID: PMC6821700 DOI: 10.1038/s41598-019-52049-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022] Open
Abstract
Guanylin, a peptide implicated in regulation of intestinal fluid secretion, is expressed in the mucosa, but the exact cellular origin remains controversial. In a new transgenic mouse model fluorescent reporter protein expression driven by the proguanylin promoter was observed throughout the small intestine and colon in goblet and Paneth(-like) cells and, except in duodenum, in mature enterocytes. In Ussing chamber experiments employing both human and mouse intestinal tissue, proguanylin was released predominantly in the luminal direction. Measurements of proguanylin expression and secretion in cell lines and organoids indicated that secretion is largely constitutive and requires ER to Golgi transport but was not acutely regulated by salt or other stimuli. Using a newly-developed proguanylin assay, we found plasma levels to be raised in humans after total gastrectomy or intestinal transplantation, but largely unresponsive to nutrient ingestion. By LC-MS/MS we identified processed forms in tissue and luminal extracts, but in plasma we only detected full-length proguanylin. Our transgenic approach provides information about the cellular origins of proguanylin, complementing previous immunohistochemical and in-situ hybridisation results. The identification of processed forms of proguanylin in the intestinal lumen but not in plasma supports the notion that the primary site of action is the gut itself.
Collapse
Affiliation(s)
- Florent Serge Dye
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Pierre Larraufie
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Richard Kay
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Tamana Darwish
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Juraj Rievaj
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Dosage Form Design & Development, AstraZeneca, Cambridge, UK
| | - Deborah A Goldspink
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Claire L Meek
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Stephen J Middleton
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Richard H Hardwick
- Barrett's Oesophagus and Oesophago-gastric Cancer, Gastroenterology Services, Addenbrooke's Hospital, Cambridge, UK
| | - Geoffrey P Roberts
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | | | - Tris Vaughan
- Department of Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Franco Ferraro
- Department of Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Benjamin G Challis
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Stephen O'Rahilly
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Maria Groves
- Department of Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge, UK.
| | - Fiona M Gribble
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Frank Reimann
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Merlino DJ, Barton JR, Charsar BA, Byrne MD, Rappaport JA, Smeyne RJ, Lepore AC, Snook AE, Waldman SA. Two distinct GUCY2C circuits with PMV (hypothalamic) and SN/VTA (midbrain) origin. Brain Struct Funct 2019; 224:2983-2999. [PMID: 31485718 DOI: 10.1007/s00429-019-01949-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/24/2019] [Indexed: 12/16/2022]
Abstract
Guanylyl cyclase C (GUCY2C) is the afferent central receptor in the gut-brain endocrine axis regulated by the anorexigenic intestinal hormone uroguanylin. GUCY2C mRNA and protein are produced in the hypothalamus, a major center regulating appetite and metabolic homeostasis. Further, GUCY2C mRNA and protein are expressed in the ventral midbrain, a principal structure regulating hedonic reward from behaviors including eating. While GUCY2C is expressed in hypothalamus and midbrain, its precise neuroanatomical organization and relationship with circuits regulating satiety remain unknown. Here, we reveal that hypothalamic GUCY2C mRNA is confined to the ventral premammillary nucleus (PMV), while in midbrain it is produced by neurons in the ventral tegmental area (VTA) and substantia nigra (SN). GUCY2C in the PMV is produced by 46% of neurons expressing anorexigenic leptin receptors, while in the VTA/SN it is produced in most tyrosine hydroxylase-immunoreactive neurons. In contrast to mRNA, GUCY2C protein is widely distributed throughout the brain in canonical sites of PMV and VTA/SN axonal projections. Selective stereotaxic ablation of PMV or VTA/SN neurons eliminated GUCY2C only in their respective canonical projection sites. Conversely, specific anterograde tracer analyses of PMV or VTA/SN neurons confirmed distinct GUCY2C-immunoreactive axons projecting to those canonical locations. Together, these findings reveal two discrete neuronal circuits expressing GUCY2C originating in the PMV in the hypothalamus and in the VTA/SN in midbrain, which separately project to other sites throughout the brain. They suggest a structural basis for a role for the GUCY2C-uroguanylin gut-brain endocrine axis in regulating homeostatic and behavioral components contributing to satiety.
Collapse
Affiliation(s)
- D J Merlino
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, 368 JAH, Philadelphia, PA, 19107, USA
| | - J R Barton
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, 368 JAH, Philadelphia, PA, 19107, USA
| | - B A Charsar
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - M D Byrne
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - J A Rappaport
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, 368 JAH, Philadelphia, PA, 19107, USA
| | - R J Smeyne
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - A C Lepore
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - A E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, 368 JAH, Philadelphia, PA, 19107, USA
| | - S A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, 368 JAH, Philadelphia, PA, 19107, USA.
| |
Collapse
|
28
|
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, Nogueiras R. Hypothalamic dopamine signaling regulates brown fat thermogenesis. Nat Metab 2019; 1:811-829. [PMID: 31579887 PMCID: PMC6774781 DOI: 10.1038/s42255-019-0099-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
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.
Collapse
Affiliation(s)
- Cintia Folgueira
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo. Hospitalario Universitario de Santiago (CHUS/SERGAS), Instituto de Investigación Sanitaria, Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Daniel Beiroa
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Begoña Porteiro
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Manon Duquenne
- Jean-Pierre Aubert Research Center (JPArc), Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm UMR-S 1172, Lille, France
| | | | - Marcos F Fondevila
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Silvia Barja-Fernández
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo. Hospitalario Universitario de Santiago (CHUS/SERGAS), Instituto de Investigación Sanitaria, Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Rosalia Gallego
- Department of Morphological Sciences, School of Medicine, University of Santiago de Compostela, S. Francisco s/n, 15782 Santiago de Compostela (A Coruña), Spain
| | - René Hernández-Bautista
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| | - Cecilia Castelao
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo. Hospitalario Universitario de Santiago (CHUS/SERGAS), Instituto de Investigación Sanitaria, Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Ana Senra
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| | - Patricia Seoane
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Noemi Gómez
- Molecular Imaging Group, Department of Psychiatry, Radiology and Public Health, Faculty of Medicine Universidade de Santiago de Compostela (USC), Santiago de Compostela 15782 Spain; Molecular Imaging Group. Health Research Institute of Santiago de Compostela (IDIS). Travesía da Choupana s/n Santiago de Compostela. Zip Code: 15706. Spain; Nuclear Medicine Department University Clinical Hospital Santiago de Compostela (SERGAS) (CHUS), Travesía Choupana s/n. Santiago de Compostela 15706 Spain
| | - Pablo Aguiar
- Molecular Imaging Group, Department of Psychiatry, Radiology and Public Health, Faculty of Medicine Universidade de Santiago de Compostela (USC), Santiago de Compostela 15782 Spain; Molecular Imaging Group. Health Research Institute of Santiago de Compostela (IDIS). Travesía da Choupana s/n Santiago de Compostela. Zip Code: 15706. Spain; Nuclear Medicine Department University Clinical Hospital Santiago de Compostela (SERGAS) (CHUS), Travesía Choupana s/n. Santiago de Compostela 15706 Spain
| | - Diana Guallar
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| | - Miguel Fidalgo
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| | - Amparo Romero-Pico
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Roger Adan
- Brain Center Rudolf Magnus, Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Clemence Blouet
- MRC Metabolic Disease Unit. Institute of Metabolic Science. University of Cambridge, UK
| | - Jose Luís Labandeira-García
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases, CIBERNED, Madrid, Spain
| | - Françoise Jeanrenaud
- Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Imre Kallo
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, HAS, 1083, Budapest, Hungary
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, HAS, 1083, Budapest, Hungary
| | - Javier Salvador
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra & IdiSNA, Pamplona, Spain
| | - Vincent Prevot
- Jean-Pierre Aubert Research Center (JPArc), Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm UMR-S 1172, Lille, France
| | - Carlos Dieguez
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Miguel Lopez
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Emmanuel Valjent
- IGF, Inserm, CNRS, Univ. Montpellier, F-34094 Montpellier, France
| | - Gema Frühbeck
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra & IdiSNA, Pamplona, Spain
| | - Luisa M Seoane
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo. Hospitalario Universitario de Santiago (CHUS/SERGAS), Instituto de Investigación Sanitaria, Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Ruben Nogueiras
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| |
Collapse
|
29
|
Waldman SA, Tenenbaum R, Foehl HC, Winkle P, Griffin P. Blunted Evoked Prouroguanylin Endocrine Secretion in Chronic Constipation. Clin Transl Gastroenterol 2019; 10:e00016. [PMID: 31318728 PMCID: PMC6708669 DOI: 10.14309/ctg.0000000000000016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/16/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Prouroguanylin (ProUGN) in the intestine is cleaved to form uroguanylin (UGN), which stimulates guanylate cyclase C (GUCY2C), inducing cyclic guanosine monophosphate signaling. Paracrine release regulates fluid secretion, contributing to bowel function, whereas endocrine secretion evoked by eating forms a gut-brain axis, controlling appetite. Whereas hormone insufficiency contributes to hyperphagia in obesity, its contribution to the pathophysiology of constipation syndromes remains unexplored. Here, we compared circulating ProUGN and UGN in healthy subjects and in patients with chronic idiopathic constipation (CIC) and patients with irritable bowel syndrome with constipation (IBS-C). METHODS Circulating ProUGN and UGN levels were measured in 60 healthy subjects, 53 patients with CIC, and 54 patients with IBS-C. After an overnight fast, the participants ingested a standardized meal; blood samples were drawn at fasting and at 30, 60, and 90 minutes thereafter, and hormone levels were quantified by enzyme-linked immunosorbent assay. RESULTS Fasting ProUGN levels were >30% lower in patients with CIC and those with IBS-C compared with healthy subjects regardless of age, sex, or disease state. After eating, ProUGN levels increased compared with fasting levels, although the rate of change was slower and maximum levels were lower in patients with CIC and those with IBS-C. Similarly, fasting UGN levels were lower in patients with CIC and those with IBS-C compared with healthy subjects. However, unlike ProUGN levels, UGN levels did not increase after eating. DISCUSSION These observations support a novel pathophysiologic model in which CIC and IBS-C reflect a contribution of ProUGN insufficiency dysregulating intestinal fluid and electrolyte secretion. TRANSLATIONAL IMPACT This study suggests that CIC and IBS-C can be treated by oral GUCY2C hormone replacement. Indeed, these observations provide a mechanistic framework for the clinical utility of oral GUCY2C ligands like plecanatide (Trulance) and linaclotide (Linzess) to treat CIC and IBS-C.
Collapse
Affiliation(s)
- Scott A. Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Henry C. Foehl
- Foehl Statistics & Analysis LLC, Philadelphia, Pennsylvania, USA
| | - Peter Winkle
- Anaheim Clinical Trials, Anaheim, California, USA
| | | |
Collapse
|
30
|
ANGPTL-4 is Associated with Obesity and Lipid Profile in Children and Adolescents. Nutrients 2019; 11:nu11061340. [PMID: 31207920 PMCID: PMC6628529 DOI: 10.3390/nu11061340] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/06/2019] [Accepted: 06/06/2019] [Indexed: 12/13/2022] Open
Abstract
Angiopoietin-like protein 4 (ANGPTL-4) regulates lipidic metabolism and affects energy homeostasis. However, its function in children with obesity remains unknown. We investigated plasma ANGPTL-4 levels in children and its relationship with body mass index (BMI) and different lipidic parameters such as free fatty acids (FFA). Plasma ANGPTL-4 levels were analyzed in two different cohorts. In the first cohort (n = 150, age 3–17 years), which included children with normal weight or obesity, we performed a cross-sectional study. In the second cohort, which included only children with obesity (n = 20, age 5–18 years) followed up for two years after an intervention for weight loss, in which we performed a longitudinal study measuring ANGPTL-4 before and after BMI-loss. In the cross-sectional study, circulating ANGPTL-4 levels were lower in children with obesity than in those with normal weight. Moreover, ANGPTL-4 presented a negative correlation with BMI, waist circumference, weight, insulin, homeostasis model assessment of insulin resistance index (HOMA index), triglycerides, and leptin, and a positive correlation with FFA and vitamin-D. In the longitudinal study, the percent change in plasma ANGPTL-4 was correlated with the percent change in FFA, total-cholesterol and high-density lipoprotein cholesterol. This study reveals a significant association of ANGPTL-4 with pediatric obesity and plasma lipid profile.
Collapse
|
31
|
Abstract
In the midst of an obesity epidemic, the promotion of brown adipose tissue (BAT) function and the browning of white adipose tissue (WAT) have emerged as promising therapeutic targets to increase energy expenditure and counteract weight gain. Despite the fact that the thermogenic potential of bone fide BAT in rodents is several orders of magnitudes higher than white fat containing brite/beige adipocytes, WAT browning represents a particularly intriguing concept in humans given the extreme amount of excess WAT in obese individuals. In addition, the clear distinction between classic brown and beige fat that has been proposed in mice does not exist in humans. In fact, studies of human BAT biopsies found controversial results suggesting both classic brown and beige characteristics. Irrespective of the true ‘color’, accumulating evidence suggests the induction of thermogenic adipocytes in human WAT depots in response to specific stimuli, highlighting that WAT browning may occur in both, mice and humans. These observations also emphasize the great plasticity of human fat depots and raise important questions about the metabolic properties of thermogenically active adipose tissue in humans and the potential therapeutic implications. We will first review the cellular and molecular aspects of selected adipose tissue browning concepts that have been identified in mouse models with emphasis on neuronal factors, the microbiome, immune cells and several hormones. We will also summarize the evidence for adipose tissue browning in humans including some experimental pharmacologic approaches.
Collapse
Affiliation(s)
- Carsten T Herz
- Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Florian W Kiefer
- Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
32
|
Uroguanylin Improves Leptin Responsiveness in Diet-Induced Obese Mice. Nutrients 2019; 11:nu11040752. [PMID: 30935076 PMCID: PMC6520813 DOI: 10.3390/nu11040752] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/27/2019] [Accepted: 03/27/2019] [Indexed: 12/17/2022] Open
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.
Collapse
|
33
|
Circulating Pro-Uroguanylin Levels In Children And Their Relation To Obesity, Sex And Puberty. Sci Rep 2018; 8:14541. [PMID: 30266914 PMCID: PMC6162323 DOI: 10.1038/s41598-018-32767-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 09/07/2018] [Indexed: 01/13/2023] Open
Abstract
Uroguanylin is a 16 amino acid peptide that constitutes a key component of the gut- brain axis with special relevance in body weight regulation. In childhood and adolescence, periods of life with notable metabolic changes; limited data exist, with measurements of pro-uroguanylin in adolescence but not in prepubertal children. This study investigates pro-uroguanylin circulating levels in children with obesity and its relationship with obesity, sex and pubertal development. We analyzed circulating prouroguanylin levels in 117 children (62) and adolescents (55), including 73 with obesity and 44 with normal weight. The pro-uroguanylin concentration is higher in lean girls during pre-puberty versus lean boys (1111 vs 635, p < 0.001). During puberty, pro-uroguanylin levels are higher in lean males with respect to lean females (1060 vs 698, p < 0.01). In girls, a negative correlation exists between pro-uroguanylin and age, Tanner stage, weight, height, BMI (body mass index), waist circumference and plasma levels of leptin and testosterone; a positive correlation was found between pro-uroguanylin and free triiodothyronine. In boys, a positive correlation was found between pro-uroguanylin and BMI and waist circumference and a negative correlation was found with high density lipoprotein-cholesterol. We conclude that a sexual dimorphism exists in circulating pro-uroguanylin levels with respect to BMI. Uroguanylin presents also an opposed circulating pattern during puberty in both sexes.
Collapse
|
34
|
p53 in AgRP neurons is required for protection against diet-induced obesity via JNK1. Nat Commun 2018; 9:3432. [PMID: 30143607 PMCID: PMC6109113 DOI: 10.1038/s41467-018-05711-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/19/2018] [Indexed: 12/13/2022] Open
Abstract
p53 is a well-known tumor suppressor that has emerged as an important player in energy balance. However, its metabolic role in the hypothalamus remains unknown. Herein, we show that mice lacking p53 in agouti-related peptide (AgRP), but not proopiomelanocortin (POMC) or steroidogenic factor-1 (SF1) neurons, are more prone to develop diet-induced obesity and show reduced brown adipose tissue (BAT) thermogenic activity. AgRP-specific ablation of p53 resulted in increased hypothalamic c-Jun N-terminal kinase (JNK) activity before the mice developed obesity, and central inhibition of JNK reversed the obese phenotype of these mice. The overexpression of p53 in the ARC or specifically in AgRP neurons of obese mice decreased body weight and stimulated BAT thermogenesis, resulting in body weight loss. Finally, p53 in AgRP neurons regulates the ghrelin-induced food intake and body weight. Overall, our findings provide evidence that p53 in AgRP neurons is required for normal adaptations against diet-induced obesity. Emerging studies suggest that p53 is an important regulator of energy metabolism, yet there is little known about the metabolic function of this tumor suppressor in the hypothalamus. Here, authors illustrate that p53, specifically in AgRP neurons, is required for adaptation to diet-induced obesity.
Collapse
|
35
|
Fernandez-Cachon ML, Pedersen SL, Rigbolt KT, Zhang C, Fabricius K, Hansen HH, Elster L, Fink LN, Schäfer M, Rhee NA, Langholz E, Wandall E, Friis SU, Vilmann P, Kristiansen VB, Schmidt C, Schreiter K, Breitschopf K, Hübschle T, Jorsal T, Vilsbøll T, Schmidt T, Theis S, Knop FK, Larsen PJ, Jelsing J. Guanylin and uroguanylin mRNA expression is increased following Roux-en-Y gastric bypass, but guanylins do not play a significant role in body weight regulation and glycemic control. Peptides 2018; 101:32-43. [PMID: 29289697 DOI: 10.1016/j.peptides.2017.12.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/18/2017] [Accepted: 12/24/2017] [Indexed: 02/07/2023]
Abstract
AIM To determine whether intestinal expression of guanylate cyclase activator 2A (GUCA2A) and guanylate cyclase activator 2B (GUCA2B) genes is regulated in obese humans following Roux-en-Y gastric bypass (RYGB), and to evaluate the corresponding guanylin (GN) and uroguanylin (UGN) peptides for potentially contributing to the beneficial metabolic effects of RYGB. METHODS Enteroendocrine cells were harvested peri- and post-RYGB, and GUCA2A/GUCA2B mRNA expression was compared. GN, UGN and their prohormones (proGN, proUGN) were administered subcutaneously in normal-weight mice to evaluate effects on food intake and glucose regulation. The effect of pro-UGN or UGN overexpression, using adeno-associated virus (AAV) vectors, was assessed in diet-induced obese (DIO) mice. Intracerebroventricular administration of GN and UGN was performed in rats for assessment of putative centrally mediated effects on food intake. GN and UGN, as well as their prohormones, were evaluated for effects on glucose-stimulated insulin secretion (GSIS) in rat pancreatic islets and perfused rat pancreas. RESULTS GUCA2A and GUCA2B mRNA expression was significantly upregulated in enteroendocrine cells after RYGB. Peripheral administration of guanylins or prohormones did not influence food intake, oral glucose tolerance, and GSIS. Central administration of GN and UGN did not affect food intake in rats. Chronic AVV-mediated overexpression of UGN and proUGN had no effect on body weight or glucose homeostasis in DIO mice. CONCLUSION GN and UGN, as well as their prohormones, do not seem to play a significant role in body weight regulation and glycemic control, suggesting that guanylin-family peptides do not show promise as targets for the treatment of obesity or diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Nicolai A Rhee
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Ebbe Langholz
- Department of Medicine, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Erik Wandall
- Department of Medicine, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Steffen U Friis
- Department of Medicine, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Peter Vilmann
- Gastro Unit, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | | | | | | | | | | | - Tina Jorsal
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | | | | | - Filip K Knop
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | | | | |
Collapse
|
36
|
Pilot Study Measuring the Novel Satiety Hormone, Pro-Uroguanylin, in Adolescents With and Without Obesity. J Pediatr Gastroenterol Nutr 2018; 66:489-495. [PMID: 29112082 PMCID: PMC5825243 DOI: 10.1097/mpg.0000000000001796] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Disruption of satiety signaling may lead to increased caloric intake and obesity. Uroguanylin, the intestinal hormone, travels as a precursor to the central nervous system where it activates guanylyl cyclase C and stimulates pro-satiety neurons. Rodent studies have demonstrated that guanylyl cyclase C-knockout mice overeat and have increased weight gain versus wild-type mice and hyper-caloric obesity diminishes uroguanylin expression. We measured circulating plasma pro-uroguanylin, along with other gastrointestinal peptides and inflammatory markers, in human adolescents with and without obesity, as a pilot study. We hypothesized that adolescents with obesity would have less circulating pro-uroguanylin than adolescents without obesity have. METHODS We recruited 24 adolescents (age 14-17 years) with and without obesity (body mass index >95% or body mass index <95%) and measured plasma pro-uroguanylin at fasting and successive time points after a meal. We measured 3 other satiety hormones and 2 inflammatory markers to characterize overall satiety signaling and highlight any link between uroguanylin and inflammation. RESULTS Female adolescents with obesity had lower circulating pro-uroguanylin levels than female adolescents without obesity; we observed no difference in males. Other measured gastrointestinal peptides varied in their differences between cohorts. Inflammatory markers were higher in female participants with obesity. CONCLUSIONS In adolescents with and without obesity, we can measure circulating pro-uroguanylin levels. In female adolescents without obesity, levels are particularly higher. Pro-uroguanylin secretion patterns differ from other circulating gastrointestinal peptides. In female adolescents with obesity, inflammation correlates with decreased pro-uroguanylin levels.
Collapse
|
37
|
Folgueira C, Barja-Fernandez S, Gonzalez-Saenz P, Pena-Leon V, Castelao C, Ruiz-Piñon M, Casanueva FF, Nogueiras R, Seoane LM. Uroguanylin: a new actor in the energy balance movie. J Mol Endocrinol 2018; 60:R31-R38. [PMID: 29203517 DOI: 10.1530/jme-17-0263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022]
Abstract
Uroguanylin (UGN) is a potential target in the fight against obesity. The mature protein is released after enzymatic cleavage from its natural precursor, proUGN. UGN is mostly produced in the gut, and its production is regulated by nutritional status. However, UGN is also produced in other tissues such as the kidneys. In the past, UGN has been widely studied as a natriuretic peptide owing to its involvement in several different pathologies such as heart failure, cancer and gastrointestinal diseases. However, recent studies have suggested that UGN also acts as a regulator of body weight homeostasis because it modulates both food intake and energy expenditure. This ultimately results in a decrease in body weight. This action is mediated by the sympathetic nervous system. Future studies should be directed at the potential effects of UGN agonists in regulating body weight in human obesity.
Collapse
Affiliation(s)
- C Folgueira
- Fisiopatología EndocrinaInstituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CiberOBN)Instituto Salud Carlos III, Madrid, Spain
- Department of PhysiologyCIMUS, USC, IDIS Santiago de Compostela, Santiago de Compostela, Spain
| | - S Barja-Fernandez
- Fisiopatología EndocrinaInstituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CiberOBN)Instituto Salud Carlos III, Madrid, Spain
| | - P Gonzalez-Saenz
- Fisiopatología EndocrinaInstituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CiberOBN)Instituto Salud Carlos III, Madrid, Spain
| | - V Pena-Leon
- Fisiopatología EndocrinaInstituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CiberOBN)Instituto Salud Carlos III, Madrid, Spain
| | - C Castelao
- Fisiopatología EndocrinaInstituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CiberOBN)Instituto Salud Carlos III, Madrid, Spain
| | - M Ruiz-Piñon
- Fisiopatología EndocrinaInstituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
- Operative Dentistry and EndodonticsUSC, Santiago de Compostela, Spain
| | - F F Casanueva
- CIBER Fisiopatología Obesidad y Nutrición (CiberOBN)Instituto Salud Carlos III, Madrid, Spain
- Laboratorio de Endocrinología Molecular y CelularUSC, Santiago de Compostela, Spain
| | - R Nogueiras
- CIBER Fisiopatología Obesidad y Nutrición (CiberOBN)Instituto Salud Carlos III, Madrid, Spain
- Department of PhysiologyCIMUS, USC, IDIS Santiago de Compostela, Santiago de Compostela, Spain
| | - L M Seoane
- Fisiopatología EndocrinaInstituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CiberOBN)Instituto Salud Carlos III, Madrid, Spain
| |
Collapse
|
38
|
Di Guglielmo MD, Perdue L, Adeyemi A, van Golen KL, Corao DU. Immunohistochemical Staining for Uroguanylin, a Satiety Hormone, is Decreased in Intestinal Tissue Specimens From Female Adolescents With Obesity. Pediatr Dev Pathol 2018; 21:285-295. [PMID: 28847213 PMCID: PMC5647253 DOI: 10.1177/1093526617722912] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gastrointestinal tract-secreted satiety hormones play a significant role in one of the largest health-care challenges for children and adults, obesity. Recent studies in mice identified a novel role for uroguanylin, the endogenous intestinal hormone that binds guanylyl cyclase C (GUCY2C), in regulating satiety via a gut-brain signaling pathway. Mice bred without GUCY2C receptors over-ate and developed obesity. We hypothesized that intestinal uroguanylin expression in pediatric patients with obesity would be lower than patients without obesity, and we attempted to examine the difference with immunohistochemistry. Retrospective chart review of gastrointestinal endoscopic procedures at an academic children's hospital identified patients with normal pathology findings on biopsy. Children aged 8-17 were included in the review; we analyzed biopsy samples from 20 matched pairs that differed only by body mass index (BMI)-for-age (average: 25%-75% vs. high: >95%). Biopsies of the duodenum, terminal ileum, ascending colon, and descending colon were subjected to immunohistochemistry for GUCY2C, uroguanylin, and the endogenous colonic hormone, guanylin. Intensity staining of all specimens was scored by a blinded pathologist. The overall staining intensity for females with high BMI-for-age was less for uroguanylin and guanylin as compared to average BMI-for-age females while GUCY2C staining was equal. Males did not exhibit different staining intensities for uroguanylin or guanylin. More matched female pairs had greater uroguanylin and guanylin staining in the average BMI-for-age cohort. The intestinal expression of uroguanylin, a key satiety hormone, appears to be diminished in female pediatric patients in the setting of obesity.
Collapse
Affiliation(s)
- Matthew D Di Guglielmo
- Department of Pediatrics, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Lacey Perdue
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
| | - Adebowale Adeyemi
- Department of Pediatrics, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Kenneth L van Golen
- Department of Molecular Biosciences, University of Delaware, Newark, Delaware
| | - Diana U Corao
- Department of Pathology, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| |
Collapse
|
39
|
Tapia P, Fernández-Galilea M, Robledo F, Mardones P, Galgani JE, Cortés VA. Biology and pathological implications of brown adipose tissue: promises and caveats for the control of obesity and its associated complications. Biol Rev Camb Philos Soc 2017; 93:1145-1164. [DOI: 10.1111/brv.12389] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 11/10/2017] [Accepted: 11/14/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Pablo Tapia
- Department of Nutrition, Diabetes and Metabolism, School of Medicine; Pontificia Universidad Católica de Chile, Marcoleta 367; Santiago, 8330024 Chile
| | - Marta Fernández-Galilea
- Department of Nutrition, Diabetes and Metabolism, School of Medicine; Pontificia Universidad Católica de Chile, Marcoleta 367; Santiago, 8330024 Chile
| | - Fermín Robledo
- Department of Nutrition, Diabetes and Metabolism, School of Medicine; Pontificia Universidad Católica de Chile, Marcoleta 367; Santiago, 8330024 Chile
| | - Pablo Mardones
- Research and Innovation Office, School of Engineering; Pontificia Universidad Católica de Chile, Marcoleta 367; Santiago, 8330024 Chile
| | - José E. Galgani
- Department of Nutrition, Diabetes and Metabolism, School of Medicine; Pontificia Universidad Católica de Chile, Marcoleta 367; Santiago, 8330024 Chile
- Departamento Ciencias de la Salud; Carrera de Nutrición y Dietética, Pontificia Universidad Católica de Chile, Marcoleta 367; Santiago, 8330024 Chile
| | - Víctor A. Cortés
- Department of Nutrition, Diabetes and Metabolism, School of Medicine; Pontificia Universidad Católica de Chile, Marcoleta 367; Santiago, 8330024 Chile
| |
Collapse
|
40
|
Antiobesity effect of Lactobacillus reuteri 263 associated with energy metabolism remodeling of white adipose tissue in high-energy-diet-fed rats. J Nutr Biochem 2017; 54:87-94. [PMID: 29329013 DOI: 10.1016/j.jnutbio.2017.11.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/26/2017] [Accepted: 11/11/2017] [Indexed: 12/20/2022]
Abstract
Obesity is a serious and costly issue to the medical welfare worldwide. Probiotics have been suggested as one of the candidates to resolve the obesity-associated problems, but how they combat obesity is not fully understood. Herein, we investigated the effects of Lactobacillus reuteri 263 (L. reuteri 263) on antiobesity using four groups of Sprague-Dawley rats (n=10/group), namely, C (normal diet with vehicle treatment), HE [high-energy diet (HED) with vehicle treatment], 1X (HED with 2.1×109 CFU/kg/day of L. reuteri 263) and 5X (HED with 1.05×1010 CFU/kg/day of L. reuteri 263), for 8 weeks. L. reuteri 263 improved the phenomenon of obesity, serum levels of proinflammatory factors and antioxidant enzymes. More importantly, L. reuteri 263 increased oxygen consumption in white adipose tissue (WAT). The mRNA expressions of thermogenesis genes uncoupling protein-1, uncoupling protein-3, carnitine palmitoyltransferase-1 and cell death-inducing DFFA-like effector-a were up-regulated in WAT of the 5X group. Moreover, L. reuteri 263 might induce browning of WAT due to the higher mRNA levels of browning-related genes peroxisome proliferator-activated receptor-γ, PR domain containing-16, Pparγ coactivator-1α, bone morphogenetic protein-7 and fibroblast growth factor-21 in the 1X and 5X groups compared to the HE group. Finally, L. reuteri 263 altered the expressions of genes involved in glucose and lipid metabolisms in WAT, including increasing the levels of glucose transporter type 4 and carbohydrate-responsive element-binding protein and decreasing the expression of Acetyl-CoA carboxylase-1. The results suggest that L. reuteri 263 may treat obesity through energy metabolism remodeling of WAT in the high-energy-diet-induced obese rats.
Collapse
|
41
|
Contreras C, Nogueiras R, Diéguez C, Rahmouni K, López M. Traveling from the hypothalamus to the adipose tissue: The thermogenic pathway. Redox Biol 2017; 12:854-863. [PMID: 28448947 PMCID: PMC5406580 DOI: 10.1016/j.redox.2017.04.019] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/08/2017] [Accepted: 04/11/2017] [Indexed: 01/13/2023] Open
Abstract
Brown adipose tissue (BAT) is a specialized tissue critical for non-shivering thermogenesis producing heat through mitochondrial uncoupling; whereas white adipose tissue (WAT) is responsible of energy storage in the form of triglycerides. Another type of fat has been described, the beige adipose tissue; this tissue emerges in existing WAT depots but with thermogenic ability, a phenomenon known as browning. Several peripheral signals relaying information about energy status act in the brain, particularly the hypothalamus, to regulate thermogenesis in BAT and browning of WAT. Different hypothalamic areas have the capacity to regulate the thermogenic process in brown and beige adipocytes through the sympathetic nervous system (SNS). This review discusses important concepts and discoveries about the central control of thermogenesis as a trip that starts in the hypothalamus, and taking the sympathetic roads to reach brown and beige fat to modulate thermogenic functions.
Collapse
Affiliation(s)
- Cristina Contreras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain.
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, Iowa 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242, USA
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain.
| |
Collapse
|
42
|
Contreras C, Nogueiras R, Diéguez C, Medina-Gómez G, López M. Hypothalamus and thermogenesis: Heating the BAT, browning the WAT. Mol Cell Endocrinol 2016; 438:107-115. [PMID: 27498420 DOI: 10.1016/j.mce.2016.08.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/30/2016] [Accepted: 08/01/2016] [Indexed: 12/20/2022]
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.
Collapse
Affiliation(s)
- Cristina Contreras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain.
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Gema Medina-Gómez
- Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Madrid, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain.
| |
Collapse
|
43
|
Blomain ES, Merlino DJ, Pattison AM, Snook AE, Waldman SA. Guanylyl Cyclase C Hormone Axis at the Intersection of Obesity and Colorectal Cancer. Mol Pharmacol 2016; 90:199-204. [PMID: 27251363 PMCID: PMC4998665 DOI: 10.1124/mol.115.103192] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 05/25/2016] [Indexed: 12/12/2022] Open
Abstract
Obesity has emerged as a principal cause of mortality worldwide, reflecting comorbidities including cancer risk, particularly in colorectum. Although this relationship is established epidemiologically, molecular mechanisms linking colorectal cancer and obesity continue to be refined. Guanylyl cyclase C (GUCY2C), a membrane-bound guanylyl cyclase expressed in intestinal epithelial cells, binds the paracrine hormones guanylin and uroguanylin, inducing cGMP signaling in colorectum and small intestine, respectively. Guanylin is the most commonly lost gene product in sporadic colorectal cancer, and its universal loss early in transformation silences GUCY2C, a tumor suppressor, disrupting epithelial homeostasis underlying tumorigenesis. In small intestine, eating induces endocrine secretion of uroguanylin, the afferent limb of a novel gut-brain axis that activates hypothalamic GUCY2C-cGMP signaling mediating satiety opposing obesity. Recent studies revealed that diet-induced obesity suppressed guanylin and uroguanylin expression in mice and humans. Hormone loss reflects reversible calorie-induced endoplasmic reticulum stress and the associated unfolded protein response, rather than the endocrine, adipokine, or inflammatory milieu of obesity. Loss of intestinal uroguanylin secretion silences the hypothalamic GUCY2C endocrine axis, creating a feed-forward loop contributing to hyperphagia in obesity. Importantly, calorie-induced guanylin loss silences the GUCY2C-cGMP paracrine axis underlying obesity-induced epithelial dysfunction and colorectal tumorigenesis. Indeed, genetically enforced guanylin replacement eliminated diet-induced intestinal tumorigenesis in mice. Taken together, these observations suggest that GUCY2C hormone axes are at the intersection of obesity and colorectal cancer. Moreover, they suggest that hormone replacement that restores GUCY2C signaling may be a novel therapeutic paradigm to prevent both hyperphagia and intestinal tumorigenesis in obesity.
Collapse
Affiliation(s)
- Erik S Blomain
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Dante J Merlino
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Amanda M Pattison
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Adam E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
44
|
Martins L, Seoane-Collazo P, Contreras C, González-García I, Martínez-Sánchez N, González F, Zalvide J, Gallego R, Diéguez C, Nogueiras R, Tena-Sempere M, López M. A Functional Link between AMPK and Orexin Mediates the Effect of BMP8B on Energy Balance. Cell Rep 2016; 16:2231-2242. [PMID: 27524625 PMCID: PMC4999418 DOI: 10.1016/j.celrep.2016.07.045] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 06/16/2016] [Accepted: 07/15/2016] [Indexed: 11/30/2022] Open
Abstract
AMP-activated protein kinase (AMPK) in the ventromedial nucleus of the hypothalamus (VMH) and orexin (OX) in the lateral hypothalamic area (LHA) modulate brown adipose tissue (BAT) thermogenesis. However, whether these two molecular mechanisms act jointly or independently is unclear. Here, we show that the thermogenic effect of bone morphogenetic protein 8B (BMP8B) is mediated by the inhibition of AMPK in the VMH and the subsequent increase in OX signaling via the OX receptor 1 (OX1R). Accordingly, the thermogenic effect of BMP8B is totally absent in ox-null mice. BMP8B also induces browning of white adipose tissue (WAT), its thermogenic effect is sexually dimorphic (only observed in females), and its impact on OX expression and thermogenesis is abolished by the knockdown of glutamate vesicular transporter 2 (VGLUT2), implicating glutamatergic signaling. Overall, our data uncover a central network controlling energy homeostasis that may be of considerable relevance for obesity and metabolic disorders. Central BMP8B modulates BAT thermogenesis and browning of WAT AMPK in the VMH mediates central BMP8B actions OX in the LHA mediates central BMP8B actions The AMPK(VMH)-OX(LHA) axis is a functional neuronal pathway regulating energy balance
Collapse
Affiliation(s)
- Luís Martins
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Patricia Seoane-Collazo
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Cristina Contreras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Ismael González-García
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Noelia Martínez-Sánchez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Francisco González
- Department of Surgery, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Service of Ophthalmology, Complejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela 15706, Spain
| | - Juan Zalvide
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
| | - Rosalía Gallego
- Department of Morphological Sciences, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain; Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Reina Sofía, 14004 Córdoba, Spain; FiDiPro Program, Department of Physiology, University of Turku, Kiinamyllynkatu10, 20520 Turku, Finland
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain.
| |
Collapse
|
45
|
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, Nogueiras R. Pharmacological and Genetic Manipulation of p53 in Brown Fat at Adult But Not Embryonic Stages Regulates Thermogenesis and Body Weight in Male Mice. Endocrinology 2016; 157:2735-49. [PMID: 27183316 DOI: 10.1210/en.2016-1209] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
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.
Collapse
Affiliation(s)
- Omar Al-Massadi
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Begoña Porteiro
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Doreen Kuhlow
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Markus Köhler
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - María J Gonzalez-Rellan
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Montserrat Garcia-Lavandeira
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Esther Díaz-Rodríguez
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Mar Quiñones
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Ana Senra
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Clara V Alvarez
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Miguel López
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Carlos Diéguez
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Tim J Schulz
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| | - Rubén Nogueiras
- Department of Physiology (O.A.-M., B.P., M.J.G.-R., M.G.-L., E.D.R., M.Q., A.S., C.V.A., M.L., C.D., R.N.), Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (O.A.-M., B.P., M.J.G.-R., M.Q., M.L., C.D., R.N.), Santiago de Compostela 15706, Spain; Department of Adipocyte Development and Nutrition (D.K., M.K., T.J.S.), German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; and German Center for Diabetes Research (T.J.S.), München-Neuherberg 85764, Germany
| |
Collapse
|
46
|
Kim GW, Lin JE, Snook AE, Aing AS, Merlino DJ, Li P, Waldman SA. Calorie-induced ER stress suppresses uroguanylin satiety signaling in diet-induced obesity. Nutr Diabetes 2016; 6:e211. [PMID: 27214655 PMCID: PMC4895379 DOI: 10.1038/nutd.2016.18] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/16/2016] [Accepted: 04/07/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND/OBJECTIVES The uroguanylin-GUCY2C gut-brain axis has emerged as one component regulating feeding, energy homeostasis, body mass and metabolism. Here, we explore a role for this axis in mechanisms underlying diet-induced obesity (DIO). SUBJECTS/METHODS Intestinal uroguanylin expression and secretion, and hypothalamic GUCY2C expression and anorexigenic signaling, were quantified in mice on high-calorie diets for 14 weeks. The role of endoplasmic reticulum (ER) stress in suppressing uroguanylin in DIO was explored using tunicamycin, an inducer of ER stress, and tauroursodeoxycholic acid (TUDCA), a chemical chaperone that inhibits ER stress. The impact of consumed calories on uroguanylin expression was explored by dietary manipulation. The role of uroguanylin in mechanisms underlying obesity was examined using Camk2a-Cre-ER(T2)-Rosa-STOP(loxP/loxP)-Guca2b mice in which tamoxifen induces transgenic hormone expression in brain. RESULTS DIO suppressed intestinal uroguanylin expression and eliminated its postprandial secretion into the circulation. DIO suppressed uroguanylin through ER stress, an effect mimicked by tunicamycin and blocked by TUDCA. Hormone suppression by DIO reflected consumed calories, rather than the pathophysiological milieu of obesity, as a diet high in calories from carbohydrates suppressed uroguanylin in lean mice, whereas calorie restriction restored uroguanylin in obese mice. However, hypothalamic GUCY2C, enriched in the arcuate nucleus, produced anorexigenic signals mediating satiety upon exogenous agonist administration, and DIO did not impair these responses. Uroguanylin replacement by transgenic expression in brain repaired the hormone insufficiency and reconstituted satiety responses opposing DIO and its associated comorbidities, including visceral adiposity, glucose intolerance and hepatic steatosis. CONCLUSIONS These studies reveal a novel pathophysiological mechanism contributing to obesity in which calorie-induced suppression of intestinal uroguanylin impairs hypothalamic mechanisms regulating food consumption through loss of anorexigenic endocrine signaling. The correlative therapeutic paradigm suggests that, in the context of hormone insufficiency with preservation of receptor sensitivity, obesity may be prevented or treated by GUCY2C hormone replacement.
Collapse
Affiliation(s)
- G W Kim
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | - J E Lin
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | - A E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | - A S Aing
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | - D J Merlino
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | - P Li
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - S A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
47
|
Guanylin and uroguanylin stimulate lipolysis in human visceral adipocytes. Int J Obes (Lond) 2016; 40:1405-15. [PMID: 27108812 DOI: 10.1038/ijo.2016.66] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 03/02/2016] [Accepted: 03/20/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND/OBJECTIVES Uroguanylin and guanylin are secreted by intestinal epithelial cells as prohormones postprandially and act on the hypothalamus to induce satiety. The impact of obesity and obesity-associated type 2 diabetes (T2D) on proguanylin and prouroguanylin expression/secretion as well as the potential role of guanylin and uroguanylin in the control of lipolysis in humans was evaluated. SUBJECTS/METHODS Circulating and gastrointestinal expression of proguanylin (GUCA2A) and prouroguanylin (GUCA2B) were measured in 134 subjects. In addition, plasma proguanylin and prouroguanylin were measured before and after weight loss achieved either by Roux-en-Y gastric bypass (RYGB) (n=24) or after a conventional diet (n=15). The effect of guanylin and uroguanylin (1-100 nmol l(-1)) on lipolysis was determined in vitro in omental adipocytes. RESULTS Circulating concentrations of prouroguanylin, but not proguanylin, were decreased in obesity in relation to adiposity. Weight loss achieved by RYGB increased plasma proguanylin and prouroguanylin. Obese T2D individuals showed higher expression of intestinal GUCA2A as well as of the receptors of the guanylin system, GUCY2C and GUCY2D, in omental adipocytes. The incubation with guanylin and uroguanylin significantly stimulated lipolysis in differentiated omental adipocytes, as evidenced by hormone-sensitive lipase phosphorylation at Ser563, an increase in fatty acids and glycerol release together with an upregulation of several lipolysis-related genes, including AQP3, AQP7, FATP1 or CD36. CONCLUSIONS Both guanylin and uroguanylin trigger lipolysis in human visceral adipocytes. Given the lipolytic action of the guanylin system on visceral adipocytes, the herein reported decrease of circulating prouroguanylin concentrations in obese patients may have a role in excessive fat accumulation in obesity.
Collapse
|
48
|
Barja-Fernández S, Folgueira C, Castelao C, Al-Massadi O, Bravo SB, Garcia-Caballero T, Leis R, Pardo M, Casanueva FF, Seoane LM. FNDC5 is produced in the stomach and associated to body composition. Sci Rep 2016; 6:23067. [PMID: 26961074 PMCID: PMC4785389 DOI: 10.1038/srep23067] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 02/25/2016] [Indexed: 12/19/2022] Open
Abstract
The fibronectin type III domain-containing protein 5 (FNDC5) discovered in 2002 has recently gained attention due to its potential role in protecting against obesity. In rat, no data exist regarding FNDC5 production and regulation in the stomach. The aim of the present work was to determine the expression of FNDC5 in the rat stomach and its potential regulation by body composition. The present data shows FNDC5 gene expression in the gastric mucosa. Immunohistochemical studies found FNDC5 immunopositivity in chief cells of gastric tissue. By the use of three different antibodies FNDC5 was found expressed in gastric mucosa and secreted by the stomach. The rate of gastric FNDC5 secretion parallels the circulating levels of FNDC5. The body fat mass increase after intervention with high fat diet coincided with a decrease in the secretion of FNDC5 from the stomach and a diminution in the FNDC5 circulating levels. In summary, the present data shows, for the first time, the expression of FNDC5 in the stomach of rats and its regulation by body composition, suggesting a potential role of gastric FNDC5 in energy homeostasis.
Collapse
Affiliation(s)
- S. Barja-Fernández
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS). Santiago de Compostela, Spain
- Departamento de Pediatría, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS). Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, Santiago de Compostela, Spain
| | - C. Folgueira
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS). Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, Santiago de Compostela, Spain
- Department of Physiology, Research Centre of Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS). Santiago de Compostela, Spain
| | - C. Castelao
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS). Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, Santiago de Compostela, Spain
| | - O. Al-Massadi
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, Santiago de Compostela, Spain
- Department of Physiology, Research Centre of Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS). Santiago de Compostela, Spain
| | - S. B. Bravo
- Unidad de Proteómica, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago CHUS/SERGAS. Santiago de Compostela, Spain
| | - T. Garcia-Caballero
- Departamento de Ciencias Morfológicas, Facultad de Medicina, Universidade de Santiago de Compostela. Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS). Santiago de Compostela, Spain
| | - R. Leis
- Departamento de Pediatría, Universidade de Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS). Santiago de Compostela, Spain
| | - M. Pardo
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, Santiago de Compostela, Spain
- Grupo Obesidomica, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS). Santiago de Compostela, Spain
| | - F. F. Casanueva
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, Santiago de Compostela, Spain
- Laboratorio de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
| | - L. M. Seoane
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS). Santiago de Compostela, Spain
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, Santiago de Compostela, Spain
| |
Collapse
|