1
|
Habek N, Ratko M, Sedmak D, Banovac I, Crljen V, Kordić M, Radmilović M, Škokić S, Tkalčić M, Mažuranić A, Bubalo P, Škavić P, Ljubić S, Rahelić D, Dugandžić A. Brain-derived uroguanylin as a regulator of postprandial brown adipose tissue activation: a potential therapeutic approach for metabolic disorders. Front Pharmacol 2025; 16:1569163. [PMID: 40351439 PMCID: PMC12062040 DOI: 10.3389/fphar.2025.1569163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/26/2025] [Indexed: 05/14/2025] Open
Abstract
Background Preclinical and clinical research of insulin resistance and glucose homeostasis in metabolic disorders are essential. In this study, we aim to determine the expression of uroguanylin (UGN) in the mouse and human brain, its regulatory mechanisms, and its significance to patients with obesity and type 2 diabetes (T2D). Methods UGN expression, regulation, and its correlation with feeding status and obesity in the mouse and human brain were analyzed at the mRNA level using RT-PCR, qPCR, and in situ hybridization and at the protein level using Western blot, ELISA, and immunohistochemistry. Brown adipose tissue (BAT) activity was measured using infrared thermography. The volume of interscapular brown adipose tissue in mice was assessed by magnetic resonance imaging. Results UGN was expressed in both the mouse and human brain, and its expression was regulated by feeding. In the human prefrontal cortex, UGN was expressed in several interneuron subpopulations across all cortical layers. In Brodmann area (BA) 10, prouroguanylin (proUGN) expression was not regulated by feeding in obesity, whereas this regulation still persisted in BA9. In mice, centrally applied UGN and its analog linaclotide, affecting the hypothalamus, induced both acute and chronic activation of BAT, which decreases the plasma glucose concentration. However, in obesity, proUGN expression was reduced in the human hypothalamus, suggesting reduced postprandial glucose consumption in BAT. Similarly, centrally applied analog of glucagon-like peptide 1 (GLP-1-liraglutide) affected proUGN expression and was associated with increased basal BAT activity but reduced BAT activation after a meal in patients with T2D receiving GLP-1 therapy. Conclusion Postprandial BAT activation is regulated by brain-derived UGN, which could serve as a novel therapeutic approach to enhance BAT activity in patients with obesity and T2D to improve postprandial glucose regulation.
Collapse
Affiliation(s)
- Nikola Habek
- 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
- 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
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivan Banovac
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Vladiana Crljen
- 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
| | | | - Marina Radmilović
- 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
| | - 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
| | - Spomenka Ljubić
- Department of Diabetes, Vuk Vrhovac University Clinic for Diabetes, Endocrinology and Metabolic Diseases, Merkur University Hospital, Zagreb, Croatia
| | - Dario Rahelić
- Department of Diabetes, Vuk Vrhovac University Clinic for Diabetes, Endocrinology and Metabolic Diseases, Merkur University Hospital, 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
|
2
|
Xiang Y, Sun J, Ma G, Dai X, Meng Y, Fu C, Zhang Y, Zhao Q, Li J, Zhang S, Zheng Z, Li X, Fu L, Li K, Qi X. Integrating Multi-Omics Data to Identify Key Functional Variants Affecting Feed Efficiency in Large White Boars. Genes (Basel) 2024; 15:980. [PMID: 39202341 PMCID: PMC11353296 DOI: 10.3390/genes15080980] [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: 06/22/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
Optimizing feed efficiency through the feed conversion ratio (FCR) is paramount for economic viability and sustainability. In this study, we integrated RNA-seq, ATAC-seq, and genome-wide association study (GWAS) data to investigate key functional variants associated with feed efficiency in pigs. Identification of differentially expressed genes in the duodenal and muscle tissues of low- and high-FCR pigs revealed that pathways related to digestion of dietary carbohydrate are responsible for differences in feed efficiency between individuals. Differential open chromatin regions identified by ATAC-seq were linked to genes involved in glycolytic and fatty acid processes. GWAS identified 211 significant single-nucleotide polymorphisms associated with feed efficiency traits, with candidate genes PPP1R14C, TH, and CTSD. Integration of duodenal ATAC-seq data and GWAS data identified six key functional variants, particularly in the 1500985-1509676 region on chromosome 2. In those regions, CTSD was found to be highly expressed in the duodenal tissues of pigs with a high feed conversion ratio, suggesting its role as a potential target gene. Overall, the integration of multi-omics data provided insights into the genetic basis of feed efficiency, offering valuable information for breeding more efficient pig breeds.
Collapse
Affiliation(s)
- Yue Xiang
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Jiahui Sun
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Guojian Ma
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Xueting Dai
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Yuan Meng
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
| | - Chong Fu
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Yan Zhang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Qiulin Zhao
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Jingjin Li
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
| | - Saixian Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
| | - Zhuqing Zheng
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Xinyun Li
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Liangliang Fu
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Kui Li
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
| | - Xiaolong Qi
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
| |
Collapse
|
3
|
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
|
4
|
Barton JR, Londregan AK, Alexander TD, Entezari AA, Covarrubias M, Waldman SA. Enteroendocrine cell regulation of the gut-brain axis. Front Neurosci 2023; 17:1272955. [PMID: 38027512 PMCID: PMC10662325 DOI: 10.3389/fnins.2023.1272955] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Enteroendocrine cells (EECs) are an essential interface between the gut and brain that communicate signals about nutrients, pain, and even information from our microbiome. EECs are hormone-producing cells expressed throughout the gastrointestinal epithelium and have been leveraged by pharmaceuticals like semaglutide (Ozempic, Wegovy), terzepatide (Mounjaro), and retatrutide (Phase 2) for diabetes and weight control, and linaclotide (Linzess) to treat irritable bowel syndrome (IBS) and visceral pain. This review focuses on role of intestinal EECs to communicate signals from the gut lumen to the brain. Canonically, EECs communicate information about the intestinal environment through a variety of hormones, dividing EECs into separate classes based on the hormone each cell type secretes. Recent studies have revealed more diverse hormone profiles and communication modalities for EECs including direct synaptic communication with peripheral neurons. EECs known as neuropod cells rapidly relay signals from gut to brain via a direct communication with vagal and primary sensory neurons. Further, this review discusses the complex information processing machinery within EECs, including receptors that transduce intraluminal signals and the ion channel complement that govern initiation and propagation of these signals. Deeper understanding of EEC physiology is necessary to safely treat devastating and pervasive conditions like irritable bowel syndrome and obesity.
Collapse
Affiliation(s)
- Joshua R. Barton
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Annie K. Londregan
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Tyler D. Alexander
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, United States
| | - Ariana A. Entezari
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Manuel Covarrubias
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, United States
| | - Scott A. Waldman
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
5
|
Fernández-Sáez EM, Losarcos M, Becerril S, Valentí V, Moncada R, Martín M, Burrell MA, Catalán V, Gómez-Ambrosi J, Mugueta C, Colina I, Silva C, Escalada J, Frühbeck G, Rodríguez A. Uroguanylin prevents hepatic steatosis, mitochondrial dysfunction and fibrosis in obesity-associated NAFLD. Metabolism 2023; 147:155663. [PMID: 37517791 DOI: 10.1016/j.metabol.2023.155663] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND The biological mediators supporting the resolution of liver steatosis, inflammation and fibrosis after bariatric surgery in patients with obesity and NAFLD remain unclear. We sought to analyze whether uroguanylin and guanylin, two gut hormones involved in the regulation of satiety, food preference and adiposity, are involved in the amelioration of obesity-associated NAFLD after bariatric surgery. METHODS Proguanylin (GUCA2A) and prouroguanylin (GUCA2B) were measured in 214 participants undergoing bariatric surgery with biopsy-proven NAFLD diagnosis. Pathways involved in lipid metabolism, mitochondrial network and fibrogenesis were evaluated in liver biopsies (n = 137). The effect of guanylin and uroguanylin on these metabolic functions was assessed in HepG2 hepatocytes and LX-2 hepatic stellate cells (HSC) under lipotoxic and profibrogenic conditions. RESULTS Plasma and hepatic expression of GUCA2B were decreased in obesity-associated NAFLD. Both GUCA2A and GUCA2B levels were increased after sleeve gastrectomy and Roux-en-Y gastric bypass in parallel to the improved liver function. The liver of patients with type 2 diabetes showed impaired mitochondrial β-oxidation, biogenesis, dynamics as well as increased fibrosis. Uroguanylin diminished the lipotoxicity in palmitate-treated HepG2 hepatocytes, evidenced by decresased steatosis and lipogenic factors, as well as increased mitochondrial network expression, AMPK-induced β-oxidation and oxygen consumption rate. Additionally, uroguanylin, but not guanylin, reversed HSC myofibroblast transdifferentiation as well as fibrogenesis after TGF-β1 stimulation. CONCLUSIONS Uroguanylin constitutes a protective factor against lipotoxicity, mitochondrial dysfunction and fibrosis. Increased GUCA2B levels might contribute to improve liver injury in patients with obesity-associated NAFLD after bariatric surgery.
Collapse
Affiliation(s)
| | - Maite Losarcos
- 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, 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
| | - Marina Martín
- Department of Pathology, Anatomy and Physiology, University of 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
| | - 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
| | - Carmen Mugueta
- Department of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
| | - Inmaculada Colina
- Department of Internal Medicine, Clínica Universidad de Navarra, 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
| | - 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
| | - 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
| | - 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
|
6
|
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
|
7
|
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 PMCID: PMC12076115 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
|
8
|
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
|
9
|
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
|
10
|
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: 10] [Impact Index Per Article: 3.3] [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
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
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
|
17
|
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: 24] [Impact Index Per Article: 4.0] [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
|
18
|
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
|
19
|
Mann EA, Sugimoto C, Williams MT, Vorhees CV. Mouse knockout of guanylyl cyclase C: Recognition memory deficits in the absence of activity changes. GENES BRAIN AND BEHAVIOR 2019; 18:e12573. [PMID: 30953414 DOI: 10.1111/gbb.12573] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/19/2019] [Accepted: 04/03/2019] [Indexed: 12/28/2022]
Abstract
Guanylyl cyclase C (GC-C) is found in brain regions where dopamine is expressed. We characterized a mouse in which GC-C was knocked out (KO) that was reported to be a model of attention deficit hyperactivity disorder (ADHD). We re-examined this model and controlled for litter effects, used 16 to 23 mice per genotype per sex and assessed an array of behavioral and neurochemical outcomes. GC-C KO mice showed no phenotypic differences from wild-type mice on most behavioral tests, or on striatal or hippocampal monoamines, and notably no evidence of an ADHD-like phenotype. KO mice were impaired on novel object recognition, had decreased tactile startle but not acoustic startle, and females had increased latency on cued training trials in the Morris water maze, but not hidden platform spatial learning trials. Open-field activity showed small differences in females but not males. The data indicate that the GC-C KO mouse with proper controls and sample sizes has a moderate cognitive and startle phenotype but has no ADHD-like phenotype.
Collapse
Affiliation(s)
- Elizabeth A Mann
- Divisions of Urology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Chiho Sugimoto
- Neurology and Cincinnati Children's Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Michael T Williams
- Neurology and Cincinnati Children's Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Charles V Vorhees
- Neurology and Cincinnati Children's Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
20
|
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
|
21
|
Mucke HA. Patent Highlights August-September 2018. Pharm Pat Anal 2019; 8:7-14. [PMID: 30869551 DOI: 10.4155/ppa-2018-0033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/06/2019] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
Differentially expressed genes in the caecal and colonic mucosa of Landrace finishing pigs with high and low food conversion ratios. Sci Rep 2017; 7:14886. [PMID: 29097775 PMCID: PMC5668291 DOI: 10.1038/s41598-017-14568-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 09/08/2017] [Indexed: 02/08/2023] Open
Abstract
The feed conversion ratio (FCR) is an essential economic trait for pig production, and is directly related to feed efficiency. Studies identifying the differential expression of functional genes involved in biological and molecular mechanisms in the intestine in relation to growth performance are rare. In this study, RNA-Seq was used to identify transcriptomes in caecal and colonic mucosal tissues in order to determine the differential expression of genes from two full-sibling pairs and two half-sibling pairs of Landrace finishing pigs with opposing FCR phenotypes. In total, 138 (comparison of high and low FCR in caecal mucosa), 64 (comparison of high and low FCR in colonic mucosa), and 165 (contrast between the caecal and colonic mucosa) differentially expressed genes were identified. Some of these genes were functionally related to energy and lipid metabolism, particularly short chain fatty acids metabolism, as well as gastrointestinal peristalsis and ion transport. Functional annotation were performed to identify differentially expressed genes, such as GUCA2A, GUCA2B, HSP70.2, NOS2, PCK1, SLCs, and CYPs, which may positively influence feed efficiency in Landrace pigs. These differentially expressed genes need to be further tested for candidate genes that are related to feed efficiency.
Collapse
|
26
|
Monteiro MP, Batterham RL. The Importance of the Gastrointestinal Tract in Controlling Food Intake and Regulating Energy Balance. Gastroenterology 2017; 152:1707-1717.e2. [PMID: 28193513 DOI: 10.1053/j.gastro.2017.01.053] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/31/2016] [Accepted: 01/03/2017] [Indexed: 12/16/2022]
Abstract
The gastrointestinal tract, the key interface between ingested nutrients and the body, plays a critical role in regulating energy homeostasis. Gut-derived signals convey information regarding incoming nutrients to the brain, initiating changes in eating behavior and energy expenditure, to maintain energy balance. Here we review hormonal, neural, and nutrient signals emanating from the gastrointestinal tract and evidence for their role in controlling feeding behavior. Mechanistic studies that have utilized pharmacologic and/or transgenic approaches targeting an individual hormone/mediator have yielded somewhat disappointing body weight changes, often leading to the hormone/mediator in question being dismissed as a potential obesity therapy. However, the recent finding of sustained weight reduction in response to systemic administration of a long-acting analog of the gut-hormone glucagon-like peptide-1 highlights the therapeutic potential of gut-derived signals acting via nonphysiologic mechanisms. Thus, we also review therapeutics strategies being utilized or developed to leverage gastrointestinal signals in order to treat obesity.
Collapse
Affiliation(s)
- Mariana P Monteiro
- Clinical and Experimental Endocrinology, Unit for Multidisciplinary Research in Biomedicine, Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Portugal; Centre for Obesity Research, University College London, London, United Kingdom; University College London Hospitals Bariatric Centre for Weight Management and Metabolic Surgery, London, United Kingdom
| | - Rachel L Batterham
- Centre for Obesity Research, University College London, London, United Kingdom; University College London Hospitals Bariatric Centre for Weight Management and Metabolic Surgery, London, United Kingdom; National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, United Kingdom.
| |
Collapse
|
27
|
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
|
28
|
Abstract
cGMP controls many cellular functions ranging from growth, viability, and differentiation to contractility, secretion, and ion transport. The mammalian genome encodes seven transmembrane guanylyl cyclases (GCs), GC-A to GC-G, which mainly modulate submembrane cGMP microdomains. These GCs share a unique topology comprising an extracellular domain, a short transmembrane region, and an intracellular COOH-terminal catalytic (cGMP synthesizing) region. GC-A mediates the endocrine effects of atrial and B-type natriuretic peptides regulating arterial blood pressure/volume and energy balance. GC-B is activated by C-type natriuretic peptide, stimulating endochondral ossification in autocrine way. GC-C mediates the paracrine effects of guanylins on intestinal ion transport and epithelial turnover. GC-E and GC-F are expressed in photoreceptor cells of the retina, and their activation by intracellular Ca(2+)-regulated proteins is essential for vision. Finally, in the rodent system two olfactorial GCs, GC-D and GC-G, are activated by low concentrations of CO2and by peptidergic (guanylins) and nonpeptidergic odorants as well as by coolness, which has implications for social behaviors. In the past years advances in human and mouse genetics as well as the development of sensitive biosensors monitoring the spatiotemporal dynamics of cGMP in living cells have provided novel relevant information about this receptor family. This increased our understanding of the mechanisms of signal transduction, regulation, and (dys)function of the membrane GCs, clarified their relevance for genetic and acquired diseases and, importantly, has revealed novel targets for therapies. The present review aims to illustrate these different features of membrane GCs and the main open questions in this field.
Collapse
Affiliation(s)
- Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
29
|
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: 37] [Impact Index Per Article: 4.1] [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
|
30
|
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
|
31
|
Blomain ES, Pattison AM, Waldman SA. GUCY2C ligand replacement to prevent colorectal cancer. Cancer Biol Ther 2016; 17:713-8. [PMID: 27104761 DOI: 10.1080/15384047.2016.1178429] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite advances in screening and prevention strategies, colorectal cancer (CRC) remains the second-leading cause of cancer-related death in the United States. Given this continued public health burden of CRC, there is a clear need for improved disease prevention. CRC initiates and progresses over decades, canonically proceeding via a series of stepwise molecular events that turn a normal epithelium into a dysfunctional epithelium, then subsequently into an adenoma, and finally an invasive adenocarcinoma. An emerging paradigm suggests that guanylyl cyclase C (GUCY2C) functions as a tumor suppressor in the intestine, and that the loss of hormone ligands for this receptor causes epithelial dysfunction and represents an important step in the disease process. In that context, GUCY2C ligand replacement therapy has been proposed as a strategy to prevent colorectal cancer, a translational opportunity that is underscored by the recent regulatory approval of the oral GUCY2C ligand linaclotide (Linzess™, Forest Laboratories and Ironwood Pharmaceuticals, Inc.).
Collapse
Affiliation(s)
- Erik S Blomain
- a Department of Pharmacology and Experimental Therapeutics , Thomas Jefferson University , Philadelphia , PA , USA
| | - Amanda M Pattison
- a Department of Pharmacology and Experimental Therapeutics , Thomas Jefferson University , Philadelphia , PA , USA
| | - Scott A Waldman
- a Department of Pharmacology and Experimental Therapeutics , Thomas Jefferson University , Philadelphia , PA , USA
| |
Collapse
|
32
|
Folgueira C, Beiroa D, Callon A, Al-Massadi O, Barja-Fernandez S, Senra A, Fernø J, López M, Dieguez C, Casanueva FF, Rohner-Jeanrenaud F, Seoane LM, Nogueiras R. Uroguanylin Action in the Brain Reduces Weight Gain in Obese Mice via Different Efferent Autonomic Pathways. Diabetes 2016; 65:421-32. [PMID: 26566631 DOI: 10.2337/db15-0889] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/06/2015] [Indexed: 11/13/2022]
Abstract
The gut-brain axis is of great importance in the control of energy homeostasis. The identification of uroguanylin (UGN), a peptide released in the intestines that is regulated by nutritional status and anorectic actions, as the endogenous ligand for the guanylyl cyclase 2C receptor has revealed a new system in the regulation of energy balance. We show that chronic central infusion of UGN reduces weight gain and adiposity in diet-induced obese mice. These effects were independent of food intake and involved specific efferent autonomic pathways. On one hand, brain UGN induces brown adipose tissue thermogenesis, as well as browning and lipid mobilization in white adipose tissue through stimulation of the sympathetic nervous system. On the other hand, brain UGN augments fecal output through the vagus nerve. These findings are of relevance as they suggest that the beneficial metabolic actions of UGN through the sympathetic nervous system do not involve nondesirable gastrointestinal adverse effects, such as diarrhea. The present work provides mechanistic insights into how UGN influences energy homeostasis and suggests that UGN action in the brain represents a feasible pharmacological target in the treatment of obesity.
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, Santiago de Compostela, Spain Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain CIBER Fisiopatología de la Obesidad y Nutrición, Spain
| | - Daniel Beiroa
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain CIBER Fisiopatología de la Obesidad y Nutrición, Spain
| | - Aurelie Callon
- Laboratory of Metabolism, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Omar Al-Massadi
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain CIBER Fisiopatología de la Obesidad y Nutrición, Spain
| | - Silvia Barja-Fernandez
- 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, Santiago de Compostela, Spain CIBER Fisiopatología de la Obesidad y Nutrición, Spain Department of Pediatrics, Center for Research in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Ana Senra
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Johan Fernø
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Miguel López
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain CIBER Fisiopatología de la Obesidad y Nutrición, Spain
| | - Carlos Dieguez
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain CIBER Fisiopatología de la Obesidad y Nutrición, Spain
| | - Felipe F Casanueva
- CIBER Fisiopatología de la Obesidad y Nutrición, Spain Department of Medicine, Center for Research in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Françoise Rohner-Jeanrenaud
- Laboratory of Metabolism, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - 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, Santiago de Compostela, Spain CIBER Fisiopatología de la Obesidad y Nutrición, Spain
| | - Ruben Nogueiras
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain CIBER Fisiopatología de la Obesidad y Nutrición, Spain
| |
Collapse
|