1
|
Galsgaard KD, Modvig IM, Holst JJ. Understanding the release mechanisms and secretion patterns for glucagon-like peptide-1 using the isolated perfused intestine as a model. Biochem Soc Trans 2025; 53:BST20241062. [PMID: 39887325 DOI: 10.1042/bst20241062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 02/01/2025]
Abstract
In the gastrointestinal (GI) tract, food is digested and absorbed while GI hormones are secreted from the enteroendocrine cells (EECs). These hormones regulate food intake, glucose homeostasis, digestion, GI motility, and metabolism. Although ECCs may express more than a single hormone, the ECCs usually secrete only one or a few hormones. The pattern of EEC secretion varies along the length of the GI tract as the different EEC types are scattered in different densities along the GI tract. Following bariatric surgery, a postprandial hypersecretion of certain GI hormones occurs which contributes to the postsurgery weight loss. Mimicking this postprandial hypersecretion of GI hormones by targeting endogenous EEC secretion, using specific modulators of receptors, ion channels, and transporters found on specific EECs, to induce weight loss is a current research aim. To achieve this, a more complete understanding of the release mechanisms, expression of receptors, transporters, and the secretion pattern of the different ECC types is needed. Using the vascularly perfused intestinal model, it is possible to obtain a detailed knowledge of these release mechanisms by evaluating the effects on secretion of blocking or stimulating specific receptors, ion channels, and transporters as well as evaluating nutrient handling and absorption in each of the different sections of the intestine. This mini-review will focus on how the isolated perfused intestine has been used in our group as a model to investigate the nutrient-induced release mechanisms of ECCs with a focus on glucagon-like peptide-1 secreting cells.
Collapse
Affiliation(s)
- Katrine D Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ida M Modvig
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, 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
| |
Collapse
|
2
|
Cho H, Lim J. The emerging role of gut hormones. Mol Cells 2024; 47:100126. [PMID: 39426686 PMCID: PMC11577206 DOI: 10.1016/j.mocell.2024.100126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/13/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024] Open
Abstract
The gut is traditionally recognized as the central organ for the digestion and absorption of nutrients, however, it also functions as a significant endocrine organ, secreting a variety of hormones such as glucagon-like peptide 1, serotonin, somatostatin, and glucocorticoids. These gut hormones, produced by specialized intestinal epithelial cells, are crucial not only for digestive processes but also for the regulation of a wide range of physiological functions, including appetite, metabolism, and immune responses. While gut hormones can exert systemic effects, they also play a pivotal role in maintaining local homeostasis within the gut. This review discusses the role of the gut as an endocrine organ, emphasizing the stimuli, the newly discovered functions, and the clinical significance of gut-secreted hormones. Deciphering the emerging role of gut hormones will lead to a better understanding of gut homeostasis, innovative treatments for disorders in the gut, as well as systemic diseases.
Collapse
Affiliation(s)
- Hyeryeong Cho
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaechul Lim
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
3
|
Holst JJ, Madsbad S, Bojsen-Møller KN, Dirksen C, Svane M. New Lessons from the gut: Studies of the role of gut peptides in weight loss and diabetes resolution after gastric bypass and sleeve gastrectomy. Peptides 2024; 176:171199. [PMID: 38552903 DOI: 10.1016/j.peptides.2024.171199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
It has been known since 2005 that the secretion of several gut hormones changes radically after gastric bypass operations and, although more moderately, after sleeve gastrectomy but not after gastric banding. It has therefore been speculated that increased secretion of particularly GLP-1 and Peptide YY (PYY), which both inhibit appetite and food intake, may be involved in the weight loss effects of surgery and for improvements in glucose tolerance. Experiments involving inhibition of hormone secretion with somatostatin, blockade of their actions with antagonists, or blockade of hormone formation/activation support this notion. However, differences between results of bypass and sleeve operations indicate that distinct mechanisms may also be involved. Although the reductions in ghrelin secretion after sleeve gastrectomy would seem to provide an obvious explanation, experiments with restoration of ghrelin levels pointed towards effects on insulin secretion and glucose tolerance rather than on food intake. It seems clear that changes in GLP-1 secretion are important for insulin secretion after bypass and appear to be responsible for postbariatric hypoglycemia in glucose-tolerant individuals; however, with time the improvements in insulin sensitivity, which in turn are secondary to the weight loss, may be more important. Changes in bile acid metabolism do not seem to be of particular importance in humans.
Collapse
Affiliation(s)
- Jens Juul Holst
- The NovoNordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Denmark.
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Denmark
| | | | - Carsten Dirksen
- Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Denmark
| | - Maria Svane
- Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Denmark
| |
Collapse
|
4
|
Berg P, Svendsen SL, Ayasse N, Sorensen MV, Leipziger J. Secretin: a hormone for HCO 3- homeostasis. Pflugers Arch 2024; 476:545-554. [PMID: 38221598 DOI: 10.1007/s00424-024-02906-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/16/2024]
Abstract
Secretin is a key hormone of the intestinal phase of digestion which activates pancreatic, bile duct and Brunner gland HCO3- secretion. Recently, the secretin receptor (SCTR) was also found in the basolateral membrane of the beta-intercalated cell (B-IC) of the collecting duct. Experimental addition of secretin triggers a pronounced activation of urinary HCO3- excretion, which is fully dependent on key functional proteins of the B-IC, namely apical pendrin and CFTR and the basolateral SCTR. Recent studies demonstrated that the SCTR knock-out mouse is unable to respond to an acute base load. Here, SCTR KO mice could not rapidly increase urine base excretion, developed prolonged metabolic alkalosis and exhibited marked compensatory hypoventilation. Here, we review the physiological effects of secretin with distinct focus on how secretin activates renal HCO3- excretion. We describe its new function as a hormone for HCO3- homeostasis.
Collapse
Affiliation(s)
- Peder Berg
- Department of Biomedicine, Physiology, Health, Aarhus University, Høegh-Guldbergsgade 10, Bld. 1115, 8000, Aarhus C, Denmark
| | - Samuel L Svendsen
- Department of Biomedicine, Physiology, Health, Aarhus University, Høegh-Guldbergsgade 10, Bld. 1115, 8000, Aarhus C, Denmark
| | - Niklas Ayasse
- Vth Department of Medicine, University Hospital Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Mads Vaarby Sorensen
- Department of Biomedicine, Physiology, Health, Aarhus University, Høegh-Guldbergsgade 10, Bld. 1115, 8000, Aarhus C, Denmark
| | - Jens Leipziger
- Department of Biomedicine, Physiology, Health, Aarhus University, Høegh-Guldbergsgade 10, Bld. 1115, 8000, Aarhus C, Denmark.
| |
Collapse
|
5
|
Rehfeld JF, Goetze JP. Gastrointestinal hormones: History, biology, and measurement. Adv Clin Chem 2024; 118:111-154. [PMID: 38280804 DOI: 10.1016/bs.acc.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2024]
Abstract
This chapter attempts to provide an all-round picture of a dynamic and major branch of modern endocrinology, i.e. the gastrointestinal endocrinology. The advances during the last half century in our understanding of the dimensions and diversity of gut hormone biology - inside as well as outside the digestive tract - are astounding. Among major milestones are the dual brain-gut relationship, i.e. the comprehensive expression of gastrointestinal hormones as potent transmitters in central and peripheral neurons; the hormonal signaling from the enteroendocrine cells to the brain and other extraintestinal targets; the role of gut hormones as growth and fertility factors; and the new era of gut hormone-derived drugs. Accordingly, gastrointestinal hormones have pathogenetic roles in major metabolic disorders (diabetes mellitus and obesity); in tumor development (common cancers, sarcomas, and neuroendocrine tumors); and in cerebral diseases (anxiety, panic attacks, and probably eating disorders). Such clinical aspects require accurate pathogenetic and diagnostic measurements of gastrointestinal hormones - an obvious responsibility for clinical chemistry/biochemistry. In order to obtain a necessary insight into today's gastrointestinal endocrinology, the chapter will first describe the advances in gastrointestinal endocrinology in a historical context. The history provides a background for the subsequent description of the present biology of gastrointestinal hormones, and its biomedical consequences - not least for clinical chemistry/biochemistry with its specific responsibility for selection of appropriate assays and reliable measurements.
Collapse
Affiliation(s)
- Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | - Jens P Goetze
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Zhang F, Tao Z, Chen C, Chow BKC. Effects of secretin gene knockout on the diversity, composition, and function of gut microbiota in adult male mice. Front Cell Infect Microbiol 2023; 13:1257857. [PMID: 38156312 PMCID: PMC10753818 DOI: 10.3389/fcimb.2023.1257857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/16/2023] [Indexed: 12/30/2023] Open
Abstract
The gut microbiota plays a vital role in maintaining gastrointestinal homeostasis, however, whether it is influenced by gut hormones remains unknown. Secretin is a well-known gastrointestinal hormone produced by enteroendocrine S cells. This study utilized 16S rRNA amplicon sequencing to characterize the effect of SCT deficiency on the gut microbiota. Our results show that systemic SCT knockout alters the composition and abundance of the mouse gut microbiota but does not affect fecal short-chain fatty acids and lipids concentrations. At the genus level, the abundance of Turicibacter, Bacteroides, Ruminococcu, Romboutsia, Asaccharobacter, and Parasutterella increased in SCT-/- mice, whereas the abundance of Akkermansia and Escherichia decreased. Functional prediction results showed that lack of SCT reduced the abundance of carbohydrate metabolism-related pathways but increased the abundance of linoleic acid metabolism and branched-chain amino acid degradation. Overall, systemic SCT knockout had only minor effects on gut microbiota composition and function in adult male mice fed a standard chow diet.
Collapse
Affiliation(s)
- Fengwei Zhang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | | | | | - Billy Kwok Chong Chow
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
7
|
Rafaqat S, Hafeez R, Mairaj R, Saleem A, Rafaqat S. Pancreatic biomarkers: role in diabetes mellitus. JOURNAL OF PANCREATOLOGY 2023; 6:169-177. [DOI: 10.1097/jp9.0000000000000137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Diabetes mellitus refers to a group of diseases that cause high blood sugar levels. The most common type is type 2 diabetes, which is caused by insulin resistance and inadequate insulin production. However, diabetes can also result from conditions affecting the exocrine pancreas. Both type 1 and type 2 diabetes patients may experience changes in their pancreatic exocrine function, leading to reduced levels of fecal elastase-1 in many cases. This review article focuses on the role of specific pancreatic biomarkers in diabetes mellitus, including cholecystokinin, trypsin, chymotrypsin, carboxypeptidase, amylase, lipase, secretin, elastase-1, and retinol-binding protein 4 about recent advances and discoveries, significant gaps in the literature, current debates, and potential directions for future research related to these biomarkers about diabetes mellitus. This review article discusses various biomarkers related to pancreatic exocrine and endocrine function and their implications in diabetes. It suggests that gut cholecystokinin may play a role in lowering glucose synthesis through a neural network and resistance to it could contribute to hyperglycemia in diabetic patients. It also discusses the use of various markers such as serum trypsin concentration, amylase and lipase levels, pancreatic elastase levels, and fasting secretin levels to assess pancreatic exocrine function. Additionally, the article explores the role of carboxypeptidase E in the endocrine and neurological systems and its association with disorders. Moreover, it also highlights the involvement of retinol-binding protein 4 in the development of type 2 diabetes and insulin resistance.
Collapse
Affiliation(s)
- Sana Rafaqat
- Department of Biotechnology (Molecular Human Genetics), Lahore College for Women University, Lahore 54000, Pakistan
| | - Ramsha Hafeez
- Department of Zoology (Fisheries and Aquaculture), Lahore College for Women University, Lahore 54000, Pakistan
| | - Rida Mairaj
- Department of Zoology (Fisheries and Nanotechnology), Lahore College for Women University, Lahore 54000, Pakistan
| | - Abeerah Saleem
- Department of Zoology (Molecular Physiology), Lahore College for Women University, 54000, Lahore, Pakistan
| | - Saira Rafaqat
- Department of Zoology (Molecular Physiology), Lahore College for Women University, 54000, Lahore, Pakistan
| |
Collapse
|
8
|
Bany Bakar R, Reimann F, Gribble FM. The intestine as an endocrine organ and the role of gut hormones in metabolic regulation. Nat Rev Gastroenterol Hepatol 2023; 20:784-796. [PMID: 37626258 DOI: 10.1038/s41575-023-00830-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 08/27/2023]
Abstract
Gut hormones orchestrate pivotal physiological processes in multiple metabolically active tissues, including the pancreas, liver, adipose tissue, gut and central nervous system, making them attractive therapeutic targets in the treatment of obesity and type 2 diabetes mellitus. Most gut hormones are derived from enteroendocrine cells, but bioactive peptides that are derived from other intestinal epithelial cell types have also been implicated in metabolic regulation and can be considered gut hormones. A deeper understanding of the complex inter-organ crosstalk mediated by the intestinal endocrine system is a prerequisite for designing more effective drugs that are based on or target gut hormones and their receptors, and extending their therapeutic potential beyond obesity and diabetes mellitus. In this Review, we present an overview of gut hormones that are involved in the regulation of metabolism and discuss their action in the gastrointestinal system and beyond.
Collapse
Affiliation(s)
- Rula Bany Bakar
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Frank Reimann
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Fiona M Gribble
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Cambridge, UK.
| |
Collapse
|
9
|
Gilliam-Vigh H, Jorsal T, Nielsen SW, Forman JL, Pedersen J, Poulsen SS, Vilsbøll T, Knop FK. Expression of Secretin and its Receptor Along the Intestinal Tract in Type 2 Diabetes Patients and Healthy Controls. J Clin Endocrinol Metab 2023; 108:e1597-e1602. [PMID: 37335970 DOI: 10.1210/clinem/dgad372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/10/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
CONTEXT The hormone secretin (SCT) is released from intestinal S cells and acts via the SCT receptor (SCTR). Circulating SCT levels increase after Roux-en-Y gastric bypass surgery and have been associated with massive weight loss and high remission rates of type 2 diabetes (T2D) linked to these operations. Exogenous SCT was recently shown to reduce ad libitum food intake in healthy volunteers. OBJECTIVE To understand SCT biology and its potential role in T2D pathophysiology, we examined the intestinal mucosal expression profile of SCT and SCTR and evaluated the density of S cells along the intestinal tract of individuals with T2D and healthy controls. METHODS Using immunohistochemistry and messenger RNA (mRNA) sequencing, we analyzed intestinal mucosa biopsies sampled along the small intestine at 30-cm intervals and from 7 well-defined anatomical sites along the large intestine (during 2 sessions of double-balloon enteroscopy) in 12 individuals with T2D and 12 healthy controls. RESULTS Both groups exhibited a progressive and similar decrease in SCT and SCTR mRNA expression and S-cell density along the small intestine, with reductions of 14, 100, and 50 times, respectively, in the ileum compared to the duodenum (used as reference). Negligible amounts of SCTR and SCT mRNA, as well as low S-cell density, were found in the large intestine. No significant differences were observed between the groups. CONCLUSION SCT and SCTR mRNA expression and S-cell density were abundant in the duodenum and decreased along the small intestine. Very low SCT and SCTR mRNA levels and S-cell numbers were observed in the large intestine, without aberrations in individuals with T2D compared to healthy controls.
Collapse
Affiliation(s)
- Hannah Gilliam-Vigh
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Tina Jorsal
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Sophie W Nielsen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Julie L Forman
- Section of Biostatistics, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, 1353 Copenhagen K, Denmark
| | - Jens Pedersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Steen S Poulsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| |
Collapse
|
10
|
Berg P, Jensen T, Andersen JF, Svendsen SL, Modvig IM, Wang T, Frische S, Chow BKC, Malte H, Holst JJ, Sørensen MV, Leipziger J. Loss of the Secretin Receptor Impairs Renal Bicarbonate Excretion and Aggravates Metabolic Alkalosis in Mice during Acute Base-Loading. J Am Soc Nephrol 2023; 34:1329-1342. [PMID: 37344929 PMCID: PMC10400107 DOI: 10.1681/asn.0000000000000173] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/12/2023] [Indexed: 06/23/2023] Open
Abstract
SIGNIFICANCE STATEMENT During acute base excess, the renal collecting duct β -intercalated cells ( β -ICs) become activated to increase urine base excretion. This process is dependent on pendrin and cystic fibrosis transmembrane regulator (CFTR) expressed in the apical membrane of β -ICs. The signal that leads to activation of this process was unknown. Plasma secretin levels increase during acute alkalosis, and the secretin receptor (SCTR) is functionally expressed in β -ICs. We find that mice with global knockout for the SCTR lose their ability to acutely increase renal base excretion. This forces the mice to lower their ventilation to cope with this challenge. Our findings suggest that secretin is a systemic bicarbonate-regulating hormone, likely being released from the small intestine during alkalosis. BACKGROUND The secretin receptor (SCTR) is functionally expressed in the basolateral membrane of the β -intercalated cells of the kidney cortical collecting duct and stimulates urine alkalization by activating the β -intercalated cells. Interestingly, the plasma secretin level increases during acute metabolic alkalosis, but its role in systemic acid-base homeostasis was unclear. We hypothesized that the SCTR system is essential for renal base excretion during acute metabolic alkalosis. METHODS We conducted bladder catheterization experiments, metabolic cage studies, blood gas analysis, barometric respirometry, perfusion of isolated cortical collecting ducts, immunoblotting, and immunohistochemistry in SCTR wild-type and knockout (KO) mice. We also perfused isolated rat small intestines to study secretin release. RESULTS In wild-type mice, secretin acutely increased urine pH and pendrin function in isolated perfused cortical collecting ducts. These effects were absent in KO mice, which also did not sufficiently increase renal base excretion during acute base loading. In line with these findings, KO mice developed prolonged metabolic alkalosis when exposed to acute oral or intraperitoneal base loading. Furthermore, KO mice exhibited transient but marked hypoventilation after acute base loading. In rats, increased blood alkalinity of the perfused upper small intestine increased venous secretin release. CONCLUSIONS Our results suggest that loss of SCTR impairs the appropriate increase of renal base excretion during acute base loading and that SCTR is necessary for acute correction of metabolic alkalosis. In addition, our findings suggest that blood alkalinity increases secretin release from the small intestine and that secretin action is critical for bicarbonate homeostasis.
Collapse
Affiliation(s)
- Peder Berg
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Tobias Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | - Ida Maria Modvig
- Department of Biomedical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Tobias Wang
- Department of Biology, Zoophysiology, Aarhus University, Aarhus, Denmark
| | | | - Billy K. C. Chow
- School of Biological Sciences, The University of Hong Kong, Hong Kong
| | - Hans Malte
- Department of Biology, Zoophysiology, Aarhus University, Aarhus, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Copenhagen University, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen University, Copenhagen, Denmark
| | | | - Jens Leipziger
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
11
|
Atanga R, Singh V, In JG. Intestinal Enteroendocrine Cells: Present and Future Druggable Targets. Int J Mol Sci 2023; 24:ijms24108836. [PMID: 37240181 DOI: 10.3390/ijms24108836] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/03/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Enteroendocrine cells are specialized secretory lineage cells in the small and large intestines that secrete hormones and peptides in response to luminal contents. The various hormones and peptides can act upon neighboring cells and as part of the endocrine system, circulate systemically via immune cells and the enteric nervous system. Locally, enteroendocrine cells have a major role in gastrointestinal motility, nutrient sensing, and glucose metabolism. Targeting the intestinal enteroendocrine cells or mimicking hormone secretion has been an important field of study in obesity and other metabolic diseases. Studies on the importance of these cells in inflammatory and auto-immune diseases have only recently been reported. The rapid global increase in metabolic and inflammatory diseases suggests that increased understanding and novel therapies are needed. This review will focus on the association between enteroendocrine changes and metabolic and inflammatory disease progression and conclude with the future of enteroendocrine cells as potential druggable targets.
Collapse
Affiliation(s)
- Roger Atanga
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Varsha Singh
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Julie G In
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
12
|
Sridhar A, Khan D, Abdelaal M, Elliott JA, Naughton V, Flatt PR, Le Roux CW, Docherty NG, Moffett CR. Differential effects of RYGB surgery and best medical treatment for obesity-diabetes on intestinal and islet adaptations in obese-diabetic ZDSD rats. PLoS One 2022; 17:e0274788. [PMID: 36137097 PMCID: PMC9499270 DOI: 10.1371/journal.pone.0274788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022] Open
Abstract
Modification of gut-islet secretions after Roux-En-Y gastric bypass (RYBG) surgery contributes to its metabolic and anti-diabetic benefits. However, there is limited knowledge on tissue-specific hormone distribution post-RYGB surgery and how this compares with best medical treatment (BMT). In the present study, pancreatic and ileal tissues were excised from male Zucker-Diabetic Sprague Dawley (ZDSD) rats 8-weeks after RYGB, BMT (daily oral dosing with metformin 300mg/kg, fenofibrate 100mg/kg, ramipril 1mg/kg, rosuvastatin 10mg/kg and subcutaneous liraglutide 0.2mg/kg) or sham operation (laparotomy). Insulin, glucagon, somatostatin, PYY, GLP-1 and GIP expression patterns were assessed using immunocytochemistry and analyzed using ImageJ. After RYGB and BMT, body weight and plasma glucose were decreased. Intestinal morphometry was unaltered by RYGB, but crypt depth was decreased by BMT. Intestinal PYY cells were increased by both interventions. GLP-1- and GIP-cell counts were unchanged by RYGB but BMT increased ileal GLP-1-cells and decreased those expressing GIP. The intestinal contents of PYY and GLP-1 were significantly enhanced by RYGB, whereas BMT decreased ileal GLP-1. No changes of islet and beta-cell area or proliferation were observed, but the extent of beta-cell apoptosis and islet integrity calculated using circularity index were improved by both treatments. Significantly decreased islet alpha-cell areas were observed in both groups, while beta- and PYY-cell areas were unchanged. RYGB also induced a decrease in islet delta-cell area. PYY and GLP-1 colocalization with glucagon in islets was significantly decreased in both groups, while co-staining of PYY with glucagon was decreased and that with somatostatin increased. These data characterize significant cellular islet and intestinal adaptations following RYGB and BMT associated with amelioration of obesity-diabetes in ZDSD rats. The differential responses observed and particularly those within islets, may provide important clues to the unique ability of RYGB to cause diabetes remission.
Collapse
Affiliation(s)
- Ananyaa Sridhar
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Dawood Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
- * E-mail:
| | - Mahmoud Abdelaal
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Jessie A. Elliott
- Department of Surgery, Trinity Centre for Health Sciences and St. James’s Hospital, Dublin, Ireland
| | - Violetta Naughton
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Peter R. Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Carel W. Le Roux
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Neil G. Docherty
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Charlotte R. Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| |
Collapse
|
13
|
Bays HE, Fitch A, Christensen S, Burridge K, Tondt J. Anti-Obesity Medications and Investigational Agents: An Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) 2022. OBESITY PILLARS 2022; 2:100018. [PMID: 37990711 PMCID: PMC10662004 DOI: 10.1016/j.obpill.2022.100018] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 11/23/2023]
Abstract
Background This "Anti-Obesity Medications and Investigational Agents: An Obesity Medicine Association Clinical Practice Statement 2022" is intended to provide clinicians an overview of Food and Drug Administration (FDA) approved anti-obesity medications and investigational anti-obesity agents in development. Methods The scientific information for this Clinical Practice Statement (CPS) is based upon published scientific citations, clinical perspectives of OMA authors, and peer review by the Obesity Medicine Association leadership. Results This CPS describes pharmacokinetic principles applicable to those with obesity, and discusses the efficacy and safety of anti-obesity medications [e.g., phentermine, semaglutide, liraglutide, phentermine/topiramate, naltrexone/bupropion, and orlistat, as well as non-systemic superabsorbent oral hydrogel particles (which is technically classified as a medical device)]. Other medications discussed include setmelanotide, metreleptin, and lisdexamfetamine dimesylate. Data regarding the use of combination anti-obesity pharmacotherapy, as well as use of anti-obesity pharmacotherapy after bariatric surgery are limited; however, published data support such approaches. Finally, this CPS discusses investigational anti-obesity medications, with an emphasis on the mechanisms of action and summary of available clinical trial data regarding tirzepatide. Conclusion This "Anti-Obesity Medications and Investigational Agents: An Obesity Medicine Association Clinical Practice Statement 2022" is one of a series of OMA CPSs designed to assist clinicians in the care of patients with pre-obesity/obesity.
Collapse
Affiliation(s)
- Harold E. Bays
- Louisville Metabolic and Atherosclerosis Research Center, University of Louisville School of Medicine, 3288 Illinois Avenue, Louisville, KY, 40213, USA
| | - Angela Fitch
- Assistant Professor of Medicine Harvard Medical School, Co-Director Massachusetts General Hospital Weight Center, Boston, MA, USA
| | - Sandra Christensen
- Integrative Medical Weight Management, 2611 NE 125th St, Suite 100B, Seattle, WA, 98125, USA
| | - Karli Burridge
- Enara Health, 16501 106th Court, Orland Park, IL, 60467, USA
- Gaining Health, 528 Pennsylvania Ave #708, Glen Ellyn, IL, 60137, USA
| | - Justin Tondt
- Department of Family and Community Medicine, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA, 23501, USA
| |
Collapse
|
14
|
QiangGuYin Modulates the OPG/RANKL/RANK Pathway by Increasing Secretin Levels during Treatment of Primary Type I Osteoporosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:7114139. [PMID: 34754319 PMCID: PMC8572595 DOI: 10.1155/2021/7114139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 10/06/2021] [Accepted: 10/15/2021] [Indexed: 11/17/2022]
Abstract
QiangGuYin (QGY) is a common Traditional Chinese medicine prescription for the treatment of osteoporosis. Previous clinical studies have found that QGY effectively improves bone mineral density (BMD) in postmenopausal women, but its underlying mechanism remains unclear. The osteoprotegerin (OPG)/receptor activator of nuclear factor kappa B ligand (RANKL)/receptor activator of nuclear factor kappa B (RANK) pathway is a classic pathway involved in osteoporosis. Secretin levels are a serum marker of osteoporosis, but their effect on the OPG/RANKL/RANK pathway has not been reported. Hence, we investigated the relationship between the OPG/RANKL/RANK pathway and secretin and further revealed the mechanism underlying the effect of QGY in the treatment of osteoporosis. Mice were divided into secretin knockdown, secretin overexpression, and corresponding control groups. Micro-computed tomography was used to detect BMD in different groups, and the results show that QGY significantly improved BMD in mice of the secretin knockdown group. To further verify this, the serum levels of OPG, RANKL, RANK, and secretin were measured by enzyme-linked immunosorbent assays, and femur levels of OPG, RANKL, RANK, and secretin were evaluated by real-time quantitative PCR and western blotting. The results show that the expression of OPG was inhibited and that of RANKL and RANK was increased in mice from the secretin knockdown group, whereas the expression of OPG was upregulated and that of RANKL and RANK was downregulated after QGY intervention. Therefore, QGY inhibited bone resorption by promoting the expression of secretin and modulating the OPG/RANKL/RANK pathway. In addition to the effect of QGY, we also revealed the general regulatory effect of secretin on the OPG/RANKL/RANK pathway. We conclude that QGY modulates the OPG/RANKL/RANK pathway by increasing secretin levels during treatment of primary type I osteoporosis. This work provides a theoretical basis for the clinical use of QGY in the treatment of osteoporosis.
Collapse
|
15
|
Schnabl K, Li Y, U-Din M, Klingenspor M. Secretin as a Satiation Whisperer With the Potential to Turn into an Obesity-curbing Knight. Endocrinology 2021; 162:6294014. [PMID: 34089599 DOI: 10.1210/endocr/bqab113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Indexed: 01/01/2023]
Abstract
The obesity pandemic requires effective preventative and therapeutic intervention strategies. Successful and sustained obesity treatment is currently limited to bariatric surgery. Modulating the release of gut hormones is considered promising to mimic bariatric surgery with its beneficial effects on food intake, body weight, and blood glucose levels. The gut peptide secretin was the first molecule to be termed a hormone; nevertheless, only recently has it been established as a legitimate anorexigenic peptide. In contrast to gut hormones that crosstalk with the brain either directly or by afferent neuronal projections, secretin mediates meal-associated brown fat thermogenesis to induce meal termination, thereby qualifying this physiological mechanism as an attractive, peripheral target for the treatment of obesity. In this perspective, it is of pivotal interest to deepen our as yet superficial knowledge on the physiological roles of secretin as well as meal-associated thermogenesis in energy balance and body weight regulation. Of note, the emerging differences between meal-associated thermogenesis and cold-induced thermogenesis must be taken into account. In fact, there is no correlation between these 2 entities. In addition, the investigation of potential effects of secretin in hedonic-driven food intake, bariatric surgery and chronic treatment using suitable application strategies to overcome pharmacokinetic limitations will provide further insight into its potential to influence energy balance. The aim of this article is to review the facts on secretin's metabolic effects, address prevailing gaps in our knowledge, and provide an overview on the opportunities and challenges of the therapeutic potential of secretin in body weight control.
Collapse
Affiliation(s)
- Katharina Schnabl
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Yongguo Li
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Mueez U-Din
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
| |
Collapse
|
16
|
Zhang Q, Hong Z, Zhu J, Zeng C, Tang Z, Wang W, Huang H. Biliopancreatic Limb Length of Small Intestinal Bypass in Non-obese Goto-Kakizaki (GK) Rats Correlates with Gastrointestinal Hormones, Adipokines, and Improvement in Type 2 Diabetes. Obes Surg 2021; 31:4419-4426. [PMID: 34312782 DOI: 10.1007/s11695-021-05604-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/27/2021] [Accepted: 07/09/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND The purpose of this study was to explore the effects on type 2 diabetes, gastrointestinal hormones, and adipokines after the small intestinal bypass of different biliopancreatic limb (BPL) lengths in non-obese type 2 diabetic rats. METHOD Small intestinal bypass with the BPL length at 10cm, 20cm, 30cm, and 40cm, respectively, and sham surgery were performed in non-obese GK rats. Fasting serum was collected at 2 days preoperatively and 1, 3, 6, and 9 weeks postoperatively. Body weight and fasting blood glucose (FBG) were measured during the experiment. Glycated hemoglobin (GHb), fasting insulin (FINS), C-peptide, ghrelin, leptin, adiponectin, and somatostatin were measured postoperatively. RESULT Rats with a bypassed length of 40cm died within 5-9 weeks. No statistically significant was observed in body weight between the sham group and the bypass groups at the 9th week postoperatively. FBG, GHb, FINS, C-peptide, and HOMA-IR in the bypass groups were lower than those in the sham group postoperatively and were negatively correlated with BPL length. Ghrelin and leptin declined compared with preoperative but were not associated with BPL length. Adiponectin of the bypass groups increased after operation and was positively correlated with BPL length. Somatostatin remained stable among groups during the experiment. CONCLUSION Ghrelin and leptin of non-obese GK rats decreased postoperatively without a linear relationship with the BPL length, while adiponectin increased with positively correlation with the BPL length. In addition, somatostatin remained steady after small intestinal bypass. Further studies are expected to confirm the effect of the BPL length of small intestinal bypass on gastrointestinal hormones and adipokines.
Collapse
Affiliation(s)
- Qiwei Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Zhi Hong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Jieyao Zhu
- Anhui Lujiang County People's Hospital, Chaohu, China
| | - Chao Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Zhen Tang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Weiqiang Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - He Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China. .,, Wuhu City, China.
| |
Collapse
|
17
|
Weight loss from caloric restriction vs Roux-en-Y gastric bypass surgery differentially regulates systemic and portal vein GDF15 levels in obese Zucker fatty rats. Physiol Behav 2021; 240:113534. [PMID: 34303715 DOI: 10.1016/j.physbeh.2021.113534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/19/2021] [Accepted: 07/19/2021] [Indexed: 01/05/2023]
Abstract
Weight loss from caloric restriction (i.e. dieting) tends to be modest and short-lived, whereas from bariatric surgeries such as Roux-en-Y gastric bypass (RYGB) is pronounced and generally sustained. The reasons behind these opposing outcomes between interventions remain unclear, but likely involve differential effects on gut-brain communication. Growth differentiation factor 15 (GDF15) is a ubiquitously-induced, centrally-acting, anorexigenic cytokine whose systemic levels are elevated under a variety of conditions associated with a negative energy balance, including in patients following RYGB. We therefore asked whether systemic and portal vein GDF15 levels differ between obese Zucker fatty rats that experienced similar weight loss from RYGB or from forced caloric restriction (CR). Compared with ad libitum fed (ALF) controls, body weight, visceral adiposity and food intake of RYGB and CR rats were markedly lower during the postoperative observation period. Both systemic and portal vein GDF15 levels in RYGB rats at postoperative day 28 were higher compared with ALF rats and particularly compared with CR rats. Further, systemic and portal vein GDF15 levels negatively correlated with body weight and food intake specifically in RYGB rats. These findings provide evidence that, unlike dieting, RYGB might achieve sustained weight loss and appetite suppression partly through increased GDF15 release from epithelial cells of the gastrointestinal tract.
Collapse
|
18
|
Albrechtsen NJW, Rehfeld JF. On premises and principles for measurement of gastrointestinal peptide hormones. Peptides 2021; 141:170545. [PMID: 33811948 DOI: 10.1016/j.peptides.2021.170545] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 11/23/2022]
Abstract
Gastrointestinal hormones are peptides, and the gastrointestinal tract is the largest endocrine organ in the body for production of peptide hormones. As a premise for accurate measurement of gastrointestinal hormones, the present review provides first an overview over the complex biology of the hormones: The structures and structural homologies; biogenetic aspects; phenotype variabilities; and cellular expression in- and outside the digestive tract. Second, the different methodological principles for measurement are discussed: Bioassay, radioimmunoassay (RIA), enzyme-linked immunosorbent assay (ELISA), mass-spectrometry (LC-MS/MS) and processing-independent analysis (PIA). Third, the variability of secretion patterns for some of the gut hormones is illustrated. Finally, the diagnostic value of gut hormone measurement is discussed. The review concludes that measurement of gastrointestinal peptide hormones is relevant not only for examination of digestive functions and diseases, but also for extra-intestinal functions. Moreover, it concludes that, so far, immunoassay technologies (RIA and ELISA) in modernized forms are still the most feasible for accurate measurements of gastrointestinal hormones in biological fluids. Mass-spectrometry technologies are promising, but still too insensitive and expensive.
Collapse
Affiliation(s)
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Denmark.
| |
Collapse
|
19
|
Yang M, Reimann F, Gribble FM. Chemosensing in enteroendocrine cells: mechanisms and therapeutic opportunities. Curr Opin Endocrinol Diabetes Obes 2021; 28:222-231. [PMID: 33449572 DOI: 10.1097/med.0000000000000614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Enteroendocrine cells (EECs) are scattered chemosensory cells in the intestinal epithelium that release hormones with a wide range of actions on intestinal function, food intake and glucose homeostasis. The mechanisms by which gut hormones are secreted postprandially, or altered by antidiabetic agents and surgical interventions are of considerable interest for future therapeutic development. RECENT FINDINGS EECs are electrically excitable and express a repertoire of G-protein coupled receptors that sense nutrient and nonnutrient stimuli, coupled to intracellular Ca2+ and cyclic adenosine monophosphate. Our knowledge of EEC function, previously developed using mouse models, has recently been extended to human cells. Gut hormone release in humans is enhanced by bariatric surgery, as well as by some antidiabetic agents including sodium-coupled glucose transporter inhibitors and metformin. SUMMARY EECs are important potential therapeutic targets. A better understanding of their chemosensory mechanisms will enhance the development of new therapeutic strategies to treat metabolic and gastrointestinal diseases.
Collapse
Affiliation(s)
- Ming Yang
- University of Cambridge, Institute of Metabolic Science and MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Cambridge, UK
| | | | | |
Collapse
|
20
|
Abstract
Bariatric and metabolic surgery has evolved from simple experimental procedures for a chronic problem associated with significant morbidity into a sophisticated multidisciplinary treatment modality rooted in biology and physiology. Although the complete mechanistic narrative of bariatric surgery cannot yet be written, significant advance in knowledge has been made in the past 2 decades. This article provides a brief overview of the most studied hypotheses and their supporting evidence. Ongoing research, especially in frontier areas, such as the microbiome, will continue to refine, and perhaps even revise, current mechanistic understanding.
Collapse
|
21
|
Discovery of small molecule positive allosteric modulators of the secretin receptor. Biochem Pharmacol 2021; 185:114451. [PMID: 33545115 DOI: 10.1016/j.bcp.2021.114451] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
The secretin receptor (SCTR) is a prototypic Class B1 G protein-coupled receptor (GPCR) that represents a key target for the development of therapeutics for the treatment of cardiovascular, gastrointestinal, and metabolic disorders. However, no non-peptidic molecules targeting this receptor have yet been disclosed. Using a high-throughput screening campaign directed at SCTR to identify small molecule modulators, we have identified three structurally related scaffolds positively modulating SCTRs. Here we outline a comprehensive study comprising a structure-activity series based on commercially available analogs of the three hit scaffold sets A (2-sulfonyl pyrimidines), B (2-mercapto pyrimidines) and C (2-amino pyrimidines), which revealed determinants of activity, cooperativity and specificity. Structural optimization of original hits resulted in analog B2, which substantially enhances signaling of truncated secretin peptides and prolongs residence time of labeled secretin up to 13-fold in a dose-dependent manner. Furthermore, we found that investigated compounds display structural similarity to positive allosteric modulators (PAMs) active at the glucagon-like peptide-1 receptor (GLP-1R), and we were able to confirm cross-recognition of that receptor by a subset of analogs. Studies using SCTR and GLP-1R mutants revealed that scaffold A, but not B and C, likely acts via two distinct mechanisms, one of which constitutes covalent modification of Cys-347GLP-1R known from GLP-1R-selective modulators. The scaffolds identified in this study might not only serve as novel pharmacologic tools to decipher SCTR- or GLP-1R-specific signaling pathways, but also as structural leads to elucidate allosteric binding sites facilitating the future development of orally available therapeutic approaches targeting these receptors.
Collapse
|
22
|
Laurila S, Rebelos E, Honka MJ, Nuutila P. Pleiotropic Effects of Secretin: A Potential Drug Candidate in the Treatment of Obesity? Front Endocrinol (Lausanne) 2021; 12:737686. [PMID: 34671320 PMCID: PMC8522834 DOI: 10.3389/fendo.2021.737686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/15/2021] [Indexed: 12/18/2022] Open
Abstract
Secretin is the first hormone that has been discovered, inaugurating the era and the field of endocrinology. Despite the initial focus, the interest in its actions faded away over the decades. However, there is mounting evidence regarding the pleiotropic beneficial effects of secretin on whole-body homeostasis. In this review, we discuss the evidence from preclinical and clinical studies based on which secretin may have a role in the treatment of obesity.
Collapse
Affiliation(s)
- Sanna Laurila
- Turku PET Centre, University of Turku, Turku, Finland
- Heart Center, Turku University Hospital, Turku, Finland
- Department of Cardiology, Satakunta Central Hospital, Pori, Finland
| | - Eleni Rebelos
- Turku PET Centre, University of Turku, Turku, Finland
| | | | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
- *Correspondence: Pirjo Nuutila,
| |
Collapse
|
23
|
Dengler DG, Sun Q, Holleran J, Pollari S, Beutel J, Brown BT, Shinoki Iwaya A, Ardecky R, Harikumar KG, Miller LJ, Sergienko EA. Development of a Testing Funnel for Identification of Small-Molecule Modulators Targeting Secretin Receptors. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2021; 26:1-16. [PMID: 32749201 PMCID: PMC8278649 DOI: 10.1177/2472555220945284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The secretin receptor (SCTR), a prototypical class B G protein-coupled receptor (GPCR), exerts its effects mainly by activating Gαs proteins upon binding of its endogenous peptide ligand secretin. SCTRs can be found in a variety of tissues and organs across species, including the pancreas, stomach, liver, heart, lung, colon, kidney, and brain. Beyond that, modulation of SCTR-mediated signaling has therapeutic potential for the treatment of multiple diseases, such as heart failure, obesity, and diabetes. However, no ligands other than secretin and its peptide analogs have been described to regulate SCTRs, probably due to inherent challenges in family B GPCR drug discovery. Here we report creation of a testing funnel that allowed targeted detection of SCTR small-molecule activators. Pursuing the strategy to identify positive allosteric modulators (PAMs), we established a unique primary screening assay employing a mixture of three orthosteric stimulators that was compared in a screening campaign testing 12,000 small-molecule compounds. Beyond that, we developed a comprehensive set of secondary assays, such as a radiolabel-free target engagement assay and a NanoBiT (NanoLuc Binary Technology)-based approach to detect β-arrestin-2 recruitment, all feasible in a high-throughput environment as well as capable of profiling ligands and hits regarding their effect on binding and receptor function. This combination of methods enabled the discovery of five promising scaffolds, four of which have been validated and further characterized with respect to their allosteric activities. We propose that our results may serve as starting points for developing the first in vivo active small molecules targeting SCTRs.
Collapse
Affiliation(s)
- Daniela G. Dengler
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Qing Sun
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - John Holleran
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Sirkku Pollari
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Jannis Beutel
- Department of Chemistry and Pharmacy, Chemikum, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Brock T. Brown
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Aki Shinoki Iwaya
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Robert Ardecky
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Kaleeckal G. Harikumar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona, USA
| | - Laurence J. Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona, USA
| | - Eduard A. Sergienko
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| |
Collapse
|
24
|
Hankir MK, Seyfried F. Do Bariatric Surgeries Enhance Brown/Beige Adipose Tissue Thermogenesis? Front Endocrinol (Lausanne) 2020; 11:275. [PMID: 32425889 PMCID: PMC7203442 DOI: 10.3389/fendo.2020.00275] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022] Open
Abstract
Bariatric surgeries induce marked and durable weight loss in individuals with morbid obesity through powerful effects on both food intake and energy expenditure. While alterations in gut-brain communication are increasingly implicated in the improved eating behavior following bariatric surgeries, less is known about the mechanistic basis for energy expenditure changes. Brown adipose tissue (BAT) and beige adipose tissue (BeAT) have emerged as major regulators of whole-body energy metabolism in humans as well as in rodents due to their ability to convert the chemical energy in circulating glucose and fatty acids into heat. In this Review, we critically discuss the steadily growing evidence from preclinical and clinical studies suggesting that Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG), the two most commonly performed bariatric surgeries, enhance BAT/BeAT thermogenesis. We address the documented mechanisms, highlight study limitations and finish by outlining unanswered questions in the subject. Further understanding how and to what extent bariatric surgeries enhance BAT/BeAT thermogenesis may not only aid in the development of improved obesity pharmacotherapies that safely and optimally target both sides of the energy balance equation, but also in the development of novel hyperglycemia and/or hyperlipidemia pharmacotherapies.
Collapse
Affiliation(s)
- Mohammed K. Hankir
- Department of Experimental Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Florian Seyfried
- Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|