1
|
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: 1] [Impact Index Per Article: 1.0] [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
|
2
|
Guanylin ligand protects the intestinal immune barrier by activating the guanylate cyclase-C signaling pathway. Acta Histochem 2022; 124:151811. [PMID: 34920371 DOI: 10.1016/j.acthis.2021.151811] [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: 05/17/2021] [Revised: 10/14/2021] [Accepted: 10/22/2021] [Indexed: 11/20/2022]
Abstract
Inflammatory bowel disease (IBD) impacts patient quality of life significantly. The dysfunction of intestinal immune barrier is closely associated with IBD. The guanylate cyclase-C (GC-C) signaling pathway activated by the guanylin (Gn) ligand is involved in the occurrence and development of IBD. However, how it regulates the intestinal immune barrier is still unclear. To investigate the effect of the GC-C pathway on intestinal mucosal immunity and provide experimental basis for seeking new therapeutic strategies for IBD, we focused on Caco-2 cells and intestinal intra-epithelial lymphocytes (IELs), which displayed inflammatory responses induced by lipopolysaccharide (LPS). GC-C activity was modulated by transfection with Gn overexpression or GC-C shRNA plasmid. Levels of Gn, GC-C, and CFTR; transepithelial electrical resistance (TER); paracellula r permeability; and levels of IL-2, IFN-γ, and secretory IgA (sIgA) were examined. The study found that after stimulation with LPS, Gn, GC-C, CFTR, TER, and sIgA levels were all significantly reduced, IL-2 and IFN-γ levels as well as paracellular permeability were significantly increased. These indicators changed inversely and significantly after transfection with the Gn overexpression vector. Compared to the vector controls, GC-C-silenced cells displayed significantly decreased levels of GC-C, CFTR, and TER and increased levels of IL-2, IFN-γ, and paracellular permeability stimulated by LPS. The results show that Gn ligand can protect the intestinal immune barrier by activating the GC-C signaling pathway, which may be helpful for the development of new treatments for IBD. DATA AVAILABILITY STATEMENT: The data used to support the findings of this study are available from the corresponding author upon request.
Collapse
|
3
|
Dye FS, Larraufie P, Kay R, Darwish T, Rievaj J, Goldspink DA, Meek CL, Middleton SJ, Hardwick RH, Roberts GP, Percival-Alwyn JL, Vaughan T, Ferraro F, Challis BG, O'Rahilly S, Groves M, Gribble FM, Reimann F. Characterisation of proguanylin expressing cells in the intestine - evidence for constitutive luminal secretion. Sci Rep 2019; 9:15574. [PMID: 31666564 PMCID: PMC6821700 DOI: 10.1038/s41598-019-52049-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022] Open
Abstract
Guanylin, a peptide implicated in regulation of intestinal fluid secretion, is expressed in the mucosa, but the exact cellular origin remains controversial. In a new transgenic mouse model fluorescent reporter protein expression driven by the proguanylin promoter was observed throughout the small intestine and colon in goblet and Paneth(-like) cells and, except in duodenum, in mature enterocytes. In Ussing chamber experiments employing both human and mouse intestinal tissue, proguanylin was released predominantly in the luminal direction. Measurements of proguanylin expression and secretion in cell lines and organoids indicated that secretion is largely constitutive and requires ER to Golgi transport but was not acutely regulated by salt or other stimuli. Using a newly-developed proguanylin assay, we found plasma levels to be raised in humans after total gastrectomy or intestinal transplantation, but largely unresponsive to nutrient ingestion. By LC-MS/MS we identified processed forms in tissue and luminal extracts, but in plasma we only detected full-length proguanylin. Our transgenic approach provides information about the cellular origins of proguanylin, complementing previous immunohistochemical and in-situ hybridisation results. The identification of processed forms of proguanylin in the intestinal lumen but not in plasma supports the notion that the primary site of action is the gut itself.
Collapse
Affiliation(s)
- Florent Serge Dye
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Pierre Larraufie
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Richard Kay
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Tamana Darwish
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Juraj Rievaj
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Dosage Form Design & Development, AstraZeneca, Cambridge, UK
| | - Deborah A Goldspink
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Claire L Meek
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Stephen J Middleton
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Richard H Hardwick
- Barrett's Oesophagus and Oesophago-gastric Cancer, Gastroenterology Services, Addenbrooke's Hospital, Cambridge, UK
| | - Geoffrey P Roberts
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | | | - Tris Vaughan
- Department of Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Franco Ferraro
- Department of Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Benjamin G Challis
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Stephen O'Rahilly
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Maria Groves
- Department of Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge, UK.
| | - Fiona M Gribble
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Frank Reimann
- Wellcome/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| |
Collapse
|
4
|
Li P, Wuthrick E, Rappaport JA, Kraft C, Lin JE, Marszalowicz G, Snook AE, Zhan T, Hyslop TM, Waldman SA. GUCY2C Signaling Opposes the Acute Radiation-Induced GI Syndrome. Cancer Res 2017; 77:5095-5106. [PMID: 28916678 PMCID: PMC5678756 DOI: 10.1158/0008-5472.can-17-0859] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/30/2017] [Accepted: 07/18/2017] [Indexed: 02/05/2023]
Abstract
High doses of ionizing radiation induce acute damage to epithelial cells of the gastrointestinal (GI) tract, mediating toxicities restricting the therapeutic efficacy of radiation in cancer and morbidity and mortality in nuclear disasters. No approved prophylaxis or therapy exists for these toxicities, in part reflecting an incomplete understanding of mechanisms contributing to the acute radiation-induced GI syndrome (RIGS). Guanylate cyclase C (GUCY2C) and its hormones guanylin and uroguanylin have recently emerged as one paracrine axis defending intestinal mucosal integrity against mutational, chemical, and inflammatory injury. Here, we reveal a role for the GUCY2C paracrine axis in compensatory mechanisms opposing RIGS. Eliminating GUCY2C signaling exacerbated RIGS, amplifying radiation-induced mortality, weight loss, mucosal bleeding, debilitation, and intestinal dysfunction. Durable expression of GUCY2C, guanylin, and uroguanylin mRNA and protein by intestinal epithelial cells was preserved following lethal irradiation inducing RIGS. Oral delivery of the heat-stable enterotoxin (ST), an exogenous GUCY2C ligand, opposed RIGS, a process requiring p53 activation mediated by dissociation from MDM2. In turn, p53 activation prevented cell death by selectively limiting mitotic catastrophe, but not apoptosis. These studies reveal a role for the GUCY2C paracrine hormone axis as a novel compensatory mechanism opposing RIGS, and they highlight the potential of oral GUCY2C agonists (Linzess; Trulance) to prevent and treat RIGS in cancer therapy and nuclear disasters. Cancer Res; 77(18); 5095-106. ©2017 AACR.
Collapse
MESH Headings
- Animals
- Apoptosis/radiation effects
- Cell Proliferation/radiation effects
- Colonic Neoplasms/enzymology
- Colonic Neoplasms/pathology
- Colonic Neoplasms/radiotherapy
- Female
- Gamma Rays/adverse effects
- Gastrointestinal Hormones/metabolism
- Gastrointestinal Tract/radiation effects
- Humans
- Irritable Bowel Syndrome/enzymology
- Irritable Bowel Syndrome/etiology
- Irritable Bowel Syndrome/prevention & control
- Lymphoma/enzymology
- Lymphoma/pathology
- Lymphoma/radiotherapy
- Male
- Melanoma, Experimental/enzymology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/radiotherapy
- Mice
- Mice, Inbred C57BL
- Natriuretic Peptides/metabolism
- Paracrine Communication/radiation effects
- Radiation Injuries, Experimental/enzymology
- Radiation Injuries, Experimental/etiology
- Radiation Injuries, Experimental/prevention & control
- Receptors, Enterotoxin
- Receptors, Guanylate Cyclase-Coupled/metabolism
- Receptors, Peptide/metabolism
- Signal Transduction/radiation effects
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Peng Li
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, The University of Florida, Gainesville, Florida
| | - Evan Wuthrick
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Jeff A Rappaport
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Crystal Kraft
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jieru E Lin
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Glen Marszalowicz
- 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
| | - Tingting Zhan
- Divisions of Clinical Pharmacology and Biostatistics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Terry M Hyslop
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
5
|
Pattison AM, Merlino DJ, Blomain ES, Waldman SA. Guanylyl cyclase C signaling axis and colon cancer prevention. World J Gastroenterol 2016; 22:8070-8077. [PMID: 27688649 PMCID: PMC5037076 DOI: 10.3748/wjg.v22.i36.8070] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/25/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related mortality and morbidity worldwide. While improved treatments have enhanced overall patient outcome, disease burden encompassing quality of life, cost of care, and patient survival has seen little benefit. Consequently, additional advances in CRC treatments remain important, with an emphasis on preventative measures. Guanylyl cyclase C (GUCY2C), a transmembrane receptor expressed on intestinal epithelial cells, plays an important role in orchestrating intestinal homeostatic mechanisms. These effects are mediated by the endogenous hormones guanylin (GUCA2A) and uroguanylin (GUCA2B), which bind and activate GUCY2C to regulate proliferation, metabolism and barrier function in intestine. Recent studies have demonstrated a link between GUCY2C silencing and intestinal dysfunction, including tumorigenesis. Indeed, GUCY2C silencing by the near universal loss of its paracrine hormone ligands increases colon cancer susceptibility in animals and humans. GUCY2C’s role as a tumor suppressor has opened the door to a new paradigm for CRC prevention by hormone replacement therapy using synthetic hormone analogs, such as the FDA-approved oral GUCY2C ligand linaclotide (Linzess™). Here we review the known contributions of the GUCY2C signaling axis to CRC, and relate them to a novel clinical strategy targeting tumor chemoprevention.
Collapse
|
6
|
Intestinal Enteroids Model Guanylate Cyclase C-Dependent Secretion Induced by Heat-Stable Enterotoxins. Infect Immun 2016; 84:3083-91. [PMID: 27481254 DOI: 10.1128/iai.00639-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 12/23/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) causes ∼20% of the acute infectious diarrhea (AID) episodes worldwide, often by producing heat-stable enterotoxins (STs), which are peptides structurally homologous to paracrine hormones of the intestinal guanylate cyclase C (GUCY2C) receptor. While molecular mechanisms mediating ST-induced intestinal secretion have been defined, advancements in therapeutics have been hampered for decades by the paucity of disease models that integrate molecular and functional endpoints amenable to high-throughput screening. Here, we reveal that mouse and human intestinal enteroids in three-dimensional ex vivo cultures express the components of the GUCY2C secretory signaling axis. ST and its structural analog, linaclotide, an FDA-approved oral secretagog, induced fluid accumulation quantified simultaneously in scores of enteroid lumens, recapitulating ETEC-induced intestinal secretion. Enteroid secretion depended on canonical molecular signaling events responsible for ETEC-induced diarrhea, including cyclic GMP (cGMP) produced by GUCY2C, activation of cGMP-dependent protein kinase (PKG), and opening of the cystic fibrosis transmembrane conductance regulator (CFTR). Importantly, pharmacological inhibition of CFTR abrogated enteroid fluid secretion, providing proof of concept for the utility of this model to screen antidiarrheal agents. Intestinal enteroids offer a unique model, integrating the GUCY2C signaling axis and luminal fluid secretion, to explore the pathophysiology of, and develop platforms for, high-throughput drug screening to identify novel compounds to prevent and treat ETEC diarrheal disease.
Collapse
|
7
|
Ikpa PT, Sleddens HFBM, Steinbrecher KA, Peppelenbosch MP, de Jonge HR, Smits R, Bijvelds MJC. Guanylin and uroguanylin are produced by mouse intestinal epithelial cells of columnar and secretory lineage. Histochem Cell Biol 2016; 146:445-55. [PMID: 27246004 PMCID: PMC5037145 DOI: 10.1007/s00418-016-1453-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2016] [Indexed: 01/12/2023]
Abstract
Guanylin (GN) and uroguanylin (UGN), through activation of guanylyl cyclase C (GCC), serve to control intestinal fluid homeostasis. Both peptides are produced in the intestinal epithelium, but their cellular origin has not been fully charted. Using quantitative PCR and an improved in situ hybridization technique (RNAscope), we have assessed the expression of GN (Guca2a), UGN (Guca2b), and GCC (Gucy2c) in mouse intestine. In the crypts of Lieberkühn, expression of Guca2a and Guca2b was restricted to cells of secretory lineage, at the crypt's base, and to a region above, previously identified as a common origin of cellular differentiation. In this compartment, comparatively uniform levels of Guca2a and Guca2b expression were observed throughout the length of the gut. In contrast, Guca2a and Guca2b expression in the villus-surface region was more variable, and reflected the distinct, but overlapping expression pattern observed previously. Accordingly, in jejunum and ileum, Guca2a and Guca2b were abundantly expressed by enterocytes, whereas in colon only Guca2a transcript was found in the surface region. In duodenum, only low levels of Guca2b transcript were observed in columnar cells, and Guca2a expression was restricted entirely to cells of the secretory lineage. Gucy2c was shown to be expressed relatively uniformly along the rostrocaudal and crypt-villus axes and was also found in the duodenal glands. Our study reveals novel aspects of the cellular localization of the GCC signaling axis that, apart from its role in the regulation of fluid balance, link it to pH regulation, cell cycle control, and host defense.
Collapse
Affiliation(s)
- Pauline T Ikpa
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Hein F B M Sleddens
- Department of Pathology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Kris A Steinbrecher
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Hugo R de Jonge
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Ron Smits
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Marcel J C Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
8
|
Brenna Ø, Furnes MW, Munkvold B, Kidd M, Sandvik AK, Gustafsson BI. Cellular localization of guanylin and uroguanylin mRNAs in human and rat duodenal and colonic mucosa. Cell Tissue Res 2016; 365:331-41. [PMID: 27044258 PMCID: PMC4943973 DOI: 10.1007/s00441-016-2393-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 03/03/2016] [Indexed: 12/11/2022]
Abstract
Guanylin (GUCA2A/Guca2a/GN) and uroguanylin (GUCA2B/Guca2b/UGN) are expressed in the gastrointestinal tract and have been implicated in ion and fluid homeostasis, satiety, abdominal pain, growth and intestinal barrier integrity. Their cellular sources are debated and include goblet cells, entero-/colonocytes, enteroendocrine (EE) cells and tuft cells. We therefore investigated the cellular sources of GN and UGN mRNAs in human and rat duodenal and colonic epithelium with in situ hybridization (ISH) to determine co-expression with Chromogranin A (CHGA/Chga/CgA; enterochromaffin [EC] cells), defensin alpha 6 (DEFA6/Defa6; Paneth cells), mucin 2 (MUC2/Muc2; goblet cells) and selected tuft cell markers. GUCA2A/Guca2a expression was localized to goblet cells and colonocytes in human and rat colon. In human duodenum, GUCA2A was expressed in Paneth cells and was scarce in villous epithelial cells. In rat duodenum, Guca2a was only localized to goblet cells. Guca2b was focally expressed in rat colon. In human and rat duodenum and in human colon, GUCA2B/Guca2b was expressed in dispersed solitary epithelial cells, some with a tuft cell-like appearance. Neither GUCA2A nor GUCA2B were co-expressed with CHGA in human duodenal cells. Consequently, EC cells are probably not the major source of human GN or UGN but other EE cells as a source of GN or UGN are not entirely excluded. No convincing overlap with tuft cell markers was found. For the first time, we demonstrate the cellular expression of GUCA2B in human duodenum. The specific cellular distribution of both GN and UGN differs between duodenum and colon and between human and rat intestines.
Collapse
Affiliation(s)
- Øystein Brenna
- Department of Gastroenterology and Hepatology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Marianne W Furnes
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjørn Munkvold
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mark Kidd
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Arne K Sandvik
- Department of Gastroenterology and Hepatology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Björn I Gustafsson
- Department of Gastroenterology and Hepatology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
9
|
Matuchansky C. Intestinal Secretory Mechanisms and Irritable Bowel Syndrome. Clin Gastroenterol Hepatol 2015; 13:2382-3. [PMID: 26164222 DOI: 10.1016/j.cgh.2015.06.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 06/23/2015] [Accepted: 06/30/2015] [Indexed: 02/07/2023]
Affiliation(s)
- Claude Matuchansky
- Lariboisière-St Louis Faculty of Medicine, Paris-Diderot University, Paris, France
| |
Collapse
|
10
|
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C. E. N. T. E. R.), College of Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
11
|
Camilleri M, Carlson P, Acosta A, Busciglio I, Nair AA, Gibbons SJ, Farrugia G, Klee EW. RNA sequencing shows transcriptomic changes in rectosigmoid mucosa in patients with irritable bowel syndrome-diarrhea: a pilot case-control study. Am J Physiol Gastrointest Liver Physiol 2014; 306:G1089-98. [PMID: 24763552 PMCID: PMC4059976 DOI: 10.1152/ajpgi.00068.2014] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Our aim was to conduct a pilot case-control study of RNA expression profile using RNA sequencing of rectosigmoid mucosa of nine females with -diarrhea-predominant irritable bowel syndrome (IBS-D) with accelerated colonic transit and nine female healthy controls. Mucosal total RNA was isolated and purified, and next-generation pair-end sequencing was performed using Illumina TruSeq. Analysis was carried out using a targeted approach toward 12 genes previously associated with IBS and a hypothesis-generating approach. Of the 12 targeted genes tested, patients with IBS-D had decreased mRNA expression of TNFSF15 (fold change controls to IBS-D: 1.53, P = 0.01). Overall, up- and downregulated mRNA expressions of 21 genes (P = 10(-5) to 10(-8); P values with false detection rates are shown) were potentially relevant to IBS-D including the following: neurotransmitters [P2RY4 (P = 0.001), vasoactive intestinal peptide (VIP, P = 0.02)]; cytokines [CCL20 (P = 0.019)]; immune function [C4BPA complement cascade (P = 0.0187)]; interferon-related [IFIT3 (P = 0.016)]; mucosal repair and cell adhesion [trefoil protein (TFF1, P = 0.012)], retinol binding protein [RBP2 (P = 0.017)]; fibronectin (FN1, P = 0.009); and ion channel functions [guanylate cyclase (GUCA2B, P = 0.017), PDZ domain-containing protein 3 (PDZD3, P = 0.029)]. Ten genes associated with functions related to pathobiology of IBS-D were validated by RT-PCR. There was significant correlation in fold changes of the selected genes (Rs = 0.73, P = 0.013). Up- or downregulation of P2RY4, GUC2AB, RBP2, FNI, and C4BPA genes were confirmed on RT-PCR, which also revealed upregulation of farnesoid X receptor (FXR) and apical sodium-coupled bile acid transporter (IBAT/ASBT). RNA-Seq and RT-PCR analysis of rectosigmoid mucosa in IBS-D show transcriptome changes that provide the rationale for validation studies to explore the role of mucosal factors in the pathobiology of IBS-D.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and
| | - Paula Carlson
- 1Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and
| | - Andres Acosta
- 1Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and
| | - Irene Busciglio
- 1Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and
| | - Asha A. Nair
- 2Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Simon J. Gibbons
- 1Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and
| | - Gianrico Farrugia
- 1Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and
| | - Eric W. Klee
- 2Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
12
|
Rozenfeld J, Tal O, Kladnitsky O, Adler L, Efrati E, Carrithers SL, Alper SL, Zelikovic I. Pendrin, a novel transcriptional target of the uroguanylin system. Cell Physiol Biochem 2013; 32:221-37. [PMID: 24429828 DOI: 10.1159/000356641] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2013] [Indexed: 12/22/2022] Open
Abstract
Guanylin (GN) and uroguanylin (UGN) are low-molecular-weight peptide hormones produced mainly in the intestinal mucosa in response to oral salt load. GN and UGN (guanylin peptides) induce secretion of electrolytes and water in both intestine and kidney. Thought to act as "intestinal natriuretic factors", GN and UGN modulate renal salt secretion by both endocrine mechanisms (linking the digestive system and kidney) and paracrine/autocrine (intrarenal) mechanisms. The cellular function of GN and UGN in intestine and proximal tubule is mediated by guanylyl cyclase C (GC-C)-, cGMP-, and G protein-dependent pathways, whereas, in principal cells of the cortical collecting duct (CCD), these peptide hormones act via GC-C-independent signaling through phospholipase A2 (PLA2). The Cl(-)/HCO(-)3 exchanger pendrin (SLC26A4), encoded by the PDS gene, is expressed in non-α intercalated cells of the CCD. Pendrin is essential for CCD bicarbonate secretion and is also involved in NaCl balance and blood pressure regulation. Our recent studies have provided evidence that pendrin-mediated anion exchange in the CCD is regulated at the transcriptional level by UGN. UGN exerts an inhibitory effect on the pendrin gene promoter likely via heat shock factor 1 (HSF1) action at a defined heat shock element (HSE) site. Recent studies have unraveled novel roles for guanylin peptides in several organ systems including involvement in appetite regulation, olfactory function, cell proliferation and differentiation, inflammation, and reproductive function. Both the guanylin system and pendrin have also been implicated in airway function. Future molecular research into the receptors and signal transduction pathways involved in the action of guanylin peptides and the pendrin anion exchanger in the kidney and other organs, and into the links between them, may facilitate discovery of new therapies for hypertension, heart failure, hepatic failure and other fluid retention syndromes, as well as for diverse diseases such as obesity, asthma, and cancer.
Collapse
Affiliation(s)
- Julia Rozenfeld
- Laboratory of Developmental Nephrology, Department of Physiology and Biophysics, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Harmel-Laws E, Mann EA, Cohen MB, Steinbrecher KA. Guanylate cyclase C deficiency causes severe inflammation in a murine model of spontaneous colitis. PLoS One 2013; 8:e79180. [PMID: 24244444 PMCID: PMC3823613 DOI: 10.1371/journal.pone.0079180] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/20/2013] [Indexed: 12/18/2022] Open
Abstract
Background Guanylate Cyclase C (GC-C; Gucy2c) is a transmembrane receptor expressed in intestinal epithelial cells. Activation of GC-C by its secreted ligand guanylin stimulates intestinal fluid secretion. Familial mutations in GC-C cause chronic diarrheal disease or constipation and are associated with intestinal inflammation and infection. Here, we investigated the impact of GC-C activity on mucosal immune responses. Methods We utilized intraperitoneal injection of lipopolysaccharide to elicit a systemic cytokine challenge and then measured pro-inflammatory gene expression in colonic mucosa. GC-C+/+ and GC-C−/− mice were bred with interleukin (IL)-10 deficient animals and colonic inflammation were assessed. Immune cell influx and cytokine/chemokine expression was measured in the colon of wildtype, IL-10−/−, GC-C+/+IL-10−/− and GC-C−/−IL-10−/− mice. GC-C and guanylin production were examined in the colon of these animals and in a cytokine-treated colon epithelial cell line. Results Relative to GC-C+/+ animals, intraperitoneal lipopolysaccharide injection into GC-C−/− mice increased proinflammatory gene expression in both whole colon tissue and in partially purified colonocyte isolations. Spontaneous colitis in GC-C−/−IL-10−/− animals was significantly more severe relative to GC-C+/+IL-10−/− mice. Unlike GC-C+/+IL-10−/− controls, colon pathology in GC-C−/−IL-10−/− animals was apparent at an early age and was characterized by severely altered mucosal architecture, crypt abscesses, and hyperplastic subepithelial lesions. F4/80 and myeloperoxidase positive cells as well as proinflammatory gene expression were elevated in GC-C−/−IL-10−/− mucosa relative to control animals. Guanylin was diminished early in colitis in vivo and tumor necrosis factor α suppressed guanylin mRNA and protein in intestinal goblet cell-like HT29-18-N2 cells. Conclusions The GC-C signaling pathway blunts colonic mucosal inflammation that is initiated by systemic cytokine burst or loss of mucosal immune cell immunosuppression. These data as well as the apparent intestinal inflammation in human GC-C mutant kindred underscore the importance of GC-C in regulating the response to injury and inflammation within the gut.
Collapse
Affiliation(s)
- Eleana Harmel-Laws
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Elizabeth A. Mann
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Mitchell B. Cohen
- Department of Pediatrics, University of Cincinnati College of Medicine Cincinnati, Ohio, United States of America
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Kris A. Steinbrecher
- Department of Pediatrics, University of Cincinnati College of Medicine Cincinnati, Ohio, United States of America
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
14
|
Sindic A. Current understanding of guanylin peptides actions. ISRN NEPHROLOGY 2013; 2013:813648. [PMID: 24967239 PMCID: PMC4045495 DOI: 10.5402/2013/813648] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 02/26/2013] [Indexed: 01/12/2023]
Abstract
Guanylin peptides (GPs) family includes guanylin (GN), uroguanylin (UGN), lymphoguanylin, and recently discovered renoguanylin. This growing family is proposed to be intestinal natriuretic peptides. After ingestion of a salty meal, GN and UGN are secreted into the intestinal lumen, where they inhibit sodium absorption and induce anion and water secretion. At the same conditions, those hormones stimulate renal electrolyte excretion by inducing natriuresis, kaliuresis, and diuresis and therefore prevent hypernatremia and hypervolemia after salty meals.
In the intestine, a well-known receptor for GPs is guanylate cyclase C (GC-C) whose activation increases intracellular concentration of cGMP. However, in the kidney of GC-C-deficient mice, effects of GPs are unaltered, which could be by new cGMP-independent signaling pathway (G-protein-coupled receptor). This is not unusual as atrial natriuretic peptide also activates two different types of receptors: guanylate cylcase A and clearance receptor which is also G-protein coupled receptor. Physiological role of GPs in other organs (liver, pancreas, lung, sweat glands, and male reproductive system) needs to be discovered. However, it is known that they are involved in pathological conditions like cystic fibrosis, asthma, intestinal tumors, kidney and heart failure, obesity, and metabolic syndrome.
Collapse
Affiliation(s)
- Aleksandra Sindic
- Department of Physiology, School of Medicine, University of Zagreb, Salata 3, 10000 Zagreb, Croatia
| |
Collapse
|
15
|
Schwabe K, Cetin Y. Guanylin and functional coupling proteins in the hepatobiliary system of rat and guinea pig. Histochem Cell Biol 2012; 137:589-97. [PMID: 22310983 DOI: 10.1007/s00418-012-0927-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2012] [Indexed: 12/13/2022]
Abstract
Guanylin, a bioactive intestinal peptide, is involved in the cystic fibrosis transmembrane conductance (CFTR)-regulated electrolyte/water secretion in various epithelia. In the present work we report on the expression and cellular localization of guanylin and its affiliated signaling and effector proteins, including guanylate cyclase C (Gucy2c), Proteinkinase GII (Pkrg2), CFTR and the solute carrier family 4, anion exchanger, member 2 (Slc4a2) in the hepatobiliary system of rat and guinea pig. Localization studies in the liver and the gallbladder revealed that guanylin is located in the secretory epithelial cells of bile ducts of the liver and of the gallbladder, while Gucy2c, Pkrg2, CFTR, and Slc4a2 are confined exclusively to the apical membrane of the same epithelial cells. Based on these findings, we assume that guanylin is synthesized as an intrinsic peptide in epithelial cells of the hepatobiliary system and released luminally into the hepatic and cystic bile to regulate electrolyte secretion by a paracrine/luminocrine signaling pathway.
Collapse
Affiliation(s)
- Karen Schwabe
- Department of Molecular Cell Biology, Institute of Anatomy and Cell Biology, Philipps-Universität Marburg, Marburg, Germany
| | | |
Collapse
|
16
|
Enteroendocrine and neuronal mechanisms in pathophysiology of acute infectious diarrhea. Dig Dis Sci 2012; 57:19-27. [PMID: 22001941 PMCID: PMC3809758 DOI: 10.1007/s10620-011-1939-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 09/30/2011] [Indexed: 12/30/2022]
Abstract
BACKGROUND While enterocyte secretion is the predominant mechanism considered responsible for secretory diarrhea in response to acute enteric infections, there are several lines of evidence that support alternative mechanisms controlling fluid and electrolyte secretion in diarrhea. AIM To review enteroendocrine and neuronal mechanisms that participate in the development of acute infectious diarrhea. RECENT ADVANCES Acute infectious diarrheas due to bacterial toxins (e.g., cholera, E. coli heat-stable enterotoxin, C. difficile) and rotavirus are all associated with secretion of transmitters from enteroendocrine cells (e.g., 5-HT) and activation of afferent neurons that stimulate submucosal secretomotor neurons. The latter secrete acetylcholine (which binds to muscarinic receptors on epithelial cells) and VIP. Involvement of nerves was demonstrated by inhibition of bacterial toxin-induced secretion by hexamethonium (nicotinic), tetrodotoxin (Na(+) channel blocker), and lidocaine (visceral/mucosal afferents). Nicotinic receptors are present on secretomotoneurons and these are activated by release of acetylcholine from enteric interneurons or extrinsic efferent fibers. Specific organisms also modify other mechanisms that may contribute to development of acute diarrhea. Thus, mucin secretion, activation of motor mechanisms, increased mucosal permeability and inhibition of bile acid absorption have been reported in specific types of acute infectious diarrhea. CONCLUSION New therapies targeting neural and transmitter mediation including 5-HT, VIP, NPY, as well as toxin receptors and channels activated during acute infectious diarrhea could usher in a novel approach to enhancing glucose-electrolyte solutions used in the treatment of acute diarrhea.
Collapse
|
17
|
Fonteles MC, do Nascimento NRF. Guanylin peptide family: history, interactions with ANP, and new pharmacological perspectives. Can J Physiol Pharmacol 2011; 89:575-85. [PMID: 21815750 DOI: 10.1139/y11-050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The guanylin family of peptides has 3 subclasses of peptides containing either 3 intramolecular disulfide bonds found in bacterial heat-stable enterotoxins (ST), or 2 disulfides observed in guanylin and uroguanylin, or a single disulfide exemplified by lymphoguanylin. These peptides bind to and activate cell-surface receptors that have intrinsic guanylate cyclase (GC) activity. These hormones are synthesized in the intestine and released both luminally and into the circulation, and are also produced within the kidney. Stimulation of renal target cells by guanylin peptides in vivo or ex vivo elicits a long-lived diuresis, natriuresis, and kaliuresis by both cGMP-dependent and independent mechanisms. Uroguanylin may act as a hormone in a novel endocrine axis linking the digestive system and kidney as well as a paracrine system intrarenally to increase sodium excretion in the postprandial period. This highly integrated and redundant mechanism allows the organism to maintain sodium balance by eliminating excess sodium in the urine. In addition, small concentrations of the atrial natriuretic peptide (ANP) can synergize with low concentrations of both guanylin or uroguanylin, which do not induce natriuresis per se, to promote significant natriuresis. Interestingly, the activation of the particulate guanylate cyclase receptors by natriuretic peptides can promote relaxation of animal and human penile erectile tissue and increase intracavernosal pressure to induce penile erection. These peptides can be prototypes for new drugs to treat erectile dysfunction, especially in patients with endothelial and nitrergic dysfunction, such as in diabetes.
Collapse
Affiliation(s)
- Manassés Claudino Fonteles
- Instituto Superior de Ciências Biomédicas (ISCB), Laboratório de Farmacologia - Universidade Estadual do Ceará (UECE), Avenida Paranjana 1700, Campus do Itaperi, CEP 60740-000, Fortaleza-CE, Brazil.
| | | |
Collapse
|
18
|
Steinbrecher KA, Harmel-Laws E, Garin-Laflam MP, Mann EA, Bezerra LD, Hogan SP, Cohen MB. Murine guanylate cyclase C regulates colonic injury and inflammation. THE JOURNAL OF IMMUNOLOGY 2011; 186:7205-14. [PMID: 21555532 DOI: 10.4049/jimmunol.1002469] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Guanylate cyclase C (GUCY2C or GC-C) and its ligands, guanylin (GUCA2A or Gn) and uroguanylin (GUCA2B or Ugn), are expressed in intestinal epithelial cells and regulate ion secretion, intestinal barrier function, and epithelial monolayer homeostasis via cGMP-dependent signaling pathways. The aim of this study was to determine whether GC-C and its ligands direct the course of intestinal inflammation. In this article, we show that dextran sodium sulfate (DSS)-induced clinical disease and histological damage to the colonic mucosa were significantly less severe in GC-C(-/-) mice and moderately reduced in Gn(-/-) animals. Relative to wild-type controls, GC-C(-/-) and Gn(-/-) mice had reduced apoptosis and increased proliferation of intestinal epithelial cells during DSS colitis. Basal and DSS-induced production of resistin-like molecule β (RELMβ) was substantially diminished in GC-C(-/-) mice. RELMβ is thought to stimulate cytokine production in macrophages in this disease model and, consistent with this, TNF-α and IFN-γ production was minimal in GC-C(-/-) animals. RELMβ and cytokine levels were similar to wild-type in Gn(-/-) mice, however. Colonic instillation of recombinant RELMβ by enema into GC-C(-/-) mice restores sensitivity to DSS-mediated mucosal injury. These findings demonstrate a novel role for GC-C signaling in facilitating mucosal wounding and inflammation, and further suggest that this may be mediated, in part, through control of RELMβ production.
Collapse
Affiliation(s)
- Kris A Steinbrecher
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Qian X, Moss NG, Fellner RC, Taylor-Blake B, Goy MF. The rat kidney contains high levels of prouroguanylin (the uroguanylin precursor) but does not express GC-C (the enteric uroguanylin receptor). Am J Physiol Renal Physiol 2011; 300:F561-73. [PMID: 21106860 PMCID: PMC3280727 DOI: 10.1152/ajprenal.00282.2010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 11/18/2010] [Indexed: 01/13/2023] Open
Abstract
The peptide uroguanylin (Ugn) regulates enteric and renal electrolyte transport. Previous studies have shown that Ugn and its receptor GC-C (a ligand-activated guanylate cyclase) are abundant in the intestine. Less is known about Ugn and GC-C expression in the kidney. Here, we identify a 9.4-kDa polypeptide in rat kidney extracts that appears, based on its biochemical and immunological properties, to be authentic prouroguanylin (proUgn). This propeptide is relatively plentiful in the kidney (~16% of intestinal levels), whereas its mRNA is marginally present (<1% of intestinal levels), and free Ugn peptide levels are below detection limits (<0.4% of renal proUgn levels). The paucity of preproUgn-encoding mRNA and free Ugn peptide raises the possibility that the kidney might absorb intact proUgn from plasma, where the concentration of propeptide greatly exceeds that of Ugn. However, immunocytochemical analysis reveals that renal proUgn is found exclusively in distal tubular segments, sites previously shown not to accumulate radiolabeled proUgn after intravascular infusions. Thus proUgn appears to be synthesized within the kidney, but the factors that determine its abundance (rates of transcription, translation, processing, and secretion) must be balanced quite differently than in the gut. Surprisingly, we also find negligible expression of GC-C in the rat kidney, a result confirmed both by RT-PCR and by functional assays that measure Ugn-activated cGMP synthesis. Taken together, these data provide evidence for an intrarenal Ugn system that differs from the well-described intestinal system in its regulatory mechanisms and in the receptor targeted by the peptide.
Collapse
Affiliation(s)
- Xun Qian
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC 27599-7545, USA
| | | | | | | | | |
Collapse
|
20
|
Moss NG, Riguera DA, Fellner RC, Cazzolla C, Goy MF. Natriuretic and antikaliuretic effects of uroguanylin and prouroguanylin in the rat. Am J Physiol Renal Physiol 2010; 299:F1433-42. [PMID: 20861080 DOI: 10.1152/ajprenal.00281.2010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The peptide uroguanylin (Ugn) is stored and released as a propeptide (proUgn) by enterochromaffin cells in the intestine, and converted to Ugn and other metabolites in the renal tubules. Both proUgn and Ugn are natriuretic, although the response to proUgn is thought to depend on its conversion to Ugn within nephrons. To assess the efficiency of intrarenal conversion of proUgn to Ugn, we measured urinary Ugn excretion in rats following intravenous infusions of proUgn or Ugn. Infusion of 2 and 10 nmol proUgn/kg body wt increased plasma proUgn concentration from 2.2 ± 0.3 to 5.6 ± 1.3 pmol/ml and to 37 ± 9.6 pmol/ml, respectively. No proUgn was detected in urine before, during, or after proUgn infusions. These two proUgn infusion doses resulted in total Ugn recovery in urine of 162 ± 64 and 206 ± 39 pmol/kg body wt (9 and 2% of the infused amount, respectively). By contrast, the same molar amounts of Ugn resulted in 1,009 ± 477 and 5,352 ± 2,133 pmol/kg body wt of Ugn in urine (recoveries of ∼50%). Unexpectedly, comparisons of natriuretic dose-response curves for each peptide showed proUgn to be about five times more potent than Ugn, despite the relatively modest amount of Ugn generated from infused proUgn. In addition, both peptides were antikaliuretic at low doses, but in this case Ugn showed greater potency than proUgn. These data do not support Ugn as the primary active principle of proUgn for regulation of renal sodium excretion. Instead, an alternative peptide fragment produced from proUgn may be responsible for natriuretic activity in the kidney, whereas Ugn itself may play an antikaliuretic role.
Collapse
Affiliation(s)
- Nicholas G Moss
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | |
Collapse
|
21
|
Bacterial heat-stable enterotoxins: translation of pathogenic peptides into novel targeted diagnostics and therapeutics. Toxins (Basel) 2010; 2:2028-54. [PMID: 22069671 PMCID: PMC3153287 DOI: 10.3390/toxins2082028] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2010] [Accepted: 08/03/2010] [Indexed: 12/13/2022] Open
Abstract
Heat-stable toxins (STs) produced by enterotoxigenic bacteria cause endemic and traveler’s diarrhea by binding to and activating the intestinal receptor guanylyl cyclase C (GC-C). Advances in understanding the biology of GC-C have extended ST from a diarrheagenic peptide to a novel therapeutic agent. Here, we summarize the physiological and pathophysiological role of GC-C in fluid-electrolyte regulation and intestinal crypt-villus homeostasis, as well as describe translational opportunities offered by STs, reflecting the unique characteristics of GC-C, in treating irritable bowel syndrome and chronic constipation, and in preventing and treating colorectal cancer.
Collapse
|
22
|
Moss NG, Riguera DA, Solinga RM, Kessler MM, Zimmer DP, Arendshorst WJ, Currie MG, Goy MF. The natriuretic peptide uroguanylin elicits physiologic actions through 2 distinct topoisomers. Hypertension 2009; 53:867-76. [PMID: 19289652 DOI: 10.1161/hypertensionaha.108.128264] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The peptide uroguanylin regulates electrolyte transport in the intestine and kidney. Human uroguanylin has 2 conformations that can be stably isolated because of their slow interconversion rate. The A isomer potently activates the guanylate cyclase C receptor found primarily in the intestine. The B isomer, by contrast, is a very weak agonist of this receptor, leading to a widely held assumption that it is physiologically irrelevant. We show here, however, that human uroguanylin B has potent natriuretic activity in the kidney. Interestingly, uroguanylin A and B both induce saluretic responses, but the activity profiles for the 2 peptides differ markedly. The uroguanylin B dose-response curve is sigmoidal with a threshold dose of approximately 10 nmol/kg of body weight, whereas uroguanylin A has a comparable threshold but a bell-shaped dose-response curve. In addition, our study indicates a unique interplay between the A and B isoforms, such that the A form at high concentrations antagonizes the natriuretic action of the B form. These data show that the kidney contains a uroguanylin receptor of which the pharmacological profile does not match that of the well-defined intestinal uroguanylin receptor (guanylate cyclase C), an observation consistent with previous studies showing that the kidney of the guanylate cyclase C knockout mouse remains responsive to uroguanylin. The results presented here also support the unconventional notion that distinct conformations of a single endocrine peptide can elicit different responses in different tissues.
Collapse
Affiliation(s)
- Nicholas G Moss
- University of North Carolina at Chapel Hill, Department of Cell and Molecular Physiology, 111 Mason Farm Rd, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Bengtsson MW, Jedstedt G, Flemström G. Duodenal bicarbonate secretion in rats: stimulation by intra-arterial and luminal guanylin and uroguanylin. Acta Physiol (Oxf) 2007; 191:309-17. [PMID: 17995576 DOI: 10.1111/j.1748-1716.2007.01759.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIM Uroguanylin and guanylin are endogenous ligands for guanylate cyclase C, an upstream regulator of the cystic fibrosis transmembrane resistance (CFTR) anion channel, and both peptides increase intestinal anion export in vitro. We have compared the effects of close intra-arterial and luminal administration of uroguanylin and guanylin on duodenal bicarbonate secretion in vivo and studied the interactions with melatonin and cholinergic stimulation. METHODS Lewis x Dark Agouti rats were anaesthetized and a segment of the proximal duodenum with intact blood supply was cannulated in situ. Mucosal bicarbonate secretion (pH stat) was continuously recorded and peptides were infused intra-arterially or added to the luminal perfusate. RESULTS Intra-arterial (50-1000 pmol kg(-1) h(-1)) as well as luminal administration (50-500 nmol L(-1)) of guanylin or uroguanylin caused dose-dependent increases in the duodenal secretion. Luminal administration induced more rapidly appearing rises in secretion and the two peptides induced secretory responses of similar shape and magnitude. The melatonin MT(2)-selective antagonist luzindole (600 nmol kg(-1)) significantly depressed the response to intra-arterial guanylins but did not affect secretion induced by luminal guanylins. Similarly, the muscarinic antagonist atropine (0.75 micromol kg(-1) followed by 0.15 micromol kg(-1) h(-1)) abolished the response to intra-arterial uroguanylin but caused only slight suppression of the response to luminal uroguanylin. CONCLUSIONS Intra-arterial as well as luminal uroguanylin and guanylin are potent stimuli of duodenal mucosal bicarbonate secretion in vivo. The response to luminal guanylins reflects an action at apical receptors. Stimulation by parenteral guanylins, in contrast, is under cholinergic influence and interacts with melatonin produced by mucosal enteroendocrine cells.
Collapse
Affiliation(s)
- M W Bengtsson
- Department of Neuroscience, Division of Physiology, Uppsala University, Uppsala, Sweden
| | | | | |
Collapse
|
24
|
Takei Y, Yuge S. The intestinal guanylin system and seawater adaptation in eels. Gen Comp Endocrinol 2007; 152:339-51. [PMID: 17561018 DOI: 10.1016/j.ygcen.2007.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Revised: 04/23/2007] [Accepted: 05/07/2007] [Indexed: 11/22/2022]
Abstract
Guanylin and uroguanylin are principal intestinal hormones secreted into the lumen to regulate ion and water absorption via a specific receptor, guanylyl cyclase-C (GC-C). As the intestine is an essential organ for seawater (SW) adaptation in teleost fishes, the intestinal guanylin system may play a critical role in SW adaptation. Molecular biological studies identified multiple guanylins (guanylin, uroguanylin and renoguanylin) and their receptors (GC-C1 and GC-C2) in eels. The relative potency of the three ligands on cGMP production in transiently expressed receptors was uroguanylin > guanylin >or= renoguanylin for CG-C1 and guanylin >or= renoguanylin > uroguanylin for GC-C2. Eel guanylin and GC-C genes are expressed exclusively in the intestine and kidney, and the level of expression is greater in SW eels than in freshwater (FW) eels except for renoguanylin. Physiological studies using Ussing chambers showed that the middle and posterior intestine are major sites of action of guanylins, where they act on the mucosal side to decrease short circuit current (I(sc)) in a dose-dependent manner. The ID(50) of guanylins for transport inhibition was 50-fold greater than that of atrial natriuretic peptide that acts from the serosal side as an endocrine hormone. However, only guanylins reversed I(sc) to levels below zero. Pharmacological analyses using various blockers showed that among transporters and channels localized on the intestinal cells of SW teleost fish, the cystic fibrosis transmembrane conductance regulator Cl(-) channel (CFTR) on the apical membrane is the major target of guanylins. Collectively, guanylins are synthesized locally in the intestine and secreted into the lumen to act on the GC-Cs in the apical membrane of eel intestinal cells. Then, intracellular cGMP production after ligand-receptor interaction activates CFTR and probably induces Cl(-) and/or HCO3- secretion into the lumen as suggested in mammals. The physiological significance of the anion secretion induced by the luminal guanylin/GC-C system on SW adaptation may rival or exceed that of the serosally derived natriuretic peptides in the euryhaline eel.
Collapse
Affiliation(s)
- Yoshio Takei
- Laboratory of Physiology, Ocean Research Institute, The University of Tokyo, 1-15-1 Minamidai, Nakano, Tokyo 164-8639, Japan.
| | | |
Collapse
|
25
|
D'Este L, Casini A, Cetin Y, Wenger T, Renda TG. Guanylin-immunoreactive cells in the female and male rat adenohypophysis and their changes under various physiological and experimental conditions. Histochem Cell Biol 2005; 123:303-13. [PMID: 15812648 DOI: 10.1007/s00418-004-0738-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2004] [Indexed: 11/29/2022]
Abstract
The peptide guanylin, first isolated from rat small intestine, is involved in the regulation of water-electrolyte transport between the intracellular and extracellular compartments of the epithelia. The main sites of guanylin expression are the intestinal, airway, or exocrine gland ductal epithelia where guanylin acts in a paracrine/luminocrine fashion. Because guanylin also circulates in the blood, sources of this peptide were sought in endocrine glands. Our group has already demonstrated the presence of guanylin-immunoreactive cells in the pars tuberalis of male rat adenohypophysis. In this study, we investigated whether guanylin-immunoreactive cells exist also in the adenohypophysial pars distalis and whether their appearance or distribution correlates with various physiological conditions in female rats or alters after gonadectomy in both sexes. These studies revealed that the rat pars distalis contains two guanylin-immunoreactive cell types, gonadotrophic cells, whose number varied notably during the estrous cycle, reached a peak in the proestrous phase, and increased consistently during pregnancy, in lactating animals, and after gonadectomy, and folliculo-stellate cells, a discrete number of which were found only in female rats at the estrous phase. These findings suggest that guanylin is involved in regulating gonadotrophic cell function. They also add important information on the controversially discussed functions of folliculo-stellate cells.
Collapse
Affiliation(s)
- Loredana D'Este
- Department of Human Anatomy, University La Sapienza, Via Alfonso Borelli, 50-00161 Rome, Italy.
| | | | | | | | | |
Collapse
|
26
|
Lorenz JN, Nieman M, Sabo J, Sanford LP, Hawkins JA, Elitsur N, Gawenis LR, Clarke LL, Cohen MB. Uroguanylin knockout mice have increased blood pressure and impaired natriuretic response to enteral NaCl load. J Clin Invest 2003; 112:1244-54. [PMID: 14561709 PMCID: PMC213491 DOI: 10.1172/jci18743] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Guanylin and uroguanylin, peptides synthesized in the intestine and kidney, have been postulated to have both paracrine and endocrine functions, forming a potential enteric-renal link to coordinate salt ingestion with natriuresis. To explore the in vivo role of uroguanylin in the regulation of sodium excretion, we created gene-targeted mice in which uroguanylin gene expression had been ablated. Northern and Western analysis confirmed the absence of uroguanylin message and protein in knockout mice, and cGMP levels were decreased in the mucosa of the small intestine. Ussing chamber analysis of jejunum revealed that Na+/H+ exchanger-mediated Na+ absorption and tissue conductance was not altered in the knockout animals, but short-circuit current, an index of electrogenic anion secretion, was reduced. Renal clearance measurements showed that uroguanylin deficiency results in impaired ability to excrete an enteral load of NaCl, primarily due to an inappropriate increase in renal Na+ reabsorption. Finally, telemetric recordings of blood pressure demonstrated increased mean arterial pressure in uroguanylin knockout animals that was independent of the level of dietary salt intake. Together, these findings establish a role for uroguanylin in an enteric-renal communication axis as well as a fundamental principle of this axis in the maintenance of salt homeostasis in vivo.
Collapse
Affiliation(s)
- John N Lorenz
- Department of Molecular and Cellular Physiology, University of Cincinnati, School of Medicine, Cincinnati, Ohio 45267-0576, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Lorenz JN, Nieman M, Sabo J, Sanford LP, Hawkins JA, Elitsur N, Gawenis LR, Clarke LL, Cohen MB. Uroguanylin knockout mice have increased blood pressure and impaired natriuretic response to enteral NaCl load. J Clin Invest 2003. [DOI: 10.1172/jci200318743] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
28
|
Yuge S, Inoue K, Hyodo S, Takei Y. A novel guanylin family (guanylin, uroguanylin, and renoguanylin) in eels: possible osmoregulatory hormones in intestine and kidney. J Biol Chem 2003; 278:22726-33. [PMID: 12684514 DOI: 10.1074/jbc.m303111200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
As the intestine is an essential organ for fish osmoregulation, the intestinal hormone guanylins may perform major functions, especially in euryhaline fish such as eels and salmonids. From the intestine of an eel, we identified cDNAs encoding three distinct guanylin-like peptides. Based on the sequence of mature peptide and sites of production, we named them guanylin, uroguanylin, and renoguanylin. Renoguanylin is a novel peptide that possesses the characteristics of both guanylin and uroguanylin and was abundantly expressed in the kidney. By immunohistochemistry, guanylin was localized exclusively in goblet cells, but not enterochromaffin cells, of the intestine. After transfer of eels from fresh water to seawater, mRNA expression of guanylin and uroguanylin did not change for 3 h, but it increased after 24 h. The increase was profound (2-6-fold) after adaptation to seawater. The expression of uroguanylin was also up-regulated in the kidney of seawater-adapted eels, but that of renoguanylin was not so prominent as other guanylins in both intestine and kidney. Collectively, the novel eel guanylin family appears to have important functions for seawater adaptation, particularly long-term adaptation. Eel guanylin may be secreted from goblet cells into the lumen with mucus in response to increased luminal osmolality and act on the epithelium to regulate water and salt absorption.
Collapse
Affiliation(s)
- Shinya Yuge
- Ocean Research Institute, The University of Tokyo, Nakano, Tokyo 164-8639, Japan.
| | | | | | | |
Collapse
|
29
|
Steinbrecher KA, Wowk SA, Rudolph JA, Witte DP, Cohen MB. Targeted inactivation of the mouse guanylin gene results in altered dynamics of colonic epithelial proliferation. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 161:2169-78. [PMID: 12466132 PMCID: PMC1850912 DOI: 10.1016/s0002-9440(10)64494-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Heat-stable enterotoxin (STa), elaborated by enterotoxigenic Echerichia coli, is a worldwide cause of secretory diarrhea in infants and travelers. Both STa and guanylin, a peptide structurally similar to STa, increase intracellular cGMP levels after binding to the same intestinal receptor, guanylate cyclase C (GC-C). Distinct from its role as an intestinal secretagogue, guanylin may also have a role in intestinal proliferation, as guanylin expression is lost in intestinal adenomas. To determine the function of guanylin in intestinal epithelia, guanylin null mice were generated using a Cre/loxP-based targeting vector. Guanylin null mice grew normally, were fertile and showed no signs of malabsorption. However, the levels of cGMP in colonic mucosa of guanylin null mice were significantly reduced. The colonic epithelial cell migration rate was increased and increased numbers of colonocytes expressing proliferating cell nuclear antigen (PCNA) were present in crypts of guanylin null mice as well. The apoptotic index was similar in guanylin null mice and littermate controls. We conclude from these studies that loss of guanylin results in increased proliferation of colonic epithelia. We speculate that the increase in colonocyte number is related to decreased levels of cGMP and that this increase in proliferation plays a role in susceptibility to intestinal adenoma formation and/or progression.
Collapse
Affiliation(s)
- Kris A Steinbrecher
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital Research Foundation, Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
30
|
Rudolph JA, Hawkins JA, Cohen MB. Proguanylin secretion and the role of negative-feedback inhibition in a villous epithelial cell line. Am J Physiol Gastrointest Liver Physiol 2002; 283:G695-702. [PMID: 12181185 DOI: 10.1152/ajpgi.00433.2001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The mechanisms of proguanylin synthesis and secretion in the intestine are incompletely understood. We designed an in vitro model to study proguanylin secretion in a model of intestinal villous epithelial cells. The C2/bbe1 cell line, a differentiated subclone of Caco-2 cells, was used to examine the direction of proguanylin secretion and the potential for feedback regulation via activators of the guanylyl cyclase C signal transduction pathway. When cells were grown on Transwell inserts, proguanylin was secreted into the apical and basolateral media, consistent with other models of intestinal guanylin secretion. Proguanylin synthesis and secretion were not decreased on activation of guanylyl cyclase C-mediated chloride secretion, implying a regulatory system other than negative-feedback inhibition. These data describe the use of C2/bbe1 cells as a model for proguanylin secretion in villous epithelial cells and demonstrate their potential use for the study of the regulatory mechanisms governing proguanylin synthesis and secretion.
Collapse
Affiliation(s)
- Jeffrey A Rudolph
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA.
| | | | | |
Collapse
|
31
|
Kunzelmann K, Mall M. Electrolyte transport in the mammalian colon: mechanisms and implications for disease. Physiol Rev 2002; 82:245-89. [PMID: 11773614 DOI: 10.1152/physrev.00026.2001] [Citation(s) in RCA: 453] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The colonic epithelium has both absorptive and secretory functions. The transport is characterized by a net absorption of NaCl, short-chain fatty acids (SCFA), and water, allowing extrusion of a feces with very little water and salt content. In addition, the epithelium does secret mucus, bicarbonate, and KCl. Polarized distribution of transport proteins in both luminal and basolateral membranes enables efficient salt transport in both directions, probably even within an individual cell. Meanwhile, most of the participating transport proteins have been identified, and their function has been studied in detail. Absorption of NaCl is a rather steady process that is controlled by steroid hormones regulating the expression of epithelial Na(+) channels (ENaC), the Na(+)-K(+)-ATPase, and additional modulating factors such as the serum- and glucocorticoid-regulated kinase SGK. Acute regulation of absorption may occur by a Na(+) feedback mechanism and the cystic fibrosis transmembrane conductance regulator (CFTR). Cl(-) secretion in the adult colon relies on luminal CFTR, which is a cAMP-regulated Cl(-) channel and a regulator of other transport proteins. As a consequence, mutations in CFTR result in both impaired Cl(-) secretion and enhanced Na(+) absorption in the colon of cystic fibrosis (CF) patients. Ca(2+)- and cAMP-activated basolateral K(+) channels support both secretion and absorption of electrolytes and work in concert with additional regulatory proteins, which determine their functional and pharmacological profile. Knowledge of the mechanisms of electrolyte transport in the colon enables the development of new strategies for the treatment of CF and secretory diarrhea. It will also lead to a better understanding of the pathophysiological events during inflammatory bowel disease and development of colonic carcinoma.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Department of Physiology and Pharmacology, University of Queensland, St. Lucia, Queensland, Brisbane, Australia.
| | | |
Collapse
|
32
|
Steinbrecher KA, Mann EA, Giannella RA, Cohen MB. Increases in guanylin and uroguanylin in a mouse model of osmotic diarrhea are guanylate cyclase C-independent. Gastroenterology 2001; 121:1191-202. [PMID: 11677212 DOI: 10.1053/gast.2001.28680] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Guanylin and uroguanylin are peptide hormones that are homologous to the diarrhea-causing Escherichia coli enterotoxins. These secretagogues are released from the intestinal epithelia into the intestinal lumen and systemic circulation and bind to the receptor guanylate cyclase C (GC-C). We hypothesized that a hypertonic diet would result in osmotic diarrhea and cause a compensatory down-regulation of guanylin/uroguanylin. METHODS Gut-to-carcass weights were used to measure fluid accumulation in the intestine. Northern and/or Western analysis was used to determine the levels of guanylin, uroguanylin, and GC-C in mice with osmotic diarrhea. RESULTS Wild-type mice fed a polyethylene glycol or lactose-based diet developed weight loss, diarrhea, and an increased gut-to-carcass ratio. Unexpectedly, 2 days on either diet resulted in increased guanylin/uroguanylin RNA and prohormone throughout the intestine, elevated uroguanylin RNA, and prohormone levels in the kidney and increased levels of circulating prouroguanylin. GC-C-deficient mice given the lactose diet reacted with higher gut-to-carcass ratios. Although they did not develop diarrhea, GC-C-sufficient and -deficient mice on the lactose diet responded with elevated levels of guanylin and uroguanylin RNA and protein. A polyethylene glycol drinking water solution resulted in diarrhea, higher gut-to-carcass ratios, and induction of guanylin and uroguanylin in both GC-C heterozygous and null animals. CONCLUSIONS We conclude that this model of osmotic diarrhea results in a GC-C-independent increase in intestinal fluid accumulation, in levels of these peptide ligands in the epithelia of the intestine, and in prouroguanylin in the kidney and blood.
Collapse
Affiliation(s)
- K A Steinbrecher
- Division of Pediatric Gastroenterology, Hepatology and Nutrition and Graduate Program in Molecular and Developmental Biology, Children's Hospital Research Foundation, Cincinnati, Ohio, USA
| | | | | | | |
Collapse
|
33
|
Di Guglielmo MD, Park J, Schulz S, Waldman SA. Nucleotide requirements for CDX2 binding to the cis promoter element mediating intestine-specific expression of guanylyl cyclase C. FEBS Lett 2001; 507:128-32. [PMID: 11684084 DOI: 10.1016/s0014-5793(01)02952-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Guanylyl cyclase C (GC-C), specifically expressed by intestinal epithelial cells, is the receptor for the Escherichia coli heat-stable enterotoxin that causes diarrhea. Tissue-specific expression of GC-C is mediated by the intestinal transcriptional regulator CDX2. This trans-activating protein regulates intestine-specific expression by binding to a critical sequence in the proximal promoter of GC-C. The precise nucleotide elements mediating CDX2 binding to promoter elements remain undefined. Several nuclear proteins form complexes with a DNA probe containing the promoter element of GC-C mediating CDX2 binding. The present study examined the nucleotide requirements in the consensus binding site and flanking regions in the cis element that mediates specific CDX2 binding to the promoter of GC-C. These studies identified seven core base pairs in the critical promoter element mediating tissue-specific expression of GC-C that are required for CDX2 binding. In addition, base pairs flanking this core sequence contribute to and are required for CDX2 recognition. These studies describe the precise nucleotide sequence within the GC-C promoter that comprises the CDX2 binding site required for intestine-specific expression.
Collapse
Affiliation(s)
- M D Di Guglielmo
- Division of Clinical Pharmacology, Departments of Medicine, Biochemistry and Molecular Pharmacology, Thomas Jefferson University, 812 Medical Office Building, 1100 Walnut Street, Philadelphia, PA 19107, USA.
| | | | | | | |
Collapse
|
34
|
Potthast R, Ehler E, Scheving LA, Sindic A, Schlatter E, Kuhn M. High salt intake increases uroguanylin expression in mouse kidney. Endocrinology 2001; 142:3087-97. [PMID: 11416031 DOI: 10.1210/endo.142.7.8274] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The intestinal peptides, guanylin and uroguanylin, may have an important role in the endocrine control of renal function. Both peptides and their receptor, guanylyl cyclase C (GC-C), are also expressed within the kidney, suggesting that they may act locally in an autocrine/paracrine fashion. However, their physiological regulation within the kidney has not been studied. To begin to address this issue, we evaluated the distribution of uroguanylin and guanylin messenger RNA (mRNA) in the mouse nephron and the regulation of renal expression by changes in dietary salt/water intake. Expression was determined in 1) wild-type mice, 2) two strains of receptor-guanylyl cyclase-deficient mice (ANP-receptor-deficient, GC-A-/-, and GC-C-deficient mice); and 3) cultured renal epithelial (M-1) cells, by RT-PCR, Northern blotting and immunocytochemistry. Renal uroguanylin messenger RNA expression was higher than guanylin and had a different distribution pattern, with highest levels in the proximal tubules, whereas guanylin was mainly expressed in the collecting ducts. Uroguanylin expression was significantly lower in GC-C-/- mice than in GC-A-/- and wild-types, suggesting that absence of a receptor was able to down-regulate ligand expression. Salt-loading (1% NaCl in drinking water) increased uroguanylin-mRNA expression by >1.8-fold but had no effect on guanylin expression. Uroguanylin but not guanylin transcripts were detected in M-1 cells and increased in response to hypertonic media (+NaCl or mannitol). Our results indicate that high-salt intake increases uroguanylin but not guanylin expression in the mouse kidney. The synthesis of these peptides by tubular epithelium may contribute to the local control of renal function and its adaptation to dietary salt.
Collapse
Affiliation(s)
- R Potthast
- Institute of Pharmacology and Toxicology and Department of Internal Medicine, Experimental Nephrology, Westfaelische Wilhelms-Universitaet Muenster, Muenster 48129, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Bold AJ, Bruneau BG. Natriuretic Peptides. Compr Physiol 2000. [DOI: 10.1002/cphy.cp070310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
36
|
Qian X, Prabhakar S, Nandi A, Visweswariah SS, Goy MF. Expression of GC-C, a receptor-guanylate cyclase, and its endogenous ligands uroguanylin and guanylin along the rostrocaudal axis of the intestine. Endocrinology 2000; 141:3210-24. [PMID: 10965892 DOI: 10.1210/endo.141.9.7644] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Members of the receptor-guanylate cyclase (rGC) family possess an intracellular catalytic domain that is regulated by an extracellular receptor domain. GC-C, an intestinally expressed rGC, was initially cloned by homology as an orphan receptor. The search for its ligands has yielded three candidates: STa (a bacterial toxin that causes traveler's diarrhea) and the endogenous peptides uroguanylin and guanylin. Here, by performing Northern and Western blots, and by measuring [125I]STa binding and STa-dependent elevation of cGMP levels, we investigate whether the distribution of GC-C matches that of its endogenous ligands in the rat intestine. We establish that 1) uroguanylin is essentially restricted to small bowel; 2) guanylin is very low in proximal small bowel, increasing to prominent levels in distal small bowel and throughout colon; 3) GC-C messenger RNA and STa-binding sites are uniformly expressed throughout the intestine; and 4) GC-C-mediated cGMP synthesis peaks at the proximal and distal extremes of the intestine (duodenum and colon), but is nearly absent in the middle (ileum). These observations suggest that GC-C's activity may be posttranslationally regulated, demonstrate that the distribution of GC-C is appropriate to mediate the actions of both uroguanylin and guanylin, and help to refine current hypotheses about the physiological role(s) of these peptides.
Collapse
Affiliation(s)
- X Qian
- Department of Cell and Molecular Physiology, University of North Carolina, Chaptel Hill 27599-7545, USA
| | | | | | | | | |
Collapse
|
37
|
Moro F, Levenez F, Nemoz-Gaillard E, Pellissier S, Plaisancie P, Cuber JC. Release of guanylin immunoreactivity from the isolated vascularly perfused rat colon. Endocrinology 2000; 141:2594-9. [PMID: 10875263 DOI: 10.1210/endo.141.7.7574] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The intestinal peptide guanylin regulates the electrolyte/water transport in the intestinal epithelium. The aim of the present study was to investigate the mechanisms that modulate its secretion in the isolated vascularly perfused rat colon by using a specific guanylin RIA. Intraarterial infusion of bethanechol (10(-4) M) or bombesin (10(-7) M) elicited a significant 6-fold increase in the release of guanylin immunoreactivity (G-IR) in the lumen. Bombesin-stimulated G-IR secretion was strongly reduced by tetrodotoxin, whereas atropine had no effect. VIP (10(-7) M) induced a moderate release of G-IR, whereas substance P, calcitonin gene-related peptide, peptide YY, somatostatin, and neurotensin were without effect. Dimethyl-PGE2 (1.4 x 10(-5) M) or interleukin-1beta (2.5 x 10(-10) M) induced a 3-fold increase in G-IR in the lumen, whereas the degranulator compound bromolasalocid did not stimulate guanylin secretion. Forskolin (10(-5) M) or sodium nitroprusside (10(-4)-10(-3) M) induced a significant release of G-IR. In contrast, PMA (10(-7) M) or ionophore A23187 (10(-6) M) did not modify basal secretion of G-IR. Upon stimulation of guanylin release with bombesin or bethanechol, an increase in G-IR in the portal effluent was also detected. The release of G-IR in the portal effluent was 40-fold lower than that of G-IR into the luminal perfusate. Additionally, analysis with gel chromatography revealed that the immunoreactive material released in the lumen or in the portal effluent coeluted with the 15-amino acid peptide originally isolated from rat intestine. In conclusion, the present data suggest that the enteric nervous system and immune cells may modulate guanylin release from the rat colon. The release of guanylin in the lumen and portal effluent suggests that this peptide may exert both luminal/paracrine and hormonal effects.
Collapse
Affiliation(s)
- F Moro
- INSERM, U-45, Hôpital Edouard Herriot, Lyon, France
| | | | | | | | | | | |
Collapse
|
38
|
Rambotti MG, Giambanco I, Spreca A. Ultracytochemical detection of guanylate cyclase C activity in alimentary tract and associated glands of the rat. Influence of pH, ATP and the ions Mg2+ and Mn2+. THE HISTOCHEMICAL JOURNAL 2000; 32:231-8. [PMID: 10872888 DOI: 10.1023/a:1004003101495] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Intestinal guanylate cyclase C is activated by guanylin, an endogenous peptide. This activity seems to be modulated by adenine nucleotides, the ions Mg2+ and Mn2+, and pH. In this study, we report an ultracytochemical method for the localization of guanylate cyclase C activity at the electron microscope level. We studied the enzymatic activity in the presence or absence of guanylin and/or ATP, in the presence of the ions Mg2+ or Mn2+, and at different pH levels. The greatest distribution of enzymatic activity was detected in samples incubated at pH 8 and 7.4 in the presence of guanylin, Mg2+ and ATP. Guanylate cyclase C activity was detected at the surface epithelium of stomach and intestine, and in liver, exocrine pancreas and parotid gland. In the intestine, enzymatic activity was more widely distributed in the duodenum than in the jejunum-ileum and colon. In the small intestine, activity was more evident in the upper portion than in the basal portion of the villus. In samples incubated at pH 8 and 7.4 in the absence of ATP, enzymatic activity was detected only in small intestine, liver and exocrine pancreas. Enzymatic activity was present in duodenum incubated at pH 8 and 7.4 in the presence of Mn2+ and in the presence or absence of ATP. No samples incubated in all these experimental conditions but at pH 5 or samples incubated in the presence of guanylin only or in the absence of guanylin, displayed guanylate cyclase C activity. Our results suggest that a complete ultracytochemical detection of guanylate cyclase C activity requires guanylin as stimulator, and incubation in the presence of Mg2+ and ATP at pH 8 and 7.4.
Collapse
Affiliation(s)
- M G Rambotti
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Italy
| | | | | |
Collapse
|
39
|
D'Este L, Kulaksiz H, Rausch U, Vaccaro R, Wenger T, Tokunaga Y, Renda TG, Cetin Y. Expression of guanylin in "pars tuberalis-specific cells" and gonadotrophs of rat adenohypophysis. Proc Natl Acad Sci U S A 2000; 97:1131-6. [PMID: 10655496 PMCID: PMC15545 DOI: 10.1073/pnas.97.3.1131] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The intestinal peptide guanylin regulates the electrolyte/water transport in the gastrointestinal epithelium by paracrine/luminocrine mechanisms. Because guanylin also circulates in the blood, we investigated the rat hypothalamo-pituitary region for expression and cellular localization of this peptide. Reverse transcriptase-PCR analyses with guanylin-specific primers revealed expression of the peptide in the pars tuberalis and pars distalis of the pituitary. Western blotting analyses in hypophyseal tissue extracts identified the expected 12.5-kDa immunoreactive peptide by using two different region-specific guanylin antisera. Light and electron microscopic immunocytochemistry with the same antisera localized guanylin in "pars tuberalis-specific cells" in the juxtaneural pars tuberalis adjacent to nerve endings and blood vessels of the hypothalamo-pituitary portal system and in gonadotrophic cells within the distal pars tuberalis and ventrolateral part of the pars distalis. The presence and cell-specific localization of guanylin within the hypothalamo-hypophyseal system indicate that this peptide may be specifically involved in paracrine and endocrine regulatory mechanisms.
Collapse
Affiliation(s)
- L D'Este
- Institute of Human Anatomy, University "La Sapienza," 00161 Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Scheving LA, Jin WH. Circadian regulation of uroguanylin and guanylin in the rat intestine. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:C1177-83. [PMID: 10600769 DOI: 10.1152/ajpcell.1999.277.6.c1177] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Uroguanylin (UGN) and guanylin (GN) are the endogenous intestinal ligands for guanylyl cyclase C (GC-C). We examined the circadian expression of UGN, GN, and GC-C in the jejunum, ileum, and proximal colon of young adult rats by Northern blot analyses. These assays revealed that UGN is more abundant in the proximal small intestine, whereas GN and GC-C are more abundant in the proximal colon. mRNA levels showed significant circadian variation for UGN (3- to 18-fold peak/trough difference), GN (2.1- to 2.8-fold peak/trough difference), and GC-C (3- to 5-fold peak/trough difference). The maximal abundance occurred in the dark period for all three mRNAs, although peak UGN and GN expression occurred later in the dark period in the jejunum relative to the ileum and colon. Immunoblot analyses using monospecific polyclonal antibodies against UGN and GN prohormones confirmed the regional and circadian variation detected by Northern assays. Thus the expression of these genes is regulated not only by histological position but also by circadian time.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal
- Blotting, Northern
- Blotting, Western
- Circadian Rhythm/physiology
- Colon/chemistry
- Colon/enzymology
- Enzyme Activators/analysis
- Enzyme Activators/immunology
- Enzyme Activators/metabolism
- Gastrointestinal Hormones
- Gene Expression/physiology
- Guanylate Cyclase/analysis
- Guanylate Cyclase/genetics
- Guanylate Cyclase/immunology
- Ileum/chemistry
- Ileum/enzymology
- Intestines/chemistry
- Intestines/enzymology
- Jejunum/chemistry
- Jejunum/enzymology
- Male
- Molecular Sequence Data
- Natriuretic Peptides
- Peptides/analysis
- Peptides/genetics
- Peptides/immunology
- Protein Precursors/analysis
- Protein Precursors/genetics
- Protein Precursors/immunology
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Enterotoxin
- Receptors, Guanylate Cyclase-Coupled
- Receptors, Peptide/analysis
- Receptors, Peptide/genetics
- Receptors, Peptide/immunology
Collapse
Affiliation(s)
- L A Scheving
- Division of Gastroenterology and Nutrition, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA.
| | | |
Collapse
|
42
|
Martin S, Adermann K, Forssmann WG, Kuhn M. Regulated, side-directed secretion of proguanylin from isolated rat colonic mucosa. Endocrinology 1999; 140:5022-9. [PMID: 10537127 DOI: 10.1210/endo.140.11.7103] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Guanylin, an activator of the guanylyl cyclase C receptor in the apical membrane of intestinal epithelium, modulates intestinal fluid and electrolyte transport. The bioactive 15-amino acid peptide originally isolated from rat intestine represents the C-terminal part of a longer, 115-residue prepropeptide. The aim of the present study was to characterize the direction and molecular form in which guanylin is secreted from the colonic mucosa, as well as the mechanisms that trigger its secretion. Isolated rat colonic mucosa was mounted in Ussing chambers, allowing the separate determination of apical and basolateral release. After HPLC purification, two different molecular forms of guanylin were identified in the apical incubation media by combining a bioassay for guanylyl cyclase C activation, a specific guanylin enzyme-linked immunosorbent assay and mass spectrometry, as well as sequence analysis: a bioactive form coeluting with synthetic 15-residue guanylin and the 94-residue propeptide, guanylin-22-115. The basal concentration of proguanylin at the apical side of epithelia was about 15-fold higher, compared with that of the small, bioactive peptide. In the basolateral incubation media, no proguanylin and only very low amounts of bioactive guanylin were detected. Incubation with carbachol led to a significant increase of about 7-fold in the release of proguanylin to both sides of the isolated epithelia. On the apical side, a concomitant increase of the small, bioactive peptide was observed; whereas, on the basolateral side, its concentration remained unchanged. Vasoactive intestinal peptide or the NO-donor S-nitroso-N-acetylpenicillamine did not affect guanylin secretion. Our results suggest that, in the intestine, guanylin is secreted mainly to the luminal side of the epithelium. The peptide is released as a 94-residue propeptide, which is then processed to a smaller, bioactive form (luminocrine secretion). Carbachol stimulates the release of proguanylin to both sides of the intestinal mucosa, but a parallel increase in the bioactive C-terminal derivative only occurs on the apical side. In vivo, the basolateral release could be a source of circulating proguanylin, which might be processed proteolytically to the active peptide in distant target tissues (endocrine secretion).
Collapse
Affiliation(s)
- S Martin
- Niedersächsisches Institut für Peptid-Forschung, Hannover, Germany
| | | | | | | |
Collapse
|
43
|
Forte LR. Guanylin regulatory peptides: structures, biological activities mediated by cyclic GMP and pathobiology. REGULATORY PEPTIDES 1999; 81:25-39. [PMID: 10395405 DOI: 10.1016/s0167-0115(99)00033-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The guanylin family of bioactive peptides consists of three endogenous peptides, including guanylin, uroguanylin and lymphoguanylin, and one exogenous peptide toxin produced by enteric bacteria. These small cysteine-rich peptides activate cell-surface receptors, which have intrinsic guanylate cyclase activity, thus modulating cellular function via the intracellular second messenger, cyclic GMP. Membrane guanylate cyclase-C is an intestinal receptor for guanylin and uroguanylin that is responsible for stimulation of Cl- and HCO3- secretion into the intestinal lumen. Guanylin and uroguanylin are produced within the intestinal mucosa to serve in a paracrine mechanism for regulation of intestinal fluid and electrolyte secretion. Enteric bacteria secrete peptide toxin mimics of uroguanylin and guanylin that activate the intestinal receptors in an uncontrolled fashion to produce secretory diarrhea. Opossum kidney guanylate cyclase is a key receptor in the kidney that may be responsible for the diuretic and natriuretic actions of uroguanylin in vivo. Uroguanylin serves in an endocrine axis linking the intestine and kidney where its natriuretic and diuretic actions contribute to the maintenance of Na+ balance following oral ingestion of NaCl. Lymphoguanylin is highly expressed in the kidney and myocardium where this unique peptide may act locally to regulate cyclic GMP levels in target cells. Lymphoguanylin is also produced in cells of the lymphoid-immune system where other physiological functions may be influenced by intracellular cyclic GMP. Observations of nature are providing insights into cellular mechanisms involving guanylin peptides in intestinal diseases such as colon cancer and diarrhea and in chronic renal diseases or cardiac disorders such as congestive heart failure where guanylin and/or uroguanylin levels in the circulation and/or urine are pathologically elevated. Guanylin peptides are clearly involved in the regulation of salt and water homeostasis, but new findings indicate that these novel peptides have diverse physiological roles in addition to those previously documented for control of intestinal and renal function.
Collapse
Affiliation(s)
- L R Forte
- The Harry S. Truman Memorial V.A. Hospital, Columbia, MO 65212, USA.
| |
Collapse
|
44
|
Date Y, Nakazato M, Yamaguchi H, Kangawa K, Kinoshita Y, Chiba T, Ueta Y, Yamashita H, Matsukura S. Enterochromaffin-like cells, a cellular source of uroguanylin in rat stomach. Endocrinology 1999; 140:2398-404. [PMID: 10218994 DOI: 10.1210/endo.140.5.6734] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Uroguanylin is an endogenous peptide ligand for guanylyl cyclase-C, an apical membrane receptor predominantly located in the gastrointestinal epithelium. It regulates intestinal and renal fluid and electrolyte transport through the second messenger, cyclic GMP. Uroguanylin messenger RNA and the peptide are present in rat stomach, but the cellular source has not been identified. We separated gastric mucosal cells by size into seven fractions (F1-F7) and enriched endocrine cells into F1-F3 using counterflow elutriation. Uroguanylin messenger RNA and peptide were found in F1-F3 by Northern blot analysis and an RIA specific for rat uroguanylin. Uroguanylin-producing cells were identified as endocrine cells by immunocytochemical methods using antisera for uroguanylin, prouroguanylin, and chromogranin A, as well as by in situ hybridization cytochemistry. Double-staining showed that uroguanylin and histamine are colocalized in enterochromaffin-like (ECL) cells that release histamine, leading to the stimulation of gastric acid secretion from parietal cells. Uroguanylin is synthesized in ECL cells. These findings should contribute to elucidating the physiological functions of ECL cells and the cyclic GMP-mediated gastric ion transport mechanism.
Collapse
Affiliation(s)
- Y Date
- Department of Internal Medicine, Miyazaki Medical College, Kiyotake, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mägert HJ, Reinecke M, David I, Raab HR, Adermann K, Zucht HD, Hill O, Hess R, Forssmann WG. Uroguanylin: gene structure, expression, processing as a peptide hormone, and co-storage with somatostatin in gastrointestinal D-cells. REGULATORY PEPTIDES 1998; 73:165-76. [PMID: 9556079 DOI: 10.1016/s0167-0115(97)01078-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Guanylin/GCAP-I and uroguanylin/GCAP-II are two structurally related peptides which play an important role in the regulation of water/electrolyte balance within the gut. In order to enable the investigation and comparison of both peptide hormones at the genomic level, we decided to clone the corresponding genes. The human gene for guanylin/GCAP-I and its 5'-flanking region have been described recently. Here, we report the three exon/two intron structure of the human uroguanylin/GCAP-II gene and its localization on chromosome 1 p35-34, as determined by radiation hybrid mapping. Together with data obtained for the guanylin/GCAP-I gene we show that these genes are localized in the same chromosomal area with other guanlyl cyclase-activating peptides like ANP etc. Northern hybridization revealed that the expression of the uroguanylin/GCAP-II gene is highest in the intestinal mucosa, especially in the ileum and colon. By means of polymerase chain reaction (PCR), an expression was also observed in the stomach where no guanylin/GCAP-I expression is detectable. Using immunohistochemical methods, uroguanylin/GCAP-II immunoreactive material was distinctly localized in D-type gastric and intestinal endocrine cells. Although the comparable data on the genomic organisation of both peptide hormones verify their high degree of relationship, this finding indicates a special task of uroguanylin/GCAP-II within the stomach, such as regulatory functions in gastric secretion. The redundant expression of the GCAP/GC-C system in the small and large intestine, however, is as yet unclear.
Collapse
Affiliation(s)
- H J Mägert
- Lower Saxony Institute for Peptide Research, Hannover, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Fan X, Hamra FK, London RM, Eber SL, Krause WJ, Freeman RH, Smith CE, Currie MG, Forte LR. Structure and activity of uroguanylin and guanylin from the intestine and urine of rats. THE AMERICAN JOURNAL OF PHYSIOLOGY 1997; 273:E957-64. [PMID: 9374682 DOI: 10.1152/ajpendo.1997.273.5.e957] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Uroguanylin and guanylin are related peptides that activate common guanylate cyclase signaling molecules in the intestine and kidney. Uroguanylin was isolated from urine and duodenum but was not detected in extracts from the colon of rats. Guanylin was identified in extracts from small and large intestine but was not detected in urine. Uroguanylin and guanylin have distinct biochemical and chromatographic properties that facilitated the separation, purification, and identification of these peptides. Northern assays revealed that mRNA transcripts for uroguanylin were more abundant in small intestine compared with large intestine, whereas guanylin mRNA levels were greater in large intestine relative to small intestine. Synthetic rat uroguanylin and guanylin had similar potencies in the activation of receptors in T84 intestinal cells. Production of uroguanylin and guanylin in the mucosa of duodenum is consistent with the postulate that both peptides influence the activity of an intracellular guanosine 3',5'-cyclic monophosphate signaling pathway that regulates the transepithelial secretion of chloride and bicarbonate in the intestinal epithelium.
Collapse
Affiliation(s)
- X Fan
- Truman Veterans Affairs Medical Center, Columbia, Missouri 65212, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Fan X, Wang Y, London RM, Eber SL, Krause WJ, Freeman RH, Forte LR. Signaling pathways for guanylin and uroguanylin in the digestive, renal, central nervous, reproductive, and lymphoid systems. Endocrinology 1997; 138:4636-48. [PMID: 9348189 DOI: 10.1210/endo.138.11.5539] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Guanylin and uroguanylin are peptides that stimulate membrane guanylate cyclases (GC) and regulate intestinal and renal function via cGMP. Complementary DNAs were isolated encoding opossum preproguanylin and a 279-amino acid portion of a receptor-guanylate cyclase expressed in opossum kidney (OK) cells (GC-OK). The tissue expression of messenger RNA transcripts for these signaling molecules were then compared. Northern and/or reverse transcription-PCR assays revealed that guanylin, uroguanylin, and GC-OK messenger RNAs are expressed in tissues within the digestive, renal, central nervous, reproductive, and lymphoid organ systems. Receptor autoradiography localized the receptors for uroguanylin and guanylin to renal proximal tubules and seminiferous tubules of testis. Synthetic guanylin and uroguanylin peptides activated the receptor-GCs in opossum kidney cortex and in cultured OK cells eliciting increased intracellular cGMP. Expression of agonist and receptor-GC signaling molecules provides a pathway for paracrine and/or autocrine regulation of cellular functions via cGMP in the digestive, renal, central nervous, reproductive, and lymphoid/immune organ systems. Uroguanylin also links the intestine and kidney in a potential endocrine axis that activates tubular receptor-GCs and influences renal function.
Collapse
Affiliation(s)
- X Fan
- The Truman Veterans Administration Medical Center, Department of Pharmacology, Missouri University School of Medicine, Columbia 65212, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Nandi A, Bhandari R, Visweswariah SS. Epitope conservation and immunohistochemical localization of the guanylin/stable toxin peptide receptor, guanylyl cyclase C. J Cell Biochem 1997; 66:500-11. [PMID: 9282328 DOI: 10.1002/(sici)1097-4644(19970915)66:4<500::aid-jcb9>3.0.co;2-p] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The heat-stable enterotoxins (ST) are a family of cysteine-rich low-molecular weight peptides produced by pathogenic bacteria, and are one of the major causes of watery diarrhea all over the world. These toxins mediate their action by binding to an intestinal cell surface receptor that is a membrane-associated guanylyl cyclase (GCC). This receptor also serves as the receptor for the recently characterised endogenous ligand, guanylin. We have expressed various domains of the receptor in Escherichia coli and used purified proteins for the generation of both polyclonal and monoclonal antibodies. While polyclonal antibodies were able to partially inhibit ST binding to the native receptor present in the T84 human colonic cell line, GCC:B10 monoclonal antibody did not interfere with ligand binding. Western blot analysis, using membranes prepared from human colonic T84 cells, detected two bands of size 160 and 140 kDa, representing alternately glycosylated forms of the receptor. Using the recombinant proteins, we could map the epitope of GCC:B10 monoclonal antibody to the intracellular domain of the receptor. We used the antibody to localize the receptor throughout the rat intestine, and in the porcine and bonnet monkey colon. We could detect receptor expression in the villus and the crypts of the duodenum, jejunum, ileum, and caecum, and in the crypts of the colon. Receptor expression was observed in cells that had earlier been shown to express cGMP-dependent kinase, but not the cystic fibrosis transmembrane regulator, a known downstream target of cGMP/G-kinase, which suggests that GCC/ cGMP could regulate additional cellular signal transduction machinery.
Collapse
Affiliation(s)
- A Nandi
- Center for Reproductive Biology and Molecular Endocrinology, Indian Institute of Science, Bangalore, India
| | | | | |
Collapse
|
49
|
Volant K, Grishina O, Descroix-Vagne M, Pansu D. Guanylin-, heat-stable enterotoxin of Escherichia coli- and vasoactive intestinal peptide-induced water and ion secretion in the rat intestine in vivo. Eur J Pharmacol 1997; 328:217-27. [PMID: 9218704 DOI: 10.1016/s0014-2999(97)83048-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The heat-stable enterotoxin of Escherichia coli binds to an intestinal receptor, guanylyl cyclase-C, and produces cGMP to induce diarrhea. Guanylin is an endogenous ligand of this receptor. In the present in vivo study, the intestinal water and ion secretion induced by mucosal application of 2 nmol/ml guanylin or 5 or 10 units/ml heat-stable enterotoxin into closed loops was compared in the rat. The characteristics of secretion induced by cAMP following intravenous perfusion of 1.2 nmol/100 g per h vasoactive intestinal peptide were compared to those induced by cGMP. Unidirectional Na+ and Cl- fluxes were estimated by addition of 22Na into the loop and i.v. injection of 36Cl. Guanylin induced less water and ion secretion than that produced by heat-stable enterotoxin in the colon, confirming the results of in vitro studies, and also in duodenum and ileum. The cAMP- or cGMP-mediated response had a similar pattern, i.e., an inhibition of Na+ absorption and an increase in anion secretion.
Collapse
Affiliation(s)
- K Volant
- Unité INSERM U 45: Systeme neuro-endocrine et épithélium intestinal normal et néoplasique, Hôpital Edouard Herriot, Lyon, France
| | | | | | | |
Collapse
|
50
|
Li Z, Perkins AG, Peters MF, Campa MJ, Goy MF. Purification, cDNA sequence, and tissue distribution of rat uroguanylin. REGULATORY PEPTIDES 1997; 68:45-56. [PMID: 9094754 DOI: 10.1016/s0167-0115(96)02103-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Guanylin, a peptide purified from rat jejunum, is thought to regulate water and electrolyte balance in the intestine. We show here, using a combination of Northern blots, Western blots, and functional assays, that guanylin and its receptor (GCC) are not distributed in parallel within the rat intestine. To investigate the possibility that there might be a second intestinal peptide that serves as a ligand for GCC, we assayed tissue extracts for the ability to stimulate cyclic GMP synthesis in a GCC-expression cell line. Duodenal extracts display a peak of biological activity that is not present in colon and that does not comigrate with guanylin or proguanylin. The activity co-purifies with a novel peptide (TIATDECELCINVACTGC) that has high homology with uroguanylin, a peptide initially purified from human and opossum urine. A rat uroguanylin cDNA clone was found to encode a propeptide whose C-terminus corresponds to our purified peptide. Northern blots with probes generated from this clone reveal that prouroguanylin mRNA is strongly expressed in proximal small intestine, but virtually absent from colon, corroborating our biochemical measurements. Taken together, these studies demonstrate an intestinal origin for uroguanylin, and show that within the intestine its distribution is complementary to that of guanylin.
Collapse
Affiliation(s)
- Z Li
- Department of Physiology, University of North Carolina, Chapel Hill 27599, USA.
| | | | | | | | | |
Collapse
|