1
|
Luo M, Liu Y, Nikolovska K, Riederer B, Patrucco E, Hofmann F, Seidler U. cGMP-dependent kinase 2, Na +/H + exchanger NHE3, and PDZ-adaptor NHERF2 co-assemble in apical membrane microdomains. Acta Physiol (Oxf) 2024; 240:e14125. [PMID: 38533975 DOI: 10.1111/apha.14125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/30/2024] [Accepted: 02/14/2024] [Indexed: 03/28/2024]
Abstract
AIM Trafficking, membrane retention, and signal-specific regulation of the Na+/H+ exchanger 3 (NHE3) are modulated by the Na+/H+ Exchanger Regulatory Factor (NHERF) family of PDZ-adapter proteins. This study explored the assembly of NHE3 and NHERF2 with the cGMP-dependent kinase II (cGKII) within detergent-resistant membrane microdomains (DRMs, "lipid rafts") during in vivo guanylate cycle C receptor (Gucy2c) activation in murine small intestine. METHODS Small intestinal brush border membranes (siBBMs) were isolated from wild type, NHE3-deficient, cGMP-kinase II-deficient, and NHERF2-deficient mice, after oral application of the heat-stable Escherichia coli toxin (STa) analog linaclotide. Lipid raft and non-raft fractions were separated by Optiprep density gradient centrifugation of Triton X-solubilized siBBMs. Confocal microscopy was performed to study NHE3 redistribution after linaclotide application in vivo. RESULTS In the WT siBBM, NHE3, NHERF2, and cGKII were strongly raft associated. The raft association of NHE3, but not of cGKII, was NHERF2 dependent. After linaclotide application to WT mice, lipid raft association of NHE3 decreased, that of cGKII increased, while that of NHERF2 did not change. NHE3 expression in the BBM shifted from a microvillar to a terminal web region. The linaclotide-induced decrease in NHE3 raft association and in microvillar abundance was abolished in cGKII-deficient mice, and strongly reduced in NHERF2-deficient mice. CONCLUSION NHE3, cGKII, and NHERF2 form a lipid raft-associated signal complex in the siBBM, which mediates the inhibition of salt and water absorption by Gucy2c activation. NHERF2 enhances the raft association of NHE3, which is essential for its close interaction with the exclusively raft-associated activated cGKII.
Collapse
Affiliation(s)
- Min Luo
- Department of Gastroenterology, Hepatology, Infectiology and Endocrinology, Hannover Medical School, Hannover, Germany
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yongjian Liu
- Department of Gastroenterology, Hepatology, Infectiology and Endocrinology, Hannover Medical School, Hannover, Germany
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Katerina Nikolovska
- Department of Gastroenterology, Hepatology, Infectiology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Brigitte Riederer
- Department of Gastroenterology, Hepatology, Infectiology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Enrico Patrucco
- Institut für Pharmakologie und Toxikologie, TU München, München, Germany
- Department of Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Franz Hofmann
- Institut für Pharmakologie und Toxikologie, TU München, München, Germany
| | - Ursula Seidler
- Department of Gastroenterology, Hepatology, Infectiology and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
2
|
Scott W, Wong IGY, Cramer J, Horton D, Basel D, Teng RJ, Muriello M, Elkadri A. Clinical course and therapeutic trial for a case of congenital secretory diarrhea due to novel GUCY2C variant. Am J Med Genet A 2024; 194:e63489. [PMID: 38058249 DOI: 10.1002/ajmg.a.63489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/24/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023]
Abstract
Chronic diarrhea presents a significant challenge for managing nutritional and electrolyte deficiencies, especially in children, given the higher stakes of impacting growth and developmental consequence. Congenital secretory diarrhea (CSD) compounds this further, particularly in the case of the activating variants of the guanylate-cyclase 2C (GUCY2C) gene. GUCY2C encodes for the guanylate-cyclase 2C (GC-C) receptor that activates the downstream cystic fibrosis transmembrane receptor (CFTR) that primarily drives the severity of diarrhea with an unclear extent of influence on other intestinal channels. Thus far, management for CSD primarily consists of mitigating nutritional, electrolyte, and volume deficiencies with no known pathophysiology-driven treatments. For activating variants of GUCY2C, experimental compounds have shown efficacy in vitro for direct inhibition of GC-C but are not currently available for clinical use. However, Crofelemer, a CFTR inhibitory modulator with negligible systemic absorption, can theoretically help to treat this type of CSD. Herein, we describe and characterize the clinical course of a premature male infant with a de novo missense variant of GUCY2C not previously reported and highly consistent with CSD. With multi-disciplinary family-directed decision-making, a treatment for CSD was evaluated for the first time to our knowledge with Crofelemer.
Collapse
Affiliation(s)
- William Scott
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ian Guo Yi Wong
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jesse Cramer
- School of Pharmacy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pharmacy, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - Darlene Horton
- Napo Pharmaceuticals, Inc, San Francisco, California, USA
| | - Donald Basel
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ru-Jeng Teng
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michael Muriello
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Abdul Elkadri
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
3
|
Lv H, Niu J, Pan W, Wang Y, Wang L, Wang M, Shi Y, Zhang G, Al Hamyari B, Wang S, Li X, Shi Y. Stool-softening effect and action mechanism of free anthraquinones extracted from Rheum palmatum L. on water deficit-induced constipation in rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117336. [PMID: 37907143 DOI: 10.1016/j.jep.2023.117336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 11/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In traditional Chinese herbal medicine, rhubarb is said to remove accumulation with purgation, clearing heat, and discharging fire. Modern pharmacology has shown that rhubarb extract has a purgative effect when given to experimental animals in an appropriate dose. However, the active components and their mechanism of action are still not clearly defined. AIM OF THE STUDY The current research aimed to evaluate the synergistic stool-softening effects and explore the action mechanism of rhubarb free anthraquinones (RhA) and their monomers on constipation in rats. MATERIALS AND METHODS A rat model of water deficit-induced constipation was established to induce constipation, and these rats were treated with RhA and its monomers. ELISA, histopathology, immunohistochemistry, qPCR and Western blotting based on network pharmacology and molecular docking were conducted to explore the possible mechanism of action of RhA and its monomers. RESULTS RhA, aloe-emodin, rhein, and chrysophanol showed stool-softening activity, and the combination of aloe-emodin and rhein had the strongest softening effect on faecal pellets. Aloe-emodin, rhein, and chrysophanol significantly increased the serum levels of vasoactive intestinal peptide (VIP), motilin (MTL), and substance P (SP), upregulated the expression of VIP, cyclase-associated protein 1 (CAP1), protein kinase A (PKA), cystic fibrosis transmembrane conductance regulator (CFTR), aquaporin 3 (AQP3), aquaporin 4 (AQP4), and aquaporin 8 (AQP8), decreased the expression of epithelial sodium channel (ENaC) and Na+/H+ exchanger 3 (NHE3), and reduced the colonic tissue concentration of Na+-K+-ATPase in the constipated rats. Osmolality of colonic fluid in model rats treated by RhA, aloe-emodin, rhein, and chrysophanol was increased. CONCLUSION Aloe-emodin, rhein, and chrysophanol were the stool-softening components of the RhA extract, and there were certain drug-interactions between the components. RhA upregulated VIP expression, activated the cyclic adenosine monophosphate protein kinase A (cAMP/PKA) pathway, and further stimulated CFTR expression while inhibiting NHE3 and ENaC expression, resulting in a hypertonic state in the colonic lumen. Water transport could then be driven by an osmotic gradient, which in turn led to the upregulation of AQP3, AQP4, and AQP8 expression. In addition, RhA likely improved gastrointestinal motility by increasing serum VIP, SP, and MTL concentrations, thus promoting faecal excretion.
Collapse
Affiliation(s)
- Huijuan Lv
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 730000, China.
| | - Jingjing Niu
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 730000, China.
| | - Wenhao Pan
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 730000, China.
| | - Yudong Wang
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 730000, China.
| | - Lifang Wang
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 730000, China.
| | - Meng Wang
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 730000, China.
| | - Yali Shi
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 730000, China.
| | - Guifang Zhang
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 730000, China.
| | - Bandar Al Hamyari
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 730000, China.
| | - Shaohua Wang
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 730000, China; Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou, 730000, China.
| | - Xuefeng Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Yanbin Shi
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 730000, China; Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
4
|
Mishra V, Sharma K, Bose A, Maisonneuve P, Visweswariah SS. The evolutionary divergence of receptor guanylyl cyclase C has implications for preclinical models for receptor-directed therapeutics. J Biol Chem 2024; 300:105505. [PMID: 38029963 PMCID: PMC7615481 DOI: 10.1016/j.jbc.2023.105505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023] Open
Abstract
Mutations in receptor guanylyl cyclase C (GC-C) cause severe gastrointestinal disease, including meconium ileus, early onset acute diarrhea, and pediatric inflammatory bowel disease that continues into adulthood. Agonists of GC-C are US Food and Drug Administration-approved drugs for the treatment of constipation and irritable bowel syndrome. Therapeutic strategies targeting GC-C are tested in preclinical mouse models, assuming that murine GC-C mimics human GC-C in its biochemical properties and downstream signaling events. Here, we reveal important differences in ligand-binding affinity and GC activity between mouse GC-C and human GC-C. We generated a series of chimeric constructs of various domains of human and mouse GC-C to show that the extracellular domain of mouse GC-C contributed to log-orders lower affinity of mouse GC-C for ligands than human GC-C. Further, the Vmax of the murine GC domain was lower than that of human GC-C, and allosteric regulation of the receptor by ATP binding to the intracellular kinase-homology domain also differed. These altered properties are reflected in the high concentrations of ligands required to elicit signaling responses in the mouse gut in preclinical models and the specificity of a GC inhibitor towards human GC-C. Therefore, our studies identify considerations in using the murine model to test molecules for therapeutic purposes that work as either agonists or antagonists of GC-C, and vaccines for the bacterial heat-stable enterotoxin that causes watery diarrhea in humans.
Collapse
Affiliation(s)
- Vishwas Mishra
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, India
| | - Kritica Sharma
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, India
| | - Avipsa Bose
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, India
| | - Pierre Maisonneuve
- UMR 5248 - Chemistry & Biology of Membranes and Nano-Objects, CNRS - Université de Bordeaux, Institut Européen de Chimie et Biologie, Pessac, France
| | - Sandhya S Visweswariah
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
5
|
Bajracharya S, Stich D, Berman J, Biank V. Congenital sodium diarrhoea caused by rare de novo activating guanylate cyclase mutation. BMJ Case Rep 2022; 15:e251632. [PMID: 36581358 PMCID: PMC9806026 DOI: 10.1136/bcr-2022-251632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 12/30/2022] Open
Abstract
A male infant with prenatal history significant for polyhydramnios requiring multiple amnioreductions with suspicion of small bowel atresia was born at 31 weeks 5 days' gestation with abdominal distension. He underwent three exploratory laparotomies and ileostomy for small bowel obstruction and was found to have fluid-filled intestinal dilatation. Serum and stool chemistries suggested sodium secretory diarrhoea. A rapid whole-exome sequencing confirmed de novo guanylate cyclase mutation variant as a cause for his congenital sodium secretory diarrhoea. He required large volume of fluid and electrolyte replacement along with total parenteral nutrition. Several medications to restore normal sodium homeostasis by targeting molecular mechanisms and pathogenesis described in previous literature failed to decrease stool output and electrolyte loss. He was discharged home at 11 months of age on total parenteral nutrition and weekly iron therapy.
Collapse
Affiliation(s)
| | - Duane Stich
- Neonatology, Advocate Children's Hospital, Park Ridge, Illinois, USA
| | - James Berman
- Pediatric Gastroenterology, Advocate Children's Hospital, Park Ridge, Illinois, USA
| | - Vincent Biank
- Pediatric Gastroenterology, NorthShore University HealthSystem, Evanston, Illinois, USA
| |
Collapse
|
6
|
Zhang Y, Tan P, Zhao Y, Ma X. Enterotoxigenic Escherichia coli: intestinal pathogenesis mechanisms and colonization resistance by gut microbiota. Gut Microbes 2022; 14:2055943. [PMID: 35358002 PMCID: PMC8973357 DOI: 10.1080/19490976.2022.2055943] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of diarrhea in children and travelers in developing countries. ETEC is characterized by the ability to produce major virulence factors including colonization factors (CFs) and enterotoxins, that bind to specific receptors on epithelial cells and induce diarrhea. The gut microbiota is a stable and sophisticated ecosystem that performs a range of beneficial functions for the host, including protection against pathogen colonization. Understanding the pathogenic mechanisms of ETEC and the interaction between the gut microbiota and ETEC represents not only a research need but also an opportunity and challenge to develop precautions for ETEC infection. Herein, this review focuses on recent discoveries about ETEC etiology, pathogenesis and clinical manifestation, and discusses the colonization resistances mediated by gut microbiota, as well as preventative strategies against ETEC with an aim to provide novel insights that can reduce the adverse effect on human health.
Collapse
Affiliation(s)
- Yucheng Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Peng Tan
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Ying Zhao
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China,CONTACT Xi Ma State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| |
Collapse
|
7
|
van Vugt AH, Bijvelds MJ, de Jonge HR, Meijsen KF, Restin T, Bryant MB, Ballauff A, Koot B, Müller T, Houwen RH, Janecke AR, Middendorp S. A Potential Treatment of Congenital Sodium Diarrhea in Patients With Activating GUCY2C Mutations. Clin Transl Gastroenterol 2021; 12:e00427. [PMID: 34797252 PMCID: PMC8604003 DOI: 10.14309/ctg.0000000000000427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Gain-of-function mutations in guanylyl cyclase C (GCC) result in persistent diarrhea with perinatal onset. We investigated a specific GCC inhibitor, SSP2518, for its potential to treat this disorder. METHODS We investigated the effect of SSP2518 on GCC-mediated intracellular cyclic guanosine monophosphate (cGMP) levels and on GCC-mediated chloride secretion in intestinal organoids from 3 patients with distinct activating GCC mutations and from controls, with and without stimulation of GCC with heat-stable enterotoxin. RESULTS Patient-derived organoids had significantly higher basal cGMP levels than control organoids, which were lowered by SSP2518 to levels found in control organoids. In addition, SSP2518 significantly reduced cGMP levels and chloride secretion in patient-derived and control organoids (P < 0.05 for all comparisons) after heat-stable enterotoxin stimulation. DISCUSSION We reported in this study that the GCC inhibitor SSP2518 normalizes cGMP levels in intestinal organoids derived from patients with GCC gain-of-function mutations and markedly reduces cystic fibrosis transmembrane conductance regulator-dependent chloride secretion, the driver of persistent diarrhea.
Collapse
Affiliation(s)
- Anke H.M. van Vugt
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht (UMCU), Utrecht University (UU), Utrecht, the Netherlands;
- Regenerative Medicine Center, UMCU, UU, Utrecht, the Netherlands;
| | - Marcel J.C. Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands;
| | - Hugo R. de Jonge
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands;
| | - Kelly F. Meijsen
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands;
| | - Tanja Restin
- Newborn Research Zürich, Department of Neonatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland;
- Institute of Physiology, University of Zurich, Zurich, Switzerland;
| | - Manuel B. Bryant
- Newborn Research Zürich, Department of Neonatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland;
| | - Antje Ballauff
- Kinder- und Jugendmedizin, Helios Klinikum, Krefeld, Germany;
| | - Bart Koot
- Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Pediatric Gastroenterology, Amsterdam, the Netherlands;
| | - Thomas Müller
- Department of Pediatrics I, Medical University Innsbruck, Innsbruck, Austria;
| | - Roderick H.J. Houwen
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht (UMCU), Utrecht University (UU), Utrecht, the Netherlands;
| | - Andreas R. Janecke
- Department of Pediatrics I, Medical University Innsbruck, Innsbruck, Austria;
- Division of Human Genetics, Medical University Innsbruck, Innsbruck, Austria
| | - Sabine Middendorp
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht (UMCU), Utrecht University (UU), Utrecht, the Netherlands;
- Regenerative Medicine Center, UMCU, UU, Utrecht, the Netherlands;
- Current affiliation: Gadeta BV, Utrecht, the Netherlands.
| |
Collapse
|
8
|
In Silico Prediction, Molecular Docking and Dynamics Studies of Steroidal Alkaloids of Holarrhena pubescens Wall. ex G. Don to Guanylyl Cyclase C: Implications in Designing of Novel Antidiarrheal Therapeutic Strategies. Molecules 2021; 26:molecules26144147. [PMID: 34299422 PMCID: PMC8305770 DOI: 10.3390/molecules26144147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 01/08/2023] Open
Abstract
The binding of heat stable enterotoxin (STa) secreted by enterotoxigenic Escherichia coli (ETEC) to the extracellular domain of guanylyl cyclase c (ECDGC-C) causes activation of a signaling cascade, which ultimately results in watery diarrhea. We carried out this study with the objective of finding ligands that would interfere with the binding of STa on ECDGC-C. With this view in mind, we tested the biological activity of a alkaloid rich fraction of Holarrhena pubescens against ETEC under in vitro conditions. Since this fraction showed significant antibacterial activity against ETEC, we decided to test the screen binding affinity of nine compounds of steroidal alkaloid type from Holarrhena pubescens against extracellular domain (ECD) by molecular docking and identified three compounds with significant binding energy. Molecular dynamics simulations were performed for all the three lead compounds to establish the stability of their interaction with the target protein. Pharmacokinetics and toxicity profiling of these leads demonstrated that they possessed good drug-like properties. Furthermore, the ability of these leads to inhibit the binding of STa to ECD was evaluated. This was first done by identifying amino acid residues of ECDGC-C binding to STa by protein-protein docking. The results were matched with our molecular docking results. We report here that holadysenterine, one of the lead compounds that showed a strong affinity for the amino acid residues on ECDGC-C, also binds to STa. This suggests that holadysenterine has the potential to inhibit binding of STa on ECD and can be considered for future study, involving its validation through in vitro assays and animal model studies.
Collapse
|
9
|
|
10
|
de Jonge HR, Ardelean MC, Bijvelds MJC, Vergani P. Strategies for cystic fibrosis transmembrane conductance regulator inhibition: from molecular mechanisms to treatment for secretory diarrhoeas. FEBS Lett 2020; 594:4085-4108. [PMID: 33113586 PMCID: PMC7756540 DOI: 10.1002/1873-3468.13971] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/22/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) is an unusual ABC transporter. It acts as an anion‐selective channel that drives osmotic fluid transport across many epithelia. In the gut, CFTR is crucial for maintaining fluid and acid‐base homeostasis, and its activity is tightly controlled by multiple neuro‐endocrine factors. However, microbial toxins can disrupt this intricate control mechanism and trigger protracted activation of CFTR. This results in the massive faecal water loss, metabolic acidosis and dehydration that characterize secretory diarrhoeas, a major cause of malnutrition and death of children under 5 years of age. Compounds that inhibit CFTR could improve emergency treatment of diarrhoeal disease. Drawing on recent structural and functional insight, we discuss how existing CFTR inhibitors function at the molecular and cellular level. We compare their mechanisms of action to those of inhibitors of related ABC transporters, revealing some unexpected features of drug action on CFTR. Although challenges remain, especially relating to the practical effectiveness of currently available CFTR inhibitors, we discuss how recent technological advances might help develop therapies to better address this important global health need.
Collapse
Affiliation(s)
- Hugo R. de Jonge
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Maria C. Ardelean
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonUK
- Department of Natural SciencesUniversity College LondonUK
| | - Marcel J. C. Bijvelds
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Paola Vergani
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonUK
| |
Collapse
|
11
|
Rappaport JA, Waldman SA. An update on guanylyl cyclase C in the diagnosis, chemoprevention, and treatment of colorectal cancer. Expert Rev Clin Pharmacol 2020; 13:1125-1137. [PMID: 32945718 DOI: 10.1080/17512433.2020.1826304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Colorectal cancer remains the second leading cause of cancer death in the United States, underscoring the need for novel therapies. Despite the successes of new targeted agents for other cancers, colorectal cancer suffers from a relative scarcity of actionable biomarkers. In this context, the intestinal receptor, guanylyl cyclase C (GUCY2C), has emerged as a promising target.Areas covered: GUCY2C regulates a tumor-suppressive signaling axis that is silenced through loss of its endogenous ligands at the earliest stages of tumorigenesis. A body of literature supports a cancer chemoprevention strategy involving reactivation of GUCY2C through FDA-approved cGMP-elevating agents such as linaclotide, plecanatide, and sildenafil. Its limited expression in extra-intestinal tissues, and retention on the surface of cancer cells, also positions GUCY2C as a target for immunotherapies to treat metastatic disease, including vaccines, chimeric antigen receptor T-cells, and antibody-drug conjugates. Likewise, GUCY2C mRNA identifies metastatic cells, enhancing colorectal cancer detection, and staging. Pre-clinical and clinical programs exploring these GUCY2C-targeting strategies will be reviewed.Expert opinion: Recent mechanistic insights characterizing GUCY2C ligand loss early in tumorigenesis, coupled with results from the first clinical trials testing GUCY2C-targeting strategies, continue to elevate GUCY2C as an ideal target for prevention, detection, and therapy.
Collapse
Affiliation(s)
- Jeffrey A Rappaport
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University , Philadelphia, PA, USA
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University , Philadelphia, PA, USA
| |
Collapse
|
12
|
Chen T, Lin R, Avula L, Sarker R, Yang J, Cha B, Tse CM, McNamara G, Seidler U, Waldman S, Snook A, Bijvelds MJC, de Jonge HR, Li X, Donowitz M. NHERF3 is necessary for Escherichia coli heat-stable enterotoxin-induced inhibition of NHE3: differences in signaling in mouse small intestine and Caco-2 cells. Am J Physiol Cell Physiol 2019; 317:C737-C748. [PMID: 31365292 DOI: 10.1152/ajpcell.00351.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a leading cause of childhood death from diarrhea and the leading cause of Traveler's diarrhea. E. coli heat-stable enterotoxin (ST) is a major virulence factor of ETEC and inhibits the brush border Na/H exchanger NHE3 in producing diarrhea. NHE3 regulation involves multiprotein signaling complexes that form on its COOH terminus. In this study, the hypothesis was tested that ST signals via members of the Na/H exchanger regulatory factor (NHERF) family of scaffolding proteins, NHERF2, which had been previously shown to have a role, and now with concentration on a role for NHERF3. Two models were used: mouse small intestine and Caco-2/BBe cells. In both models, ST rapidly increased intracellular cGMP, inhibited NHE3 activity, and caused a quantitatively similar decrease in apical expression of NHE3. The transport effects were NHERF3 and NHERF2 dependent. Also, mutation of the COOH-terminal amino acids of NHERF3 supported that NHERF3-NHERF2 heterodimerization was likely to account for this dual dependence. The ST increase in cGMP in both models was partially dependent on NHERF3. The intracellular signaling pathways by which ST-cGMP inhibits NHE3 were different in mouse jejunum (activation of cGMP kinase II, cGKII) and Caco-2 cells, which do not express cGKII (elevation of intracellular Ca2+ concentration [Ca2+]i). The ST elevation of [Ca2+]i was from intracellular stores and was dependent on NHERF3-NHERF2. This study shows that intracellular signaling in the same diarrheal model in multiple cell types may be different; this has implications for therapeutic strategies, which often assume that models have similar signaling mechanisms.
Collapse
Affiliation(s)
- Tiane Chen
- Departments of Physiology and Medicine, Gastroenterology Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ruxian Lin
- Departments of Physiology and Medicine, Gastroenterology Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Leela Avula
- Departments of Physiology and Medicine, Gastroenterology Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rafiquel Sarker
- Departments of Physiology and Medicine, Gastroenterology Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jianbo Yang
- Departments of Physiology and Medicine, Gastroenterology Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Boyoung Cha
- Departments of Physiology and Medicine, Gastroenterology Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chung Ming Tse
- Departments of Physiology and Medicine, Gastroenterology Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - George McNamara
- Departments of Physiology and Medicine, Gastroenterology Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ursula Seidler
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Scott Waldman
- Division of Clinical Pharmacology, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Adam Snook
- Division of Clinical Pharmacology, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Marcel J C Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Hugo R de Jonge
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Xuhang Li
- Departments of Physiology and Medicine, Gastroenterology Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mark Donowitz
- Departments of Physiology and Medicine, Gastroenterology Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
13
|
Jiang X, Xia S, He X, Ma H, Feng Y, Liu Z, Wang W, Tian M, Chen H, Peng F, Wang L, Zhao P, Ge J, Liu D. Targeting peptide‐enhanced antibody and CD11c+dendritic cells to inclusion bodies expressing protective antigen against ETEC in mice. FASEB J 2018; 33:2836-2847. [DOI: 10.1096/fj.201800289rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Xinpeng Jiang
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
- Postdoctoral WorkstationHeilongjiang Academy of Agricultural SciencesHarbinChina
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - Shuang Xia
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - Xinmiao He
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Hong Ma
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Yanzhong Feng
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Ziguang Liu
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Wentao Wang
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Ming Tian
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
- Postdoctoral WorkstationHeilongjiang Academy of Agricultural SciencesHarbinChina
| | - Heshu Chen
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Fugang Peng
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Liang Wang
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Peng Zhao
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - Junwei Ge
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - Di Liu
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| |
Collapse
|
14
|
Waldman SA, Camilleri M. Guanylate cyclase-C as a therapeutic target in gastrointestinal disorders. Gut 2018; 67:1543-1552. [PMID: 29563144 PMCID: PMC6204952 DOI: 10.1136/gutjnl-2018-316029] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/19/2018] [Accepted: 02/26/2018] [Indexed: 12/16/2022]
Abstract
Functional gastrointestinal disorders (FGIDs) and IBDs are two of the most prevalent disorders of the GI tract and consume a significant proportion of healthcare resources. Recent studies have shown that membrane-bound guanylate cyclase-C (GC-C) receptors lining the GI tract may serve as novel therapeutic targets in the treatment of FGIDs and IBDs. GC-C receptor activation by its endogenous paracrine hormones uroguanylin and guanylin, and the resulting intracellular production of its downstream effector cyclic GMP, occurs in a pH-dependent manner and modulates key physiological functions. These include fluid and electrolyte homeostasis, maintenance of the intestinal barrier, anti-inflammatory activity and regulation of epithelial regeneration. Studies of the GC-C paracrine signalling axis have revealed the therapeutic potential of these receptors in treating GI disorders, including chronic idiopathic constipation and irritable bowel syndrome-constipation. This review focuses on the evolving understanding of GC-C function in health and disease, and strategies for translating these principles into new treatments for FGIDs and IBDs.
Collapse
Affiliation(s)
- Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Michael Camilleri
- Clinical Enteric Neurosciences Translational and Epidemiological Research (CENTER), Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
15
|
Bijvelds MJC, Tresadern G, Hellemans A, Smans K, Nieuwenhuijze NDA, Meijsen KF, Bongartz JP, Ver Donck L, de Jonge HR, Schuurkes JAJ, De Maeyer JH. Selective inhibition of intestinal guanosine 3',5'-cyclic monophosphate signaling by small-molecule protein kinase inhibitors. J Biol Chem 2018; 293:8173-8181. [PMID: 29653944 PMCID: PMC5971447 DOI: 10.1074/jbc.ra118.002835] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/11/2018] [Indexed: 11/06/2022] Open
Abstract
The guanosine 3',5'-cyclic monophosphate (cGMP)-dependent protein kinase II (cGKII) serine/threonine kinase relays signaling through guanylyl cyclase C (GCC) to control intestinal fluid homeostasis. Here, we report the discovery of small-molecule inhibitors of cGKII. These inhibitors were imidazole-aminopyrimidines, which blocked recombinant human cGKII at submicromolar concentrations but exhibited comparatively little activity toward the phylogenetically related protein kinases cGKI and cAMP-dependent protein kinase (PKA). Whereas aminopyrimidyl motifs are common in protein kinase inhibitors, molecular modeling of these imidazole-aminopyrimidines in the ATP-binding pocket of cGKII indicated an unconventional binding mode that directs their amine substituent into a narrow pocket delineated by hydrophobic residues of the hinge and the αC-helix. Crucially, this set of residues included the Leu-530 gatekeeper, which is not conserved in cGKI and PKA. In intestinal organoids, these compounds blocked cGKII-dependent phosphorylation of the vasodilator-stimulated phosphoprotein (VASP). In mouse small intestinal tissue, cGKII inhibition significantly attenuated the anion secretory response provoked by the GCC-activating bacterial heat-stable toxin (STa), a frequent cause of infectious secretory diarrhea. In contrast, both PKA-dependent VASP phosphorylation and intestinal anion secretion were unaffected by treatment with these compounds, whereas experiments with T84 cells indicated that they weakly inhibit the activity of cAMP-hydrolyzing phosphodiesterases. As these protein kinase inhibitors are the first to display selective inhibition of cGKII, they may expedite research on cGMP signaling and may aid future development of therapeutics for managing diarrheal disease and other pathogenic syndromes that involve cGKII.
Collapse
Affiliation(s)
- Marcel J C Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, P. O. Box 2040, 3000CA Rotterdam, The Netherlands.
| | - Gary Tresadern
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Ann Hellemans
- Shire-Movetis NV, Veedijk 58, B-2300 Turnhout, Belgium
| | - Karine Smans
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Natascha D A Nieuwenhuijze
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, P. O. Box 2040, 3000CA Rotterdam, The Netherlands
| | - Kelly F Meijsen
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, P. O. Box 2040, 3000CA Rotterdam, The Netherlands
| | - Jean-Pierre Bongartz
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Luc Ver Donck
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Hugo R de Jonge
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, P. O. Box 2040, 3000CA Rotterdam, The Netherlands
| | | | | |
Collapse
|
16
|
Arora K, Huang Y, Mun K, Yarlagadda S, Sundaram N, Kessler MM, Hannig G, Kurtz CB, Silos-Santiago I, Helmrath M, Palermo JJ, Clancy JP, Steinbrecher KA, Naren AP. Guanylate cyclase 2C agonism corrects CFTR mutants. JCI Insight 2017; 2:93686. [PMID: 28978796 DOI: 10.1172/jci.insight.93686] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/29/2017] [Indexed: 01/06/2023] Open
Abstract
Cystic fibrosis (CF) is a genetic disorder in which epithelium-generated fluid flow from the lung, intestine, and pancreas is impaired due to mutations disrupting CF transmembrane conductance regulator (CFTR) channel function. CF manifestations of the pancreas and lung are present in the vast majority of CF patients, and 15% of CF infants are born with obstructed gut or meconium ileus. However, constipation is a significantly underreported outcome of CF disease, affecting 47% of the CF patients, and management becomes critical in the wake of increasing life span of CF patients. In this study, we unraveled a potentially novel molecular role of a membrane-bound cyclic guanosine monophosphate-synthesizing (cGMP-synthesizing) intestinal enzyme, guanylate cyclase 2C (GCC) that could be targeted to ameliorate CF-associated intestinal fluid deficit. We demonstrated that GCC agonism results in functional rescue of murine F508del/F508del and R117H/R117H Cftr and CFTR mutants in CF patient-derived intestinal spheres. GCC coexpression and activation facilitated processing and ER exit of F508del CFTR and presented a potentially novel rescue modality in the intestine, similar to the CF corrector VX-809. Our findings identify GCC as a biological CFTR corrector and potentiator in the intestine.
Collapse
Affiliation(s)
- Kavisha Arora
- Division of Pulmonary Medicine, Department of Pediatrics, and
| | - Yunjie Huang
- Division of Pulmonary Medicine, Department of Pediatrics, and
| | - Kyushik Mun
- Division of Pulmonary Medicine, Department of Pediatrics, and
| | | | - Nambirajan Sundaram
- Department of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | | | | | | | | | - Michael Helmrath
- Department of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Joseph J Palermo
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, and
| | - John P Clancy
- Division of Pulmonary Medicine, Department of Pediatrics, and
| | - Kris A Steinbrecher
- Division of Gastroenterology, Hepatology and Nutrition, CCHMC, Cincinnati, Ohio, USA
| | | |
Collapse
|
17
|
Guanylate Cyclase C Activation Shapes the Intestinal Microbiota in Patients with Familial Diarrhea and Increased Susceptibility for Crohn's Disease. Inflamm Bowel Dis 2017; 23:1752-1761. [PMID: 28902124 DOI: 10.1097/mib.0000000000001264] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND With 25% prevalence of Crohn's disease, Familial GUCY2C diarrhea syndrome (FGDS) is a monogenic disorder potentially suited to study initiating factors in inflammatory bowel disease (IBD). We aimed to characterize the impact of an activating GUCY2C mutation on the gut microbiota in patients with FGDS controlling for Crohn's disease status and to determine whether changes share features with those observed in unrelated patients with IBD. METHODS Bacterial DNA from fecal samples collected from patients with FGDS (N = 20), healthy relatives (N = 11), unrelated healthy individuals (N = 263), and IBD controls (N = 46) was subjected to sequencing of the V3-V4 region of the 16S rRNA gene to determine gut microbiota composition. Food frequency questionnaires were obtained from patients with FGDS and their relatives. RESULTS Compared with healthy controls, FGDS displayed prominent changes in many microbial lineages including increase in Enterobacteriaceae, loss of Bifidobacterium and Faecalibacterium prausnitzii but an unchanged intraindividual (alpha) diversity. The depletion of F. prausnitzii is in line with what is typically observed in Crohn's disease. There was no significant difference in the dietary profile between the patients and related controls. The gut microbiota in related and unrelated healthy controls was also similar, suggesting that diet and familial factors do not explain the gut microbiota alterations in FGDS. CONCLUSIONS The findings support that the activating mutation in GUCY2C creates an intestinal environment with a major influence on the microbiota, which could contribute to the increased susceptibility to IBD in patients with FGDS.
Collapse
|
18
|
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: 2.4] [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
|
19
|
Zomer-van Ommen DD, Pukin AV, Fu O, Quarles van Ufford LH, Janssens HM, Beekman JM, Pieters RJ. Functional Characterization of Cholera Toxin Inhibitors Using Human Intestinal Organoids. J Med Chem 2016; 59:6968-72. [DOI: 10.1021/acs.jmedchem.6b00770] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Domenique D. Zomer-van Ommen
- Department
of Pediatric Pulmonology, University Medical Centre Utrecht, Lundlaan
6, 3508 GA Utrecht, The Netherlands
| | - Aliaksei V. Pukin
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Ou Fu
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Linda H.C. Quarles van Ufford
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Hettie M. Janssens
- Department
of Pediatric Pulmonology, Erasmus Medical Center/Sophia Children’s Hospital, Wytemaweg 80, 3015
CN Rotterdam, The Netherlands
| | - Jeffrey M. Beekman
- Department
of Pediatric Pulmonology, University Medical Centre Utrecht, Lundlaan
6, 3508 GA Utrecht, The Netherlands
| | - Roland J. Pieters
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
20
|
Beltrán AR, Carraro-Lacroix LR, Bezerra CNA, Cornejo M, Norambuena K, Toledo F, Araos J, Pardo F, Leiva A, Sanhueza C, Malnic G, Sobrevia L, Ramírez MA. Escherichia coli Heat-Stable Enterotoxin Mediates Na+/H+ Exchanger 4 Inhibition Involving cAMP in T84 Human Intestinal Epithelial Cells. PLoS One 2015; 10:e0146042. [PMID: 26713849 PMCID: PMC4699896 DOI: 10.1371/journal.pone.0146042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/11/2015] [Indexed: 01/05/2023] Open
Abstract
The enterotoxigenic Escherichia coli strains lead to diarrhoea in humans due to heat-labile and heat-stable (STa) enterotoxins. STa increases Cl-release in intestinal cells, including the human colonic carcinoma T84 cell line, involving increased cGMP and membrane alkalization due to reduced Na+/H+ exchangers (NHEs) activity. Since NHEs modulate intracellular pH (pHi), and NHE1, NHE2, and NHE4 are expressed in T84 cells, we characterized the STa role as modulator of these exchangers. pHi was assayed by the NH4Cl pulse technique and measured by fluorescence microscopy in BCECF-preloaded cells. pHi recovery rate (dpHi/dt) was determined in the absence or presence of 0.25 μmol/L STa (30 minutes), 25 μmol/L HOE-694 (concentration inhibiting NHE1 and NHE2), 500 μmol/L sodium nitroprusside (SNP, spontaneous nitric oxide donor), 100 μmol/L dibutyryl cyclic GMP (db-cGMP), 100 nmol/L H89 (protein kinase A inhibitor), or 10 μmol/L forskolin (adenylyl cyclase activator). cGMP and cAMP were measured in cell extracts by radioimmunoassay, and buffering capacity (ßi) and H+ efflux (JH+) was determined. NHE4 protein abundance was determined by western blotting. STa and HOE-694 caused comparable reduction in dpHi/dt and JH+ (~63%), without altering basal pHi (range 7.144-7.172). STa did not alter ßi value in a range of 1.6 pHi units. The dpHi/dt and JH+ was almost abolished (~94% inhibition) by STa + HOE-694. STa effect was unaltered by db-cGMP or SNP. However, STa and forskolin increased cAMP level. STa-decreased dpHi/dt and JH+ was mimicked by forskolin, and STa + HOE-694 effect was abolished by H89. Thus, incubation of T84 cells with STa results in reduced NHE4 activity leading to a lower capacity of pHi recovery requiring cAMP, but not cGMP. STa effect results in a causal phenomenon (STa/increased cAMP/increased PKA activity/reduced NHE4 activity) ending with intracellular acidification that could have consequences in the gastrointestinal cells function promoting human diarrhoea.
Collapse
Affiliation(s)
- Ana R. Beltrán
- Cellular Physiology Laboratory, Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Antofagasta 1270300, Chile
- Department of Education, Faculty of Education, Universidad de Antofagasta, Antofagasta 1270300, Chile
| | - Luciene R. Carraro-Lacroix
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 3550308–1009, Brazil
| | - Camila N. A. Bezerra
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 3550308–1009, Brazil
| | - Marcelo Cornejo
- Cellular Physiology Laboratory, Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Antofagasta 1270300, Chile
| | - Katrina Norambuena
- Cellular Physiology Laboratory, Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Antofagasta 1270300, Chile
| | - Fernando Toledo
- Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán 3780000, Chile
| | - Joaquín Araos
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Fabián Pardo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Andrea Leiva
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Carlos Sanhueza
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Gerhard Malnic
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 3550308–1009, Brazil
| | - Luis Sobrevia
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Queensland, Australia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
- * E-mail: (MAR); (LS)
| | - Marco A. Ramírez
- Cellular Physiology Laboratory, Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Antofagasta 1270300, Chile
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
- * E-mail: (MAR); (LS)
| |
Collapse
|