1
|
Singh P, Sayuk GS, Rosenbaum DP, Edelstein S, Kozuka K, Chang L. An Overview of the Effects of Tenapanor on Visceral Hypersensitivity in the Treatment of Irritable Bowel Syndrome with Constipation. Clin Exp Gastroenterol 2024; 17:87-96. [PMID: 38617992 PMCID: PMC11016248 DOI: 10.2147/ceg.s454526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 03/22/2024] [Indexed: 04/16/2024] Open
Abstract
Background Patients with irritable bowel syndrome with constipation (IBS-C) experience persistent abdominal pain, a common symptom leading to greater healthcare utilization and reports of treatment non-response. Clinically significant improvements in abdominal pain were observed in clinical trials of tenapanor, a first-in-class inhibitor of sodium/hydrogen exchanger isoform 3 (NHE3), for the treatment of IBS-C in adults. Aim This narrative review reports the current knowledge about visceral hypersensitivity as a mechanism for abdominal pain in patients with IBS-C and explores the published evidence for hypothesized mechanisms by which tenapanor may reduce visceral hypersensitivity leading to the observed clinical response of decreased abdominal pain. Findings Abdominal pain is experienced through activation and signaling of nociceptive dorsal root ganglia that innervate the gut. These sensory afferent neurons may become hypersensitized through signaling of transient receptor potential cation channel subfamily V member 1 (TRPV1), resulting in reduced action potential thresholds. TRPV1 signaling is also a key component of the proinflammatory cascade involving mast cell responses to macromolecule exposure following permeation through the intestinal epithelium. Indirect evidence of this pathway is supported by observations of higher pain in association with increased intestinal permeability in patients with IBS. Tenapanor reduces intestinal sodium absorption, leading to increased water retention in the intestinal lumen, thereby improving gastrointestinal motility. In animal models of visceral hypersensitivity, tenapanor normalized visceromotor responses and normalized TRPV1-mediated nociceptive signaling. Conclusion By improving gastrointestinal motility, decreasing intestinal permeability and inflammation, and normalizing nociception through decreased TRPV1 signaling, tenapanor may reduce visceral hypersensitivity, leading to less abdominal pain in patients with IBS-C. Therapies that have demonstrated effects on visceral hypersensitivity may be the future direction for meaningful abdominal pain relief for patients with IBS-C.
Collapse
Affiliation(s)
- Prashant Singh
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Gregory S Sayuk
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | - Lin Chang
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| |
Collapse
|
2
|
Mullens W, Damman K, Dhont S, Banerjee D, Bayes-Genis A, Cannata A, Chioncel O, Cikes M, Ezekowitz J, Flammer AJ, Martens P, Mebazaa A, Mentz RJ, Miró Ò, Moura B, Nunez J, Ter Maaten JM, Testani J, van Kimmenade R, Verbrugge FH, Metra M, Rosano GMC, Filippatos G. Dietary sodium and fluid intake in heart failure. A clinical consensus statement of the Heart Failure Association of the ESC. Eur J Heart Fail 2024; 26:730-741. [PMID: 38606657 DOI: 10.1002/ejhf.3244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
Sodium and fluid restriction has traditionally been advocated in patients with heart failure (HF) due to their sodium and water avid state. However, most evidence regarding the altered sodium handling, fluid homeostasis and congestion-related signs and symptoms in patients with HF originates from untreated patient cohorts and physiological investigations. Recent data challenge the beneficial role of dietary sodium and fluid restriction in HF. Consequently, the European Society of Cardiology HF guidelines have gradually downgraded these recommendations over time, now advising for the limitation of salt intake to no more than 5 g/day in patients with HF, while contemplating fluid restriction of 1.5-2 L/day only in selected patients. Therefore, the objective of this clinical consensus statement is to provide advice on fluid and sodium intake in patients with acute and chronic HF, based on contemporary evidence and expert opinion.
Collapse
Affiliation(s)
- Wilfried Mullens
- Department of Cardiology, Ziekenhuis Oost-Limburg A.V, Genk, Belgium
- Hasselt University, Hasselt, Belgium
| | - Kevin Damman
- University of Groningen, Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Sebastiaan Dhont
- Department of Cardiology, Ziekenhuis Oost-Limburg A.V, Genk, Belgium
- Hasselt University, Hasselt, Belgium
| | - Debasish Banerjee
- Renal and Transplantation Unit, St George's University Hospitals National Health Service Foundation Trust, London, UK
| | - Antoni Bayes-Genis
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, CIBERCV, Barcelona, Spain
| | - Antonio Cannata
- School of Cardiovascular Medicine and Sciences, King's College London, London, UK
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases, University of Medicine Carol Davila, Bucharest, Romania
| | - Maja Cikes
- Department of Cardiovascular Diseases, University of Zagreb School of Medicine & University Hospital Center Zagreb, Zagreb, Croatia
| | - Justin Ezekowitz
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Canadian VIGOUR Centre, University of Alberta, Edmonton, AB, Canada
| | - Andreas J Flammer
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Pieter Martens
- Department of Cardiology, Ziekenhuis Oost-Limburg A.V, Genk, Belgium
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | | - Òscar Miró
- Department of Emergency, Hospital Clínic, 'Processes and Pathologies, Emergencies Research Group' IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Brenda Moura
- Hospital das Forças Armadas and Cintesis - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Julio Nunez
- Cardiology Department and Heart Failure Unit, Hospital Clínico Universitario de Valencia, University of Valencia, INCLIVA, Valencia, Spain
| | - Jozine M Ter Maaten
- University of Groningen, Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Jeffrey Testani
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Roland van Kimmenade
- Department of Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frederik H Verbrugge
- Centre for Cardiovascular Diseases, University Hospital Brussels, Jette, Belgium
- Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Jette, Belgium
| | - Marco Metra
- Cardiology, ASST Spedali Civili, and Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Giuseppe M C Rosano
- Cardiology Clinical Academic Group, Molecular and Clinical Research Institute, St Georges University of London, London, UK
- Cardiology, San Raffaele Cassino, Rome, Italy
| | | |
Collapse
|
3
|
ZHANG QY, GUO Y, JIANG XL, LIU X, ZHAO SG, ZHOU XL, YANG ZW. Intestinal Cckbr-specific knockout mouse as a novel model of salt-sensitive hypertension via sodium over-absorption. J Geriatr Cardiol 2023; 20:538-547. [PMID: 37576480 PMCID: PMC10412539 DOI: 10.26599/1671-5411.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023] Open
Abstract
OBJECTIVES To investigate the value of CCKBRfl/fl villin-Cre mice as a mouse model of salt-sensitive hypertension (SSH). METHODS In the first part, 2-month-old CCKBRfl/fl villin-Cre mice (CKO) and control CCKBRfl/fl mice (WT) were fed with normal diet (0.4% NaCl) or high salt diet (4% NaCl), separately for 6 weeks. In the rescue study, one week of hydrochlorothiazide or saline injection were treated with the CKO mice fed high salt diet. The blood pressure, biochemical indexes, and the expression of small intestinal sodium transporters (NHE3, NKCC1, eNaC) was detected. The organ injury markers (MMP2/MMP9) and the histopathological changes of kidneys were observed, whereas the changes of duodenal sodium absorption were detected by small intestinal perfusion in vivo. RESULTS The CCKBRfl/fl villin-Cre mice with high salt intake exhibited high blood pressure, increased duodenal sodium absorption and urinary sodium excretion, and with renal injury. The protein expression of NHE3, NKCC1 and eNaC were also significant increase in the intestine of CKO-HS mice. Treatment with hydrochlorothiazide remarkably attenuated the elevated blood pressure by high salt absorption in the CCKBRfl/fl villin-Cre mice, but no significant histopathological changes were observed. CONCLUSIONS These results support a crucial role of intestinal Cckbr deficiency on SSH development and the diuretic antihypertension effect in CCKBRfl/fl villin-Cre mice. The CCKBRfl/fl villin-Cre mice with the high salt intake may serve as a stable model of salt-sensitive hypertensive induced by sodium overloading.
Collapse
Affiliation(s)
- Qiong-Yu ZHANG
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan GUO
- Emergency Department, Taihe County People’s Hospital, Taihe County, Anhui Province, China
| | - Xiao-Liang JIANG
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical Collage (PUMC), Beijing, China
| | - Xing LIU
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical Collage (PUMC), Beijing, China
| | - Shu-Guang ZHAO
- Emergency Department, Taihe County People’s Hospital, Taihe County, Anhui Province, China
| | - Xian-Liang ZHOU
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Wei YANG
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical Collage (PUMC), Beijing, China
| |
Collapse
|
4
|
Yi H, Wang Z, Yang B, Yang X, Gao K, Xiong Y, Wu Q, Qiu Y, Hu S, Wang L, Jiang Z. Effects of zinc oxide and condensed tannins on the growth performance and intestinal health of weaned piglets in ETEC-challenged environment. Front Microbiol 2023; 14:1181519. [PMID: 37180229 PMCID: PMC10172512 DOI: 10.3389/fmicb.2023.1181519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023] Open
Abstract
This experiment was conducted to evaluate effects of zine oxide (ZnO) and condensed tannins (CT), independently or in combination, on the growth performance and intestinal health of weaned piglets in enterotoxigenic Escherichia coli (ETEC-K88)-challenged environment. Randomly divided 72 weaned piglets into 4 groups. Dietary treatments included the following: basic diet group (CON), 1,500 mg/kg zinc oxide group (ZnO), 1,000 mg/kg condensed tannins group (CT), and 1,500 mg/kg zinc oxide +1,000 mg/kg condensed tannins group (ZnO + CT). Dietary ZnO supplementation decreased diarrhea rate from 0 to 14 days, 15 to 28 days, and 0 to 28 days (p < 0.05) and no significant on growth performance. The effect of CT on reducing diarrhea rate and diarrhea index was similar to the results of ZnO. Compared with the CON group, ZnO increased the ileum villus height and improved intestinal barrier function by increasing the content of mucin 2 (MUC-2) in jejunum and ileum mucosa and the mRNA expression of zonula occludens-1 (ZO-1) in jejunum (p < 0.05) and the expression of Occludin in duodenum and ileum (p < 0.05). The effects of CT on intestinal barrier function genes were similar to that of ZnO. Moreover, the mRNA expression of cystic fibrosis transmembrane conductance regulator (CFTR) in jejunum and ileum was reduced in ZnO group (p < 0.05). And CT was also capable of alleviating diarrhea by decreasing CFTR expression and promote water reabsorption by increasing AQP3 expression (p < 0.05). In addition, pigs receiving ZnO diet had higher abundance of phylum Bacteroidetes, and genera Prevotella, and lower phylum Firmicutes and genera Lactobacillus in colonic contents. These results indicated that ZnO and CT can alleviate diarrhea and improve intestinal barrier function of weaned pigs in ETEC-challenged environment. In addition, the application of ZnO combined with CT did not show synergistic effects on piglet intestinal health and overall performance. This study provides a theoretical basis for the application of ZnO in weaning piglet production practices, we also explored effects of CT on the growth performance and intestinal health of weaned piglets in ETEC-challenged environment.
Collapse
|
5
|
Peritore-Galve FC, Kaji I, Smith A, Walker LM, Shupe JA, Washington MK, Algood HMS, Dudeja PK, Goldenring JR, Lacy DB. Increased intestinal permeability and downregulation of absorptive ion transporters Nhe3, Dra, and Sglt1 contribute to diarrhea during Clostridioides difficile infection. Gut Microbes 2023; 15:2225841. [PMID: 37350393 PMCID: PMC10291935 DOI: 10.1080/19490976.2023.2225841] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 06/09/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND & AIM Clostridioides difficile infection (CDI) is the leading cause of hospital-acquired diarrhea and pseudomembranous colitis. Two protein toxins, TcdA and TcdB, produced by C. difficile are the major determinants of disease. However, the pathophysiological causes of diarrhea during CDI are not well understood. Here, we investigated the effects of C. difficile toxins on paracellular permeability and apical ion transporters in the context of an acute physiological infection. METHODS We studied intestinal permeability and apical membrane transporters in female C57BL/6J mice. Üssing chambers were used to measure paracellular permeability and ion transporter function across the intestinal tract. Infected intestinal tissues were analyzed by immunofluorescence microscopy and RNA-sequencing to uncover mechanisms of transporter dysregulation. RESULTS Intestinal permeability was increased through the size-selective leak pathway in vivo during acute CDI in a 2-day-post infection model. Chloride secretory activity was reduced in the cecum and distal colon during infection by decreased CaCC and CFTR function, respectively. SGLT1 activity was significantly reduced in the cecum and colon, accompanied by ablated SGLT1 expression in colonocytes and increased luminal glucose concentrations. SGLT1 and DRA expression was ablated by either TcdA or TcdB during acute infection, but NHE3 was decreased in a TcdB-dependent manner. The localization of key proteins that link filamentous actin to the ion transporters in the apical plasma membrane was unchanged. However, Sglt1, Nhe3, and Dra were drastically reduced at the transcript level, implicating downregulation of ion transporters in the mechanism of diarrhea during CDI. CONCLUSIONS CDI increases intestinal permeability and decreases apical abundance of NHE3, SGLT1, and DRA. This combination likely leads to dysfunctional water and solute absorption in the large bowel, causing osmotic diarrhea. These findings provide insights into the pathophysiological mechanisms underlying diarrhea and may open novel avenues for attenuating CDI-associated diarrhea.
Collapse
Affiliation(s)
- F. Christopher Peritore-Galve
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Anna Smith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lauren M. Walker
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John A. Shupe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M. Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Holly M. Scott Algood
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Pradeep K. Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Veterans Affairs, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - James R. Goldenring
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
- Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - D. Borden Lacy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
6
|
Nylander O, Sjöblom M, Sedin J, Dahlgren D. Effects of α2-adrenoceptor stimulation on luminal alkalinisation and net fluid flux in rat duodenum. PLoS One 2022; 17:e0273208. [PMID: 36006975 PMCID: PMC9409570 DOI: 10.1371/journal.pone.0273208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 08/03/2022] [Indexed: 12/03/2022] Open
Abstract
The sympathetic nervous system is highly involved in the regulation of gastrointestinal functions such as luminal alkalinisation and fluid absorption. However, the exact mechanisms are not clear. This study aimed to delineate how α2-adrenergic receptor stimulation reduces duodenal luminal alkalinisation and induces net fluid absorption. This was tested by perfusing the duodenum of anesthetized rats with isotonic solutions devoid of Cl- and/or Na+, in the absence and presence of the α2-adrenoceptor agonist clonidine. The clonidine was also studied in rats treated with dimethylamiloride (a Na+/H+ exchange inhibitor), vasoactive intestinal peptide, and the nicotinic receptor antagonist hexamethonium. Clonidine reduced luminal alkalinisation and induced net fluid absorption. The Cl--free solution decreased luminal alkalinisation and abolished net fluid absorption, but did not prevent clonidine from doing so. Both the Na+-free solution and luminal dimethylamiloride increased luminal alkalinisation and abolished net fluid absorption, effects counteracted by clonidine. The NaCl-free solution (D-mannitol) did not affect luminal alkalinisation, but reduced net fluid absorption. Clonidine reduced luminal alkalinisation and induced net fluid absorption in rats perfused luminally with mannitol. However, clonidine did not affect the vasoactive intestinal peptide-induced increase in luminal alkalinisation or fluid secretion. Pre-treatment with hexamethonium abolished the effects of clonidine on luminal alkalinisation and net fluid flux. In summary, our in vivo experiments showed that clonidine-induced reduction in luminal alkalinisation and induction of net fluid absorption was unrelated to luminal Na+ and Cl-, or to apical Na+/H+ or Cl-/HCO3- exchangers. Instead, clonidine seems to exert its effects via suppression of nicotinic receptor-activated acetylcholine secretomotor neurons.
Collapse
Affiliation(s)
- Olof Nylander
- Division of Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Markus Sjöblom
- Division of Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - John Sedin
- Division of Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - David Dahlgren
- Division of Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
7
|
Nikolovska K, Seidler UE, Stock C. The Role of Plasma Membrane Sodium/Hydrogen Exchangers in Gastrointestinal Functions: Proliferation and Differentiation, Fluid/Electrolyte Transport and Barrier Integrity. Front Physiol 2022; 13:899286. [PMID: 35665228 PMCID: PMC9159811 DOI: 10.3389/fphys.2022.899286] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/19/2022] [Indexed: 12/11/2022] Open
Abstract
The five plasma membrane Na+/H+ exchanger (NHE) isoforms in the gastrointestinal tract are characterized by distinct cellular localization, tissue distribution, inhibitor sensitivities, and physiological regulation. NHE1 (Slc9a1) is ubiquitously expressed along the gastrointestinal tract in the basolateral membrane of enterocytes, but so far, an exclusive role for NHE1 in enterocyte physiology has remained elusive. NHE2 (Slc9a2) and NHE8 (Slc9a8) are apically expressed isoforms with ubiquitous distribution along the colonic crypt axis. They are involved in pHi regulation of intestinal epithelial cells. Combined use of a knockout mouse model, intestinal organoid technology, and specific inhibitors revealed previously unrecognized actions of NHE2 and NHE8 in enterocyte proliferation and differentiation. NHE3 (Slc9a3), expressed in the apical membrane of differentiated intestinal epithelial cells, functions as the predominant nutrient-independent Na+ absorptive mechanism in the gut. The new selective NHE3 inhibitor (Tenapanor) allowed discovery of novel pathophysiological and drug-targetable NHE3 functions in cystic-fibrosis associated intestinal obstructions. NHE4, expressed in the basolateral membrane of parietal cells, is essential for parietal cell integrity and acid secretory function, through its role in cell volume regulation. This review focuses on the expression, regulation and activity of the five plasma membrane Na+/H+ exchangers in the gastrointestinal tract, emphasizing their role in maintaining intestinal homeostasis, or their impact on disease pathogenesis. We point to major open questions in identifying NHE interacting partners in central cellular pathways and processes and the necessity of determining their physiological role in a system where their endogenous expression/activity is maintained, such as organoids derived from different parts of the gastrointestinal tract.
Collapse
|
8
|
Nwia SM, Li XC, Leite APDO, Hassan R, Zhuo JL. The Na +/H + Exchanger 3 in the Intestines and the Proximal Tubule of the Kidney: Localization, Physiological Function, and Key Roles in Angiotensin II-Induced Hypertension. Front Physiol 2022; 13:861659. [PMID: 35514347 PMCID: PMC9062697 DOI: 10.3389/fphys.2022.861659] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/25/2022] [Indexed: 01/29/2023] Open
Abstract
The sodium (Na+)/hydrogen (H+) exchanger 3 (NHE3) is one of the most important Na+/H+ antiporters in the small intestines of the gastrointestinal tract and the proximal tubules of the kidney. The roles of NHE3 in the regulation of intracellular pH and acid-base balance have been well established in cellular physiology using in vitro techniques. Localized primarily on the apical membranes in small intestines and proximal tubules, the key action of NHE3 is to facilitate the entry of luminal Na+ and the extrusion of intracellular H+ from intestinal and proximal tubule tubular epithelial cells. NHE3 is, directly and indirectly, responsible for absorbing the majority of ingested Na+ from small and large intestines and reabsorbing >50% of filtered Na+ in the proximal tubules of the kidney. However, the roles of NHE3 in the regulation of proximal tubular Na+ transport in the integrative physiological settings and its contributions to the basal blood pressure regulation and angiotensin II (Ang II)-induced hypertension have not been well studied previously due to the lack of suitable animal models. Recently, novel genetically modified mouse models with whole-body, kidney-specific, or proximal tubule-specific deletion of NHE3 have been generated by us and others to determine the critical roles and underlying mechanisms of NHE3 in maintaining basal body salt and fluid balance, blood pressure homeostasis, and the development of Ang II-induced hypertension at the whole-body, kidney, or proximal tubule levels. The objective of this invited article is to review, update, and discuss recent findings on the critical roles of intestinal and proximal tubule NHE3 in maintaining basal blood pressure homeostasis and their potential therapeutic implications in the development of angiotensin II (Ang II)-dependent hypertension.
Collapse
Affiliation(s)
- Sarah M. Nwia
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Xiao Chun Li
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Ana Paula de Oliveira Leite
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Rumana Hassan
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jia Long Zhuo
- Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, United States,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States,*Correspondence: Jia Long Zhuo,
| |
Collapse
|
9
|
Jacobs JW, Leadbetter MR, Bell N, Koo-McCoy S, Carreras CW, He L, Kohler J, Kozuka K, Labonté ED, Navre M, Spencer AG, Charmot D. Discovery of Tenapanor: A First-in-Class Minimally Systemic Inhibitor of Intestinal Na +/H + Exchanger Isoform 3. ACS Med Chem Lett 2022; 13:1043-1051. [PMID: 35859876 PMCID: PMC9290029 DOI: 10.1021/acsmedchemlett.2c00037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
![]()
We present herein
the design, synthesis, and optimization of gut-restricted
inhibitors of Na+/H+ exchanger isoform 3 (NHE3).
NHE3 is predominantly expressed in the kidney and gastrointestinal
tract where it acts as the major absorptive sodium transporter. We
desired minimally systemic agents that would block sodium absorption
in the gastrointestinal tract but avoid exposure in the kidney. Starting
with a relatively low-potency highly bioavailable hit compound (1), potent and minimally absorbed NHE3 inhibitors were designed,
culminating with the discovery of tenapanor (28). Tenapanor
has been approved by the U.S. Food and Drug Administration (FDA) for
the treatment of irritable bowel syndrome with constipation in adults.
Collapse
Affiliation(s)
- Jeffrey W. Jacobs
- Ardelyx, Inc., 400 Fifth Avenue, Suite 210, Waltham, Massachusetts 02451, United States
| | - Michael R. Leadbetter
- Ardelyx, Inc., 400 Fifth Avenue, Suite 210, Waltham, Massachusetts 02451, United States
| | - Noah Bell
- Ardelyx, Inc., 400 Fifth Avenue, Suite 210, Waltham, Massachusetts 02451, United States
| | - Samantha Koo-McCoy
- Ardelyx, Inc., 400 Fifth Avenue, Suite 210, Waltham, Massachusetts 02451, United States
| | | | - Limin He
- Ardelyx, Inc., 400 Fifth Avenue, Suite 210, Waltham, Massachusetts 02451, United States
| | - Jill Kohler
- Ardelyx, Inc., 400 Fifth Avenue, Suite 210, Waltham, Massachusetts 02451, United States
| | - Kenji Kozuka
- Ardelyx, Inc., 400 Fifth Avenue, Suite 210, Waltham, Massachusetts 02451, United States
| | - Eric D. Labonté
- Ardelyx, Inc., 400 Fifth Avenue, Suite 210, Waltham, Massachusetts 02451, United States
| | - Marc Navre
- Ardelyx, Inc., 400 Fifth Avenue, Suite 210, Waltham, Massachusetts 02451, United States
| | - Andrew G. Spencer
- Ardelyx, Inc., 400 Fifth Avenue, Suite 210, Waltham, Massachusetts 02451, United States
| | - Dominique Charmot
- Ardelyx, Inc., 400 Fifth Avenue, Suite 210, Waltham, Massachusetts 02451, United States
| |
Collapse
|
10
|
Donowitz M. Has a physiologic function for NHE2 finally been identified? Acta Physiol (Oxf) 2022; 234:e13792. [PMID: 35094506 PMCID: PMC11283681 DOI: 10.1111/apha.13792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Mark Donowitz
- Department of Medicine and Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Xue J, Thomas L, Murali SK, Levi M, Fenton RA, Dominguez Rieg JA, Rieg T. Enhanced phosphate absorption in intestinal epithelial cell-specific NHE3 knockout mice. Acta Physiol (Oxf) 2022; 234:e13756. [PMID: 34978760 PMCID: PMC9286053 DOI: 10.1111/apha.13756] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/16/2021] [Accepted: 01/01/2022] [Indexed: 12/11/2022]
Abstract
Aims The kidneys play a major role in maintaining Pi homeostasis. Patients in later stages of CKD develop hyperphosphatemia. One novel treatment option is tenapanor, an intestinal‐specific NHE3 inhibitor. To gain mechanistic insight into the role of intestinal NHE3 in Pi homeostasis, we studied tamoxifen‐inducible intestinal epithelial cell‐specific NHE3 knockout (NHE3IEC‐KO) mice. Methods Mice underwent dietary Pi challenges, and hormones as well as urinary/plasma Pi were determined. Intestinal 33P uptake studies were conducted in vivo to compare the effects of tenapanor and NHE3IEC‐KO. Ex vivo Pi transport was measured in everted gut sacs and brush border membrane vesicles. Intestinal and renal protein expression of Pi transporters were determined. Results On the control diet, NHE3IEC‐KO mice had similar Pi homeostasis, but a ~25% reduction in FGF23 compared with control mice. Everted gut sacs and brush border membrane vesicles showed enhanced Pi uptake associated with increased Npt2b expression in NHE3IEC‐KO mice. Acute oral Pi loading resulted in higher plasma Pi in NHE3IEC‐KO mice. Tenapanor inhibited intestinal 33P uptake acutely but then led to hyper‐absorption at later time points compared to vehicle. In response to high dietary Pi, plasma Pi and FGF23 increased to higher levels in NHE3IEC‐KO mice which was associated with greater Npt2b expression. Reduced renal Npt2c and a trend for reduced Npt2a expression were unable to correct for higher plasma Pi. Conclusion Intestinal NHE3 has a significant contribution to Pi homeostasis. In contrast to effects described for tenapanor on Pi homeostasis, NHE3IEC‐KO mice show enhanced, rather than reduced, intestinal Pi uptake.
Collapse
Affiliation(s)
- Jianxiang Xue
- Department of Molecular Pharmacology and Physiology Morsani College of Medicine University of South Florida Tampa Florida USA
| | - Linto Thomas
- Department of Molecular Pharmacology and Physiology Morsani College of Medicine University of South Florida Tampa Florida USA
| | | | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology Georgetown University Washington District of Columbia USA
| | | | - Jessica A. Dominguez Rieg
- Department of Molecular Pharmacology and Physiology Morsani College of Medicine University of South Florida Tampa Florida USA
- James A. Haley Veterans' Hospital Tampa Florida USA
| | - Timo Rieg
- Department of Molecular Pharmacology and Physiology Morsani College of Medicine University of South Florida Tampa Florida USA
- James A. Haley Veterans' Hospital Tampa Florida USA
| |
Collapse
|
12
|
Elzinga J, van der Lugt B, Belzer C, Steegenga WT. Characterization of increased mucus production of HT29-MTX-E12 cells grown under Semi-Wet interface with Mechanical Stimulation. PLoS One 2021; 16:e0261191. [PMID: 34928974 PMCID: PMC8687553 DOI: 10.1371/journal.pone.0261191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 11/24/2021] [Indexed: 12/29/2022] Open
Abstract
The intestinal mucus layer plays a crucial role in human health. To study intestinal mucus function and structure in vitro, the mucus-producing intestinal cell line HT29-MTX-E12 has been commonly used. However, this cell line produces only low amounts of the intestine-specific MUC2. It has been shown previously that HT29-MTX-E12 cells cultured under Semi-Wet interface with Mechanical Stimulation (SWMS) produced higher amounts of MUC2, concomitant with a thicker mucus layer, compared to cells cultured conventionally. However, it remains unknown which underlying pathways are involved. Therefore, we aimed to further explore the cellular processes underlying the increased MUC2 production by HT29-MTX-E12 cells grown under SWMS conditions. Cells grown on Transwell membranes for 14 days under static and SWMS conditions (after cell seeding and attachment) were subjected to transcriptome analysis to investigate underlying molecular pathways at gene expression level. Caco-2 and LS174T cell lines were included as references. We characterized how SWMS conditions affected HT29-MTX-E12 cells in terms of epithelial barrier integrity, by measuring transepithelial electrical resistance, and cell metabolism, by monitoring pH and lactate production per molecule glucose of the conditioned medium. We confirmed higher MUC2 production under SWMS conditions at gene and protein level and demonstrated that this culturing method primarily stimulated cell growth. In addition, we also found evidence for a more aerobic cell metabolism under SWMS, as shown previously for similar models. In summary, we suggest different mechanisms by which MUC2 production is enhanced under SWMS and propose potential applications of this model in future studies.
Collapse
Affiliation(s)
- Janneke Elzinga
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Benthe van der Lugt
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Wilma T Steegenga
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
13
|
Kovesdy CP, Adebiyi A, Rosenbaum D, Jacobs JW, Quarles LD. Novel Treatments from Inhibition of the Intestinal Sodium-Hydrogen Exchanger 3. Int J Nephrol Renovasc Dis 2021; 14:411-420. [PMID: 34880650 PMCID: PMC8646223 DOI: 10.2147/ijnrd.s334024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/11/2021] [Indexed: 12/31/2022] Open
Abstract
Plasma membrane sodium–hydrogen exchangers (NHE) transport Na+ into cells in exchange for H+. While there are nine isoforms of NHE in humans, this review focuses on the NHE3 isoform, which is abundantly expressed in the gastrointestinal tract, where it plays a key role in acid–base balance and water homeostasis. NHE3 inhibition in the small intestine results in luminal sodium and water retention, leading to a general decrease in paracellular water flux and diffusional driving force, reduced intestinal sodium absorption, and increased stool sodium excretion. The resulting softer and more frequent stools are the rationale for the development of tenapanor as a novel, first-in-class NHE3 inhibitor to treat irritable bowel syndrome with constipation. NHE3 also has additional therapeutic implications in nephrology. Inhibition of intestinal NHE3 also lowers blood pressure by reducing intestinal sodium absorption. Perhaps, the most novel effect is its ability to decrease intestinal phosphate absorption by inhibiting the paracellular phosphate absorption pathway. Therefore, selective pharmacological inhibition of NHE3 could be a potential therapeutic strategy to treat not only heart failure and hypertension but also hyperphosphatemia. This review presents an overview of the molecular and physiological functions of NHE3 and discusses how these functions translate to potential clinical applications in nephrology.
Collapse
Affiliation(s)
- Csaba P Kovesdy
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Adebowale Adebiyi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | | - L Darryl Quarles
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
14
|
Yee J, Rosenbaum D, Jacobs JW, Sprague SM. Small Intestinal Phosphate Absorption: Novel Therapeutic Implications. Am J Nephrol 2021; 52:522-530. [PMID: 34515051 DOI: 10.1159/000518110] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/24/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) affects approximately 15% of adults in the USA. As CKD progresses, urinary phosphate excretion decreases and results in phosphate retention and, eventually, hyperphosphatemia. As hyperphosphatemia is associated with numerous adverse outcomes, including increased cardiovascular mortality, reduction in phosphorus concentrations is a guideline-recommended, established clinical practice. Dietary phosphate restriction, dialysis, and phosphate binders are currently the only options for phosphate management. However, many patients with hyperphosphatemia have phosphorus concentrations >5.5 mg/dL, despite treatment. SUMMARY This review pre-sents recent advances in the understanding of intestinal phosphate absorption and therapeutic implications. Dietary phosphate is absorbed in the intestine through two distinct pathways, paracellular absorption and transcellular transport. Recent evidence indicates that the paracellular route accounts for 65-80% of total phosphate absorbed. Thus, the paracellular pathway is the dominant mechanism of phosphate absorption. Tenapanor is a first-in-class, non-phosphate binder that inhibits the sodium-hydrogen exchanger 3 or solute carrier family 9 member 3 (SLC9A3) encoded by the SLC9A3 gene, and blocks paracellular phosphate absorption. Key Messages: Targeted inhibition of sodium-hydrogen exchanger 3 effectively reduces paracellular permeability of phosphate. Novel therapies that target the paracellular pathway may improve phosphate control in chronic kidney disease.
Collapse
Affiliation(s)
- Jerry Yee
- Nephrology and Hypertension, Henry Ford Hospital, Detroit, Michigan, USA
| | | | | | - Stuart M Sprague
- Division of Nephrology and Hypertension, NorthShore University Health System, Evanston, Illinois, USA
| |
Collapse
|
15
|
Zhou K, Amiri M, Salari A, Yu Y, Xu H, Seidler U, Nikolovska K. Functional characterization of the sodium/hydrogen exchanger 8 and its role in proliferation of colonic epithelial cells. Am J Physiol Cell Physiol 2021; 321:C471-C488. [PMID: 34288721 DOI: 10.1152/ajpcell.00582.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Intestinal NaCl, HCO3-, and fluid absorption are strongly dependent on apical Na+/H+ exchange. The intestine expresses three presumably apical sodium-hydrogen exchanger (NHE) isoforms: NHE2, NHE3, and NHE8. We addressed the role of NHE8 [solute carrier 9A8 (SLC9A8)] and its interplay with NHE2 (SLC9A2) in luminal proton extrusion during acute and chronic enterocyte acidosis and studied the differential effects of NHE8 and NHE2 on enterocyte proliferation. In contrast to NHE3, which was upregulated in differentiated versus undifferentiated colonoids, the expression of NHE2 and NHE8 remained constant during differentiation of colonoids and Caco2Bbe cells. Heterogeneously expressed Flag-tagged rat (r)Nhe8 and human (h)NHE8 translocated to the apical membrane of Caco2Bbe cells. rNhe8 and hNHE8, when expressed in NHE-deficient PS120 fibroblasts showed higher sensitivity to HOE642 compared to NHE2. Lentiviral shRNA knockdown of endogenous NHE2 in Caco2Bbe cells (C2Bbe/shNHE2) resulted in a decreased steady-state intracellular pH (pHi), an increased NHE8 mRNA expression, and augmented NHE8-mediated apical NHE activity. Lentiviral shRNA knockdown of endogenous NHE8 in Caco2Bbe cells (C2Bbe/shNHE8) resulted in a decreased steady-state pHi as well, accompanied by decreased NHE2 mRNA expression and activity, which together contributed to reduced apical NHE activity in the NHE8-knockdown cells. Chronic acidosis increased NHE8 but not NHE2 mRNA expression. Alterations in NHE2 and NHE8 expression/activity affected proliferation, with C2Bbe/shNHE2 cells having lower and C2Bbe/shNHE8 having higher proliferative capacity, accompanied by amplified ERK1/2 signaling pathway and increased EGFR expression in the latter cell line. Thus, both Na+/H+ exchangers have distinct functions during cellular homeostasis by triggering different signaling pathways to regulate cellular proliferation and pHi control.
Collapse
Affiliation(s)
- Kunyan Zhou
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Mahdi Amiri
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Azam Salari
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Yan Yu
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Hua Xu
- Department of Pediatrics, University of Arizona Health Science Center, Tucson, Arizona
| | - Ursula Seidler
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Katerina Nikolovska
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
16
|
New Insights into the Critical Importance of Intratubular Na +/H + Exchanger 3 and Its Potential Therapeutic Implications in Hypertension. Curr Hypertens Rep 2021; 23:34. [PMID: 34110521 DOI: 10.1007/s11906-021-01152-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW The sodium (Na+) and hydrogen (H+) exchanger 3 (NHE3), known as solute carrier family 9 member 3 (SLC9A3), mediates active transcellular Na+ and bicarbonate reabsorption in the small intestine of the gut and proximal tubules of the kidney. The purpose of this article is to review and discuss recent findings on the critical roles of intestinal and proximal tubule NHE3 in maintaining basal blood pressure (BP) homeostasis and their potential therapeutic implications in the development of angiotensin II (Ang II)-dependent hypertension. RECENT FINDINGS Recently, our and other laboratories have generated or used novel genetically modified mouse models with whole-body, kidney-specific, or proximal tubule-specific deletion of NHE3 to determine the critical roles and underlying mechanisms of NHE3 in maintaining basal BP homeostasis and the development of Ang II-induced hypertension at the whole-body, kidney, or proximal tubule levels. The new findings demonstrate that NHE3 contributes to about 10 to 15 mmHg to basal blood pressure levels, and that deletion of NHE3 at the whole-kidney or proximal tubule level, or pharmacological inhibition of NHE3 at the kidney level with an orally absorbable NHE3 inhibitor AVE-0657, attenuates ~ 50% of Ang II-induced hypertension in mice. The results support the proof-of-concept hypothesis that NHE3 plays critical roles in physiologically maintaining normal BP and in the development of Ang II-dependent hypertension. Our results also strongly suggest that NHE3 in the proximal tubules of the kidney may be therapeutically targeted to treat poorly controlled hypertension in humans.
Collapse
|
17
|
Efficacy of Tenapanor in Treating Patients With Irritable Bowel Syndrome With Constipation: A 26-Week, Placebo-Controlled Phase 3 Trial (T3MPO-2). Am J Gastroenterol 2021; 116:1294-1303. [PMID: 33337659 PMCID: PMC8183489 DOI: 10.14309/ajg.0000000000001056] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Tenapanor is a first-in-class, minimally absorbed, small-molecule inhibitor of the gastrointestinal sodium/hydrogen exchanger isoform 3. This phase 3 trial assessed the long-term efficacy and safety of tenapanor 50 mg b.i.d. for the treatment of patients with irritable bowel syndrome with constipation (IBS-C). METHODS In this randomized double-blind study (ClinicalTrials.gov identifier: NCT02686138), patients with IBS-C received tenapanor 50 mg b.i.d. or placebo b.i.d. for 26 weeks. The primary endpoint was the proportion of patients who had a reduction of ≥30.0% in average weekly worst abdominal pain and an increase of ≥1 weekly complete spontaneous bowel movement from baseline, both in the same week, for ≥6 of the first 12 treatment weeks (6/12-week combined responder). RESULTS Of the 620 randomized patients with IBS-C, 593 (95.6%) were included in the intention-to-treat analysis set (tenapanor: n = 293; placebo: n = 300) and 481 patients (77.6%) completed the 26-week treatment period. In the intention-to-treat analysis set (mean age: 45.4 years; 82.1% women), a significantly greater proportion of patients treated with tenapanor were 6/12-week combined responders than those treated with placebo (36.5% vs 23.7%; P < 0.001). Abdominal symptoms and global symptoms of IBS were significantly improved with tenapanor compared with placebo. Diarrhea, the most common adverse event, was typically transient and mild to moderate in severity. Diarrhea led to study drug discontinuation for 19 (6.5%) and 2 patients (0.7%) receiving tenapanor and placebo, respectively. DISCUSSION Tenapanor 50 mg b.i.d. improved IBS-C symptoms over 26 weeks and was generally well tolerated, offering a potential new long-term treatment option for patients with IBS-C (see Visual abstract, Supplementary Digital Content 1, http://links.lww.com/AJG/B797).
Collapse
|
18
|
Stephens CE, Whittamore JM, Hatch M. The role of NHE3 (Slc9a3) in oxalate and sodium transport by mouse intestine and regulation by cAMP. Physiol Rep 2021; 9:e14828. [PMID: 33904662 PMCID: PMC8077127 DOI: 10.14814/phy2.14828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/01/2021] [Accepted: 03/09/2021] [Indexed: 12/14/2022] Open
Abstract
Intestinal oxalate transport involves Cl−/HCO3− exchangers but how this transport is regulated is not currently known. NHE3 (Slc9a3), an apical Na+/H+ exchanger, is an established target for regulation of electroneutral NaCl absorption working in concert with Cl−/HCO3− exchangers. To test whether NHE3 could be involved in regulation of intestinal oxalate transport and renal oxalate handling we compared urinary oxalate excretion rates and intestinal transepithelial fluxes of 14C‐oxalate and 22Na+ between NHE3 KO and wild‐type (WT) mice. NHE3 KO kidneys had lower creatinine clearance suggesting reduced GFR, but urinary oxalate excretion rates (µmol/24 h) were similar compared to the WT but doubled when expressed as a ratio of creatinine. Intestinal transepithelial fluxes of 14C‐oxalate and 22Na+ were measured in the distal ileum, cecum, and distal colon. The absence of NHE3 did not affect basal net transport rates of oxalate or sodium across any intestinal section examined. Stimulation of intracellular cAMP with forskolin (FSK) and 3‐isobutyl‐1‐methylxanthine (IBMX) led to an increase in net oxalate secretion in the WT distal ileum and cecum and inhibition of sodium absorption in the cecum and distal colon. In NHE3 KO cecum, cAMP stimulation of oxalate secretion was impaired suggesting the possibility of a role for NHE3 in this process. Although, there is little evidence for a role of NHE3 in basal intestinal oxalate fluxes, NHE3 may be important for cAMP stimulation of oxalate in the cecum and for renal handling of oxalate.
Collapse
Affiliation(s)
- Christine E Stephens
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jonathan M Whittamore
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Marguerite Hatch
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
19
|
Exploring the impact of intestinal ion transport on the gut microbiota. Comput Struct Biotechnol J 2020; 19:134-144. [PMID: 33425246 PMCID: PMC7773683 DOI: 10.1016/j.csbj.2020.12.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota and the host are intimately connected. The host physiology dictates the intestinal environment through regulation of pH, ion concentration, mucus production, etc., all of which exerts a selective pressure on the gut microbiota. Since different regions of the gastrointestinal tract are characterized by their own physicochemical conditions, distinct microbial communities are present in these locations. While it is widely accepted that the intestinal microbiome influences the host (tight junctions, cytokine/immune responses, diarrhea, etc.), the reciprocal interaction of the host on the microbiome is under-explored. This review aims to address these gaps in knowledge by focusing on how the host intestinal ion transport influences the luminal environment and thereby modulates the gut microbiota composition.
Collapse
Key Words
- CFTR
- CFTR, cystic fibrosis transmembrane regulator
- ClC, chloride channel
- DRA
- DRA, down-regulated in adenoma
- ENaC, epithelial Na+ channel
- GI, gastrointestinal
- GLUT2
- GLUT2, glucose transporter 2
- Gastrointestinal
- Ion transport
- Microbiome
- Microbiota
- NHE2
- NHE2, sodium-hydrogen exchanger isoform 2
- NHE3
- NHE3, sodium-hydrogen exchanger isoform 3
- NKCC1, Na+-K+-2Cl− co-transporter
- OTUs, operational taxonomic units
- SGLT1, sodium glucose co-transporter 1
Collapse
|
20
|
Deciphering ion transporters, kinases and PDZ-adaptor molecules that mediate guanylate cyclase C agonist-dependent intestinal fluid loss in vivo. Biochem Pharmacol 2020; 178:114040. [DOI: 10.1016/j.bcp.2020.114040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/13/2020] [Indexed: 01/12/2023]
|
21
|
Na +-Coupled Nutrient Cotransport Induced Luminal Negative Potential and Claudin-15 Play an Important Role in Paracellular Na + Recycling in Mouse Small Intestine. Int J Mol Sci 2020; 21:ijms21020376. [PMID: 31936130 PMCID: PMC7013606 DOI: 10.3390/ijms21020376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/27/2019] [Accepted: 12/30/2019] [Indexed: 12/26/2022] Open
Abstract
Many nutrients are absorbed via Na+ cotransport systems, and therefore it is predicted that nutrient absorption mechanisms require a large amount of luminal Na+. It is thought that Na+ diffuses back into the lumen via paracellular pathways to support Na+ cotransport absorption. However, direct experimental evidence in support of this mechanism has not been shown. To elucidate this, we took advantage of claudin-15 deficient (cldn15-/-) mice, which have been shown to have decreased paracellular Na+ permeability. We measured glucose-induced currents (ΔIsc) under open- and short-circuit conditions and simultaneously measured changes in unidirectional 22Na+ fluxes (ΔJNa) in Ussing chambers. Under short-circuit conditions, application of glucose resulted in an increase in ΔIsc and unidirectional mucosal to serosal 22Na+ (∆JNaMS) flux in both wild-type and cldn15-/- mice. However, under open-circuit conditions, ΔIsc was observed but ∆JNaMS was strongly inhibited in wild-type but not in cldn15-/- mice. In addition, in the duodenum of mice treated with cholera toxin, paracellular Na+ conductance was decreased and glucose-induced ∆JNaMS increment was observed under open-circuit conditions. We concluded that the Na+ which is absorbed by Na+-dependent glucose cotransport is recycled back into the lumen via paracellular Na+ conductance through claudin-15, which is driven by Na+ cotransport induced luminal negativity.
Collapse
|
22
|
The role of splanchnic congestion and the intestinal microenvironment in the pathogenesis of advanced heart failure. Curr Opin Support Palliat Care 2019; 13:24-30. [PMID: 30640740 DOI: 10.1097/spc.0000000000000414] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW Right-sided heart failure, which is often present in the setting of advanced heart failure, is associated with cardiac cachexia, the cardiorenal syndrome, and adverse outcomes. Improved understanding of venous congestion of the splanchnic circulation, which may play a key role in the pathogenesis of right-sided heart failure, could lead to novel therapeutics to ameliorate heart failure. Here we provide an overview of right-sided heart failure, splanchnic hemodynamics, fluid homeostasis, and the intestinal microenvironment. We review recent literature to describe pathophysiologic mechanisms and possible therapeutics. RECENT FINDINGS Several possible mechanisms centered around upregulation of sodium-hydrogen exchanger-3 (NHE3) may form a causal link between right ventricular dysfunction, splanchnic congestion, and worsening heart failure. These include an anaerobic environment in enterocytes, resulting in reduced intracellular pH; increased sodium absorption by the gut via NHE3; decreased pH at the intestinal brush border thus altering the gut microbiome profile; increased bacterial synthesis of trimethylamine N-oxide; and decreased bacterial synthesis of short-chain fatty acids causing abnormal intestinal barrier function. SUMMARY Splanchnic congestion in the setting of right-sided heart failure may serve an important role in the pathogenesis of advanced heart failure, and further exploration of these mechanisms may lead to new therapeutic advances.
Collapse
|
23
|
Wang JL, Zhao L, Zhu J, Wang DK, Ren MJ, Wang M, Liu Y, Boron WF, Chen LM. Expression, Localization, and Effect of High Salt Intake on Electroneutral Na +/HCO 3 - Cotransporter NBCn2 in Rat Small Intestine: Implication in Intestinal NaCl Absorption. Front Physiol 2019; 10:1334. [PMID: 31736772 PMCID: PMC6828735 DOI: 10.3389/fphys.2019.01334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/07/2019] [Indexed: 12/20/2022] Open
Abstract
The electroneutral Na+/HCO3 - cotransporter NBCn2 (SLC4A10) of solute carrier family 4 (SLC4) plays important physiological and pathological roles in the body. Our previous study showed that NBCn2 is expressed on the protein level in the small intestine of rat. Here, by reverse-transcription polymerase chain reaction (PCR), we identified a novel full-length NBCn2 variant, i.e., NBCn2-K, from rat small intestine. By pHi measurement with Xenopus oocytes, the activity of NBCn2-K is not significantly different from NBCn2-G. By western blotting, NBCn2 and the Na+/H+ exchanger NHE3 (SLC9A3) are predominantly expressed in the jejunum of rat small intestine. By immunofluorescence, NBCn2 and NHE3 are localized at the apical domain of the jejunum. NaCl overload decreases the expression of NBCn2 by 56% and that of NHE3 by 40% in the small intestine. We propose that NBCn2 is involved in the transepithelial NaCl absorption in the small intestine, and that the down-regulation of NBCn2 by NaCl represents an adaptive response to high salt intake in rat.
Collapse
Affiliation(s)
- Jin-Lin Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhao
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Jun Zhu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Deng-Ke Wang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Mei-Juan Ren
- Key Laboratory of Molecular Biophysics of the Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Walter F. Boron
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Li-Ming Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
King AJ, Siegel M, He Y, Nie B, Wang J, Koo-McCoy S, Minassian NA, Jafri Q, Pan D, Kohler J, Kumaraswamy P, Kozuka K, Lewis JG, Dragoli D, Rosenbaum DP, O'Neill D, Plain A, Greasley PJ, Jönsson-Rylander AC, Karlsson D, Behrendt M, Strömstedt M, Ryden-Bergsten T, Knöpfel T, Pastor Arroyo EM, Hernando N, Marks J, Donowitz M, Wagner CA, Alexander RT, Caldwell JS. Inhibition of sodium/hydrogen exchanger 3 in the gastrointestinal tract by tenapanor reduces paracellular phosphate permeability. Sci Transl Med 2019; 10:10/456/eaam6474. [PMID: 30158152 DOI: 10.1126/scitranslmed.aam6474] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 10/31/2017] [Accepted: 03/06/2018] [Indexed: 12/16/2022]
Abstract
Hyperphosphatemia is common in patients with chronic kidney disease and is increasingly associated with poor clinical outcomes. Current management of hyperphosphatemia with dietary restriction and oral phosphate binders often proves inadequate. Tenapanor, a minimally absorbed, small-molecule inhibitor of the sodium/hydrogen exchanger isoform 3 (NHE3), acts locally in the gastrointestinal tract to inhibit sodium absorption. Because tenapanor also reduces intestinal phosphate absorption, it may have potential as a therapy for hyperphosphatemia. We investigated the mechanism by which tenapanor reduces gastrointestinal phosphate uptake, using in vivo studies in rodents and translational experiments on human small intestinal stem cell-derived enteroid monolayers to model ion transport physiology. We found that tenapanor produces its effect by modulating tight junctions, which increases transepithelial electrical resistance (TEER) and reduces permeability to phosphate, reducing paracellular phosphate absorption. NHE3-deficient monolayers mimicked the phosphate phenotype of tenapanor treatment, and tenapanor did not affect TEER or phosphate flux in the absence of NHE3. Tenapanor also prevents active transcellular phosphate absorption compensation by decreasing the expression of NaPi2b, the major active intestinal phosphate transporter. In healthy human volunteers, tenapanor (15 mg, given twice daily for 4 days) increased stool phosphorus and decreased urinary phosphorus excretion. We determined that tenapanor reduces intestinal phosphate absorption predominantly through reduction of passive paracellular phosphate flux, an effect mediated exclusively via on-target NHE3 inhibition.
Collapse
Affiliation(s)
| | | | - Ying He
- Ardelyx Inc., Fremont, CA 94555, USA
| | | | - Ji Wang
- Ardelyx Inc., Fremont, CA 94555, USA
| | | | | | | | - Deng Pan
- Ardelyx Inc., Fremont, CA 94555, USA
| | | | | | | | | | | | | | - Debbie O'Neill
- University of Alberta, Edmonton, Alberta T6G 1C9, Canada
| | - Allein Plain
- University of Alberta, Edmonton, Alberta T6G 1C9, Canada
| | - Peter J Greasley
- Cardiovascular and Metabolic Disease (CVMD) Translational Medicine Unit, Early Clinical Development, Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca Gothenburg, 431 50 Mölndal, Sweden
| | | | - Daniel Karlsson
- Bioscience, CVMD, IMED Biotech Unit, AstraZeneca Gothenburg, 431 50 Mölndal, Sweden
| | - Margareta Behrendt
- Bioscience, CVMD, IMED Biotech Unit, AstraZeneca Gothenburg, 431 50 Mölndal, Sweden
| | - Maria Strömstedt
- Bioscience, CVMD, IMED Biotech Unit, AstraZeneca Gothenburg, 431 50 Mölndal, Sweden
| | | | - Thomas Knöpfel
- Institute of Physiology, University of Zurich and National Center of Competence in Research Kidney Control of Homeostasis, CH-8057 Zurich, Switzerland
| | - Eva M Pastor Arroyo
- Institute of Physiology, University of Zurich and National Center of Competence in Research Kidney Control of Homeostasis, CH-8057 Zurich, Switzerland
| | - Nati Hernando
- Institute of Physiology, University of Zurich and National Center of Competence in Research Kidney Control of Homeostasis, CH-8057 Zurich, Switzerland
| | - Joanne Marks
- Department of Neuroscience, Physiology and Pharmacology, University College London, Royal Free Campus, London NW3 2PF, UK
| | - Mark Donowitz
- Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich and National Center of Competence in Research Kidney Control of Homeostasis, CH-8057 Zurich, Switzerland
| | | | | |
Collapse
|
25
|
Yu Y, Seidler A, Zhou K, Yuan Z, Yeruva S, Amiri M, Yun CC, Nikolovska K, Seidler U. Expression, Localization and Functional Activity of the Major Na⁺/H⁺ Exchange Isoforms Expressed in the Intestinal Cell Line Caco-2BBe. Cell Physiol Biochem 2019; 52:1017-1038. [PMID: 30977986 PMCID: PMC7104346 DOI: 10.33594/000000070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 02/25/2019] [Indexed: 12/15/2022] Open
Abstract
Background/Aims Enterocytes express a number of NHE isoforms with presumed localization in the apical (NHE2, 3 and 8) or basolateral (NHE1) membrane. Functional activity and localization of enterocyte NHE isoforms were assessed using fully differentiated Caco-2BBe cells, whose genetic expression profile closely resembles mature enterocytes. Methods The activity of the different NHEs was analyzed by fluorometric pHi-metry in a perfusion chamber with separate apical and basolateral perfusion, using specific inhibitors and shRNA knockdown of NHE2. The expression of the NHEs and of other relevant acid extrusion transporters was quantified by qPCR. Results Quantitative comparison of the mRNA expression levels of the different NHE isoforms in 14 day-differentiated Caco-2BBe cells showed the following order: NHE2>NHE8>NHE3>NHE1. Acid-activated NHE exchange rates in the basolateral membrane were >6-fold higher than in the apical membrane. 79 ± 3 % of the acid-activated basolateral Na+/H+ exchange rate displayed a NHE1-typical inhibitor profile, and no NHE2/3/8 typical activity could be observed. Analysis of the apical Na+/H+ exchange rates revealed that approximately 51 ± 3 % of the total apical activity displayed a NHE2/8-typical inhibitor profile and 31 ± 6 % a NHE3-typical inhibitor profile. Because no selective NHE2 inhibitor is available, a stable NHE2 knockdown cell line (C2NHE2KD) was generated. C2NHE2KD displayed a reduced NHE2-typical apical Na+/H+ exchange rate and maintained a lower steady-state pHi, despite high expression levels of other acid extruders, in particular NBCn1 (Slc4a7). Conclusion Differentiated Caco-2BBe cells display particularly high mRNA expression levels of NHE2, which can be functionally identified in the apical membrane. Although at low intracellular pH, NHE2 transport rate was far lower than that of NHE1. NHE2 activity was nevertheless essential for the maintenance of the steady-state pHi of these cells.
Collapse
Affiliation(s)
- Yan Yu
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Anna Seidler
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Kunyan Zhou
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Zhenglin Yuan
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Sunil Yeruva
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Mahdi Amiri
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Chris C Yun
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | | | - Ursula Seidler
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany,
| |
Collapse
|
26
|
Liu Y, Wen H, Qi X, Zhang X, Zhang K, Fan H, Tian Y, Hu Y, Li Y. Genome-wide identification of the Na+/H+ exchanger gene family in Lateolabrax maculatus and its involvement in salinity regulation. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2019; 29:286-298. [DOI: 10.1016/j.cbd.2019.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/03/2019] [Accepted: 01/04/2019] [Indexed: 01/21/2023]
|
27
|
Stephens CE, Whittamore JM, Hatch M. 125 Iodide as a surrogate tracer for epithelial chloride transport by the mouse large intestine in vitro. Exp Physiol 2019; 104:334-344. [PMID: 30615234 PMCID: PMC6397055 DOI: 10.1113/ep087445] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/02/2019] [Indexed: 12/25/2022]
Abstract
NEW FINDINGS What is the central question of this study? The tracer 36 Cl- , currently used to measure transepithelial Cl- fluxes, has become prohibitively expensive, threatening its future use. 125 Iodide, previously validated alongside 36 Cl- as a tracer of Cl- efflux by cells, has not been tested as a surrogate for 36 Cl- across epithelia. What is the main finding and its importance? We demonstrate that 125 I- can serve as an inexpensive replacement for measuring Cl- transport across mouse large intestine, tracking Cl- transport in response to cAMP stimulation (inducing Cl- secretion) in the presence and absence of the main gastrointestinal Cl- -HCO3- exchanger, DRA. ABSTRACT Chloride transport is important for driving fluid secretion and absorption by the large intestine, with dysregulation resulting in diarrhoea-associated pathologies. The radioisotope 36 Cl- has long been used as a tracer to measure epithelial Cl- transport but is prohibitively expensive. 125 Iodide has been used as an alternative to 36 Cl- in some transport assays but has never been validated as an alternative for tracing bidirectional transepithelial Cl- fluxes. The goal of this study was to validate 125 I- as an alternative to 36 Cl- for measurement of Cl- transport by the intestine. Simultaneous fluxes of 36 Cl- and 125 I- were measured across the mouse caecum and distal colon. Net Cl- secretion was induced by the stimulation of cAMP with a cocktail of forskolin (FSK) and 3-isobutyl-1-methylxanthine (IBMX). Unidirectional fluxes of 125 I- correlated well with 36 Cl- fluxes after cAMP-induced net Cl- secretion, occurring predominantly through a reduction in the absorptive mucosal-to-serosal Cl- flux rather than by stimulation of the secretory serosal-to-mucosal Cl- flux. Correlations between 125 I- fluxes and 36 Cl- fluxes were maintained in epithelia from mice lacking DRA (Slc26a3), the main Cl- -HCO3- exchanger responsible for Cl- absorption by the large intestine. Lower rates of Cl- and I- absorption in the DRA knockout intestine suggest that DRA might have a previously unrecognized role in iodide uptake. This study validates that 125 I- traces transepithelial Cl- fluxes across the mouse large intestine, provides insights into the mechanism of net Cl- secretion and suggests that DRA might be involved in intestinal iodide absorption.
Collapse
Affiliation(s)
- Christine E Stephens
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jonathan M Whittamore
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Marguerite Hatch
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
28
|
The role of SLC9A3 in Taiwanese patients with congenital bilateral absence of vas deferens (CBAVD). J Formos Med Assoc 2019; 118:1576-1583. [PMID: 30797621 DOI: 10.1016/j.jfma.2019.01.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/29/2018] [Accepted: 01/29/2019] [Indexed: 02/05/2023] Open
Abstract
Congenital bilateral absence of vas deferens (CBAVD) is a special entity in obstructive azoospermia. Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene are involved in Taiwanese CBAVD but most heterozygous 5T variant. The solute carrier family 9 isoform 3 (SLC9A3) is the Na+/H+ exchanger, which interacts with CFTR and regulates the Ca2+ homeostasis. Loss of SLC9A3 decreases CFTR protein and causes obstructive azoospermia in mice. It also causes mal-reabsorption by the efferent tubules, which leads to the obstructive phenomenon and eventually results in testicular atrophy. In 6-month old SLC9A3 deficiency mice, the atrophy of their vas deferens and seminal vesicles become more prominent. Decreases of CFTR expression in the reproductive organ in the SLC9A3 deficient (-/-) mice prove the interaction between CFTR and SLC9A3 in the reproductive tract. Most of Taiwanese CBAVD have at least one variant of SLC9A3 deletion and CFTR IVS8-5T, which co-contribute to Taiwanese CBAVD. The report indicates SLC9A3 deficiency can reverse the pathological changes in the gastrointestinal tract of CF mice. Further research can explore the definite mechanism of SLC9A3 and its role interacting with CFTR in different organ systems, which can contribute to novel treatment for the patients with cystic fibrosis and CBAVD.
Collapse
|
29
|
Saurette M, Alexander RT. Intestinal phosphate absorption: The paracellular pathway predominates? Exp Biol Med (Maywood) 2019; 244:646-654. [PMID: 30764666 DOI: 10.1177/1535370219831220] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
IMPACT STATEMENT This review summarizes the work on transcellular intestinal phosphate absorption, arguing why this pathway is not the predominant pathway in humans consuming a "Western" diet. We then highlight the recent evidence which is strongly consistent with paracellular intestinal phosphate absorption mediating the bulk of intestinal phosphate absorption in humans.
Collapse
Affiliation(s)
- Matthew Saurette
- 1 Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2R7, Canada.,2 The Women's & Children's Health Research Institute, Edmonton, Alberta T6G 1C9, Canada
| | - R Todd Alexander
- 1 Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2R7, Canada.,2 The Women's & Children's Health Research Institute, Edmonton, Alberta T6G 1C9, Canada.,3 Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| |
Collapse
|
30
|
Yang Z, Ran L, Yuan P, Yang Y, Wang K, Xie L, Huang S, Liu J, Song Z. EGFR as a Negative Regulatory Protein Adjusts the Activity and Mobility of NHE3 in the Cell Membrane of IPEC-J2 Cells With TGEV Infection. Front Microbiol 2018; 9:2734. [PMID: 30483239 PMCID: PMC6243134 DOI: 10.3389/fmicb.2018.02734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/25/2018] [Indexed: 11/13/2022] Open
Abstract
Transmissible gastroenteritis (TGE) has caused devastating economic losses to the swine industry worldwide, despite extensive research focusing on the pathogenesis of virus infection. The molecular pathogenic mechanism of TGEV-induced diarrhea in piglets is unknown. Intestinal diarrhea is closely related to the function of the Na+/H+ exchanger protein NHE3 in the brush border membrane of small intestine epithelial cells. The epidermal growth factor receptor (EGFR) may act to regulate NHE3 expression. In addition, EGFR may promote viral invasion of host cells. The present study aimed to determine whether NHE3 activity is regulated by altering EGFR expression to affect Na+ absorption in TGEV-infected intestinal epithelial cells. Porcine intestinal epithelial cells were used as models for TGEV infection. The results showed that Na+ absorption and NHE3 expression levels decreased in TGEV-infected cells. Proliferation of TGEV within IPEC-J2 cells could be inhibited by treatment with the EGFR inhibitor AG1478 and knockdown; resulting in recovery of Na+ absorption in TGEV infected cells and increasing the activity and expression of NHE3. Moreover, we demonstrated that NHE3 activity was regulated through the EGFR/ERK pathway. Importantly, NHE3 mobility on the plasma membrane of TGEV infected cells was significantly weaker than that in normal cells, and EGFR inhibition and knockdown recovered this mobility. Our research indicated that NHE3 activity was negatively regulated by EGFR in TGEV-infected intestinal epithelial cells.
Collapse
Affiliation(s)
- Zhou Yang
- Department of Veterinary Medicine, Southwest University, Chongqing, China
| | - Ling Ran
- Department of Veterinary Medicine, Southwest University, Chongqing, China
| | - Peng Yuan
- Department of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yang Yang
- Department of Veterinary Medicine, Southwest University, Chongqing, China
| | - Kai Wang
- Department of Veterinary Medicine, Southwest University, Chongqing, China
| | - Luyi Xie
- Department of Veterinary Medicine, Southwest University, Chongqing, China
| | - Shilei Huang
- Department of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jia Liu
- Department of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zhenhui Song
- Department of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
31
|
Ishizuka N, Nakayama M, Watanabe M, Tajima H, Yamauchi Y, Ikari A, Hayashi H. Luminal Na + homeostasis has an important role in intestinal peptide absorption in vivo. Am J Physiol Gastrointest Liver Physiol 2018; 315:G799-G809. [PMID: 30138575 DOI: 10.1152/ajpgi.00099.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal cell line studies indicated luminal Na+ homeostasis is essential for proton-coupled peptide absorption, because the driving force of PepT1 activity is supported by the apical Na+/H+ exchanger NHE3. However, there is no direct evidence demonstrating the importance of in vivo luminal Na+ for peptide absorption in animal experiments. To investigate the relationship between luminal Na+ homeostasis and peptide absorption, we took advantage of claudin 15-deficient (cldn15-/-) mice, whereby Na+ homeostasis is disrupted. We quantitatively assessed the intestinal segment responsible for peptide absorption using radiolabeled nonhydrolyzable dipeptide (glycylsarcosine, Gly-Sar) and nonabsorbable fluid phase marker polyethylene glycol (PEG) 4000 in vivo. In wild-type (WT) mice, the concentration ratio of Gly-Sar to PEG 4000 decreased in the upper jejunum, suggesting the upper jejunum is responsible for peptide absorption. Gly-Sar absorption was decreased in the jejunum of cldn15-/- mice. To elucidate the mechanism underlining these impairments, a Gly-Sar-induced short-circuit ( Isc) current was measured. In WT mice, increments of Gly-Sar-induced Isc were inhibited by the luminal application of a NHE3-specific inhibitor S3226 in a dose-dependent fashion. In contrast to in vivo experiments, robust Gly-Sar-induced Isc increments were observed in the jejunal mucosa of cldn15-/- mice. Gly-Sar-induced Isc was inhibited by S3226 or a reduction of luminal Na+ concentration, which mimics low luminal Na+ concentrations in vivo . Our study demonstrates that luminal Na+ homeostasis is important for peptide absorption in native epithelia and that there is a cooperative functional relationship between PepT1 and NHE3. NEW & NOTEWORTHY Our study is the first to demonstrate that luminal Na+ homeostasis is important for proton-coupled peptide absorption in in vivo animal experiments.
Collapse
Affiliation(s)
- Noriko Ishizuka
- Laboratory of Physiology School of Food and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka , Japan
| | - Michiko Nakayama
- Laboratory of Physiology School of Food and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka , Japan
| | - Miki Watanabe
- Laboratory of Physiology School of Food and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka , Japan
| | - Haruna Tajima
- Laboratory of Physiology School of Food and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka , Japan
| | - Yuri Yamauchi
- Laboratory of Physiology School of Food and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka , Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University , Gifu , Japan
| | - Hisayoshi Hayashi
- Laboratory of Physiology School of Food and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka , Japan
| |
Collapse
|
32
|
McHugh DR, Cotton CU, Moss FJ, Vitko M, Valerio DM, Kelley TJ, Hao S, Jafri A, Drumm ML, Boron WF, Stern RC, McBennett K, Hodges CA. Linaclotide improves gastrointestinal transit in cystic fibrosis mice by inhibiting sodium/hydrogen exchanger 3. Am J Physiol Gastrointest Liver Physiol 2018; 315:G868-G878. [PMID: 30118317 PMCID: PMC9925117 DOI: 10.1152/ajpgi.00261.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastrointestinal dysfunction in cystic fibrosis (CF) is a prominent source of pain among patients with CF. Linaclotide, a guanylate cyclase C (GCC) receptor agonist, is a US Food and Drug Administration-approved drug prescribed for chronic constipation but has not been widely used in CF, as the cystic fibrosis transmembrane conductance regulator (CFTR) is the main mechanism of action. However, anecdotal clinical evidence suggests that linaclotide may be effective for treating some gastrointestinal symptoms in CF. The goal of this study was to determine the effectiveness and mechanism of linaclotide in treating CF gastrointestinal disorders using CF mouse models. Intestinal transit, chloride secretion, and intestinal lumen fluidity were assessed in wild-type and CF mouse models in response to linaclotide. CFTR and sodium/hydrogen exchanger 3 (NHE3) response to linaclotide was also evaluated. Linaclotide treatment improved intestinal transit in mice carrying either F508del or null Cftr mutations but did not induce detectable Cl- secretion. Linaclotide increased fluid retention and fluidity of CF intestinal contents, suggesting inhibition of fluid absorption. Targeted inhibition of sodium absorption by the NHE3 inhibitor tenapanor produced improvements in gastrointestinal transit similar to those produced by linaclotide treatment, suggesting that inhibition of fluid absorption by linaclotide contributes to improved gastrointestinal transit in CF. Our results demonstrate that linaclotide improves gastrointestinal transit in CF mouse models by increasing luminal fluidity through inhibiting NHE3-mediated sodium absorption. Further studies are necessary to assess whether linaclotide could improve CF intestinal pathologies in patients. GCC signaling and NHE3 inhibition may be therapeutic targets for CF intestinal manifestations. NEW & NOTEWORTHY Linaclotide's primary mechanism of action in alleviating chronic constipation is through cystic fibrosis transmembrane conductance regulator (CFTR), negating its use in patients with cystic fibrosis (CF). For the first time, our findings suggest that in the absence of CFTR, linaclotide can improve fluidity of the intestinal lumen through the inhibition of sodium/hydrogen exchanger 3. These findings suggest that linaclotide could improve CF intestinal pathologies in patients.
Collapse
Affiliation(s)
- Daniel R. McHugh
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Calvin U. Cotton
- 2Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio,3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Fraser J. Moss
- 2Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Megan Vitko
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Dana M. Valerio
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Thomas J. Kelley
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio,4Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Shuyu Hao
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Anjum Jafri
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Mitchell L. Drumm
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio,3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Walter F. Boron
- 2Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio,5Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio,6Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Robert C. Stern
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio,7Rainbow Babies and Children’s Hospital, Cleveland, Ohio
| | - Kimberly McBennett
- 3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio,7Rainbow Babies and Children’s Hospital, Cleveland, Ohio
| | - Craig A. Hodges
- 1Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio,3Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
33
|
Lu YY, Liang J, Chen SX, Wang BX, Yuan H, Li CT, Wu YY, Wu YF, Shi XG, Gao J, Hou SZ. Phloridzin alleviate colitis in mice by protecting the intestinal brush border and improving the expression of sodium glycogen transporter 1. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
34
|
Abstract
Inflammatory bowel diseases broadly categorized into Crohn's disease (CD) and ulcerative colitis (UC), are chronic inflammatory disorders of the gastrointestinal tract with increasing prevalence worldwide. The etiology of the disease is complex and involves a combination of genetic, environmental, immunological and gut microbial factors. Recurring and bloody diarrhea is the most prevalent and debilitating symptom in IBD. The pathogenesis of IBD-associated diarrhea is multifactorial and is essentially an outcome of mucosal damage caused by persistent inflammation resulting in dysregulated intestinal ion transport, impaired epithelial barrier function and increased accessibility of the pathogens to the intestinal mucosa. Altered expression and/or function of epithelial ion transporters and channels is the principle cause of electrolyte retention and water accumulation in the intestinal lumen leading to diarrhea in IBD. Aberrant barrier function further contributes to diarrhea via leak-flux mechanism. Mucosal penetration of enteric pathogens promotes dysbiosis and exacerbates the underlying immune system further perpetuating IBD associated-tissue damage and diarrhea. Here, we review the mechanisms of impaired ion transport and loss of epithelial barrier function contributing to diarrhea associated with IBD.
Collapse
Affiliation(s)
- Arivarasu N Anbazhagan
- a Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago , IL , USA
| | - Shubha Priyamvada
- a Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago , IL , USA
| | - Waddah A Alrefai
- a Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago , IL , USA.,b Jesse Brown VA Medical Center , Chicago , IL , USA
| | - Pradeep K Dudeja
- a Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago , IL , USA.,b Jesse Brown VA Medical Center , Chicago , IL , USA
| |
Collapse
|
35
|
Xu H, Ghishan FK, Kiela PR. SLC9 Gene Family: Function, Expression, and Regulation. Compr Physiol 2018; 8:555-583. [PMID: 29687889 DOI: 10.1002/cphy.c170027] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The Slc9 family of Na+ /H+ exchangers (NHEs) plays a critical role in electroneutral exchange of Na+ and H+ in the mammalian intestine as well as other absorptive and secretory epithelia of digestive organs. These transport proteins contribute to the transepithelial Na+ and water absorption, intracellular pH and cellular volume regulation as well as the electrolyte, acid-base, and fluid volume homeostasis at the systemic level. They also influence the function of other membrane transport mechanisms, affect cellular proliferation and apoptosis as well as cell migration, adherence to the extracellular matrix, and tissue repair. Additionally, they modulate the extracellular milieu to facilitate other nutrient absorption and to regulate the intestinal microbial microenvironment. Na+ /H+ exchange is inhibited in selected gastrointestinal diseases, either by intrinsic factors (e.g., bile acids, inflammatory mediators) or infectious agents and associated bacterial toxins. Disrupted NHE activity may contribute not only to local and systemic electrolyte imbalance but also to the disease severity via multiple mechanisms. In this review, we describe the cation proton antiporter superfamily of Na+ /H+ exchangers with a particular emphasis on the eight SLC9A isoforms found in the digestive tract, followed by a more integrative description in their roles in each of the digestive organs. We discuss regulatory mechanisms that determine the function of Na+ /H+ exchangers as pertinent to the digestive tract, their regulation in pathological states of the digestive organs, and reciprocally, the contribution of dysregulated Na+ /H+ exchange to the disease pathogenesis and progression. © 2018 American Physiological Society. Compr Physiol 8:555-583, 2018.
Collapse
Affiliation(s)
- Hua Xu
- Department of Pediatrics, Steele Children's Research Center, University of Arizona, Tucson, Arizona, USA
| | - Fayez K Ghishan
- Department of Pediatrics, Steele Children's Research Center, University of Arizona, Tucson, Arizona, USA
| | - Pawel R Kiela
- Department of Pediatrics, Steele Children's Research Center, University of Arizona, Tucson, Arizona, USA.,Department of Immunobiology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
36
|
Das S, Jayaratne R, Barrett KE. The Role of Ion Transporters in the Pathophysiology of Infectious Diarrhea. Cell Mol Gastroenterol Hepatol 2018; 6:33-45. [PMID: 29928670 PMCID: PMC6007821 DOI: 10.1016/j.jcmgh.2018.02.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/26/2018] [Indexed: 12/12/2022]
Abstract
Every year, enteric infections and associated diarrhea kill millions of people. The situation is compounded by increases in the number of enteric pathogens that are acquiring resistance to antibiotics, as well as (hitherto) a relative paucity of information on host molecular targets that may contribute to diarrhea. Many forms of diarrheal disease depend on the dysregulation of intestinal ion transporters, and an associated imbalance between secretory and absorptive functions of the intestinal epithelium. A number of major transporters have been implicated in the pathogenesis of diarrheal diseases and thus an understanding of their expression, localization, and regulation after infection with various bacteria, viruses, and protozoa likely will prove critical in designing new therapies. This article surveys our understanding of transporters that are modulated by specific pathogens and the mechanism(s) involved, thereby illuminating targets that might be exploited for new therapeutic approaches.
Collapse
Key Words
- ATP, adenosine triphosphate
- ATPase, adenosine triphosphatase
- CDI, Clostridium difficile infection
- CFTR, cystic fibrosis transmembrane conductance regulator
- CLCA1, chloride channel accessory 1
- CT, cholera toxin
- CXCR2, C-X-C motif chemokine receptor 2
- DRA, down-regulated in adenoma
- Diarrhea
- ENaC, epithelial sodium channel
- EPEC, enteropathogenic Escherichia coli
- ETEC, enterotoxigenic Escherichia coli
- Enteric Pathogen
- Epithelium
- EspG, Escherichia coli secreted protein G
- GPR39, G-protein coupled receptor 39
- Ion Transport
- KCC, potassium-chloride cotransporter
- LPA, lysophosphatidic acid
- LT, heat-labile toxin
- NHE, sodium/hydrogen exchanger
- NHERF2, sodium/hydrogen exchanger regulatory factor 2
- NKCC, sodium-potassium-2 chloride cotransporter
- ORT, oral rehydration therapy
- PKC, protein kinase C
- SGLT1, sodium-glucose cotransporter 1
- SLC, solute carrier
- ST, heat-stabile toxin
- TNF, tumor necrosis factor
- Tcd, Clostridium difficile toxin
- ZnR, zinc sensing receptor
- cAMP, adenosine 3′,5′-cyclic monophosphate
Collapse
Affiliation(s)
- Soumita Das
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, California
| | - Rashini Jayaratne
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, California
| | - Kim E. Barrett
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, California,Correspondence Address correspondence to: Kim E. Barrett, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0063. fax: (858) 246-1788.
| |
Collapse
|
37
|
Sircana A, De Michieli F, Parente R, Framarin L, Leone N, Berrutti M, Paschetta E, Bongiovanni D, Musso G. Gut microbiota, hypertension and chronic kidney disease: Recent advances. Pharmacol Res 2018; 144:390-408. [PMID: 29378252 DOI: 10.1016/j.phrs.2018.01.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/29/2017] [Accepted: 01/22/2018] [Indexed: 02/07/2023]
Abstract
A large number of different microbial species populates intestine. Extensive research has studied the entire microbial population and their genes (microbiome) by using metagenomics, metatranscriptomics and metabolomic analysis. Studies suggest that the imbalances of the microbial community causes alterations in the intestinal homeostasis, leading to repercussions on other systems: metabolic, nervous, cardiovascular, immune. These studies have also shown that alterations in the structure and function of the gut microbiota play a key role in the pathogenesis and complications of Hypertension (HTN) and Chronic Kidney Disease (CKD). Increased blood pressure (BP) and CKD are two leading risk factors for cardiovascular disease and their treatment represents a challenge for the clinicians. In this Review, we discuss mechanisms whereby gut microbiota (GM) and its metabolites act on downstream cellular targets to contribute to the pathogenesis of HTN and CKD, and potential therapeutic implications.
Collapse
Affiliation(s)
- Antonio Sircana
- Unità Operativa di Cardiologia, Azienda Ospedaliero Universitaria, Sassari, Italy; Department of Medical Sciences, San Giovanni Battista Hospital, Turin, Italy
| | - Franco De Michieli
- HUMANITAS Gradenigo, University of Turin, Turin, Italy; Department of Medical Sciences, San Giovanni Battista Hospital, Turin, Italy
| | - Renato Parente
- HUMANITAS Gradenigo, University of Turin, Turin, Italy; Department of Medical Sciences, San Giovanni Battista Hospital, Turin, Italy
| | - Luciana Framarin
- HUMANITAS Gradenigo, University of Turin, Turin, Italy; Department of Medical Sciences, San Giovanni Battista Hospital, Turin, Italy
| | - Nicola Leone
- HUMANITAS Gradenigo, University of Turin, Turin, Italy; Department of Medical Sciences, San Giovanni Battista Hospital, Turin, Italy
| | - Mara Berrutti
- HUMANITAS Gradenigo, University of Turin, Turin, Italy; Department of Medical Sciences, San Giovanni Battista Hospital, Turin, Italy
| | - Elena Paschetta
- HUMANITAS Gradenigo, University of Turin, Turin, Italy; Department of Medical Sciences, San Giovanni Battista Hospital, Turin, Italy
| | - Daria Bongiovanni
- HUMANITAS Gradenigo, University of Turin, Turin, Italy; Department of Medical Sciences, San Giovanni Battista Hospital, Turin, Italy
| | - Giovanni Musso
- HUMANITAS Gradenigo, University of Turin, Turin, Italy; Department of Medical Sciences, San Giovanni Battista Hospital, Turin, Italy.
| |
Collapse
|
38
|
Visceral Congestion in Heart Failure: Right Ventricular Dysfunction, Splanchnic Hemodynamics, and the Intestinal Microenvironment. Curr Heart Fail Rep 2018; 14:519-528. [PMID: 29075956 DOI: 10.1007/s11897-017-0370-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Visceral venous congestion of the gut may play a key role in the pathogenesis of right-sided heart failure (HF) and cardiorenal syndromes. Here, we review the role of right ventricular (RV) dysfunction, visceral congestion, splanchnic hemodynamics, and the intestinal microenvironment in the setting of right-sided HF. We review recent literature on this topic, outline possible mechanisms of disease pathogenesis, and discuss potential therapeutics. RECENT FINDINGS There are several mechanisms linking RV-gut interactions via visceral venous congestion which could result in (1) hypoxia and acidosis in enterocytes, which may lead to enhanced sodium-hydrogen exchanger 3 (NHE3) expression with increased sodium and fluid retention; (2) decreased luminal pH in the intestines, which could lead to alteration of the gut microbiome which could increase gut permeability and inflammation; (3) alteration of renal hemodynamics with triggering of the cardiorenal syndrome; and (4) altered phosphate metabolism resulting in increased pulmonary artery stiffening, thereby increasing RV afterload. A wide variety of therapeutic interventions that act on the RV, pulmonary vasculature, intestinal microenvironment, and the kidney could alter these pathways and should be tested in patients with right-sided HF. The RV-gut axis is an important aspect of HF pathogenesis that deserves more attention. Modulation of the pathways interconnecting the right heart, visceral congestion, and the intestinal microenvironment could be a novel avenue of intervention for right-sided HF.
Collapse
|
39
|
Xiong R, Li W, Li Y, Zheng K, Zhang T, Gao M, Li Y, Hu L, Hu C. Er Shen Wan extract reduces diarrhea and regulates AQP 4 and NHE 3 in a rat model of spleen-kidney Yang deficiency-induced diarrhea. Biomed Pharmacother 2018; 98:834-846. [PMID: 29571254 DOI: 10.1016/j.biopha.2018.01.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/02/2018] [Accepted: 01/03/2018] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Er Shen Wan (ESW), a traditional Chinese medicinal formula comprised of Psoraleae Fructus (Babchi seeds, from Psoralea corylifolia Linn.) and Myristicae Semen (Nutmeg, from Myristica fragrans Houtt.), is widely used to treat spleen-kidney Yang deficiency (SKYD)-induced diarrhea. Previous studies have demonstrated preliminarily that the petroleum ether extract of ESW (ESWP) exhibits significant anti-diarrheal activity. The present study aimed to evaluate the anti-diarrhea activity of ESWP and to explore the underlying mechanisms with respect to fluid metabolism in a rat model of SKYD-induced diarrhea. MATERIALS AND METHODS A high-performance liquid chromatography-diode array detector (HPLC-DAD) approach was developed and validated for qualitative and quantitative analyses of the main constituents of ESWP. SKYD model rats were established and treated with an effective dose (3.5?g/kg) of the extract for two weeks. Anti-diarrheal activity and stool properties were observed. After the experiment, the appearance and histology of the intestines were evaluated. Serum levels of neuropeptide Y (NPY) and vasoactive intestinal peptide (VIP) were also determined. Furthermore, to characterize the regulation of aquaporin-4 (AQP 4) and Na+/H+ exchanger isoform 3 (NHE 3) in the colon, quantitative real-time RT-PCR (qRT-PCR), immunohistochemistry (IHC) and Western blotting (WB) were employed to detect mRNA and protein expression levels. RESULTS In the rat models, oral ESWP administration significantly reduced the diarrhea score and the number and weight of wet stools. Jejunal and ileac histological damage was impeded, and the histology score decreased. Serum VIP levels were significantly decreased, in contrast to NPY levels. In addition, AQP 4 and NHE 3 expression levels increased significantly. CONCLUSIONS These results showed that ESWP's anti-diarrheal effect might at least partially involve the regulation of hormones intimately involved in maintaining fluid and electrolyte levels, as well as by increasing AQP 4 and NHE 3 expression levels and enhancing the absorption of Na+ and water.
Collapse
Affiliation(s)
- Rui Xiong
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China
| | - Wenbing Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China; Sichuan Neo-Green Pharmaceutical Technology Development Co., Ltd., Chengdu, 610081, PR China
| | - Yidan Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China
| | - Kaixuan Zheng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China
| | - Tingting Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China
| | - Mingyang Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China
| | - Yun Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China; College of Pharmacy, Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, PR China
| | - Lin Hu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China
| | - Changjiang Hu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China; Sichuan Neo-Green Pharmaceutical Technology Development Co., Ltd., Chengdu, 610081, PR China.
| |
Collapse
|
40
|
Tapia R, Kralicek SE, Hecht GA. EPEC effector EspF promotes Crumbs3 endocytosis and disrupts epithelial cell polarity. Cell Microbiol 2017; 19. [PMID: 28618099 DOI: 10.1111/cmi.12757] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/19/2017] [Accepted: 06/09/2017] [Indexed: 12/12/2022]
Abstract
Enteropathogenic Escherichia coli (EPEC) uses a type III secretion system to inject effector proteins into host intestinal epithelial cells causing diarrhoea. EPEC infection redistributes basolateral proteins β1-integrin and Na+ /K+ ATPase to the apical membrane of host cells. The Crumbs (Crb) polarity complex (Crb3/Pals1/Patj) is essential for epithelial cell polarisation and tight junction (TJ) assembly. Here, we demonstrate that EPEC displaces Crb3 and Pals1 from the apical membrane to the cytoplasm of cultured intestinal epithelial cells and colonocytes of infected mice. In vitro studies show that EspF, but not Map, alters Crb3, whereas both effectors modulate Pals1. EspF perturbs polarity formation in cyst morphogenesis assays and induces endocytosis and apical redistribution of Na+ /K+ ATPase. EspF binds to sorting nexin 9 (SNX9) causing membrane remodelling in host cells. Infection with ΔespF/pespFD3, a mutant strain that ablates EspF binding to SNX9, or inhibition of dynamin, attenuates Crb3 endocytosis caused by EPEC. In addition, infection with ΔespF/pespFD3 has no impact on Na+ /K+ ATPase endocytosis. These data support the hypothesis that EPEC perturbs apical-basal polarity in an EspF-dependent manner, which would contribute to EPEC-associated diarrhoea by disruption of TJ and altering the crucial positioning of membrane transporters involved in the absorption of ions and solutes.
Collapse
Affiliation(s)
- Rocio Tapia
- Department of Medicine and Division of Gastroenterology and Nutrition, Loyola University Chicago, Chicago, IL, USA
| | - Sarah E Kralicek
- Department of Medicine and Division of Gastroenterology and Nutrition, Loyola University Chicago, Chicago, IL, USA
| | - Gail A Hecht
- Department of Medicine and Division of Gastroenterology and Nutrition, Loyola University Chicago, Chicago, IL, USA.,Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA.,Edward Hines Jr. VA Hospital, Hines, IL, USA
| |
Collapse
|
41
|
Soleiman AA, Thameem F, Khan I. Mechanism of down regulation of Na-H exchanger-2 in experimental colitis. PLoS One 2017; 12:e0176767. [PMID: 28493993 PMCID: PMC5426621 DOI: 10.1371/journal.pone.0176767] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/17/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The Na-H exchanger [NHE] performs an electroneutral uptake of NaCl and water from the lumen of the gastrointestinal tract. There are several distinct NHE isoforms, some of which show an altered expression in the inflammatory bowel diseases (IBD). In this study, we examined a role of NHE-2 in experimental colitis. METHODS Colitis was induced in male Sprague-Dawley rats by intra-rectal administration of trinitrobenzenesulphonic acid (TNBS). On day 6 post-TNBS, the animals were sacrificed, colonic and ileal segments were taken out, cleaned with phosphate buffered saline and used in this study. RESULTS There was a significant decrease in the level of NHE-2 protein as measured by ECL western blot analysis and confocal immunofluorescence microscopy. The levels of NHE-2 mRNA and heteronuclear RNA measured by an end-point RT-PCR and a real time PCR were also decreased significantly in the inflamed colon. However, there was no change in the level of NHE-2 protein in response to in vitro TNF-α treatment of uninflamed rat colonic segment. These changes were selective and localized to the colon as actin, an internal control, remained unchanged. Confocal immunofluorescence microscopy revealed co-localization of NHE-2 and NHE-3 in the brush borders of colonic epithelial cells. Inflamed colon showed a significant increase in myeloperoxidase activity and colon hypertrophy. In addition, there was a significant decrease in body weight and goblet cells' mucin staining in the TNBS treated colon. These changes were not conspicuous in the non-inflamed ileum. CONCLUSIONS These findings demonstrate suppression of NHE-2 expression on the brush borders in the colonic epithelial cells which is regulated transcriptionally. However a role of TNF-α in the regulation of NHE-2 is discounted in the present model of colitis. This decrease in the NHE-2 expression will lead to a loss of electrolyte and water uptake thus contributing to the symptoms associated with IBD.
Collapse
Affiliation(s)
- Amal Ali Soleiman
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Jabriya, Kuwait
| | - Farook Thameem
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Jabriya, Kuwait
| | - Islam Khan
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Jabriya, Kuwait
- * E-mail:
| |
Collapse
|
42
|
Fenton RA, Poulsen SB, de la Mora Chavez S, Soleimani M, Dominguez Rieg JA, Rieg T. Renal tubular NHE3 is required in the maintenance of water and sodium chloride homeostasis. Kidney Int 2017; 92:397-414. [PMID: 28385297 DOI: 10.1016/j.kint.2017.02.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 01/25/2017] [Accepted: 02/02/2017] [Indexed: 11/30/2022]
Abstract
The sodium/proton exchanger isoform 3 (NHE3) is expressed in the intestine and the kidney, where it facilitates sodium (re)absorption and proton secretion. The importance of NHE3 in the kidney for sodium chloride homeostasis, relative to the intestine, is unknown. Constitutive tubule-specific NHE3 knockout mice (NHE3loxloxCre) did not show significant differences compared to control mice in body weight, blood pH or bicarbonate and plasma sodium, potassium, or aldosterone levels. Fluid intake, urinary flow rate, urinary sodium/creatinine, and pH were significantly elevated in NHE3loxloxCre mice, while urine osmolality and GFR were significantly lower. Water deprivation revealed a small urinary concentrating defect in NHE3loxloxCre mice on a control diet, exaggerated on low sodium chloride. Ten days of low or high sodium chloride diet did not affect plasma sodium in control mice; however, NHE3loxloxCre mice were susceptible to low sodium chloride (about -4 mM) or high sodium chloride intake (about +2 mM) versus baseline, effects without differences in plasma aldosterone between groups. Blood pressure was significantly lower in NHE3loxloxCre mice and was sodium chloride sensitive. In control mice, the expression of the sodium/phosphate co-transporter Npt2c was sodium chloride sensitive. However, lack of tubular NHE3 blunted Npt2c expression. Alterations in the abundances of sodium/chloride cotransporter and its phosphorylation at threonine 58 as well as the abundances of the α-subunit of the epithelial sodium channel, and its cleaved form, were also apparent in NHE3loxloxCre mice. Thus, renal NHE3 is required to maintain blood pressure and steady-state plasma sodium levels when dietary sodium chloride intake is modified.
Collapse
Affiliation(s)
- Robert A Fenton
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Søren B Poulsen
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark; VA San Diego Healthcare System, San Diego, California, USA
| | | | - Manoocher Soleimani
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio, USA; Research Services, Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Jessica A Dominguez Rieg
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Timo Rieg
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA.
| |
Collapse
|
43
|
Afsar B, Vaziri ND, Aslan G, Tarim K, Kanbay M. Gut hormones and gut microbiota: implications for kidney function and hypertension. ACTA ACUST UNITED AC 2016; 10:954-961. [PMID: 27865823 DOI: 10.1016/j.jash.2016.10.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
Abstract
Increased blood pressure (BP) and chronic kidney disease are two leading risk factors for cardiovascular disease. Increased sodium intake is one of the most important risk factors for development of hypertension. Recent data have shown that gut influences kidney function and BP by variety of mechanisms. Various hormones and peptides secreted from gut such as gastrin, glucocorticoids, Glucagon-like peptide-1 impact on kidney function and BP especially influencing sodium absorption from gut. These findings stimulate scientist to find new therapeutic options such as tenapanor for treatment of hypertension by blocking sodium absorption from gut. The gastrointestinal tract is also occupied by a huge community of microbes (microbiome) that under normal condition has a symbiotic relationship with the host. Alterations in the structure and function of the gut microbiota have been shown to play a key role in the pathogenesis and complications of numerous diseases including hypertension. Based on these data, in this review, we provide a summary of the available data on the role of gut and gut microbiota in regulation of BP and kidney function.
Collapse
Affiliation(s)
- Baris Afsar
- Division of Nephrology, Department of Medicine, Konya Numune State Hospital, Konya, Turkey
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, Department of Medicine, Schools of Medicine and Biological Science, University of California, Irvine, CA, USA
| | - Gamze Aslan
- Department of Cardiology, Koc University School of Medicine, Istanbul, Turkey
| | - Kayhan Tarim
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
44
|
Shawki A, Engevik MA, Kim RS, Knight PB, Baik RA, Anthony SR, Worrell RT, Shull GE, Mackenzie B. Intestinal brush-border Na+/H+ exchanger-3 drives H+-coupled iron absorption in the mouse. Am J Physiol Gastrointest Liver Physiol 2016; 311:G423-30. [PMID: 27390324 PMCID: PMC5076011 DOI: 10.1152/ajpgi.00167.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/29/2016] [Indexed: 01/31/2023]
Abstract
Divalent metal-ion transporter-1 (DMT1), the principal mechanism by which nonheme iron is taken up at the intestinal brush border, is energized by the H(+)-electrochemical potential gradient. The provenance of the H(+) gradient in vivo is unknown, so we have explored a role for brush-border Na(+)/H(+) exchanger (NHE) isoforms by examining iron homeostasis and intestinal iron handling in mice lacking NHE2 or NHE3. We observed modestly depleted liver iron stores in NHE2-null (NHE2(-/-)) mice stressed on a low-iron diet but no change in hematological or blood iron variables or the expression of genes associated with iron metabolism compared with wild-type mice. Ablation of NHE3 strongly depleted liver iron stores, regardless of diet. We observed decreases in blood iron variables but no overt anemia in NHE3-null (NHE3(-/-)) mice on a low-iron diet. Intestinal expression of DMT1, the apical surface ferrireductase cytochrome b reductase-1, and the basolateral iron exporter ferroportin was upregulated in NHE3(-/-) mice, and expression of liver Hamp1 (hepcidin) was suppressed compared with wild-type mice. Absorption of (59)Fe from an oral dose was substantially impaired in NHE3(-/-) compared with wild-type mice. Our data point to an important role for NHE3 in generating the H(+) gradient that drives DMT1-mediated iron uptake at the intestinal brush border.
Collapse
Affiliation(s)
- Ali Shawki
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio; Systems Biology and Physiology Program, University of Cincinnati College of Medicine, Cincinnati, Ohio; and
| | - Melinda A Engevik
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio; Systems Biology and Physiology Program, University of Cincinnati College of Medicine, Cincinnati, Ohio; and
| | - Robert S Kim
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Patrick B Knight
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Rusty A Baik
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Sarah R Anthony
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Roger T Worrell
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio; Systems Biology and Physiology Program, University of Cincinnati College of Medicine, Cincinnati, Ohio; and
| | - Gary E Shull
- Systems Biology and Physiology Program, University of Cincinnati College of Medicine, Cincinnati, Ohio; and Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Bryan Mackenzie
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio; Systems Biology and Physiology Program, University of Cincinnati College of Medicine, Cincinnati, Ohio; and
| |
Collapse
|
45
|
Asghar MN, Priyamvada S, Nyström JH, Anbazhagan AN, Dudeja PK, Toivola DM. Keratin 8 knockdown leads to loss of the chloride transporter DRA in the colon. Am J Physiol Gastrointest Liver Physiol 2016; 310:G1147-54. [PMID: 27125276 PMCID: PMC4935477 DOI: 10.1152/ajpgi.00354.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 04/23/2016] [Indexed: 02/06/2023]
Abstract
Keratins (K) are intermediate filament proteins important in protection from stress. The roles of keratins in the intestine are not clear, but K8 knockout (K8(-/-)) mice develop a Th2-type colonic inflammation, epithelial hyperproliferation, and mild diarrhea caused by a keratin level-dependent decrease in short-circuit current and net sodium and chloride absorption in the distal colon. The lack of K8 leads to mistargeting or altered levels of membrane proteins in colonocytes; however, the main transporter responsible for the keratin-related ion transport defect is unknown. We here analyzed protein and mRNA levels of candidate ion transporters CFTR, PAT-1, NHE-3, and DRA in ileum, cecum, and proximal and distal colon. Although no differences were observed for CFTR, PAT-1, or NHE-3, DRA mRNA levels were decreased by three- to fourfold and DRA protein was almost entirely lost in K8(-/-) cecum and proximal and distal colon compared with K8(+/+), whereas the levels in ileum were normal. In K8(+/-) mice, DRA mRNA levels were unaltered, while decreased DRA protein levels were detected in the proximal colon. Immunofluorescence staining confirmed the loss of DRA in K8(-/-) distal colon, while K8(+/-) displayed a similar but more patchy apical DRA distribution compared with K8(+/+) DRA was similarly decreased when K8 was knocked down in Caco-2 cells, confirming that K8 levels modulate DRA levels in an inflammation-independent manner. Taken together, the loss of DRA in the K8(-/-) mouse colon and cecum explains the dramatic chloride transport defect and diarrheal phenotype after K8 inactivation and identifies K8 as a novel regulator of DRA.
Collapse
Affiliation(s)
- M. Nadeem Asghar
- 1Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University and Turku Center for Disease Modeling, University of Turku, Turku, Finland; and
| | - Shubha Priyamvada
- 2University of Illinois at Chicago, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Joel H. Nyström
- 1Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University and Turku Center for Disease Modeling, University of Turku, Turku, Finland; and
| | | | - Pradeep K. Dudeja
- 2University of Illinois at Chicago, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Diana M. Toivola
- 1Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University and Turku Center for Disease Modeling, University of Turku, Turku, Finland; and
| |
Collapse
|
46
|
Priyamvada S, Gomes R, Gill RK, Saksena S, Alrefai WA, Dudeja PK. Mechanisms Underlying Dysregulation of Electrolyte Absorption in Inflammatory Bowel Disease-Associated Diarrhea. Inflamm Bowel Dis 2015; 21:2926-35. [PMID: 26595422 PMCID: PMC4662046 DOI: 10.1097/mib.0000000000000504] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Inflammatory bowel diseases (IBDs), including Crohn's disease and ulcerative colitis, are chronic relapsing inflammatory disorders of the gastrointestinal tract. Chronic inflammation of the intestine affects the normal fluid and electrolyte absorption leading to diarrhea, the hallmark symptom of IBD. The management of IBD-associated diarrhea still remains to be a challenge, and extensive studies over the last 2 decades have focused on investigating the molecular mechanisms underlying IBD-associated diarrhea. These studies have shown that the predominant mechanism of diarrhea in IBD involves impairment of electroneutral NaCl absorption, with very little role if any played by anion secretion. The electroneutral NaCl absorption involves coupled operation of Na/H exchanger 3 (NHE3 or SLC9A3) and Cl/HCO3 exchanger DRA (Down Regulated in Adenoma, or SLC26A3). Increasing evidence now supports the critical role of a marked decrease in NHE3 and DRA function and/or expression in IBD-associated diarrhea. This review provides a detailed analysis of the current knowledge related to alterations in NHE3 and DRA function and expression in IBD including the mechanisms underlying these observations and highlights the potential of these transporters as important and novel therapeutic targets.
Collapse
Affiliation(s)
- Shubha Priyamvada
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Rochelle Gomes
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Ravinder K. Gill
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Seema Saksena
- Jesse Brown VA Medical Center, University of Illinois at Chicago, Chicago, IL
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Waddah A. Alrefai
- Jesse Brown VA Medical Center, University of Illinois at Chicago, Chicago, IL
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Pradeep K. Dudeja
- Jesse Brown VA Medical Center, University of Illinois at Chicago, Chicago, IL
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
47
|
Bile acids inhibit Na⁺/H⁺ exchanger and Cl⁻/HCO₃⁻ exchanger activities via cellular energy breakdown and Ca²⁺ overload in human colonic crypts. Pflugers Arch 2015; 467:1277-90. [PMID: 25015415 DOI: 10.1007/s00424-014-1560-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 06/17/2014] [Indexed: 12/17/2022]
Abstract
Bile acids play important physiological role in the solubilisation and absorption of dietary lipids. However, under pathophysiological conditions, such as short bowel syndrome, they can reach the colon in high concentrations inducing diarrhoea. In this study, our aim was to characterise the cellular pathomechanism of bile-induced diarrhoea using human samples. Colonic crypts were isolated from biopsies of patients (controls with negative colonoscopic findings) and of cholecystectomised/ileum-resected patients with or without diarrhoea. In vitro measurement of the transporter activities revealed impaired Na⁺/H⁺ exchanger (NHE) and Cl⁻/HCO₃⁻ exchanger (CBE) activities in cholecystectomised/ileum-resected patients suffering from diarrhoea, compared to control patients. Acute treatment of colonic crypts with 0.3 mM chenodeoxycholate caused dose-dependent intracellular acidosis; moreover, the activities of acid/base transporters (NHE and CBE) were strongly impaired. This concentration of chenodeoxycholate did not cause morphological changes in colonic epithelial cells, although significantly reduced the intracellular ATP level, decreased mitochondrial transmembrane potential and caused sustained intracellular Ca²⁺ elevation. We also showed that chenodeoxycholate induced Ca²⁺ release from the endoplasmic reticulum and extracellular Ca²⁺ influx contributing to the Ca²⁺ elevation. Importantly, our results suggest that the chenodeoxycholate-induced inhibition of NHE activities was ATP-dependent, whereas the inhibition of CBE activity was mediated by the sustained Ca²⁺ elevation. We suggest that bile acids inhibit the function of ion transporters via cellular energy breakdown and Ca²⁺ overload in human colonic epithelial cells, which can reduce fluid and electrolyte absorption in the colon and promote the development of diarrhea.
Collapse
|
48
|
Yang J, Zhao X, Patel A, Potru R, Azizi-Ghannad S, Dolinger M, Cao J, Bartholomew C, Mazurkiewicz J, Conti D, Jones D, Huang Y, Zhu XC. Rapamycin Inhibition of mTOR Reduces Levels of the Na+/H+ Exchanger 3 in Intestines of Mice and Humans, Leading to Diarrhea. Gastroenterology 2015; 149:151-62. [PMID: 25836987 PMCID: PMC4849539 DOI: 10.1053/j.gastro.2015.03.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 03/11/2015] [Accepted: 03/25/2015] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS The immunosuppressant rapamycin frequently causes noninfectious diarrhea in organ transplant recipients. We investigated the mechanisms of this process. METHODS We performed a retrospective analysis of renal transplant recipients treated with rapamycin from 2003 through 2010 at Albany Medical College, collecting data on serum levels of rapamycin. Levels of the Na+/H+ exchanger 3 (NHE3) were measured in human ileal biopsy specimens from patients who did and did not receive rapamycin (controls), in ileum tissues from rats or mice given rapamycin, and in mice with intestine-specific disruption of mammalian target of rapamycin (Mtor) (mTOR(f/f):Villin-cre mice) or Atg7 (Atg7(flox/flox); Villin-Cre). Exchange activity and intestinal water absorption were measured using a pH-sensitive dye and small intestine perfusion, respectively. RESULTS Episodes of noninfectious diarrhea occurred in organ recipients after increases in serum levels of rapamycin. The expression of NHE3 was reduced in the ileal brush border of patients with diarrhea. In rats and mice, continuous administration of low doses of rapamycin reduced levels of NHE3 in intestinal tissues; this effect was not observed in mice with intestinal deletion of ATG7, indicating that autophagy is required for the reduction. Administration of single high doses of rapamycin to mice, to model the spikes in rapamycin levels that occur in patients with severe diarrheal episodes, resulted in reduced phosphorylation of S6 and AKT in ileal tissues, indicating inhibition of the mTOR complex (mTORC1 and mTORC2). The intestines of mice with intestine-specific deletion of mTOR were dilated and contained large amounts of liquid stools; they also had reduced levels of total NHE3 and NHERF1 compared with control mice. We observed a significant reduction in Na(+)/H(+) exchange activity in ileum tissues from these mice. CONCLUSIONS Rapamycin inhibition of mTOR reduces levels of NHE3 and Na(+)/H(+) exchange activity in intestinal tissues of patients and rodents. This process appears to require the autophagic activity mediated by ATG7. Loss of mTOR regulation of NHE3 could mediate the development of diarrhea in patients undergoing rapamycin therapy.
Collapse
Affiliation(s)
- Jun Yang
- Department of Medicine, Division of Gastroenterology and Hepatology, Albany Medical College, Albany, New York; Center of Cardiovascular Sciences, Albany Medical College, Albany, New York
| | - Xiaofeng Zhao
- Department of Medicine, Division of Gastroenterology and Hepatology, Albany Medical College, Albany, New York; Center of Cardiovascular Sciences, Albany Medical College, Albany, New York
| | - Archana Patel
- Department of Medicine, Division of Gastroenterology and Hepatology, Albany Medical College, Albany, New York
| | - Rachana Potru
- Department of Medicine, Division of Gastroenterology and Hepatology, Albany Medical College, Albany, New York
| | - Sadra Azizi-Ghannad
- Department of Medicine, Division of Gastroenterology and Hepatology, Albany Medical College, Albany, New York
| | - Michael Dolinger
- Department of Medicine, Division of Gastroenterology and Hepatology, Albany Medical College, Albany, New York
| | - James Cao
- Center of Cardiovascular Sciences, Albany Medical College, Albany, New York
| | - Catherine Bartholomew
- Department of Medicine, Division of Gastroenterology and Hepatology, Albany Medical College, Albany, New York
| | - Joseph Mazurkiewicz
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York
| | - David Conti
- Department of Transplant Surgery, Albany Medical College, Albany, New York
| | - David Jones
- Department of Pathology, Albany Medical College, Albany, New York
| | - Yunfei Huang
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York
| | - Xinjun Cindy Zhu
- Department of Medicine, Division of Gastroenterology and Hepatology, Albany Medical College, Albany, New York; Center of Cardiovascular Sciences, Albany Medical College, Albany, New York.
| |
Collapse
|
49
|
Krishnan D, Liu L, Wiebe SA, Casey JR, Cordat E, Alexander RT. Carbonic anhydrase II binds to and increases the activity of the epithelial sodium-proton exchanger, NHE3. Am J Physiol Renal Physiol 2015; 309:F383-92. [PMID: 26041446 DOI: 10.1152/ajprenal.00464.2014] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 05/20/2015] [Indexed: 02/08/2023] Open
Abstract
Two-thirds of sodium filtered by the renal glomerulus is reabsorbed from the proximal tubule via a sodium/proton exchanger isoform 3 (NHE3)-dependent mechanism. Since sodium and bicarbonate reabsorption are coupled, we postulated that the molecules involved in their reabsorption [NHE3 and carbonic anhydrase II (CAII)] might physically and functionally interact. Consistent with this, CAII and NHE3 were closely associated in a renal proximal tubular cell culture model as revealed by a proximity ligation assay. Direct physical interaction was confirmed in solid-phase binding assays with immobilized CAII and C-terminal NHE3 glutathione-S-transferase fusion constructs. To assess the effect of CAII on NHE3 function, we expressed NHE3 in a proximal tubule cell line and measured NHE3 activity as the rate of intracellular pH recovery, following an acid load. NHE3-expressing cells had a significantly greater rate of intracellular pH recovery than controls. Inhibition of endogenous CAII activity with acetazolamide significantly decreased NHE3 activity, indicating that CAII activates NHE3. To ascertain whether CAII binding per se activates NHE3, we expressed NHE3 with wild-type CAII, a catalytically inactive CAII mutant (CAII-V143Y), or a mutant unable to bind other transporters (CAII-HEX). NHE3 activity increased upon wild-type CAII coexpression, but not in the presence of the CAII V143Y or HEX mutant. Together these studies support an association between CAII and NHE3 that alters the transporter's activity.
Collapse
Affiliation(s)
- Devishree Krishnan
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; and
| | - Lei Liu
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Shane A Wiebe
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; and
| | - Joseph R Casey
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; and
| | - Emmanuelle Cordat
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; and
| | - R Todd Alexander
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; and Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
50
|
Engevik MA, Engevik KA, Yacyshyn MB, Wang J, Hassett DJ, Darien B, Yacyshyn BR, Worrell RT. Human Clostridium difficile infection: inhibition of NHE3 and microbiota profile. Am J Physiol Gastrointest Liver Physiol 2015; 308:G497-509. [PMID: 25552580 PMCID: PMC4422371 DOI: 10.1152/ajpgi.00090.2014] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Clostridium difficile infection (CDI) is principally responsible for hospital acquired, antibiotic-induced diarrhea and colitis and represents a significant financial burden on our healthcare system. Little is known about C. difficile proliferation requirements, and a better understanding of these parameters is critical for development of new therapeutic targets. In cell lines, C. difficile toxin B has been shown to inhibit Na(+)/H(+) exchanger 3 (NHE3) and loss of NHE3 in mice results in an altered intestinal environment coupled with a transformed gut microbiota composition. However, this has yet to be established in vivo in humans. We hypothesize that C. difficile toxin inhibits NHE3, resulting in alteration of the intestinal environment and gut microbiota. Our results demonstrate that CDI patient biopsy specimens have decreased NHE3 expression and CDI stool has elevated Na(+) and is more alkaline compared with stool from healthy individuals. CDI stool microbiota have increased Bacteroidetes and Proteobacteria and decreased Firmicutes phyla compared with healthy subjects. In vitro, C. difficile grows optimally in the presence of elevated Na(+) and alkaline pH, conditions that correlate to changes observed in CDI patients. To confirm that inhibition of NHE3 was specific to C. difficile, human intestinal organoids (HIOs) were injected with C. difficile or healthy and CDI stool supernatant. Injection of C. difficile and CDI stool decreased NHE3 mRNA and protein expression compared with healthy stool and control HIOs. Together these data demonstrate that C. difficile inhibits NHE3 in vivo, which creates an altered environment favored by C. difficile.
Collapse
Affiliation(s)
- Melinda A. Engevik
- 1Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Kristen A. Engevik
- 1Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Mary Beth Yacyshyn
- 3Department of Medicine Division of Digestive Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Jiang Wang
- 4Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Daniel J. Hassett
- 2Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Benjamin Darien
- 5Department of Animal Health and Biomedical Sciences, University Wisconsin, Madison, Wisconsin; and
| | - Bruce R. Yacyshyn
- 3Department of Medicine Division of Digestive Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio; ,6Digestive Health Center of Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Roger T. Worrell
- 1Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio; ,6Digestive Health Center of Cincinnati Children's Hospital, Cincinnati, Ohio
| |
Collapse
|