1
|
Liu Y, Reyes E, Castillo-Azofeifa D, Klein OD, Nystul T, Barber DL. Intracellular pH dynamics regulates intestinal stem cell lineage specification. Nat Commun 2023; 14:3745. [PMID: 37353491 PMCID: PMC10290085 DOI: 10.1038/s41467-023-39312-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 06/06/2023] [Indexed: 06/25/2023] Open
Abstract
Intracellular pH dynamics is increasingly recognized to regulate myriad cell behaviors. We report a finding that intracellular pH dynamics also regulates adult stem cell lineage specification. We identify an intracellular pH gradient in mouse small intestinal crypts, lowest in crypt stem cells and increasing along the crypt column. Disrupting this gradient by inhibiting H+ efflux by Na+/H+ exchanger 1 abolishes crypt budding and blocks differentiation of Paneth cells, which are rescued with exogenous WNT. Using single-cell RNA sequencing and lineage tracing we demonstrate that intracellular pH dynamics acts downstream of ATOH1, with increased pH promoting differentiation toward the secretory lineage. Our findings indicate that an increase in pH is required for the lineage specification that contributes to crypt maintenance, establishing a role for intracellular pH dynamics in cell fate decisions within an adult stem cell lineage.
Collapse
Affiliation(s)
- Yi Liu
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Efren Reyes
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, 94143, USA
| | - David Castillo-Azofeifa
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, 94143, USA
- Immunology Discovery, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Todd Nystul
- Departments of Anatomy, University of California San Francisco, San Francisco, CA, 94143, USA.
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
2
|
Salari A, Zhou K, Nikolovska K, Seidler U, Amiri M. Human Colonoid-Myofibroblast Coculture for Study of Apical Na +/H + Exchangers of the Lower Cryptal Neck Region. Int J Mol Sci 2023; 24:ijms24054266. [PMID: 36901695 PMCID: PMC10001859 DOI: 10.3390/ijms24054266] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
Cation and anion transport in the colonocyte apical membrane is highly spatially organized along the cryptal axis. Because of lack of experimental accessibility, information about the functionality of ion transporters in the colonocyte apical membrane in the lower part of the crypt is scarce. The aim of this study was to establish an in vitro model of the colonic lower crypt compartment, which expresses the transit amplifying/progenitor (TA/PE) cells, with accessibility of the apical membrane for functional study of lower crypt-expressed Na+/H+ exchangers (NHEs). Colonic crypts and myofibroblasts were isolated from human transverse colonic biopsies, expanded as three-dimensional (3D) colonoids and myofibroblast monolayers, and characterized. Filter-grown colonic myofibroblast-colonic epithelial cell (CM-CE) cocultures (myofibroblasts on the bottom of the transwell and colonocytes on the filter) were established. The expression pattern for ion transport/junctional/stem cell markers of the CM-CE monolayers was compared with that of nondifferentiated (EM) and differentiated (DM) colonoid monolayers. Fluorometric pHi measurements were performed to characterize apical NHEs. CM-CE cocultures displayed a rapid increase in transepithelial electrical resistance (TEER), paralleled by downregulation of claudin-2. They maintained proliferative activity and an expression pattern resembling TA/PE cells. The CM-CE monolayers displayed high apical Na+/H+ exchange activity, mediated to >80% by NHE2. Human colonoid-myofibroblast cocultures allow the study of ion transporters that are expressed in the apical membrane of the nondifferentiated colonocytes of the cryptal neck region. The NHE2 isoform is the predominant apical Na+/H+ exchanger in this epithelial compartment.
Collapse
Affiliation(s)
- Azam Salari
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Kunyan Zhou
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Department of Thyroid Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Katerina Nikolovska
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Ursula Seidler
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Correspondence: (U.S.); (M.A.); Tel.: +49-511-532-9427 (U.S.); Fax: +49-511-532-8428 (U.S.)
| | - Mahdi Amiri
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Correspondence: (U.S.); (M.A.); Tel.: +49-511-532-9427 (U.S.); Fax: +49-511-532-8428 (U.S.)
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Nikolovska K, Cao L, Hensel I, Di Stefano G, Seidler A, Zhou K, Qian J, Singh AK, Riederer B, Seidler U. Sodium/hydrogen-exchanger-2 modulates colonocyte lineage differentiation. Acta Physiol (Oxf) 2022; 234:e13774. [PMID: 34985202 DOI: 10.1111/apha.13774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/12/2021] [Accepted: 01/01/2022] [Indexed: 12/11/2022]
Abstract
AIM The sodium/hydrogen exchanger 2 (NHE2) is an intestinal acid extruder with crypt-predominant localization and unresolved physiological significance. Our aim was to decipher its role in colonic epithelial cell proliferation, differentiation and electrolyte transport. METHODS Alterations induced by NHE2-deficiency were addressed in murine nhe2-/- and nhe2+/+ colonic crypts and colonoids, and NHE2-knockdown and control Caco2Bbe cells using pH-fluorometry, gene expression analysis and immunofluorescence. RESULTS pHi -measurements along the colonic cryptal axis revealed significantly decreased intracellular pH (pHi ) in the middle segment of nhe2-/- compared to nhe2+/+ crypts. Increased Nhe2 mRNA expression was detected in murine colonoids in the transiently amplifying/progenitor cell stage (TA/PE). Lack of Nhe2 altered the differentiation programme of colonic epithelial cells with reduced expression of absorptive lineage markers alkaline phosphatase (iAlp), Slc26a3 and transcription factor hairy and enhancer-of-split 1 (Hes1), but increased expression of secretory lineage markers Mucin 2, trefoil factor 3 (Tff3), enteroendocrine marker chromogranin A and murine atonal homolog 1 (Math1). Enterocyte differentiation was found to be pHi dependent with acidic pHi reducing, and alkaline pHi stimulating the expression of enterocyte differentiation markers in Caco2Bbe cells. A thicker mucus layer, longer crypts and an expanded brush border membrane zone of sodium/hydrogen exchanger 3 (NHE3) abundance may explain the lack of inflammation and the normal fluid absorptive rate in nhe2-/- colon. CONCLUSIONS The results suggest that NHE2 expression is activated when colonocytes emerge from the stem cell niche. Its activity increases progenitor cell pHi and thereby supports absorptive enterocyte differentiation.
Collapse
Affiliation(s)
- Katerina Nikolovska
- Department of Gastroenterology, Hepatology and Endocrinology Hannover Medical School Hannover Germany
| | - Li Cao
- Department of Gastroenterology, Hepatology and Endocrinology Hannover Medical School Hannover Germany
- Department of Gastroenterology Tongji Hospital Huazhong University Wuhan China
| | - Inga Hensel
- Department of Gastroenterology, Hepatology and Endocrinology Hannover Medical School Hannover Germany
| | - Gabriella Di Stefano
- Department of Gastroenterology, Hepatology and Endocrinology Hannover Medical School Hannover Germany
| | - Anna Elisabeth Seidler
- Department of Gastroenterology, Hepatology and Endocrinology Hannover Medical School Hannover Germany
| | - Kunyan Zhou
- Department of Gastroenterology, Hepatology and Endocrinology Hannover Medical School Hannover Germany
| | - Jiajie Qian
- Department of Gastroenterology, Hepatology and Endocrinology Hannover Medical School Hannover Germany
- Department of Transplantation and Hepatobiliary Surgery First Affiliated Hospital of Zheijang University Hangzhou China
| | - Anurag Kumar Singh
- Department of Gastroenterology, Hepatology and Endocrinology Hannover Medical School Hannover Germany
- Department of Physiological Chemistry University of Halle Halle (Saale) Germany
| | - Brigitte Riederer
- Department of Gastroenterology, Hepatology and Endocrinology Hannover Medical School Hannover Germany
| | - Ursula Seidler
- Department of Gastroenterology, Hepatology and Endocrinology Hannover Medical School Hannover Germany
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Amiri M, Seidler UE, Nikolovska K. The Role of pH i in Intestinal Epithelial Proliferation-Transport Mechanisms, Regulatory Pathways, and Consequences. Front Cell Dev Biol 2021; 9:618135. [PMID: 33553180 PMCID: PMC7862550 DOI: 10.3389/fcell.2021.618135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/04/2021] [Indexed: 01/07/2023] Open
Abstract
During the maturation of intestinal epithelial cells along the crypt/surface axis, a multitude of acid/base transporters are differentially expressed in their apical and basolateral membranes, enabling processes of electrolyte, macromolecule, nutrient, acid/base and fluid secretion, and absorption. An intracellular pH (pHi)-gradient is generated along the epithelial crypt/surface axis, either as a consequence of the sum of the ion transport activities or as a distinctly regulated entity. While the role of pHi on proliferation, migration, and tumorigenesis has been explored in cancer cells for some time, emerging evidence suggests an important role of the pHi in the intestinal stem cells (ISCs) proliferative rate under physiological conditions. The present review highlights the current state of knowledge about the potential regulatory role of pHi on intestinal proliferation and differentiation.
Collapse
|
9
|
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
|
10
|
Pedersen SF, Counillon L. The SLC9A-C Mammalian Na +/H + Exchanger Family: Molecules, Mechanisms, and Physiology. Physiol Rev 2019; 99:2015-2113. [PMID: 31507243 DOI: 10.1152/physrev.00028.2018] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Na+/H+ exchangers play pivotal roles in the control of cell and tissue pH by mediating the electroneutral exchange of Na+ and H+ across cellular membranes. They belong to an ancient family of highly evolutionarily conserved proteins, and they play essential physiological roles in all phyla. In this review, we focus on the mammalian Na+/H+ exchangers (NHEs), the solute carrier (SLC) 9 family. This family of electroneutral transporters constitutes three branches: SLC9A, -B, and -C. Within these, each isoform exhibits distinct tissue expression profiles, regulation, and physiological roles. Some of these transporters are highly studied, with hundreds of original articles, and some are still only rudimentarily understood. In this review, we present and discuss the pioneering original work as well as the current state-of-the-art research on mammalian NHEs. We aim to provide the reader with a comprehensive view of core knowledge and recent insights into each family member, from gene organization over protein structure and regulation to physiological and pathophysiological roles. Particular attention is given to the integrated physiology of NHEs in the main organ systems. We provide several novel analyses and useful overviews, and we pinpoint main remaining enigmas, which we hope will inspire novel research on these highly versatile proteins.
Collapse
Affiliation(s)
- S F Pedersen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; and Université Côte d'Azur, CNRS, Laboratoire de Physiomédecine Moléculaire, LP2M, France, and Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France
| | - L Counillon
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; and Université Côte d'Azur, CNRS, Laboratoire de Physiomédecine Moléculaire, LP2M, France, and Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
Laczkó D, Rosztóczy A, Birkás K, Katona M, Rakonczay Z, Tiszlavicz L, Róka R, Wittmann T, Hegyi P, Venglovecz V. Role of ion transporters in the bile acid-induced esophageal injury. Am J Physiol Gastrointest Liver Physiol 2016; 311:G16-31. [PMID: 27198194 DOI: 10.1152/ajpgi.00159.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 04/20/2016] [Indexed: 01/31/2023]
Abstract
Barrett's esophagus (BE) is considered to be the most severe complication of gastro-esophageal reflux disease (GERD), in which the prolonged, repetitive episodes of combined acidic and biliary reflux result in the replacement of the squamous esophageal lining by columnar epithelium. Therefore, the acid-extruding mechanisms of esophageal epithelial cells (EECs) may play an important role in the defense. Our aim was to identify the presence of acid/base transporters on EECs and to investigate the effect of bile acids on their expressions and functions. Human EEC lines (CP-A and CP-D) were acutely exposed to bile acid cocktail (BAC) and the changes in intracellular pH (pHi) and Ca(2+) concentration ([Ca(2+)]i) were measured by microfluorometry. mRNA and protein expression of ion transporters was investigated by RT-PCR, Western blot, and immunohistochemistry. We have identified the presence of a Na(+)/H(+) exchanger (NHE), Na(+)/HCO3 (-) cotransporter (NBC), and a Cl(-)-dependent HCO3 (-) secretory mechanism in CP-A and CP-D cells. Acute administration of BAC stimulated HCO3 (-) secretion in both cell lines and the NHE activity in CP-D cells by an inositol triphosphate-dependent calcium release. Chronic administration of BAC to EECs increased the expression of ion transporters compared with nontreated cells. A similar expression pattern was observed in biopsy samples from BE compared with normal epithelium. We have shown that acute administration of bile acids differently alters ion transport mechanisms of EECs, whereas chronic exposure to bile acids increases the expression of acid/base transporters. We speculate that these adaptive processes of EECs represent an important mucosal defense against the bile acid-induced epithelial injury.
Collapse
Affiliation(s)
- Dorottya Laczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary; First Department of Medicine, University of Szeged, Szeged, Hungary
| | - András Rosztóczy
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Klaudia Birkás
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Máté Katona
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Zoltán Rakonczay
- First Department of Medicine, University of Szeged, Szeged, Hungary; Department of Pathophysiology, University of Szeged, Szeged, Hungary
| | | | - Richárd Róka
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Tibor Wittmann
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Péter Hegyi
- First Department of Medicine, University of Szeged, Szeged, Hungary; MTA-SZTE Translational Gastroenterology Research Group, University of Szeged, Szeged, Hungary; and Institute for Translational Medicine and First Department of Medicine, University of Pécs, Pécs, Hungary
| | - Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary;
| |
Collapse
|
15
|
Walker NM, Liu J, Stein SR, Stefanski CD, Strubberg AM, Clarke LL. Cellular chloride and bicarbonate retention alters intracellular pH regulation in Cftr KO crypt epithelium. Am J Physiol Gastrointest Liver Physiol 2016; 310:G70-80. [PMID: 26542396 PMCID: PMC4719062 DOI: 10.1152/ajpgi.00236.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/29/2015] [Indexed: 01/31/2023]
Abstract
Cystic fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator (CFTR), an anion channel providing a major pathway for Cl(-) and HCO3 (-) efflux across the apical membrane of the epithelium. In the intestine, CF manifests as obstructive syndromes, dysbiosis, inflammation, and an increased risk for gastrointestinal cancer. Cftr knockout (KO) mice recapitulate CF intestinal disease, including intestinal hyperproliferation. Previous studies using Cftr KO intestinal organoids (enteroids) indicate that crypt epithelium maintains an alkaline intracellular pH (pHi). We hypothesized that Cftr has a cell-autonomous role in downregulating pHi that is incompletely compensated by acid-base regulation in its absence. Here, 2',7'-bis(2-carboxyethyl)-5(6)-carboxyfluorescein microfluorimetry of enteroids showed that Cftr KO crypt epithelium sustains an alkaline pHi and resistance to cell acidification relative to wild-type. Quantitative real-time PCR revealed that Cftr KO enteroids exhibit downregulated transcription of base (HCO3 (-))-loading proteins and upregulation of the basolateral membrane HCO3 (-)-unloader anion exchanger 2 (Ae2). Although Cftr KO crypt epithelium had increased Ae2 expression and Ae2-mediated Cl(-)/HCO3 (-) exchange with maximized gradients, it also had increased intracellular Cl(-) concentration relative to wild-type. Pharmacological reduction of intracellular Cl(-) concentration in Cftr KO crypt epithelium normalized pHi, which was largely Ae2-dependent. We conclude that Cftr KO crypt epithelium maintains an alkaline pHi as a consequence of losing both Cl(-) and HCO3 (-) efflux, which impairs pHi regulation by Ae2. Retention of Cl(-) and an alkaline pHi in crypt epithelium may alter several cellular processes in the proliferative compartment of Cftr KO intestine.
Collapse
Affiliation(s)
- Nancy M. Walker
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; and
| | - Jinghua Liu
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; and
| | - Sydney R. Stein
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; and
| | - Casey D. Stefanski
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; and ,2Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Ashlee M. Strubberg
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; and ,2Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Lane L. Clarke
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; and ,2Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
16
|
Fuster DG, Alexander RT. Traditional and emerging roles for the SLC9 Na+/H+ exchangers. Pflugers Arch 2013; 466:61-76. [PMID: 24337822 DOI: 10.1007/s00424-013-1408-8] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/14/2013] [Accepted: 11/20/2013] [Indexed: 10/25/2022]
Abstract
The SLC9 gene family encodes Na(+)/H(+) exchangers (NHEs). These transmembrane proteins transport ions across lipid bilayers in a diverse array of species from prokaryotes to eukaryotes, including plants, fungi, and animals. They utilize the electrochemical gradient of one ion to transport another ion against its electrochemical gradient. Currently, 13 evolutionarily conserved NHE isoforms are known in mammals [22, 46, 128]. The SLC9 gene family (solute carrier classification of transporters: www.bioparadigms.org) is divided into three subgroups [46]. The SLC9A subgroup encompasses plasmalemmal isoforms NHE1-5 (SLC9A1-5) and the predominantly intracellular isoforms NHE6-9 (SLC9A6-9). The SLC9B subgroup consists of two recently cloned isoforms, NHA1 and NHA2 (SLC9B1 and SLC9B2, respectively). The SLC9C subgroup consist of a sperm specific plasmalemmal NHE (SLC9C1) and a putative NHE, SLC9C2, for which there is currently no functional data [46]. NHEs participate in the regulation of cytosolic and organellar pH as well as cell volume. In the intestine and kidney, NHEs are critical for transepithelial movement of Na(+) and HCO3(-) and thus for whole body volume and acid-base homeostasis [46]. Mutations in the NHE6 or NHE9 genes cause neurological disease in humans and are currently the only NHEs directly linked to human disease. However, it is becoming increasingly apparent that members of this gene family contribute to the pathophysiology of multiple human diseases.
Collapse
Affiliation(s)
- Daniel G Fuster
- Division of Nephrology, Hypertension and Clinical Pharmacology and Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland,
| | | |
Collapse
|
17
|
Roginiel AC, Kohut DL, Kaur S, Saleh AMA, Weber T, Geibel P, Singh H, Geibel JP. Effect of NSAIDs on Na⁺/H⁺ exchanger activity in rat colonic crypts. Am J Physiol Cell Physiol 2013; 305:C512-8. [PMID: 23739181 DOI: 10.1152/ajpcell.00303.2012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs; 1) are widely recommended for several acute and chronic conditions. For example, both indomethacin and aspirin are taken for pain relief. Aspirin is also used for prevention of myocardial infarction, and indomethacin can be administered orally or as a suppository for patients with rheumatoid disease and other chronic inflammatory states. However, use of NSAIDs can cause damage to the mucosal barrier surrounding the gastrointestinal (GI) tract, increasing the risk of ulcer formation. While microencapsulation of NSAIDs has been shown to reduce upper GI injury, sustained release in the lower GI tract and colon may cause epithelial erosion due to increased acidification. The use of suppositories has also been linked to rectal and lower GI bleeding. In this study, we investigated the role of NSAIDs aspirin and indomethacin on Na⁺/H⁺ exchanger (NHE) activity in rat colonic crypts. By comparing average rates of pH recovery between control and NSAID perfusion runs, we were able to determine that both aspirin and indomethacin increase hydrogen extrusion into the colonic lumen. Through treatment with 5-ethylisopropyl amiloride (EIPA), amiloride, and zoniporide dihydrochloride, we further demonstrated that indomethacin specifically enhances proton excretion through regulation of apical NHE-3 and NHE-2 and to a lesser extent on basolateral NHE-1 and NHE-4. Our results suggest that clinical exposure to NSAIDs may affect colonic tissue at the site of selected NHE isoforms, resulting in modulation of transport and barrier function.
Collapse
Affiliation(s)
- Aliya C Roginiel
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Cohen L, Asraf H, Sekler I, Hershfinkel M. Extracellular pH regulates zinc signaling via an Asp residue of the zinc-sensing receptor (ZnR/GPR39). J Biol Chem 2012; 287:33339-50. [PMID: 22879599 DOI: 10.1074/jbc.m112.372441] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Zinc activates a specific Zn(2+)-sensing receptor, ZnR/GPR39, and thereby triggers cellular signaling leading to epithelial cell proliferation and survival. Epithelial cells that express ZnR, particularly colonocytes, face frequent changes in extracellular pH that are of physiological and pathological implication. Here we show that the ZnR/GPR39-dependent Ca(2+) responses in HT29 colonocytes were maximal at pH 7.4 but were reduced by about 50% at pH 7.7 and by about 62% at pH 7.1 and were completely abolished at pH 6.5. Intracellular acidification did not attenuate ZnR/GPR39 activity, indicating that the pH sensor of this protein is located on an extracellular domain. ZnR/GPR39-dependent activation of extracellular-regulated kinase (ERK)1/2 or AKT pathways was abolished at acidic extracellular pH of 6.5. A similar inhibitory effect was monitored for the ZnR/GPR39-dependent up-regulation of Na(+)/H(+) exchange activity at pH 6.5. Focusing on residues putatively facing the extracellular domain, we sought to identify the pH sensor of ZnR/GPR39. Replacing the histidine residues forming the Zn(2+) binding site, His(17) or His(19), or other extracellular-facing histidines to alanine residues did not abolish the pH dependence of ZnR/GPR39. In contrast, replacing Asp(313) with alanine resulted in similar Ca(2+) responses triggered by ZnR/GPR39 at pH 7.4 or 6.5. This mutant also showed similar activation of ERK1/2 and AKT pathways, and ZnR-dependent up-regulation of Na(+)/H(+) exchange at pH 7.4 and pH 6.5. Substitution of Asp(313) to His or Glu residues restored pH sensitivity of the receptor. This indicates that Asp(313), which was shown to modulate Zn(2+) binding, is an essential residue of the pH sensor of GPR39. In conclusion, ZnR/GPR39 is tuned to sense physiologically relevant changes in extracellular pH that thus regulate ZnR-dependent signaling and ion transport activity.
Collapse
Affiliation(s)
- Limor Cohen
- Department of Morphology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer-Sheva 84105, Israel
| | | | | | | |
Collapse
|
19
|
Zinc sensing receptor signaling, mediated by GPR39, reduces butyrate-induced cell death in HT29 colonocytes via upregulation of clusterin. PLoS One 2012; 7:e35482. [PMID: 22545109 PMCID: PMC3335870 DOI: 10.1371/journal.pone.0035482] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 03/16/2012] [Indexed: 02/06/2023] Open
Abstract
Zinc enhances epithelial proliferation, protects the digestive epithelial layer and has profound antiulcerative and antidiarrheal roles in the colon. Despite the clinical significance of this ion, the mechanisms linking zinc to these cellular processes are poorly understood. We have previously identified an extracellular Zn2+ sensing G-protein coupled receptor (ZnR) that activates Ca2+ signaling in colonocytes, but its molecular identity as well as its effects on colonocytes' survival remained elusive. Here, we show that Zn2+, by activation of the ZnR, protects HT29 colonocytes from butyrate induced cell death. Silencing of the G-protein coupled receptor GPR39 expression abolished ZnR-dependent Ca2+ release and Zn2+-dependent survival of butyrate-treated colonocytes. Importantly, GPR39 also mediated ZnR-dependent upregulation of Na+/H+ exchange activity as this activity was found in native colon tissue but not in tissue obtained from GPR39 knock-out mice. Although ZnR-dependent upregulation of Na+/H+ exchange reduced the cellular acid load induced by butyrate, it did not rescue HT29 cells from butyrate induced cell death. ZnR/GPR39 activation however, increased the expression of the anti-apoptotic protein clusterin in butyrate-treated cells. Furthermore, silencing of clusterin abolished the Zn2+-dependent survival of HT29 cells. Altogether, our results demonstrate that extracellular Zn2+, acting through ZnR, regulates intracellular pH and clusterin expression thereby enhancing survival of HT29 colonocytes. Moreover, we identify GPR39 as the molecular moiety of ZnR in HT29 and native colonocytes.
Collapse
|
20
|
Liu J, Walker NM, Cook MT, Ootani A, Clarke LL. Functional Cftr in crypt epithelium of organotypic enteroid cultures from murine small intestine. Am J Physiol Cell Physiol 2012; 302:C1492-503. [PMID: 22403785 DOI: 10.1152/ajpcell.00392.2011] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Physiological studies of intact crypt epithelium have been limited by problems of accessibility in vivo and dedifferentiation in standard primary culture. Investigations of murine intestinal stem cells have recently yielded a primary intestinal culture in three-dimensional gel suspension that recapitulates crypt structure and epithelial differentiation (Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, Van Es JH, Abo A, Kujala P, Peters PJ, Clevers H. Nature 459: 262-265, 2009). We investigated the utility of murine intestinal crypt cultures (termed "enteroids") for physiological studies of crypt epithelium by focusing on the transport activity of the cystic fibrosis transmembrane conductance regulator Cftr. Enteroids had multiple crypts with well-differentiated goblet and Paneth cells that degranulated on exposure to the muscarinic agonist carbachol. Modified growth medium provided a crypt proliferation rate, as measured by 5-ethynyl-2'-deoxyuridine labeling, which was similar to proliferation in vivo. Immunoblots demonstrated equivalent Cftr expression in comparisons of freshly isolated crypts with primary and passage 1 enteroids. Apparent enteroid differences in mRNA expression of other transporters were primarily associated with villous epithelial contamination of freshly isolated crypts. Microelectrode analysis revealed cAMP-stimulated membrane depolarization in enteroid epithelium from wild-type (WT) but not Cftr knockout (KO) mice. Morphological and microfluorimetric studies, respectively, demonstrated Cftr-dependent cell shrinkage and lower intracellular pH in WT enteroid epithelium in contrast to Cftr KO epithelium or WT epithelium treated with Cftr inhibitor 172. We conclude that crypt epithelium of murine enteroids exhibit Cftr expression and activity that recapitulates crypt epithelium in vivo. Enteroids provide a primary culture model that is suitable for physiological studies of regenerating crypt epithelium.
Collapse
Affiliation(s)
- Jinghua Liu
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | | | | | | | | |
Collapse
|
21
|
Abstract
Na(+) and Cl(-) movement across the intestinal epithelium occurs by several interconnected mechanisms: (a) nutrient-coupled Na(+) absorption, (b) electroneutral NaCl absorption, (c) electrogenic Cl(-) secretion by CFTR, and (d) electrogenic Na(+) absorption by ENaC. All these transport modes require a favorable electrochemical gradient maintained by the basolateral Na(+)/K(+)-ATPase, a Cl(-) channel, and K(+) channels. Electroneutral NaCl absorption is observed from the small intestine to the distal colon. This transport is mediated by apical Na(+)/H(+) (NHE2/3) and Cl(-)/HCO(3)(-) (Slc26a3/a6 and others) exchangers that provide the major route of NaCl absorption. Electroneutral NaCl absorption and Cl(-) secretion by CFTR are oppositely regulated by the autonomic nerve system, the immune system, and the endocrine system via PKAα, PKCα, cGKII, and/or SGK1. This integrated regulation requires the formation of macromolecular complexes, which are mediated by the NHERF family of scaffold proteins and involve internalization of NHE3. Through use of knockout mice and human mutations, a more detailed understanding of the integrated as well as subtle regulation of electroneutral NaCl absorption by the mammalian intestine has emerged.
Collapse
Affiliation(s)
- Akira Kato
- Biological Sciences, Tokyo Institute of Technology, Yokohama, Japan.
| | | |
Collapse
|
22
|
Guan Y, Watson AJM, Marchiando AM, Bradford E, Shen L, Turner JR, Montrose MH. Redistribution of the tight junction protein ZO-1 during physiological shedding of mouse intestinal epithelial cells. Am J Physiol Cell Physiol 2011; 300:C1404-14. [PMID: 21346149 DOI: 10.1152/ajpcell.00270.2010] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We questioned how tight junctions contribute to intestinal barrier function during the cell shedding that is part of physiological cell renewal. Intravital confocal microscopy studied the jejunal villus epithelium of mice expressing a fluorescent zonula occludens 1 (ZO-1) fusion protein. Vital staining also visualized the cell nucleus (Hoechst staining) or local permeability to luminal constituents (Lucifer Yellow; LY). In a cell fated to be shed, ZO-1 redistributes from the tight junction toward the apical and then basolateral cell region. ZO-1 rearrangement occurs 15 ± 6 min (n = 28) before movement of the cell nucleus from the epithelial layer. During cell extrusion, permeation of luminal LY extends along the lateral intercellular spaces of the shedding cell only as far as the location of ZO-1. Within 3 min after detachment from the epithelial layer, nuclear chromatin condenses. After cell loss, a residual patch of ZO-1 remains in the space previously occupied by the departed cell, and the size of the patch shrinks to 14 ± 2% (n = 15) of the original cell space over 20 min. The duration of cell shedding measured by nucleus movement (14 ± 1 min) is much less than the total duration of ZO-1 redistribution at the same sites (45 ± 2 min). In about 15% of cell shedding cases, neighboring epithelial cells also undergo extrusion with a delay of 5-10 min. With the use of normal mice, ZO-1 immunofluorescent staining of fixed tissue confirmed ZO-1 redistribution and the presence of ZO-1 patches beneath shedding cells. Immunostaining also showed that redistribution of ZO-1 occurred without corresponding mixing of apical and basolateral membrane domains as marked by ezrin or E-cadherin. ZO-1 redistribution is the earliest cellular event yet identified as a herald of physiological cell shedding, and redistribution of tight junction function along the lateral plasma membrane sustains epithelial barrier during cell shedding.
Collapse
Affiliation(s)
- Yanfang Guan
- Dept. of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0576, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Loss of the NHE2 Na+/H+ exchanger in mice results in dilation of folliculo-stellate cell canaliculi. J Biomed Biotechnol 2011; 2011:510827. [PMID: 21274460 PMCID: PMC3025390 DOI: 10.1155/2011/510827] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 11/23/2010] [Indexed: 11/18/2022] Open
Abstract
Genetic ablation of the NHE2 Na+/H+ exchanger causes gastric achlorhydria, absorptive defects in kidney and colon, and low fertility. Here we show that NHE2 is expressed in the pituitary, with the highest mRNA expression in pars distalis and lower expression in pars intermedia. In pars distalis of NHE2-null mice, prominent cyst-like dilatations of folliculo-stellate (FS) cell canaliculi developed with age, and there were increased FS cell area, accumulation of lipid in FS cell cytoplasm, redundancies in FS cell basement membrane, and other changes. The expansion of the canaliculi indicates that NHE2 is a major absorptive Na+/H+ exchanger in the luminal membranes lining the extensive network of channels formed by FS cells, which may provide a means of intrapituitary communication. The results suggest that NHE2 contributes to homeostatic regulation of the volume and composition of the canalicular fluid and may counter the secretory activity of the CFTR Cl− channel, which is known to be expressed in pituitary.
Collapse
|
24
|
Bachmann O, Juric M, Seidler U, Manns MP, Yu H. Basolateral ion transporters involved in colonic epithelial electrolyte absorption, anion secretion and cellular homeostasis. Acta Physiol (Oxf) 2011; 201:33-46. [PMID: 20528802 DOI: 10.1111/j.1748-1716.2010.02153.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Electrolyte transporters located in the basolateral membrane of the colonic epithelium are increasingly appreciated as elaborately regulated components of specific transport functions and cellular homeostasis: During electrolyte absorption, Na(+) /K(+) ATPase, Cl⁻ conductance, Cl⁻/HCO₃⁻ exchange, K(+) /Cl⁻ cotransport and K(+) channels are candidates for basolateral Na(+) , Cl⁻ and K(+) extrusion. The process of colonic anion secretion involves basolateral Na(+) /K(+) /2Cl⁻ , and probably also Na(+) /HCO₃⁻ cotransport, as well as Na(+) /K(+) ATPase and K(+) channels to supply substrate, stabilize the membrane potential and generate driving force respectively. Together with a multitude of additional transport systems, Na(+) /H(+) exchange and Na(+) /HCO₃⁻ cotransport have been implicated in colonocyte pH(i) and volume homeostasis. The purpose of this article is to summarize recently gathered information on the molecular identity, function and regulation of the involved basolateral transport systems in native tissue. Furthermore, we discuss how these findings can help to integrate these systems into the transport function and the cellular homoeostasis of colonic epithelial cells. Finally, disturbances of basolateral electrolyte transport during disease states such as mucosal inflammation will be reviewed.
Collapse
Affiliation(s)
- O Bachmann
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Germany.
| | | | | | | | | |
Collapse
|
25
|
Seidler U, Song P, Xiao F, Riederer B, Bachmann O, Chen M. Recent advances in the molecular and functional characterization of acid/base and electrolyte transporters in the basolateral membranes of gastric and duodenal epithelial cells. Acta Physiol (Oxf) 2011; 201:3-20. [PMID: 20331540 DOI: 10.1111/j.1748-1716.2010.02107.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
All segments of the gastrointestinal tract are comprised of an elaborately folded epithelium that expresses a variety of cell types and performs multiple secretory and absorptive functions. While the apical membrane expresses the electrolyte transporters that secrete or absorb electrolytes and water, basolateral transporters regulate the secretory or absorptive rates. During gastric acid formation, Cl⁻/HCO₃⁻ and Na(+) /H(+) exchange and other transporters secure Cl⁻ re-supply as well as pH and volume regulation. Gastric surface cells utilize ion transporters to secrete HCO₃⁻, maintain pH(i) during a luminal acid load and repair damaged surface areas during the process of epithelial restitution. Na(+)/H(+) exchange and Na(+)/HCO₃⁻ cotransport serve basolateral acid/base import for gastroduodenal HCO₃⁻ secretion. The gastric and duodenal epithelium also absorbs salt and water. Recent molecular information on novel ion transporters expressed in the gastric and duodenal epithelium has exploded; however, a function has not been found yet for all transporters. The purpose of this review is to summarize current knowledge on the molecular identity and cellular function of basolateral ion transporters in the gastric and duodenal epithelium.
Collapse
Affiliation(s)
- U Seidler
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Germany.
| | | | | | | | | | | |
Collapse
|
26
|
Guan YF, Pritts TA, Montrose MH. Ischemic post-conditioning to counteract intestinal ischemia/reperfusion injury. World J Gastrointest Pathophysiol 2010; 1:137-43. [PMID: 21607154 PMCID: PMC3097957 DOI: 10.4291/wjgp.v1.i4.137] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 09/29/2010] [Accepted: 10/06/2010] [Indexed: 02/06/2023] Open
Abstract
Intestinal ischemia is a severe disorder with a variety of causes. Reperfusion is a common occurrence during treatment of acute intestinal ischemia but the injury resulting from ischemia/reperfusion (IR) may lead to even more serious complications from intestinal atrophy to multiple organ failure and death. The susceptibility of the intestine to IR-induced injury (IRI) appears from various experimental studies and clinical settings such as cardiac and major vascular surgery and organ transplantation. Whereas oxygen free radicals, activation of leukocytes, failure of microvascular perfusion, cellular acidosis and disturbance of intracellular homeostasis have been implicated as important factors in the pathogenesis of intestinal IRI, the mechanisms underlying this disorder are not well known. To date, increasing attention is being paid in animal studies to potential pre- and post-ischemia treatments that protect against intestinal IRI such as drug interference with IR-induced apoptosis and inflammation processes and ischemic pre-conditioning. However, better insight is needed into the molecular and cellular events associated with reperfusion-induced damage to develop effective clinical protection protocols to combat this disorder. In this respect, the use of ischemic post-conditioning in combination with experimentally prolonged acidosis blocking deleterious reperfusion actions may turn out to have particular clinical relevance.
Collapse
|
27
|
Talbot C, Lytle C. Segregation of Na/H exchanger-3 and Cl/HCO3 exchanger SLC26A3 (DRA) in rodent cecum and colon. Am J Physiol Gastrointest Liver Physiol 2010; 299:G358-67. [PMID: 20466943 DOI: 10.1152/ajpgi.00151.2010] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The colon is believed to absorb NaCl via the coupled operation of apical Na/H exchanger-3 (NHE3) and Cl/HCO(3) exchanger SLC26A3 (DRA). Efficient coupling requires that NHE3 and DRA operate in close proximity within common luminal and cytosolic microenvironments. Thus we examined whether these proteins coexist along the apical margin of surface enterocytes by quantitative immunofluorescence microscopy in consecutive colon segments from nonfasted mice and rats. The cecocolonic profiles of NHE3 and DRA expression were roughly inverse; NHE3 was highest in proximal colon (PC) and negligible in distal colon, whereas DRA was absent in early PC and highest in the late midcolon, and DRA was prominent in the cecum whereas NHE3 was not. NHE3 and DRA coexisted only in the middle third of the colon. The consequences of unpaired NHE3/DRA expression on mucosal surface (subscript MS) pH and Na(+) concentration ([Na(+)]) were assessed in nonfasted rats in situ using miniature electrodes. In the cecum, where only DRA is expressed, pH(MS) was approximately 7.5, markedly higher than underlaying stool (6.3), consistent with net HCO(3)(-) secretion. In the early PC, where NHE3 is not expressed with DRA, pH(MS) was acidic (6.2), consistent with unopposed H(+) secretion. [Na(+)](MS) was approximately 60 mM in the cecum, decreased along the PC to approximately 20 mM, and declined further to approximately 10 mM distally. Cl(-) was secreted into the PC, then reabsorbed distally. Our results suggest a model in which 1) unpaired DRA activity in the cecum maintains an alkaline mucosal surface that could neutralize fermentative H(+); 2) unpaired NHE3 activity in the early PC preserves an acidic mucosal surface that could energize short-chain fatty acid absorption; and 3) coupled NHE3/DRA activities in the midcolon allow for vigorous NaCl absorption at a neutral pH(MS).
Collapse
|
28
|
Volume density, distribution, and ultrastructure of secretory and basolateral membranes and mitochondria predict parietal cell secretory (dys)function. J Biomed Biotechnol 2010; 2010:394198. [PMID: 20339514 PMCID: PMC2842899 DOI: 10.1155/2010/394198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 11/03/2009] [Accepted: 12/07/2009] [Indexed: 11/24/2022] Open
Abstract
Acid secretion in gastric parietal cells requires highly coordinated membrane transport and vesicle trafficking. Histologically, consensus defines acid secretion as the ratio of the volume density (Vd) of canalicular and apical membranes (CAMs) to tubulovesicular (TV) membranes, a value which varies widely under normal conditions. Examination of numerous achlorhydric mice made it clear that this paradigm is discrepant when used to assess most mice with genetic mutations affecting acid secretion. Vd of organelles in parietal cells of 6 genetically engineered mouse strains was obtained to identify a stable histological phenotype of acid secretion. We confirmed that CAM to TV ratio fairly represented secretory activity in untreated and secretion-inhibited wild-type (WT) mice and in NHE2−/− mice as well, though the response was significantly attenuated in the latter. However, high CAM to TV ratios wrongly posed as active acid secretion in AE2−/−, GHKAα−/−, and NHE4−/− mice. Achlorhydric genotypes also had a significantly higher Vd of basolateral membrane than WT mice, and reduced Vd of mitochondria and canaliculi. The Vd of mitochondria, and ratio of the Vd of basolateral membranes/Vd of mitochondria were preferred predictors of the level of acid secretion. Alterations in acid secretion, then, cause significant changes not only in the Vd of secretory membranes but also in mitochondria and basolateral membranes.
Collapse
|
29
|
Mechanisms of the regulation of the intestinal Na+/H+ exchanger NHE3. J Biomed Biotechnol 2010; 2010:238080. [PMID: 20011065 PMCID: PMC2789519 DOI: 10.1155/2010/238080] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Accepted: 09/11/2009] [Indexed: 01/25/2023] Open
Abstract
A major of Na+ absorptive process in the proximal part of intestine and kidney is electroneutral exchange of Na+ and H+ by Na+/H+ exchanger type 3 (NHE3). During the past decade, significant advance has been achieved in the mechanisms of NHE3 regulation. A bulk of the current knowledge on Na+/H+ exchanger regulation is based on heterologous expression of mammalian Na+/H+ exchangers in Na+/H+ exchanger deficient fibroblasts, renal epithelial, and intestinal epithelial cells. Based on the reductionist's approach, an understanding of NHE3 regulation has been greatly advanced. More recently, confirmations of in vitro studies have been made using animals deficient in one or more proteins but in some cases unexpected findings have emerged. The purpose of this paper is to provide a brief overview of recent progress in the regulation and functions of NHE3 present in the luminal membrane of the intestinal tract.
Collapse
|
30
|
Catalán MA, Nakamoto T, Gonzalez-Begne M, Camden JM, Wall SM, Clarke LL, Melvin JE. Cftr and ENaC ion channels mediate NaCl absorption in the mouse submandibular gland. J Physiol 2009; 588:713-24. [PMID: 20026617 DOI: 10.1113/jphysiol.2009.183541] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cystic fibrosis is caused by mutations in CFTR, the cystic fibrosis transmembrane conductance regulator gene. Disruption of CFTR-mediated anion conductance results in defective fluid and electrolyte movement in the epithelial cells of organs such as the pancreas, airways and sweat glands, but the function of CFTR in salivary glands is unclear. Salivary gland acinar cells produce an isotonic, plasma-like fluid, which is subsequently modified by the ducts to produce a hypotonic, NaCl-depleted final saliva. In the present study we investigated whether submandibular salivary glands (SMGs) in F508 mice (Cftr(F/F)) display ion transport defects characteristic of cystic fibrosis in other tissues. Immunolocalization and whole-cell recordings demonstrated that Cftr and the epithelial Na(+) (ENaC) channels are co-expressed in the apical membrane of submandibular duct cells, consistent with the significantly higher saliva [NaCl] observed in vivo in Cftr(F/F) mice. In contrast, Cftr and ENaC channels were not detected in acinar cells, nor was saliva production affected in Cftr(F/F) mice, implying that Cftr contributes little to the fluid secretion process in the mouse SMG. To identify the source of the NaCl absorption defect in Cftr(F/F) mice, saliva was collected from ex vivo perfused SMGs. Cftr(F/F) glands secreted saliva with significantly increased [NaCl]. Moreover, pharmacological inhibition of either Cftr or ENaC in the ex vivo SMGs mimicked the Cftr(F/F) phenotype. In summary, our results demonstrate that NaCl absorption requires and is likely to be mediated by functionally dependent Cftr and ENaC channels localized to the apical membranes of mouse salivary gland duct cells.
Collapse
Affiliation(s)
- Marcelo A Catalán
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14620, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Guan Y, Worrell RT, Pritts TA, Montrose MH. Intestinal ischemia-reperfusion injury: reversible and irreversible damage imaged in vivo. Am J Physiol Gastrointest Liver Physiol 2009; 297:G187-96. [PMID: 19407214 PMCID: PMC2889629 DOI: 10.1152/ajpgi.90595.2008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The early events in an intestinal ischemic episode have been difficult to evaluate. Using in vivo microscopy we have analyzed in real-time the effects of short (15 min) and long (40-50 min) ischemia with subsequent reperfusion (IR), evaluating structure, integrity, and functioning of the mouse jejunal mucosa while monitoring blood flow by confocal microscopy. IR was imposed by inflation/deflation of a vascular occluder, and blood flow was monitored and confirmed with scanning confocal imaging. After short ischemia, villus tip cells revealed a rapid increase (23%) in the intracellular NAD(P)H concentration (confocal autofluorescence microscopy), and the pH-sensitive probe BCECF showed a biphasic response of the intracellular pH (pH(i)), quickly alkalinizing from the resting value of 6.8 +/- 0.1 to 7.1 +/- 0.1 but then strongly acidifying to 6.3 +/- 0.1. Upon reperfusion, values returned toward control. In contrast, results were heterogeneous after long IR. During long ischemia, one-third of the epithelial cells remained viable with reversible changes upon reperfusion, but remaining cells lost membrane integrity (Lucifer Yellow uptake, LY) and had membrane blebs during ischemia. These effects became more pronounced as the reperfusion interval progressed when cells exhibited more severely affected NAD(P)H and pH(i) values, larger blebs, and more LY uptake and eventually were shed from the villus. Results from stereo microscopy suggest that these irreversible effects of IR may have occurred as a result of incomplete restorations of local blood flow, especially at the antimesenteric side of the intestine. We conclude that the adverse effects of short ischemia on the jejunum epithelium are fully reversible during the reperfusion interval. However, after long ischemia, reperfusion cannot restore normal structure and functioning of a majority of cells, which deteriorate further. Our results provide a basis for defining the cellular events that cause tissue to transit from reversible to irreversible damage during IR.
Collapse
Affiliation(s)
- Yanfang Guan
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0576, USA.
| | - Roger T. Worrell
- Departments of Molecular and Cellular Physiology and Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Timothy A. Pritts
- Departments of Molecular and Cellular Physiology and Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marshall H. Montrose
- Departments of Molecular and Cellular Physiology and Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
32
|
Sullivan S, Alex P, Dassopoulos T, Zachos NC, Iacobuzio-Donahue C, Donowitz M, Brant SR, Cuffari C, Harris ML, Datta LW, Conklin A, Chen Y, Li X. Downregulation of sodium transporters and NHERF proteins in IBD patients and mouse colitis models: potential contributors to IBD-associated diarrhea. Inflamm Bowel Dis 2009; 15:261-74. [PMID: 18942765 PMCID: PMC2627787 DOI: 10.1002/ibd.20743] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND One of the most common symptoms among patients with inflammatory bowel disease (IBD) is diarrhea, which is thought to be contributed by changes in electrolyte transport associated with intestinal inflammation. This study was designed to test the hypothesis that intestinal Na(+)-related transporters/channels and their regulatory proteins may be downregulated as a potential contributor to IBD-associated diarrhea. METHODS SDS-PAGE and Western blotting and/or confocal immunomicroscopy were used to examine the expression of Na(+)/H(+)-exchangers 1-3 (NHE1-3), epithelial Na(+) channel (ENaC), Na(+)/K(+)-ATPase, the intracellular Cl(-) channel 5 (ClC-5), and NHE3 regulatory factors (NHERF1,2) in ileal and colonic pinch biopsies from IBD patients and noninflammatory controls, as well as from colonic mucosa of dextran sodium sulfate (DSS)- and TNBS-induced acute murine IBD models. RESULTS NHE1,3 (but not NHE2), beta-ENaC, Na(+)/K(+)-ATPase-alpha, ClC-5, and NHERF1 were all downregulated in sigmoid mucosal biopsies from most cases of active UC and/or CD compared to controls. NHE3 was also decreased in ileal mucosal biopsies of active CD, as well as in approximately 50% of sigmoid biopsies from inactive UC or CD. Importantly, similar downregulation of NHE1,3, beta-ENaC, and NHERF1,2 was also observed in the mouse colon (but not ileum) of DSS- and TNBS-induced colitis. CONCLUSIONS IBD-associated diarrhea may be due to a coordinated downregulation of multiple Na(+) transporter and related regulatory proteins, including NHE1,3, Na(+)/K(+)-ATPase, and ENaC, as well as NHERF1,2, and ClC-5, all of which are involved directly or indirectly in intestinal Na(+) absorption.
Collapse
Affiliation(s)
- Sean Sullivan
- GI Division/Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205,Dept of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Philip Alex
- GI Division/Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | | | - Nicholas C. Zachos
- GI Division/Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | | | - Mark Donowitz
- GI Division/Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Steven R. Brant
- GI Division/Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Carmen Cuffari
- Dept of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Mary L. Harris
- GI Division/Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Lisa Wu Datta
- GI Division/Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - aurie Conklin
- GI Division/Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205,Dept of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Yueping Chen
- GI Division/Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Xuhang Li
- GI Division/Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205,Address correspondence to: Xuhang Li, Ph.D. 720 Rutland Ave, 918 Ross Research Bldg GI Division, Department of Medicine Johns Hopkins University School of Medicine Baltimore, MD 21205 Tel. 443-287-4804 Fax. 410-955-9677 E-mail:
| |
Collapse
|
33
|
Moeser AJ, Nighot PK, Ryan KA, Simpson JE, Clarke LL, Blikslager AT. Mice lacking the Na+/H+ exchanger 2 have impaired recovery of intestinal barrier function. Am J Physiol Gastrointest Liver Physiol 2008; 295:G791-7. [PMID: 18719001 PMCID: PMC4838133 DOI: 10.1152/ajpgi.00538.2007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ischemic injury induces breakdown of the intestinal barrier. Recent studies in porcine postischemic tissues indicate that inhibition of NHE2 results in enhanced recovery of barrier function in vitro via a process involving interepithelial tight junctions. To further study this process, recovery of barrier function was assessed in wild-type (NHE2(+/+)) and NHE2(-/-) mice in vivo and wild-type mice in vitro. Mice were subjected to complete mesenteric ischemia in vivo, after which barrier function was measured by blood-to-lumen mannitol clearance over a 3-h recovery period or measurement of transepithelial electrical resistance (TER) in Ussing chambers immediately following ischemia. Tissues were assessed for expression of select junctional proteins. Compared with NHE2(+/+) mice, NHE2(-/-) mice had greater intestinal permeability during the postischemic recovery process. In contrast to prior porcine studies, pharmacological inhibition of NHE2 in postischemic tissues from wild-type mice also resulted in significant reductions in TER. Mucosa from NHE2(-/-) mice displayed a shift of occludin and claudin-1 expression to the Triton-X-soluble membrane fractions and showed disruption of occludin and claudin-1 localization patterns following injury. This was qualitatively and quantitatively recovered in NHE2(+/+) mice compared with NHE2(-/-) mice by the end of the 3-h recovery period. Serine phosphorylation of occludin and claudin-1 was downregulated in NHE2(-/-) postischemia compared with wild-type mice. These data indicate an important role for NHE2 in recovery of barrier function in mice via a mechanism involving tight junctions.
Collapse
|
34
|
Heitzmann D, Warth R. Physiology and pathophysiology of potassium channels in gastrointestinal epithelia. Physiol Rev 2008; 88:1119-82. [PMID: 18626068 DOI: 10.1152/physrev.00020.2007] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Epithelial cells of the gastrointestinal tract are an important barrier between the "milieu interne" and the luminal content of the gut. They perform transport of nutrients, salts, and water, which is essential for the maintenance of body homeostasis. In these epithelia, a variety of K(+) channels are expressed, allowing adaptation to different needs. This review provides an overview of the current literature that has led to a better understanding of the multifaceted function of gastrointestinal K(+) channels, thereby shedding light on pathophysiological implications of impaired channel function. For instance, in gastric mucosa, K(+) channel function is a prerequisite for acid secretion of parietal cells. In epithelial cells of small intestine, K(+) channels provide the driving force for electrogenic transport processes across the plasma membrane, and they are involved in cell volume regulation. Fine tuning of salt and water transport and of K(+) homeostasis occurs in colonic epithelia cells, where K(+) channels are involved in secretory and reabsorptive processes. Furthermore, there is growing evidence for changes in epithelial K(+) channel expression during cell proliferation, differentiation, apoptosis, and, under pathological conditions, carcinogenesis. In the future, integrative approaches using functional and postgenomic/proteomic techniques will help us to gain comprehensive insights into the role of K(+) channels of the gastrointestinal tract.
Collapse
Affiliation(s)
- Dirk Heitzmann
- Institute of Physiology and Clinic and Policlinic for Internal Medicine II, Regensburg, Germany
| | | |
Collapse
|
35
|
Claiborne JB, Choe KP, Morrison-Shetlar AI, Weakley JC, Havird J, Freiji A, Evans DH, Edwards SL. Molecular detection and immunological localization of gill Na+/H+ exchanger in the dogfish (Squalus acanthias). Am J Physiol Regul Integr Comp Physiol 2007; 294:R1092-102. [PMID: 18094061 DOI: 10.1152/ajpregu.00718.2007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The dogfish (Squalus acanthias) can make rapid adjustments to gill acid-base transfers to compensate for internal acidosis/alkalosis. Branchial Na+/H+ exchange (NHE) has been postulated as one mechanism driving the excretion of H+ following acidosis. We have cloned gill cDNA that includes an open reading frame coding for a 770-residue protein most homologous (approximately 71%) to mammalian NHE2. RT-PCR revealed NHE2 transcripts predominantly in gill, stomach, rectal gland, intestine, and kidney. In situ hybridization with an antisense probe against NHE2 in gill sections revealed a strong mRNA signal from a subset of interlamellar and lamellae cells. We developed dogfish-specific polyclonal antibodies against NHE2 that detected a approximately 70-kDa protein in Western blots and immunologically recognized branchial cells having two patterns of protein expression. Cytoplasmic and apical NHE2 immunoreactivity were observed in cells coexpressing basolateral Na+-K+-ATPase. Other large ovoid cells more generally staining for NHE2 also were strongly positive for basolateral H+-ATPase. Gill mRNA levels for NHE2 and H+-ATPase did not change following systemic acidosis (as measured by quantitative PCR 2 h after a 1- or 2-meq/kg acid infusion). These data indicate that posttranslational adjustments of NHE2 and other transport systems (e.g., NHE3) following acidosis may be of importance in the short-term pH adjustment and net branchial H+ efflux observed in vivo. NHE2 may play multiple roles in the gills, involved with H+ efflux from acid-secreting cells, basolateral H+ reabsorption for pHi regulation, and in parallel with H+-ATPase for the generation of HCO3(-) in base-secreting cells.
Collapse
Affiliation(s)
- James B Claiborne
- Department of Biology, Georgia Southern University, Statesboro, GA 30460, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
NHE3 is the brush-border (BB) Na+/H+exchanger of small intestine, colon, and renal proximal tubule which is involved in large amounts of neutral Na+absorption. NHE3 is a highly regulated transporter, being both stimulated and inhibited by signaling that mimics the postprandial state. It also undergoes downregulation in diarrheal diseases as well as changes in renal disorders. For this regulation, NHE3 exists in large, multiprotein complexes in which it associates with at least nine other proteins. This review deals with short-term regulation of NHE3 and the identity and function of its recognized interacting partners and the multiprotein complexes in which NHE3 functions.
Collapse
Affiliation(s)
- Mark Donowitz
- Department of Medicine, GI Division, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| | | |
Collapse
|
37
|
Hua P, Xu H, Uno JK, Lipko MA, Dong J, Kiela PR, Ghishan FK. Sp1 and Sp3 mediate NHE2 gene transcription in the intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2007; 293:G146-53. [PMID: 17379926 DOI: 10.1152/ajpgi.00443.2006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Our previous studies have identified a minimal Sp1-driven promoter region (nt -36/+116) directing NHE2 expression in mouse renal epithelial cells. However, this minimal promoter region was not sufficient to support active transcription of NHE2 gene in the intestinal epithelial cells, suggesting the need for additional upstream regulatory elements. In the present study, we used nontransformed rat intestinal epithelial (RIE) cells as a model to identify the minimal promoter region and transcription factors necessary for the basal transcription of rat NHE2 gene in the intestinal epithelial cells. We identified a region within the rat NHE2 gene promoter located within nt -67/-43 upstream of transcription initiation site as indispensable for the promoter function in intestinal epithelial cells. Mutations at nt -56/-51 not only abolished the DNA-protein interaction in this region, but also completely abolished NHE2 gene promoter activity in RIE cells. Supershift assays revealed that Sp1 and Sp3 interact with this promoter region, but, contrary to the minimal promoter indispensable for renal expression of NHE2, both transcription factors expressed individually in Drosophila SL2 cells activated rat NHE2 gene promoter. Moreover, Sp1 was a weaker transactivator and when coexpressed in SL2 cells it reduced Sp3-mediated NHE2 basal promoter activity. Furthermore, DNase I footprinting confirmed that nt -58/-51 is protected by nuclear protein from RIE cells. We conclude that the mechanism of basal control of rat NHE2 gene promoter activity is different in the renal and intestinal epithelium, with Sp3 being the major transcriptional activator of NHE2 gene transcription in the intestinal epithelial cells.
Collapse
Affiliation(s)
- Ping Hua
- Department of Pediatrics, Steele Memorial Children's Research Center, 1501 N. Campbell Ave., Tucson, AZ 85724, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Gens JS, Dou H, Tackett L, Kong SS, Chu S, Montrose MH. Different ionic conditions prompt NHE2 and NHE3 translocation to the plasma membrane. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1768:1023-35. [PMID: 17303069 PMCID: PMC1974857 DOI: 10.1016/j.bbamem.2007.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Revised: 12/31/2006] [Accepted: 01/04/2007] [Indexed: 11/29/2022]
Abstract
We tested whether NHE3 and NHE2 Na(+)/H(+) exchanger isoforms were recruited to the plasma membrane (PM) in response to changes in ion homeostasis. NHE2-CFP or NHE3-CFP fusion proteins were functional Na(+)/H(+) exchangers when transiently expressed in NHE-deficient PS120 fibroblasts. Confocal morphometry of cells whose PM was labeled with FM4-64 measured the fractional amount of fusion protein at the cell surface. In resting cells, 10-20% of CFP fluorescence was at PM and stable over time. A protocol commonly used to activate the Na(+)/H(+) exchange function (NH(4)-prepulse acid load sustained in Na(+)-free medium), increased PM percentages of PM NHE3-CFP and NHE2-CFP. Separation of cellular acidification from Na(+) removal revealed that only NHE3-CFP translocated when medium Na(+) was removed, and only NHE2-CFP translocated when the cell was acidified. NHE2/NHE3 chimeric proteins demonstrate that the Na(+)-removal response element resides predominantly in the NHE3 cytoplasmic tail and is distinct from the acidification response sequence of NHE2.
Collapse
Affiliation(s)
- J. Scott Gens
- Biocomplexity Institute, Indiana University, Bloomington, Indiana, 47405
| | - Hongwei Dou
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267
| | - Lixuan Tackett
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202-5120
| | - Shen-Shen Kong
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202-5120
| | - Shaoyou Chu
- Eli Lilly and Company, Indianapolis, Indiana, 46225
| | - Marshall H. Montrose
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202-5120
- Corresponding Author: Marshall H. Montrose, Mail address: Department of Molecular and Cellular Physiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, Telephone number: (513)-558-5636, FAX number: (513)-558-5738, E-mail:
| |
Collapse
|