Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 May 11.
Published in final edited form as: Curr Opin Gastroenterol. 2009 Mar;25(2):87–91. doi: 10.1097/MOG.0b013e3283260900

Ion transport in the intestine

Pawel R Kiela a,b, Fayez K Ghishan a
PMCID: PMC4427515  NIHMSID: NIHMS686598  PMID: 19528875

Abstract

Purpose of review

In recent years, the field of intestinal physiology has witnessed significant progress in our understanding of the expression and function of ion transport proteins and their genes under physiological and pathophysiological conditions. This review will present some of these most recent advances in the small intestinal ion transport mechanisms.

Recent findings

One of the new and exciting aspects of this field has been the integration of function and structure of several intestinal transport processes. This is well exemplified by the discussed intricacies of intestinal bicarbonate secretion as well as the role of scaffolding PDZ proteins interacting with several transporters. We also discuss some of the most recent data pointing to the role of ion transporters in the pathogenesis of inflammation-associated diarrhea and their potential role in the maintenance of epithelial integrity.

Summary

Mouse models deficient in some of the key genes encoding ion transporters and their adapter proteins continue to provide important clues into intestinal transport processes. Several of the new in-vivo findings revise or complement past paradigms, many of which were derived from in-vitro approaches. New data on the interdependent functions of multiple transporters, as exemplified here by intestinal bicarbonate secretion, increase the complexity of the intestinal ion transport mechanisms and continue to contribute to a more integrated view of the transport phenomena in the gut. Data from patients and mouse models of intestinal inflammation also increase our understanding of the pathophysiology of inflammation-associated diarrhea.

Keywords: anion exchange, bicarbonate secretion, inflammation, Na+/H+ exchange, PDZ proteins

Introduction

Research on intestinal and epithelial transport in general can be viewed historically as proceeding in phases. The first phase was represented by physiological studies on solute transport without the knowledge of the molecular identity of the transport proteins and their genes. In the next stage, with the advent of molecular biology, recombinant DNA techniques, and later successful genome sequencing projects, most of the critical transport genes have been identified, cloned, and characterized for their tissue and cellular distribution, substrate specificity, transport kinetics, gene organization, transcriptional and post-transcriptional control under basal conditions and as regulated by physiological stimuli. Later, with the advent of gene targeting technologies and evolving animal models of human disorders, intestinal transport physiologists focused their attention on evaluating the physiological roles of intestinal transporters in mice carrying targeted inactivating mutations, and on regulation of intestinal transport processes under pathophysiological conditions related to an array of disorders affecting electrolyte homeostasis in the gut. Another new and exciting aspect of recent progress in this field has been the integration of function and structure of several intestinal transport processes. Although the realization that many of the membrane transporters do not function independently is not a new concept, many recent studies explain it with significantly more biochemical detail, highlighted by the role of common scaffolding proteins participating in the formation of membrane-associated signaling complexes effectively regulating the activity, cellular localization, and degradation rate of the intestinal transport proteins.

Integration of transporter function in duodenal bicarbonate secretion

One prime example of the complexity and integration of function in intestinal ion transport is bicarbonate secretion. Duodenal HCO3 secretion is a critical nonstructural protection mechanism against the acidic gastric output, against changes in luminal pH, which could adversely affect mucin viscosity and bacterial binding, and against mucosal ulcerations in the otherwise ‘leaky’ duodenal epithelium. Several key transport pathways participate in this defense mechanism, resulting not only in neutralization of the luminal content, but also providing high intracellular buffering capacity to counteract the effects of highly acidic millieu. These include basolateral uptake and enzymatic generation of HCO3, and its luminal secretion, functions secondarily affected by abundantly expressed apical and basolateral Na+/H+ exchangers. HCO3 entry into the epithelial cells is provided by the activity of the basolateral Na+/HCO3 cotransporter NBC1 along with NHE1 [1]. Intracellularly, bicarbonate is generated from the ambient CO2 by the activity of several soluble or membrane-bound isoforms of carbonic anhydrase, although the relative contribution of each of these enzymes is still not defined. Duodenal bicarbonate secretion has been believed to be mediated by transport mechanisms provided by CFTR, and three anion/HCO3 exchangers, SLC26A3 (DRA: downregulated in adenoma), SLC26A6 (PAT1), and SLC4A9 (AE4). The contribution of each of these four pathways to basal and acid stimulated HCO3 secretion as well as their mutual relationship has been a subject of debate in recent years.

CFTR is expressed primarily in the crypt cells whereas the three anion exchangers are expressed at different levels in the brush border membrane of the villus epithelial cells. In a recent report Simpson et al. [2••], using in-vitro techniques, measured anion exchange in the upper villus epithelium of wild type, SLC26A3, SLC26A6, and SLC4A9 knockout mice. AE4 had relatively low expression and did not significantly contribute to basal Cl/HCO3 or SO42−/HCO3 exchange or to changes in intracellular pH (pHi). DRA-deficient mouse epithelium displayed a 30–40% reduction of Cl – but not SO42−-dependent HCO3 flux. Interestingly, the increase in pHi in DRA−/− epithelia suggested that this cytoplasmic alkalinization may be due to disrupted coupling between DRA and Na+/H+ exchange activities. Indeed, a very recent study by Walker et al. [3] elegantly confirmed that DRA couples with NHE3 for electroneutral NaCl absorption across the mammalian small intestine. It remains unclear whether other apical NHE isoforms are in similar relationship with DRA. We have reported that NHE2 (SLC9A2) contributes to small intestinal Na+/H+ exchange, albeit with a rapid decline with age [4]. Similarly, the role of NHE8, also expressed on the apical membrane of the small intestinal epithelial cells [5] has not yet been elucidated in this context. The third anion/HCO3 exchanger, PAT-1, is the most versatile in terms of substrate specificity, demonstrating Cl, SO42−, oxalate, formate, HCO3, and OH transport [6]. In the quoted study by Simpson et al. [2••], PAT-1 was responsible for 65–80% of the Cl -dependent HCO3 flux, and for essentially all of the SO42−-dependent HCO3 flux in the villus epithelial cells. Interestingly, contrary to the observed cytoplasmic alkalinization in DRA-deficient mice, baseline pHi was reduced in PAT-1−/− mice, a phenomenon that could not be attributed to changes in Na+/H+ activity. The authors speculated that PAT-1 might function as a bidirectional exchanger, whereby membrane depolarization, e.g., resulting from electroneutral Na+/H+ exchange would favor PAT-1-mediated HCO3 influx. This would imply that PAT-1 stoichiometry for anion/HCO3 exchange may be electrogenic under certain conditions. This indeed has been described in other systems, although it may not be universal across species, cell type, or the exchange substrates [7-9].

Tightly controlled conditions of an in-vitro study such as Simpson’s [2••] are both an asset and a disadvantage. They do not account for the neural tone, regional (crypt-villus) distribution and activity of transporters, hormone and neurotransmitter receptors or the levels of their ligands under basal and acid-stimulated conditions, etc. Importantly, such focused designs may miss cooperative aspects of transporter function, such as that reported for CFTR and anion/HCO3 exchangers. There is a number of reports emphasizing the role of CFTR in the duodenal bicarbonate transport, which includes not only the passive facilitating coupling effect of CFTR-mediated Cl secretion on Cl/HCO3 exchange, but also active and direct HCO3 conductance. CFTR’s anion selectivity is not intrinsically fixed, but can undergo a dynamic shift to conduct bicarbonate by a process involving ATP hydrolysis, especially when activated by glutamate. A recent article by Singh et al. [10••] attempted to address some of the issues related to the roles of PAT-1 and CFTR in a more physiologically relevant in-vivo settings using luminal perfusion in anesthetized mice. This study utilized CFTR and SLC26A6 knockout mice to investigate the resting, luminal acid stimulated, and high plasma HCO3-stimulated duodenal HCO3 secretion. Contrary to the in-vitro study by Simpson et al. [2••], PAT-1 deficiency resulted in only a slight reduction in basal duodenal HCO3 secretion (by approximately 18% under normal plasma HCO3 concentration), but it almost completely eliminated the increase in HCO3 secretion in response to intraarterial perfusion with bicarbonate. CFTR deficiency similarly blunted the response to high arterial bicarbonate, but it also reduced basal HCO3 secretion by as much as 70%. PAT-1 status had little effect on luminal acidification-induced bicarbonate secretion, suggesting that in vivo, the role of PAT-1 is limited to basal rather than acid-stimulated HCO3 secretion, and that its contribution may be highly dependent on the systemic acid –base status. CFTR deficiency, on the contrary, completely eliminated the effects of luminal acidification on bicarbonate secretion, an effect that was not influenced by arterial HCO3 infusion.

This, like several other studies, points to a critical role of CFTR and its coupling with anion/HCO3 exchange in basal and particularly in acid-stimulated bicarbonate secretion. This finding, however, also takes us back to the ‘CFTR paradox’ formulated by J.D. Kaunitz, that is the lack of mucosal ulcerations in cystic fibrosis patients despite a defect in duodenal HCO3 secretion. It is not inconceivable, that the role of CFTR in murine duodenal mucosa is much greater than that of human, particularly that there are significant species differences in the distribution of CFTR-expressing cells in mice vs. humans or rats. Unlike in humans, mice lack the scattered highly expressing epithelial cell, with more prominent expression in Brunner glands, crypts, and on the villi. CFTR expression in the villus cells is significantly greater than in humans or rats [11]. This could partially explain the phenotypic differences in cystic fibrosis symptoms in human patients and CFTR knockout mice, with the latter more affected by intestinal dysfunction. It has also been postulated, that in duodenal epithelium, CFTR deficiency may be partially protective against an acidic insult due to intracellular accumulation of HCO3 and increased cytosolic buffering capacity. A recent article by Mizumori et al. [12] adds another dimension to the explanation of this ‘CFTR paradox’ by demonstrating that in rats, CFTR inhibition results in increased NHE3 activity. The authors postulate that NHE3-mediated proton extrusion could neutralize luminal HCO3 and facilitate CO2 absorption, which could then serve as a substrate for carbonic anhydrases, or for basolateral NBC1 transporter. An alternative, though not mutually exclusive theory is that HCO3 is taken up by the reverse activity of PAT-1, as we described earlier in this review. One complicating factor stems from a recent demonstration of coupling of NHE3 and DRA activities in the murine small intestine [3]. One could extrapolate the results of this study and hypothesize that an increase in NHE3 activity in response to CFTR inhibition or ablation would be accompanied by DRA-mediated HCO3 secretion, which could, at least to a degree, counteract PAT-1 mediated HCO3 uptake.

In conclusion, the emerging picture of small intestinal ion transport is increasingly complex, with multiple players acting in a synchronized and interdependent manner. New models of targeted gene ablation continue to unravel novel aspects of this integration, as exemplified by the described mechanism of duodenal bicarbonate secretion and absorption.

PDZ protein family and their role in intestinal ion transport

Another plane of structural and functional integration of intestinal ion transporters is provided by PDZ domain-containing proteins. These domains constitute one of the largest families of interaction domains and function by binding the C termini of their target proteins [13,14]. They serve as adapter proteins between integral membrane and cytoskeletal proteins, and have been shown to bind directly to actin in vitro. A group of these proteins, highly concentrated in the apical aspect of polarized epithelial cells, has attracted considerable attention in the field of intestinal and renal ion transport; this includes NHERF-1 (also known as NHERF, EBP50), NHERF-2 (E3KARP), NHERF-3 (CAP70, PDZ-dc1), NHERF-4 (IKEPP), CAL (CFTR-associated ligand), and Shank2/ CortBP1. A comprehensive (change to lower case) overview of this topic is well beyond the limits of this brief article and the reader is referred to two recent reviews published by Lamprecht and Seidler [15], and Donowitz and Li [16]. Here, we will discuss some of the most recent developments describing the structural and functional relationship between PDZ proteins and intestinal ion transporters.

NHERF-1 has been demonstrated to associate with multiple ion transporters in a heterologous expression systems, including the discussed NHE3, DRA, PAT-1, and CFTR. Although NHERF-1 and NHERF-2 were initially described as mediating the inhibitory effects of cAMP on NHE3 activity and Na+ uptake in vitro, and as potentially viable targets of antidiarrheal therapies, a recently published report utilizing NHERF-1 deficient mice demonstrates reduced small intestinal fluid absorption in vivo, as well as reduced in-vitro Na+ absorption in isolated jejunal and colonic, though not ileal, mucosa [17••]. Moreover, contrary to renal proximal tubules [18••] and jejunum [17••], cAMP-mediated inhibition of both, ileal Na+/H+ exchange and fluid absorption remained unaffected by NHERF-1 ablation [17••,18••]. These new findings point to a tissue-specific and segment-specific involvement of NHERF-1 in the regulation of water and electrolyte transport, as well as to a potentially profound impact of this scaffolding protein in diarrheal disorders. Indeed, a very recent report by Sullivan et al. [19••] suggests that inflammation (IBD)–associated diarrhea may be related to reduced expression of NHERF-1 and NHERF-2 in the ileal and colonic mucosa.

NHERF-1 and NHERF-2 are known to enhance CFTR-mediated Cl secretion due to the anchoring of protein kinase A (PKA) to the CFTR complex. NHERF-1 and NHERF-2 have also been described to reduce CFTR endosomal recycling and lysosomal degradation by interfering with the negative effects of CAL, a phenomenon facilitated by the recently illustrated low affinity of the CAL–CFTR interaction compared with NHERF–CFTR binding [20]. This mechanism of CFTR regulation may explain the observed decrease in forskolin-stimulated Cl secretion in NHERF-1-deficient mice [17••]. Somewhat unexpectedly, however, NHERF-2 deficiency had no effect on cyclic nucleotide-induced CFTR activation in mouse jejunum, a phenomenon that may be related to the described very low expression of this adapter protein in the mouse small intestinal epithelium [21].

In summary, some of the new in-vivo findings revisit and revise the roles of the PDZ proteins in epithelial ion transport, bringing new complexity to the involvement of these crucial adapter proteins in intestinal electrolyte and water homeostasis. The unraveling of the intricate network of functional and structural relationships continues to provide clues into their tissue-specific, segment-specific, and transporter-specific roles.

Regulation of intestinal ion transport in inflammatory states

In conjunction with the kidney, the gastrointestinal tract is uniquely positioned, both structurally and functionally, to maintain the homeostatic control of electrolyte balance and the ensuing fluid homeostasis and passive mucosal defense. Under normal circumstances in an adult human, the gastrointestinal tract handles an average luminal daily load of 9 L of water and 800 mEq of Na+ originating from ingestion as well as secretory fluids. Small intestinal absorption of water is largely secondary and controlled by ion flux across the epithelial barrier. Small intestine has a substantial capacity to absorb osmolytes and water, with nearly 80% of the daily load absorbed and approximately 2 L/day of resulting ileocecal flow. Large intestine has the capacity to absorb 1.5–1.9 L/day, although in pathological states with defective small intestinal absorption, the initiated adaptive mechanisms allow the colon to compensate with absorption rate reaching 5–6 L/day. Exceeding this maximal capacity will result in diarrhea, commonly observed in response to intestinal inflammation such as in chronic inflammatory bowel diseases or secondary to microbial infections. Pathogenic mechanisms involved in infectious diarrhea have been recently reviewed by Navaneethan and Giannella [22] and will not be discussed in detail. The reader is also referred to a recent review on the altered ion transport in IBD by Martinez –Augustin et al. [23]. Several key findings have been recently described that contribute to our understanding of the pathogenesis of inflammation-associated diarrhea. Intestinal electroneutral Na+/H+ exchange, mediated primarily by NHE3, is known to be negatively affected by colonic inflammation and inflammatory mediators (nitric oxide, TNFa, IFN-g) as well as enter-opathogenic bacteria [24]. In a recent report, Sullivan et al. [19••] describes that in ileal or sigmoid biopsies of a considerable number of patients with active Crohn’s disease or ulcerative colitis, there is reduced expression of NHE3, NHERF-1, NHE1, epithelial sodium channel (EnaC) (Na+ channel), and CLC-5 protein, with the latter suggested to participate in endosomal NHE3 trafficking rather than epithelial Cl conductance. These findings were recapitulated in mouse models of chemically induced colitis [19••], thus further highlighting and explaining the role of altered transepithelial Na+ and fluid transport in IBD-associated diarrhea. Colitis or systemic inflammation may also affect small intestinal ion transport as recently demonstrated for ileal bicarbonate transport [25] and NHE8 expression in the jejunum (unpublished data and [26]). Our group has also recently described spontaneous development of distal colitis in NHE3-deficient mice, and suggested the role of NHE3 as a modifier gene in intestinal inflammation [27]. We have also presented data demonstrating severe susceptibility of NHE3 knockout mice to DSS-induced mucosal injury, which unprecedently extends to the small intestine (unpublished data and [28••]). These findings, combined, strongly suggest a vital role of NHE3 not only in the pathogenesis of inflammation-associated diarrhea, but also in maintenance of epithelial integrity and perhaps in modulating mucosal inflammation and injury when down regulated.

In summary, intestinal inflammation is associated with defects in epithelial barrier function and ion flux, both contributing to impaired fluid homeostasis and diarrhea. Disturbances in ion and fluid absorption are less well characterized but the emerging data points to both the electroneutral and electrogenic Na+ absorption. Infammation and inflammatory mediators are associated with changes in the expression and function of key epithelial transporters, including the Na+/K+ ATPase, sodium/potassium/chloride cotransporter 1 (NKCC1), sodium/hydrogen exchangers, and the ENaC. There is also emerging data suggesting that those changes may negatively feed back to the ability to maintain epithelial integrity in the face of microbial or noxious challenge. Further studies will be needed to provide a more comprehensive picture of the role of ion transport in intestinal inflammation.

Conclusion

Although recent months have not yielded major break-throughs in the field of physiology and pathophysiology of intestinal ion transport, gene targeting and generated mouse models deficient in some of the key genes encoding transporters and their adapter proteins continue to provide important clues into intestinal transport processes. Several of the new in-vivo findings revise or complement past paradigms, many of which were derived mostly from in-vitro approaches. Novel information on the interdependent functions of multiple transporters, as exemplified here by intestinal bicarbonate secretion, increases the complexity of the intestinal ion transport mechanisms and continues to contribute to a more integrated view of the transport phenomena in the gut. Data from patients and mouse models of intestinal inflammation also increase our understanding of the pathophysiology and etiology of inflammation-associated diarrhea.

Acknowledgements

Work in the authors’ laboratory is supported by the National Institutes of Health (2R01DK041274).

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

• of special interest

•• of outstanding interest

Additional references related to this topic can also be found in the Current World Literature section in this issue (pp. 160 –161).

  • 1.Jacob P, Christiani S, Rossmann H, et al. Role of Na(+)HCO(3)(−) cotransporter NBC1, Na(+)/H(+) exchanger NHE1, and carbonic anhydrase in rabbit duodenal bicarbonate secretion. Gastroenterology. 2000;119:406–419. doi: 10.1053/gast.2000.9358. [DOI] [PubMed] [Google Scholar]
  • 2.Simpson JE, Schweinfest CW, Shull GE, et al. PAT-1 (Slc26a6) is the predominant apical membrane Cl−/HCO3− exchanger in the upper villous epithelium of the murine duodenum. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1079–1088. doi: 10.1152/ajpgi.00354.2006. •• This in-vitro study demonstrates that in the upper duodenal villi, PAT-1 is the major contributor to basal Cl−/HCO3− and SO42−/HCO3− exchange and that it plays a significant role in regulating intracellular pH. [DOI] [PubMed] [Google Scholar]
  • 3.Walker NM, Simpson JE, Yen PF, et al. Down-regulated in Adenoma Cl/HCO(3) Exchanger Couples with Na/H Exchanger 3 for NaCl Absorption in Murine Small Intestine. Gastroenterology. 2008;135:1645–1653. doi: 10.1053/j.gastro.2008.07.083. • These studies utilizing DRA−/− and PAT-1−/− mice provide functional evidence that DRA is the major Cl−/HCO3− exchanger coupled with NHE3 for electroneutral NaCl absorption across mammalian small intestine. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Collins JF, Kiela PR, Xu H, et al. Increased NHE2 expression in rat intestinal epithelium during ontogeny is transcriptionally mediated. Am J Physiol. 1998;275:C1143–1150. doi: 10.1152/ajpcell.1998.275.4.C1143. [DOI] [PubMed] [Google Scholar]
  • 5.Xu H, Chen H, Dong J, et al. Gastrointestinal distribution and kinetic characterization of the sodium-hydrogen exchanger isoform 8 (NHE8) Cell Physiol Biochem. 2008;21:109–116. doi: 10.1159/000113752. • This paper demonstrates expression of a novel nhe isoform (NHE8) along the human and mouse gastrointestinal tract and provides evidence of Na+/H+ exchange kinetics different from other apically expressed NHE isoforms. [DOI] [PubMed] [Google Scholar]
  • 6.Dorwart MR, Shcheynikov N, Yang D, Muallem S. The solute carrier 26 family of proteins in epithelial ion transport. Physiology (Bethesda) 2008;23:104–114. doi: 10.1152/physiol.00037.2007. [DOI] [PubMed] [Google Scholar]
  • 7.Shcheynikov N, Wang Y, Park M, et al. Coupling modes and stoichiometry of Cl−/HCO3− exchange by slc26a3 and slc26a6. J Gen Physiol. 2006;127:511–524. doi: 10.1085/jgp.200509392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Niederer SA, Swietach P, Wilson DA, et al. Measuring and modeling chloridehydroxyl exchange in the Guinea-pig ventricular myocyte. Biophys J. 2008;94:2385–2403. doi: 10.1529/biophysj.107.118885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Clark JS, Vandorpe DH, Chernova MN, et al. Species differences in Cl− affinity and in electrogenicity of SLC26A6-mediated oxalate/Cl-exchange correlate with the distinct human and mouse susceptibilities to nephrolithiasis. J Physiol. 2008;586:1291–1306. doi: 10.1113/jphysiol.2007.143222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Singh AK, Sjoblom M, Zheng W, et al. CFTR and its key role in in vivo resting and luminal acid-induced duodenal HCO3− secretion. Acta Physiol (Oxf) 2008;193:357–365. doi: 10.1111/j.1748-1716.2008.01854.x. ••This report provides in-vivo evidence for coupled activities of Slc26a6 (PAT-1) with CFTR in murine basal, but not acid-stimulated duodenal bicarbonate secretion. [DOI] [PubMed] [Google Scholar]
  • 11.Ameen N, Alexis J, Salas P. Cellular localization of the cystic fibrosis transmembrane conductance regulator in mouse intestinal tract. Histochem Cell Biol. 2000;114:69–75. doi: 10.1007/s004180000164. [DOI] [PubMed] [Google Scholar]
  • 12.Mizumori M, Choi Y, Guth PH, et al. CFTR inhibition augments NHE3 activity during luminal high CO2 exposure in rat duodenal mucosa. Am J Physiol Gastrointest Liver Physiol. 2008;294:G1318–1327. doi: 10.1152/ajpgi.00025.2008. [DOI] [PubMed] [Google Scholar]
  • 13.Chen JR, Chang BH, Allen JE, et al. Predicting PDZ domain-peptide interactions from primary sequences. Nat Biotechnol. 2008;26:1041–1045. doi: 10.1038/nbt.1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Pawson T, Nash P. Assembly of cell regulatory systems through protein interaction domains. Science. 2003;300:445–452. doi: 10.1126/science.1083653. [DOI] [PubMed] [Google Scholar]
  • 15.Lamprecht G, Seidler U. The emerging role of PDZ adapter proteins for regulation of intestinal ion transport. Am J Physiol Gastrointest Liver Physiol. 2006;291:G766–777. doi: 10.1152/ajpgi.00135.2006. • A comprehensive review covering the roles of PDZ proteins in the regulation of intestinal anion secretion and electroneutral NaCl absorption. [DOI] [PubMed] [Google Scholar]
  • 16.Donowitz M, Li X. Regulatory binding partners and complexes of NHE3. Physiol Rev. 2007;87:825–872. doi: 10.1152/physrev.00030.2006. • A more recent review focuses on the roles of PDZ proteins and their signaling partners in the regulation of NHE3 trafficking and activity. [DOI] [PubMed] [Google Scholar]
  • 17.Broere N, Chen M, Cinar A, et al. Defective jejunal and colonic salt absorption and alteredNa(+)/H (+) exchanger 3 (NHE3) activity in NHE regulatory factor 1 (NHERF1) adaptor protein-deficient mice. Pflugers Arch. 2009 doi: 10.1007/s00424-008-0579-1. in press).•• This study provides in-vivo evidence for the segment-specific involvement of NHERF-1 in intestinal salt absorption, NHE3 activity and membrane expression, and postulates its limited involvement in regulation of NHE3 by cyclic nucleotides in vivo. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Murtazina R, Kovbasnjuk O, Zachos NC, et al. Tissue-specific regulation of sodium/proton exchanger isoform 3 activity in Na(+)/H(+) exchanger regulatory factor 1 (NHERF1) null mice. cAMP inhibition is differentially dependent on NHERF1 and exchange protein directly activated by cAMP in ileum versus proximal tubule. J Biol Chem. 2007;282:25141–25151. doi: 10.1074/jbc.M701910200. •• This report compares the role of NHERF-1 in cyclic nucleotide-mediated downregulation of NHE3 in the renal proximal tubules and ileum, and suggests that cAMP inhibition of NHE3 activity in these two tissues is mediated by different mechanisms. [DOI] [PubMed] [Google Scholar]
  • 19.Sullivan S, Alex P, Dassopoulos T, et al. Downregulation of sodium transporters and NHERF proteins in IBD patients and mouse colitis models: potential contributors to IBD-associated diarrhea. Inflamm Bowel Dis. 2009;15:261–274. doi: 10.1002/ibd.20743. •• Data derived from human patients and mouse models of colitis demonstrates that IBD-associated diarrhea may be due to a coordinated downregulation of multiple Na+ transporters and related regulatory proteins, including NHE1, NHE3, Na+/K+-ATPase, and ENaC, as well as NHERF1, 2, and ClC-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Cushing PR, Fellows A, Villone D, et al. The relative binding affinities of PDZ partners for CFTR: a biochemical basis for efficient endocytic recycling. Biochemistry. 2008;47:10084–10098. doi: 10.1021/bi8003928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Broere N, Hillesheim J, Tuo B, et al. Cystic fibrosis transmembrane conductance regulator activation is reduced in the small intestine of Na+/H+ exchanger 3 regulatory factor 1 (NHERF-1)-but Not NHERF-2-deficient mice. J Biol Chem. 2007;282:37575–37584. doi: 10.1074/jbc.M704878200. • These studies, performed with NHERF-1 and NHERF-2 knockout mice, provide evidence that NHERF-1 but not NHERF-2 is involved in the formation of CFTR-containing functional complexes responsible for membrane targeting and fine tuning of CFTR function as a cyclic nucleotide-regulated anion channel. [DOI] [PubMed] [Google Scholar]
  • 22.Navaneethan U, Giannella RA. Mechanisms of infectious diarrhea. Nat Clin Pract Gastroenterol Hepatol. 2008;5:637–647. doi: 10.1038/ncpgasthep1264. • A useful review of the mechanisms of diarrhea caused by various pathogens. [DOI] [PubMed] [Google Scholar]
  • 23.Martinez-Augustin O, Romero-Calvo I, Suarez MD, et al. Molecular bases of impaired water and ion movements in inflammatory bowel diseases. Inflamm Bowel Dis. 2009;15:114–127. doi: 10.1002/ibd.20579. • A recent summary on the disturbances in intestinal ion transport in inflammatory bowel diseases. [DOI] [PubMed] [Google Scholar]
  • 24.Hodges K, Alto NM, Ramaswamy K, et al. The enteropathogenic Escherichia coli effector protein EspF decreases sodium hydrogen exchanger 3 activity. Cell Microbiol. 2008;10:1735–1745. doi: 10.1111/j.1462-5822.2008.01163.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhang H, Ameen N, Melvin JE, Vidyasagar S. Acute inflammation alters bicarbonate transport in mouse ileum. J Physiol. 2007;581:1221–1233. doi: 10.1113/jphysiol.2007.129262. • This interesting, though somewhat limited study, demonstrates that CD3 antibodymediated inflammation alters ileal bicarbonate transport in mice and changes primarily electroneutral Cl−/HCO3− exchange to a predominantly electrogenic CFTR-mediated anion secretion. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Xu H, Chen H, Dong J, Ghishan FK. TNF-a Inhibits Human Intestinal NHE8 Gene Expression in Caco-2 via Tyrosine Kinase Pathway. In Digestive Disease Week Meeting/109th Annual Meeting of the American-Gastroenterological-Association; San Diego, CA: May 17-22, 2008. 2008. p. A-56. [Google Scholar]
  • 27.Laubitz D, Larmonier CB, Bai A, et al. Colonic gene expression profile in NHE3-deficient mice: evidence for spontaneous distal colitis. Am J Physiol Gastrointest Liver Physiol. 2008;295:G63–G77. doi: 10.1152/ajpgi.90207.2008. • In this report we demonstrated for the first time, that NHE3 deficiency results in inflammatory changes in the distal colon, DATA suggesting a novel role for NHE3 beyond vectorial Na+ transport. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kiela PR, Larmonier CB, Midura-Kiela MT, Thurston RD, Ghishan FK. Hypersensitivity to DSS-induced mucosal injury in NHE3 knockout mice. In Digestive Disease Week Meeting/108th Annual Meeting of the American-Gastroenterological-Association; Washington, DC, USA: May 19-24, 2007. 2007. p. A396. Gastroenterology. •• In this study, we demonstrate that changes in the small intestinal and colonic gene expression profile dramatically alter the mucosal response to DSS-mediated epithelial injury, and for the first time show that the small intestine is affected by DSS dependent on the NHE3 status. [Google Scholar]

RESOURCES