Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Regina Engelhardt is active.

Publication


Featured researches published by Regina Engelhardt.


Kidney International | 2008

Fructose-induced hypertension: essential role of chloride and fructose absorbing transporters PAT1 and Glut5

Anurag Singh; Hassane Amlal; Patrick J. Haas; Ulrike Dringenberg; Stacey Fussell; Sharon Barone; Regina Engelhardt; Jian Zuo; Ursula Seidler; Manoocher Soleimani

Increased dietary fructose in rodents recapitulates many aspects of the Metabolic Syndrome with hypertension, insulin resistance and dyslipidemia. Here we show that fructose increased jejunal NaCl and water absorption which was significantly decreased in mice whose apical chloride/base exchanger Slc26a6 (PAT1, CFEX) was knocked out. Increased dietary fructose intake enhanced expression of this transporter as well as the fructose-absorbing transporter Slc2a5 (Glut5) in the small intestine of wild type mice. Fructose feeding decreased salt excretion by the kidney and resulted in hypertension, a response almost abolished in the knockout mice. In parallel studies, a chloride-free diet blocked fructose-induced hypertension in Sprague Dawley rats. Serum uric acid remained unchanged in animals on increased fructose intake with hypertension. We suggest that fructose-induced hypertension is likely caused by increased salt absorption by the intestine and kidney and the transporters Slc26a6 and Slc2a5 are essential in this process.


Pflügers Archiv: European Journal of Physiology | 2009

Defective jejunal and colonic salt absorption and alteredNa +/H+ exchanger 3 (NHE3) activity in NHE regulatory factor 1 (NHERF1) adaptor protein-deficient mice

N. Broere; Mingmin Chen; Ayhan Cinar; Anurag Singh; Jutta Hillesheim; Brigitte Riederer; Maria Lünnemann; I. Rottinghaus; Anja Krabbenhöft; Regina Engelhardt; Brigitte Rausch; E. J. Weinman; Mark Donowitz; Ann L. Hubbard; Olivier Kocher; H.R. de Jonge; Boris M. Hogema; Ursula Seidler

We investigated the role of the Na+/H+ exchanger regulatory factor 1 (NHERF1) on intestinal salt and water absorption, brush border membrane (BBM) morphology, and on the NHE3 mRNA expression, protein abundance, and transport activity in the murine intestine. NHERF1-deficient mice displayed reduced jejunal fluid absorption in vivo, as well as an attenuated in vitro Na+ absorption in isolated jejunal and colonic, but not of ileal, mucosa. However, cAMP-mediated inhibition of both parameters remained intact. Acid-activated NHE3 transport rate was reduced in surface colonocytes, while its inhibition by cAMP and cGMP was normal. Immunodetection of NHE3 revealed normal NHE3 localization in the BBM of NHERF1 null mice, but NHE3 abundance, as measured by Western blot, was significantly reduced in isolated BBM from the small and large intestines. Furthermore, the microvilli in the proximal colon, but not in the small intestine, were significantly shorter in NHERF1 null mice. Additional knockout of PDZK1 (NHERF3), another member of the NHERF family of adaptor proteins, which binds to both NHE3 and NHERF1, further reduced basal NHE3 activity and caused complete loss of cAMP-mediated NHE3 inhibition. An activator of the exchange protein activated by cAMP (EPAC) had no effect on jejunal fluid absorption in vivo, but slightly inhibited NHE3 activity in surface colonocytes in vitro. In conclusion, NHERF1 has segment-specific effects on intestinal salt absorption, NHE3 transport rates, and NHE3 membrane abundance without affecting mRNA levels. However, unlike PDZK1, NHERF1 is not required for NHE3 regulation by cyclic nucleotides.


American Journal of Physiology-cell Physiology | 2010

The switch of intestinal Slc26 exchangers from anion absorptive to HCO3- secretory mode is dependent on CFTR anion channel function

Anurag K. Singh; Brigitte Riederer; Mingmin Chen; Fang Xiao; Anja Krabbenhöft; Regina Engelhardt; Olof Nylander; Manoocher Soleimani; Ursula Seidler

CFTR has been recognized to function as both an anion channel and a key regulator of Slc26 anion transporters in heterologous expression systems. Whether this regulatory relationship between CFTR and Slc26 transporters is seen in native intestine, and whether this effect is coupled to CFTR transport function or other features of this protein, has not been studied. The duodena of anesthetized CFTR-, NHE3-, Slc26a6-, and Scl26a3-deficient mice and wild-type (WT) littermates were perfused, and duodenal bicarbonate (HCO(3)(-)) secretion (DBS) and fluid absorptive or secretory rates were measured. The selective NHE3 inhibitor S1611 or genetic ablation of NHE3 significantly reduced fluid absorptive rates and increased DBS. Slc26a6 (PAT1) or Slc26a3 (DRA) ablation reduced the S1611-induced DBS increase and reduced fluid absorptive rates, suggesting that the effect of S1611 or NHE3 ablation on HCO(3)(-) secretion may be an unmasking of Slc26a6- and Slc26a3-mediated Cl(-)/HCO(3)(-) exchange activity. In the absence of CFTR expression or after application of the CFTR(inh)-172, fluid absorptive rates were similar to those of WT, but S1611 induced virtually no increase in DBS, demonstrating that CFTR transport activity, and not just its presence, is required for Slc26-mediated duodenal HCO(3)(-) secretion. A functionally active CFTR is an absolute requirement for Slc26-mediated duodenal HCO(3)(-) secretion, but not for Slc26-mediated fluid absorption, in which these transporters operate in conjunction with the Na(+)/H(+) exchanger NHE3. This suggests that Slc26a6 and Slc26a3 need proton recycling via NHE3 to operate in the Cl(-) absorptive mode and Cl(-) exit via CFTR to operate in the HCO(3)(-) secretory mode.


Pflügers Archiv: European Journal of Physiology | 2007

Sodium and chloride absorptive defects in the small intestine in Slc26a6 null mice

Ursula Seidler; Ingrid Rottinghaus; Jutta Hillesheim; Mingmin Chen; Brigitte Riederer; Anja Krabbenhöft; Regina Engelhardt; Martin Wiemann; Zhaouhui Wang; Sharon Barone; Michael P. Manns; Manoocher Soleimani

PAT1 (Slc26a6) is located on the apical membrane of the small intestinal villi, but its role for salt absorption has not been studied. To ascertain the role of Slc26a6 in jejunal sodium and chloride absorption, and its interplay with NHE3, muscle-stripped jejuna from Slc26a6+/+ and −/− and NHE3 +/+ and −/− mice were mounted in Ussing chambers and electrical parameters, and 36Cl− and 22Na+ fluxes were measured. In parallel studies, expression of the apical Na+/H+ exchanger (NHE3) was examined by immunofluorescence labeling and immunoblot analysis in brush border membrane (BBM). In the basal state, net Cl− and Na+ fluxes were absorptive in Slc26a6−/− and +/+ jejuni, but significantly decreased in −/− animals. Upon forskolin addition, net Na+ absorption decreased, Isc strongly increased, and net Cl− flux became secretory in Slc26a6−/− and +/+ jejuni. When luminal glucose was added to activate Na+/glucose cotransport, concomitant Cl− absorption was significantly reduced in Slc26a6 −/− jejuni, while Na+ absorption increased to the same degree in Slc26a6 −/− and +/+ jejuni. Identical experiments in NHE3-deficient jejuni also showed reduced Na+ and Cl− absorption. Results further demonstrated that the lack of NHE3 rendered Na+ and Cl− absorption unresponsive to inhibition by cAMP, but did not affect glucose-driven Na+ and Cl− absorption. Immunoblotting revealed comparable NHE3 abundance and distribution in apical membranes in Slc26a6−/− and +/+ mice. The data strongly suggests that Slc26a6 acts in concert with NHE3 in electroneutral salt absorption in the small intestine. Slc26a6 also serves to absorb Cl− during glucose-driven salt absorption.


Gastroenterology | 2008

Localization, Trafficking, and Significance for Acid Secretion of Parietal Cell Kir4.1 and KCNQ1 K+ Channels

Marc Kaufhold; Anja Krabbenhöft; Penghong Song; Regina Engelhardt; Brigitte Riederer; Michael Fährmann; Nikolaj Klöcker; Winfried Beil; Michael P. Manns; Susan J. Hagen; Ursula Seidler

BACKGROUND & AIMS K(+) recycling at the apical membrane of gastric parietal cells is a prerequisite for gastric acid secretion. Two K(+) channels are currently being considered for this function, namely KCNQ1 and inwardly rectifying K(+) channels (Kir). This study addresses the subcellular localization, trafficking, and potential functional significance of KCNQ1 and Kir4.1 channels during stimulated acid secretion. METHODS The effect of pharmacologic KCNQ1 blockade on acid secretion was studied in cultured rat and rabbit parietal cells and in isolated mouse gastric mucosa. The subcellular localization of KCNQ1 and Kir4.1 was determined in highly purified membrane fractions by Western blot analysis as well as in fixed and living cells by confocal microscopy. RESULTS In cultured parietal cells and in isolated gastric mucosa, a robust acid secretory response was seen after complete pharmacologic blockade of KCNQ1. Both biochemical and morphologic data demonstrate that Kir4.1 and KCNQ1 colocalize with the H(+)/K(+)-ATPase but do so in different tubulovesicular pools. All Kir4.1 translocates to the apical membrane after stimulation in contrast to only a fraction of KCNQ1, which mostly remains cytoplasmic. CONCLUSIONS Acid secretion can be stimulated after complete pharmacologic blockade of KCNQ1 activity, suggesting that additional apical K(+) channels regulate gastric acid secretion. The close association of Kir4.1 channels with H(+)/K(+)-ATPase in the resting and stimulated membrane suggests a possible role for Kir4.1 channels during the acid secretory cycle.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2010

Gene ablation for PEPT1 in mice abolishes the effects of dipeptides on small intestinal fluid absorption, short-circuit current, and intracellular pH

Mingmin Chen; Anurag K. Singh; Fang Xiao; Ulrike Dringenberg; Jian Wang; Regina Engelhardt; Sunil Yeruva; Isabel Rubio-Aliaga; Anna-Maria Näßl; Gabor Kottra; Hannelore Daniel; Ursula Seidler

PEPT1 function in mouse intestine has not been assessed by means of electrophysiology and methods to assess its role in intracellular pH and fluid homeostasis. Therefore, the effects of the dipeptide glycilsarcosin (Gly-Sar) on jejunal fluid absorption and villous enterocyte intracellular pH (pH(i)) in vivo, as well as on enterocyte[(14)C]Gly-Sar uptake, short-circuit current (I(sc)) response, and enterocyte pH(i) in vitro were determined in wild-type and PEPT1-deficient mice and in mice lacking PEPT1. Immunohistochemistry for PEPT1 failed to detect any protein in enterocyte apical membranes in Slc15a1(-/-) animals. Saturable Gly-Sar uptake in Slc15a1(-/-) everted sac preparations was no longer detectable. Similarly, Gly-Sar-induced jejunal I(sc) response in vitro was abolished. The dipeptide-induced increase in fluid absorption in vivo was also abolished in animals lacking PEPT1. Since PEPT1 acts as an acid loader in enterocytes, enterocyte pH(i) was measured in vivo by two-photon microscopy in SNARF-4-loaded villous enterocytes of exteriorized jejuni in anesthetized mice, as well as in BCECF-loaded enterocytes of microdissected jejunal villi. Gly-Sar-induced pH(i) decrease was no longer observed in the absence of PEPT1. A reversal of the proton gradient across the luminal membrane did not significantly diminish Gly-Sar-induced I(sc) response, whereas a depolarization of the apical membrane potential by high K(+) or via Na(+)-K(+)-ATPase inhibition strongly diminished Gly-Sar-induced I(sc) responses. This study demonstrates for the first time that proton-coupled electrogenic dipeptide uptake in the native small intestine, mediated by PEPT1, relies on the negative apical membrane potential as the major driving force and contributes significantly to intestinal fluid transport.


Acta Physiologica | 2014

Slc26a3 deficiency is associated with loss of colonic HCO3− secretion, absence of a firm mucus layer and barrier impairment in mice

Fang Xiao; Q. Yu; Junhua Li; Malin E. V. Johansson; Anurag Singh; Weiliang Xia; Brigitte Riederer; Regina Engelhardt; Marshall H. Montrose; Manoocher Soleimani; Dean Tian; Gang Xu; Gunnar C. Hansson; Ursula Seidler

Downregulated in adenoma (DRA, Slc26a3) is a member of the solute carrier family 26 (SLC26), family of anion transporters, which is mutated in familial chloride‐losing diarrhoea (CLD). Besides Cl−‐rich diarrhoea, CLD patients also have a higher‐than‐average incidence of intestinal inflammation. In a search for potential explanations for this clinical finding, we investigated colonic electrolyte transport, the mucus layer and susceptibility against dextran sodium sulphate (DSS)‐induced colitis in Slc26a3−/− mice.


The Journal of Physiology | 2013

Molecular transport machinery involved in orchestrating luminal acid-induced duodenal bicarbonate secretion in vivo

Anurag K. Singh; Yongjian Liu; Brigitte Riederer; Regina Engelhardt; Basant Kumar Thakur; Manoocher Soleimani; Ursula Seidler

•  Acid damage of the proximal duodenum is a key pathogenic factor in duodenal ulcer disease as well as in the intestinal manifestations of cystic fibrosis. •  Short contact of healthy duodenal mucosa with acid results in long‐lasting stimulation of proximal duodenal bicarbonate secretion. •  While the complex neural, paracrine, humoral and luminocrine regulation of this acid‐induced bicarbonate secretory response, as well as its protective role, has been studied in some detail, little is known about the molecular identity of the involved ion transporters or intracellular signalling. •  Using genetically engineered mouse models, we found that the anion exchanger DRA (Slc26a3), the anion conductances Slc26a9 and cystic fibrosis transmembrane conductance regulator, and the Na+/H+ exchanger isoform 3 play essential roles in orchestrating the acid‐induced duodenal bicarbonate secretory response. These transporters are differentially controlled by signalling mechanisms along the crypt–villus axis. •  These findings provide a better understanding of the pathophysiology of peptic damage to the duodenum and may provide novel treatment strategies.


Critical Care Medicine | 2014

The role of pancreatic ductal secretion in protection against acute pancreatitis in mice

Petra Pallagi; Zsolt Balla; Anurag Singh; Sándor Dósa; Béla Iványi; Zoltán Kukor; Adél Tóth; Brigitte Riederer; Yongjian Liu; Regina Engelhardt; K. Jármay; Andrea Szabó; Ágnes Janovszky; George Perides; Viktória Venglovecz; József Maléth; Tibor Wittmann; Tamás Takács; Michael A. Gray; Attila Gácser; Péter Hegyi; Ursula Seidler; Zoltán Rakonczay

Objectives:A common potentially fatal disease of the pancreas is acute pancreatitis, for which there is no treatment. Most studies of this disorder focus on the damage to acinar cells since they are assumed to be the primary target of multiple stressors affecting the pancreas. However, increasing evidence suggests that the ducts may also have a crucial role in induction of the disease. To test this hypothesis, we sought to determine the specific role of the duct in the induction of acute pancreatitis using well-established disease models and mice with deletion of the Na+/H+ exchanger regulatory factor-1 that have selectively impaired ductal function. Design:Randomized animal study. Setting:Animal research laboratory. Subjects:Wild-type and Na+/H+ exchanger regulatory factor-1 knockout mice. Interventions:Acute necrotizing pancreatitis was induced by i.p. administration of cerulein or by intraductal administration of sodium taurocholate. The pancreatic expression of Na+/H+ exchanger regulatory factor-1 and cystic fibrosis transmembrane conductance regulator (a key player in the control of ductal secretion) was analyzed by immunohistochemistry. In vivo pancreatic ductal secretion was studied in anesthetized mice. Functions of pancreatic acinar and ductal cells as well as inflammatory cells were analyzed in vitro. Measurements and Main Results:Deletion of Na+/H+ exchanger regulatory factor-1 resulted in gross mislocalization of cystic fibrosis transmembrane conductance regulator, causing marked reduction in pancreatic ductal fluid and bicarbonate secretion. Importantly, deletion of Na+/H+ exchanger regulatory factor-1 had no deleterious effect on functions of acinar and inflammatory cells. Deletion of Na+/H+ exchanger regulatory factor-1, which specifically impaired ductal function, increased the severity of acute pancreatitis in the two mouse models tested. Conclusions:Our findings provide the first direct evidence for the crucial role of ductal secretion in protecting the pancreas from acute pancreatitis and strongly suggest that improved ductal function should be an important modality in prevention and treatment of the disease.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2012

Telomere shortening is associated with reduced duodenal HCO3− secretory but normal gastric acid secretory capacity in aging mice

Biguang Tuo; Zhenyu Ju; Brigitte Riederer; Regina Engelhardt; Michael P. Manns; K. Lenhard Rudolph; Ursula Seidler

The incidence of duodenal ulcer, especially Helicobacter pylori-negative duodenal ulcer, strongly increases with age. In humans, telomere length shortening is considered to be one critical factor in cellular senescence and organ survival. In this study, we compared basal and stimulated gastric acid and duodenal HCO(3)(-) secretory rates in aged late-generation (G(3)) telomerase-deficient (mTERC(-/-)) mice, which are characterized by severe telomere dysfunction due to the inability to elongate telomeres during cell division. We found that basal and forskolin-stimulated HCO(3)(-) secretion and short-circuit current (I(sc)) in isolated duodenal mucosa of G(3) mTERC(-/-) mice were markedly reduced compared with age-matched wild-type mice. In contrast, basal and forskolin-stimulated acid secretory rates in isolated G(3) mTERC(-/-) gastric mucosa were not significantly altered. Correspondingly, duodenal mucosa of G(3) mTERC(-/-) mice showed slimming and shortening of villi, whereas gastric mucosal histology was not significantly altered. However, the ratios of cystic fibrosis transmembrane conductance regulator (CFTR) and solute-linked carrier 26 gene family (Slc26a6) mRNA expression in relation to cytokeratin-18 were not altered in duodenal mucosa. The further knockout of p21, which is a downstream effector of telomere shortening-induced senescence, rescued villus atrophy of duodenal mucosa, and basal and forskolin-stimulated duodenal HCO(3)(-) secretion and I(sc) in mTERC(-/-) p21(-/-) double-knockout mice were not different from wild-type controls. In conclusion, genetic ablation of telomerase resulted in p21-dependent duodenal mucosal atrophy and reduced duodenal HCO(3)(-) secretory capacity, whereas gastric morphology and acid secretory function were preserved. This suggests that telomere shortening during aging may result in an imbalance between aggressive and protective secretions against duodenal mucosa and thus predispose to ulcer formation.

Collaboration


Dive into the Regina Engelhardt's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anurag K. Singh

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Mingmin Chen

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anurag Singh

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fang Xiao

Hannover Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge