Network


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

Hotspot


Dive into the research topics where Edward J. Weinman is active.

Publication


Featured researches published by Edward J. Weinman.


Nature | 1998

The beta2-adrenergic receptor interacts with the Na+/H+-exchanger regulatory factor to control Na+/H+ exchange.

Randy A. Hall; Richard T. Premont; Chung-Wai Chow; Jeremy T. Blitzer; Julie A. Pitcher; Audrey Claing; Robert H. Stoffel; Larry S. Barak; Shirish Shenolikar; Edward J. Weinman; Sergio Grinstein; Robert J. Lefkowitz

Stimulation of β2-adrenergic receptors on the cell surface by adrenaline or noradrenaline leads to alterations in the metabolism, excitability, differentiation and growth of many cell types. These effects have traditionally been thought to be mediated exclusively by receptor activation of intracellular G proteins. However, certain physiological effects of β2-adrenergic receptor stimulation, notably the regulation of cellular pH by modulation of Na+/H+ exchanger (NHE) function, do not seem to be entirely dependent on G-protein activation. We report here a direct agonist-promoted association of the β2-adrenergic receptor with the Na+/H+ exchanger regulatory factor (NHERF), a protein that regulates the activity of the Na+/H+ exchanger type 3 (NHE3). NHERF binds to the β2-adrenergic receptor by means of a PDZ-domain-mediated interaction with the last few residues of the carboxy-terminal cytoplasmic domain of the receptor. Mutation ofthe final residue of the β2-adrenergic receptor from leucine toalanine abolishes the receptors interaction with NHERF andalso markedly alters β2-adrenergic receptor regulation of NHE3 in cells without altering receptor-mediated activation of adenylyl cyclase. Our findings indicate that agonist-dependent β2-adrenergic receptor binding of NHERF plays a role in β2-adrenergic receptor-mediated regulation of Na+/H+ exchange.


Journal of Clinical Investigation | 1995

Characterization of a protein cofactor that mediates protein kinase A regulation of the renal brush border membrane Na(+)-H+ exchanger.

Edward J. Weinman; Deborah Steplock; Yiping Wang; Shirish Shenolikar

Activation of cAMP-dependent protein kinase A inhibits the renal proximal tubule brush border membrane Na(+)-H+ exchanger by a process involving participation of a regulatory cofactor (NHE-RF) that is distinct from the transporter itself. Recent studies from this laboratory reported a partial amino acid sequence of this putative cofactor (Weinman, E. J., D. H. Steplock, and S. Shenolikar. 1993. J. Clin. Invest. 92:1781-1786). The present experiments detail the structure of the NHE-RF protein as determined from molecular cloning studies. A codon-biased oligonucleotide probe to a portion of the amino acid sequence of the putative cofactor was used to isolate a 1.9-kb cDNA from a rabbit renal library. The encoded protein is 358 amino acids in length and is rich in proline residues. Search of existing data bases indicates that NHE-RF is a unique protein. Using a reticulocyte lysate, the cDNA translated a product of approximately 44 kD, which was recognized by an affinity-purified polyclonal antibody to NHE-RF. Potential phosphorylation sites for protein kinase A are present. The mRNA for the protein is expressed in kidney, proximal small intestine, and liver. Reverse transcription/PCR studies in the kidney indicate the presence of mRNA for NHE-RF in several distinct nephron segments including the proximal tubule.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Targeted disruption of the mouse NHERF-1 gene promotes internalization of proximal tubule sodium-phosphate cotransporter type IIa and renal phosphate wasting

Shirish Shenolikar; James W. Voltz; Charles Minkoff; James B. Wade; Edward J. Weinman

Na+/H+ exchanger regulatory factor (NHERF)-1 and NHERF-2, two structurally related protein adapters containing tandem PSD-95/Discs large/ZO-1 (PDZ) domains, were identified as essential factors for protein kinase A-mediated inhibition of the sodium-hydrogen exchanger, NHE3. NHERF-1 and NHERF-2 also bound other cellular targets including the sodium-phosphate cotransporter type IIa encoded by the NPT2 gene. Targeted disruption of the mouse NHERF-1 gene eliminated NHERF-1 expression in kidney and other tissues of the mutant mice without altering NHERF-2 levels in these tissues. NHERF-1 (+/−) and (−/−) male mice maintained normal blood electrolytes but showed increased urinary excretion of phosphate when compared with wild-type (+/+) animals. Although the overall levels of renal NHERF-1 targets, NHE3 and Npt2, were unchanged in the mutant mice, immunocytochemistry showed that the Npt2 protein was aberrantly localized at internal sites in the renal proximal tubule cells. The mislocalization of Npt2 paralleled a reduction in the transporter protein in renal brush–border membranes isolated from the mutant mice. In contrast, NHE3 was appropriately localized at the apical surface of proximal tubules in both wild-type and mutant mice. These data suggested that NHERF-1 played a unique role in the apical targeting and/or trafficking of Npt2 in the mammalian kidney, a function not shared by NHERF-2 or other renal PDZ proteins. Phosphate wasting seen in the NHERF-1(−/−) null mice provided a new experimental system for defining the role of PDZ adapters in the hormonal control of ion transport and renal disease.


Journal of Clinical Investigation | 2009

Differential roles of NHERF1, NHERF2, and PDZK1 in regulating CFTR-mediated intestinal anion secretion in mice

Anurag Singh; Brigitte Riederer; Anja Krabbenhöft; Brigitte Rausch; Janina Bonhagen; Ulrich Lehmann; Hugo R. de Jonge; Mark Donowitz; C. Chris Yun; Edward J. Weinman; Olivier Kocher; Boris M. Hogema; Ursula Seidler

The epithelial anion channel CFTR interacts with multiple PDZ domain-containing proteins. Heterologous expression studies have demonstrated that the Na+/H+ exchanger regulatory factors, NHERF1, NHERF2, and PDZK1 (NHERF3), modulate CFTR membrane retention, conductivity, and interactions with other transporters. To study their biological roles in vivo, we investigated CFTR-dependent duodenal HCO3- secretion in mouse models of Nherf1, Nherf2, and Pdzk1 loss of function. We found that Nherf1 ablation strongly reduced basal as well as forskolin-stimulated (FSK-stimulated) HCO3- secretory rates and blocked beta2-adrenergic receptor (beta2-AR) stimulation. Conversely, Nherf2-/- mice displayed augmented FSK-stimulated HCO3- secretion. Furthermore, although lysophosphatidic acid (LPA) inhibited FSK-stimulated HCO3- secretion in WT mice, this effect was lost in Nherf2-/- mice. Pdzk1 ablation reduced basal, but not FSK-stimulated, HCO3- secretion. In addition, laser microdissection and quantitative PCR revealed that the beta2-AR and the type 2 LPA receptor were expressed together with CFTR in duodenal crypts and that colocalization of the beta2-AR and CFTR was reduced in the Nherf1-/- mice. These data suggest that the NHERF proteins differentially modulate duodenal HCO3- secretion: while NHERF1 is an obligatory linker for beta2-AR stimulation of CFTR, NHERF2 confers inhibitory signals by coupling the LPA receptor to CFTR.


Journal of Biological Chemistry | 1998

The Role of NHERF and E3KARP in the cAMP-mediated Inhibition of NHE3

Georg Lamprecht; Edward J. Weinman; C. H. Chris Yun

NHE3 is the apically located Na+/H+ exchanger in the gut and in the renal proximal tubule. Acute inhibition of this transporter by cAMP requires the presence of either of two NHE3-associated proteins, NHERF or E3KARP. It has been suggested that these proteins either directly regulate NHE3 activity after being phosphorylated by protein kinase A (PKA) or that they may serve as adapters that localize PKA near NHE3. We studied the role of NHERF and E3KARP in opossum kidney cells, which endogenously express NHE3, NHERF, and ezrin and display cAMP-dependent inhibition of NHE3. In vivophosphorylation studies showed that NHERF is a phosphoprotein under basal conditions, but does not change its phosphorylation state after 8-bromo-cAMP treatment, and that E3KARP is not phosphorylated at all. Co-immunoprecipitation showed that NHERF and E3KARP bind both NHE3 and ezrin. Using cAMP analogs it was demonstrated that NHE3 activity, measured as sodium-dependent recovery of the intracellular pH after intracellular acidification, is inhibited by PKA type II. Because others have shown that ezrin binds PKA type II and that NHE3 is phosphorylated by PKA we suggest that NHERF and E3KARP are adapters that link NHE3 to ezrin, thereby localizing PKA near NHE3 to allow NHE3 phosphorylation.


Proceedings of the National Academy of Sciences of the United States of America | 2002

PDZ-domain interactions and apical expression of type IIa Na/Pi cotransporters

Nati Hernando; Nadine Déliot; Serge M. Gisler; Eleanor D. Lederer; Edward J. Weinman; Jürg Biber; Heini Murer

Type IIa Na/Pi cotransporters are expressed in renal proximal brush border and are the major determinants of inorganic phosphate (Pi) reabsorption. Their carboxyl-terminal tail contains information for apical expression, and interacts by means of its three terminal amino acids with several PSD95/DglA/ZO-1-like domain (PDZ)-containing proteins. Two of these proteins, NaPi-Cap1 and Na/H exchanger-regulatory factor 1 (NHERF1), colocalize with the cotransporter in the proximal brush border. We used opossum kidney cells to test the hypothesis of a potential role of PDZ-interactions on the apical expression of the cotransporter. We found that opossum kidney cells contain NaPi-Cap1 and NHERF1 mRNAs. For NHERF1, an apical location of the protein could be documented; this location probably reflects interaction with the cytoskeleton by means of the MERM-binding domain. Overexpression of PDZ domains involved in interaction with the cotransporter (PDZ-1/NHERF1 and PDZ-3/NaPi-Cap1) had a dominant–negative effect, disturbing the apical expression of the cotransporter without affecting the actin cytoskeleton or the basolateral expression of Na/K-ATPase. These data suggest an involvement of PDZ-interactions on the apical expression of type IIa cotransporters.


Oncogene | 2001

Expanding the role of NHERF, a PDZ-domain containing protein adapter, to growth regulation.

James W. Voltz; Edward J. Weinman; Shirish Shenolikar

NHERF (Na+/H+ exchanger regulatory factor or NHERF-1) and E3KARP (NHE3 kinase A regulatory protein or NHERF-2) are structurally related protein adapters that are highly expressed in epithelial tissues. NHERF proteins contain two tandem PDZ domains and a C-terminal sequence that binds several members of the ERM (ezrin-radixin-moesin) family of membrane-cytoskeletal adapters. Although identified as a regulator of NHE3, recent evidence points to a broadening role for NHERF in the function, localization and/or turnover of G-protein coupled receptors, platelet-derived growth factor receptor and ion transporters such as CFTR, Na/Pi cotransporter, Na/HCO3 cotransporter and Trp (calcium) channels. NHERF also recruits non-membrane proteins such as the c-Yes/YAP-65 complex, members of the phospholipase Cβ family and the GRK6A protein kinase to apical surface of polarized epithelial cells where they regulate or respond to membrane signals. While two distinct models have been proposed for NHERFs role in signal transduction, the common theme is NHERFs ability to bring together membrane and non-membrane proteins to regulate cell metabolism and growth. NHERF overexpression in human breast cancers and mutations in NHERF targets, such as CFTR and merlin, the product of Neurofibromatosis NF2 tumor suppressor gene, that impair NHERF binding suggest that aberrant NHERF function contributes to human disease.


Journal of The American Society of Nephrology | 2002

The Serum and Glucocorticoid-Inducible Kinase SGK1 and the Na+/H+ Exchange Regulating Factor NHERF2 Synergize to Stimulate the Renal Outer Medullary K+ Channel ROMK1

C. Chris Yun; Monica Palmada; Hamdy M. Embark; Olga Fedorenko; Yuxi Feng; Guido Henke; Iwan Setiawan; Christoph Boehmer; Edward J. Weinman; Sabrina Sandrasagra; Christoph Korbmacher; Philip Cohen; David A. Pearce; Florian Lang

Mineralocorticoids stimulate Na(+) reabsorption and K(+) secretion in principal cells of connecting tubule and collecting duct. The involved ion channels are ENaC and ROMK1, respectively. In Xenopus oocytes, the serum and glucocorticoid-sensitive kinase SGK1 has been shown to increase ENaC activity by enhancing its abundance in the plasma membrane. With the same method, ROMK1 appeared to be insensitive to regulation by SGK1. On the other hand, ROMK1 has been shown to colocalize with NHERF2, a protein mediating targeting and trafficking of transport proteins into the cell membrane. The present study has been performed to test whether NHERF2 is required for regulation of ROMK1 by SGK1. Coexpression of neither NHERF2 nor SGK1 with ROMK1 increases ROMK1 activity. However, coexpression of NHERF2 and SGK1 together with ROMK1 markedly increases K(+) channel activity. The combined effect of SGK1 and NHERF2 does not significantly alter the I/V relation of the channel but increases the abundance of the channel in the membrane and decreases the decay of channel activity after inhibition of vesicle insertion with brefeldin. Coexpression of NHERF2 and SGK1 does not modify cytosolic pH but leads to a slight shift of pK(a) of ROMK1 to more acidic values. In conclusion, NHERF2 and SGK1 interact to enhance ROMK1 activity in large part by enhancing the abundance of channel protein within the cell membrane. This interaction allows the integration of genomic regulation and activation of SGK1 and NHERF2 in the control of ROMK1 activity and renal K(+) excretion.


The Journal of Physiology | 2001

Na+‐H+ exchanger 3 (NHE3) is present in lipid rafts in the rabbit ileal brush border: a role for rafts in trafficking and rapid stimulation of NHE3

Xuhang Li; Thierry Galli; Sharon Leu; James B. Wade; Edward J. Weinman; George P H Leung; Alice Cheong; Daniel Louvard; Mark Donowitz

1 Rabbit ileal Na+‐absorbing cell Na+‐H+ exchanger 3 (NHE3) was shown to exist in three pools in the brush border (BB), including a population in lipid rafts. Approximately 50 % of BB NHE3 was associated with Triton X‐100‐soluble fractions and the other ∼50 % with Triton X‐100‐insoluble fractions; ∼33 % of the detergent‐insoluble NHE3 was present in cholesterol‐enriched lipid microdomains (rafts). 2 The raft pool of NHE3 was involved in the stimulation of BB NHE3 activity with epidermal growth factor (EGF). Both EGF and clonidine treatments were associated with a rapid increase in the total amount of BB NHE3. This EGF‐ and clonidine‐induced increase of BB NHE3 was associated with an increase in the raft pool of NHE3 and to a smaller extent with an increase in the total detergent‐insoluble fraction, but there was no change in the detergent‐soluble pool. In agreement with the rapid increase in the amount of NHE3 in the BB, EGF also caused a rapid stimulation of BB Na+‐H+ exchange activity. 3 Disrupting rafts by removal of cholesterol with methyl‐β‐cyclodextrin (MβCD) or destabilizing the actin cytoskeleton with cytochalasin D decreased the amount of NHE3 in early endosomes isolated by OptiPrep gradient fractionation. Specifically, NHE3 was shown to associate with endosomal vesicles immunoisolated by anti‐EEA1 (early endosomal autoantigen 1) antibody‐coated magnetic beads and the endosome‐associated NHE3 was decreased by cytochalasin D and MβCD treatment. 4 We conclude that: (i) a pool of ileal BB NHE3 exists in lipid rafts; (ii) EGF and clonidine increase the amount of BB NHE3; (iii) lipid rafts and to a lesser extent, the cytoskeleton, but not the detergent‐soluble NHE3 pool, are involved in the EGF‐ and clonidine‐induced acute increase in amount of BB NHE3; (iv) lipid rafts and the actin cytoskeleton play important roles in the basal endocytosis of BB NHE3.


Journal of Clinical Investigation | 1993

CAMP-mediated inhibition of the renal brush border membrane Na+-H+ exchanger requires a dissociable phosphoprotein cofactor.

Edward J. Weinman; Deborah Steplock; Shirish Shenolikar

Prior studies have suggested that protein kinase A (PKA)-mediated inhibition of the rabbit renal brush border membrane (BBM) Na(+)-H+ exchanger involves a regulatory protein that is distinct from the transporter. This putative regulatory protein was purified by column chromatography and SDS-PAGE, and a partial primary amino acid sequence was determined. An affinity-purified polyclonal antibody to a synthetic peptide representing a sequence of the protein recognized a polypeptide of 55 kD in BBM but not in basolateral membrane. The antibody immunoprecipitated a PKA substrate of a similar molecular mass from detergent-solubilized BBM proteins. When assayed after reconstitution, PKA in the presence of ATP and Mg2+ did not inhibit Na(+)-H+ exchange transport in a fraction of solubilized BBM proteins eluting from an anion exchange column between 0.2 and 0.4 M NaCl (fraction B). Coreconstitution of fraction B with the immunoprecipitated 55-kD protein restored the inhibitory effect of PKA (change = 42%, P < 0.05). By contrast, Na(+)-H+ exchange transport in total solubilized BBM proteins was inhibited 25% (P < 0.05) by PKA, ATP, and Mg2+. This effect was abolished by immunodepletion of the cAMP regulatory protein (change = +5%, P = NS). These findings provide evidence that the regulation of renal BBM Na(+)-H+ exchange transport by PKA is affected by repletion and depletion of a specific protein. This suggests that PKA-mediated inhibition of the renal BBM Na(+)-H+ exchanger requires participation of a regulatory protein that is distinct from the transporter itself.

Collaboration


Dive into the Edward J. Weinman's collaboration.

Top Co-Authors

Avatar

Shirish Shenolikar

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark Donowitz

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pedro A. Jose

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Andrew M. Kahn

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge