Network


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

Hotspot


Dive into the research topics where Emanuel E. Strehler is active.

Publication


Featured researches published by Emanuel E. Strehler.


Current Molecular Medicine | 2004

Calcium Pumps of Plasma Membrane and Cell Interior

Emanuel E. Strehler; Marek Treiman

Calcium entering the cell from the outside or from intracellular organelles eventually must be returned to the extracellular milieu or to intracellular storage organelles. The two major systems capable of pumping Ca2+ against its large concentration gradient out of the cell or into the sarco/endoplasmatic reticulum are the plasma membrane Ca2+ ATPases (PMCAs) and the sarco/endoplasmic reticulum Ca2+ ATPases (SERCAs), respectively. In mammals, multigene families code for these Ca2+ pumps and additional isoform subtypes are generated via alternative splicing. PMCA and SERCA isoforms show developmental-, tissue- and cell type-specific patterns of expression. Different PMCA and SERCA isoforms are characterized by different regulatory and kinetic properties that likely are optimized for the distinct functional tasks fulfilled by each pump in setting resting cytosolic or intra-organellar Ca2+ levels, and in shaping intracellular Ca2+ signals with spatial and temporal resolution. The loss or malfunction of specific Ca2+ pump isoforms is associated with defects such as deafness, ataxia or heart failure. Understanding the involvement of different Ca2+ pump isoforms in the pathogenesis of disease allows their identification as therapeutic targets for the development of selective strategies to prevent or combat the progression of these disorders.


The Journal of Membrane Biology | 1991

RECENT ADVANCES IN THE MOLECULAR CHARACTERIZATION OF PLASMA MEMBRANE CA2+ PUMPS

Emanuel E. Strehler

Since its discovery some 25 years ago (Schatzmann, 1966) the plasma membrane Ca 2+ pump (PMCA) has constantly gained in importance as a model membrane protein to study the structural, functional, regulatory and genetic basis of ATP-driven cation transport. The success in the characterization of this enzyme parallels that made in the molecular analysis of other complex low-abundance proteins spanning the lipid bilayer and reflects to a large extent the impressive technical progress made during the past two decades in all areas of modern biological research. It is not surprising, therefore, that several reviews have already been devoted mostly, if not exclusively, to the various aspects of the plasma membrane Ca 2+ pump. Rather than trying to resummarize the contents of previous reviews, the present contribution will focus on those developments concerning the characterization of the Ca 2 § pump which have only recently (i.e., during the past three to five years) been added to our existing views on this important enzyme. Accordingly, no attempt will be made to present a comprehensive picture of the subject; for more detailed information on different aspects of the plasma membrane Ca 2+ pump the reader is referred to previously published reviews and references found therein. A recent summary of the general properties, and particularly of ion transportrelated characteristics, of the pump can be found in Garrahan and Rega (1990), and a synopsis of the problems related to the mechanism of calcium pumping has been given by Jencks (1989). A limited selection of further reviews published during the past decade includes contributions by Sarkadi [1980 (emphasis on early results on the enzymatic properties


Annals of the New York Academy of Sciences | 2007

Plasma Membrane Ca2+ ATPases as Dynamic Regulators of Cellular Calcium Handling

Emanuel E. Strehler; Ariel J. Caride; Adelaida G. Filoteo; Yuning Xiong; John T. Penniston; Ágnes Enyedi

Abstract:  Plasma membrane Ca2+ ATPases (PMCAs) are essential components of the cellular toolkit to regulate and fine‐tune cytosolic Ca2+ concentrations. Historically, the PMCAs have been assigned a housekeeping role in the maintenance of intracellular Ca2+ homeostasis. More recent work has revealed a perplexing multitude of PMCA isoforms and alternative splice variants, raising questions about their specific role in Ca2+ handling under conditions of varying Ca2+ loads. Studies on the kinetics of individual isoforms, combined with expression and localization studies suggest that PMCAs are optimized to function in Ca2+ regulation according to tissue‐ and cell‐specific demands. Different PMCA isoforms help control slow, tonic Ca2+ signals in some cells and rapid, efficient Ca2+ extrusion in others. Localized Ca2+ handling requires targeting of the pumps to specialized cellular locales, such as the apical membrane of cochlear hair cells or the basolateral membrane of kidney epithelial cells. Recent studies suggest that alternatively spliced regions in the PMCAs are responsible for their unique targeting, membrane localization, and signaling cross‐talk. The regulated deployment and retrieval of PMCAs from specific membranes provide a dynamic system for a cell to respond to changing needs of Ca2+ regulation.


Journal of Biological Chemistry | 2002

Plasma membrane Ca2+ ATPase isoform 2b interacts preferentially with Na+/H+ exchanger regulatory factor 2 in apical plasma membranes

Steven J. DeMarco; Emanuel E. Strehler

Spatial and temporal regulation of Ca2+ signaling require the assembly of multiprotein complexes linking molecules involved in Ca2+ influx, sensing, buffering, and extrusion. Recent evidence indicates that plasma membrane Ca2+ ATPases (PMCAs) participate in the control of local Ca2+ fluxes, but the mechanism of multiprotein complex formation of specific PMCAs is poorly understood. Using the PMCA2b COOH-terminal tail as bait in a yeast two-hybrid screen, we identified the PSD-95, Dlg,ZO-1 (PDZ) domain-containing Na+/H+exchanger regulatory factor-2 (NHERF2) as an interacting partner. Protein pull-down and coimmunoprecipitation experiments using recombinant PMCA2b and PMCA4b as well as NHERF1 and NHERF2 showed that the interaction of PMCA2b with NHERF2 was specific and selective. PMCA4b did not interact with either of the NHERFs, and PMCA2b selectively preferred NHERF2 over NHERF1. Green fluorescent protein-tagged PMCA2b was expressed at the apical membrane in Madin-Darby canine kidney epithelial cells, where it colocalized with apically targeted NHERF2. Our study identifies NHERF2 as the first specific PDZ partner for PMCA2b not shared with PMCA4b, and demonstrates that PMCA splice forms differing only minimally in their COOH-terminal residues interact with unique PDZ proteins. NHERFs have been implicated in the targeting, retention and regulation of membrane proteins including the β2-adrenergic receptor, cystic fibrosis transmembrane conductance regulator, and Trp4 Ca2+channel, and NHERF2 is now shown to also interact with PMCA2b. This interaction may allow the functional assembly of PMCA2b in a multiprotein Ca2+ signaling complex, facilitating integrated cross-talk between local Ca2+ influx and efflux.


Journal of Biological Chemistry | 2003

Alternative splicing of the first intracellular loop of plasma membrane Ca2+ ATPase isoform 2 alters its membrane targeting

Emanuel E. Strehler

Plasma membrane Ca2+-ATPases (PMCAs) are involved in local Ca2+ signaling and in the spatial control of Ca2+ extrusion, but how different PMCA isoforms are targeted to specific membrane domains is unknown. In polarized MDCK epithelial cells, a green fluorescent protein-tagged PMCA4b construct was targeted to the basolateral membrane, whereas a green fluorescent protein-tagged PMCA2b construct was localized to both the apical and basolateral domain. The PDZ protein-binding COOH-terminal tail of PMCA2b was not responsible for its apical membrane localization, as a chimeric pump made of an NH2-terminal portion from PMCA4 and a COOH-terminal tail from PMCA2b was targeted to the basolateral domain. Deletion of the last six residues of the COOH terminus of either PMCA2b or PMCA4b did not alter their membrane targeting, suggesting that PDZ protein interactions are not essential for proper membrane localization of the pumps. Instead, we found that alternative splicing affecting the first cytosolic loop determined apical membrane targeting of PMCA2. Only the “w” form, which contains a 45-amino acid residue insertion, showed prominent apical membrane localization. By contrast, the x and z splice variants containing insertions of 14 and 0 residues, respectively, localized to the basolateral membrane. The w splice insert was the crucial determinant of apical PMCA2 localization, and this was independent of the splice configuration at the COOH-terminal end of the pump; both PMCA2w/b and PMCA2w/a showed prominent apical targeting, whereas PMCA2x/b, PMCA2z/b, and PMCA2z/a were confined to the basolateral membrane. These data report the first differential effect of alternative splicing within the first cytosolic loop of PMCA2 and help explain the selective enrichment of specific PMCA2 isoforms in specialized membrane compartments such as stereocilia of auditory hair cells.


Biochemical Society Transactions | 2007

Plasma-membrane Ca2+ pumps: Structural diversity as the basis for functional versatility

Emanuel E. Strehler; Adelaida G. Filoteo; John T. Penniston; Ariel J. Caride

Plasma-membrane calcium pumps [PMCAs (plasma-membrane Ca(2+)-ATPases)] expel Ca(2+) from eukaryotic cells to maintain overall Ca(2+) homoeostasis and to provide local control of intracellular Ca(2+) signalling. Recent work indicates functional versatility among PMCA isoforms, with specific pumps being essential for cochlear hair cell function, sperm motility, feedback signalling in the heart and pre- and post-synaptic Ca(2+) regulation in neurons. The functional versatility of PMCAs is due to differences in their regulation by CaM (calmodulin), kinases and other signalling proteins, as well as to their differential targeting and retention in defined plasma membrane domains. The basis for this is the structural diversity of PMCAs. In mammals, four genes encode PMCA isoforms 1-4, and each of these has multiple variants generated by alternative RNA splicing. The alternatively spliced regions are intimately involved in the regulatory interactions and differential membrane localization of the pumps. The alternatively spliced C-terminal tail acts as an autoinhibitory domain by interacting with the catalytic core of the pump. The degree of inhibition and the kinetics of interaction with the major activator CaM differ between PMCA variants. This translates into functional differences in how PMCAs handle Ca(2+) signals of different magnitude and frequency. Accumulating evidence thus demonstrates how structural diversity provides functional versatility in the PMCAs.


Neuron | 2002

Bradykinin and ATP Accelerate Ca2+ Efflux from Rat Sensory Neurons via Protein Kinase C and the Plasma Membrane Ca2+ Pump Isoform 4

Yuriy M. Usachev; Steven J. DeMarco; Colin Campbell; Emanuel E. Strehler; Stanley A. Thayer

Modulation of Ca(2+) channels by neurotransmitters provides critical control of neuronal excitability and synaptic strength. Little is known about regulation of the Ca(2+) efflux pathways that counterbalance Ca(2+) influx in neurons. We demonstrate that bradykinin and ATP significantly facilitate removal of action potential-induced Ca(2+) loads by stimulating plasma membrane Ca(2+)-ATPases (PMCAs) in rat sensory neurons. This effect was mimicked in the soma and axonal varicosities by phorbol esters and was blocked by antagonists of protein kinase C (PKC). Reduced expression of PMCA isoform 4 abolished, and overexpression of isoform 4b enhanced, PKC-dependent facilitation of Ca(2+) efflux. This acceleration of PMCA4 underlies the shortening of the action potential afterhyperpolarization produced by activation of bradykinin and purinergic receptors. Thus, isoform-specific modulation of PMCA-mediated Ca(2+) efflux represents a novel mechanism to control excitability in sensory neurons.


Journal of Neurochemistry | 2006

Glutamate-induced protease-mediated loss of plasma membrane Ca2+ pump activity in rat hippocampal neurons

William J. Pottorf; Tanner M. Johanns; Stephen M. Derrington; Emanuel E. Strehler; Ágnes Enyedi; Stanley A. Thayer

Ca2+ dysregulation is a hallmark of excitotoxicity, a process that underlies multiple neurodegenerative disorders. The plasma membrane Ca2+ ATPase (PMCA) plays a major role in clearing Ca2+ from the neuronal cytoplasm. Here, we show that the rate of PMCA‐mediated Ca2+ efflux from rat hippocampal neurons decreased following treatment with an excitotoxic concentration of glutamate. PMCA‐mediated Ca2+ extrusion following a brief train of action potentials exhibited an exponential decay with a mean time constant (τ) of 8.8 ± 0.2 s. Four hours following the start of a 30 min treatment with 200 µm glutamate, a second population of cells emerged with slowed recovery kinetics (τ = 16.5 ± 0.3 s). Confocal imaging of cells expressing an enhanced green fluorescent protein (EGFP)‐PMCA4b fusion protein revealed that glutamate treatment internalized EGFP and that cells with reduced plasma membrane fluorescence had impaired Ca2+ clearance. Treatment with inhibitors of the Ca2+‐activated protease calpain protected PMCA function and prevented EGFP‐PMCA internalization. PMCA internalization was triggered by activation of NMDA receptors and was less pronounced for a non‐toxic concentration of glutamate relative to one that produces excitotoxicity. PMCA isoform 2 also internalized following exposure to glutamate, although the Na+/K+ ATPase did not. These data suggest that glutamate exposure initiated protease‐mediated internalization of PMCAs with a corresponding loss of function that may contribute to the Ca2+ dysregulation that accompanies excitotoxicity.


The Journal of Comparative Neurology | 2002

Cell-specific expression of plasma membrane calcium ATPase isoforms in retinal neurons.

Steven J. Demarco; Juliette Johnson; Emanuel E. Strehler; David R. Copenhagen

Ca2+ extrusion by high‐affinity plasma membrane calcium ATPases (PMCAs) is a principal mechanism for the clearance of Ca2+ from the cytosol. The PMCA family consists of four isoforms (PMCA1–4). Little is known about the selective expression of these isoforms in brain tissues or about the physiological function conferred upon neurons by any given isoform. We investigated the cellular and subcellular distribution of PMCA isoforms in a mammalian retina. Mouse photoreceptors, cone bipolar cells and horizontal cells, which respond to light with a graded polarization, express isoform 1 (PMCA1) of the PMCA family. PMCA2 is localized to rod bipolar cells, horizontal cells, amacrine cells, and ganglion cells, and PMCA3 is predominantly expressed in spiking neurons, including both amacrine and ganglion cells but is also found in horizontal cells. PMCA4 was found to be selectively expressed in both synaptic layers. Optical measurements of Ca2+ clearance showed that PMCAs mediate Ca2+ extrusion in both rod and cone bipolar cells. In addition, we found that rod bipolar cells, but not cone bipolar cells possess a prominent Na+/Ca2+ exchange mechanism. We conclude that PMCA isoforms are selectively expressed in retinal neurons and that processes of Ca2+ clearance are different in rod and cone bipolar cells. J. Comp. Neurol. 451:1–21, 2002.


The Journal of Comparative Neurology | 2003

Isoform-specific distribution of the plasma membrane Ca2+ ATPase in the rat brain

Alain Burette; Julia M. Rockwood; Emanuel E. Strehler; Richard J. Weinberg

Regulation of cytoplasmic calcium is crucial both for proper neuronal function and cell survival. The concentration of Ca2+ in cytoplasm of a neuron at rest is 10,000 times lower than in the extracellular space, pointing to the importance of the transporters that extrude intracellular Ca2+. The family of plasma membrane calcium‐dependent ATPases (PMCAs) represent a major component of the Ca2+ regulatory system. However, little information is available on the regional and cellular distribution of these calcium pumps. We used immunohistochemistry to investigate the distribution of each of the four PMCA isoforms (PMCA1–4) in the rat brain. Each isoform exhibited a remarkably precise and distinct pattern of distribution. In many cases, PMCA isoforms in a single brain structure were differentially expressed within different classes of neurons, and within different subcellular compartments. These data show that each isoform is independently organized and suggest that PMCAs may play a more complex role in calcium homeostasis than generally recognized. J. Comp. Neurol. 467:464–476, 2003.

Collaboration


Dive into the Emanuel E. Strehler's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ágnes Enyedi

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Hans M. Eppenberger

École Polytechnique Fédérale de Lausanne

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alain Burette

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge