Network


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

Hotspot


Dive into the research topics where David R. Stevens is active.

Publication


Featured researches published by David R. Stevens.


Nature | 2001

Hyperpolarization-activated channels HCN1 and HCN4 mediate responses to sour stimuli

David R. Stevens; Reinhard Seifert; Bernd Bufe; Frank Müller; Elisabeth Kremmer; Renate Gauss; Wolfgang Meyerhof; U. Benjamin Kaupp; Bernd Lindemann

Sour taste is initiated by protons acting at receptor proteins or channels. In vertebrates, transduction of this taste quality involves several parallel pathways. Here we examine the effects of sour stimuli on taste cells in slices of vallate papilla from rat. From a subset of cells, we identified a hyperpolarization-activated current that was enhanced by sour stimulation at the taste pore. This current resembled Ih found in neurons and cardio-myocytes, a current carried by members of the family of hyperpolarization-activated and cyclic-nucleotide-gated (HCN) channels. We show by in situ hybridization and immunohistochemistry that HCN1 and HCN4 are expressed in a subset of taste cells. By contrast, gustducin, the G-protein involved in bitter and sweet taste, is not expressed in these cells. Lowering extracellular pH causes a dose-dependent flattening of the activation curve of HCN channels and a shift in the voltage of half-maximal activation to more positive voltages. Our results indicate that HCN channels are gated by extracellular protons and may act as receptors for sour taste.


Current Biology | 2005

Identification of the Minimal Protein Domain Required for Priming Activity of Munc13-1

David R. Stevens; Zheng-Xing Wu; Ulf Matti; Harald J. Junge; Claudia Schirra; Ute Becherer; Sonja M. Wojcik; Nils Brose; Jens Rettig

Most nerve cells communicate with each other through synaptic transmission at chemical synapses. The regulated exocytosis of neurotransmitters, hormones, and peptides occurs at specialized membrane areas through Ca2+-triggered fusion of secretory vesicles with the plasma membrane . Prior to fusion, vesicles are docked at the plasma membrane and must then be rendered fusion-competent through a process called priming. The molecular mechanism underlying this priming process is most likely the formation of the SNARE complex consisting of Syntaxin 1, SNAP-25, and Synaptobrevin 2. Members of the Munc13 protein family consisting of Munc13-1, -2, -3, and -4 were found to be absolutely required for this priming process . In the present study, we identified the minimal Munc13-1 domain that is responsible for its priming activity. Using Munc13-1 deletion constructs in an electrophysiological gain-of-function assay of chromaffin-granule secretion, we show that priming activity is mediated by the C-terminal residues 1100-1735 of Munc13-1, which contains both Munc13-homology domains and the C-terminal C2 domain. Priming by Munc13-1 appears to require its interaction with Syntaxin 1 because point mutants that do not bind Syntaxin 1 do not prime chromaffin granules.


The Journal of Neuroscience | 2005

The role of snapin in neurosecretion: Snapin knock-out mice exhibit impaired calcium-dependent exocytosis of large dense-core vesicles in chromaffin cells

Jin Hua Tian; Zaeng Xing Wu; Michael Unzicker; Li Lu; Qian Cai; Cuiling Li; Claudia Schirra; Ulf Matti; David R. Stevens; Chuxia Deng; Jens Rettig; Zu-Hang Sheng

Identification of the molecules that regulate the priming of synaptic vesicles for fusion and the structural coupling of the calcium sensor with the soluble N-ethyl maleimide sensitive factor adaptor protein receptor (SNARE)-based fusion machinery is critical for understanding the mechanisms underlying calcium-dependent neurosecretion. Snapin binds to synaptosomal-associated protein 25 kDa (SNAP-25) and enhances the association of the SNARE complex with synaptotagmin. In the present study, we abolished snapin expression in mice and functionally evaluated the role of Snapin in neuroexocytosis. We found that the association of synaptotagmin-1 with SNAP-25 in brain homogenates of snapin mutant mice is impaired. Consequently, the absence of Snapin in embryonic chromaffin cells leads to a significant reduction of calcium-dependent exocytosis resulting from a decreased number of vesicles in releasable pools. Overexpression of Snapin fully rescued this inhibitory effect in the mutant cells. Furthermore, Snapin is relatively enriched in the purified large dense-core vesicles of chromaffin cells and associated with synaptotagmin-1. Thus, our biochemical and electrophysiological studies using snapin knock-out mice demonstrate that Snapin plays a critical role in modulating neurosecretion by stabilizing the release-ready vesicles.


The Journal of Neuroscience | 2008

CAPS Facilitates Filling of the Rapidly Releasable Pool of Large Dense-Core Vesicles

Yuanyuan Liu; Claudia Schirra; David R. Stevens; Ulf Matti; Dina Speidel; Detlef Hof; Dieter Bruns; Nils Brose; Jens Rettig

Calcium-activator protein for secretion (CAPS) is a cytosolic protein that associates with large dense-core vesicles and is involved in their secretion. Mammals express two CAPS isoforms, which share a similar domain structure including a Munc13 homology domain that is believed to be involved in the priming of secretory vesicles. A variety of studies designed to perturb CAPS function indicate that CAPS is involved in the secretion of large dense-core vesicles, but where in the secretory pathway CAPS acts is still under debate. Mice in which one allele of the CAPS-1 gene is deleted exhibit a deficit in catecholamine secretion from chromaffin cells. We have examined catecholamine secretion from chromaffin cells in which both CAPS genes were deleted and show that the deletion of both CAPS isoforms causes a strong reduction in the pool of rapidly releasable chromaffin granules and of sustained release during ongoing stimulation. We conclude that CAPS is required for the adequate refilling and/or maintenance of a rapidly releasable granule pool.


The Journal of Neuroscience | 2004

Effects of PKA-Mediated Phosphorylation of Snapin on Synaptic Transmission in Cultured Hippocampal Neurons

Pratima Thakur; David R. Stevens; Zu-Hang Sheng; Jens Rettig

Use-dependent activation of protein kinase A (PKA) modulates transmitter release, contributing to synaptic plasticity. Snapin, a PKA substrate in neurons, associates with the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex, and its phosphorylation leads to increased binding of synaptotagmin to the SNARE complex. We investigated the role of PKA-dependent phosphorylation of Snapin in hippocampal neurons. Overexpression of Snapin S50D, a mutant mimicking the phosphorylated state, resulted in a decreased number of readily releasable vesicles. In addition, both the release probability of individual vesicles and the depression rate during high-frequency stimulation were increased. Overexpression of Snapin S50A, a mutant that cannot be phosphorylated, did not alter the size of the pool or the probability of release. Furthermore, dialysis of Sp-cAMPS, a nonhydrolyzable analog of cAMP that will promote phosphorylation by PKA, also led to increased synaptic depression in cells overexpressing wild-type Snapin. These results establish Snapin as an important target of PKA in CNS synapses and indicate a role for Snapin in the plasticity of transmitter release.


Journal of Cell Biology | 2010

Two distinct secretory vesicle-priming steps in adrenal chromaffin cells

Yuanyuan Liu; Claudia Schirra; Ludwig Edelmann; Ulf Matti; Jeong-Seop Rhee; Detlef Hof; Dieter Bruns; Nils Brose; Heiko Rieger; David R. Stevens; Jens Rettig

The calcium-dependent activator proteins for secretion, CAPS1 and CAPS2, facilitate syntaxin opening during synaptic vesicle priming.


Frontiers in Synaptic Neuroscience | 2011

Vesicle Pools: Lessons from Adrenal Chromaffin Cells

David R. Stevens; Claudia Schirra; Ute Becherer; Jens Rettig

The adrenal chromaffin cell serves as a model system to study fast Ca2+-dependent exocytosis. Membrane capacitance measurements in combination with Ca2+ uncaging offers a temporal resolution in the millisecond range and reveals that catecholamine release occurs in three distinct phases. Release of a readily releasable (RRP) and a slowly releasable (SRP) pool are followed by sustained release, due to maturation, and release of vesicles which were not release-ready at the start of the stimulus. Trains of depolarizations, a more physiological stimulus, induce release from a small immediately releasable pool of vesicles residing adjacent to calcium channels, as well as from the RRP. The SRP is poorly activated by depolarization. A sequential model, in which non-releasable docked vesicles are primed to a slowly releasable state, and then further mature to the readily releasable state, has been proposed. The docked state, dependent on membrane proximity, requires SNAP-25, synaptotagmin, and syntaxin. The ablation or modification of SNAP-25 and syntaxin, components of the SNARE complex, as well as of synaptotagmin, the calcium sensor, and modulators such complexins and Snapin alter the properties and/or magnitudes of different phases of release, and in particular can ablate the RRP. These results indicate that the composition of the SNARE complex and its interaction with modulatory molecules drives priming and provides a molecular basis for different pools of releasable vesicles.


Journal of the American Chemical Society | 2012

New photolabile BAPTA-based Ca2+ cages with improved photorelease.

Jiaxi Cui; Radu A. Gropeanu; David R. Stevens; Jens Rettig; Aránzazu del Campo

The efficient synthesis, physicochemical and photolytical properties of a photoactivable BAPTA-based Ca(2+) cage containing two photosensitive o-nitrobenzhydryl groups attached to the aromatic core are described. Ca(2+) release in living cells was evaluated. The double substitution with the chromophores caused a significant improvement of the Ca(2+) release properties of nitr-T versus singly substituted reported nitr-x derivatives without compromising Ca(2+)/Mg(2+) selectivity or pH insensitivity. Our results demonstrate a general strategy to improve light-triggered Ca(2+) release which may result in more efficient, selective, and pH-insensitive photolabile Ca(2+) chelators.


eLife | 2015

Identification of a Munc13-sensitive step in chromaffin cell large dense-core vesicle exocytosis

Kwun-nok Mimi Man; Cordelia Imig; Alexander M. Walter; Paulo S. Pinheiro; David R. Stevens; Jens Rettig; Jakob B. Sørensen; Benjamin H. Cooper; Nils Brose; Sonja M. Wojcik

It is currently unknown whether the molecular steps of large dense-core vesicle (LDCV) docking and priming are identical to the corresponding reactions in synaptic vesicle (SV) exocytosis. Munc13s are essential for SV docking and priming, and we systematically analyzed their role in LDCV exocytosis using chromaffin cells lacking individual isoforms. We show that particularly Munc13-2 plays a fundamental role in LDCV exocytosis, but in contrast to synapses lacking Munc13s, the corresponding chromaffin cells do not exhibit a vesicle docking defect. We further demonstrate that ubMunc13-2 and Munc13-1 confer Ca2+-dependent LDCV priming with similar affinities, but distinct kinetics. Using a mathematical model, we identify an early LDCV priming step that is strongly dependent upon Munc13s. Our data demonstrate that the molecular steps of SV and LDCV priming are very similar while SV and LDCV docking mechanisms are distinct. DOI: http://dx.doi.org/10.7554/eLife.10635.001


Cell Calcium | 2012

Regulated exocytosis in chromaffin cells and cytotoxic T lymphocytes: How similar are they?

Ute Becherer; Moritz R. Medart; Claudia Schirra; Elmar Krause; David R. Stevens; Jens Rettig

Chromaffin cells from the adrenal medulla secrete catecholamines into the blood stream as part of the fight-or-flight response. Cytotoxic T lymphocytes from the immune system release cytotoxic substances to kill antigen-presenting cells. While at first glance these two cell types do not seem to have much in common, evidence from human diseases indicates that the molecular mechanisms of exocytosis of the respective granules share many similar features. In this review we highlight the similarities and differences of individual aspects of granule maturation and release in both cell types. In addition, we discuss established and putative molecules involved in distinct steps and suggest technical approaches which might facilitate future studies in chromaffin cells and cytotoxic T lymphocytes.

Collaboration


Dive into the David R. Stevens's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge