Network


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

Hotspot


Dive into the research topics where Ulrike Janssen-Bienhold is active.

Publication


Featured researches published by Ulrike Janssen-Bienhold.


European Journal of Neuroscience | 2004

Functional expression of connexin57 in horizontal cells of the mouse retina

Sonja Hombach; Ulrike Janssen-Bienhold; Goran Söhl; Timm Schubert; Heinrich Büssow; Thomas Ott; Reto Weiler; Klaus Willecke

Horizontal cells are interneurons of the vertebrate retina that exhibit strong electrical and tracer coupling but the identity of the channel‐forming connexins has remained elusive. Here we show that horizontal cells of the mouse retina express connexin57 (Cx57). We have generated Cx57‐deficient mice by replacing the Cx57 coding region with a lacZ reporter gene, expressed under control of the endogenous Cx57 promoter. These mice were fertile and showed no obvious anatomical or behavioural abnormalities. Cx57 mRNA was expressed in the retina of wild‐type littermates but was absent from the retina of Cx57‐deficient mice. Previously reported results that the Cx57 gene was very weakly expressed in several other mouse tissues turned out to be unspecific. Cx57 mRNA is abundantly expressed in the retina and weakly in the thymus of adult mice but absent in all other adult tissues tested, including brain. Furthermore, Cx57 is expressed in embryonic kidney at E16.5 to E18.5 days post‐conception, as indicated by the pattern of lacZ expression. Within the retina, lacZ signals were assigned exclusively to horizontal cells based on co‐localization with cell‐type‐specific marker proteins. Microinjection of Neurobiotin into horizontal cells of isolated retinae revealed less than 1% of tracer coupling in Cx57‐deficient retinae compared with wild‐type controls. Cx57 is the first connexin identified in mammalian horizontal cells and the first connexin whose expression is apparently restricted to only one type of neuron.


The Journal of Neuroscience | 2005

Deletion of Connexin45 in Mouse Retinal Neurons Disrupts the Rod/Cone Signaling Pathway between AII Amacrine and ON Cone Bipolar Cells and Leads to Impaired Visual Transmission

Stephan Maxeiner; Karin Dedek; Ulrike Janssen-Bienhold; Josef Ammermüller; Hendrik Brune; Taryn Kirsch; Mario Pieper; Joachim Degen; Olaf Krüger; Klaus Willecke; Reto Weiler

Connexin45 (Cx45) is known to be expressed in the retina, but its functional analysis was problematic because general deletion of Cx45 coding DNA resulted in cardiovascular defects and embryonic lethality at embryonic day 10.5. We generated mice with neuron-directed deletion of Cx45 and concomitant activation of the enhanced green fluorescent protein (EGFP). EGFP labeling was observed in bipolar, amacrine, and ganglion cell populations. Intracellular microinjection of fluorescent dyes in EGFP-labeled somata combined with immunohistological markers revealed Cx45 expression in both ON and OFF cone bipolar cells. The scotopic electroretinogram of mutant mice revealed a normal a-wave but a 40% reduction in the b-wave amplitude, similar to that found in Cx36-deficient animals, suggesting a possible defect in the rod pathway of visual transmission. Indeed, neurotransmitter coupling between AII amacrine cells and Cx45-expressing cone bipolar cells was disrupted in Cx45-deficient mice. These data suggest that both Cx45 and Cx36 participate in the formation of functional heterotypic electrical synapses between these two types of retinal neurons that make up the major rod pathway.


The Journal of Neuroscience | 2004

Expression of connexin36 in cone pedicles and OFF-cone bipolar cells of the mouse retina

Andreas Feigenspan; Ulrike Janssen-Bienhold; Sheriar G. Hormuzdi; Hannah Monyer; Joachim Degen; Goran Söhl; Klaus Willecke; Josef Ammermüller; Reto Weiler

Transgenic technology, immunocytochemistry, electrophysiology, intracellular injection techniques, and reverse transcription PCR were combined to study the expression of neuronal connexin36 (Cx36) in the outer plexiform layer of the mouse retina. Transgenic animals expressed either a fusion protein of full-length Cx36 with enhanced green fluorescent protein (EGFP) attached at the C terminus or exon 2 of Cx36 was replaced byβ-galactosidase (β-gal). In the outer nuclear layer,β-gal-positive cell bodies, which were confined to the most distal region close to the outer limiting membrane, displayed immunoreactivity against S-cone opsin. Cx36–EGFP puncta colocalized with cone pedicles, which were visualized by intracellular injection. In reverse transcriptase PCR experiments, Cx36 mRNA was never detected in samples of rods harvested from the outer nuclear layer. These results strongly suggest expression of Cx36 in cones but not in rods. In vertical sections, Cx36 expression in the vitreal part of the outer plexiform layer was characterized by a patchy distribution. Immunocytochemistry with antibodies against the neurokinin-3 receptor and the potassium channel HCN4 (hyperpolarization-activated cyclic nucleotide-gated potassium channel) displayed clusters of the Cx36 label on the dendrites of OFF-cone bipolar cells. In horizontal sections, these clusters of Cx36 appeared as round or oval-shaped groups of individual puncta, and they were always aligned with the base of cone pedicles. Double-labeling experiments and single-cell reverse transcriptase PCR ruled out expression of Cx36 in horizontal cells and rod bipolar cells. At light microscopic resolution, we found close association of Cx36–EGFP with the AMPA-type glutamate receptor subunit GluR1 but not with GluR2–GluR4, the kainate receptor subunit GluR5, or the metabotropic glutamate receptor mGluR6.


Journal of Cell Science | 2006

Ablation of connexin43 in uterine smooth muscle cells of the mouse causes delayed parturition

Britta Döring; Oksana Shynlova; Prudence Tsui; Dominik Eckardt; Ulrike Janssen-Bienhold; Franz Hofmann; Susanne Feil; Robert Feil; Stephen J. Lye; Klaus Willecke

Gap junctions are characteristically increased in the myometrium during term and preterm delivery and are thought to be essential for the development of uterine contractions during labour. Expression of connexin43 (Cx43), the major myometrial gap junction protein, is increased during delivery. We have generated a mouse mutant (Cx43fl/fl:SM-CreERT2), in which the coding region of Cx43 can be specifically deleted in smooth muscle cells at any given time point by application of tamoxifen. By this approach, we were able to study long-term effects on myometrial functions that are necessary for parturition as well as gap junction intercellular communication in primary myometrial cell cultures. We found a prolongation of the pregnancy in 82% of tamoxifen-treated Cx43fl/fl:SM-CreERT2 mice as well as decreased dye coupling in cultured primary myocytes of these animals. Other parturition-specific parameters such as the regulation of oxytocin receptor, prostaglandin F receptor or progesterone remained unchanged. Our results indicate the important function of Cx43 during parturition in the living animal and suggest further strategies to investigate the role of connexins in uterine contractility in transgenic mice.


The Journal of Comparative Neurology | 1998

Distribution of connexin43 immunoreactivity in the retinas of different vertebrates.

Ulrike Janssen-Bienhold; Rolf Dermietzel; Reto Weiler

The distribution of Connexin43 (Cx43) was examined by immunoblotting and immunofluorescence microscopy in the retinas of five different vertebrates by using a C‐terminal specific peptide antibody. The specificity of the antibody was proved on immunoblots, in which it showed cross reactivity with a 43‐kDa protein in rat heart homogenates as well as in homogenates of rabbit, rat, chicken, turtle, and fish (carp and zebrafish) retinas.


PLOS Biology | 2011

Synaptic Transmission from Horizontal Cells to Cones Is Impaired by Loss of Connexin Hemichannels

Lauw J. Klaassen; Ziyi Sun; Marvin N. Steijaert; Petra Bolte; Iris Fahrenfort; Trijntje Sjoerdsma; Jan Klooster; Yvonne Claassen; Colleen R. Shields; Huub M. M. ten Eikelder; Ulrike Janssen-Bienhold; Georg Zoidl; Douglas G. McMahon; Maarten Kamermans

In the vertebrate retina, horizontal cells generate the inhibitory surround of bipolar cells, an essential step in contrast enhancement. For the last decades, the mechanism involved in this inhibitory synaptic pathway has been a major controversy in retinal research. One hypothesis suggests that connexin hemichannels mediate this negative feedback signal; another suggests that feedback is mediated by protons. Mutant zebrafish were generated that lack connexin 55.5 hemichannels in horizontal cells. Whole cell voltage clamp recordings were made from isolated horizontal cells and cones in flat mount retinas. Light-induced feedback from horizontal cells to cones was reduced in mutants. A reduction of feedback was also found when horizontal cells were pharmacologically hyperpolarized but was absent when they were pharmacologically depolarized. Hemichannel currents in isolated horizontal cells showed a similar behavior. The hyperpolarization-induced hemichannel current was strongly reduced in the mutants while the depolarization-induced hemichannel current was not. Intracellular recordings were made from horizontal cells. Consistent with impaired feedback in the mutant, spectral opponent responses in horizontal cells were diminished in these animals. A behavioral assay revealed a lower contrast-sensitivity, illustrating the role of the horizontal cell to cone feedback pathway in contrast enhancement. Model simulations showed that the observed modifications of feedback can be accounted for by an ephaptic mechanism. A model for feedback, in which the number of connexin hemichannels is reduced to about 40%, fully predicts the specific asymmetric modification of feedback. To our knowledge, this is the first successful genetic interference in the feedback pathway from horizontal cells to cones. It provides direct evidence for an unconventional role of connexin hemichannels in the inhibitory synapse between horizontal cells and cones. This is an important step in resolving a long-standing debate about the unusual form of (ephaptic) synaptic transmission between horizontal cells and cones in the vertebrate retina.


European Journal of Neuroscience | 2006

Localization of heterotypic gap junctions composed of connexin45 and connexin36 in the rod pathway of the mouse retina

Karin Dedek; Konrad Schultz; Mario Pieper; Petra Dirks; Stephan Maxeiner; Klaus Willecke; Reto Weiler; Ulrike Janssen-Bienhold

The primary rod pathway in mammals contains gap junctions between AII amacrine cells and ON cone bipolar cells which relay the rod signal into the cone pathway under scotopic conditions. Two gap junctional proteins, connexin36 (Cx36) and connexin45 (Cx45), appear to play a pivotal role in this pathway because lack of either protein leads to an impairment of visual transmission under scotopic conditions. To investigate whether these connexins form heterotypic gap junctions between ON cone bipolar and AII amacrine cells, we used newly developed Cx45 antibodies and studied the cellular and subcellular distribution of this protein in the mouse retina. Specificity of the Cx45 antibodies was determined, among others, by Western blot and immunostaining of mouse heart, where Cx45 is abundantly expressed. In mouse retina, Cx45 immunosignals were detected in both plexiform layers and the ganglion cell layer. Double staining for Cx45 and Cx36 revealed a partial overlap in the punctate patterns in the ON sublamina of the inner plexiform layer of the retina. We quantified the distributions of these two connexins in the ON sublamina, and detected 30% of the Cx45 signals to be co‐localized with or in close apposition to Cx36 signals. Combining immunostaining and intracellular dye injection revealed an overlap or tight association of Cx36 and Cx45 signals on the terminals of injected AII amacrine and two types of ON cone bipolar cells. Our results provide direct evidence for heterotypic gap junctions composed of Cx36 and Cx45 between AII amacrine and certain types of ON cone bipolar cells.


Experimental Cell Research | 2008

A new conditional mouse mutant reveals specific expression and functions of connexin36 in neurons and pancreatic beta-cells

Kerstin Wellershaus; Joachim Degen; Jim Deuchars; Martin Theis; Anne Charollais; Dorothée Caille; Benoit R. Gauthier; Ulrike Janssen-Bienhold; Stephan Sonntag; Pedro Luis Herrera; Paolo Meda; Klaus Willecke

Connexin36 (Cx36) is the main connexin isoform expressed in neurons of the central nervous system (CNS) and in pancreatic beta-cells, i.e. two types of excitable cells that share - in spite of their different origins - a number of common features. Previous studies on Cx36 deficient mice have documented that loss of Cx36 resulted in phenotypic abnormalities in both the CNS and the pancreas which, however, could not be attributed to specific cell types due to the general deletion nature of the animal model used. Attempts to address this limitation using cell type specific deletions generated by the Cre/loxP strategy have so far been complicated by the lack of Cx36 expression from the floxed allele. We have now generated a conditional Cx36 deficient mouse mutant in which the coding region of Cx36 is flanked by loxP sites, followed by a cyan fluorescent protein (CFP) reporter gene. Here we show that Cx36 was still expressed from the floxed allele in neurons and pancreatic beta-cells. In these cells, a 30-60% decrease of this protein, relative to the expression level of the wildtype allele, did not significantly perturb cell coupling. The deletion of Cx36 by ubiquitously and cell type specifically expressed Cre recombinases revealed that CFP functions as a reliable reporter for Cx36 expression in brain neurons and to some extent in retina neurons, but not in pancreas. Loss of Cx36 by Cre-mediated recombination was documented at transcript and protein levels. Cell type specific deletion of Cx36 in the endocrine pancreas revealed major alterations in the basal as well as the glucose-induced insulin secretion, hence specifically attributing to pancreatic Cx36 an important regulatory role in the control of beta-cell function. Cell type specific deletion of Cx36 in the CNS by suitable Cre recombinases should also help to elucidate the functional role of Cx36 in different neuronal subtypes.


PLOS ONE | 2014

Identification of a common non-apoptotic cell death mechanism in hereditary retinal degeneration.

Blanca Arango-Gonzalez; Dragana Trifunović; Ayse Sahaboglu; Katharina Kranz; Stylianos Michalakis; Pietro Farinelli; Susanne Koch; Fred Koch; Sandra Cottet; Ulrike Janssen-Bienhold; Karin Dedek; Martin Biel; Eberhart Zrenner; Thomas Euler; Per Ekström; Marius Ueffing; François Paquet-Durand

Cell death in neurodegenerative diseases is often thought to be governed by apoptosis; however, an increasing body of evidence suggests the involvement of alternative cell death mechanisms in neuronal degeneration. We studied retinal neurodegeneration using 10 different animal models, covering all major groups of hereditary human blindness (rd1, rd2, rd10, Cngb1 KO, Rho KO, S334ter, P23H, Cnga3 KO, cpfl1, Rpe65 KO), by investigating metabolic processes relevant for different forms of cell death. We show that apoptosis plays only a minor role in the inherited forms of retinal neurodegeneration studied, where instead, a non-apoptotic degenerative mechanism common to all mutants is of major importance. Hallmark features of this pathway are activation of histone deacetylase, poly-ADP-ribose-polymerase, and calpain, as well as accumulation of cyclic guanosine monophosphate and poly-ADP-ribose. Our work thus demonstrates the prevalence of alternative cell death mechanisms in inherited retinal degeneration and provides a rational basis for the design of mutation-independent treatments.


Visual Neuroscience | 2001

Identification and localization of connexin26 within the photoreceptor-horizontal cell synaptic complex.

Ulrike Janssen-Bienhold; Konrad Schultz; Alexandra Gellhaus; Peter Schmidt; Josef Ammermüller; Reto Weiler

Connexin26 (Cx26) is a member of the family of integral membrane proteins that normally form intercellular gap junctional channels. We have used Western blotting, immunofluorescence, immunoelectron microscopy, and single-cell reverse-transcriptase polymerase chain reaction amplification (RT-PCR) to analyze the expression and cellular localization of Cx26 in the carp retina. In the outer plexiform layer, strong clustered Cx26 immunolabeling was concentrated at and restricted to the terminal dendrites of horizontal cells. Single-cell RT-PCR confirmed the expression of Cx26 in carp retinal horizontal cells. 248-bp fragments amplified from cDNAs of four different horizontal cells were cloned and each nucleotide sequence encodes a protein fragment (AA 104-185) with highly significant homology to rat and mouse Cx26. Immunoelectron microscopy revealed that only the invaginating dendrites of horizontal cells in intimate lateral association with the presynaptic ribbon complex were labeled. No labeling was found at the photoreceptor membrane and there was no septalaminar structure, indicative of gap junctions, between photoreceptors and horizontal cells. The focal location of Cx26 at the membrane of the dendritic tips of horizontal cells and the lack of gap junctional morphology suggests that Cx26 might form hemichannels.

Collaboration


Dive into the Ulrike Janssen-Bienhold's collaboration.

Top Co-Authors

Avatar

Reto Weiler

University of Oldenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karin Dedek

University of Oldenburg

View shared research outputs
Top Co-Authors

Avatar

Petra Bolte

University of Oldenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Helena Greb

University of Oldenburg

View shared research outputs
Researchain Logo
Decentralizing Knowledge