Network


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

Hotspot


Dive into the research topics where Enno Klussmann is active.

Publication


Featured researches published by Enno Klussmann.


Journal of Biological Chemistry | 1999

Protein Kinase A Anchoring Proteins Are Required for Vasopressin-mediated Translocation of Aquaporin-2 into Cell Membranes of Renal Principal Cells

Enno Klussmann; Kenan Maric; Burkhard Wiesner; Michael Beyermann; Walter Rosenthal

The antidiuretic hormone arginine-vasopressin (AVP) regulates water reabsorption in renal collecting duct principal cells by inducing a cAMP-dependent translocation of water channels (aquaporin-2, AQP-2) from intracellular vesicles into the apical cell membranes. In subcellular fractions from primary cultured rat inner medullary collecting duct (IMCD) cells, enriched for intracellular AQP-2-bearing vesicles, catalytic protein kinase A (PKA) subunits and several protein kinase A anchoring proteins (AKAPs) were detected. In nonstimulated IMCD cells the majority of AQP-2 staining was detected intracellularly but became mainly localized within the cell membrane after stimulation with AVP or forskolin. Quantitative analysis revealed that preincubation of the cells with the synthetic peptide S-Ht31, which prevents the binding between AKAPs and regulatory subunits of PKA, strongly inhibited AQP-2 translocation in response to forskolin. Preincubation of the cells with the PKA inhibitor H89 prior to forskolin stimulation abolished AQP-2 translocation. In contrast to H89, S-Ht31 did not affect the catalytic activity of PKA. These data demonstrate that not only the activity of PKA, but also its tethering to subcellular compartments, are prerequisites for cAMP-dependent AQP-2 translocation.


EMBO Reports | 2007

AKAP complex regulates Ca2+ re‐uptake into heart sarcoplasmic reticulum

Birgitte Lygren; Cathrine R. Carlson; Katja Santamaria; Valentina Lissandron; Theresa McSorley; Jessica Litzenberg; Dorothea Lorenz; Burkhard Wiesner; Walter Rosenthal; Manuela Zaccolo; Kjetil Taskén; Enno Klussmann

The β‐adrenergic receptor/cyclic AMP/protein kinase A (PKA) signalling pathway regulates heart rate and contractility. Here, we identified a supramolecular complex consisting of the sarcoplasmic reticulum Ca2+‐ATPase (SERCA2), its negative regulator phospholamban (PLN), the A‐kinase anchoring protein AKAP18δ and PKA. We show that AKAP18δ acts as a scaffold that coordinates PKA phosphorylation of PLN and the adrenergic effect on Ca2+ re‐uptake. Inhibition of the compartmentalization of this cAMP signalling complex by specific molecular disruptors interferes with the phosphorylation of PLN. This prevents the subsequent release of PLN from SERCA2, thereby affecting the Ca2+ re‐uptake into the sarcoplasmic reticulum induced by adrenergic stimuli.


Journal of Biological Chemistry | 2001

An Inhibitory Role of Rho in the Vasopressin-mediated Translocation of Aquaporin-2 into Cell Membranes of Renal Principal Cells

Enno Klussmann; Grazia Tamma; Dorothea Lorenz; Burkhard Wiesner; Kenan Maric; Fred Hofmann; Klaus Aktories; Giovanna Valenti; Walter Rosenthal

Vasopressin regulates water reabsorption in renal collecting duct principal cells by a cAMP-dependent translocation of the water channel aquaporin-2 (AQP2) from intracellular vesicles into the cell membrane. In the present work primary cultured inner medullary collecting duct cells were used to study the role of the proteins of the Rho family in the translocation of AQP2. Clostridium difficile toxin B, which inhibits all members of the Rho family, Clostridium limosum C3 toxin, which inactivates only Rho, and the Rho kinase inhibitor, Y-27632, induced both depolymerization of actin stress fibers and AQP2 translocation in the absence of vasopressin. The data suggest an inhibitory role of Rho in this process, whereby constitutive membrane localization is prevented in resting cells. Expression of constitutively active RhoA induced formation of actin stress fibers and abolished AQP2 translocation in response to elevation of intracellular cAMP, confirming the inhibitory role of Rho. Cytochalasin D induced both depolymerization of the F-actin cytoskeleton and AQP2 translocation, indicating that depolymerization of F-actin is sufficient to induce AQP2 translocation. Thus Rho is likely to control the intracellular localization of AQP2 via regulation of the F-actin cytoskeleton.


International Review of Cell and Molecular Biology | 2010

Mechanisms of protein kinase A anchoring

Philipp Skroblin; Solveig Grossmann; Gesa Schäfer; Walter Rosenthal; Enno Klussmann

The second messenger cyclic adenosine monophosphate (cAMP), which is produced by adenylyl cyclases following stimulation of G-protein-coupled receptors, exerts its effect mainly through the cAMP-dependent serine/threonine protein kinase A (PKA). Due to the ubiquitous nature of the cAMP/PKA system, PKA signaling pathways underlie strict spatial and temporal control to achieve specificity. A-kinase anchoring proteins (AKAPs) bind to the regulatory subunit dimer of the tetrameric PKA holoenzyme and thereby target PKA to defined cellular compartments in the vicinity of its substrates. AKAPs promote the termination of cAMP signals by recruiting phosphodiesterases and protein phosphatases, and the integration of signaling pathways by binding additional signaling proteins. AKAPs are a heterogeneous family of proteins that only display similarity within their PKA-binding domains, amphipathic helixes docking into a hydrophobic groove formed by the PKA regulatory subunit dimer. This review summarizes the current state of information on compartmentalized cAMP/PKA signaling with a major focus on structural aspects, evolution, diversity, and (patho)physiological functions of AKAPs and intends to outline newly emerging directions of the field, such as the elucidation of AKAP mutations and alterations of AKAP expression in human diseases, and the validation of AKAP-dependent protein-protein interactions as new drug targets. In addition, alternative PKA anchoring mechanisms employed by noncanonical AKAPs and PKA catalytic subunit-interacting proteins are illustrated.


Journal of Biological Chemistry | 2004

Identification of a Novel A-kinase Anchoring Protein 18 Isoform and Evidence for Its Role in the Vasopressin-induced Aquaporin-2 Shuttle in Renal Principal Cells

Volker Henn; Bayram Edemir; Eduard Stefan; Burkhard Wiesner; Dorothea Lorenz; Franziska Theilig; Roland Schmitt; Lutz Vossebein; Grazia Tamma; Michael Beyermann; Eberhard Krause; Friedrich W. Herberg; Giovana Valenti; S. Bachmann; Walter Rosenthal; Enno Klussmann

Arginine vasopressin (AVP) increases the water permeability of renal collecting duct principal cells by inducing the fusion of vesicles containing the water channel aquaporin-2 (AQP2) with the plasma membrane (AQP2 shuttle). This event is initiated by activation of vasopressin V2 receptors, followed by an elevation of cAMP and the activation of protein kinase A (PKA). The tethering of PKA to subcellular compartments by protein kinase A anchoring proteins (AKAPs) is a prerequisite for the AQP2 shuttle. During the search for AKAP(s) involved in the shuttle, a new splice variant of AKAP18, AKAP18δ, was identified. AKAP18δ functions as an AKAP in vitro and in vivo. In the kidney, it is mainly expressed in principal cells of the inner medullary collecting duct, closely resembling the distribution of AQP2. It is present in both the soluble and particulate fractions derived from renal inner medullary tissue. Within the particulate fraction, AKAP18δ was identified on the same intracellular vesicles as AQP2 and PKA. AVP not only recruited AQP2, but also AKAP18δ to the plasma membrane. The elevation of cAMP caused the dissociation of AKAP18δ and PKA. The data suggest that AKAP18δ is involved in the AQP2 shuttle.


Traffic | 2007

A Role of Myosin Vb and Rab11-FIP2 in the Aquaporin-2 Shuttle

Pavel I. Nedvetsky; Eduard Stefan; Sebastian Frische; Katja Santamaria; Burkhard Wiesner; Giovanna Valenti; John A. Hammer; Søren Nielsen; James R. Goldenring; Walter Rosenthal; Enno Klussmann

Arginine‐vasopressin (AVP) regulates water reabsorption in renal collecting duct principal cells. Its binding to Gs‐coupled vasopressin V2 receptors increases cyclic AMP (cAMP) and subsequently elicits the redistribution of the water channel aquaporin‐2 (AQP2) from intracellular vesicles into the plasma membrane (AQP2 shuttle), thereby facilitating water reabsorption from primary urine. The AQP2 shuttle is a paradigm for cAMP‐dependent exocytic processes. Using sections of rat kidney, the AQP2‐expressing cell line CD8, and primary principal cells, we studied the role of the motor protein myosin Vb, its vesicular receptor Rab11, and the myosin Vb‐ and Rab11‐binding protein Rab11‐FIP2 in the AQP2 shuttle. Myosin Vb colocalized with AQP2 intracellularly in resting and at the plasma membrane in AVP‐treated cells. Rab11 was found on AQP2‐bearing vesicles. A dominant‐negative myosin Vb tail construct and Rab11‐FIP2 lacking the C2 domain (Rab11‐FIP2‐ΔC2), which disrupt recycling, caused condensation of AQP2 in a Rab11‐positive compartment and abolished the AQP2 shuttle. This effect was dependent on binding of myosin Vb tail and Rab11‐FIP2‐ΔC2 to Rab11. In summary, we identified myosin Vb as a motor protein involved in AQP2 recycling and show that myosin Vb‐ and Rab11‐FIP2‐dependent recycling of AQP2 is an integral part of the AQP2 shuttle.


Biochemical Journal | 2006

High-affinity AKAP7δ–protein kinase A interaction yields novel protein kinase A-anchoring disruptor peptides

Christian Hundsrucker; Gerd Krause; Michael Beyermann; Anke Prinz; Bastian Zimmermann; Oliver Diekmann; Dorothea Lorenz; Eduard Stefan; Pavel I. Nedvetsky; Margitta Dathe; Frank Christian; Theresa McSorley; Eberhard Krause; George McConnachie; Friedrich W. Herberg; John D. Scott; Walter Rosenthal; Enno Klussmann

PKA (protein kinase A) is tethered to subcellular compartments by direct interaction of its regulatory subunits (RI or RII) with AKAPs (A kinase-anchoring proteins). AKAPs preferentially bind RII subunits via their RII-binding domains. RII-binding domains form structurally conserved amphipathic helices with unrelated sequences. Their binding affinities for RII subunits differ greatly within the AKAP family. Amongst the AKAPs that bind RIIalpha subunits with high affinity is AKAP7delta [AKAP18delta; K(d) (equilibrium dissociation constant) value of 31 nM]. An N-terminally truncated AKAP7delta mutant binds RIIalpha subunits with higher affinity than the full-length protein presumably due to loss of an inhibitory region [Henn, Edemir, Stefan, Wiesner, Lorenz, Theilig, Schmidtt, Vossebein, Tamma, Beyermann et al. (2004) J. Biol. Chem. 279, 26654-26665]. In the present study, we demonstrate that peptides (25 amino acid residues) derived from the RII-binding domain of AKAP7delta bind RIIalpha subunits with higher affinity (K(d)=0.4+/-0.3 nM) than either full-length or N-terminally truncated AKAP7delta, or peptides derived from other RII binding domains. The AKAP7delta-derived peptides and stearate-coupled membrane-permeable mutants effectively disrupt AKAP-RII subunit interactions in vitro and in cell-based assays. Thus they are valuable novel tools for studying anchored PKA signalling. Molecular modelling indicated that the high affinity binding of the amphipathic helix, which forms the RII-binding domain of AKAP7delta, with RII subunits involves both the hydrophobic and the hydrophilic faces of the helix. Alanine scanning (25 amino acid peptides, SPOT technology, combined with RII overlay assays) of the RII binding domain revealed that hydrophobic amino acid residues form the backbone of the interaction and that hydrogen bond- and salt-bridge-forming amino acid residues increase the affinity of the interaction.


Journal of Biological Chemistry | 2011

Small Molecule AKAP-Protein Kinase A (PKA) Interaction Disruptors That Activate PKA Interfere with Compartmentalized cAMP Signaling in Cardiac Myocytes

Frank Christian; Márta Szaszák; Sabine Friedl; Stephan Drewianka; Dorothea Lorenz; Andrey C. da Costa Goncalves; Jens Furkert; Carolyn Vargas; Peter Schmieder; Frank Götz; Kerstin Zühlke; Marie Moutty; Hendrikje Göttert; Mangesh Joshi; Bernd Reif; Hannelore Haase; Ingo Morano; Solveig Grossmann; Anna Klukovits; Judit Verli; Róbert Gáspár; Claudia Noack; Martin W. Bergmann; Robert S. Kass; Kornelia Hampel; Dmitry Kashin; Hans Gottfried Genieser; Friedrich W. Herberg; Debbie Willoughby; Dermot M. F. Cooper

A-kinase anchoring proteins (AKAPs) tether protein kinase A (PKA) and other signaling proteins to defined intracellular sites, thereby establishing compartmentalized cAMP signaling. AKAP-PKA interactions play key roles in various cellular processes, including the regulation of cardiac myocyte contractility. We discovered small molecules, 3,3′-diamino-4,4′-dihydroxydiphenylmethane (FMP-API-1) and its derivatives, which inhibit AKAP-PKA interactions in vitro and in cultured cardiac myocytes. The molecules bind to an allosteric site of regulatory subunits of PKA identifying a hitherto unrecognized region that controls AKAP-PKA interactions. FMP-API-1 also activates PKA. The net effect of FMP-API-1 is a selective interference with compartmentalized cAMP signaling. In cardiac myocytes, FMP-API-1 reveals a novel mechanism involved in terminating β-adrenoreceptor-induced cAMP synthesis. In addition, FMP-API-1 leads to an increase in contractility of cultured rat cardiac myocytes and intact hearts. Thus, FMP-API-1 represents not only a novel means to study compartmentalized cAMP/PKA signaling but, due to its effects on cardiac myocytes and intact hearts, provides the basis for a new concept in the treatment of chronic heart failure.


International Journal of Cancer | 1997

Low O-acetylation of sialyl-LEx contributes to its overexpression in colon carcinoma metastases

Benno Mann; Enno Klussmann; Valérie Vandamme-Feldhaus; Matthias Iwersen; Marie-Luise Hanski; Ernst-Otto Riecken; Heinz J. Buhr; Roland Schauer; Young S. Kim; Christoph Hanski

Two factors potentially determining the consistent over‐expression of sialyl‐Lex antigen in colon carcinoma and metastases were investigated: (i) the expression of the mucins MUC1 and MUC2, known to carry sialyl‐Lex, by Northern blotting; (ii) the extent of sialic acid O‐acetylation, by Western blotting and HPLC. RNA and sialyl‐Lex‐positive mucins were purified from normal colonic mucosa (N), primary carcinomas (T) and their liver metastases (M). Northern blots showed that mRNA expression both of MUC1 and of MUC2 decreases during the progression of the disease, and is lowest in metastatic tissue. The expression of mucin‐bound sialyl‐Lex increased strongly from N to T and, to a lesser extent, to M. After alkali treatment of the mucins these differences disappeared, indicating that the total amount of mucin‐bound sialyl‐Lex is the same in the 3 types of tissues. The O‐acetylation of mucin‐bound sialyl‐Lex gradually decreased from N to M. HPLC analysis showed that in N about 70%, in T 45% and in M only 20% of mucin‐bound sialic acids are O‐acetylated. Thus, the increase of sialyl‐Lex detectable during colon‐carcinoma progression is due to diminished O‐acetylation and not to increased expression of mucin protein cores. The decrease of O‐acetylation is therefore the primary chemical alteration contributing to colon carcinoma‐associated overexpression of sialyl‐Lex. Int. J. Cancer 72:258–264, 1997.


Handbook of experimental pharmacology | 2008

Direct AKAP-mediated protein-protein interactions as potential drug targets

Christian Hundsrucker; Enno Klussmann

A-kinase-anchoring proteins (AKAPs) are a diverse family of about 50 scaffolding proteins. They are defined by the presence of a structurally conserved protein kinase A (PKA)-binding domain. AKAPs tether PKA and other signalling proteins such as further protein kinases, protein phosphatases and phosphodiesterases by direct protein-protein interactions to cellular compartments. Thus, AKAPs form the basis of signalling modules that integrate cellular signalling processes and limit these to defined sites. Disruption of AKAP functions by gene targeting, knockdown approaches and, in particular, pharmacological disruption of defined AKAP-dependent protein-protein interactions has revealed key roles of AKAPs in numerous processes, including the regulation of cardiac myocyte contractility and vasopressin-mediated water reabsorption in the kidney. Dysregulation of such processes causes diseases, including cardiovascular and renal disorders. In this review, we discuss AKAP functions elucidated by gene targeting and knockdown approaches, but mainly focus on studies utilizing peptides for disruption of direct AKAP-mediated protein-protein interactions. The latter studies point to direct AKAP-mediated protein-protein interactions as targets for novel drugs.

Collaboration


Dive into the Enno Klussmann'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
Researchain Logo
Decentralizing Knowledge