Network


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

Hotspot


Dive into the research topics where Werner Müller-Esterl is active.

Publication


Featured researches published by Werner Müller-Esterl.


Journal of Clinical Investigation | 2006

Targeting the heme-oxidized nitric oxide receptor for selective vasodilatation of diseased blood vessels

Johannes-Peter Stasch; Peter Schmidt; Pavel I. Nedvetsky; Tatiana Y. Nedvetskaya; H S Arun Kumar; Sabine Meurer; Martin Deile; Ashraf Taye; Andreas Knorr; Harald Lapp; Helmut Müller; Yagmur Turgay; Christiane Rothkegel; Adrian Tersteegen; Barbara Kemp-Harper; Werner Müller-Esterl; Harald Schmidt

ROS are a risk factor of several cardiovascular disorders and interfere with NO/soluble guanylyl cyclase/cyclic GMP (NO/sGC/cGMP) signaling through scavenging of NO and formation of the strong oxidant peroxynitrite. Increased oxidative stress affects the heme-containing NO receptor sGC by both decreasing its expression levels and impairing NO-induced activation, making vasodilator therapy with NO donors less effective. Here we show in vivo that oxidative stress and related vascular disease states, including human diabetes mellitus, led to an sGC that was indistinguishable from the in vitro oxidized/heme-free enzyme. This sGC variant represents what we believe to be a novel cGMP signaling entity that is unresponsive to NO and prone to degradation. Whereas high-affinity ligands for the unoccupied heme pocket of sGC such as zinc-protoporphyrin IX and the novel NO-independent sGC activator 4-[((4-carboxybutyl){2-[(4-phenethylbenzyl)oxy]phenethyl}amino) methyl [benzoic]acid (BAY 58-2667) stabilized the enzyme, only the latter activated the NO-insensitive sGC variant. Importantly, in isolated cells, in blood vessels, and in vivo, BAY 58-2667 was more effective and potentiated under pathophysiological and oxidative stress conditions. This therapeutic principle preferentially dilates diseased versus normal blood vessels and may have far-reaching implications for the currently investigated clinical use of BAY 58-2667 as a unique diagnostic tool and highly innovative vascular therapy.


Journal of Biological Chemistry | 1996

The Serum Protein α2-HS Glycoprotein/Fetuin Inhibits Apatite Formation in Vitro and in Mineralizing Calvaria Cells A POSSIBLE ROLE IN MINERALIZATION AND CALCIUM HOMEOSTASIS

Thorsten Schinke; Christiane Amendt; Andreas Trindl; Oliver Pöschke; Werner Müller-Esterl; Willi Jahnen-Dechent

We present data suggesting a function of α2-HS glycoproteins/fetuins in serum and in mineralization, namely interference with calcium salt precipitation. Fetuins occur in high serum concentration during fetal life. They accumulate in bones and teeth as a major fraction of noncollagenous bone proteins. The expression pattern in fetal mice confirms that fetuin is predominantly made in the liver and is accumulated in the mineralized matrix of bones. We arrived at a hypothesis on the molecular basis of fetuin function in bones using primary rat calvaria osteoblast cultures and salt precipitation assays. Our results indicate that fetuins inhibit apatite formation both in cell culture and in the test tube. This inhibitory effect is mediated by acidic amino acids clustering in cystatin-like domain D1. Fetuins account for roughly half of the capacity of serum to inhibit salt precipitation. We propose that fetuins inhibit phase separation in serum and modulate apatite formation during mineralization.


Journal of Cell Biology | 2001

Functional specialization of calreticulin domains

Kimitoshi Nakamura; Anna Zuppini; Serge Arnaudeau; Jeffery Lynch; Irfan Ahsan; Ryoko Krause; Sylvia Papp; Humbert De Smedt; Jan B. Parys; Werner Müller-Esterl; Daniel Pablo Lew; Karl-Heinz Krause; Nicolas Demaurex; Michal Opas; Marek Michalak

Calreticulin is a Ca2+-binding chaperone in the endoplasmic reticulum (ER), and calreticulin gene knockout is embryonic lethal. Here, we used calreticulin-deficient mouse embryonic fibroblasts to examine the function of calreticulin as a regulator of Ca2+ homeostasis. In cells without calreticulin, the ER has a lower capacity for Ca2+ storage, although the free ER luminal Ca2+ concentration is unchanged. Calreticulin-deficient cells show inhibited Ca2+ release in response to bradykinin, yet they release Ca2+ upon direct activation with the inositol 1,4,5-trisphosphate (InsP3). These cells fail to produce a measurable level of InsP3 upon stimulation with bradykinin, likely because the binding of bradykinin to its cell surface receptor is impaired. Bradykinin binding and bradykinin-induced Ca2+ release are both restored by expression of full-length calreticulin and the N + P domain of the protein. Expression of the P + C domain of calreticulin does not affect bradykinin-induced Ca2+ release but restores the ER Ca2+ storage capacity. Our results indicate that calreticulin may play a role in folding of the bradykinin receptor, which affects its ability to initiate InsP3-dependent Ca2+ release in calreticulin-deficient cells. We concluded that the C domain of calreticulin plays a role in Ca2+ storage and that the N domain may participate in its chaperone functions.


Journal of Biological Chemistry | 1997

Cloning and Targeted Deletion of the Mouse Fetuin Gene

Willi Jahnen-Dechent; Thorsten Schinke; Andreas Trindl; Werner Müller-Esterl; Fred Sablitzky; Sibylle Kaiser; Manfred Blessing

We proposed that the α2-Heremans Schmid glycoprotein/fetuin family of serum proteins inhibits unwanted mineralization. To test this hypothesis in animals, we cloned the mouse fetuin gene and generated mice lacking fetuin. The gene consists of seven exons and six introns. The cystatin-like domains D1 and D2 of mouse fetuin are encoded by three exons each, whereas a single terminal exon encodes the carboxyl-terminal domain D3. The promoter structure is well conserved between rat and mouse fetuin genes within the regions shown to bind transcription factors in the rat system. Expression studies demonstrated that mice homozygous for the gene deletion lacked fetuin protein and that mice heterozygous for the null mutation produced roughly half the amount of fetuin protein produced by wild-type mice. Fetuin-deficient mice were fertile and showed no gross anatomical abnormalities. However, the serum inhibition of apatite formation was compromised in these mice as well as in heterozygotes. In addition, some homozygous fetuin-deficient female ex-breeders developed ectopic microcalcifications in soft tissues. These results corroborate a role for fetuin in serum calcium homeostasis. The fact that generalized ectopic calcification did not occur in fetuin-deficient mice proves that additional inhibitors of phase separation exist in serum.


The FASEB Journal | 2001

NOSIP, a novel modulator of endothelial nitric oxide synthase activity

Jürgen Dedio; Peter König; Paulus Wohlfart; C. Schroeder; Wolfgang Kummer; Werner Müller-Esterl

Production of nitric oxide (NO) in endothelial cells is regulated by direct interactions of endothelial nitric oxide synthase (eNOS) with effector proteins such as Ca2+‐calmodulin, by posttranslational modifications such as phosphorylation via protein kinase B, and by translocation of the enzyme from the plasma membrane caveolae to intracellular compartments. Reversible acylation of eNOS is thought to contribute to the intracellular trafficking of the enzyme;however, protein factor(s) that govern the translocation of the enzyme are still unknown. Here we have used the yeast two‐hybrid system and identified a novel 34 kDa protein, termed NOSIP (eNOS interacting protein), which avidly binds to the carboxyl‐terminal region of the eNOS oxygenase domain. Coimmunoprecipitation studies demonstrated the specific interaction of eNOS and NOSIP in vitro and in vivo, and complex formation was inhibited by a synthetic peptide of the caveolin‐1 scaffolding domain. NO production was significantly reduced in eNOS‐expressing CHO cells (CHO‐eNOS) that transiently overexpressed NOSIP. Stimulation with the calcium ionophore A23187 induced the reversible translocation of eNOS from the detergentinsoluble to the detergent‐soluble fractions of CHO‐eNOS, and this translocation was completelyprevented by transient coexpression of NOSIP in CHO‐eNOS. Immunofluorescence studies revealed a prominent plasma membrane staining for eNOS in CHO‐eNOS that was abolished in the presence of NOSIP. Subcellular fractionation studies identified eNOS in the caveolin‐rich membrane fractions of CHO‐eNOS, and coexpression of NOSIP caused a shift of eNOS to intracellular compartments. We conclude that NOSIP is a novel type of modulator that promotes translocation of eNOS from the plasma membrane to intracellular sites, thereby uncoupling eNOS from plasma membrane caveolae and inhibiting NO synthesis.—Dedio, J., König, P., Wohlfart, P., Schroeder, C., Kummer, W., Muller‐Esterl, W. NOSIP, a novel modulator of endothelial nitric oxide synthase activity. FASEB J. 15, 79–89 (2001)


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

NOSTRIN: A protein modulating nitric oxide release and subcellular distribution of endothelial nitric oxide synthase

Kirstin Zimmermann; Nils Opitz; Jürgen Dedio; Christoph Renné; Werner Müller-Esterl; Stefanie Oess

Activity and localization of endothelial nitric oxide synthase (eNOS) is regulated in a remarkably complex fashion, yet the complex molecular machinery mastering stimulus-induced eNOS translocation and trafficking is poorly understood. In a search by the yeast two-hybrid system using the eNOS oxygenase domain as bait, we have identified a previously uncharacterized eNOS-interacting protein, dubbed NOSTRIN (for eNOS traffic inducer). NOSTRIN contains a single polypeptide chain of 506-aa residues of 58 kDa with an N-terminal cdc15 domain and a C-terminal SH3 domain. NOSTRIN mRNA is abundant in highly vascularized tissues such as placenta, kidney, lung, and heart, and NOSTRIN protein is expressed in vascular endothelial cells. Coimmunoprecipitation experiments demonstrated the eNOS–NOSTRIN interaction in vitro and in vivo, and NOSTRINs SH3 domain was essential and sufficient for eNOS binding. NOSTRIN colocalized extensively with eNOS at the plasma membrane of confluent human umbilical venous endothelial cells and in punctate cytosolic structures of CHO-eNOS cells. NOSTRIN overexpression induced a profound redistribution of eNOS from the plasma membrane to vesicle-like structures matching the NOSTRIN pattern and at the same time led to a significant inhibition of NO release. We conclude that NOSTRIN contributes to the intricate protein network controlling activity, trafficking, and targeting of eNOS.


Molecular Microbiology | 1996

Assembly of human contact phase proteins and release of bradykinin at the surface of curli-expressing Escherichia coli.

Abdelhakim Ben Nasr; Arne Olsén; Ulf Sjöbring; Werner Müller-Esterl; Lars Björck

Previous work has demonstrated that most strains of the human pathogen Streptococcus pyogenes bind kininogens through M protein, a fibrous surface protein and virulence determinant. Here we find that strains of several other pathogenic bacterial species, both Gram‐positive and Gram‐negative, isolated from patients with sepsis, also bind kininogens, especially H‐kininogen (HK). The most pronounced interaction was seen between HK and Escherichia coli. Among clinical isolates of E. coli, the majority of the entero‐haemorrhagic, enterotoxigenic, and sepsis strains, but none of the enteroinvasive and enteropathogenic strains, bound HK. Binding of HK to E. coli correlated with the expression of curli, another fibrous bacterial surface protein, and the binding of HK to purified curli was specific, saturable, and of high affinity; Ka = 9 107M‐1. Other contact phase proteins such as factor XI, factor XII, and prekallikrein bound to curliated E. coli, but not to an isogenic curli‐deficient mutant strain, suggesting that contact phase activation may occur at the surface of curliated bacteria. Kininogens are also precursor molecules of the vasoactive kinins. When incubated with human plasma, curli‐expressing bacteria absorbed HK. Addition of purified plasma kallikrein to the HK‐loaded bacteria resulted in a rapid and efficient release of bradykinin from surface‐bound HK. The assembly of contact phase factors at the surface of pathogenic bacteria and the release of the potent proinflammatory and vasoactive peptide bradykinin, should have a major impact on the host‐microbe relationship and may contribute to bacterial pathogencity and virulence.


Journal of Biological Chemistry | 1996

Ligand-induced Phosphorylation/Dephosphorylation of the Endogenous Bradykinin B2 Receptor from Human Fibroblasts

Andree Blaukat; Said Abd Alla; Martin J. Lohse; Werner Müller-Esterl

We have studied the ligand-induced phosphorylation/dephosphorylation of the bradykinin B2 receptor endogenously expressed in human HF-15 fibroblasts. An antiserum (AS346) to a synthetic peptide (CRS36), derived from the extreme carboxyl terminus of the human B2 receptor, precipitated the receptor from solubilized membranes of HF-15 cells that had been labeled with [32P]orthophosphate. A low basal level of B2 receptor phosphorylation was found in the absence of a ligand. Stimulation of the cells with the B2 receptor agonists bradykinin, [Lys0,Hyp3]bradykinin, kallidin, and T-kinin resulted in a rapid and efficient phosphorylation of the receptor. The B2 receptor antagonist HOE140 and the B1 receptor agonist des-Arg9-bradykinin failed to induce significant phosphorylation of the B2 receptor. Phosphoamino acid analysis revealed that the B2 receptor is phosphorylated on serine and threonine, but not on tyrosine residues. The ligand-induced phosphorylation of the receptor was concentration-dependent, with an apparent EC50 of 33 nM, and peaked at 1 min after challenge. The kinin-stimulated phosphorylation of the B2 receptor was rapid and transient and paralleled the kinetics of desensitization/resensitization of the receptor as followed by [Ca2+]i release and radioligand binding assay, respectively. The ligand-induced phosphorylation of the B2 receptor was independent of the protein kinase C pathway. In vitro experiments suggest βARK1 (-drenergic eceptor inase) as a candidate kinase that could mediate the homologous B2 receptor phosphorylation. Inhibitors of protein phosphatases 1 and 2A effectively blocked the dephosphorylation, but did not affect the internalization of the B2 receptor, whereas inhibitors of receptor internalization delayed its dephosphorylation. These finding point to a role of ligand-induced phosphorylation in the desensitization and redistribution of the bradykinin receptor in human fibroblasts.


Journal of Immunology | 2009

Activation of mammalian target of rapamycin controls the loss of TCRzeta in lupus T cells through HRES-1/Rab4-regulated lysosomal degradation.

David Fernandez; Tiffany Telarico; Eduardo Bonilla; Qing Li; Sanjay K. Banerjee; Frank A. Middleton; Paul E. Phillips; Mary K. Crow; Stefanie Oess; Werner Müller-Esterl; Andras Perl

Persistent mitochondrial hyperpolarization (MHP) and enhanced calcium fluxing underlie aberrant T cell activation and death pathway selection in systemic lupus erythematosus. Treatment with rapamycin, which effectively controls disease activity, normalizes CD3/CD28-induced calcium fluxing but fails to influence MHP, suggesting that altered calcium fluxing is downstream or independent of mitochondrial dysfunction. In this article, we show that activity of the mammalian target of rapamycin (mTOR), which is a sensor of the mitochondrial transmembrane potential, is increased in lupus T cells. Activation of mTOR was inducible by NO, a key trigger of MHP, which in turn enhanced the expression of HRES-1/Rab4, a small GTPase that regulates recycling of surface receptors through early endosomes. Expression of HRES-1/Rab4 was increased in CD4+ lupus T cells, and in accordance with its dominant impact on the endocytic recycling of CD4, it was inversely correlated with diminished CD4 expression. HRES-1/Rab4 overexpression was also inversely correlated with diminished TCRζ protein levels. Pull-down studies revealed a direct interaction of HRES-1/Rab4 with CD4 and TCRζ. Importantly, the deficiency of the TCRζ chain and of Lck and the compensatory up-regulation of FcεRIγ and Syk, which mediate enhanced calcium fluxing in lupus T cells, were reversed in patients treated with rapamcyin in vivo. Knockdown of HRES-1/Rab4 by small interfering RNA and inhibitors of lysosomal function augmented TCRζ protein levels in vitro. The results suggest that activation of mTOR causes the loss of TCRζ in lupus T cells through HRES-1/Rab4-dependent lysosomal degradation.


Circulation | 1999

Angiotensin-Converting Enzyme Inhibitor Ramiprilat Interferes With the Sequestration of the B2 Kinin Receptor Within the Plasma Membrane of Native Endothelial Cells

Thomas Benzing; Ingrid Fleming; Andree Blaukat; Werner Müller-Esterl; Rudi Busse

BACKGROUND ACE (kininase II) inhibitors have been shown to exert their beneficial cardiovascular effects via the inhibition of both angiotensin II formation and bradykinin breakdown. Because recent evidence suggests that ACE inhibitors may also interfere with B2 kinin receptor signaling and thus enhance the vascular response to bradykinin, we examined whether the distribution of B2 kinin receptors within the plasma membrane of native endothelial cells is affected by an ACE inhibitor. METHODS AND RESULTS Localization of the B2 kinin receptor in membranes prepared from native porcine aortic endothelial cells was evaluated by means of specific [3H]bradykinin binding and immunoprecipitation of the B2 receptor from isolated membranes. Effects of bradykinin and ramiprilat on intracellular signaling were determined by monitoring the activation of the extracellularly regulated kinases Erk1 and Erk2 as well as [Ca2+]i increases in fura 2-loaded endothelial cells. Stimulation of native endothelial cells with bradykinin 100 nmol/L resulted in the time-dependent sequestration of the B2 receptor to caveolin-rich (CR) membranes, which was maximal after 5 minutes. Pretreatment with ramiprilat 100 nmol/L for 15 minutes significantly attenuated the recovery of B2 kinin receptors in CR membranes while increasing that from membranes lacking caveolin. This effect was not due to the inhibition of bradykinin degradation, because no effect was seen in the presence of an inhibitory concentration of the synthetic ACE substrate hippuryl-L-histidyl-L-leucine. Ramiprilat also decreased [3H]bradykinin binding to CR membranes when applied either before or after bradykinin stimulation. Moreover, ramiprilat resulted in reactivation of the B2 receptor in bradykinin-stimulated cells and induced a second peak in [Ca2+]i and reactivation of Erk1/2. CONCLUSIONS The ACE inhibitor ramiprilat interferes with the targeting of the B2 kinin receptor to CR membrane domains in native endothelial cells. Therefore, effects other than the inhibition of kininase II may account for the effects of ramiprilat and other ACE inhibitors on the vascular system.

Collaboration


Dive into the Werner Müller-Esterl's collaboration.

Top Co-Authors

Avatar

Stefanie Oess

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ingrid Fleming

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Rudi Busse

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Sylke Pioch

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ivan Dikic

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge