Network


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

Hotspot


Dive into the research topics where Susanne Ullrich is active.

Publication


Featured researches published by Susanne Ullrich.


Nature | 2013

T-helper-1-cell cytokines drive cancer into senescence

Heidi Braumüller; Thomas Wieder; Ellen Brenner; Sonja Aßmann; Matthias Hahn; Mohammed Alkhaled; Karin Schilbach; Frank Essmann; Manfred Kneilling; Christoph M. Griessinger; Felicia Ranta; Susanne Ullrich; Ralph Mocikat; Kilian Braungart; Tarun Mehra; Birgit Fehrenbacher; Julia Berdel; Heike Niessner; Friedegund Meier; Maries van den Broek; Hans-Ulrich Häring; Rupert Handgretinger; Leticia Quintanilla-Martinez; Falko Fend; Marina Pesic; Jürgen Bauer; Lars Zender; Martin Schaller; Klaus Schulze-Osthoff; Martin Röcken

Cancer control by adaptive immunity involves a number of defined death and clearance mechanisms. However, efficient inhibition of exponential cancer growth by T cells and interferon-γ (IFN-γ) requires additional undefined mechanisms that arrest cancer cell proliferation. Here we show that the combined action of the T-helper-1-cell cytokines IFN-γ and tumour necrosis factor (TNF) directly induces permanent growth arrest in cancers. To safely separate senescence induced by tumour immunity from oncogene-induced senescence, we used a mouse model in which the Simian virus 40 large T antigen (Tag) expressed under the control of the rat insulin promoter creates tumours by attenuating p53- and Rb-mediated cell cycle control. When combined, IFN-γ and TNF drive Tag-expressing cancers into senescence by inducing permanent growth arrest in G1/G0, activation of p16INK4a (also known as CDKN2A), and downstream Rb hypophosphorylation at serine 795. This cytokine-induced senescence strictly requires STAT1 and TNFR1 (also known as TNFRSF1A) signalling in addition to p16INK4a. In vivo, Tag-specific T-helper 1 cells permanently arrest Tag-expressing cancers by inducing IFN-γ- and TNFR1-dependent senescence. Conversely, Tnfr1−/− Tag-expressing cancers resist cytokine-induced senescence and grow aggressively, even in TNFR1-expressing hosts. Finally, as IFN-γ and TNF induce senescence in numerous murine and human cancers, this may be a general mechanism for arresting cancer progression.


Molecular Microbiology | 2005

Intracellular multiplication of Legionella pneumophila depends on host cell amino acid transporter SLC1A5

Hagen Wieland; Susanne Ullrich; Florian Lang; Birgid Neumeister

The infectious agent of Legionnaires’ disease, Legionella (L) pneumophila, multiplies intracellularly in eukaryotic cells. This study has been performed to explore the nutrient requirements of L. pneumophila during intracellular replication. In human monocytes, bacterial replication rate was reduced by 76% in defined medium lacking l‐cysteine, l‐glutamine or l‐serine. SLC1A5 (hATB0,+), a neutral amino acid transporter, was upregulated in the host cells after infection with L. pneumophila. Inhibition of SLC1A5 by BCH, a competitive inhibitor of amino acid uptake as well as siRNA silencing of the slc1a5 gene blocked intracellular multiplication of L. pneumophila without compromising viability of host cells. These observations suggest that replication of L. pneumophila depends on the function of host cell SLC1A5.


British Journal of Pharmacology | 2011

Fenamates as TRP channel blockers: mefenamic acid selectively blocks TRPM3.

Chihab Klose; Isabelle Straub; Marc Riehle; Felicia Ranta; Dietmar Krautwurst; Susanne Ullrich; Wolfgang Meyerhof; Christian Harteneck

BACKGROUND AND PURPOSE Fenamates are N‐phenyl‐substituted anthranilic acid derivatives clinically used as non‐steroid anti‐inflammatory drugs in pain treatment. Reports describing fenamates as tools to interfere with cellular volume regulation attracted our attention based on our interest in the role of the volume‐modulated transient receptor potential (TRP) channels TRPM3 and TRPV4.


Journal of Medicinal Chemistry | 2011

Identification of a potent and selective free fatty acid receptor 1 (FFA1/GPR40) agonist with favorable physicochemical and in vitro ADME properties.

Elisabeth Christiansen; Christian Urban; Manuel Grundmann; Maria E. Due-Hansen; Ellen Hagesaether; Johannes Schmidt; Leonardo Pardo; Susanne Ullrich; Evi Kostenis; Matthias U. Kassack; Trond Ulven

The free fatty acid receptor 1 (FFA1, also known as GPR40) enhances glucose-stimulated insulin secretion from pancreatic β-cells and is recognized as an interesting new target for treatment of type 2 diabetes. Several series of selective FFA1 agonists are already known. Most of these are derived from free fatty acids (FFAs) or glitazones and are relatively lipophilic. Aiming for the development of potent, selective, and less lipophilic FFA1 agonists, the terminal phenyl of a known compound series was replaced by nitrogen containing heterocycles. This resulted in the identification of 37, a selective FFA1 agonist with potent activity on recombinant human FFA1 receptors and on the rat insulinoma cell line INS-1E, optimal lipophilicity, and excellent in vitro permeability and metabolic stability.


FEBS Letters | 1989

Galanin inhibits insulin secretion by direct interference with exocytosis.

Susanne Ullrich; Claes B. Wollheim

Electrically permeabilized RINm5F cells were used to study whether galanin inhibits insulin secretion distally to the generation of soluble second messengers. Ca2+‐induced insulin secretion was inhibited by the neuropeptide in a dose‐dependent manner. Galanin appears to act via a G‐protein as persussis toxin treatment abolished the effect. GTP (100 μM), GDP (100 μM) and a low dose of GTPγS (10 μM) did not affect galanin‐mediated inhibition of secretion. In contrast, at 100 μM, GTPγS attenuated and GDPβS abolished the effect of the peptide. We conclude that galanin inhibits exocytosis directly by a mechanism involving a G‐protein.


Diabetes | 2010

Overexpression of Kinase-Negative Protein Kinase Cδ in Pancreatic β-Cells Protects Mice From Diet-Induced Glucose Intolerance and β-Cell Dysfunction

Anita M. Hennige; Felicia Ranta; Isabel Heinzelmann; Martina Düfer; Diana Michael; Heidi Braumüller; Stefan Z. Lutz; Reiner Lammers; Gisela Drews; Fatima Bosch; Hans-Ulrich Häring; Susanne Ullrich

OBJECTIVE In vitro models suggest that free fatty acid–induced apoptotic β-cell death is mediated through protein kinase C (PKC)δ. To examine the role of PKCδ signaling in vivo, transgenic mice overexpressing a kinase-negative PKCδ (PKCδKN) selectively in β-cells were generated and analyzed for glucose homeostasis and β-cell survival. RESEARCH DESIGN AND METHODS Mice were fed a standard or high-fat diet (HFD). Blood glucose and insulin levels were determined after glucose loads. Islet size, cleaved caspase-3, and PKCδ expression were estimated by immunohistochemistry. In isolated islet cells apoptosis was assessed with TUNEL/TO-PRO3 DNA staining and the mitochondrial potential by rhodamine-123 staining. Changes in phosphorylation and subcellular distribution of forkhead box class O1 (FOXO1) were analyzed by Western blotting and immunohistochemistry. RESULTS PKCδKN mice were protected from HFD-induced glucose intolerance. This was accompanied by increased insulin levels in vivo, by an increased islet size, and by a reduced staining of β-cells for cleaved caspase-3 compared with wild-type littermates. In accordance, long-term treatment with palmitate increased apoptotic cell death of isolated islet cells from wild-type but not from PKCδKN mice. PKCδKN overexpression protected islet cells from palmitate-induced mitochondrial dysfunction and inhibited nuclear accumulation of FOXO1 in mouse islet and INS-1E cells. The inhibition of nuclear accumulation of FOXO1 by PKCδKN was accompanied by an increased phosphorylation of FOXO1 at Ser256 and a significant reduction of FOXO1 protein. CONCLUSIONS Overexpression of PKCδKN in β-cells protects from HFD-induced β-cell failure in vivo by a mechanism that involves inhibition of fatty acid–mediated apoptosis, inhibition of mitochondrial dysfunction, and inhibition of FOXO1 activation.


Journal of Biological Chemistry | 2011

Conjugated linoleic acids mediate insulin release through islet G protein coupled receptor FFA1/GPR40

Johannes Schmidt; Kathrin Liebscher; Nicole Merten; Manuel Grundmann; M. Mielenz; H. Sauerwein; Elisabeth Christiansen; Maria E. Due-Hansen; Trond Ulven; Susanne Ullrich; Jesus Gomeza; Christel Drewke; Evi Kostenis

Among dietary components, conjugated linoleic acids (CLAs) have attracted considerable attention as weight loss supplements in the Western world because they reduce fat stores and increase muscle mass. However, a number of adverse effects are also ascribed to the intake of CLAs such as aggravation of insulin resistance and the risk of developing diabetes. However, the mechanisms accounting for the effects of CLAs on glucose homeostasis are incompletely understood. Herein we provide evidence that CLAs specifically activate the cell surface receptor FFA1, an emerging therapeutic target to treat type 2 diabetes. Using different recombinant cellular systems engineered to stably express FFA1 and a set of diverse functional assays including the novel, label-free non-invasive dynamic mass redistribution technology (Corning® Epic® biosensor), both CLA isomers cis-9, trans-11-CLA and trans-10, cis-12-CLA were found to activate FFA1 in vitro at concentrations sufficient to also account for FFA1 activation in vivo. Each CLA isomer markedly increased glucose-stimulated insulin secretion in insulin-producing INS-1E cells that endogenously express FFA1 and in primary pancreatic β-cells of wild type but not FFA1−/− knock-out mice. Our findings establish a clear mechanistic link between CLAs and insulin production and identify the cell surface receptor FFA1 as a molecular target for CLAs, explaining their acute stimulatory effects on insulin secretion in vivo. CLAs are also revealed as insulinotropic components in widely used nutraceuticals, a finding with significant implication for development of FFA1 modulators to treat type 2 diabetes.


ACS Medicinal Chemistry Letters | 2010

Structure−Activity Study of Dihydrocinnamic Acids and Discovery of the Potent FFA1 (GPR40) Agonist TUG-469

Elisabeth Christiansen; Maria E. Due-Hansen; Christian Urban; Nicole Merten; Michael Pfleiderer; Kasper K. Karlsen; Sanne S. Rasmussen; Mette Steensgaard; Alexandra Hamacher; Johannes Schmidt; Christel Drewke; Rasmus Koefoed Petersen; Karsten Kristiansen; Susanne Ullrich; Evi Kostenis; Matthias U. Kassack; Trond Ulven

The free fatty acid 1 receptor (FFA1 or GPR40), which is highly expressed on pancreatic β-cells and amplifies glucose-stimulated insulin secretion, has emerged as an attractive target for the treatment of type 2 diabetes. Several FFA1 agonists containing the para-substituted dihydrocinnamic acid moiety are known. We here present a structure-activity relationship study of this compound family suggesting that the central methyleneoxy linker is preferable for the smaller compounds, whereas the central methyleneamine linker gives higher potency to the larger compounds. The study resulted in the discovery of the potent and selective full FFA1 agonist TUG-469 (29).


Diabetes | 2013

Reevaluation of Fatty Acid Receptor 1 as a Drug Target for the Stimulation of Insulin Secretion in Humans

Robert Wagner; Gabriele Kaiser; Felicia Gerst; Elisabeth Christiansen; Maria E. Due-Hansen; Manuel Grundmann; Fausto Machicao; Andreas Peter; Evi Kostenis; Trond Ulven; Andreas Fritsche; Hans-Ulrich Häring; Susanne Ullrich

The role of free fatty acid receptor 1 (FFAR1/GPR40) in glucose homeostasis is still incompletely understood. Small receptor agonists stimulating insulin secretion are undergoing investigation for the treatment of type 2 diabetes. Surprisingly, genome-wide association studies did not discover diabetes risk variants in FFAR1. We reevaluated the role of FFAR1 in insulin secretion using a specific agonist, FFAR1-knockout mice and human islets. Nondiabetic individuals were metabolically phenotyped and genotyped. In vitro experiments indicated that palmitate and a specific FFAR1 agonist, TUG-469, stimulate glucose-induced insulin secretion through FFAR1. The proapoptotic effect of chronic exposure of β-cells to palmitate was independent of FFAR1. TUG-469 was protective, whereas inhibition of FFAR1 promoted apoptosis. In accordance with the proapoptotic effect of palmitate, in vivo cross-sectional observations demonstrated a negative association between fasting free fatty acids (NEFAs) and insulin secretion. Because NEFAs stimulate secretion through FFAR1, we examined the interaction of genetic variation in FFAR1 with NEFA and insulin secretion. The inverse association of NEFA and secretion was modulated by rs1573611 and became steeper for carriers of the minor allele. In conclusion, FFAR1 agonists support β-cell function, but variation in FFAR1 influences NEFA effects on insulin secretion and therefore could affect therapeutic efficacy of FFAR1 agonists.


ACS Medicinal Chemistry Letters | 2013

Discovery of TUG-770: A Highly Potent Free Fatty Acid Receptor 1 (FFA1/GPR40) Agonist for Treatment of Type 2 Diabetes

Elisabeth Christiansen; Steffen V. F. Hansen; Christian Urban; Brian D. Hudson; Edward T. Wargent; Manuel Grundmann; Laura Jenkins; Mohamed S. Zaibi; Claire J. Stocker; Susanne Ullrich; Evi Kostenis; Matthias U. Kassack; Graeme Milligan; Michael A. Cawthorne; Trond Ulven

Free fatty acid receptor 1 (FFA1 or GPR40) enhances glucose-stimulated insulin secretion from pancreatic β-cells and currently attracts high interest as a new target for the treatment of type 2 diabetes. We here report the discovery of a highly potent FFA1 agonist with favorable physicochemical and pharmacokinetic properties. The compound efficiently normalizes glucose tolerance in diet-induced obese mice, an effect that is fully sustained after 29 days of chronic dosing.

Collaboration


Dive into the Susanne Ullrich's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Florian Lang

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge