Network


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

Hotspot


Dive into the research topics where Teresa F. Ackermann is active.

Publication


Featured researches published by Teresa F. Ackermann.


Nature Medicine | 2013

Acid sphingomyelinase–ceramide system mediates effects of antidepressant drugs

Erich Gulbins; Monica Palmada; Martin Reichel; Anja Lüth; Christoph Böhmer; Davide Amato; Christian P. Müller; Carsten H. Tischbirek; Teja W. Groemer; Ghazaleh Tabatabai; Katrin Anne Becker; Philipp Tripal; Sven Staedtler; Teresa F. Ackermann; Johannes van Brederode; Christian Alzheimer; Michael Weller; Undine E. Lang; Burkhard Kleuser; Heike Grassmé; Johannes Kornhuber

Major depression is a highly prevalent severe mood disorder that is treated with antidepressants. The molecular targets of antidepressants require definition. We investigated the role of the acid sphingomyelinase (Asm)-ceramide system as a target for antidepressants. Therapeutic concentrations of the antidepressants amitriptyline and fluoxetine reduced Asm activity and ceramide concentrations in the hippocampus, increased neuronal proliferation, maturation and survival and improved behavior in mouse models of stress-induced depression. Genetic Asm deficiency abrogated these effects. Mice overexpressing Asm, heterozygous for acid ceramidase, treated with blockers of ceramide metabolism or directly injected with C16 ceramide in the hippocampus had higher ceramide concentrations and lower rates of neuronal proliferation, maturation and survival compared with controls and showed depression-like behavior even in the absence of stress. The decrease of ceramide abundance achieved by antidepressant-mediated inhibition of Asm normalized these effects. Lowering ceramide abundance may thus be a central goal for the future development of antidepressants.


Cellular Physiology and Biochemistry | 2011

EMD638683, a novel SGK inhibitor with antihypertensive potency.

Teresa F. Ackermann; Krishna M. Boini; Norbert Beier; Wolfgang Scholz; Thomas Fuchß; Florian Lang

The serum- and glucocorticoid-inducible kinase 1 (SGK1) is transcriptionally upregulated by mineralocorticoids and activated by insulin. The kinase enhances renal tubular Na+-reabsorption and accounts for blood pressure increase following high salt diet in mice made hyperinsulinemic by dietary fructose or fat. The present study describes the in vitro and in vivo efficacy of a novel SGK1 inhibitor (EMD638683). EMD638683 was tested in vitro by determination of SGK1-dependent phosphorylation of NDRG1 (N-Myc downstream-regulated gene 1) in human cervical carcinoma HeLa-cells. In vivo EMD638683 (4460 ppm in chow, i.e. approx. 600 mg/kg/day) was administered to mice drinking tap water or isotonic saline containing 10% fructose. Blood pressure was determined by the tail cuff method, and urinary electrolyte (flame photometry) concentrations determined in metabolic cages. In vitro testing disclosed EMD638683 as a SGK1 inhibitor with an IC50 of 3 µM. Within 24 hours in vivo EMD638683 treatment significantly decreased blood pressure in fructose/saline-treated mice but not in control animals or in SGK1 knockout mice. EMD638683 failed to alter the blood pressure in SGK1 knockout mice. Following chronic (4 weeks) fructose/high salt treatment, additional EMD638683 treatment again decreased blood pressure. EMD638683 thus abrogates the salt sensitivity of blood pressure in hyperinsulinism without appreciably affecting blood pressure in the absence of hyperinsulinism. EMD638683 tended to increase fluid intake and urinary excretion of Na+, significantly increased urinary flow rate and significantly decreased body weight. Conclusion: EMD638683 could serve as a template for drugs counteracting hypertension in individuals with type II diabetes and metabolic syndrome.


American Journal of Physiology-renal Physiology | 2010

Hyperaldosteronism in Klotho-deficient mice

Stephanie S. Fischer; Daniela S. Kempe; Christina Leibrock; Rexhep Rexhepaj; Balasaheb Siraskar; Krishna M. Boini; Teresa F. Ackermann; Michael Föller; Berthold Hocher; Kevin P. Rosenblatt; Makoto Kuro-o; Florian Lang

Klotho is a membrane protein participating in the inhibitory effect of FGF23 on the formation of 1,25-dihydroxyvitamin-D(3) [1,25(OH)(2)D(3)]. It participates in the regulation of renal tubular phosphate reabsorption and stimulates renal tubular Ca(2+) reabsorption. Klotho hypomorphic mice (klotho(hm)) suffer from severe growth deficit, rapid aging, and early death, events largely reversed by a vitamin D-deficient diet. The present study explored the role of Klotho deficiency in mineral and electrolyte metabolism. To this end, klotho(hm) mice and wild-type mice (klotho(+/+)) were subjected to a normal (D(+)) or vitamin D-deficient (D(-)) diet or to a vitamin D-deficient diet for 4 wk and then to a normal diet (D(-/+)). At the age of 8 wk, body weight was significantly lower in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice, klotho(hm)D(-) mice, and klotho(hm)D(-/+) mice. Plasma concentrations of 1,25(OH)(2)D(3,) adrenocorticotropic hormone (ACTH), antidiuretic hormone (ADH), and aldosterone were significantly higher in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice. Plasma volume was significantly smaller in klotho(hm)D(-/+) mice, and plasma urea, Ca(2+), phosphate and Na(+), but not K(+) concentrations were significantly higher in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice. The differences were partially abrogated by a vitamin D-deficient diet. Moreover, the hyperaldosteronism was partially reversed by Ca(2+)-deficient diet. Ussing chamber experiments revealed a marked increase in amiloride-sensitive current across the colonic epithelium, pointing to enhanced epithelial sodium channel (ENaC) activity. A salt-deficient diet tended to decrease and a salt-rich diet significantly increased the life span of klotho(hm)D(+) mice. In conclusion, the present observation disclose that the excessive formation of 1,25(OH)(2)D(3) in Klotho-deficient mice results in extracellular volume depletion, which significantly contributes to the shortening of life span.


Cellular Physiology and Biochemistry | 2010

Hyperactivity and enhanced curiosity of mice expressing PKB/SGK-resistant glycogen synthase kinase-3 (GSK-3).

Teresa F. Ackermann; Daniela S. Kempe; Florian Lang; Undine E. Lang

Background: Mounting evidence suggests that bipolar disorder symptoms could be favorably influenced by modification of glycogen synthase kinase-3 (GSK-3) activity. Specifically, the well known antimanic and mood stabilizing medications lithium, valproate, olanzapine and clozapine have been shown to inhibit GSK-3 activity. GSK-3 is phosphorylated and thus inhibited by protein kinase B (PKB/Akt) and serum and glucocorticoid inducible kinase (SGK) isoforms. The present study explored, whether PKB/SGK-dependent GSK-3 regulation influences the behavior of mice. Methods: Gene-targeted knockin mice with mutated and thus PKB/SGK-resistant GSK-3Α, Β (gsk-3KI) were compared to corresponding wild type mice (gsk-3WT). The mice were analyzed by open-field, light-dark (LD-) box, O-maze, emergence test, object exploration test and forced swimming test (FST). Results: In open-field, LD-box and O-maze, gsk-3KI mice displayed a hyperactive and more curious phenotype when compared to wild type mice. Speed and total distance moved as well as rearings were significantly increased in gsk-3KI compared to gsk-3WT mice. In the O-maze, gsk-3KI mice tended to travel a larger distance in the open, unprotected area thangsk-3WT mice, and performed significantly more unprotected head dips suggesting decreased anxiety behavior. In the forced swimming test, the immobility time was significantly decreased in gsk-3KI mice indicating a phenotype less prone to depression. Moreover, gsk-3KImice were less sensitive to the application of chronic mild stress and showed a decreased HPA axis activity. Conclusions: The present observations disclose a significant role of PKB/SGK-dependent regulation of GSK-3 in the control of activity, anxiety and proneness to depression. Accordingly, mice expressing SGK/PKB resistant GSK-3 may be a valuable model of hyperactivity and mania.


Cellular Physiology and Biochemistry | 2008

Phosphatidylinositide Dependent Kinase Deficiency Increases Anxiety and Decreases GABA and Serotonin Abundance in the Amygdala

Teresa F. Ackermann; Heide Hörtnagl; David P. Wolfer; Giovanni Colacicco; Reinhard Sohr; Florian Lang; Rainer Hellweg; Undine E. Lang

Pathological anxiety is paralleled by deficits in serotonergic and GABAergic neurotransmission in the amygdala. Conversely, anxiety disorders and depression may be reversed by brain-derived neurotrophic factor (BDNF). BDNF signaling involves Phosphatidylinositol 3-Kinase / 3-phosphoinositide-dependent protein kinase 1 (PI3K/PDK1). We thus hypothesized that impaired function of PDK1 might be associated with increased anxiety and concomitant neurotransmitter changes. Here we used the hypomorphic PDK1hm mouse to investigate anxiety behavior in different settings: PDK1hm mice differed from Wt littermates PDK1WT in several behavioral measures related to anxiety and exploration, namely in the open field, dark-light box, O-maze and startle response. Further we analyzed the brain substrate underlying this phenotype and found significantly decreased GABA, taurine and serotonin concentrations in the amygdala and olfactory bulb of PDK1hm mice, while BDNF and nerve growth factor (NGF) concentrations were not significantly different between PDK1hm and PDK1WT mice. These results suggest that impaired PI3K signaling in the PDK1hm mouse reduces concentrations of GABA and serotonin in anxiety related brain regions and can serve as a molecular substrate for behavior indicative for anxious and depressive-like mood states.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2009

Enhanced insulin sensitivity of gene targeted mice lacking functional KCNQ1

Krishna M. Boini; Dirk Graf; Anita M. Hennige; Saisudha Koka; Daniela S. Kempe; Kan Wang; Teresa F. Ackermann; Michael Föller; Volker Vallon; Karl Pfeifer; Erwin Schleicher; Susanne Ullrich; Hans-Ulrich Häring; Dieter Häussinger; Florian Lang

The pore-forming K+-channel alpha-subunit KCNQ1 is expressed in a wide variety of tissues including heart, skeletal muscle, liver, and epithelia. Most recent evidence revealed an association of the KCNQ1 gene with the susceptibility to type 2 diabetes. KCNQ1 participates in the regulation of cell volume, which is, in turn, critically important for the regulation of metabolism by insulin. The present study explored the influence of KCNQ1 on insulin-induced cellular K+ uptake and glucose metabolism. Insulin (100 nM)-induced K+ uptake was determined in isolated perfused livers from KCNQ1-deficient mice (kcnq1(-/-)) and their wild-type littermates (kcnq1(+/+)). Moreover, plasma glucose and insulin levels, intraperitoneal glucose (3 g/kg) tolerance, insulin (0.15 U/kg)-induced hypoglycemia, and peripheral uptake of radiolabeled 3H-deoxy-glucose were determined in both genotypes. Insulin-stimulated hepatocellular K+ uptake was significantly more sustained in isolated perfused livers from kcnq1(-/-) mice than from kcnq1(+/+)mice. The decline of plasma glucose concentration following an intraperitoneal injection of insulin was again significantly more sustained in kcnq1(-/-) than in kcnq1(+/+) mice. Both fasted and nonfasted plasma glucose and insulin concentrations were significantly lower in kcnq1(-/-) than in kcnq1(+/+)mice. Following an intraperitoneal glucose injection, the peak plasma glucose concentration was significantly lower in kcnq1(-/-) than in kcnq1(+/+)mice. Uptake of 3H-deoxy-glucose into skeletal muscle, liver, kidney and lung tissue was significantly higher in kcnq1(-/-) than in kcnq1(+/+)mice. In conclusion, KCNQ1 counteracts the stimulation of cellular K+ uptake by insulin and thereby influences K+-dependent insulin signaling on glucose metabolism. The observations indicate that KCNQ1 is a novel molecule affecting insulin sensitivity of glucose metabolism.


Iubmb Life | 2009

Relative resistance of SGK1 knockout mice against chemical carcinogenesis

Kan Wang; Michael Föller; Shuchen Gu; Madhuri Bhandaru; Teresa F. Ackermann; Krishna M. Boini; Andreas F. Mack; Karin Klingel; Rosario Amato; Nicola Perrotti; Dietmar Kuhl; Jürgen Behrens; Christos Stournaras; Florian Lang

The serum and glucocorticoid inducible kinase SGK1 was originally cloned from mammary tumor cells. SGK1 was found to be up‐regulated in a variety of tumors, but down‐regulated in several distinct tumors. Thus, evidence for a role of SGK1 in tumor growth remained conflicting. According to in vitro observations, SGK1 is up‐regulated by the oncogene β‐catenin and negatively regulates the proapoptotic transcription factor FOXO3a, which in turn stimulates transcription of the Bcl2‐interacting mediator BIM. This study aimed to define the role of SGK1 in colon carcinoma in vivo. SGK1 knockout mice (sgk1−/−) and their wild type littermates (sgk1+/+) were subjected to chemical cancerogenesis (intraperitoneal injection of 20 mg/kg 1,2‐dimethylhydrazine followed by three cycles of 30 g/L synthetic dextran sulfate sodium for 7 days). Moreover, SGK1 was silenced in HEK293 cells. FOXO3a and BIM protein abundance was determined by Western blotting and immunohistochemistry. Following chemical cancerogenesis, sgk1−/−mice developed significantly less colonic tumors than sgk1+/+mice. According to Western blotting and immunohistochemistry, SGK1 deficiency enhanced the expression of FOXO3a and BIM both, in vitro and in vivo. SGK1 deficiency counteracts the development of colonic tumors, an effect at least in part due to up‐regulation of FOXO3a and BIM.


Cellular Physiology and Biochemistry | 2013

Inhibition of Colonic Tumor Growth by the Selective SGK Inhibitor EMD638683

Syeda T. Towhid; Guilai Liu; Teresa F. Ackermann; Norbert Beier; Wolfgang Scholz; Thomas Fuchß; Mahmoud Toulany; Hans-Peter Rodemann; Florian Lang

Background: The serum and glucocorticoid inducible kinase SGK1, which was originally cloned from mammary tumor cells, is highly expressed in some but not all tumors. SGK1 confers survival to several tumor cells. Along those lines, the number of colonic tumors following chemical carcinogenesis was decreased in SGK1 knockout mice. Recently, a highly selective SGK inhibitor (EMD638683) has been developed. The present study explored whether EMD638683 affects survival of colon carcinoma cells in vitro and impacts on development of colonic tumors in vivo. Methods: Colon carcinoma (Caco-2) cells were exposed to EMD638683 with or without exposure to radiation (3 Gray) and cell volume was estimated from forward scatter, phosphatidylserine exposure from annexin V binding, mitochondrial potential from JC-9 fluorescence, caspase 3 activity from CaspGlow Fluorescein staining, DNA degradation from propidium iodide staining as well as late apoptosis from annexin-V FITC and propidium iodide double staining. In vivo tumor growth was determined in wild type mice subjected to chemical carcinogenesis (intraperitoneal injection of 20 mg/kg 1,2-dimethylhydrazine followed by three cycles of 30 g/L synthetic dextran sulfate sodium in drinking water for 7 days). Results: EMD638683 treatment significantly augmented the radiation-induced decrease of forward scatter, increase of phosphatidylserine exposure, decrease of mitochondrial potential, increase of caspase 3 activity, increase of DNA fragmentation and increase of late apoptosis. The in vivo development of tumors following chemical carcinogenesis was significantly blunted by treatment with EMD638683. Conclusions: EMD638683 promotes radiation-induced suicidal death of colon tumor cells in vitro and decreases the number of colonic tumors following chemical carcinogenesis in vivo.


Scientific Reports | 2015

Accelerated apoptotic death and in vivo turnover of erythrocytes in mice lacking functional mitogen- and stress-activated kinase MSK1/2

Elisabeth Lang; Rosi Bissinger; Abul Fajol; Madhuri S. Salker; Yogesh Singh; Christine Zelenak; Mehrdad Ghashghaeinia; Shuchen Gu; Kashif Jilani; Adrian Lupescu; Kathleen M. S. E. Reyskens; Teresa F. Ackermann; Michael Föller; Erwin Schleicher; William P. Sheffield; J. Simon C. Arthur; Florian Lang; Syed M. Qadri

The mitogen- and stress-activated kinase MSK1/2 plays a decisive role in apoptosis. In analogy to apoptosis of nucleated cells, suicidal erythrocyte death called eryptosis is characterized by cell shrinkage and cell membrane scrambling leading to phosphatidylserine (PS) externalization. Here, we explored whether MSK1/2 participates in the regulation of eryptosis. To this end, erythrocytes were isolated from mice lacking functional MSK1/2 (msk−/−) and corresponding wild-type mice (msk+/+). Blood count, hematocrit, hemoglobin concentration and mean erythrocyte volume were similar in both msk−/− and msk+/+ mice, but reticulocyte count was significantly increased in msk−/− mice. Cell membrane PS exposure was similar in untreated msk−/− and msk+/+ erythrocytes, but was enhanced by pathophysiological cell stressors ex vivo such as hyperosmotic shock or energy depletion to significantly higher levels in msk−/− erythrocytes than in msk+/+ erythrocytes. Cell shrinkage following hyperosmotic shock and energy depletion, as well as hemolysis following decrease of extracellular osmolarity was more pronounced in msk−/− erythrocytes. The in vivo clearance of autologously-infused CFSE-labeled erythrocytes from circulating blood was faster in msk−/− mice. The spleens from msk−/− mice contained a significantly greater number of PS-exposing erythrocytes than spleens from msk+/+ mice. The present observations point to accelerated eryptosis and subsequent clearance of erythrocytes leading to enhanced erythrocyte turnover in MSK1/2-deficient mice.


Cellular Physiology and Biochemistry | 2009

APC sensitive gastric acid secretion.

Anand Rotte; Madhuri Bhandaru; Michael Föller; Raja Biswas; Andreas F. Mack; Björn Friedrich; Rexhep Rexhepaj; Teresa F. Ackermann; Krishna M. Boini; Karl Kunzelmann; Jürgen Behrens; Florian Lang

Adenomatous polyposis coli (APC) is a tumor suppressor gene inactivated in familial adenomatous polyposis and sporadic colorectal cancer. Mice carrying a loss-of-function mutation in the apc gene (apc<sup>Min/+</sup>) spontaneously develop gastrointestinal tumors. APC fosters degradation of β-catenin, which in turn upregulates the serum- and glucocorticoid-inducible kinase SGK1. SGK1 stimulates KCNQ1, which is required for luminal K<sup>+</sup> recycling and thus for gastric acid secretion. BCECF-fluorescence was utilized to determine gastric acid secretion in isolated gastric glands from apc<sup>Min/+</sup> mice and their wild type littermates (apc<sup>+/+</sup>). Western blotting was employed to analyse β-catenin and SGK1 expression and immunohistochemistry to determine KCNQ1 protein abundance. β-catenin and SGK1 expression were enhanced in apc<sup>Min/+</sup> mice. Cytosolic pH was similar in apc<sup>Min/+</sup> mice and apc<sup>+/+</sup> mice. Na<sup>+</sup>-independent pH recovery following an ammonium pulse (ΔpH/min), which reflects H<sup>+</sup>/K<sup>+</sup> ATPase activity, was, however, significantly faster in apc<sup>Min/+</sup> mice than in apc<sup>+/+</sup>mice. In both genotypes ΔpH/min was abolished in the presence of H<sup>+</sup>/K<sup>+</sup> ATPase inhibitor omeprazole (100 μM). Treatment of apc<sup>Min/+</sup> and apc<sup>+/+</sup>mice with 5 μM forskolin 15 minutes prior to the experiment or increase in local K<sup>+</sup>-concentrations to 35 mM (replacing Na<sup>+</sup>/NMDG) significantly increased ΔpH/min and abrogated the differences between genotypes. The increase of ΔpH/min in apc<sup>Min/+</sup>mice required SGK1, as it was abolished by additional knockout of SGK1 (apc<sup>Min/+</sup>/sgk1<sup>-/-</sup>). In conclusion, basal gastric acid secretion is significantly enhanced in apc<sup>Min/+</sup>mice, pointing to a role of APC in the regulation of gastric acid secretion. The effect of APC requires H<sup>+</sup>/K<sup>+</sup> ATPase activity and is at least partially due to SGK1-dependent upregulation of KCNQ1.

Collaboration


Dive into the Teresa F. Ackermann's collaboration.

Top Co-Authors

Avatar

Florian Lang

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Krishna M. Boini

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kan Wang

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shuchen Gu

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar

Anand Rotte

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge