@article{AhmadWolberEckardtetal.2012, author = {Ahmad, Ruhel and Wolber, Wanja and Eckardt, Sigrid and Koch, Philipp and Schmitt, Jessica and Semechkin, Ruslan and Geis, Christian and Heckmann, Manfred and Br{\"u}stle, Oliver and McLaughlin, John K. and Sir{\´e}n, Anna-Leena and M{\"u}ller, Albrecht M.}, title = {Functional Neuronal Cells Generated by Human Parthenogenetic Stem Cells}, series = {PLoS One}, volume = {7}, journal = {PLoS One}, number = {8}, doi = {10.1371/journal.pone.0042800}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-130268}, pages = {e42800}, year = {2012}, abstract = {Parent of origin imprints on the genome have been implicated in the regulation of neural cell type differentiation. The ability of human parthenogenetic (PG) embryonic stem cells (hpESCs) to undergo neural lineage and cell type-specific differentiation is undefined. We determined the potential of hpESCs to differentiate into various neural subtypes. Concurrently, we examined DNA methylation and expression status of imprinted genes. Under culture conditions promoting neural differentiation, hpESC-derived neural stem cells (hpNSCs) gave rise to glia and neuron-like cells that expressed subtype-specific markers and generated action potentials. Analysis of imprinting in hpESCs and in hpNSCs revealed that maternal-specific gene expression patterns and imprinting marks were generally maintained in PG cells upon differentiation. Our results demonstrate that despite the lack of a paternal genome, hpESCs generate proliferating NSCs that are capable of differentiation into physiologically functional neuron-like cells and maintain allele-specific expression of imprinted genes. Thus, hpESCs can serve as a model to study the role of maternal and paternal genomes in neural development and to better understand imprinting-associated brain diseases.}, language = {en} } @article{BavendiekBerlinerAguirreDavilaetal.2019, author = {Bavendiek, Udo and Berliner, Dominik and Aguirre D{\´a}vila, Lukas and Schwab, Johannes and Maier, Lars and Philipp, Sebastian A. and Rieth, Andreas and Westenfeld, Ralf and Piorkowski, Christopher and Weber, Kristina and H{\"a}nselmann, Anja and Oldhafer, Maximiliane and Schallhorn, Sven and von der Leyen, Heiko and Schr{\"o}der, Christoph and Veltmann, Christian and St{\"o}rk, Stefan and B{\"o}hm, Michael and Koch, Armin and Bauersachs, Johann}, title = {Rationale and design of the DIGIT-HF trial (DIGitoxin to Improve ouTcomes in patients with advanced chronic Heart Failure): a randomized, double-blind, placebo-controlled study}, series = {European Journal of Heart Failure}, volume = {21}, journal = {European Journal of Heart Failure}, organization = {DIGIT-HF Investigators and Committees}, doi = {10.1002/ejhf.1452}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-221548}, pages = {676-684}, year = {2019}, abstract = {Aims Despite recent advances in the treatment of chronic heart failure (HF), mortality and hospitalizations still remain high. Additional therapies to improve mortality and morbidity are urgently needed. The efficacy of cardiac glycosides - although regularly used for HF treatment - remains unclear. DIGIT-HF was designed to demonstrate that digitoxin on top of standard of care treatment improves mortality and morbidity in patients with HF and a reduced ejection fraction (HFrEF). Methods Patients with chronic HF, New York Heart Association (NYHA) functional class III-IV and left ventricular ejection fraction (LVEF) ≤ 40\%, or patients in NYHA functional class II and LVEF ≤ 30\% are randomized 1:1 in a double-blind fashion to treatment with digitoxin (target serum concentration 8-18 ng/mL) or matching placebo. Randomization is stratified by centre, sex, NYHA functional class (II, III, or IV), atrial fibrillation, and treatment with cardiac glycosides at baseline. A total of 2190 eligible patients will be included in this clinical trial (1095 per group). All patients receive standard of care treatment recommended by expert guidelines upon discretion of the treating physician. The primary outcome is a composite of all-cause mortality or hospital admission for worsening HF (whatever occurs first). Key secondary endpoints are all-cause mortality, hospital admission for worsening HF, and recurrent hospital admission for worsening HF. Conclusion The DIGIT-HF trial will provide important evidence, whether the cardiac glycoside digitoxin reduces the risk for all-cause mortality and/or hospital admission for worsening HF in patients with advanced chronic HFrEF on top of standard of care treatment.}, language = {en} }