Network


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

Hotspot


Dive into the research topics where Katharina Drews is active.

Publication


Featured researches published by Katharina Drews.


Stem Cells | 2014

HIF1α Modulates Cell Fate Reprogramming Through Early Glycolytic Shift and Upregulation of PDK1–3 and PKM2

Alessandro Prigione; Nadine Rohwer; Sheila Hoffmann; Barbara Mlody; Katharina Drews; Raul Bukowiecki; Katharina Blümlein; Erich E. Wanker; Markus Ralser; Thorsten Cramer; James Adjaye

Reprogramming somatic cells to a pluripotent state drastically reconfigures the cellular anabolic requirements, thus potentially inducing cancer‐like metabolic transformation. Accordingly, we and others previously showed that somatic mitochondria and bioenergetics are extensively remodeled upon derivation of induced pluripotent stem cells (iPSCs), as the cells transit from oxidative to glycolytic metabolism. In the attempt to identify possible regulatory mechanisms underlying this metabolic restructuring, we investigated the contributing role of hypoxia‐inducible factor one alpha (HIF1α), a master regulator of energy metabolism, in the induction and maintenance of pluripotency. We discovered that the ablation of HIF1α function in dermal fibroblasts dramatically hampers reprogramming efficiency, while small molecule‐based activation of HIF1α significantly improves cell fate conversion. Transcriptional and bioenergetic analysis during reprogramming initiation indicated that the transduction of the four factors is sufficient to upregulate the HIF1α target pyruvate dehydrogenase kinase (PDK) one and set in motion the glycolytic shift. However, additional HIF1α activation appears critical in the early upregulation of other HIF1α‐associated metabolic regulators, including PDK3 and pyruvate kinase (PK) isoform M2 (PKM2), resulting in increased glycolysis and enhanced reprogramming. Accordingly, elevated levels of PDK1, PDK3, and PKM2 and reduced PK activity could be observed in iPSCs and human embryonic stem cells in the undifferentiated state. Overall, the findings suggest that the early induction of HIF1α targets may be instrumental in iPSC derivation via the activation of a glycolytic program. These findings implicate the HIF1α pathway as an enabling regulator of cellular reprogramming. Stem Cells 2014;32:364–376


Molecular Therapy | 2012

Valproic Acid Confers Functional Pluripotency to Human Amniotic Fluid Stem Cells in a Transgene-free Approach

Dafni Moschidou; Sayandip Mukherjee; Michael P. Blundell; Katharina Drews; Gemma N. Jones; Hassan Abdulrazzak; Beata Nowakowska; Anju Phoolchund; Kenneth Lay; T Selvee Ramasamy; Mara Cananzi; Daniel Nettersheim; M.H.F. Sullivan; Jennifer M. Frost; Gudrun E. Moore; Joris Vermeesch; Nicholas M. Fisk; Adrian J. Thrasher; Anthony Atala; James Adjaye; Hubert Schorle; Paolo De Coppi; Pascale V. Guillot

Induced pluripotent stem cells (iPSCs) with potential for therapeutic applications can be derived from somatic cells via ectopic expression of a set of limited and defined transcription factors. However, due to risks of random integration of the reprogramming transgenes into the host genome, the low efficiency of the process, and the potential risk of virally induced tumorigenicity, alternative methods have been developed to generate pluripotent cells using nonintegrating systems, albeit with limited success. Here, we show that c-KIT+ human first-trimester amniotic fluid stem cells (AFSCs) can be fully reprogrammed to pluripotency without ectopic factors, by culture on Matrigel in human embryonic stem cell (hESC) medium supplemented with the histone deacetylase inhibitor (HDACi) valproic acid (VPA). The cells share 82% transcriptome identity with hESCs and are capable of forming embryoid bodies (EBs) in vitro and teratomas in vivo. After long-term expansion, they maintain genetic stability, protein level expression of key pluripotency factors, high cell-division kinetics, telomerase activity, repression of X-inactivation, and capacity to differentiate into lineages of the three germ layers, such as definitive endoderm, hepatocytes, bone, fat, cartilage, neurons, and oligodendrocytes. We conclude that AFSC can be utilized for cell banking of patient-specific pluripotent cells for potential applications in allogeneic cellular replacement therapies, pharmaceutical screening, and disease modeling.


Journal of Visualized Experiments | 2012

Preparation of Mouse Embryonic Fibroblast Cells Suitable for Culturing Human Embryonic and Induced Pluripotent Stem Cells

Justyna Jozefczuk; Katharina Drews; James Adjaye

In general, human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs)1 can be cultured under variable conditions. However, it is not easy to establish an effective system for culturing these cells. Since the culture conditions can influence gene expression that confers pluripotency in hESCs and hiPSCs, the optimization and standardization of the culture method is crucial. The establishment of hESC lines was first described by using MEFs as feeder cells and fetal bovine serum (FBS)-containing culture medium2. Next, FBS was replaced with knockout serum replacement (KSR) and FGF2, which enhances proliferation of hESCs3. Finally, feeder-free culture systems enable culturing cells on Matrigel-coated plates in KSR-containing conditioned medium (medium conditioned by MEFs)4. Subsequently, hESCs culture conditions have moved towards feeder-free culture in chemically defined conditions5-7. Moreover, to avoid the potential contamination by pathogens and animal proteins culture methods using xeno-free components have been established8. To obtain improved conditions mouse feeder cells have been replaced with human cell lines (e.g. fetal muscle and skin cells9, adult skin cells10, foreskin fibroblasts11-12, amniotic mesenchymal cells13). However, the efficiency of maintaining undifferentiated hESCs using human foreskin fibroblast-derived feeder layers is not as high as that from mouse feeder cells due to the lower level of secretion of Activin A14. Obviously, there is an evident difference in growth factor production by mouse and human feeder cells. Analyses of the transcriptomes of mouse and human feeder cells revealed significant differences between supportive and non-supportive cells. Exogenous FGF2 is crucial for maintaining self-renewal of hESCs and hiPSCs, and has been identified as a key factor regulating the expression of Tgfβ1, Activin A and Gremlin (a BMP antagonist) in feeder cells. Activin A has been shown to induce the expression of OCT4, SOX2, and NANOG in hESCs15-16. For long-term culture, hESCs and hiPSCs can be grown on mitotically inactivated MEFs or under feeder-free conditions in MEF-CM (MEF-Conditioned Medium) on Matrigel-coated plates to maintain their undifferentiated state. Success of both culture conditions fully depends on the quality of the feeder cells, since they directly affect the growth of hESCs. Here, we present an optimized method for the isolation and culture of mouse embryonic fibroblasts (MEFs), preparation of conditioned medium (CM) and enzyme-linked immunosorbent assay (ELISA) to assess the levels of Activin A within the media.


Current Stem Cell Research & Therapy | 2013

Molecular Signature of Human amniotic Fluid Stem Cells During Fetal Development

Dafni Moschidou; Katharina Drews; Ayad Eddaoudi; James Adjaye; Paolo De Coppi; Pascale V. Guillot

Mid-gestation c-KIT(+) amniotic fluid stem cells (AFSC) have an intermediate phenotype between embryonic and adult stem cells and are easy to reprogram to pluripotency. We previously showed that 1st trimester AFSC can be reprogrammed to functional pluripotency in a transgene-free approach. Despite both parental populations sharing a common phenotype, expressing CD29, CD44, CD73, CD90, CD105, SSEA4 and OCT4, 2nd trimester AFSC, contrary to 1st trimester cells, do not express NANOG, SSEA3, TRA-1-60 and TRA-1-81, and have slower growth kinetics. Here, we used the Illumina Beadstudio microarray platform to analyse the transcriptome of 1st and 2nd trimester AFSC and show a unique 1st trimester AFSC-specific gene expression signature consisting of 366 genes and a larger set of 603 genes common with hESC compared to 496 genes overlapping between 2nd trimester AFSC and hESC. We conclude that both populations are related but distinct to each other as well as to hESC.


Scientific Data | 2015

Multi-omic profiles of human non-alcoholic fatty liver disease tissue highlight heterogenic phenotypes

Wasco Wruck; Karl Kashofer; Samrina Rehman; Andriani Daskalaki; Daniela Berg; Ewa Gralka; Justyna Jozefczuk; Katharina Drews; Vikash Pandey; Christian R. A. Regenbrecht; Christoph Wierling; Paola Turano; Ulrike Korf; Kurt Zatloukal; Hans Lehrach; Hans V. Westerhoff; James Adjaye

Non-alcoholic fatty liver disease (NAFLD) is a consequence of sedentary life style and high fat diets with an estimated prevalence of about 30% in western countries. It is associated with insulin resistance, obesity, glucose intolerance and drug toxicity. Additionally, polymorphisms within, e.g., APOC3, PNPLA3, NCAN, TM6SF2 and PPP1R3B, correlate with NAFLD. Several studies have already investigated later stages of the disease. This study explores the early steatosis stage of NAFLD with the aim of identifying molecular mechanisms underlying the etiology of NAFLD. We analyzed liver biopsies and serum samples from patients with high- and low-grade steatosis (also pre-disease states) employing transcriptomics, ELISA-based serum protein analyses and metabolomics. Here, we provide a detailed description of the various related datasets produced in the course of this study. These datasets may help other researchers find new clues for the etiology of NAFLD and the mechanisms underlying its progression to more severe disease states.


The International Journal of Developmental Biology | 2012

Comparative molecular portraits of human unfertilized oocytes and primordial germ cells at 10 weeks of gestation.

Ferdinand Diedrichs; Barbara Mlody; Peggy Matz; Heiko Fuchs; Lukas Chavez; Katharina Drews; James Adjaye

Primordial germ cells (PGCs) are precursors of gametes and share several features in common with pluripotent stem cells, such as alkaline phosphatase activity and the expression of pluripotency-associated genes such as OCT4 and NANOG. PGCs are able to differentiate into oocytes and spermatogonia and establish totipotency after fertilization. However, our knowledge of human germ cell development is still fragmentary. In this study, we have carried out genome-wide comparisons of the transcriptomes and molecular portraits of human male PGCs (mPGCs), female PGCs (fPGCs) and unfertilized oocytes. We detected 9210 genes showing elevated expression in fPGCs, 9184 in mPGCs and 9207 in oocytes, with 6342 of these expressed in common. As well as known germ cell-related genes such as BLIMP1/PRDM1, PIWIL2, VASA/DDX4, DAZL, STELLA/DPPA3 and LIN28, we also identified 465 novel non-annotated genes with orthologs in the mouse. A plethora of olfactory receptor-encoding genes were detected in all samples, which would suggest their involvement not only in sperm chemotaxis, but also in the development of female germ cells and oocytes. We anticipate that our data might increase our meagre knowledge of the genes and associated signaling pathways operative during germ cell development. This in turn might aid in the development of strategies enabling better differentiation and molecular characterisation of germ cells derived from either embryonic or induced pluripotent stem cells. Ultimately, this would have a profound relevance for reproductive as well as regenerative medicine.


Stem Cell Research | 2015

Generation of iPSC lines from primary human amniotic fluid cells.

Katharina Drews; Peggy Matz; James Adjaye

By means of retroviral transduction using the four Yamanaka-factors OCT4, SOX2, KLF4 and c-MYC primary human amniotic fluid cells (AFCs) were reprogrammed into several iPSC lines. Pluripotency was confirmed both in vitro and in vivo. A comparative transcriptome analysis of the AF-derived iPSC line 41 and the human embryonic stem cell lines (H1 and H9) revealed a Pearson correlation of 0.953 and 0.941 respectively.


Stem Cell Research & Therapy | 2015

Association between in vivo bone formation and ex vivo migratory capacity of human bone marrow stromal cells

Rikke K. Andersen; Walid Zaher; Kenneth H. Larsen; Nicholas Ditzel; Katharina Drews; Wasco Wruck; James Adjaye; Basem Abdallah; Moustapha Kassem


Archive | 2016

Classifying distinct grades of human non-alcoholic fatty liver disease employing a systems biology approach

Wasco Wruck; Karl Kashofer; Samrina Rehman; Andriani Daskalaki; Daniela Berg; Ewa Gralka; Justyna Jozefczuk; Katharina Drews; Vikash Pandey; Christian R. A. Regenbrecht; Christoph Wierling; Paola Turano; Ulrike Korf; Kurt Zatloukal; Hans Lehrach; Hans V. Westerhoff; James Adjaye


Archive | 2012

Viral and non-viral generation and characterization of induced pluripotent stem cells from human amniotic fluid cells

Katharina Drews; Hans Lehrach; Petra Knaus

Collaboration


Dive into the Katharina Drews's collaboration.

Top Co-Authors

Avatar

James Adjaye

University of Düsseldorf

View shared research outputs
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

Peggy Matz

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Ulrike Korf

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge