Network


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

Hotspot


Dive into the research topics where Kerstin Otte is active.

Publication


Featured researches published by Kerstin Otte.


Biotechnology Advances | 2015

The art of CHO cell engineering: A comprehensive retrospect and future perspectives

Simon Fischer; René Handrick; Kerstin Otte

Chinese hamster ovary (CHO) cells represent the most frequently applied host cell system for industrial manufacturing of recombinant protein therapeutics. CHO cells are capable of producing high quality biologics exhibiting human-like post-translational modifications in gram quantities. However, production processes for biopharmaceuticals using mammalian cells still suffer from cellular limitations such as limited growth, low productivity and stress resistance as well as higher expenses compared to bacterial or yeast based expression systems. Besides bioprocess, media and vector optimizations, advances in host cell engineering technologies comprising introduction, knock-out or post-transcriptional silencing of engineering genes have paved the way for remarkable achievements in CHO cell line development. Furthermore, thorough analysis of cellular pathways and mechanisms important for bioprocessing steadily unravels novel target molecules which might be addressed by functional genomic tools in order to establish superior production cell factories. This review provides a comprehensive summary of the most fundamental achievements in CHO cell engineering over the past three decades. Finally, the authors discuss the potential of novel and innovative methodologies that might contribute to further enhancement of existing CHO based production platforms for biopharmaceutical manufacturing in the future.


Cell Proliferation | 1998

Transcriptional regulation and biological significance of the insulin like growth factor II gene

Wilhelm Engström; A. Shokrai; Kerstin Otte; M. Granerus; Åsa Gessbo; P. Bierke; A. Madej; M. Sjolund; Andrew Ward

Abstract. The insulin like growth factors I and II are the most ubiquitous in the mammalian embryo. Moreover they play a pivotal role in the development and growth of tumours. The bioavailability of these growth factors is regulated on a transcriptional as well as on a posttranslational level. The expression of non‐signalling receptors as well as binding proteins does further tune the local concentration of IGFs. This paper aims at reviewing how the transcription of the IGF genes is regulated. The biological significance of these control mechanisms will be discussed.


Biotechnology Journal | 2014

A functional high‐content miRNA screen identifies miR‐30 family to boost recombinant protein production in CHO cells

Simon Fischer; Theresa Buck; Andreas Wagner; Carolin Ehrhart; Julia Giancaterino; Samuel Mang; Matthias Schad; Sven Mathias; Armaz Aschrafi; René Handrick; Kerstin Otte

The steady improvement of mammalian cell factories for the production of biopharmaceuticals is a key challenge for the biotechnology community. Recently, small regulatory microRNAs (miRNAs) were identified as novel targets for optimizing Chinese hamster ovary (CHO) production cells as they do not add any translational burden to the cell while being capable of regulating entire physiological pathways. The aim of the present study was to elucidate miRNA function in a recombinant CHO-SEAP cell line by means of a genome-wide high-content miRNA screen. This screen revealed that out of the 1, 139 miRNAs examined, 21% of the miRNAs enhanced cell-specific SEAP productivity mainly resulting in elevated volumetric yields, while cell proliferation was accelerated by 5% of the miRNAs. Conversely, cell death was diminished by 13% (apoptosis) or 4% (necrosis) of all transfected miRNAs. Besides these large number of identified target miRNAs, the outcome of our studies suggest that the entire miR-30 family substantially improves bioprocess performance of CHO cells. Stable miR-30 over expressing cells outperformed parental cells by increasing SEAP productivity or maximum cell density of approximately twofold. Our results highlight the application of miRNAs as powerful tools for CHO cell engineering, identified the miR-30 family as a critical component of cell proliferation, and support the notion that miRNAs are powerful determinants of cell viability.


Journal of Biotechnology | 2013

Breaking limitations of complex culture media: Functional non-viral miRNA delivery into pharmaceutical production cell lines

Simon Fischer; Andreas Wagner; Aron Kos; Armaz Aschrafi; René Handrick; Juergen Hannemann; Kerstin Otte

MicroRNAs (miRNAs) are promising targets for cell engineering through modulation of crucial cellular pathways. An effective introduction of miRNAs into the cell is a prerequisite to reliably study microRNA function. Previously, non-viral delivery of nucleic acids has been demonstrated to be cell type as well as culture medium dependent. Due to their importance for biopharmaceutical research and manufacturing, Chinese hamster ovary (CHO) and Cevecs Amniocyte Production (CAP) cells were used as host cell lines to investigate transfection reagents with respect to successful delivery of small non-coding RNAs (ncRNAs) and their ability to allow for biological activity of miRNAs and small interfering RNAs (siRNAs) within the cell. In the present study, we screened numerous transfection reagents for their suitability to successfully deliver miRNA mimics into CHO DG44 and CAP cells. Our investigation revealed that the determination of transfection efficiency for a given transfection reagent alone is not sufficient to draw conclusions about its ability to maintain the functionality of the miRNA. We could show that independent from high transfection rates observed for several reagents only one was suitable for efficient introduction of functional miRNA mimics into cells cultured in complex protein production media. We provide evidence for the functionality of transferred ncRNAs by demonstrating siRNA-mediated changes in protein levels and cellular phenotype as well as decreased twinfilin-1 (twf-1) transcript levels by its upstream miR-1 regulator. Furthermore, the process could be shown to be scalable which has important implications for biotechnological applications.


RNA Biology | 2015

Unveiling the principle of microRNA-mediated redundancy in cellular pathway regulation

Simon Fischer; René Handrick; Armaz Aschrafi; Kerstin Otte

Understanding the multifaceted nature of microRNA (miRNA) function in mammalian cells is still a challenge. Commonly accepted principles of cooperativity and multiplicity of miRNA function imply that individual mRNAs can be targeted by several miRNAs whereas a single miRNA may concomitantly regulate a subset of different genes. However, there is a paucity of information whether multiple miRNAs regulate critical cellular events and thereby acting redundantly. To gain insight into this notion, we conducted an unbiased high-content miRNA screen by individually introducing 1139 miRNA mimics into Chinese hamster ovary (CHO) cells. We discovered that 66% of all miRNAs significantly impacted on proliferation, protein expression, apoptosis and necrosis. In summary, we provide evidence for a substantial degree of redundancy among miRNAs to maintain cellular homeostasis.


Mammalian Genome | 1997

FISH mapping of the IGF2 gene in horse and donkey : detection of homoeology with HSA11

Terje Raudsepp; Kerstin Otte; Björn Rozell; B. P. Chowdhary

Three genomic subclones derived from a phage clone containing the equine IGF2 gene were used to FISH map the gene on horse (ECA) and donkey (EAS) metaphase chromosomes. The gene mapped on ECA 12q13 band and is the first locus mapped to this horse chromosome. In donkey the gene mapped very terminal on the long arm of one small submetacentric chromosome that shows almost identical DAPI-banding pattern with ECA12. This is the first locus mapped in donkey genome. Cross species chromosome painting of equine metaphase chromosomes with human Chromosome (Chr) 11-specific probe showed homoeology of this human chromosome with ECA12 and ECA7. The novel ECA12 comparative painting results are thus in accordance with the localization of the equine IGF2 gene. Comparison of the hitherto known physical locations of IGF2 in different species, viz. human, cattle, sheep, horse, and donkey, shows that this gene tends to maintain a terminal location on the chromosome arm.


Biotechnology and Bioengineering | 2015

miR-2861 as novel HDAC5 inhibitor in CHO cells enhances productivity while maintaining product quality.

Simon Fischer; Albert Jesuran Paul; Andreas Wagner; Sven Mathias; Melanie Geiss; Franziska Schandock; Martin Domnowski; Jörg Zimmermann; René Handrick; Friedemann Hesse; Kerstin Otte

Histone deacetylase (HDAC) inhibitors have been exploited for years to improve recombinant protein expression in mammalian production cells. However, global HDAC inhibition is associated with negative effects on various cellular processes. microRNAs (miRNAs) have been shown to regulate gene expression in almost all eukaryotic cell types by controlling entire cellular pathways. Since miRNAs recently have gained much attention as next‐generation cell engineering tool to improve Chinese hamster ovary (CHO) cell factories, we were interested if miRNAs are able to specifically repress HDAC expression in CHO cells to circumvent limitations of unspecific HDAC inhibition. We discovered a novel miRNA in CHO cells, miR‐2861, which was shown to enhance productivity in various recombinant CHO cell lines. Furthermore, we demonstrate that miR‐2861 might post‐transcriptionally regulate HDAC5 in CHO cells. Intriguingly, siRNA‐mediated HDAC5 suppression could be demonstrated to phenocopy pro‐productive effects of miR‐2861 in CHO cells. This supports the notion that miRNA‐induced inhibition of HDAC5 may contribute to productivity enhancing effects of miR‐2861. Furthermore, since product quality is fundamental to safety and functionality of biologics, we examined the effect of HDAC inhibition on critical product quality attributes. In contrast to unspecific HDAC inhibition using VPA, enforced expression of miR‐2861 did not negatively influence antibody aggregation or N‐glycosylation. Our findings highlight the superiority of miRNA‐mediated inhibition of specific HDACs and present miR‐2861 as novel cell engineering tool for improving CHO manufacturing cells. Biotechnol. Bioeng. 2015;112: 2142–2153.


Journal of Biotechnology | 2015

Enhanced protein production by microRNA-30 family in CHO cells is mediated by the modulation of the ubiquitin pathway.

Simon Fischer; Sven Mathias; Simone Schaz; Verena Vanessa Emmerling; Theresa Buck; Michael Kleemann; Matthias Hackl; Johannes Grillari; Armaz Aschrafi; René Handrick; Kerstin Otte

Functional genomics represent a valuable approach to improve culture performance of Chinese hamster ovary (CHO) cell lines for biopharmaceutical manufacturing. Recent advances in applied microRNA (miRNAs) research suggest that these small non-coding RNAs are critical for the regulation of cell phenotypes in CHO cells. However, the notion that individual miRNAs usually control the expression of hundreds of different genes makes miRNA target identification highly complex. We have recently reported that the entire miR-30 family enhances recombinant protein production in CHO cells. To better understand the pro-productive effects of this miRNA family, we set out to identify their downstream target genes in CHO cells. Computational target prediction combined with a comprehensive functional validation enabled the discovery of a set of twenty putative target genes for all productivity enhancing miR-30 family members. We demonstrate that all miR-30 isoforms contribute to the regulation of the ubiquitin pathway in CHO cells by directly targeting the ubiquitin E3 ligase S-phase kinase-associated protein 2 (Skp2). Finally, we provide several lines of evidence that miR-30-mediated modulation of the ubiquitin pathway may enhance recombinant protein expression in CHO cells. In summary, this study supports the importance of non-coding RNAs, especially of miRNAs, in the context of cell line engineering.


Biotechnology and Bioengineering | 2016

Temperature‐sensitive miR‐483 is a conserved regulator of recombinant protein and viral vector production in mammalian cells

Verena Vanessa Emmerling; Simon Fischer; Fabian Stiefel; Karlheinz Holzmann; René Handrick; Friedemann Hesse; Markus Hörer; Stefan Kochanek; Kerstin Otte

Cell engineering and bioprocess optimizations such as low temperature cultivation represent powerful tools to improve cellular performance and product yields of mammalian production cells. Besides monoclonal antibodies (mABs), novel biotherapeutic formats such as viral vectors will gain increasing importance. Here, we demonstrate that similar to Chinese hamster ovary (CHO) cells, product yields of recombinant adeno‐associated virus (rAAV) producing HeLa cells can be markedly increased by low temperature cultivation. MicroRNAs (miRNAs) are small non‐coding RNAs that critically regulate cell phenotypes. We thus investigated differential miRNA expression in response to mild hypothermia in CHO and HeLa production cells. We discovered miR‐483 to be substantially up‐regulated upon temperature down‐shift in both cell types. Functional validation experiments revealed that introduction of miR‐483 mimics led to a significant increase in both rAAV and mAB production in HeLa and CHO cells, respectively. Furthermore, inhibition of miR‐483 up‐regulation during mild hypothermia significantly decreased product yields, suggesting that miR‐483 is a key regulator of cellular productivity in mammalian cells. In addition, miRNA target gene identification indicated that miR‐483 might regulate genes directly involved in cellular survival and protein expression. Our results highlight that miR‐483 is a valuable tool for product‐independent engineering of mammalian production cells. Biotechnol. Bioeng. 2016;113: 830–841.


Endocrinology | 2016

GATA4 Regulates Blood-Testis Barrier Function and Lactate Metabolism in Mouse Sertoli Cells.

Anja Schrade; Antti Kyrönlahti; Oyediran Akinrinade; Marjut Pihlajoki; Simon Fischer; Verena Martinez Rodriguez; Kerstin Otte; Vidya Velagapudi; Jorma Toppari; David B. Wilson; Markku Heikinheimo

Conditional deletion of Gata4 in Sertoli cells (SCs) of adult mice has been shown to increase permeability of the blood-testis barrier (BTB) and disrupt spermatogenesis. To gain insight into the molecular underpinnings of these phenotypic abnormalities, we assessed the impact of Gata4 gene silencing in cell culture models. Microarray hybridization identified genes dysregulated by siRNA-mediated inhibition of Gata4 in TM4 cells, an immortalized mouse SC line. Differentially expressed genes were validated by quantitative RT-PCR analysis of primary cultures of Gata4(flox/flox) mouse SCs that had been subjected to cre-mediated recombination in vitro. Depletion of GATA4 in TM4 cells and primary SCs was associated with altered expression of genes involved in key facets of BTB maintenance, including tight/adherens junction formation (Tjp1, Cldn12, Vcl, Tnc, Csk) and extracellular matrix reorganization (Lamc1, Col4a1, Col4a5, Mmp10, Mmp23, Timp2). Western blotting and immunocytochemistry demonstrated reduced levels of tight junction protein-1, a prototypical tight junction protein, in GATA4-depleted cells. These changes were accompanied by a loss of morphologically recognizable junctional complexes and a decline in epithelial membrane resistance. Furthermore, Gata4 gene silencing was associated with altered expression of Hk1, Gpi1, Pfkp, Pgam1, Gls2, Pdk3, Pkd4, and Ldhb, genes regulating the production of lactate, a key nutrient that SCs provide to developing germ cells. Comprehensive metabolomic profiling demonstrated impaired lactate production in GATA4-deficient SCs. We conclude that GATA4 plays a pivotal role in the regulation of BTB function and lactate metabolism in mouse SCs.

Collaboration


Dive into the Kerstin Otte's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

René Handrick

University of Applied Sciences Biberach

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Björn Rozell

Swedish University of Agricultural Sciences

View shared research outputs
Top Co-Authors

Avatar

Wilhelm Engström

Swedish University of Agricultural Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fabian Stiefel

University of Applied Sciences Biberach

View shared research outputs
Top Co-Authors

Avatar

Sven Mathias

Radboud University Nijmegen

View shared research outputs
Researchain Logo
Decentralizing Knowledge