@article{VottelerCarvajalBerrioPudlasetal.2012, author = {Votteler, Miriam and Carvajal Berrio, Daniel A. and Pudlas, Marieke and Walles, Heike and Schenke-Layland, Katja}, title = {Non-contact, Label-free Monitoring of Cells and Extracellular Matrix using Raman Spectroscopy}, series = {Journal of Visual Expression}, volume = {63}, journal = {Journal of Visual Expression}, number = {e3977}, doi = {10.3791/3977}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-124569}, year = {2012}, abstract = {Non-destructive, non-contact and label-free technologies to monitor cell and tissue cultures are needed in the field of biomedical research.1-5 However, currently available routine methods require processing steps and alter sample integrity. Raman spectroscopy is a fast method that enables the measurement of biological samples without the need for further processing steps. This laser-based technology detects the inelastic scattering of monochromatic light.6 As every chemical vibration is assigned to a specific Raman band (wavenumber in cm-1), each biological sample features a typical spectral pattern due to their inherent biochemical composition.7-9 Within Raman spectra, the peak intensities correlate with the amount of the present molecular bonds.1 Similarities and differences of the spectral data sets can be detected by employing a multivariate analysis (e.g. principal component analysis (PCA)).10 Here, we perform Raman spectroscopy of living cells and native tissues. Cells are either seeded on glass bottom dishes or kept in suspension under normal cell culture conditions (37 °C, 5\% CO2) before measurement. Native tissues are dissected and stored in phosphate buffered saline (PBS) at 4 °C prior measurements. Depending on our experimental set up, we then either focused on the cell nucleus or extracellular matrix (ECM) proteins such as elastin and collagen. For all studies, a minimum of 30 cells or 30 random points of interest within the ECM are measured. Data processing steps included background subtraction and normalization.}, language = {en} } @article{JakobEbertRudertetal.2012, author = {Jakob, Franz and Ebert, Regina and Rudert, Maximilian and N{\"o}th, Ulrich and Walles, Heike and Docheva, Denitsa and Schieker, Matthias and Meinel, Lorenz and Groll, J{\"u}rgen}, title = {In situ guided tissue regeneration in musculoskeletal diseases and aging}, series = {Cell and Tissue Research}, volume = {347}, journal = {Cell and Tissue Research}, number = {3}, doi = {10.1007/s00441-011-1237-z}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-124738}, pages = {725-735}, year = {2012}, abstract = {In situ guided tissue regeneration, also addressed as in situ tissue engineering or endogenous regeneration, has a great potential for population-wide "minimal invasive" applications. During the last two decades, tissue engineering has been developed with remarkable in vitro and preclinical success but still the number of applications in clinical routine is extremely small. Moreover, the vision of population-wide applications of ex vivo tissue engineered constructs based on cells, growth and differentiation factors and scaffolds, must probably be deemed unrealistic for economic and regulation-related issues. Hence, the progress made in this respect will be mostly applicable to a fraction of post-traumatic or post-surgery situations such as big tissue defects due to tumor manifestation. Minimally invasive procedures would probably qualify for a broader application and ideally would only require off the shelf standardized products without cells. Such products should mimic the microenvironment of regenerating tissues and make use of the endogenous tissue regeneration capacities. Functionally, the chemotaxis of regenerative cells, their amplification as a transient amplifying pool and their concerted differentiation and remodeling should be addressed. This is especially important because the main target populations for such applications are the elderly and diseased. The quality of regenerative cells is impaired in such organisms and high levels of inhibitors also interfere with regeneration and healing. In metabolic bone diseases like osteoporosis, it is already known that antagonists for inhibitors such as activin and sclerostin enhance bone formation. Implementing such strategies into applications for in situ guided tissue regeneration should greatly enhance the efficacy of tailored procedures in the future.}, language = {en} } @article{RackwitzEdenReppenhagenetal.2012, author = {Rackwitz, Lars and Eden, Lars and Reppenhagen, Stephan and Reichert, Johannes C. and Jakob, Franz and Walles, Heike and Pullig, Oliver and Tuan, Rocky S. and Rudert, Maximilian and N{\"o}th, Ulrich}, title = {Stem cell- and growth factor-based regenerative therapies for avascular necrosis of the femoral head}, series = {Stem Cell Research \& Therapy}, volume = {3}, journal = {Stem Cell Research \& Therapy}, number = {7}, doi = {10.1186/scrt98}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-135413}, year = {2012}, abstract = {Avascular necrosis (AVN) of the femoral head is a debilitating disease of multifactorial genesis, predominately affects young patients, and often leads to the development of secondary osteoarthritis. The evolving field of regenerative medicine offers promising treatment strategies using cells, biomaterial scaffolds, and bioactive factors, which might improve clinical outcome. Early stages of AVN with preserved structural integrity of the subchondral plate are accessible to retrograde surgical procedures, such as core decompression to reduce the intraosseous pressure and to induce bone remodeling. The additive application of concentrated bone marrow aspirates, ex vivo expanded mesenchymal stem cells, and osteogenic or angiogenic growth factors (or both) holds great potential to improve bone regeneration. In contrast, advanced stages of AVN with collapsed subchondral bone require an osteochondral reconstruction to preserve the physiological joint function. Analogously to strategies for osteochondral reconstruction in the knee, anterograde surgical techniques, such as osteochondral transplantation (mosaicplasty), matrix-based autologous chondrocyte implantation, or the use of acellular scaffolds alone, might preserve joint function and reduce the need for hip replacement. This review summarizes recent experimental accomplishments and initial clinical findings in the field of regenerative medicine which apply cells, growth factors, and matrices to address the clinical problem of AVN.}, language = {en} } @article{MollReboredoSchwarzetal.2013, author = {Moll, Corinna and Reboredo, Jenny and Schwarz, Thomas and Appelt, Antje and Sch{\"u}rlein, Sebastian and Walles, Heike and Nietzer, Sarah}, title = {Tissue Engineering of a Human 3D in vitro Tumor Test System}, series = {Journal of Visualized Experiments}, volume = {78}, journal = {Journal of Visualized Experiments}, number = {e50460}, doi = {10.3791/50460}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-132277}, year = {2013}, abstract = {Cancer is one of the leading causes of death worldwide. Current therapeutic strategies are predominantly developed in 2D culture systems, which inadequately reflect physiological conditions in vivo. Biological 3D matrices provide cells an environment in which cells can self-organize, allowing the study of tissue organization and cell differentiation. Such scaffolds can be seeded with a mixture of different cell types to study direct 3D cell-cell-interactions. To mimic the 3D complexity of cancer tumors, our group has developed a 3D in vitro tumor test system. Our 3D tissue test system models the in vivo situation of malignant peripheral nerve sheath tumors (MPNSTs), which we established with our decellularized porcine jejunal segment derived biological vascularized scaffold (BioVaSc). In our model, we reseeded a modified BioVaSc matrix with primary fibroblasts, microvascular endothelial cells (mvECs) and the S462 tumor cell line For static culture, the vascular structure of the BioVaSc is removed and the remaining scaffold is cut open on one side (Small Intestinal Submucosa SIS-Muc). The resulting matrix is then fixed between two metal rings (cell crowns). Another option is to culture the cell-seeded SIS-Muc in a flow bioreactor system that exposes the cells to shear stress. Here, the bioreactor is connected to a peristaltic pump in a self-constructed incubator. A computer regulates the arterial oxygen and nutrient supply via parameters such as blood pressure, temperature, and flow rate. This setup allows for a dynamic culture with either pressure-regulated pulsatile or constant flow. In this study, we could successfully establish both a static and dynamic 3D culture system for MPNSTs. The ability to model cancer tumors in a more natural 3D environment will enable the discovery, testing, and validation of future pharmaceuticals in a human-like model.}, language = {en} } @article{RamachandranSchirmerMuenstetal.2015, author = {Ramachandran, Sarada Devi and Schirmer, Katharina and M{\"u}nst, Bernhard and Heinz, Stefan and Ghafoory, Shahrouz and W{\"o}lfl, Stefan and Simon-Keller, Katja and Marx, Alexander and {\O}ie, Cristina Ionica and Ebert, Matthias P. and Walles, Heike and Braspenning, Joris and Breitkopf-Heinlein, Katja}, title = {In Vitro Generation of Functional Liver Organoid-Like Structures Using Adult Human Cells}, series = {PLoS One}, volume = {10}, journal = {PLoS One}, number = {10}, doi = {10.1371/journal.pone.0139345}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-139552}, pages = {e0139345}, year = {2015}, abstract = {In this study we used differentiated adult human upcyte (R) cells for the in vitro generation of liver organoids. Upcyte (R) cells are genetically engineered cell strains derived from primary human cells by lenti-viral transduction of genes or gene combinations inducing transient proliferation capacity (upcyte (R) process). Proliferating upcyte (R) cells undergo a finite number of cell divisions, i.e., 20 to 40 population doublings, but upon withdrawal of proliferation stimulating factors, they regain most of the cell specific characteristics of primary cells. When a defined mixture of differentiated human upcyte (R) cells (hepatocytes, liver sinusoidal endothelial cells (LSECs) and mesenchymal stem cells (MSCs)) was cultured in vitro on a thick layer of Matrigel\(^{TM}\), they self-organized to form liver organoid-like structures within 24 hours. When further cultured for 10 days in a bioreactor, these liver organoids show typical functional characteristics of liver parenchyma including activity of cytochromes P450, CYP3A4, CYP2B6 and CYP2C9 as well as mRNA expression of several marker genes and other enzymes. In summary, we hereby describe that 3D functional hepatic structures composed of primary human cell strains can be generated in vitro. They can be cultured for a prolonged period of time and are potentially useful ex vivo models to study liver functions.}, language = {en} } @article{GordonDaneshianBouwstraetal.2015, author = {Gordon, Sarah and Daneshian, Mardas and Bouwstra, Joke and Caloni, Francesca and Constant, Samuel and Davies, Donna E. and Dandekar, Gudrun and Guzman, Carlos A. and Fabian, Eric and Haltner, Eleonore and Hartung, Thomas and Hasiwa, Nina and Hayden, Patrick and Kandarova, Helena and Khare, Sangeeta and Krug, Harald F. and Kneuer, Carsten and Leist, Marcel and Lian, Guoping and Marx, Uwe and Metzger, Marco and Ott, Katharina and Prieto, Pilar and Roberts, Michael S. and Roggen, Erwin L. and Tralau, Tewes and van den Braak, Claudia and Walles, Heike and Lehr, Claus-Michael}, title = {Non-animal models of epithelial barriers (skin, intestine and lung) in research, industrial applications and regulatory toxicology}, series = {ALTEX: Alternatives to Animal Experimentation}, volume = {32}, journal = {ALTEX: Alternatives to Animal Experimentation}, number = {4}, doi = {10.14573/altex.1510051}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-144275}, pages = {327-378}, year = {2015}, abstract = {Models of the outer epithelia of the human body namely the skin, the intestine and the lung have found valid applications in both research and industrial settings as attractive alternatives to animal testing. A variety of approaches to model these barriers are currently employed in such fields, ranging from the utilization of ex vivo tissue to reconstructed in vitro models, and further to chip-based technologies, synthetic membrane systems and, of increasing current interest, in silico modeling approaches. An international group of experts in the field of epithelial barriers was convened from academia, industry and regulatory bodies to present both the current state of the art of non-animal models of the skin, intestinal and pulmonary barriers in their various fields of application, and to discuss research-based, industry-driven and regulatory-relevant future directions for both the development of new models and the refinement of existing test methods. Issues of model relevance and preference, validation and standardization, acceptance, and the need for simplicity versus complexity were focal themes of the discussions. The outcomes of workshop presentations and discussions, in relation to both current status and future directions in the utilization and development of epithelial barrier models, are presented by the attending experts in the current report.}, language = {en} } @article{RamachandranVivaresKlieberetal.2015, author = {Ramachandran, Sarada D. and Vivar{\`e}s, Aur{\´e}lie and Klieber, Sylvie and Hewitt, Nicola J. and Muenst, Bernhard and Heinz, Stefan and Walles, Heike and Braspenning, Joris}, title = {Applicability of second-generation upcyte\(^{®}\) human hepatocytes for use in CYP inhibition and induction studies}, series = {Pharmacology Research \& Perspectives}, volume = {3}, journal = {Pharmacology Research \& Perspectives}, number = {5}, doi = {10.1002/prp2.161}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-149564}, pages = {e00161}, year = {2015}, abstract = {Human upcyte\(^{®}\) hepatocytes are proliferating hepatocytes that retain many characteristics of primary human hepatocytes. We conducted a comprehensive evaluation of the application of second-generation upcyte\(^{®}\) hepatocytes from four donors for inhibition and induction assays using a selection of reference inhibitors and inducers. CYP1A2, CYP2B6, CYP2C9, and CYP3A4 were reproducibly inhibited in a concentration-dependent manner and the calculated IC\(_{50}\) values for each compound correctly classified them as potent inhibitors. Upcyte\(^{®}\) hepatocytes were responsive to prototypical CYP1A2, CYP2B6, CYP2C9, and CYP3A4 inducers, confirming that they have functional AhR-, CAR-, and PXR-mediated CYP regulation. A panel of 11 inducers classified as potent, moderate or noninducers of CYP3A4 and CYP2B6 were tested. There was a good fit of data from upcyte\(^{®}\) hepatocytes to three different predictive models for CYP3A4 induction, namely the Relative Induction Score (RIS), AUC\(_{u}\)/F\(_{2}\), and C\(_{max,u}\)/Ind\(_{50}\). In addition, PXR (rifampicin) and CAR-selective (carbamazepine and phenytoin) inducers of CYP3A4 and CYP2B6 induction, respectively, were demonstrated. In conclusion, these data support the use of second-generation upcyte\(^{®}\) hepatocytes for CYP inhibition and induction assays. Under the culture conditions used, these cells expressed CYP activities that were equivalent to or higher than those measured in primary human hepatocyte cultures, which could be inhibited or induced by prototypical CYP inhibitors and inducers, respectively. Moreover, they can be used to predict in vivo CYP3A4 induction potential using three prediction models. Bulk availability of cells from multiple donors makes upcyte\(^{®}\) hepatocytes suitable for DDI screening, as well as more in-depth mechanistic investigations.}, language = {en} } @article{GroeberSchoberSchmidetal.2016, author = {Groeber, Florian and Schober, Lena and Schmid, Freia F. and Traube, Andrea and Kolbus-Hernandez, Silvia and Daton, Karolina and Hoffmann, Sebastian and Petersohn, Dirk and Schaefer-Korting, Monika and Walles, Heike and Mewes, Karsten R.}, title = {Catch-up validation study of an in vitro skin irritation test method based on an open source reconstructed epidermis (phase II)}, series = {Toxicology in Vitro}, volume = {36}, journal = {Toxicology in Vitro}, doi = {10.1016/j.tiv.2016.07.008}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-187311}, pages = {254-261}, year = {2016}, abstract = {To replace the Draize skin irritation assay (OECD guideline 404) several test methods based on reconstructed human epidermis (RHE) have been developed and were adopted in the OECD test guideline 439. However, all validated test methods in the guideline are linked to RHE provided by only three companies. Thus,the availability of these test models is dependent on the commercial interest of the producer. To overcome this limitation and thus to increase the accessibility of in vitro skin irritation testing, an open source reconstructed epidermis (OS-REp) was introduced. To demonstrate the capacity of the OS-REp in regulatory risk assessment, a catch-up-validation study was performed. The participating laboratories used in-house generated OS-REp to assess the set of 20 reference substances according to the performance standards amending the OECD test guideline 439. Testing was performed under blinded conditions. The within-laboratory reproducibility of 87\% and the inter-laboratory reproducibility of 85\% prove a high reliability of irritancy testing using the OS-REp protocol. In addition, the prediction capacity was with an accuracy of 80\% comparable to previous published RHE based test protocols. Taken together the results indicate that the OS-REp test method can be used as a standalone alternative skin irritation test replacing the OECD test guideline 404.}, language = {en} } @article{KunzWolfSchulzeetal.2016, author = {Kunz, Meik and Wolf, Beat and Schulze, Harald and Atlan, David and Walles, Thorsten and Walles, Heike and Dandekar, Thomas}, title = {Non-Coding RNAs in Lung Cancer: Contribution of Bioinformatics Analysis to the Development of Non-Invasive Diagnostic Tools}, series = {Genes}, volume = {8}, journal = {Genes}, number = {1}, doi = {10.3390/genes8010008}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-147990}, pages = {8}, year = {2016}, abstract = {Lung cancer is currently the leading cause of cancer related mortality due to late diagnosis and limited treatment intervention. Non-coding RNAs are not translated into proteins and have emerged as fundamental regulators of gene expression. Recent studies reported that microRNAs and long non-coding RNAs are involved in lung cancer development and progression. Moreover, they appear as new promising non-invasive biomarkers for early lung cancer diagnosis. Here, we highlight their potential as biomarker in lung cancer and present how bioinformatics can contribute to the development of non-invasive diagnostic tools. For this, we discuss several bioinformatics algorithms and software tools for a comprehensive understanding and functional characterization of microRNAs and long non-coding RNAs.}, language = {en} } @article{RosenbaumSchickWollbornetal.2016, author = {Rosenbaum, Corinna and Schick, Martin Alexander and Wollborn, Jakob and Heider, Andreas and Scholz, Claus-J{\"u}rgen and Cecil, Alexander and Niesler, Beate and Hirrlinger, Johannes and Walles, Heike and Metzger, Marco}, title = {Activation of Myenteric Glia during Acute Inflammation In Vitro and In Vivo}, series = {PLoS One}, volume = {11}, journal = {PLoS One}, number = {3}, doi = {10.1371/journal.pone.0151335}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-146544}, pages = {e0151335}, year = {2016}, abstract = {Background Enteric glial cells (EGCs) are the main constituent of the enteric nervous system and share similarities with astrocytes from the central nervous system including their reactivity to an inflammatory microenvironment. Previous studies on EGC pathophysiology have specifically focused on mucosal glia activation and its contribution to mucosal inflammatory processes observed in the gut of inflammatory bowel disease (IBD) patients. In contrast knowledge is scarce on intestinal inflammation not locally restricted to the mucosa but systemically affecting the intestine and its effect on the overall EGC network. Methods and Results In this study, we analyzed the biological effects of a systemic LPS-induced hyperinflammatory insult on overall EGCs in a rat model in vivo, mimicking the clinical situation of systemic inflammation response syndrome (SIRS). Tissues from small and large intestine were removed 4 hours after systemic LPS-injection and analyzed on transcript and protein level. Laser capture microdissection was performed to study plexus-specific gene expression alterations. Upon systemic LPS-injection in vivo we observed a rapid and dramatic activation of Glial Fibrillary Acidic Protein (GFAP)-expressing glia on mRNA level, locally restricted to the myenteric plexus. To study the specific role of the GFAP subpopulation, we established flow cytometry-purified primary glial cell cultures from GFAP promotor-driven EGFP reporter mice. After LPS stimulation, we analyzed cytokine secretion and global gene expression profiles, which were finally implemented in a bioinformatic comparative transcriptome analysis. Enriched GFAP+ glial cells cultured as gliospheres secreted increased levels of prominent inflammatory cytokines upon LPS stimulation. Additionally, a shift in myenteric glial gene expression profile was induced that predominantly affected genes associated with immune response. Conclusion and Significance Our findings identify the myenteric GFAP-expressing glial subpopulation as particularly susceptible and responsive to acute systemic inflammation of the gut wall and complement knowledge on glial involvement in mucosal inflammation of the intestine.}, language = {en} }