Metanavigation:

Hier finden Sie den Zugang zur Notfallseite, Kontaktinformationen, Barrierefreiheits-Einstellungen, die Sprachwahl und die Suchfunktion.

Navigation öffnen

ECRT Projekte

Das ECRT will insbesondere den wissenschaftlichen Nachwuchs beim Aufbau eigener Forschungsvorhaben unterstützen. Dafür bietet das ECRT Einstein Kickboxen mit einer Startfinanzierung für erste Experimente zur Validierung der Forschungsideen sowie ECRT Research oder Consumable Grants zur Weiterentwicklung der Forschungsvorhaben.

Die Zusammenfassungen der Projekte stehen nur auf Englsich zur Verfügung.

Sie befinden sich hier:

Frühere ECRT Consumable Grants

ECRT Consumable Grants - Förderperiode 2022

Homeostatic proliferation of naïve CD4+ T cells, induces epigenetic changes, which contributes to immune aging

Aging is associated with a functional decline of the immune system, which is linked to the inferior immune response of elderly people to infections and vaccinations. Naïve T cells (TN) are important for reactivity against new infections and vaccines. In humans however, the generation of new TN clones ceases as early as puberty. To maintain a high number of CD4+ TN, the cells must undergo activation-independent proliferation, known as homeostatic proliferation. CD4+ TN which lack the surface marker CD31 (CD31-), are enriched for cells with a greater homeostatic proliferation history compared to CD31 expressing TN (CD31+). Indeed, the number of CD31+ CD4+ TN decreases with age as well as the telomere length in TN, demonstrating that cells from older individuals experienced a higher number of homeostatic proliferation events.
Our working hypothesis is that the accumulation of homeostatic proliferation events over the life-time of a human being in CD4+ TN induces changes in the cell’s epigenome, which impact T cell function and contribute to the impaired immune responses in elderly. Indeed, our preliminary data show that accumulation of homeostatic proliferation events induces stable epigenetic changes in ex vivo derived CD4 TN of young healthy donors.With the support of the Einstein funding we want to show that these differences increase in elderly healthy donors and further characterize the potential differences in transcriptional regulation in CD31+ and CD31- TN.

Team: Dania Hamo, Mai Phan, Andreas Magg

Funding: ECRT Consumable Grant - Young Scientists

Immune modulation as a potential treatment strategy against periprosthetic joint infection

Periprosthetic joint infection (PJI) is a severe complication occurring in approximately 1-5% of all patients after total joint replacement. Despite successful revision surgery, risk of aseptic loosening is significantly elevated due to the impaired capability of bone regeneration. Programmed cell death protein-1 (PD-1), a receptor expressed on various cell types, has been recently shown to affect the bone metabolism, specifically on osteoclasts. We hypothesize that targeting PD-1 is a promising strategy to improve bone quality during PJI. Here we plan to introduce a bone explant culture mimicking in vivo environment. This model will allow us to test the overall change of the bony scaffold and the behavior of the different cells inside ex vivo under the influence of PD-1 inhibitor. Understanding the phenomenon and related mechanisms will provide us deeper knowledge on this unique therapeutic approach to improve implant survival.

Team: Yi Ren, Arne Kienzle, Denise Jahn, Weijie Du

Funding: ECRT Consumable Grant - Young Scientists

Oxalate-induced RyR dysfunction as a substrate for cardiac arrhythmias and contractile impairment

Prevalence estimates indicate that approximately 13% of the US adult population suffers from CKD. In 2019, the G-BA registered 93.089 dialysis patients in Germany. Both conditions are associated with an increased cardiovascular risk. In patients undergoing dialysis, sudden cardiac death is the most frequent cause of death (~25%). Tailored therapies related to underlying disease mechanisms may prove to be effective in reducing cardiovascular morbidity and mortality in these patients.

Reduced renal function leads to increased levels of plasma oxalate (POx). Several lines of evidence indicate that oxalate may have detrimental effects on the cardiovascular system. For example, post-hoc analysis of the German Diabetes Dialysis study unveiled a 62% higher risk for sudden cardiac death and 40% for cardiovascular events in dialysis patients of the highest Pox quartile compared to the lowest quartile. Elevated Pox concentrations are associated with cardiac fibrosis and altered hemodynamic parameters (pulse wave velocity, blood pressure). While these reports are intriguing, mechanisms of myocardial oxalate homeostasis and potential toxicity remain elusive.

Our previous experiments indicate that oxalate induces RyR dysfunction in cardiomyocytes in-vitro, which is considered key component of cardiac arrhythmogenesis and contractile function. In this proposal, we hypothesize that elevated concentrations of plasma oxalate affect cardiac contractility and susceptibility to arrhythmias through functional impairment of the cardiac ryanodine receptor (RyR) in-vivo.

Team: David Bode, Gerlineke Hawkins-van der Clingel, Cristian Sotomayor-Flores

Funding: ECRT Consumable Grant - Young Scientists

Deciphering the cancer stem cell niche – The road to standardize patient-derived lung cancer or-ganoid culture

Lung cancer is characterized by potent and individual drug resistance due to genetic and molecular diversity. Patient-derived lung cancer organoids (PDLCOs), 3D multicellular structures derived from patient tumors, could serve as preclinical biomarkers of individual responses to therapies. Lung cancer has been suggested to originate from cancer stem cells (CSCs). CSCs are stimulated by niche factors, secreted by non-epithelial cells in vivo. These niche factors need to be substituted in PDLCO cultures as organoids comprise only epithelial cells. However, heterogeneous trajectories of lung CSCs, such as gene expression, impede standardized and efficient PDLCO culture so far. We could identify putative trajectories of lung CSCs and their niche factor requirements. In our project, we plan to generate PDLCOs with four different growth factor cocktails (artificial niche). We will perform RNA sequencing of organoids and primary tumors. Comparing the transcriptomic signature of the individual CSC niche with the respective growth factor cocktail that successfully enables organoid growth (=successful mimicking of the niche) will enable us to predict the specific growth factors requirements for different tumor / CSC signatures. With our proof of concept approach, we could individualize protocols and efficiently generate PDLCOs as platforms for personalized medicine.

Team: Lukas Ehlen, Martin Szyska, Michael Schmück-Henneresse, Martí Farrera I Sal, Janine Arndt

Funding: ECRT Consumable Grant - Advanced Scientists

On the other side of the table – Defining the pathogenic and regenerative role of renal transplant tubular epithelial cells in ischemia-reperfusion-injury and alloreactivity

In our previous Einstein Kickbox project, we developed an assay using cells from the urine of kidney transplant patients to measure transplant reactive T cells. This allows the monitoring of immune reactions towards the allograft and the detection of acute rejection episodes, enabling personalized medicine. With the Einstein Kickbox consumable grant, we aim to deepen our understanding of the kidney transplant tubular epithelial cells (TEC). After explantation, the donor kidney is subject to warm and cold ischemia. The severity of the subsequent organ inflammation determines future immune reactivity and fibrosis. A clinical symptom of these processes is a delayed graft function, which is associated with a reduced long-term graft and patient survival. Thus, the goal of this project is to investigate the role of transplant TEC in pathologies and regeneration following kidney transplantation. Therefore, we will use the consumable grant to perform comprehensive analyses of the urine-derived transplant TEC. We will analyze changes in protein expression and epigenetic regulation in different patient cohorts (kidney transplant patients with and without delayed graft function or acute rejection and healthy donors) and different conditions (inflammation and hypoxia). All in all, we plan to gain knowledge about the pathogenic or regenerative capacities of kidney cells in transplantation and open new doors for future translational research projects and new therapeutic approaches.

Team: Constantin Thieme, Nina Babel, Julia Polánsky-Biskup, Toralf Roch, Luka Queitsch

Funding: ECRT Consumable Grant - Advanced Scientists

Cell-mediated depletion of residual TCR+ cells for allogeneic CAR T cell products

T lymphocytes are immune cells that defend the body from pathogens and cancer. They can be genetically modified to express designed receptors such as Chimeric Antigen Receptors (CARs), enabling them to recognize antigens of choice. Such CAR T cells have been employed in the treatment of several oncological disorders, the best clinical example being CD19-specific CAR T cells used to treat B cell derived leukaemia and lymphoma.

We recently improved a method for efficient manufacturing of T cell receptor (TCR) replaced CAR T cells. We employ CRISPR-Cas9 to introduce a CAR transgene into the TCR alpha chain constant locus (TRAC). To this end, sgRNA/Cas9 complexes and DNA molecules carrying a CAR transgene are co-transfected into T cells. By exploitation of Homology-directed DNA Repair, the CAR transgene can be brought under the transcriptional control of the TRAC promoter. CAR integration at this site furthermore prevents formation of functional TCRs.

This method mainly produces TCR-replaced CAR T cells carrying the CAR as their only specificity-determining receptor, and TCR-/CAR-double negative T cells, incapable of any antigen-specific action. While these cells would be fit for allogeneic use, there are also small fractions of unedited and CAR+/TCR+ double positive T cells. These remaining TCR+ cells could potentially induce Graft-vs-Host disease if transfused in an allogeneic setting. This project aims to eradicate all remaining TCR+ contaminants in order to generate safe off-the-shelf CAR T cell products.

T lymphocytes are immune cells that defend the body from pathogens and cancer. They can be genetically modified to express designed receptors such as Chimeric Antigen Receptors (CARs), enabling them to recognize antigens of choice. Such CAR T cells have been employed in the treatment of several oncological disorders, the best clinical example being CD19-specific CAR T cells used to treat B cell derived leukaemia and lymphoma.

We recently improved a method for efficient manufacturing of T cell receptor (TCR) replaced CAR T cells. We employ CRISPR-Cas9 to introduce a CAR transgene into the TCR alpha chain constant locus (TRAC). To this end, sgRNA/Cas9 complexes and DNA molecules carrying a CAR transgene are co-transfected into T cells. By exploitation of Homology-directed DNA Repair, the CAR transgene can be brought under the transcriptional control of the TRAC promoter. CAR integration at this site furthermore prevents formation of functional TCRs.

This method mainly produces TCR-replaced CAR T cells carrying the CAR as their only specificity-determining receptor, and TCR-/CAR-double negative T cells, incapable of any antigen-specific action. While these cells would be fit for allogeneic use, there are also small fractions of unedited and CAR+/TCR+ double positive T cells. These remaining TCR+ cells could potentially induce Graft-vs-Host disease if transfused in an allogeneic setting. This project aims to eradicate all remaining TCR+ contaminants in order to generate safe off-the-shelf CAR T cell products.

Team: Jonas Kath, Weijie Du, Magdi Elsallab, Stefania Martini

Funding: ECRT Consumable Grant

ECRT Consumable Grants - Förderperiode 2021

A virus-free approach to generate safe and effective CAR-T cell products for third-party use in patients with solid tumours

T cells are a major part of our adaptive immunity, being able to eliminate infected or transformed target cells and are increasingly exploited as powerful tools in oncology. They can be redirected to recognize and eliminate cancer cells using Chimeric Antigen Receptors (CARs), fusion proteins of antibodies for antigen recognition and T cell-specific signaling domains. While already effective in patients suffering from certain blood cancers, the generation of an individual autologous CAR-T-Cell product for a single patient is a difficult and costly endeavor. Treatment delay due to production processes and high costs hinder their broad application in the clinic. This kickbox application aims to test a novel approach to create CAR-T-Cell product from healthy donors that are unable to attack the patients cells via their endogenous T cell receptors in an allogenic treatment setting of solid tumors. Therefore, this "next generation" of CAR-T cells may enable the generation of potent but cost-effective treatments of solid tumours.

Team: Dimitrios L. Wagner, Jonas Kath, Lennart Ostendorf

Funding: ECRT Consumable Grant

Unravel fibronectin tensional state in a 3D physiological microenvironment – the impact of inflammatory signaling on early ECM formation

In regeneration, the cellular deposition of ECM is crucially linked with properties of the surrounding microenvironment. However, in an injury situation the ECM secreting fibroblast-like cells are influenced not only by the mechanical cues in their 3D microenvironment, but also by the inflammatory response orchestrating the healing process. We here suggest a novel, more holistic approach to reflect the versatile stimuli acting through immunological and mechanical cues on fibroblast-like cells upon injury. We aim to define whether Fn deposition and tensional state are related to mechanical properties and immune signaling to better understand beneficial and adverse healing scenarios. Understanding this complex interplay of immune response, 3D microenvironment mechanics and cell function will provide further insight into modeling of degenerative diseases and benefit regenerative approaches involving cell delivery using biomaterials.

Team: Matthias Kollert, Georgios Kotsaris, Julia Mehl, Christian Bucher

Funding: ECRT Consumable Grant

ECRT Consumable Grants - Förderperiode 2019

Adrenal Differentiation and Organoid Generation from Induced Pluripotent Stem Cells (iPSCs)

The adrenal cortex produces steroid hormones (aldosterone from the outermost zona glomerulosa, cortisol from the zona fasciculata and sex steroids from the zona reticularis). Disorders affecting the adrenal cortex represent a major health burden. They include primary aldosteronism (overproduction of aldosterone, a major cause of secondary hypertension), Cushing syndrome (overproduction of cortisol) and adrenal insufficiency (Addison’s disease). In development, the adrenal cortex is formed from the intermediate mesoderm. In adults, according to the centripetal migration model, glomerulosa cells undergo lineage conversion into zona fasciculata and reticularis cells and eventually apoptosis at the corticomedullary junction.

In this project, we aim to address two challenges in adrenocortical research and therapy: the lack of adequate disease models and the inadequate replacement of adrenocortical hormones in states of deficiency. Human adrenocortical cancer cell lines, for example H295R, have been used commonly as a model system for aldosterone production. However, these cells produce steroids that originate from multiple zones of the adrenal gland and lack the cellular organization that is characteristic of the adrenal cortex in vivo. Moreover, animal models, especially rodents, are not ideal models for studying adrenal pathology due to differences in the development, structure and physiology between humans and rodents. From a clinical perspective, the conventional therapy for patients with adrenal insufficiency is hormone replacement, which is far from optimal because it does not mimic the circadian (morning peak) and stressinduced pattern of cortisol secretion; this could be addressed by the transplantation of cells or organoids that produce hormones in a physiological fashion.

Since the first generation of human iPSCs, a variety of cell lineages and several complex mini-organs (intestinal, brain and kidney organoids) have been generated. However, adrenal organoids so far have not been reported. We here propose to develop human induced pluripotent stem cell (hiPSC)-derived functional adrenal lineages and adrenocortical organoids, which can be used to model adrenal disorders and potentially for future cell- and organoid-based therapy. In prior studies, the main activator used to convert fate from adult and pluripotent stem cells to steroidogenic cells was NR5A1 (encoding steroidogenic factor 1 or SF-1), which not only plays a role in adrenal, but also in gonadal development. Although hiPSCs have been differentiated into steroid-producing cells (Sonoyama et al. Endocrinology 2012), these cells lacked expression of CYP11B2, encoding aldosterone synthase. As part of the Einstein Kickbox, we optimized differentiation of hiPSCs towards intermediate mesoderm. We will now investigate expression profiles on the single-cell level in adrenal development to identify candidate differentiation conditions from intermediate mesoderm towards adrenocortical cells beyond the established SF-1. We will optimize differentiation conditions in order to obtain cells that represent the three zones of the adrenal cortex, followed by adrenal organoid generation using defined niche factors and an appropriate extracellular matrix, and subsequently examined in vitro as a functional mini-organ. As a proof-of-principle experiment, we propose to repair a disease-causing CLCN2 mutation in iPSCs generated from individuals with familial hyperaldosteronism (Scholl et al. Nat Genect 2018). Adrenal cells and / or organoids will be generated in parallel from iPSCs in which the mutation has been repaired as well as their isogenic controls. Subsequently, the generated adrenal organoids will serve as adequate tissue models to investigate the mechanism of primary aldosteronism. Similar models of other adrenocortical diseases can be used in the future to screen for drugs as personalized interventions, a completely novel approach. For rare genetic diseases of the adrenal cortex, future autotransplantation strategies would be highly promising.

Team: Kieu Nhi Tran Vo, Sun Jun Oh, Valeria Fernandez Vallone

Funding: Einstein Kickbox - Young Scientists & ECRT Consumable Grant

Native human joint model for in-vitro toxicologic screening

OA is the most prevalent disease of the human joint and has a massive socioeconomic impact. There is an extensive need for medical innovations which stop and/or reverse the cartilage degeneration. Focal cartilage defects can be regenerated with autologous chondrocyte transplantation – a technology developed by the laboratory for tissue engineering. For disseminated OA however, the only “curative” treatment available is the surgical implantation of an endoprosthesis. Intraarticular injections of corticosteroids are known for lowering inflammation for a limited amount of time – but do not slow down disease progression. The Laboratory for Tissue Engineering is currently developing a regenerative therapy based on mesenchymal stromal cells (MSCs), which can be applied in all stages of OA. The therapy aims to recruit MSCs into the joint by intraarticular injections of the chemokine CCL25 - which then create a regenerative micro-environment for the damaged cartilage. In an OA-animal model, 20% decreased cartilage degeneration was observed after 4 weeks of subsequent CCL25 injections.

Unfortunately, very few is known about adverse effects of chemokine signaling. An injection bares completely unknown risks and as research in cytostatic drugs showed, the effects of drugs on cells in monolayer culture are highly incongruent to effects in-vivo. When talking about effects, the target cell always has to be regarded in the context of the tissue it is embedded in. Extracellular matrix and adjacent cells heavily influence cell behavior – from receptor expression to migration pattern. Hence, models are required which represent the native tissue situation as well as possible. This may lower the time and resources spent before a go/no-go decision is reached - as well as reducing the number of required experiments on animals. Thus, this project aims to build a model of the human joint from native human tissue to expose the target cells with the therapeutic agent – effectively screening for any form of undesired tissue behavior. The model shall consist of tissue slices from living human joint tissue which is resected during endoprosthesis implantation. These will be cultivated in hanging inserts for three consecutive weeks und after establishment exposed to chemokines to assess any histologic disintegrity and inflammatory reactions. The project is designated to make use of Charité exceptionally good relation between clinic and research and establish live tissue culture long-term for other subjects of research in this field.

So far, we determined a high viability of the tissue slice over a 3-week time span using biochemichal and optical approaches Histological analysis showed highly conserved tissue integrity and cartilage-typical cell behavior. Next milestones will be final characterization of the model by testing its reactivity towards externally applied stimuli - assessing tissue slices feasibility as an advanced in vitro model for screening adverse effects of innovative therapeutics. Finally, we aim to stimulate the osteochondral tissue with CCL25 creating a more informed a go/no-go decision to go forward with the development of the therapy.

Team: Jacob Spinnen, Michael Sittinger, Tilo Dehne, Anja Kühl, Hendrik Mei

Funding: Einstein Kickbox - Young Scientists & ECRT Consumable Grant

Culturing human B cells in a 3D microfluidic bone marrow device

B cells are challenging to study; complex to culture and thus it is virtually impossible to follow their maturation path in vitro. This is mainly due to difficulties accessing their site of origin: the bone marrow (BM). Information on B cell development is mainly available from mouse and rat studies. However, in recent years it became clear that murine models are insufficient to mimic B cell biology in humans. Differences such as the role of BCR signaling; along with ethical concerns make animal studies in B cell biology questionable. Thus, the aim of the projected work is the development of a reliable human BM system which could hold the potential to study B cells close to an in vivo - situation. In a previous project we developed a 3D bone/BM organ-on-a-chip system. In the course of this project, by chance, we found that the system maintained a CD19 + (B-lymphocyte antigen) cell population over a time course of 28 days. We will further investigate the subpopulations in depth using flow cytometry and mass cytometry (CyTOF) and fine-tune the microenvironment to ultimately establish a selfrenewing, stable and heterogeneous B cell population. If successful, the benefits in terms of the models’ applicability in basic research are pronounced. The model could give completely new insights into B cell development in humans. In addition, the possibilities of translating it into therapeutic approaches (culturing B cells for clinical use) and for commercialization are immense.

Team: Melanie Ort, Janosch Schoon, Christine Consentius, Alessandro Camoneschi, Sven Geißler, Anastasia Rakow

Funding: Einsten Kickbox - Young Scientists & ECRT Consumable Grant

Visualization of the endogenous BMP receptor ALK2 in endothelial cells using CRISPR/Cas9 and induced pluripotent stem cell technology (iPSC-GenEd)

Generation of induced pluripotent stem cells (iPSCs) from somatic cells offers a great opportunity to study basic mechanisms in development, adult tissue homeostasis and to model human disease in vitro. Genome editing in iPSCs is a powerful investigative technique, which enables to introduce precise changes in the genome.

As information about the nanoscale localization and function of BMP receptors is still lacking due to absence of functional specific antibodies, we aim to apply CRISPR/Cas9 genome editing to address a major unsolved question in the research field of growth factor signaling.

To address these questions, we aim to generate genetically engineered human iPSC lines, containing an endogenously GFP tagged BMP type I receptor ALK2 to enable direct receptor visualization.
Mutations in the ALK2 gene cause the rare disorder Fibrodysplasia ossificans progressive (FOP), which leads to severe extra skeletal, heterotopic ossification (HO). Shedding light of the subcellular localization of ALK2 will accelerate the understanding of BMP receptor biology and will be beneficial for the development of more targeted treatment strategies for FOP.

Thus, we will use iPSCs from FOP patients and healthy control donors in order to generate WT and FOP ALK2-GFP iPSC lines to solve the question about the subcellular function of the mutant and WT ALK2 receptor. A genetically engineered FOP iPSC line enables the differentiation to disease relevant cell types, such as vascular endothelial cells (ECs), which contribute to HO via endothelial to mesenchymal transition (EndMT). Elucidating a possible disease-related mislocalization and/or lateral mobility of ALK2 in ECs by super resolution microscopy may be a novel approach for therapeutic intervention.

Team: Susanne Hildebrandt, Petra Knaus, Harald Stachelscheid, Jacob Piehler

Funding: Einstein Kickbox - Young Scientists & ECRT Consumable Grant

 

ECRT Consumable Grants - Förderperiode 2018

Decoding the aged matrix - consequences of cellular senescence for tissue patterning and stem cell invasion

Bone fractures are one of the most frequent orthopedic problems requiring medical treatment. Facing an aging population, specifically in the western world, a better understanding of how bone regeneration processes are altered with age is essential to develop novel treatment strategies satisfying the clinical need. We have recently developed a toolbox to simulate and study extracellular matrix (ECM) formation in vitro by cultivating primary human fibroblasts inside 3D macroporous collagen scaffolds. We propose that under compromised conditions such as aging, these early tissue formation processes are altered (e.g. by ECM secretion pattern, ROS level, cellular tension) with further consequences for stem- and progenitor cell migration, proliferation and differentiation.

Team:  Erik Brauer, Sophie Schreivogel, Daniela Mau, Uwe Kornak, Ansgar Petersen

Funding Scheme: Einstein Kickbox - Young Scientists & ECRT Consumable Grant

Regenerative potential of adipose tissue - Unraveling the crosstalk between adipocytes and hypertrophic scar tissue

Our project is based on observations from plastic surgery: autologous fat grafts lead to a significant reduction of scar tissue. We will attempt to identify the fat components responsible for this phenomenon, as well as the molecular pathways they work within. Therefore, we will study the interactions between myofibroblasts, a cell type already known to play a crucial role in wound healing, adipocytes and the adipocyte secretome. Since wound healing processes include changes to the extracellular matrix, we will analyze matrix remodeling alongside changes to the intracellular signaling of myofibroblasts cultured in 2D and 3D. Ideally, we would like to incorporate a pathological phenotype into our studies. To this end, a research stay in the group of Susan Gibbs, Vrije University Amsterdam, has been organized during the Kickbox period, during which we will work with fibroblasts isolated from hypertrophic scars. 

TeamKatharina Hörst, Uwe von Fritschen, Nan Ma, Susan Gibbs, Sarah Hedtrich

Funding Scheme: Einstein Kickbox - Young Scientists & ECRT Consumable Grant

Role of mechanics and tissue architecture in pancreatic cancer and post-operative complications

Team: Evi Lippens, Rosa Schmuck, Amaia Cipitria, Dag Wulsten, Daniela Garske

Funding Scheme: Einstein Kickbox - Young Scientists & ECRT Consumable Grant