@article{SchuetzeRoehringVorlovaetal.2015, author = {Sch{\"u}tze, Friedrich and R{\"o}hring, Florian and Vorlov{\´a}, Sandra and G{\"a}tzner, Sabine and Kuhn, Anja and Erg{\"u}n, S{\"u}leyman and Henke, Erik}, title = {Inhibition of lysyl oxidases improves drug diffusion and increases efficacy of cytotoxic treatment in 3D tumor models}, series = {Scientific Reports}, volume = {5}, journal = {Scientific Reports}, number = {17576}, doi = {10.1038/srep17576}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-145109}, year = {2015}, abstract = {Tumors are characterized by a rigid, highly cross-linked extracellular matrix (ECM), which impedes homogeneous drug distribution and potentially protects malignant cells from exposure to therapeutics. Lysyl oxidases are major contributors to tissue stiffness and the elevated expression of these enzymes observed in most cancers might influence drug distribution and efficacy. We examined the effect of lysyl oxidases on drug distribution and efficacy in 3D in vitro assay systems. In our experiments elevated lysyl oxidase activity was responsible for reduced drug diffusion under hypoxic conditions and consequently impaired cytotoxicity of various chemotherapeutics. This effect was only observed in 3D settings but not in 2D-cell culture, confirming that lysyl oxidases affect drug efficacy by modification of the ECM and do not confer a direct desensitizing effect. Both drug diffusion and efficacy were strongly enhanced by inhibition of lysyl oxidases. The results from the in vitro experiments correlated with tumor drug distribution in vivo, and predicted response to therapeutics in murine tumor models. Our results demonstrate that lysyl oxidase activity modulates the physical barrier function of ECM for small molecule drugs influencing their therapeutic efficacy. Targeting this process has the potential to significantly enhance therapeutic efficacy in the treatment of malignant diseases.}, language = {en} } @article{HorderGuazaLasherasGrummeletal.2021, author = {Horder, Hannes and Guaza Lasheras, Mar and Grummel, Nadine and Nadernezhad, Ali and Herbig, Johannes and Erg{\"u}n, S{\"u}leyman and Teßmar, J{\"o}rg and Groll, J{\"u}rgen and Fabry, Ben and Bauer-Kreisel, Petra and Blunk, Torsten}, title = {Bioprinting and differentiation of adipose-derived stromal cell spheroids for a 3D breast cancer-adipose tissue model}, series = {Cells}, volume = {10}, journal = {Cells}, number = {4}, doi = {10.3390/cells10040803}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-236496}, year = {2021}, abstract = {Biofabrication, including printing technologies, has emerged as a powerful approach to the design of disease models, such as in cancer research. In breast cancer, adipose tissue has been acknowledged as an important part of the tumor microenvironment favoring tumor progression. Therefore, in this study, a 3D-printed breast cancer model for facilitating investigations into cancer cell-adipocyte interaction was developed. First, we focused on the printability of human adipose-derived stromal cell (ASC) spheroids in an extrusion-based bioprinting setup and the adipogenic differentiation within printed spheroids into adipose microtissues. The printing process was optimized in terms of spheroid viability and homogeneous spheroid distribution in a hyaluronic acid-based bioink. Adipogenic differentiation after printing was demonstrated by lipid accumulation, expression of adipogenic marker genes, and an adipogenic ECM profile. Subsequently, a breast cancer cell (MDA-MB-231) compartment was printed onto the adipose tissue constructs. After nine days of co-culture, we observed a cancer cell-induced reduction of the lipid content and a remodeling of the ECM within the adipose tissues, with increased fibronectin, collagen I and collagen VI expression. Together, our data demonstrate that 3D-printed breast cancer-adipose tissue models can recapitulate important aspects of the complex cell-cell and cell-matrix interplay within the tumor-stroma microenvironment}, language = {en} } @article{KleefeldtBoemmelBroedeetal.2019, author = {Kleefeldt, Florian and B{\"o}mmel, Heike and Broede, Britta and Thomsen, Michael and Pfeiffer, Verena and W{\"o}rsd{\"o}rfer, Philipp and Karnati, Srikanth and Wagner, Nicole and Rueckschloss, Uwe and Erg{\"u}n, S{\"u}leyman}, title = {Aging-related carcinoembryonic antigen-related cell adhesion molecule 1 signaling promotes vascular dysfunction}, series = {Aging Cell}, volume = {2019}, journal = {Aging Cell}, number = {18}, doi = {10.1111/acel.13025}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-201231}, pages = {e13025}, year = {2019}, abstract = {Aging is an independent risk factor for cardiovascular diseases and therefore of particular interest for the prevention of cardiovascular events. However, the mechanisms underlying vascular aging are not well understood. Since carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is crucially involved in vascular homeostasis, we sought to identify the role of CEACAM1 in vascular aging. Using human internal thoracic artery and murine aorta, we show that CEACAM1 is upregulated in the course of vascular aging. Further analyses demonstrated that TNF-α is CEACAM1-dependently upregulated in the aging vasculature. Vice versa, TNF-α induces CEACAM1 expression. This results in a feed-forward loop in the aging vasculature that maintains a chronic pro-inflammatory milieu. Furthermore, we demonstrate that age-associated vascular alterations, that is, increased oxidative stress and vascular fibrosis, due to increased medial collagen deposition crucially depend on the presence of CEACAM1. Additionally, age-dependent upregulation of vascular CEACAM1 expression contributes to endothelial barrier impairment, putatively via increased VEGF/VEGFR-2 signaling. Consequently, aging-related upregulation of vascular CEACAM1 expression results in endothelial dysfunction that may promote atherosclerotic plaque formation in the presence of additional risk factors. Our data suggest that CEACAM1 might represent an attractive target in order to delay physiological aging and therefore the transition to vascular disorders such as atherosclerosis.}, language = {en} } @article{JordanJaeckleScheidtetal.2021, author = {Jordan, Martin C. and J{\"a}ckle, Veronika and Scheidt, Sebastian and Gilbert, Fabian and H{\"o}lscher-Doht, Stefanie and Erg{\"u}n, S{\"u}leyman and Meffert, Rainer H. and Heintel, Timo M.}, title = {Trans-obturator cable fixation of open book pelvic injuries}, series = {Scientific Reports}, volume = {11}, journal = {Scientific Reports}, number = {1}, doi = {10.1038/s41598-021-92755-2}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-261212}, year = {2021}, abstract = {Operative treatment of ruptured pubic symphysis by plating is often accompanied by complications. Trans-obturator cable fixation might be a more reliable technique; however, have not yet been tested for stabilization of ruptured pubic symphysis. This study compares symphyseal trans-obturator cable fixation versus plating through biomechanical testing and evaluates safety in a cadaver experiment. APC type II injuries were generated in synthetic pelvic models and subsequently separated into three different groups. The anterior pelvic ring was fixed using a four-hole steel plate in Group A, a stainless steel cable in Group B, and a titan band in Group C. Biomechanical testing was conducted by a single-leg-stance model using a material testing machine under physiological load levels. A cadaver study was carried out to analyze the trans-obturator surgical approach. Peak-to-peak displacement, total displacement, plastic deformation and stiffness revealed a tendency for higher stability for trans-obturator cable/band fixation but no statistical difference to plating was detected. The cadaver study revealed a safe zone for cable passage with sufficient distance to the obturator canal. Trans-obturator cable fixation has the potential to become an alternative for symphyseal fixation with less complications.}, language = {en} } @article{JordanBroeerFischeretal.2022, author = {Jordan, Martin C. and Br{\"o}er, David and Fischer, Christian and Heilig, Philipp and Gilbert, Fabian and H{\"o}lscher-Doht, Stefanie and Kalogirou, Charis and Popp, Kevin and Grunz, Jan-Peter and Huflage, Henner and Jakubietz, Rafael G. and Erg{\"u}n, S{\"u}leyman and Meffert, Rainer H.}, title = {Development and preclinical evaluation of a cable-clamp fixation device for a disrupted pubic symphysis}, series = {Communications Medicine}, volume = {2}, journal = {Communications Medicine}, number = {1}, doi = {10.1038/s43856-022-00227-z}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-299800}, year = {2022}, abstract = {Background Traumatic separation of the pubic symphysis can destabilize the pelvis and require surgical fixation to reduce symphyseal gapping. The traditional approach involves open reduction and the implantation of a steel symphyseal plate (SP) on the pubic bone to hold the reposition. Despite its widespread use, SP-fixation is often associated with implant failure caused by screw loosening or breakage. Methods To address the need for a more reliable surgical intervention, we developed and tested two titanium cable-clamp implants. The cable served as tensioning device while the clamp secured the cable to the bone. The first implant design included a steel cable anterior to the pubic symphysis to simplify its placement outside the pelvis, and the second design included a cable encircling the pubic symphysis to stabilize the anterior pelvic ring. Using highly reproducible synthetic bone models and a limited number of cadaver specimens, we performed a comprehensive biomechanical study of implant stability and evaluated surgical feasibility. Results We were able to demonstrate that the cable-clamp implants provide stability equivalent to that of a traditional SP-fixation but without the same risks of implant failure. We also provide detailed ex vivo evaluations of the safety and feasibility of a trans-obturator surgical approach required for those kind of fixation. Conclusion We propose that the developed cable-clamp fixation devices may be of clinical value in treating pubic symphysis separation.}, language = {en} } @article{HenkeNandigamaErguen2020, author = {Henke, Erik and Nandigama, Rajender and Erg{\"u}n, S{\"u}leyman}, title = {Extracellular matrix in the tumor microenvironment and its impact on cancer therapy}, series = {Frontiers in Molecular Biosciences}, volume = {6}, journal = {Frontiers in Molecular Biosciences}, number = {160}, issn = {2296-889X}, doi = {10.3389/fmolb.2019.00160}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-199341}, year = {2020}, abstract = {Solid tumors are complex organ-like structures that consist not only of tumor cells but also of vasculature, extracellular matrix (ECM), stromal, and immune cells. Often, this tumor microenvironment (TME) comprises the larger part of the overall tumor mass. Like the other components of the TME, the ECM in solid tumors differs significantly from that in normal organs. Intratumoral signaling, transport mechanisms, metabolisms, oxygenation, and immunogenicity are strongly affected if not controlled by the ECM. Exerting this regulatory control, the ECM does not only influence malignancy and growth of the tumor but also its response toward therapy. Understanding the particularities of the ECM in solid tumor is necessary to develop approaches to interfere with its negative effect. In this review, we will also highlight the current understanding of the physical, cellular, and molecular mechanisms by which the pathological tumor ECM affects the efficiency of radio-, chemo-, and immunotherapy. Finally, we will discuss the various strategies to target and modify the tumor ECM and how they could be utilized to improve response to therapy.}, language = {en} } @article{KleefeldtUpcinBoemmeletal.2022, author = {Kleefeldt, Florian and Upcin, Berin and B{\"o}mmel, Heike and Schulz, Christian and Eckner, Georg and Allmanritter, Jan and Bauer, Jochen and Braunger, Barbara and Rueckschloss, Uwe and Erg{\"u}n, S{\"u}leyman}, title = {Bone marrow-independent adventitial macrophage progenitor cells contribute to angiogenesis}, series = {Cell Death \& Disease}, volume = {13}, journal = {Cell Death \& Disease}, number = {3}, doi = {10.1038/s41419-022-04605-2}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-299724}, year = {2022}, abstract = {Pathological angiogenesis promotes tumor growth, metastasis, and atherosclerotic plaque rupture. Macrophages are key players in these processes. However, whether these macrophages differentiate from bone marrow-derived monocytes or from local vascular wall-resident stem and progenitor cells (VW-SCs) is an unresolved issue of angiogenesis. To answer this question, we analyzed vascular sprouting and alterations in aortic cell populations in mouse aortic ring assays (ARA). ARA culture leads to the generation of large numbers of macrophages, especially within the aortic adventitia. Using immunohistochemical fate-mapping and genetic in vivo-labeling approaches we show that 60\% of these macrophages differentiate from bone marrow-independent Ly6c\(^{+}\)/Sca-1\(^{+}\) adventitial progenitor cells. Analysis of the NCX\(^{-/-}\) mouse model that genetically lacks embryonic circulation and yolk sac perfusion indicates that at least some of those progenitor cells arise yolk sac-independent. Macrophages represent the main source of VEGF in ARA that vice versa promotes the generation of additional macrophages thereby creating a pro-angiogenetic feedforward loop. Additionally, macrophage-derived VEGF activates CD34\(^{+}\) progenitor cells within the adventitial vasculogenic zone to differentiate into CD31\(^{+}\) endothelial cells. Consequently, depletion of macrophages and VEGFR2 antagonism drastically reduce vascular sprouting activity in ARA. In summary, we show that angiogenic activation induces differentiation of macrophages from bone marrow-derived as well as from bone marrow-independent VW-SCs. The latter ones are at least partially yolk sac-independent, too. Those VW-SC-derived macrophages critically contribute to angiogenesis, making them an attractive target to interfere with pathological angiogenesis in cancer and atherosclerosis as well as with regenerative angiogenesis in ischemic cardiovascular disorders.}, language = {en} } @article{KarnatiSeimetzKleefeldtetal.2021, author = {Karnati, Srikanth and Seimetz, Michael and Kleefeldt, Florian and Sonawane, Avinash and Madhusudhan, Thati and Bachhuka, Akash and Kosanovic, Djuro and Weissmann, Norbert and Kr{\"u}ger, Karsten and Erg{\"u}n, S{\"u}leyman}, title = {Chronic Obstructive Pulmonary Disease and the Cardiovascular System: Vascular Repair and Regeneration as a Therapeutic Target}, series = {Frontiers in Cardiovascular Medicine}, volume = {8}, journal = {Frontiers in Cardiovascular Medicine}, issn = {2297-055X}, doi = {10.3389/fcvm.2021.649512}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-235631}, year = {2021}, abstract = {Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity and mortality worldwide and encompasses chronic bronchitis and emphysema. It has been shown that vascular wall remodeling and pulmonary hypertension (PH) can occur not only in patients with COPD but also in smokers with normal lung function, suggesting a causal role for vascular alterations in the development of emphysema. Mechanistically, abnormalities in the vasculature, such as inflammation, endothelial dysfunction, imbalances in cellular apoptosis/proliferation, and increased oxidative/nitrosative stress promote development of PH, cor pulmonale, and most probably pulmonary emphysema. Hypoxemia in the pulmonary chamber modulates the activation of key transcription factors and signaling cascades, which propagates inflammation and infiltration of neutrophils, resulting in vascular remodeling. Endothelial progenitor cells have angiogenesis capabilities, resulting in transdifferentiation of the smooth muscle cells via aberrant activation of several cytokines, growth factors, and chemokines. The vascular endothelium influences the balance between vaso-constriction and -dilation in the heart. Targeting key players affecting the vasculature might help in the development of new treatment strategies for both PH and COPD. The present review aims to summarize current knowledge about vascular alterations and production of reactive oxygen species in COPD. The present review emphasizes on the importance of the vasculature for the usually parenchyma-focused view of the pathobiology of COPD.}, language = {en} } @article{DoganScheuringWagneretal.2021, author = {Dogan, Leyla and Scheuring, Ruben and Wagner, Nicole and Ueda, Yuichiro and Schmidt, Sven and W{\"o}rsd{\"o}rfer, Philipp and Groll, J{\"u}rgen and Erg{\"u}n, S{\"u}leyman}, title = {Human iPSC-derived mesodermal progenitor cells preserve their vasculogenesis potential after extrusion and form hierarchically organized blood vessels}, series = {Biofabrication}, volume = {13}, journal = {Biofabrication}, number = {4}, doi = {10.1088/1758-5090/ac26ac}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-254046}, year = {2021}, abstract = {Post-fabrication formation of a proper vasculature remains an unresolved challenge in bioprinting. Established strategies focus on the supply of the fabricated structure with nutrients and oxygen and either rely on the mere formation of a channel system using fugitive inks or additionally use mature endothelial cells and/or peri-endothelial cells such as smooth muscle cells for the formation of blood vessels in vitro. Functional vessels, however, exhibit a hierarchical organization and multilayered wall structure that is important for their function. Human induced pluripotent stem cell-derived mesodermal progenitor cells (hiMPCs) have been shown to possess the capacity to form blood vessels in vitro, but have so far not been assessed for their applicability in bioprinting processes. Here, we demonstrate that hiMPCs, after formulation into an alginate/collagen type I bioink and subsequent extrusion, retain their ability to give rise to the formation of complex vessels that display a hierarchical network in a process that mimics the embryonic steps of vessel formation during vasculogenesis. Histological evaluations at different time points of extrusion revealed the initial formation of spheres, followed by lumen formation and further structural maturation as evidenced by building a multilayered vessel wall and a vascular network. These findings are supported by immunostainings for endothelial and peri-endothelial cell markers as well as electron microscopic analyses at the ultrastructural level. Moreover, endothelial cells in capillary-like vessel structures deposited a basement membrane-like matrix at the basal side between the vessel wall and the alginate-collagen matrix. After transplantation of the printed constructs into the chicken chorioallantoic membrane (CAM) the printed vessels connected to the CAM blood vessels and get perfused in vivo. These results evidence the applicability and great potential of hiMPCs for the bioprinting of vascular structures mimicking the basic morphogenetic steps of de novo vessel formation during embryogenesis.}, language = {en} } @article{LuetkensErguenHuflageetal.2021, author = {Luetkens, Karsten Sebastian and Erg{\"u}n, S{\"u}leyman and Huflage, Henner and Kunz, Andreas Steven and Gietzen, Carsten Herbert and Conrads, Nora and Pennig, Lenhard and Goertz, Lukas and Bley, Thorsten Alexander and Gassenmaier, Tobias and Grunz, Jan-Peter}, title = {Dose reduction potential in cone-beam CT imaging of upper extremity joints with a twin robotic x-ray system}, series = {Scientific Reports}, volume = {11}, journal = {Scientific Reports}, number = {1}, doi = {10.1038/s41598-021-99748-1}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-270429}, year = {2021}, abstract = {Cone-beam computed tomography is a powerful tool for 3D imaging of the appendicular skeleton, facilitating detailed visualization of bone microarchitecture. This study evaluated various combinations of acquisition and reconstruction parameters for the cone-beam CT mode of a twin robotic x-ray system in cadaveric wrist and elbow scans, aiming to define the best possible trade-off between image quality and radiation dose. Images were acquired with different combinations of tube voltage and tube current-time product, resulting in five scan protocols with varying volume CT dose indices: full-dose (FD; 17.4 mGy), low-dose (LD; 4.5 mGy), ultra-low-dose (ULD; 1.15 mGy), modulated low-dose (mLD; 0.6 mGy) and modulated ultra-low-dose (mULD; 0.29 mGy). Each set of projection data was reconstructed with three convolution kernels (very sharp [Ur77], sharp [Br69], intermediate [Br62]). Five radiologists subjectively assessed the image quality of cortical bone, cancellous bone and soft tissue using seven-point scales. Irrespective of the reconstruction kernel, overall image quality of every FD, LD and ULD scan was deemed suitable for diagnostic use in contrast to mLD (very sharp/sharp/intermediate: 60/55/70\%) and mULD (0/3/5\%). Superior depiction of cortical and cancellous bone was achieved in FD\(_{Ur77}\) and LD\(_{Ur77}\) examinations (p < 0.001) with LD\(_{Ur77}\) scans also providing favorable bone visualization compared to FD\(_{Br69}\) and FD\(_{Br62}\) (p < 0.001). Fleiss' kappa was 0.618 (0.594-0.641; p < 0.001), indicating substantial interrater reliability. In this study, we demonstrate that considerable dose reduction can be realized while maintaining diagnostic image quality in upper extremity joint scans with the cone-beam CT mode of a twin robotic x-ray system. Application of sharper convolution kernels for image reconstruction facilitates superior display of bone microarchitecture.}, language = {en} }