Network


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

Hotspot


Dive into the research topics where Nicole Krott is active.

Publication


Featured researches published by Nicole Krott.


Tissue Engineering | 2002

Efficient Transfection Method for Primary Cells

Astrid Hamm; Nicole Krott; Ines Breibach; Rüdiger Blindt; Anja K. Bosserhoff

Transfection of primary cells and stem cells is a problem in the laboratory routine and further in tissue engineering and gene therapy. Most methods working effectively for cell lines in culture fail to transfect primary cells. Here we describe the use of the Nucleofector technology developed by amaxa biosystems. We were able to transfect primary human melanocytes, human coronary smooth muscle cells, human chondrocytes, and human mesenchymal stem cells with high efficiencies (28.9-45.3%). All primary cell types failed to be transfected satisfactorily by methods based on liposome-mediated transfection in our hands. The viability of the transfected cells varied between 11.2% and 75% in comparison to untreated cells. Only 200,000 cells per transfection sample were needed. In summary, this method presents an effective and fast mean for transfection of primary and stem cells demonstrated by four cell types which are only transfected with low efficiency by other methods.


Journal of Vascular Research | 2002

Characterization of Differential Gene Expression in Quiescent and Invasive Human Arterial Smooth Muscle Cells

Rüdiger Blindt; Felix Vogt; Daniel Lamby; Ute Zeiffer; Nicole Krott; Kristina Hilger-Eversheim; Peter Hanrath; Jürgen vom Dahl; Anja-Katrin Bosserhoff

Proliferation, migration and invasion of smooth muscle cells (SMCs) are essential pathogenic processes in the development of a broad spectrum of cardiovascular disorders, like arteriosclerosis, restenosis after percutaneous transluminal angioplasty and stent implantation as well as transplant vessel disease. As an in vitro model mimicking these processes, the Boyden chamber was employed to characterize the diverging migratory and invasive potentials of proliferating and nonproliferating human arterial SMCs (haSMCs). Using this model, differential gene expression of both phenotypes was analyzed by a cDNA array system (Clontech human cardiovascular array). With these arrays, 558 cardiovascular-associated genes could be compared. Further, gene expression was exactly quantified by real-time RT-PCR. Protein expression was analyzed by ELISA and Western blotting. In total, 47 genes were differentially expressed more than 1.5 times. Most of the differentially regulated genes in this study were associated with the extracellular matrix (ECM) and cell motility. In detail, the respective groups were matrix-organizing proteins, ECM proteins, cell adhesion proteins, extracellular communication and cytoskeleton motility proteins. Genes known to be differentially regulated during haSMC migration and invasion, like TIMP 2, TIMP 3, and MMP 3, were confirmed by the array data. Reduced expression of several cytoskeletal proteins, like vimentin, fibronectin, cytokeratins and β1 integrin, was shown in the invasive phenotype. Further, angio-associated protein, alpha E-catenin and atrial brain natriuretic peptide receptor were downregulated whereas TFPI 2 was strongly upregulated in invasive haSMCs. In conclusion, several relevant potential candidate genes for the quiescent and the invasive SMC phenotype were identified and genes already known to be differentially regulated by previous analysis were confirmed.


Cardiovascular Research | 2002

Upregulation of the cytoskeletal-associated protein Moesin in the neointima of coronary arteries after balloon angioplasty: a new marker of smooth muscle cell migration?

Rüdiger Blindt; Ute Zeiffer; Nicole Krott; Karsten Filzmaier; Meinolf Voss; Peter Hanrath; Jürgen vom Dahl; Anja-Katrin Bosserhoff

Migrating cells like coronary smooth muscle cells in restenosis change their cell shape and form cellular protrusions called filopodia. A prerequisite for filopodia formation is the rearrangement of the actin cytoskeleton. An essential role of the 78-kDa protein Moesin is described for Rho- and Rac-dependent assembly of actin filaments. In vivo Moesin is not observed in mature smooth muscle cells. The objective of this study was to demonstrate that Moesin is upregulated in migrating coronary smooth muscle cells during restenosis development. In vivo expression of Moesin was upregulated in neointimal coronary smooth muscle cells of dilated porcine coronary arteries compared to the undilated left circumflex coronary artery of the same swine. Concordant to these results Moesin expression was upregulated in migrating and invading human arterial smooth muscle cells in vitro analyzed by FACS, Western blotting and RT-PCR. In addition, the invasive potential of Moesin-positive Mel Im cells transfected with Moesin sense DNA increased by 28% as compared to mock-transfected control, whereas antisense transfected cells had a decreased invasive potential of 32%. Transfection of Moesin-negative HepG2 with Moesin sense cDNA increased the invasive potential by 43%. Finally, transfection of human arterial smooth muscle cells with Moesin sense cDNA caused an increased invasive potential of 30%. Transfection of haSMCs with antisense cDNA decreased the invasive potential by 37% in comparison to mock-transfected control. These results demonstrate for the first time an upregulation of Moesin expression in coronary smooth muscle cells of the neointima after arterial injury. The increased migrative and invasive potential of cells transfected with Moesin confirmed the functional role of Moesin in cell migration. This indicates an important role of Moesin during restenosis development.


Journal of the American College of Cardiology | 2008

Blockade of Angio-Associated Migratory Cell Protein Inhibits Smooth Muscle Cell Migration and Neointima Formation in Accelerated Atherosclerosis

Felix Vogt; Alma Zernecke; Marie E. Beckner; Nicole Krott; Anja-Katrin Bosserhoff; Rainer Hoffmann; Marc A.M.J. Zandvoort; Thomas Jahnke; Malte Kelm; Christian Weber; Rüdiger Blindt

OBJECTIVES The aim of this study was to elucidate the role of angio-associated migratory cell protein (AAMP) for the migration of vascular smooth muscle cells (SMCs) and for the development of neointimal hyperplasia after vascular injury. BACKGROUND Although AAMP has been shown to participate in angiogenesis and cancerogenesis and is predominantly expressed in cells with a migratory phenotype, involvement of AAMP during neointima (NI) formation after arterial injury has not been analyzed previously. METHODS The AAMP content in SMCs was examined using 2-photon laser-scanning microscopy and subcellular fractioning. Migratory potential of SMCs transiently transfected with AAMP expression vectors, transfected with small interfering ribonucleic acid (siRNA), or treated with antirecombinant angio-associated migratory cell protein-antibody (anti-rAAMP-ab) was examined using transwell migration chamber assays. Expression of AAMP was determined in the atherogenic apolipoprotein E knockout (apoE(-/-)) mouse model and in the porcine coronary restenosis model by immunohistochemistry and by Western blot. ApoE(-/-) mice were treated intraperitoneally with anti-rAAMP-ab, and wire-injured carotid arteries were examined. RESULTS Angio-associated migratory cell protein is localized in the membrane of SMCs, and its expression is enhanced in NI-derived SMCs. The AAMP overexpression increases, while both treatment with anti-rAAMP-ab and transfection with siRNA decreases SMC migration. Knockdown of AAMP decreases RhoA activity in the membrane fraction of SMCs. The AAMP expression by SMCs is enhanced in both animal models. Anti-rAAMP-ab reduces neointimal SMC density at 1 week and NI formation at 4 weeks in apoE(-/-) mice without affecting proliferation of SMCs. CONCLUSIONS These data reveal an important functional role of AAMP in the migration of SMCs, identifying AAMP as a potential target to limit lesion formation after injury.


Coronary Artery Disease | 2002

Decrease of vascular smooth muscle cell locomotion by abciximab, but not tirofiban: a possible role of different affinity to alpha v beta 3 integrins

Rüdiger Blindt; Anja-Katrin Bosserhoff; Nicole Krott; Felix Vogt; Peter Hanrath; Lütfü Demircan; Jürgen vom Dahl

AimThe EPISTENT and EPIC studies demonstrated a reduction of clinically driven re-interventions after percutaneous transluminal coronary angioplasty (PTCA) and stent implantation in patients treated with abciximab, while for tirofiban no similar effects could be demonstrated. This may be explained by the different effects on the migratory and invasive potential of vascular smooth muscle cells (VSMCs) by integrin alpha v beta 3 blockade. Therefore, the objective of this study was to compare the effectiveness of abciximab and tirofiban to affect VSMC migration and invasion. MethodsVascular smooth muscle cells were treated with abciximab (0.1–1 μg/ml), tirofiban (0.1–1 μg/ml), and the alpha v beta 3 specific antibody LM609 (1–5 μg/ml), that was used as a positive control during the assay (treatment) over 24 h before the assay (pre-treatment), or before and during the assay (combined treatment). Sodium 3′-[1-(phenylaminocarbonyl)-3,4-tetrazolium]-bis (4-methoxyy-6-nitro) benzene sulfonic acid (XTT)-assay and cell counting measured the influence of the substances on VSMC proliferation. Using a Boyden Chamber model, the capability of VSMCs for migration and invasion was tested with different chemo-attractants and barriers. ResultsAny influence of the platelet glycoprotein (GP) IIb/IIIa receptor (integrin alpha IIb beta 3) antagonists on VSMC proliferation could be excluded. After combined treatment, abciximab demonstrated a dose-dependent inhibition of migration (IC50 = 33 μg/ml) and invasion (IC50 = 0.5 μg/ml) of VSMCs. Administration during the assay without pre-treatment inhibited migration similarly (IC50 = 32 μg/ml) but invasion to a significant lower extent (IC50 = 44 μg/ml). Administration of tirofiban during the assay with or without pre-treatment had no inhibitory effect on VSMC migration and invasion. Pre-treatment alone with one of the substances also did not alter VSMC migration or invasion. ConclusionAbciximab administration in physiological concentrations was capable of significantly inhibiting the migratory and invasive potential of VSMCs, while for tirofiban no similar effect could be demonstrated.


Journal of the American College of Cardiology | 2013

TCT-824 Improved Safety And Efficacy Of A Novel Dual cRGD- And Everolimus-coated Coronary Stent Compared To Everolimus-eluting Stents In The Porcine Model

Felix Vogt; Rüdiger Blindt; Rainer Hoffmann; Nicole Krott; Nikolaus Marx

Drug-eluting stents markedly reduce restenosis; however, re-endothelialization is delayed. Integrin-binding cyclic Arg-Gly-Asp (cRGD) peptide-loaded stents offer potential for enhanced early and late endothelial recovery by endothelial progenitor cell (EPC) anchorage. Here, a novel dual cRGD and


Journal of Biomedical Materials Research Part B | 2012

Introduction of a high‐throughput double‐stent animal model for the evaluation of biodegradable vascular stents

Mauricio Borinski; Christian Flege; Fabian Schreiber; Nicole Krott; Thomas Gries; Elisa A. Liehn; Rüdiger Blindt; Nikolaus Marx; Felix Vogt

Current stent system efficacy for the treatment of coronary artery disease is hampered by in-stent restenosis (ISR) rates of up to 20% in certain high-risk settings and by the risk of stent thrombosis, which is characterized by a high mortality rate. In theory, biodegradable vascular devices exhibit crucial advantages. Most absorbable implant materials are based on poly-L-lactic acid (PLLA) owing to its mechanical properties; however, PLLA might induce an inflammatory reaction in the vessel wall. Evaluation of biodegradable implant efficacy includes a long-term examination of tissue response; therefore, a simple in vivo tool for thorough biocompatibility and biodegradation evaluation would facilitate future stent system development. Rats have been used for the study of in vivo degradation processes, and stent implantation into the abdominal aorta of rats is a proven model for stent evaluation. Here, we report the transformation of the porcine double-stent animal model into the high-throughput rat abdominal aorta model. As genetic manipulation of rats was introduced recently, this novel method presents a powerful tool for future in vivo biodegradable candidate stent biocompatibility and biodegradation characterization in a reliable simple model of coronary ISR.


Journal of the American College of Cardiology | 2006

A novel drug-eluting stent coated with an integrin-binding cyclic Arg-Gly-Asp peptide inhibits neointimal hyperplasia by recruiting endothelial progenitor cells

Rüdiger Blindt; Felix Vogt; Irina Astafieva; Christian Fach; Mihail Hristov; Nicole Krott; Berthold Seitz; Aphrodite Kapurniotu; Connie Kwok; Manfred Dewor; Anja-Katrin Bosserhoff; Jürgen Bernhagen; Peter Hanrath; Rainer Hoffmann; Christian Weber


European Heart Journal | 2004

Long-term assessment of a novel biodegradable paclitaxel-eluting coronary polylactide stent

Felix Vogt; Andreas Stein; Gösta Rettemeier; Nicole Krott; Rainer Hoffmann; Jürgen vom Dahl; Anja-Katrin Bosserhoff; Walter Michaeli; Peter Hanrath; Christian Weber; Rüdiger Blindt


Cardiovascular Research | 2004

Downregulation of N-cadherin in the neointima stimulates migration of smooth muscle cells by RhoA deactivation

Rüdiger Blindt; Anja-Katrin Bosserhoff; Julia Dammers; Nicole Krott; Lütfü Demircan; Rainer Hoffmann; Peter Hanrath; Christian Weber; Felix Vogt

Collaboration


Dive into the Nicole Krott's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Felix Vogt

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar

Anja-Katrin Bosserhoff

University of Erlangen-Nuremberg

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

Ute Zeiffer

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge