Network


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

Hotspot


Dive into the research topics where Sabine Neuss is active.

Publication


Featured researches published by Sabine Neuss.


Small | 2009

Gold Nanoparticles of Diameter 1.4 nm Trigger Necrosis by Oxidative Stress and Mitochondrial Damage

Yu Pan; Annika Leifert; David Ruau; Sabine Neuss; Jörg Bornemann; Günter Schmid; Wolfgang Brandau; Ulrich Simon; Willi Jahnen-Dechent

Gold nanoparticles (AuNPs) are generally considered nontoxic, similar to bulk gold, which is inert and biocompatible. AuNPs of diameter 1.4 nm capped with triphenylphosphine monosulfonate (TPPMS), Au1.4MS, are much more cytotoxic than 15-nm nanoparticles (Au15MS) of similar chemical composition. Here, major cell-death pathways are studied and it is determined that the cytotoxicity is caused by oxidative stress. Indicators of oxidative stress, reactive oxygen species (ROS), mitochondrial potential and integrity, and mitochondrial substrate reduction are all compromised. Genome-wide expression profiling using DNA gene arrays indicates robust upregulation of stress-related genes after 6 and 12 h of incubation with a 2 x IC50 concentration of Au1.4MS but not with Au15MS nanoparticles. The caspase inhibitor Z-VAD-fmk does not rescue the cells, which suggests that necrosis, not apoptosis, is the predominant pathway at this concentration. Pretreatment of the nanoparticles with reducing agents/antioxidants N-acetylcysteine, glutathione, and TPPMS reduces the toxicity of Au1.4MS. AuNPs of similar size but capped with glutathione (Au1.1GSH) likewise do not induce oxidative stress. Besides the size dependency of AuNP toxicity, ligand chemistry is a critical parameter determining the degree of cytotoxicity. AuNP exposure most likely causes oxidative stress that is amplified by mitochondrial damage. Au1.4MS nanoparticle cytotoxicity is associated with oxidative stress, endogenous ROS production, and depletion of the intracellular antioxidant pool.


Stem Cells | 2004

Functional expression of HGF and HGF receptor/c-met in adult human mesenchymal stem cells suggests a role in cell mobilization, tissue repair, and wound healing.

Sabine Neuss; Eva Becher; Michael Wöltje; Lothar Tietze; Willi Jahnen-Dechent

Human mesenchymal stem cells (hMSC) are adult stem cells with multipotent capacities. The ability of mesenchymal stem cells to differentiate into many cell types, as well as their high ex vivo expansion potential, makes these cells an attractive therapeutic tool for cell transplantation and tissue engineering. hMSC are thought to contribute to tissue regeneration, but the signals governing their mobilization, diapedesis into the bloodstream, and migration into the target tissue are largely unknown. Here we report that hepatocyte growth factor (HGF) and the cognate receptor HGFR/c‐met are expressed in hMSC, on both the RNA and the protein levels. The expression of HGF was downregulated by transforming growth factor beta. HGF stimulated chemotactic migration but not proliferation of hMSC. Therefore the HGF/c‐met signaling system may have an important role in hMSC recruitment sites of tissue regeneration. The controlled regulation of HGF/c‐met expression may be beneficial in tissue engineering and cell therapy employing hMSC.


Biomaterials | 2010

The osteogenic differentiation of adult bone marrow and perinatal umbilical mesenchymal stem cells and matrix remodelling in three-dimensional collagen scaffolds.

Rebekka K. Schneider; Andrea Puellen; Rafael Kramann; Kerstin Raupach; Jörg Bornemann; Ruth Knuechel; Alberto Perez-Bouza; Sabine Neuss

Adult human mesenchymal stem cells from bone marrow (BM-MSC) represent a promising source for skeletal regeneration. Perinatal MSC from Whartons Jelly of the umbilical cord (UC-MSC) are expected to possess enhanced differentiation capacities due to partial expression of pluripotency markers. For bone tissue engineering, it is important to analyse in vitro behaviour of stem cell/biomaterial hybrids concerning in vivo integration into injured tissue via migration, matrix remodelling and differentiation. This study compares the cell-mediated remodelling of three-dimensional collagen I/III gels during osteogenic differentiation of both cell types. When activated through collagen contact and subjected to osteogenic differentiation, UC-MSC differ from BM-MSC in expression and synthesis of extracellular matrix (ECM) proteins as shown by histology, immunohistochemistry, Western Blot analysis and realtime-RT-PCR. The biosynthetic activity was accompanied in both cell types by the ultrastructural appearance of hydroxyapatite/calcium crystals and osteogenic gene induction. Following secretion of matrix metalloproteinases (MMP), both MSC types migrated into and colonised the collagenous matrix causing matrix strengthening and contraction. These results indicate that UC-MSC and BM-MSC display all features needed for effective bone fracture healing. The expression of ECM differs in both cell types considerably, suggesting different mechanisms for bone formation and significant impact for bone tissue engineering.


BMC Neuroscience | 2009

Neural differentiation potential of human bone marrow-derived mesenchymal stromal cells: misleading marker gene expression.

Katrin Montzka; Nina Lassonczyk; Beate Tschöke; Sabine Neuss; Tobias Führmann; Rachelle Franzen; Ralf Smeets; Gary Brook; Michael Wöltje

BackgroundIn contrast to pluripotent embryonic stem cells, adult stem cells have been considered to be multipotent, being somewhat more restricted in their differentiation capacity and only giving rise to cell types related to their tissue of origin. Several studies, however, have reported that bone marrow-derived mesenchymal stromal cells (MSCs) are capable of transdifferentiating to neural cell types, effectively crossing normal lineage restriction boundaries. Such reports have been based on the detection of neural-related proteins by the differentiated MSCs. In order to assess the potential of human adult MSCs to undergo true differentiation to a neural lineage and to determine the degree of homogeneity between donor samples, we have used RT-PCR and immunocytochemistry to investigate the basal expression of a range of neural related mRNAs and proteins in populations of non-differentiated MSCs obtained from 4 donors.ResultsThe expression analysis revealed that several of the commonly used marker genes from other studies like nestin, Enolase2 and microtubule associated protein 1b (MAP1b) are already expressed by undifferentiated human MSCs. Furthermore, mRNA for some of the neural-related transcription factors, e.g. Engrailed-1 and Nurr1 were also strongly expressed. However, several other neural-related mRNAs (e.g. DRD2, enolase2, NFL and MBP) could be identified, but not in all donor samples. Similarly, synaptic vesicle-related mRNA, STX1A could only be detected in 2 of the 4 undifferentiated donor hMSC samples. More significantly, each donor sample revealed a unique expression pattern, demonstrating a significant variation of marker expression.ConclusionThe present study highlights the existence of an inter-donor variability of expression of neural-related markers in human MSC samples that has not previously been described. This donor-related heterogeneity might influence the reproducibility of transdifferentiation protocols as well as contributing to the ongoing controversy about differentiation capacities of MSCs. Therefore, further studies need to consider the differences between donor samples prior to any treatment as well as the possibility of harvesting donor cells that may be inappropriate for transplantation strategies.


Biomaterials | 2012

3D co-culture of hematopoietic stem and progenitor cells and mesenchymal stem cells in collagen scaffolds as a model of the hematopoietic niche.

Isabelle Leisten; Rafael Kramann; Mónica S. Ventura Ferreira; Manfred Bovi; Sabine Neuss; Patrick Ziegler; Wolfgang Wagner; Ruth Knüchel; Rebekka K. Schneider

Here, we propose a collagen-based three-dimensional (3D) environment for hematopoietic stem and progenitor cells (HPC) with mesenchymal stem cells (MSC) derived either from bone marrow (BM) or umbilical cord (UC), to recapitulate the main components of the BM niche. Mechanisms described for HPC homeostasis were systematically analyzed in comparison to the conventional liquid HPC culture. The 3D-cultivation allows dissecting two sub-populations of HPC: (I) HPC in suspension above the collagen gel and (II) migratory HPC in the collagen fibres of the collagen gel. The different sites represent distinct microenvironments with significant impact on HPC fate. HPC in niche I (suspension) are proliferative and a dynamic culture containing HPC (CD34(+)/CD38(-)), maturing myeloid cells (CD38(+), CD13(+), CAE(+)) and natural killer (NK) cells (CD56(+)). In contrast, HPC in niche II showed clonal growth with significant high levels of the primitive CD34(+)/CD38(-) phenotype with starting myeloid (CD13(+), CAE(+)) differentiation, resembling the endosteal part of the BM niche. In contrast, UC-MSC are not adequate for HSC expansion as they significantly enhance HPC proliferation and lineage commitment. In conclusion, the 3D-culture system using collagen and BM-MSC enables HPC expansion and provides a potential platform to dissect regulatory mechanisms in hematopoiesis.


Experimental Hematology | 2011

Synergistic effects of growth factors and mesenchymal stromal cells for expansion of hematopoietic stem and progenitor cells

Thomas Walenda; Gudrun Bokermann; Mónica S. Ventura Ferreira; Daniela Piroth; Thomas Hieronymus; Sabine Neuss; Martin Zenke; Anthony D. Ho; Albrecht M. Müller; Wolfgang Wagner

OBJECTIVE The number of hematopoietic stem and progenitor cells (HPCs) per cord blood unit is limited, and this can result in delayed engraftment or graft failure. In vitro expansion of HPCs provides a perspective to overcome these limitations. Cytokines as well as mesenchymal stromal cells (MSCs) have been shown to support HPCs ex vivo expansion, but a systematic analysis of their interplay remains elusive. MATERIALS AND METHODS Twenty different combinations of growth factors (stem cell factor [SCF], thrombopoietin [TPO], fibroblast growth factor-1 [FGF-1], angiopoietin-like 5, and insulin-like growth factor-binding protein 2), either with or without MSC coculture were systematically compared for their ability to support HPC expansion. CD34(+) cells were stained with carboxyfluorescein diacetate N-succinimidyl ester to monitor cell division history in conjunction with immunophenotype. Colony-forming unit frequencies and hematopoietic reconstitution of nonobese diabetic severe combined immunodeficient mice were also assessed. RESULTS Proliferation of HPCs was stimulated by coculture with MSCs. This was further enhanced in combination with SCF, TPO, and FGF-1. Moreover, these conditions maintained expression of primitive surface markers for more than four cell divisions. Colony-forming unit-initiating cells were not expanded without stromal support, whereas an eightfold increase was reached by simultaneous cytokine-treatment and MSC coculture. Importantly, in comparison to expansion without stromal support, coculture with MSCs significantly enhanced hematopoietic chimerism in a murine transplantation model. CONCLUSIONS The supportive effect of MSCs on hematopoiesis can be significantly increased by addition of specific recombinant growth factors; especially in combination with SCF, TPO, and FGF-1.


Cell Transplantation | 2008

Long-Term Survival and Bipotent Terminal Differentiation of Human Mesenchymal Stem Cells (hMSC) in Combination with a Commercially Available Three-Dimensional Collagen Scaffold:

Sabine Neuss; Rebekah Stainforth; Jochen Salber; P. Schenck; Manfred Bovi; Ruth Knüchel; Alberto Perez-Bouza

Researchers working in the field of tissue engineering ideally combine autologous cells and biocompatible scaffolds to replace defect tissues/organs. Due to their differentiation capacity, mesenchym-derived stem cells, such as human mesenchymal stem cells (hMSC), are a promising autologous cell source for the treatment of human diseases. As natural precursors for mesenchymal tissues, hMSC are particularly suitable for bone, cartilage, and adipose tissue replacement. In this study a detailed histological and ultrastructural analysis of long-term cultured and terminally differentiated hMSC on 3D collagen scaffolds was performed. Standardized 2D differentiation protocols for hMSC into adipocytes and osteoblasts were adapted for long-term 3D in vitro cultures in porous collagen matrices. After a 50-day culture period, large numbers of mature adipocytes and osteoblasts were clearly identifiable within the scaffolds. The adipocytes exhibited membrane free lipid vacuoles. The osteoblasts were arranged in close association with hydroxyapatite crystals, which were deposited on the surrounding fibers. The collagen matrix was remodeled and adopted a contracted and curved form. Human MSC survive long-term culture within these scaffolds and could be terminally differentiated into adipocytes and osteoblasts. Thus, the combination of hMSC and this particular collagen scaffold is a possible candidate for bone and adipose tissue replacement strategies.


Differentiation | 2010

Long-term survival and characterisation of human umbilical cord-derived mesenchymal stem cells on dermal equivalents.

Rebekka K. Schneider; Andrea Püllen; Rafael Kramann; Jörg Bornemann; Ruth Knüchel; Sabine Neuss; Alberto Perez-Bouza

During early embryogenesis, mesenchymal cells arise from the primitive epithelium and can revert to an epithelial phenotype by passing through mesenchymal-to-epithelial transition (MET). Mesenchymal stem cells (MSC) of the Whartons Jelly of the umbilical cord (UC-MSC) express pluripotency markers underlining their primitive developmental state. As mesenchymal stem cells from bone marrow (BM-MSC) possess a strong propensity to ameliorate mesenchymal tissue damage, UC-MSC might also be able to differentiate into cells apart from the mesoderm, allowing replacement of ectodermal and mesodermal tissues. In this study, we analysed the possible epidermal differentiation of UC-MSC on dermal equivalents (DEs) consisting of collagen I/III with dermal fibroblasts and subjected to the culture conditions for tissue engineering of skin with keratinocytes. The culture conditions were further modified by pre-treating the cells with 5-azacytidine or by supplementing the medium with all trans retinoic acid. Interestingly, a subpopulation of UC-MSC (29%) co-expressed pan-cytokeratin (epithelial marker; pan-CK) and vimentin (mesenchymal marker) after isolation. Under the three-dimensional conditions of skin, the number of pan-CK(+)-cells increased to >30% after 21 days of cultivation, while under osteogenic culture conditions the cells were pan-CK-negative, thus showing the influence of the artificial niche. Nevertheless, the pan-CK-expression was neither accompanied by typical epithelial morphology nor expression of other epidermal markers. The pan-CK-detection can be explained by the expression of cytokeratins in myofibroblasts. UC-MSC expressed alpha-smooth muscle actin after isolation and displayed all features of functional myofibroblasts like morphology, cell-mediated contraction of a collagen gel and production of components of the extracellular matrix (ECM). The treatment with all trans retinoic acid or 5-azacytidine could neither induce an epidermal differentiation nor enhance the myofibroblastic differentiation. Concluding, UC-MSC might be an interesting cell source to support the regeneration of wounds by their differentiation into myofibroblasts and their extensive synthesis of ECM components.


Biomaterials | 2010

The role of biomaterials in the direction of mesenchymal stem cell properties and extracellular matrix remodelling in dermal tissue engineering

Rebekka K. Schneider; Julia Anraths; Rafael Kramann; Jörg Bornemann; Manfred Bovi; Ruth Knüchel; Sabine Neuss

Recently, a new generation of dermal equivalents (DE) was presented which are solely generated on a human fibroblast-derived dermal matrix. Since human mesenchymal stem cells from bone marrow (BM-MSC) and Whartons Jelly of the umbilical cord (UC-MSC) are characterised by a distinct biosynthetic and paracrine activity, they are an appealing alternative approach for generating cell-based DE. This study compares the epithelial-mesenchymal interaction and extracellular matrix (ECM) remodelling of cell-based and collagen-based DE using fibroblasts, BM-MSC or UC-MSC, respectively, in co-culture with the keratinocyte cell line HaCaT. While fibroblast-based DE exhibit normal matrix synthesis, proliferation and differentiation of keratinocytes, mesenchymal stem cell-based DE resulted in excessive production of inhomogenous matrix aggregates, loss of polarisation of the epidermal cell layer and an inconstant paracrine activity. In contrast, collagen-embedded MSC revealed a homogenous growth pattern as well as a constant expression of growth factors and ECM proteins without a negative influence on the epidermal layer as shown by histology, electron microscopy, immunohistochemistry and realtime-RT-PCR. These results indicate the necessity of an instructive biomaterial-based scaffold to direct stem cell differentiation, proliferation, paracrine activity as well as regulation of ECM deposition.


Cells Tissues Organs | 2010

Secretion of fibrinolytic enzymes facilitates human mesenchymal stem cell invasion into fibrin clots.

Sabine Neuss; Rebekka K. Schneider; Lothar Tietze; Ruth Knüchel; Willi Jahnen-Dechent

Adult human mesenchymal stem cells (hMSC) are involved in wound healing and regeneration of mesodermal tissue, but the underlying homing mechanisms are not well understood. Fibrin clot formation is associated with most wound healing processes and potentially guides the recruitment of hMSC. The objective of this study is the investigation of a fibrinolytic capacity, which is required for hMSC to migrate into a wounded tissue and thus to contribute to tissue regeneration. Using RT-PCR, semiquantitative real-time PCR and ELISA, we detected key components of the fibrinolytic cascade, including the urokinase plasminogen activator (uPA) and its receptor (uPAR), the tissue plasminogen activator (tPA) and the plasminogen activator inhibitor (PAI), suggesting a strong fibrinolytic activity of hMSC. To test this activity in a functional assay, we cultured fibrin-embedded hMSC in vitro for 7 days. The cells efficiently dissolved the surrounding fibrin mesh into the fibrin degradation products, the fibrinopeptides. The fibrinolytic activity of hMSC and human dermal fibroblasts, known to be critically involved in skin wound extracellular matrix remodeling, was similar. Our results suggest that a high intrinsic fibrinolytic capacity of hMSC mediates the invasion into a fibrin clot of a wounded tissue.

Collaboration


Dive into the Sabine Neuss's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge