Network


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

Hotspot


Dive into the research topics where Eddy Van de Leur is active.

Publication


Featured researches published by Eddy Van de Leur.


Obstetrics & Gynecology | 1998

Interleukin-1beta and interleukin-8 concentrations in the Lower uterine segment during parturition at term

Matthias Winkler; Dagmar-Christiane Fischer; Michaela Hlubek; Eddy Van de Leur; Hans Dieter Haubeck; Werner Rath

Objective To assess the roles of interleukin-1β, interleukin-8, and fibroblasts in the lower uterine segment during parturition. Methods Lower uterine segment biopsy specimens were obtained from 36 women undergoing cesarean delivery at various stages of cervical dilation (less than 2 cm, n = 8; 2 to less than 4 cm, n = 9; 4-6 cm, n = 10; more than 6 cm, n = 9). The concentrations of interleukin-1β and interleukin-8 in protein extracts prepared from the tissue samples were measured by enzyme immunoassays. The effect of incubation with interleukin-1β (30 U/mL) on interleukin-8 secretion by lower uterine segment fibroblasts in vitro also was determined. Results The median interleukin-1β concentration in the specimens increased from 1.3 pg/mg of total protein at less than 2 cm of dilation to 22.2 pg/mg of total protein at 4-6 cm of dilation (P < .05). No further increase was detectable after 6 cm of dilation. The interleukin-8 concentration increased from 17.2 pg/mg of total protein at less than 2 cm of dilation to 2080.7 pg/mg of total protein at 4-6 cm of dilation (P < .05), thus paralleling the increase in interleukin-1β concentration. Interleukin-1β induced a significant increase in interleukin-8 secretion by fibroblasts in vitro, from 0.8 ng/106 cells to 35.6 ng/106 cells. Conclusion The increase in interleukin-8 concentration in the lower uterine segment during parturition may be induced by interleukin-1β and fibroblasts may be one of the sources of this interleukin-8.


Biochimica et Biophysica Acta | 2014

The anti-fibrotic effects of CCN1/CYR61 in primary portal myofibroblasts are mediated through induction of reactive oxygen species resulting in cellular senescence, apoptosis and attenuated TGF-β signaling.

Erawan Borkham-Kamphorst; Christian Schaffrath; Eddy Van de Leur; U Haas; Lidia Tihaa; Steffen K. Meurer; Yulia A. Nevzorova; Christian Liedtke; Ralf Weiskirchen

UNLABELLED Cysteine-rich protein 61 (CCN1/CYR61) is a CCN (CYR61, CTGF (connective tissue growth factor), and NOV (Nephroblastoma overexpressed gene)) family matricellular protein comprising six secreted CCN proteins in mammals. CCN1/CYR61 expression is associated with inflammation and injury repair. Recent studies show that CCN1/CYR61 limits fibrosis in models of cutaneous wound healing by inducing cellular senescence in myofibroblasts of the granulation tissue which thereby transforms into an extracellular matrix-degrading phenotype. We here investigate CCN1/CYR61 expression in primary profibrogenic liver cells (i.e., hepatic stellate cells and periportal myofibroblasts) and found an increase of CCN1/CYR61 expression during early activation of hepatic stellate cells that declines in fully transdifferentiated myofibroblasts. By contrast, CCN1/CYR61 levels found in primary parenchymal liver cells (i.e., hepatocytes) were relatively low compared to the levels exhibited in hepatic stellate cells and portal myofibroblasts. In models of ongoing liver fibrogenesis, elevated levels of CCN1/CYR61 were particularly noticed during early periods of insult, while expression declined during prolonged phases of fibrogenesis. We generated an adenovirus type 5 encoding CCN1/CYR61 (i.e., Ad5-CMV-CCN1/CYR61) and overexpressed CCN1/CYR61 in primary portal myofibroblasts. Interestingly, overexpressed CCN1/CYR61 significantly inhibited production of collagen type I at both mRNA and protein levels as evidenced by quantitative real-time polymerase chain reaction, Western blot and immunocytochemistry. CCN1/CYR61 further induces production of reactive oxygen species (ROS) leading to dose-dependent cellular senescence and apoptosis. Additionally, we demonstrate that CCN1/CYR61 attenuates TGF-β signaling by scavenging TGF-β thereby mitigating in vivo liver fibrogenesis in a bile duct ligation model. CONCLUSION In line with dermal fibrosis and scar formation, CCN1/CYR61 is involved in liver injury repair and tissue remodeling. CCN1/CYR61 gene transfer into extracellular matrix-producing liver cells is therefore potentially beneficial in liver fibrotic therapy.


Biochimica et Biophysica Acta | 2013

Protective effects of lipocalin-2 (LCN2) in acute liver injury suggest a novel function in liver homeostasis ☆ ☆☆

Erawan Borkham-Kamphorst; Eddy Van de Leur; Henning W. Zimmermann; Karlin Raja Karlmark; Lidia Tihaa; U Haas; Frank Tacke; Thorsten Berger; Tak W. Mak; Ralf Weiskirchen

Lipocalin-2 is expressed under pernicious conditions such as intoxication, infection, inflammation and other forms of cellular stress. Experimental liver injury induces rapid and sustained LCN2 production by injured hepatocytes. However, the precise biological function of LCN2 in liver is still unknown. In this study, LCN2(-/-) mice were exposed to short term application of CCl4, lipopolysaccharide and Concanavalin A, or subjected to bile duct ligation. Subsequent injuries were assessed by liver function analysis, qRT-PCR for chemokine and cytokine expression, liver tissue Western blot, histology and TUNEL assay. Serum LCN2 levels from patients suffering from liver disease were assessed and evaluated. Acute CCl4 intoxication showed increased liver damage in LCN2(-/-) mice indicated by higher levels of aminotransferases, and increased expression of inflammatory cytokines and chemokines such as IL-1β, IL-6, TNF-α and MCP-1/CCL2, resulting in sustained activation of STAT1, STAT3 and JNK pathways. Hepatocytes of LCN2(-/-) mice showed lipid droplet accumulation and increased apoptosis. Hepatocyte apoptosis was confirmed in the Concanavalin A and lipopolysaccharide models. In chronic models (4weeks bile duct ligation or 8weeks CCl4 application), LCN2(-/-) mice showed slightly increased fibrosis compared to controls. Interestingly, serum LCN2 levels in diseased human livers were significantly higher compared to controls, but no differences were observed between cirrhotic and non-cirrhotic patients. Upregulation of LCN2 is a reliable indicator of liver damage and has significant hepato-protective effect in acute liver injury. LCN2 levels provide no correlation to the degree of liver fibrosis but show significant positive correlation to inflammation instead.


BMC Cell Biology | 2000

Comparative evaluation of gene delivery devices in primary cultures of rat hepatic stellate cells and rat myofibroblasts

Ralf Weiskirchen; Jens Kneifel; Sabine Weiskirchen; Eddy Van de Leur; Dagmar Kunz; Axel M. Gressner

BackgroundThe hepatic stellate cell is the primary cell type responsible for the excessive formation and deposition of connective tissue elements during the development of hepatic fibrosis in chronically injured liver. Culturing quiescent hepatic stellate cells on plastic causes spontaneous activation leading to a myofibroblastic phenotype similar to that seen in vivo. This provides a simple model system for studying activation and transdifferentiation of these cells. The introduction of exogenous DNA into these cells is discussed controversially mainly due to the lack of systematic analysis. Therefore, we examined comparatively five nonviral, lipid-mediated gene transfer methods and adenoviral based infection, as potential tools for efficient delivery of DNA to rat hepatic stellate cells and their transdifferentiated counterpart, i.e. myofibroblasts. Transfection conditions were determined using enhanced green fluorescent protein as a reporter expressed under the transcriptional control of the human cytomegalovirus immediate early gene 1 promoter/enhancer.ResultsWith the use of chemically enhanced transfection methods, the highest relative efficiency was obtained with FuGENE™6 gene mediated DNA transfer. Quantitative evaluation of representative transfection experiments by flow cytometry revealed that approximately 6% of the rat hepatic stellate cells were transfected. None of the transfection methods tested was able to mediate gene delivery to rat myofibroblasts. To analyze if rat hepatic stellate cells and myofibroblasts are susceptible to adenoviral infection, we have inserted the transgenic expression cassette into a recombinant adenoviral type 5 genome as replacement for the E1 region. Viral particles of this replication-deficient Ad5-based reporter are able to infect 100% of rat hepatic stellate cells and myofibroblasts, respectively.ConclusionsOur results indicate that FuGENE™6-based methods may be optimized sufficiently to offer a feasible approach for gene transfer into rat hepatic stellate cells. The data further demonstrate that adenoviral mediated transfer is a promising approach for gene delivery to these hepatic cells.


Biochemical and Biophysical Research Communications | 2015

NLRP3 inflammasome expression is driven by NF-κB in cultured hepatocytes.

Sorina Georgiana Boaru; Erawan Borkham-Kamphorst; Eddy Van de Leur; Eric Lehnen; Christian Liedtke; Ralf Weiskirchen

The inflammasomes are cytoplasmic multiprotein complexes that are responsible for activation of inflammatory reactions. In principle, there are four individual inflammasome branches (NLRP1, NLRP3, NLRC4/NALP4, and AIM2) that mediate the cleavage and activation of Caspase-1 and IL-1β that in turn lead to a complex network of cellular reactions initiating local and systemic inflammatory reactions. We have recently shown that NLRP3 expression is virtually absent in primary cultured hepatocytes and that in vitro the stimulation of hepatocytes with lipopolysaccharides results in strong activation of NLRP3 expression. We here demonstrate that this activation can be blocked by the NF-κB activation inhibitor QNZ or by infection with an adenoviral expression vector constitutively expressing a superrepressor of NF-κB. We show that QNZ blocks NF-κB-dependent expression of TNF-α, IL-1β and NLRP3. Likewise, the superrepressor of NF-κB prevents expression of NLRP3 and significantly reduces expression of inflammatory marker genes in liver cells. In a primary murine hepatoma cells, the concomitant depletion of NEMO and Caspase-8 resulted in a significant suppression of NLRP3 expression after Lipopolysaccharide challenge. Moreover, we demonstrate that a 1.3-kbp fragment located in close proximity of the most upstream transcriptional start site of the human NLRP3 gene that harbours one putative octamer NF-κB binding site renders LPS sensitivity in reporter gene assay. We conclude that NF-κB signalling is a necessary prerequisite for proper activation of the NLRP3 inflammasome in primary hepatocytes.


Biochimica et Biophysica Acta | 1980

Interaction of synthetic polynucleotides with small rat liver ribosomal subunits possessing low and highly phosphorylated protein S6

Axel M. Gressner; Eddy Van de Leur

The interaction of some synthetic mRNAs (polyuridylate, polyadenylate, polycytidylate) with small rat liver ribosomal subunits which have protein S6 in different states of phosphorylation was studied by BioGel column chromatography, affinity chromatography on poly(U)-Sepharose 4B, and continuous diafiltration at 4 degrees C. 40-S subunits with low phosphorylated protein S6 (isolated from normal liver) and small subunits with highly phosphorylated protein S6 (from galactosamine-, thioacetamide-, dimethylnitrosamine-, puromycin-, and cycloheximide-treated livers) bind initially equal amounts of poly(U) but the dissociation of the radioactive polyuridylate occurs much more rapidly and to a greater extent from the low than from the highly phosphorylated type of subunits. From control- and galactosamine-4-S subunits 62% and 22%, respectively, of originally bound [3H]poly(U) was removed. The release of initially bound poly(A) from 40-S subunits of galactosamine-treated liver ws retarded but reached finally the same level as with control liver ribosomal subunits (removal of 40% of bound [3H]poly(A)). No differences between low and highly phosphorylated subunits were observed with poly(C). If the dissociation reaction was performed at 22 degrees C instead of 4 degrees C the differences in the release of poly(U) described above disappeared.


Liver International | 2004

CSRP2, TIMP-1, and SM22alpha promoter fragments direct hepatic stellate cell-specific transgene expression in vitro, but not in vivo.

J. Herrmann; Monica Arias; Eddy Van de Leur; Axel M. Gressner; Ralf Weiskirchen

Background/Aims: The activation of hepatic stellate cells (HSC) and their transdifferentiation into myofibroblasts (MFB) is the key step for development of liver fibrosis. Over the past several years, significant progress has been made in the understanding of the critical pathways involved incells undergoing activation. Cellular activation in the course of transdifferentiation involves, among other biochemical modifications, functionally relevant changes in the control of gene expression. These include the up‐regulation of transcription factors, different extracellular matrix proteins, cell adhesion molecules, smooth muscle specific genes, and proteins involved in matrix remodelling, or cytoskeletal organization. The corresponding regulatory elements of these genes have afforded us the opportunity to express transgenes with antifibrotic potential in a cell type‐ and/or transdifferentiation‐dependent manner.


Obstetrics & Gynecology | 2000

Regulation of interleukin-8 synthesis in human lower uterine segment fibroblasts by cytokines and growth factors

Matthias Winkler; Werner Rath; Dagmar-Christiane Fischer; Eddy Van de Leur; Hans-Dieter Haubeck

Objective To investigate the influence of lipopolysaccharide, cytokines, growth factors, and progesterone on the synthesis of interleukin-8 by human lower uterine segment fibroblasts. Methods Fibroblasts derived from a lower uterine segment biopsy specimen obtained from a woman undergoing elective cesarean delivery at term were exposed to lipopolysaccharide, interleukin-1β, transforming growth factor-β1, platelet-derived growth factor-AB, and combinations of these substances. All experiments were performed in the absence and presence of progesterone. The concentration of interleukin-8 in the culture medium was determined by enzyme immunoassay after 24 hours. Results Compared with controls (0.71 ± 0.04 ng interleukin-8/106 cells), fibroblasts exposed to lipopolysaccharide, transforming growth factor-β1, or platelet-derived growth factor-AB exhibited no increase, or at most, only a minor but significant increase, in interleukin-8 secretion. Incubation with interleukin-1β led to a moderate increase, whereas the combinations interleukin-1β/transforming growth factor-β1 (105.0 ± 7.5 ng interleukin-8/106 cells) and interleukin-1β/platelet-derived growth factor-AB (387.3 ± 25.6 ng interleukin-8/106 cells) increased interleukin-8 secretion dramatically. No further increase was observed with the combination interleukin-1β/platelet-derived growth factor-AB/transforming growth factor-β1. When progesterone was added, interleukin-8 secretion decreased significantly by 16–34%, depending on the stimulator, or did not change. Conclusion The findings indicate that interleukin-8 secretion by human lower uterine segment fibroblasts in vitro is upregulated by interleukin-1β, transforming growth factor-β1, and platelet-derived growth factor-AB in a synergistic fashion. Because interleukin-8 mediates the invasion of neutrophils into the cervical stroma, this may be an important mechanism controlling cervical dilatation during parturition.


Journal of Cell Communication and Signaling | 2012

CCN3/NOV small interfering RNA enhances fibrogenic gene expression in primary hepatic stellate cells and cirrhotic fat storing cell line CFSC

Erawan Borkham-Kamphorst; Claudia R.C. van Roeyen; Eddy Van de Leur; Jürgen Floege; Ralf Weiskirchen

Nephroblastoma overexpressed gene encodes a matricellular protein (CCN3/NOV) of the CCN family, comprising CCN1 (CYR61), CCN2 (CTGF), CCN4 (WISP-1), CCN5 (WISP-2), and CCN6 (WISP-3). CCN proteins are involved in the regulation of mitosis, adhesion, apoptosis, extracellular matrix production, growth arrest and migration in multiple cell types. Compared to CCN2/CTGF, known as a profibrotic protein, the biological role of CCN3/NOV in liver fibrosis remains obscure. In this study we showed ccn3/nov mRNA to increase dramatically following hepatic stellate cell activation, reaching peak levels in fully transdifferentiated myofibroblasts. In models of experimental hepatic fibrosis, CCN3/NOV increased significantly at the mRNA and protein levels. CCN3/NOV was found mainly in non-parenchymal cells along the areas of tissue damage and repair. In the bile-duct ligation model, CCN3/NOV was localized mainly along portal tracts, while the repeated application of carbon tetrachloride resulted in CCN3/NOV expression mainly in the centrilobular areas. In contrast to CCN2/CTGF, the profibrotic cytokines platelet-derived growth factor-B and -D as well as transforming growth factor-β suppressed CCN3/NOV expression. In vitro, CCN3/NOV siRNA attenuated migration in the cirrhotic fat storing cell line CFSC well in line with in vivo findings that various types of cells expressing CCN3/NOV migrate into the area of tissue damage and regeneration. The suppression of CCN3/NOV enhanced expression of profibrotic marker proteins, such as α-smooth muscle actin, collagen type I, fibronectin, CCN2/CTGF and TIMP-1 in primary rat hepatic stellate cells and in CFSC. We further found that adenoviral overexpression of CCN2/CTGF suppressed CCN3/NOV expression, while the overexpression of CCN3/NOV as well as the suppression of CCN3/NOV by targeting siRNAs both resulted in enhanced CCN2/CTGF expression. These results indicate the complexity of CCN actions that are far beyond the classic Yin/Yang interplay.


Cellular Signalling | 2016

CCN1/CYR61 overexpression in hepatic stellate cells induces ER stress-related apoptosis.

Erawan Borkham-Kamphorst; Bettina Therese Steffen; Eddy Van de Leur; U Haas; Lidia Tihaa; Scott L. Friedman; Ralf Weiskirchen

CCN1/CYR61 is a matricellular protein of the CCN family, comprising six secreted proteins specifically associated with the extracellular matrix (ECM). CCN1 acts as an enhancer of the cutaneous wound healing process by preventing hypertrophic scar formation through induction of myofibroblast senescence. In liver fibrosis, the senescent cells are primarily derived from activated hepatic stellate cells (HSC) that initially proliferate in response to liver damage and are the major source of ECM. We investigate here the possible use of CCN1 as a senescence inducer to attenuate liver fibrogenesis by means of adenoviral gene transfer in primary HSC, myofibroblasts (MFB) and immortalized HSC lines (i.e. LX-2, CFSC-2G). Infection with Ad5-CMV-CCN1 induced large amounts of CCN1 protein in all these cells, resulting in an overload of the endoplasmic reticulum (ER) and in a compensatory unfolded protein response (UPR). The UPR resulted in upregulation of ER chaperones including BIP/Grp78, Grp94 and led to an activation of IRE1α as evidenced by spliced XBP1 mRNA with IRE1α-induced JNK phosphorylation. The UPR arm PERK and eIF2a was phosphorylated, combined with significant CHOP upregulation. Ad5-CMV-CCN1 induced HSC apoptosis that was evident by proteolytic cleavage of caspase-12, caspase-9 and the executor caspase-3 and positive TUNEL stain. Remarkably, Ad5-CMV-CCN1 effectively reduced collagen type I mRNA expression and protein. We conclude that the matricellular protein CCN1 gene transfer induces HSC apoptosis through ER stress and UPR.

Collaboration


Dive into the Eddy Van de Leur's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

U Haas

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar

H. Greiling

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar

Lidia Tihaa

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J. Herrmann

RWTH Aachen University

View shared research outputs
Researchain Logo
Decentralizing Knowledge