Network


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

Hotspot


Dive into the research topics where Eliza Wiercinska is active.

Publication


Featured researches published by Eliza Wiercinska.


Cancer Research | 2010

Matrix Metalloproteinase-14 (MT1-MMP)–Mediated Endoglin Shedding Inhibits Tumor Angiogenesis

Lukas J.A.C. Hawinkels; Patricia Kuiper; Eliza Wiercinska; Hein W. Verspaget; Zhen Liu; Evangelia Pardali; Cornelis F. M. Sier; Peter ten Dijke

Endoglin is a transforming growth factor-beta coreceptor with a crucial role in angiogenesis. A soluble form of endoglin is present in the circulation, but the role of soluble endoglin (sEndoglin) is poorly understood. In addition, the endoglin shedding mechanism is not known. Therefore, we examined the role of sEndoglin in tumor angiogenesis and the mechanism by which the extracellular domain of endoglin is released from the membrane.In colorectal cancer specimens, we observed high endothelial endoglin protein expression, accompanied with slightly lower sEndoglin levels in the circulation, compared with healthy controls. In vitro analysis using endothelial sprouting assays revealed that sEndoglin reduced spontaneous and vascular endothelial growth factor-induced endothelial sprouting. Human umbilical vascular endothelial cells were found to secrete high levels of sEndoglin. Endoglin shedding was inhibited by matrix metalloproteinase (MMP) inhibitors and MMP-14 short hairpin RNA, indicating MMP-14 as the major endoglin shedding protease. Coexpression of endoglin and membrane-bound MMP-14 led to a strong increase in sEndoglin levels. Endoglin shedding required a direct interaction between endoglin and membrane-localized MMP-14. Using cleavage site mutants, we determined that MMP-14 cleaved endoglin at a site in close proximity to the transmembrane domain. Taken together, this study shows that MMP-14 mediates endoglin shedding, which may regulate the angiogenic potential of endothelial cells in the (colorectal) tumor microenvironment.


Breast Cancer Research and Treatment | 2011

The TGF-β/Smad pathway induces breast cancer cell invasion through the up-regulation of matrix metalloproteinase 2 and 9 in a spheroid invasion model system

Eliza Wiercinska; Hildegonda P. H. Naber; Evangelia Pardali; Gabri van der Pluijm; Hans van Dam; Peter ten Dijke

Transforming growth factor-β (TGF-β) has opposing roles in breast cancer progression by acting as a tumor suppressor in the initial phase, but stimulating invasion and metastasis at later stages. In contrast to the mechanisms by which TGF-β induces growth arrest, the pathways that mediate tumor invasion are not well understood. Here, we describe a TGF-β-dependent invasion assay system consisting of spheroids of MCF10A1 normal breast epithelial cells (M1) and RAS-transformed (pre-)malignant derivatives (M2 and M4) embedded in collagen gels. Both basal and TGF-β-induced invasion of these cell lines was found to correlate with their tumorigenic potential; M4 showing the most aggressive behavior and M1 showing the least. Basal invasion was strongly inhibited by the TGF-β receptor kinase inhibitor SB-431542, indicating the involvement of autocrine TGF-β or TGF-β-like activity. TGF-β-induced invasion in premalignant M2 and highly malignant M4 cells was also inhibited upon specific knockdown of Smad3 or Smad4. Interestingly, both a broad spectrum matrix metalloproteinase (MMP) inhibitor and a selective MMP2 and MMP9 inhibitor mitigated TGF-β-induced invasion of M4 cells, while leaving basal invasion intact. In line with this, TGF-β was found to strongly induce MMP2 and MMP9 expression in a Smad3- and Smad4-dependent manner. This collagen-embedded spheroid system therefore offers a valuable screening model for TGF-β/Smad- and MMP2- and MMP9-dependent breast cancer invasion.


Hepatology | 2006

Id1 is a critical mediator in TGF‐β–induced transdifferentiation of rat hepatic stellate cells

Eliza Wiercinska; Lucia Wickert; Bernd Denecke; Harun M. Said; Jafar Hamzavi; Axel M. Gressner; Midori Thorikay; Peter ten Dijke; Peter R. Mertens; Katja Breitkopf; Steven Dooley

Transforming growth factor (TGF)‐β is critically involved in the activation of hepatic stellate cells (HSCs) that occurs during the process of liver damage, for example, by alcohol, hepatotoxic viruses, or aflatoxins. Overexpression of the TGF‐β antagonist Smad7 inhibits transdifferentiation and arrests HSCs in a quiescent stage. Additionally, bile duct ligation (BDL)‐induced fibrosis is ameliorated by introducing adenoviruses expressing Smad7 with down‐regulated collagen and α‐smooth muscle actin (α‐SMA) expression. The aim of this study was to further characterize the molecular details of TGF‐β pathways that control the transdifferentiation process. In an attempt to elucidate TGF‐β target genes responsible for fibrogenesis, an analysis of Smad7‐dependent mRNA expression profiles in HSCs was performed, resulting in the identification of the inhibitor of differentiation 1 (Id1) gene. Ectopic Smad7 expression in HSCs strongly reduced Id1 mRNA and protein expression. Conversely, Id1 overexpression in HSCs enhanced cell activation and circumvented Smad7‐dependent inhibition of transdifferentiation. Moreover, knock‐down of Id1 in HSCs interfered with α‐SMA fiber formation, indicating a pivotal role of Id1 for fibrogenesis. Treatment of HSCs with TGF‐β1 led to increased Id1 protein expression, which was not directly mediated by the ALK5/Smad2/3, but the ALK1/Smad1 pathway. In vivo, Id1 expression and Smad1 phosphorylation were co‐induced during fibrogenesis. In conclusion, Id1 is identified as TGF‐β/ALK1/Smad1 target gene in HSCs and represents a critical mediator of transdifferentiation that might be involved in hepatic fibrogenesis. (HEPATOLOGY 2006;43:1032–1041.)


Molecular Cell | 2010

TMEPAI, a Transmembrane TGF-β-Inducible Protein, Sequesters Smad Proteins from Active Participation in TGF-β Signaling

Yukihide Watanabe; Susumu Itoh; Toshiyasu Goto; Eriko Ohnishi; Masako Inamitsu; Fumiko Itoh; Kiyotoshi Satoh; Eliza Wiercinska; Weiwen Yang; Liang Shi; Aya Tanaka; Naoko Nakano; A. Mieke Mommaas; Hiroshi Shibuya; Peter ten Dijke; Mitsuyasu Kato

Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine of key importance for controlling embryogenesis and tissue homeostasis. How TGF-beta signals are attenuated and terminated is not well understood. Here, we show that TMEPAI, a direct target gene of TGF-beta signaling, antagonizes TGF-beta signaling by interfering with TGF-beta type I receptor (TbetaRI)-induced R-Smad phosphorylation. TMEPAI can directly interact with R-Smads via a Smad interaction motif. TMEPAI competes with Smad anchor for receptor activation for R-Smad binding, thereby sequestering R-Smads from TbetaRI kinase activation. In mammalian cells, ectopic expression of TMEPAI inhibited TGF-beta-dependent regulation of plasminogen activator inhibitor-1, JunB, cyclin-dependent kinase inhibitors, and c-myc expression, whereas specific knockdown of TMEPAI expression prolonged duration of TGF-beta-induced Smad2 and Smad3 phosphorylation and concomitantly potentiated cellular responsiveness to TGF-beta. Consistently, TMEPAI inhibits activin-mediated mesoderm formation in Xenopus embryos. Therefore, TMEPAI participates in a negative feedback loop to control the duration and intensity of TGF-beta/Smad signaling.


Oncogene | 2014

Interaction with colon cancer cells hyperactivates TGF-β signaling in cancer-associated fibroblasts

Lukas J.A.C. Hawinkels; Madelon Paauwe; Hein W. Verspaget; Eliza Wiercinska; J M van der Zon; K van der Ploeg; Pim J. Koelink; Jan H.N. Lindeman; Wilma E. Mesker; P. ten Dijke; Cornelis F. M. Sier

The interaction between epithelial cancer cells and cancer-associated fibroblasts (CAFs) has a major role in cancer progression and eventually in metastasis. In colorectal cancer (CRC), CAFs are present in high abundance, but their origin and functional interaction with epithelial tumor cells has not been elucidated. In this study we observed strong activation of the transforming growth factor-β (TGF-β)/Smad signaling pathway in CRC CAFs, accompanied by decreased signaling in epithelial tumor cells. We evaluated the TGF-β1 response and the expression of target genes including matrix metalloproteinases (MMPs) and plasminogen activator inhibitor (PAI)-1 of various epithelial CRC cell lines and primary CAFs in vitro. TGF-β1 stimulation caused high upregulation of MMPs, PAI-1 and TGF-β1 itself. Next we showed that incubation of CAFs with conditioned medium (CM) from epithelial cancer cells led to hyperactivation of the TGF-β signaling pathway, enhanced expression of target genes like PAI-1, and the expression of α-smooth muscle actin (α-SMA). We propose that the interaction of tumor cells with resident fibroblasts results in hyperactivated TGF-β1 signaling and subsequent transdifferentiation of the fibroblasts into α-SMA-positive CAFs. In turn this leads to cumulative production of TGF-β and proteinases within the tumor microenvironment, creating a cancer-promoting feedback loop.


Journal of Cellular and Molecular Medicine | 2006

Liver fibrogenesis due to cholestasis is associated with increased Smad7 expression and Smad3 signaling.

H. Seyhan; Jafar Hamzavi; Eliza Wiercinska; Axel M. Gressner; Peter R. Mertens; J. Kopp; R. E. Horch; Katja Breitkopf; Steven Dooley

Background/Aims: Profibrogenic TGF‐β signaling in hepatic stellate cells is modulated during transdifferentiation. Strategies to abrogate TGF‐β effects provide promising antifibrotic results, however, in vivo data regarding Smad activation during fibrogenesis are scarce. Methods: Here, liver fibrosis was assessed subsequent to bile duct ligation by determining liver enzymes in serum and collagen deposition in liver tissue. Activated hepatic stellate cells were identified by immunohistochemistry and immunoblots for alpha smooth muscle actin. Cellular localization of Smad3 and Smad7 proteins was demonstrated by immunohistochemistry. RTPCR for Smad4 and Smad7 was conducted with total RNA and Northern blot analysis for Smad7 with mRNA. Whole liver lysates were prepared to detect Smad2/3/4 and phospho‐ Smad2/3 by Western blotting. Results: Cholestasis induces TGF‐β signaling via Smad3 in vivo, whereas Smad2 phosphorylation was only marginally increased. Smad4 expression levels were unchanged. Smad7 expression was continuously increasing with duration of cholestasis. Hepatocytes of fibrotic lesions exhibited nuclear staining Smad3. In contrast to this, Smad7 expression was localized to activated hepatic stellate cells. Conclusions: Hepatocytes of damaged liver tissue display increased TGF‐β signaling via Smad3. Further, negative feedback regulation of TGF‐β signaling by increased Smad7 expression in activated hepatic stellate cells occurs, however does not interfere with fibrogenesis.


Growth Factors Journal | 2009

Transforming growth factor-beta induces nerve growth factor expression in pancreatic stellate cells by activation of the ALK-5 pathway.

Stephan L. Haas; Brit Fitzner; Robert Jaster; Eliza Wiercinska; Haristi Gaitantzi; Ralf Jesenowski; J.-Matthias Löhr; Manfred V. Singer; Steven Dooley; Katja Breitkopf

Nerve growth factor (NGF), a survival factor for neurons enforces pain by sensitizing nociceptors. Also in the pancreas, NGF was associated with pain and it can stimulate the proliferation of pancreatic cancer cells. Hepatic stellate cells (HSC) respond to NGF with apoptosis. Transforming growth factor (TGF)-β, one of the strongest pro-fibrogenic activators of pancreatic stellate cells (PSC) induced NGF and its two receptors in an immortalized human cell line (ihPSC) and primary rat PSC (prPSC) as determined by RT-PCR, western blot, and immunofluorescence. In contrast to HSC, PSC expressed both NGF receptors, although p75NTR expression was weak in prPSC. In contrast to ihPSC TGF-β activated both Smad signaling cascades in prPSC. NGF secretion was diminished by the activin-like kinase (ALK)-5 inhibitor SB431542, indicating the predominant role of ALK5 in activating the NGF system in PSC. While NGF did not affect proliferation or survival of PSC it induced expression of Inhibitor of Differentiation-1. We conclude that under conditions of upregulated TGF-β, like fibrosis, NGF levels will also increase in PSC which might contribute to pancreatic wound healing responses.


Leukemia | 2013

The novel CXCR4 antagonist POL5551 mobilizes hematopoietic stem and progenitor cells with greater efficiency than Plerixafor

Darja Karpova; Katrin Dauber; Gabriele Spohn; Doreen Chudziak; Eliza Wiercinska; Miriam Schulz; Allison R. Pettit; Jean-Pierre Levesque; Barbara Romagnoli; Kalpana Patel; Eric Chevalier; Klaus Dembowsky; Halvard-Björn Bönig

Mobilized blood has supplanted bone marrow (BM) as the primary source of hematopoietic stem cells for autologous and allogeneic stem cell transplantation. Pharmacologically enforced egress of hematopoietic stem cells from BM, or mobilization, has been achieved by directly or indirectly targeting the CXCL12/CXCR4 axis. Shortcomings of the standard mobilizing agent, granulocyte colony-stimulating factor (G-CSF), administered alone or in combination with the only approved CXCR4 antagonist, Plerixafor, continue to fuel the quest for new mobilizing agents. Using Protein Epitope Mimetics technology, a novel peptidic CXCR4 antagonist, POL5551, was developed. In vitro data presented herein indicate high affinity to and specificity for CXCR4. POL5551 exhibited rapid mobilization kinetics and unprecedented efficiency in C57BL/6 mice, exceeding that of Plerixafor and at higher doses also of G-CSF. POL5551-mobilized stem cells demonstrated adequate transplantation properties. In contrast to G-CSF, POL5551 did not induce major morphological changes in the BM of mice. Moreover, we provide evidence of direct POL5551 binding to hematopoietic stem and progenitor cells (HSPCs) in vivo, strengthening the hypothesis that CXCR4 antagonists mediate mobilization by direct targeting of HSPCs. In summary, POL5551 is a potent mobilizing agent for HSPCs in mice with promising therapeutic potential if these data can be corroborated in humans.


Cellular Oncology | 2012

BMP-7 inhibits TGF-β-induced invasion of breast cancer cells through inhibition of integrin β(3) expression

Hildegonda P. H. Naber; Eliza Wiercinska; Evangelia Pardali; Theo van Laar; Ella Nirmala; Anders Sundqvist; Hans van Dam; Geertje van der Horst; Gabri van der Pluijm; Bertrand Heckmann; Erik H. J. Danen; Peter ten Dijke

BackgroundThe transforming growth factor (TGF)-β superfamily comprises cytokines such as TGF-β and Bone Morphogenetic Proteins (BMPs), which have a critical role in a multitude of biological processes. In breast cancer, high levels of TGF-β are associated with poor outcome, whereas inhibition of TGF-β-signaling reduces metastasis. In contrast, BMP-7 inhibits bone metastasis of breast cancer cells.MethodsIn this study, we investigated the effect of BMP-7 on TGF-β-induced invasion in a 3 dimensional invasion assay.ResultsBMP-7 inhibited TGF-β-induced invasion of the metastatic breast cancer cell line MCF10CA1a, but not of its premalignant precursor MCF10AT in a spheroid invasion model. The inhibitory effect appears to be specific for BMP-7, as its closest homolog, BMP-6, did not alter the invasion of MCF10CA1a spheroids. To elucidate the mechanism by which BMP-7 inhibits TGF-β-induced invasion, we analyzed invasion-related genes. BMP-7 inhibited TGF-β-induced expression of integrin αvβ3 in the spheroids. Moreover, targeting of integrins by a chemical inhibitor or knockdown of integrin β3 negatively affected TGF-β-induced invasion. On the other hand, overexpression of integrin β3 counteracted the inhibitory effect of BMP7 on TGF-β-induced invasion.ConclusionThus, BMP-7 may exert anti-invasive actions by inhibiting TGF-β-induced expression of integrin β3.


Cell Reports | 2015

Discovery and characterization of an endogenous CXCR4 antagonist.

Onofrio Zirafi; Kyeong-Ae Kim; Ludger Ständker; Katharina Mohr; Daniel Sauter; Anke Heigele; Silvia F. Kluge; Eliza Wiercinska; Doreen Chudziak; Rudolf Richter; Barbara Moepps; Peter Gierschik; Virag Vas; Hartmut Geiger; Markus Lamla; Tanja Weil; Timo Burster; Andreas Zgraja; François Daubeuf; Nelly Frossard; Muriel Hachet-Haas; Fabian Heunisch; Christoph Reichetzeder; Jean-Luc Galzi; Javier Pérez-Castells; Angeles Canales-Mayordomo; Jesús Jiménez-Barbero; Guillermo Giménez-Gallego; Marion Schneider; James Shorter

CXCL12-CXCR4 signaling controls multiple physiological processes and its dysregulation is associated with cancers and inflammatory diseases. To discover as-yet-unknown endogenous ligands of CXCR4, we screened a blood-derived peptide library for inhibitors of CXCR4-tropic HIV-1 strains. This approach identified a 16 amino acid fragment of serum albumin as an effective and highly specific CXCR4 antagonist. The endogenous peptide, termed EPI-X4, is evolutionarily conserved and generated from the highly abundant albumin precursor by pH-regulated proteases. EPI-X4 forms an unusual lasso-like structure and antagonizes CXCL12-induced tumor cell migration, mobilizes stem cells, and suppresses inflammatory responses in mice. Furthermore, the peptide is abundant in the urine of patients with inflammatory kidney diseases and may serve as a biomarker. Our results identify EPI-X4 as a key regulator of CXCR4 signaling and introduce proteolysis of an abundant precursor protein as an alternative concept for chemokine receptor regulation.

Collaboration


Dive into the Eliza Wiercinska's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter ten Dijke

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Halvard Bonig

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Darja Karpova

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Gabriele Spohn

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

S Dooley

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar

Lukas J.A.C. Hawinkels

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge