Network


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

Hotspot


Dive into the research topics where Florence Schaffner is active.

Publication


Featured researches published by Florence Schaffner.


Nature | 2008

Dendritic cell PAR1-S1P3 signalling couples coagulation and inflammation

Frank Niessen; Florence Schaffner; Christian Furlan-Freguia; Rafal Pawlinski; Gourab Bhattacharjee; Jerold Chun; Claudia K. Derian; Patricia Andrade-Gordon; Hugh Rosen; Wolfram Ruf

Defining critical points of modulation across heterogeneous clinical syndromes may provide insight into new therapeutic approaches. Coagulation initiated by the cytokine-receptor family member known as tissue factor is a hallmark of systemic inflammatory response syndromes in bacterial sepsis and viral haemorrhagic fevers, and anticoagulants can be effective in severe sepsis with disseminated intravascular coagulation. The precise mechanism coupling coagulation and inflammation remains unresolved. Here we show that protease-activated receptor 1 (PAR1) signalling sustains a lethal inflammatory response that can be interrupted by inhibition of either thrombin or PAR1 signalling. The sphingosine 1-phosphate (S1P) axis is a downstream component of PAR1 signalling, and by combining chemical and genetic probes for S1P receptor 3 (S1P3) we show a critical role for dendritic cell PAR1–S1P3 cross-talk in regulating amplification of inflammation in sepsis syndrome. Conversely, dendritic cells sustain escalated systemic coagulation and are the primary hub at which coagulation and inflammation intersect within the lymphatic compartment. Loss of dendritic cell PAR1–S1P3 signalling sequesters dendritic cells and inflammation into draining lymph nodes, and attenuates dissemination of interleukin-1β to the lungs. Thus, activation of dendritic cells by coagulation in the lymphatics emerges as a previously unknown mechanism that promotes systemic inflammation and lethality in decompensated innate immune responses.


Neoplasia | 2004

Inhibition of tumor growth and angiogenesis by soluble EphB4.

Georg Martiny-Baron; Thomas Korff; Florence Schaffner; Norbert Esser; Stefan Eggstein; Dieter Marmé; Hellmut G. Augustin

EphB receptors and their ephrinB ligands play a key role in the formation of a regular vascular system. Recent studies have also shown the involvement of Eph/ephrin interactions in malignant tumor progression and angiogenesis. We have generated soluble monomeric EphB4 (sEphB4)-expressing A375 melanoma cells to study the effect of dominant negatively acting sEphB4 on tumor growth and angiogenesis. Soluble EphB4-expressing A375 tumors grown subcutaneously in nude mice show dramatically reduced tumor growth compared to control tumors. The proliferative capacity of sEphB4-expressing cells in monolayer culture is not altered. Yet, sEphB4-expressing A375 cells cannot establish proper cell-cell contacts in three-dimensional spheroids. However, sEphB4 transfectants have reduced proliferation and apoptosis rates when grown in three-dimensional culture in vitro or in subcutaneous tumors in vivo. Analysis of the vascular phenotype of the tumors revealed a reduction of intratumoral microvessel density in sEphB4-expressing tumors. Corresponding to these mouse experiments, a matched pair analysis of EphB4 and ephrinB2 expression in human colon carcinomas revealed significantly upregulated levels of EphB4 expression compared to adjacent normal tissue. Taken together, the data identify dual effects of sEphB4 on the tumor and the vascular compartment that collectively inhibit tumor growth.


Nature | 2012

Tissue factor and PAR1 promote microbiota-induced intestinal vascular remodelling

Christoph Reinhardt; Mattias Bergentall; Thomas U. Greiner; Florence Schaffner; Gunnel Östergren-Lundén; Lars C. Petersen; Wolfram Ruf; Fredrik Bäckhed

The gut microbiota is a complex ecosystem that has coevolved with host physiology. Colonization of germ-free (GF) mice with a microbiota promotes increased vessel density in the small intestine, but little is known about the mechanisms involved. Tissue factor (TF) is the membrane receptor that initiates the extrinsic coagulation pathway, and it promotes developmental and tumour angiogenesis. Here we show that the gut microbiota promotes TF glycosylation associated with localization of TF on the cell surface, the activation of coagulation proteases, and phosphorylation of the TF cytoplasmic domain in the small intestine. Anti-TF treatment of colonized GF mice decreased microbiota-induced vascular remodelling and expression of the proangiogenic factor angiopoietin-1 (Ang-1) in the small intestine. Mice with a genetic deletion of the TF cytoplasmic domain or with hypomorphic TF (F3) alleles had a decreased intestinal vessel density. Coagulation proteases downstream of TF activate protease-activated receptor (PAR) signalling implicated in angiogenesis. Vessel density and phosphorylation of the cytoplasmic domain of TF were decreased in small intestine from PAR1-deficient (F2r−/−) but not PAR2-deficient (F2rl1−/−) mice, and inhibition of thrombin showed that thrombin–PAR1 signalling was upstream of TF phosphorylation. Thus, the microbiota-induced extravascular TF–PAR1 signalling loop is a novel pathway that may be modulated to influence vascular remodelling in the small intestine.


Cancer Research | 2008

Protease-Activated Receptor (PAR) 2, but not PAR1, Signaling Promotes the Development of Mammary Adenocarcinoma in Polyoma Middle T Mice

Henri H. Versteeg; Florence Schaffner; Marjolein Kerver; Lesley G. Ellies; Patricia Andrade-Gordon; Barbara M. Mueller; Wolfram Ruf

The G protein-coupled protease-activated receptors (PAR) are key signaling components for proteases in vascular biology and tumor progression. To address the contributions of PAR1 and PAR2 to breast cancer development, we established cohorts of mouse mammary tumor virus-polyoma middle T (PyMT) PAR1(-/-) and PAR2(-/-) mice, considering that the PyMT model recapitulates aspects of human disease. Appearance of palpable tumors, tumor expansion, and metastasis was indistinguishable between wild-type and PAR1(-/-) mice. PAR1(-/-) breast cancer cells were no longer responsive to thrombin in vitro, excluding compensatory up-regulation of alternative thrombin receptors and indicating that thrombin-PAR1 signaling is dispensable in breast tumor microenvironments. In contrast, palpable tumors and multifocal disease developed slower in PAR2(-/-) mice, and as a consequence of delayed tumor onset, metastasis was reduced. Analysis of early tumors showed persistence of adenomas with delayed appearance of vascularized adenocarcinomas in PAR2(-/-) mice. Furthermore, CXCL1 production by early PAR2(-/-) tumors was reduced. These results are consistent with previous xenograft data that implicated breast cancer PAR2 signaling in the induction of proangiogenic growth factors and chemokines. This study establishes that protease signaling contributes to mammary tumor development and that PAR2, rather than the thrombin receptor PAR1, plays a crucial role in the angiogenic switch.


Journal of Thrombosis and Haemostasis | 2011

Tissue Factor and Cell Signalling in Cancer Progression and Thrombosis

Wolfram Ruf; Jennifer Disse; Tatiana C. Carneiro-Lobo; Naho Yokota; Florence Schaffner

Summary.  The close link between coagulation activation and clinical cancer is well established and recent progress has defined underlying molecular pathways by which tumour cells interact with the haemostatic system to promote cancer progression. Tumour type‐specific oncogenic transformations cause constitutive and hypoxia‐dependent upregulation of tissue factor (TF) in cancer cells, but TF expressed by vascular, stromal and inflammatory cells also contributes to the procoagulant character of the tumour microenvironment. A growing body of genetic and pharmacological evidence implicates signalling by protease activated receptors (PARs) and specifically by tumour cell‐expressed TF‐VIIa‐PAR2 in the induction of an array of proangiogenic and immune modulating cytokines, chemokines and growth factors. Specific inhibition of this pathway results in attenuated tumour growth and angiogenesis. PARs are increasingly recognised as targets for proteases outside the coagulation system and emerging evidence indicates that alternative protease signalling pathways synergise with the coagulation system to promote tumour growth, angiogenesis and metastasis. The elucidation of new therapeutic targets in tumour‐promoting protease signalling pathways requires new diagnostic approaches to identify patients that will benefit from tailored therapy targeting procoagulant or signalling aspects of the TF pathway.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2009

Tissue Factor and PAR2 Signaling in the Tumor Microenvironment

Florence Schaffner; Wolfram Ruf

Diverse oncogenic transformations result in the constitutive expression of tissue factor (TF) in cancer cells. The local and systemic activation of the coagulation cascade has long been a recognized hallmark for aggressive cancer, but genetic mouse models and new experimental therapeutics have only recently demonstrated crucial roles for TF initiated cell signaling in the pathogenesis of cancer. On tumor cells, the TF-VIIa binary complex mediates activation of protease activated receptor (PAR) 2 and thereby shapes the tumor microenvironment by inducing an array of proangiogenic and immune modulating cytokines, chemokines, and growth factors. PAR2 also uniquely triggers tumor cell migration by G protein-independent pathways through beta-arrestin scaffolding. Metastatic tumor cells use additional signaling networks of the coagulation cascade by activating PAR1 through thrombin or the ternary TF-VIIa-Xa signaling complex in the vascular and potentially lymphatic system. Selective antagonists of TF-VIIa-PAR2 signaling may be used as antiangiogenic therapy without increasing the risk of bleeding, whereas coagulation and associated signaling pathways on platelets and other host cells may be targeted for therapeutic benefit in advanced cancer and metastatic disease.


Blood | 2010

Cooperation of tissue factor cytoplasmic domain and PAR2 signaling in breast cancer development

Florence Schaffner; Henri H. Versteeg; Anja Schillert; Naho Yokota; Lars C. Petersen; Barbara M. Mueller; Wolfram Ruf

Constitutive expression of tissue factor (TF) by cancer cells triggers local activation of the coagulation cascade and promotes breast cancer progression through cell signaling involving protease activated receptor (PAR)2. In human breast cancer, TF and PAR2 are up-regulated and TF cytoplasmic domain phosphorylation is correlated with relapse. Here we show that cancer cell PAR2 signaling promotes angiogenesis independent of PAR2 phosphorylation at the recognized β-arrestin recruitment site. Similar to PAR2(-/-) mice, TF cytoplasmic domain-deleted (TF(ΔCT)) mice have delayed spontaneous breast cancer development in the polyoma middle T model. Simultaneous deletion of PAR2 in TF(ΔCT) mice did not further delay tumor appearance, consistent with overlapping roles of TF and PAR2 in promoting the angiogenic switch in early stages of breast cancer. In advanced carcinomas, tumor-associated macrophages were reduced in TF(ΔCT) and TF(ΔCT)/PAR2(-/-) mice, and increased tumor vessel diameters of TF(ΔCT) mice were partially reversed by PAR2-deficiency, indicating that the TF cytoplasmic domain has additional roles that are interdependent with PAR2 signaling in regulating host angiogenic responses. These experiments demonstrate a crosstalk of tumor cell TF cytoplasmic domain and PAR2 signaling and provide a possible mechanism for the close correlation between TF phosphorylation and cancer recurrence of TF and PAR2-positive clinical breast cancer.


Thrombosis Research | 2010

Tissue factor in cancer progression and angiogenesis

Wolfram Ruf; Naho Yokota; Florence Schaffner

Constitutive expression of tissue factor (TF) by cancer cells triggers local and systemic activation of the coagulation cascade and is a major cause of cancer-associated thrombosis. Primary breast cancer biopsies show a marked upregulation of TF and protease activated receptor (PAR) 2, as well as increased TF cytoplasmic domain phosphorylation that is correlated with cancer relapse. TF signaling involving PAR2 and integrins has multiple effects on angiogenesis and tumor progression. The non-coagulant, alternatively spliced form of TF retains an integrin-binding site and, upon deposition into the tumor stroma, stimulates angiogenesis by ligating endothelial integrins alpha(v)beta(3) and alpha(6)beta(1). On tumor cells, full-length TF is constitutively associated with laminin-binding beta(1) integrins that support TF-VIIa-PAR2 signaling leading to upregulation of pro-angiogenic and immune modulatory cytokines and growth factors. Deficiency of PAR2, but not of the thrombin receptor PAR1, delays spontaneous breast cancer development and the angiogenic switch in mice. In addition, human xenograft breast cancer growth and angiogenesis is suppressed by selective antibody inhibition of TF-VIIa-PAR2 signaling, but not by blocking TF initiated coagulation. Thus, interruption of TF signaling represents a potential anti-angiogenic strategy that does not carry an increased risk of bleeding associated with prolonged inhibition of the TF coagulation pathway.


International Journal of Cancer | 2009

Evidence for tissue factor phosphorylation and its correlation with protease-activated receptor expression and the prognosis of primary breast cancer

Lisa Rydén; Dorthe Grabau; Florence Schaffner; Per-Ebbe Jönsson; Wolfram Ruf; Mattias Belting

Tissue factor (TF)‐mediated protease‐activated receptor (PAR)‐2 signaling is associated with a promigratory, invasive and proangiogenic phenotype in experimental models of breast cancer and has been mechanistically coupled to phosphorylation of the TF cytoplasmic domain (pTF). However, the clinical relevance of these findings is unknown. Here, we provide the first in vivo evidence of TF phosphorylation in experimental as well as clinical breast cancer tumors. pTF was demonstrated in MDA‐MB‐231 xenografts and in tumors from the MMTV‐PyMT transgene model of spontaneous murine breast adenocarcinoma. Tumors from PAR‐2–deficient transgenic mice were negative for pTF, thus linking pTF to PAR‐2 signaling. The clinical correlation between TF, pTF, PAR‐1, PAR‐2 and vascular endothelial growth factor (VEGF)‐A was determined by immunohistochemistry on tumors from a cohort of 172 consecutive primary breast cancer patients, with a median follow‐up time of 50 months. In 160 evaluable patient tumors, pTF was associated with TF (p = 0.01) and cancer cell expression of PAR‐1 (p = 0.001), PAR‐2 (p = 0.014) and VEGF‐A (p = 0.003) using χ2 test. PAR‐2 and VEGF‐A were coexpressed (p = 0.013) and associated with a more aggressive phenotype. Interestingly, all patients experiencing recurrences had tumors expressing pTF and PAR‐2, and pTF alone as well as coexpression of pTF and PAR‐2 were significantly correlated with shorter recurrence‐free survival (log rank test, p = 0.04 and p = 0.02, respectively). This study provides the first evidence to link PAR‐2 expression and TF phosphorylation to clinical data in human breast cancer. In conjunction with experimental tumor models, these data support an important role of TF‐PAR‐2 signaling in breast cancer recurrence.


Cancers | 2013

Integrin α5β1, the Fibronectin Receptor, as a Pertinent Therapeutic Target in Solid Tumors

Florence Schaffner; Anne Marie Ray; Monique Dontenwill

Integrins are transmembrane heterodimeric proteins sensing the cell microenvironment and modulating numerous signalling pathways. Changes in integrin expression between normal and tumoral cells support involvement of specific integrins in tumor progression and aggressiveness. This review highlights the current knowledge about α5β1 integrin, also called the fibronectin receptor, in solid tumors. We summarize data showing that α5β1 integrin is a pertinent therapeutic target expressed by tumoral neovessels and tumoral cells. Although mainly evaluated in preclinical models, α5β1 integrin merits interest in particular in colon, breast, ovarian, lung and brain tumors where its overexpression is associated with a poor prognosis for patients. Specific α5β1 integrin antagonists will be listed that may represent new potential therapeutic agents to fight defined subpopulations of particularly aggressive tumors.

Collaboration


Dive into the Florence Schaffner's collaboration.

Top Co-Authors

Avatar

Wolfram Ruf

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Naho Yokota

Yokohama City University

View shared research outputs
Top Co-Authors

Avatar

Hellmut G. Augustin

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tatiana C. Carneiro-Lobo

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Henri H. Versteeg

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dennis Pfaff

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge