Network


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

Hotspot


Dive into the research topics where Sabrina Danilin is active.

Publication


Featured researches published by Sabrina Danilin.


Cancer Research | 2006

The phosphoinositide 3-kinase/Akt pathway: a new target in human renal cell carcinoma therapy.

Carole Sourbier; Véronique Lindner; H. Lang; Abdelali Agouni; Eric Schordan; Sabrina Danilin; Sylvie Rothhut; Didier Jacqmin; Jean-Jacques Helwig; Thierry Massfelder

Metastatic renal cell carcinoma is resistant to current therapies. The phosphoinositide 3-kinase (PI3K)/Akt signaling cascade induces cell growth, cell transformation, and neovascularization. We evaluated whether targeting this pathway could be of therapeutic value against human renal cell carcinoma. The activation of the PI3K/Akt pathway and its role in renal cell carcinoma progression was evaluated in vitro in seven human cell lines by Western blot, cell counting, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, terminal deoxyribonucleotide transferase-mediated nick-end labeling assays, and fluorescence-activated cell sorting analysis, using two PI3K inhibitors, LY294002 and wortmannin, as well as by transfection with various Akt constructs and through Akt knockdown by small interfering RNA (siRNA). In vivo nude mice bearing human renal cell carcinoma tumor xenografts were treated with LY294002 (75 mg/kg/wk, 4 weeks, i.p.). Tumor growth was measured and tumors were subjected to Western blot and immunohistochemical analysis. Akt was constitutively activated in all cell lines. Constitutive phosphorylation of glycogen synthase kinase-3 (GSK-3) was observed in all cell lines, whereas forkhead transcription factor and mammalian target of rapamycin, although expressed, were not constitutively phosphorylated. Exposure to LY294002 or wortmannin decreased Akt activation and GSK-3 phosphorylation and reduced cell growth by up to 70% through induction of cell apoptosis. These effects were confirmed by transfection experiments with Akt constructs or Akt siRNA. Importantly, LY294002 induced up to 50% tumor regression in mice through tumor cell apoptosis. Tumor neovascularization was significantly increased by LY294002 treatment. Blood chemistries showed no adverse effects of the treatment. Our results suggest an important role of PI3K/Akt inhibitors as a potentially useful treatment for patients with renal cell carcinoma.


Molecular Cancer | 2009

The sonic hedgehog signaling pathway is reactivated in human renal cell carcinoma and plays orchestral role in tumor growth

Valérian Dormoy; Sabrina Danilin; Véronique Lindner; Lionel Thomas; Sylvie Rothhut; Catherine Coquard; Jean-Jacques Helwig; Didier Jacqmin; H. Lang; Thierry Massfelder

BackgroundHuman clear cell renal cell carcinoma (CRCC) remains resistant to therapies. Recent advances in Hypoxia Inducible Factors (HIF) molecular network led to targeted therapies, but unfortunately with only limited clinical significance. Elucidating the molecular processes involved in kidney tumorigenesis and resistance is central to the development of improved therapies, not only for kidney cancer but for many, if not all, cancer types. The oncogenic PI3K/Akt, NF-kB and MAPK pathways are critical for tumorigenesis. The sonic hedgehog (SHH) signaling pathway is crucial to normal development.ResultsBy quantitative RT-PCR and immunoblot, we report that the SHH signaling pathway is constitutively reactivated in tumors independently of the von Hippel-Lindau (VHL) tumor suppressor gene expression which is inactivated in the majority of CRCC. The inhibition of the SHH signaling pathway by the specific inhibitor cyclopamine abolished CRCC cell growth as assessed by cell counting, BrdU incorporation studies, fluorescence-activated cell sorting and β-galactosidase staining. Importantly, inhibition of the SHH pathway induced tumor regression in nude mice through inhibition of cell proliferation and neo-vascularization, and induction of apoptosis but not senescence assessed by in vivo studies, immunoblot and immunohistochemistry. Gli1, cyclin D1, Pax2, Lim1, VEGF, and TGF-β were exclusively expressed in tumors and were shown to be regulated by SHH, as evidenced by immunoblot after SHH inhibition. Using specific inhibitors and immunoblot, the activation of the oncogenic PI3K/Akt, NF-kB and MAPK pathways was decreased by SHH inhibition.ConclusionsThese findings support targeting SHH for the treatment of CRCC and pave the way for innovative and additional investigations in a broad range of cancers.


Cancer Research | 2007

Targeting the Nuclear Factor-κB Rescue Pathway Has Promising Future in Human Renal Cell Carcinoma Therapy

Carole Sourbier; Sabrina Danilin; Véronique Lindner; Jacques Steger; Sylvie Rothhut; Nicolas Meyer; Didier Jacqmin; Jean-Jacques Helwig; H. Lang; Thierry Massfelder

Metastatic renal cell carcinoma (RCC) remains refractory to therapies. The nuclear factor-kappaB (NF-kappaB) transcription factor is involved in cell growth, cell motility, and vascularization. We evaluated whether targeting NF-kappaB could be of therapeutic and prognostic values in human RCC. The activation of the NF-kappaB pathway in human RCC cells and tumors was investigated by Western blot. In vitro, the effects of BAY 11-7085 and sulfasalazine, two NF-kappaB inhibitors, on tumor cell growth were investigated by cell counting, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analysis, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling, and fluorescence-activated cell sorting. Their specificity toward NF-kappaB was analyzed by Western blot, confocal microscopy, NF-kappaB small interfering RNA, and NF-kappaB transcription assay. In vivo, the effects of BAY 11-7085 on the growth of human RCC tumors were investigated in nude mice. A tissue microarray (TMA) containing 241 cases of human RCC with 12 to 22 years of clinical follow-up and corresponding normal tissues was built up to assess prognostic significance of activated NF-kappaB. NF-kappaB is constitutively activated in cultured cells expressing or not the von Hippel-Lindau (VHL) tumor suppressor gene as a consequence of Akt kinase activation and in tumors. In vitro and in vivo NF-kappaB inhibition blocked tumor cell growth by inducing cell apoptosis. On the TMA, NF-kappaB activation was correlated with tumor dimension but was not found to be an independent prognostic factor for patient survival. This report provides strong evidence that the mechanisms responsible for the intrinsic resistance of RCC cells to apoptosis converge on NF-kappaB independently of VHL expression and that targeting this pathway has great anticancer potential.


OncoImmunology | 2012

Myeloid-derived suppressor cells expand during breast cancer progression and promote tumor-induced bone destruction.

Sabrina Danilin; Alyssa R. Merkel; Joshua R. Johnson; Rachelle W. Johnson; James R. Edwards; Julie A. Sterling

Myeloid-derived suppressor cells (MDSCs), identified as Gr1+CD11b+ cells in mice, expand during cancer and promote tumor growth, recurrence and burden. However, little is known about their role in bone metastases. We hypothesized that MDSCs may contribute to tumor-induced bone disease, and inoculated breast cancer cells into the left cardiac ventricle of nude mice. Disease progression was monitored weekly by X-ray and fluorescence imaging and MDSCs expansion by fluorescence-activated cell sorting. To explore the contribution of MDSCs to bone metastasis, we co-injected mice with tumor cells or PBS into the left cardiac ventricle and Gr1+CD11b+ cells isolated from healthy or tumor-bearing mice into the left tibia. MDSCs didn’t induce bone resorption in normal mice, but increased resorption and tumor burden significantly in tumor-bearing mice. In vitro experiments showed that Gr1+CD11b+ cells isolated from normal and tumor-bearing mice differentiate into osteoclasts when cultured with RANK ligand and macrophage colony-stimulating factor, and that MDSCs from tumor-bearing mice upregulate parathyroid hormone-related protein (PTHrP) mRNA levels in cancer cells. PTHrP upregulation is likely due to the 2-fold increase in transforming growth factor β expression that we observed in MDSCs isolated from tumor-bearing mice. Importantly, using MDSCs isolated from GFP-expressing animals, we found that MDSCs differentiate into osteoclast-like cells in tumor-bearing mice as evidenced by the presence of GFP+TRAP+ cells. These results demonstrate that MDSCs expand in breast cancer bone metastases and induce bone destruction. Furthermore, our data strongly suggest that MDSCs are able to differentiate into osteoclasts in vivo and that this is stimulated in the presence of tumors.


Carcinogenesis | 2010

Role of the RNA-binding protein HuR in human renal cell carcinoma

Sabrina Danilin; Carole Sourbier; Lionel Thomas; Véronique Lindner; Sylvie Rothhut; Valérian Dormoy; Jean-Jacques Helwig; Didier Jacqmin; H. Lang; Thierry Massfelder

Human conventional renal cell carcinoma (CRCC) remains resistant to therapy. The RNA-binding protein HuR regulates the stability and/or translation of multiple messenger RNAs involved in malignant transformation. In this study, we aimed to evaluate the potential role of HuR in this pathology. Using seven human CRCC cell lines expressing or not the von Hippel-Lindau (VHL) tumor suppressor gene as well as 15 normal/renal cell carcinoma tumor pairs, we showed that HuR is overexpressed in all tumors independently of the VHL status. Futhermore, HuR cytoplasmic presence appears to be more common in early tumor stages, suggesting a role in tumor promotion. We then assessed the effect of HuR knockdown using small interfering RNA in cultured cell and in tumor-bearing mice. Both in vitro and in vivo, we observed that cell growth was inhibited by 60% and that this effect was obtained through an inhibition of cell proliferation and an induction of cell apoptosis. Finally, we found that expression of vascular endothelium growth factor, tumor growth factor-beta and of the hypoxia-induced transcription factor-2alpha as well as the constitutive activation of the oncogenic phosphoinositide 3-kinase/Akt, nuclear factor-kappaB and mitogen-activated protein kinase pathways were decreased in HuR-depleted cells and tumors. All these results suggest a pivotal role for HuR in human CRCC.


Oncotarget | 2016

Establishment of a large panel of patient-derived preclinical models of human renal cell carcinoma

H. Lang; Claire Béraud; Audrey Bethry; Sabrina Danilin; Véronique Lindner; Catherine Coquard; Sylvie Rothhut; Thierry Massfelder

The objective of the present work was to establish a large panel of preclinical models of human renal cell carcinoma (RCC) directly from patients, faithfully reproducing the biological features of the original tumor. RCC tissues (all stages/subtypes) were collected for 8 years from 336 patients undergoing surgery, xenografted subcutaneously in nude mice, and serially passaged into new mice up to 13 passages. Tissue samples from the primary tumor and tumors grown in mice through passages were analyzed for biological tissue stability by histopathology, mRNA profiling, von Hippel-Lindau gene sequencing, STR fingerprinting, growth characteristics and response to current therapies. Metastatic models were also established by orthotopic implantation and analyzed by imagery. We established a large panel of 30 RCC models (passage > 3, 8.9% success rate). High tumor take rate was associated with high stage and grade. Histopathologic, molecular and genetic characteristics were preserved between original tumors and case-matched xenografts. The models reproduced the sensitivity to targeted therapies observed in the clinic. Overall, these models constitute an invaluable tool for the clinical design of efficient therapies, the identification of predictive biomarkers and translational research.


International Journal of Cancer | 2015

Targeting FAK scaffold functions inhibits human renal cell carcinoma growth.

Claire Béraud; Valérian Dormoy; Sabrina Danilin; Véronique Lindner; Audrey Bethry; Mazène Hochane; Catherine Coquard; Mariette Barthelmebs; Didier Jacqmin; H. Lang; Thierry Massfelder

Human conventional renal cell carcinoma (CCC) remains resistant to current therapies. Focal Adhesion Kinase (FAK) is upregulated in many epithelial tumors and clearly implicated in nearly all facets of cancer. However, only few reports have assessed whether FAK may be associated with renal tumorigenesis. In this study, we investigated the potential role of FAK in the growth of human CCC using a panel of CCC cell lines expressing or not the von Hippel‐Lindau (VHL) tumor suppressor gene as well as normal/tumoral renal tissue pairs. FAK was found constitutively expressed in human CCC both in culture cells and freshly harvested tumors obtained from patients. We showed that CCC cell growth was dramatically reduced in FAK‐depleted cells or after FAK inhibition with various inhibitors and this effect was obtained through inhibition of cell proliferation and induction of cell apoptosis. Additionally, our results indicated that FAK knockdown decreased CCC cell migration and invasion. More importantly, depletion or pharmacological inhibition of FAK substantially inhibited tumor growth in vivo. Interestingly, investigations of the molecular mechanism revealed loss of FAK phosphorylation during renal tumorigenesis impacting multiple signaling pathways. Taken together, our findings reveal a previously uncharacterized role of FAK in CCC whereby FAK exerts oncogenic properties through a non canonical signaling pathway involving its scaffolding kinase‐independent properties. Therefore, targeting the FAK scaffold may represent a promising approach for developing innovative and highly specific therapies in human CCC.


American Journal of Physiology-cell Physiology | 2018

Parathyroid hormone-related protein modulates mice mesangial cells inflammation and blunts apoptosis by enhancing Cox-2 expression

Mazène Hochane; Denis Raison; Catherine Coquard; Claire Béraud; Audrey Bethry; Sabrina Danilin; Thierry Massfelder; Mariette Barthelmebs

Injury of mesangial cells (MC) is a prominent feature of glomerulonephritis. Activated MC secrete inflammatory mediators that induce cell apoptosis. Parathyroid hormone-related peptide (PTHrP) is a locally active cytokine that enhances cell survival and is upregulated by proinflammatory factors in many cell types. The aim of this study was to analyze the regulation of PTHrP expression by inflammatory cytokines and to evaluate whether PTHrP itself acts as a proinflammatory and/or survival factor on male murine MC in primary culture. Our results showed that IL-1β (10 ng/ml) and TNF-α (10 ng/ml) rapidly and transiently upregulated PTHrP expression in MC. The effects of IL-1β were both transcriptional and posttranscriptional, with stabilization of the PTHrP mRNA by human antigen R (HuR). Proteome profiler arrays showed that PTHrP itself enhanced cytokines within 2 h in cell lysates, mainly IL-17, IL-16, IL-1α, and IL-6. PTHrP also stimulated sustained expression (2-4 h) of chemokines, mainly regulated upon activation normal T cell expressed and secreted (RANTES)/C-C motif chemokine 5 (CCL5) and macrophage inflammatory protein-2 (MIP-2)/C-X-C motif chemokine 2 (CXCL2), thymus and activation-regulated chemokine (TARC)/CCL17, and interferon-inducible T cell α-chemoattractant (I-TAC)/CXCL11. Moreover, PTHrP markedly enhanced cyclooxygenase-2 (COX-2) expression and elicited its autoinduction through the activation of the NF-κB pathway. PTHrP induced MC survival via the COX-2 products, and PTHrP overexpression in MC blunted the apoptotic effects of IL-1β and TNF-α. Altogether, these findings suggest that PTHrP functions as a booster of glomerular inflammatory processes and may be a negative feedback loop preserving MC survival.


The Journal of Urology | 2009

RNA-BINDING PROTEIN HUR BLOCKADE HAS POTENT ANTI-TUMOR ACTIVITIES IN HUMAN RENAL CELL CARCINOMA BOTH IN VITRO AND IN VIVO

Sabrina Danilin; Lionel Thomas; Thomas Charles; Carole Sourbier; Véronique Lindner; Jean-Jacques Helwig; Didier Jacqmin; H. Lang; Thierry Massfelder

INTRODUCTION AND OBJECTIVE: Human renal cell carcinoma (RCC) remains resistant to therapy. Clear cell RCC (CCC) is the major sub-type (>75%). The RNA-binding protein HuR is crucial for stabilization and/or translation of ARE-containing labile mRNAs such as those encoding VEGF orTGF, that are involved in malignant transformation. The potential role of HuR in human CCC growth is unknown and was evaluated in vitro and in vivo. METHODS: We used 7 human CCC cell lines expressing or not the von Hippel-Lindau (VHL) tumor suppressor gene, as well as RCC tumors (7 pT1 and 8 pT3) and corresponding normal tissues. The total and subcellular expression of HuR was assessed by western blot. In vitro, the effect of HuR on cell proliferation and death was analyzed by cell counting, BrdU incorporation studies, and FACS in cells transfected with HuR siRNA (siHuR) or with control siRNA (siCtl). In vivo in nude mice bearing human 786-0 RCC tumors, the effects of HuR silencing on tumor growth was measured after intratumoral injection of siHuR or siCtl (10 mice/group, 10 nmol/kg, 2 times/week, 3 weeks). Tumor cell proliferation and death and vascularization in treated mice were analyzed by Ki67, TUNEL and factor VIII stainings. The expression of VEGF, TGF, and HIF-2 , as well as the activation of the survival pathways PI3K/Akt, NFkB and MAPK in cells and xenografted tumors treated with siHuR or siCtl was quantified by ELISA or western blot. RESULTS: The analysis of HuR expression in cells and tumorsnonmalignant tissues pairs showed that HuR is overexpressed in tumors and that the cytoplasmic presence of HuR is more common in early stages. No differences in HuR expression and localization were observed depending on the VHL status. Cell growth was inhibited by more than 50% when HuR was down-regulated. This effect was obtained through inhibition of cell proliferation and induction of apoptosis. Injection of siHuR in nude mice inhibited tumor growth by 60% compared to tumors injected with siCtl, at least through decrease in cell proliferation and increase in cell apoptosis. The effect on vascularization is under investigation. As a consequence of HuR down-regulation, the expression of VEGF and TGFwas decreased by 50% in both cells and tumors. Very interestingly, the expression of HIF-2 as well as the activaction of PI3K/Akt, NFB and MAPK pathways were also decreased in HuR-depleted cells and tumors. CONCLUSIONS: These results argue for an important role for HuR in human CCC growth and may lead to the design of new therapeutics.


Oncogene | 2018

The Lim1 oncogene as a new therapeutic target for metastatic human renal cell carcinoma

Imène Hamaidi; Catherine Coquard; Sabrina Danilin; Valérian Dormoy; Claire Béraud; Sylvie Rothhut; Mariette Barthelmebs; Nadia Benkirane-Jessel; Véronique Lindner; H. Lang; Thierry Massfelder

Metastatic clear cell renal cell carcinoma (CCC) remains incurable despite advances in the development of anti-angiogenic targeted therapies and the emergence of immune checkpoint inhibitors. We have previously shown that the sonic hedgehog-Gli signaling pathway is oncogenic in CCC allowing us to identify the developmental Lim1 transcription factor as a Gli target and as a new oncogene in CCC regulating cell proliferation and apoptosis, and promoting tumor growth. In this previous study, preliminary in vitro results also suggested that Lim1 may be implicated in metastatic spread. Here we investigated the potential pro-metastatic role of Lim1 in advanced CCC (1) in vitro using a panel of CCC cell lines expressing or not the von Hippel-Lindau (VHL) tumor suppressor gene either naturally or by gene transfer and (2) ex vivo in 30 CCC metastatic tissues, including lymph nodes, lung, skin, bone, and adrenal metastases, and (3) in vivo, using a metastatic model by intravenous injection of siRNA-transfected cells into Balb/c nude. Our in vitro results reveal that Lim1 knockdown time-dependently decreased CCC cell motility, migration, invasion, and clonogenicity by up to 50% regardless of their VHL status. Investigating the molecular machinery involved in these processes, we identified a large panel of Lim1 targets known to be involved in cell adhesion (paxillin and fibronectin), epithelial-mesenchymal transition (Twist1/2 and snail), invasion (MMP1/2/3/8/9), and metastatic progression (CXCR4, SDF-1, and ANG-1). Importantly, Lim1 was found constitutively expressed in all metastatic tissues. The H-score in metastatic tissues being significantly superior to the score in the corresponding primary tumor tissues (P value = 0.009). Furthermore, we showed that Lim1 silencing decreases pulmonary metastasis development in terms of number and size in the in vivo metastatic model of human CCC. Taken together, these experiments strengthen the potential therapeutic value of Lim1 targeting as a promising novel approach for treating metastatic human CCC.

Collaboration


Dive into the Sabrina Danilin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

H. Lang

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Didier Jacqmin

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar

Sylvie Rothhut

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jean-Jacques Helwig

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Carole Sourbier

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Claire Béraud

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar

Lionel Thomas

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge