Network


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

Hotspot


Dive into the research topics where Florence Boutillon is active.

Publication


Featured researches published by Florence Boutillon.


American Journal of Physiology-endocrinology and Metabolism | 2012

Prolactin receptor antagonism in mouse anterior pituitary: effects on cell turnover and prolactin receptor expression

Jimena Ferraris; Florence Boutillon; Marie Bernadet; Adriana Seilicovich; Vincent Goffin; Daniel Pisera

Since anterior pituitary expresses prolactin receptors, prolactin secreted by lactotropes could exert autocrine or paracrine actions on anterior pituitary cells. In fact, it has been observed that prolactin inhibits its own expression by lactotropes. Our hypothesis is that prolactin participates in the control of anterior pituitary cell turnover. In the present study, we explored the action of prolactin on proliferation and apoptosis of anterior pituitary cells and its effect on the expression of the prolactin receptor. To determine the activity of endogenous prolactin, we evaluated the effect of the competitive prolactin receptor antagonist Δ1-9-G129R-hPRL in vivo, using transgenic mice that constitutively and systemically express this antagonist. The weight of the pituitary gland and the anterior pituitary proliferation index, determined by BrdU incorporation, were higher in transgenic mice expressing the antagonist than in wild-type littermates. In addition, blockade of prolactin receptor in vitro by Δ1-9-G129R-hPRL increased proliferation and inhibited apoptosis of somatolactotrope GH3 cells and of primary cultures of male rat anterior pituitary cells, including lactotropes. These results suggest that prolactin acts as an autocrine/paracrine antiproliferative and proapoptotic factor in the anterior pituitary gland. In addition, anterior pituitary expression of the long isoform of the prolactin receptor, measured by real-time PCR, increased about 10-fold in transgenic mice expressing the prolactin receptor antagonist, whereas only a modest increase in the S3 short-isoform expression was observed. These results suggest that endogenous prolactin may regulate its own biological actions in the anterior pituitary by inhibiting the expression of the long isoform of the prolactin receptor. In conclusion, our observations suggest that prolactin is involved in the maintenance of physiological cell renewal in the anterior pituitary. Alterations in this physiological role of prolactin could contribute to pituitary tumor development.


PLOS ONE | 2014

Prolactin Induces Apoptosis of Lactotropes in Female Rodents

Jimena Ferraris; Sandra Zárate; Gabriela Jaita; Florence Boutillon; Marie Bernadet; Julien Auffret; Adriana Seilicovich; Nadine Binart; Vincent Goffin; Daniel Pisera

Anterior pituitary cell turnover occurring during female sexual cycle is a poorly understood process that involves complex regulation of cell proliferation and apoptosis by multiple hormones. In rats, the prolactin (PRL) surge that occurs at proestrus coincides with the highest apoptotic rate. Since anterior pituitary cells express the prolactin receptor (PRLR), we aimed to address the actual role of PRL in the regulation of pituitary cell turnover in cycling females. We showed that acute hyperprolactinemia induced in ovariectomized rats using PRL injection or dopamine antagonist treatment rapidly increased apoptosis and decreased proliferation specifically of PRL producing cells (lactotropes), suggesting a direct regulation of these cell responses by PRL. To demonstrate that apoptosis naturally occurring at proestrus was regulated by transient elevation of endogenous PRL levels, we used PRLR-deficient female mice (PRLRKO) in which PRL signaling is totally abolished. According to our hypothesis, no increase in lactotrope apoptotic rate was observed at proestrus, which likely contributes to pituitary tumorigenesis observed in these animals. To decipher the molecular mechanisms underlying PRL effects, we explored the isoform-specific pattern of PRLR expression in cycling wild type females. This analysis revealed dramatic changes of long versus short PRLR ratio during the estrous cycle, which is particularly relevant since these isoforms exhibit distinct signaling properties. This pattern was markedly altered in a model of chronic PRLR signaling blockade involving transgenic mice expressing a pure PRLR antagonist (TGΔ1–9-G129R-hPRL), providing evidence that PRL regulates the expression of its own receptor in an isoform-specific manner. Taken together, these results demonstrate that i) the PRL surge occurring during proestrus is a major proapoptotic signal for lactotropes, and ii) partial or total deficiencies in PRLR signaling in the anterior pituitary may result in pituitary hyperplasia and eventual prolactinoma development, as observed in TGΔ1–9-G129R-hPRL and PRLRKO mice, respectively.


Journal of the National Cancer Institute | 2016

The Role of Prolactin in Bone Metastasis and Breast Cancer Cell-Mediated Osteoclast Differentiation.

Sutherland A; Amanda Forsyth; Yingying Cong; Grant L; Juan Th; Lee Jk; Alexander C. Klimowicz; Stephanie K. Petrillo; Hu J; Alexander K. Chan; Florence Boutillon; Goffin; Cay Egan; Patricia A. Tang; Cai L; Don Morris; Anthony M. Magliocco; Carrie S. Shemanko

BACKGROUND Metastasis to the bone is a deleterious aspect of breast cancer and is a preferred site that results in bone loss. Hormones such as prolactin (PRL) have not yet been studied for their role in modulating the secondary tumor bone microenvironment. METHODS We used quantitative immunohistochemistry with 134 samples of human primary breast cancer and 17 matched primary breast cancers and bone metastases. A Cox proportional hazards regression model was fitted to evaluate the associations between high prolactin receptor (PRLR) expression and time to bone metastasis, adjusting for estrogen receptor status, lymph node status, and chemotherapy status. We assessed osteoclast differentiation, osteoclast size, and measured pit formation in dentine slices. Statistical tests were two-sided. RESULTS High PRLR expression in the primary breast tumor was associated with a shorter time to metastasis that includes bone (PRLRAQUA Max-per 100 unit hazard ratio = 1.04, 95% confidence interval = 1.00 to 1.07, P = .03). We observed the PRLR in rare samples of bone metastases and matched primary breast cancer. PRL treatment of breast cancer cells induced osteoclast differentiation and bone lysis via secreted factors and was abrogated by a PRLR antagonist (delta1-9-G129R-hPRL). We demonstrated that sonic hedgehog is a PRL-regulated cytokine in breast cancer cells and part of the mechanism that induces osteoclast differentiation. CONCLUSIONS Our evidence indicates that PRL-PRLR can escalate the impact of breast cancer on bone metastasis and that the presence of the PRLR in the tumor microenvironment of breast cancer bone metastasis has the potential to modulate the microenvironment to induce lytic osteoclast formation.


Cancer Research | 2017

Vitamin D3 Prevents Calcium-Induced Progression of Early-Stage Prostate Tumors by Counteracting TRPC6 and Calcium Sensing Receptor Upregulation

Sophie Bernichtein; Natascha Pigat; Nicolas Barry Delongchamps; Florence Boutillon; Virginie Verkarre; Philippe Camparo; Edouard Reyes-Gomez; Arnaud Mejean; Stéphane Oudard; Eve M. Lepicard; Mélanie Viltard; Jean-Claude Souberbielle; Gérard Friedlander; Thierry Capiod; Vincent Goffin

Active surveillance has emerged as an alternative to immediate treatment for men with low-risk prostate cancer. Accordingly, identification of environmental factors that facilitate progression to more aggressive stages is critical for disease prevention. Although calcium-enriched diets have been speculated to increase prostate cancer risk, their impact on early-stage tumors remains unexplored. In this study, we addressed this issue with a large interventional animal study. Mouse models of fully penetrant and slowly evolving prostate tumorigenesis showed that a high calcium diet dramatically accelerated the progression of prostate intraepithelial neoplasia, by promoting cell proliferation, micro-invasion, tissue inflammation, and expression of acknowledged prostate cancer markers. Strikingly, dietary vitamin D prevented these calcium-triggered tumorigenic effects. Expression profiling and in vitro mechanistic studies showed that stimulation of PC-3 cells with extracellular Ca2+ resulted in an increase in cell proliferation rate, store-operated calcium entry (SOCE) amplitude, cationic channel TRPC6, and calcium sensing receptor (CaSR) expression. Notably, administration of the active vitamin D metabolite calcitriol reversed all these effects. Silencing CaSR or TRPC6 expression in calcium-stimulated PC3 cells decreased cell proliferation and SOCE. Overall, our results demonstrate the protective effects of vitamin D supplementation in blocking the progression of early-stage prostate lesions induced by a calcium-rich diet. Cancer Res; 77(2); 355-65. ©2016 AACR.


The Journal of Pathology | 2017

A rare castration-resistant progenitor cell population is highly enriched in Pten-null prostate tumours

Lucila Sackmann Sala; Florence Boutillon; Giulia Menara; Andréa De Goyon-Pélard; Mylène Leprévost; Julie Codzamanian; Natalie Lister; Jan Pencik; Ashlee K. Clark; Nicolas Cagnard; Christine Bole-Feysot; Richard Moriggl; Gail P. Risbridger; Renea A. Taylor; Lukas Kenner; Jacques Emmanuel Guidotti; Vincent Goffin

Castration‐resistant prostate cancer is a lethal disease. The cell type(s) that survive androgen deprivation remain poorly described, despite global efforts to understand the various mechanisms of therapy resistance. We recently identified in wild‐type (WT) mouse prostates a rare population of luminal progenitor cells that we called LSCmed according to their FACS profile (Lin−/Sca‐1+/CD49fmed). Here, we investigated the prevalence and castration resistance of LSCmed in various mouse models of prostate tumourigenesis (Pb‐PRL, Ptenpc−/−, and Hi‐Myc mice). LSCmed prevalence is low (∼8%, similar to WT) in Hi‐Myc mice, where prostatic androgen receptor signalling is unaltered, but is significantly higher in the two other models, where androgen receptor signalling is decreased, rising up to more than 80% in Ptenpc−/− prostates. LSCmed tolerate androgen deprivation and persist or are enriched 2–3 weeks after castration. The tumour‐initiating properties of LSCmed from Ptenpc−/− mice were demonstrated by regeneration of tumours in vivo. Transcriptomic analysis revealed that LSCmed represent a unique cell entity as their gene expression profile is different from luminal and basal/stem cells, but shares markers of each. Their intrinsic androgen signalling is markedly decreased, explaining why LSCmed tolerate androgen deprivation. This also illuminates why Ptenpc−/− tumours are castration‐resistant since LSCmed represent the most prevalent cell type in this model. We validated CK4 as a specific marker for LSCmed on sorted cells and prostate tissues by immunostaining, allowing for the detection of LSCmed in various mouse prostate specimens. In castrated Ptenpc−/− prostates, there was significant proliferation of CK4+ cells, further demonstrating their key role in castration‐resistant prostate cancer progression. Taken together, this study identifies LSCmed as a probable source of prostate cancer relapse after androgen deprivation and as a new therapeutic target for the prevention of castrate‐resistant prostate cancer. Copyright


Journal of Endocrinology | 2014

Prolactin receptor antagonism uncouples lipids from atherosclerosis susceptibility

Ronald J. van der Sluis; Tim van den Aardweg; Anne Q. Reuwer; Marcel T Twickler; Florence Boutillon; Miranda Van Eck; Vincent Goffin; Menno Hoekstra

The pituitary-derived hormone prolactin has been suggested to stimulate the development of atherosclerosis and cardiovascular disease through its effects on metabolism and inflammation. In this study, we aimed to challenge the hypothesis that inhibition of prolactin function may beneficially affect atherosclerosis burden. Hereto, atherosclerosis-susceptible LDL receptor (Ldlr) knockout mice were transplanted with bone marrow from transgenic mice expressing the pure prolactin receptor antagonist Del1-9-G129R-hPRL or their non-transgenic littermates as control. Recipient mice expressing Del1-9-G129R-hPRL exhibited a decrease in plasma cholesterol levels (-29%; P<0.05) upon feeding a Western-type diet (WTD), which could be attributed to a marked decrease (-47%; P<0.01) in the amount of cholesterol esters associated with pro-atherogenic lipoproteins VLDL/LDL. By contrast, Del1-9-G129R-hPRL-expressing mice did not display any change in the susceptibility for atherosclerosis after 12 weeks of WTD feeding. Both the absolute atherosclerotic lesion size (223 ± 33 × 10(3) μm(2) for Del1-9-G129R-hPRL vs 259 ± 32 × 10(3) μm(2) for controls) and the lesional macrophage and collagen contents were not different between the two groups of bone marrow recipients. Importantly, Del1-9-G129R-hPRL exposure increased levels of circulating neutrophils (+91%; P<0.05), lymphocytes (+55%; P<0.05), and monocytes (+43%; P<0.05), resulting in a 49% higher (P<0.01) total blood leukocyte count. In conclusion, we have shown that prolactin receptor signaling inhibition uncouples the plasma atherogenic index from atherosclerosis susceptibility in Ldlr knockout mice. Despite an associated decrease in VLDL/LDL cholesterol levels, application of the prolactin receptor antagonist Del1-9-G129R-hPRL does not alter the susceptibility for initial development of atherosclerotic lesions probably due to the parallel increase in circulating leukocyte concentrations.


General and Comparative Endocrinology | 2015

Human and murine prostate basal/stem cells are not direct targets of prolactin

Lucila Sackmann-Sala; Antoine Angelergues; Florence Boutillon; Bruno d’Acremont; Marc Maidenberg; S. Oudard; Vincent Goffin

Local overexpression of prolactin (PRL) in the prostate of Pb-PRL transgenic mice induces benign prostate tumors exhibiting marked amplification of the epithelial basal/stem cell compartment. However, PRL-activated intracellular signaling seems to be restricted to luminal cells, suggesting that basal/stem cells may not be direct targets of PRL. Given their described role as prostate cancer-initiating cells, it is important to understand the mechanisms that regulate basal/stem cells. In this study, we evaluated whether PRL can act directly on these cells, by growing them as prostaspheres. For this, primary 3D prostasphere cultures were prepared from unfractionated cells isolated from freshly harvested human and mouse benign prostate tissues and subjected to PRL stimulation in vitro. None of the various concentrations of PRL tested showed any effects on the sizes or numbers of the prostaspheres generated. In addition, neither activation of canonical PRL-induced signaling pathways (Stat5, Stat3 or Erk1/2) nor increased expression of the proliferation marker Ki-67 were detected by immunostaining in PRL-stimulated prostaspheres. Consistent with the absence of response, PRL receptor mRNA levels were generally undetectable in mouse sphere cells. We conclude that human and mouse prostate basal/stem cells are not direct targets of PRL action. The observed amplification of basal/stem cells in Pb-PRL prostates might be due to paracrine mechanisms originating from PRL action on other cell compartments. Our current efforts are aimed at unraveling these mechanisms.


Endocrinology | 2018

ATM Is Required for the Prolactin-Induced HSP90-Mediated Increase in Cellular Viability and Clonogenic Growth After DNA Damage

Ödül Karayazi Atici; Anna Urbanska; Sesha Gopal Gopinathan; Florence Boutillon; Vincent Goffin; Carrie S. Shemanko

Prolactin (PRL) acts as a survival factor for breast cancer cells, but the PRL signaling pathway and the mechanism are unknown. Previously, we identified the master chaperone, heat shock protein 90 (HSP90) α, as a prolactin-Janus kinase 2 (JAK2)-signal transducer and activator of transcription 5 (STAT5) target gene involved in survival, and here we investigated the role of HSP90 in the mechanism of PRL-induced viability in response to DNA damage. The ataxia-telangiectasia mutated kinase (ATM) protein plays a critical role in the cellular response to double-strand DNA damage. We observed that PRL increased viability of breast cancer cells treated with doxorubicin or etoposide. The increase in cellular resistance is specific to the PRL receptor, because the PRL receptor antagonist, Δ1-9-G129R-hPRL, prevented the increase in viability. Two different HSP90 inhibitors, 17-allylamino-17-demethoxygeldanamycin and BIIB021, reduced the PRL-mediated increase in cell viability of doxorubicin-treated cells and led to a decrease in JAK2, ATM, and phosphorylated ATM protein levels. Inhibitors of JAK2 (G6) and ATM (KU55933) abolished the PRL-mediated increase in cell viability of DNA-damaged cells, supporting the involvement of each, as well as the crosstalk of ATM with the PRL pathway in the context of DNA damage. Drug synergism was detected between the ATM inhibitor (KU55933) and doxorubicin and between the HSP90 inhibitor (BIIB021) and doxorubicin. Short interfering RNA directed against ATM prevented the PRL-mediated increase in cell survival in two-dimensional cell culture, three-dimensional collagen gel cultures, and clonogenic cell survival, after doxorubicin treatment. Our results indicate that ATM contributes to the PRL-JAK2-STAT5-HSP90 pathway in mediating cellular resistance to DNA-damaging agents.


Molecular Cancer Research | 2016

Abstract B56: Prolactin promotes breast cancer to bone metastasis and breast cancer cell-mediated osteoclast differentiation

Amanda Forsyth; Ashley Sutherland; Yingying Cong; Laurel Grant; Tzu-Hua Juan; Jae K. Lee; Alexander C. Klimowicz; Stephanie K. Petrillo; Jinghui Hu; Angela Chan; Florence Boutillon; Vincent Goffin; Cay Egan; Patricia A. Tang; Li Cai; Don Morris; Anthony M. Magliocco; Carrie S. Shemanko

The hormones prolactin (PRL), estrogen and progesterone have long been studied for their role in the primary breast tumor but not yet in modulating the secondary tumor microenvironment of the bone. Metastasis to the bone is a debilitating aspect of many cancers, including breast cancer, where it is a preferred site of metastasis that results in bone loss. Breast cancer cells release osteolytic factors that induce the breakdown of bone, which releases growth factors and calcium that create a vicious cycle of metastatic tumor growth. Using quantitative immunohistochemistry (AQUA) (n=134), we determined that high PRL-receptor expression in the primary tumor was associated with a shorter time to bone metastasis (PRLR AQUA Max Hazard ratio=1.04, 95% Hazard Ratio confidence limits 1.00-1.07, p=0.03/multivariable Cox proportional hazards model), indicating their treatment failure may be related to the PRL-receptor. We also identified the PRL-receptor on rare samples of matched primary and bone metastases. In an analysis of advanced breast cancer patients, we also detected the PRL-receptor in circulating tumor cells of the blood. PRL treatment of breast cancer cells induced osteoclast differentiation and bone lysis via presumed secreted factors, and interestingly these effects were abrogated by a PRL-receptor-antagonist (delta1-9-G129R-hPRL). We identified sonic hedgehog as part of the molecular mechanism by which PRL and the PRL-receptor induce breast cancer cells to directly promote the differentiation of osteoclast cells capable of bone resorption. This molecular mechanism identifies key potential therapeutic targets to ameliorate the devastating effects of breast cancer to bone metastasis and potential predictive biomarkers. Citation Format: Amanda Forsyth, Ashley Sutherland, Yingying Cong, Laurel Grant, Tzu-Hua Juan, Jae K. Lee, Alexander Klimowicz, Stephanie K. Petrillo, Jinghui Hu, Angela Chan, Florence Boutillon, Vincent Goffin, Cay Egan, Patricia A. Tang, Li Cai, Don Morris, Anthony Magliocco, Carrie S. Shemanko. Prolactin promotes breast cancer to bone metastasis and breast cancer cell-mediated osteoclast differentiation. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Breast Cancer Research; Oct 17-20, 2015; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(2_Suppl):Abstract nr B56.


American Journal of Pathology | 2014

Prolactin-Induced Prostate Tumorigenesis Links Sustained Stat5 Signaling with the Amplification of Basal/Stem Cells and Emergence of Putative Luminal Progenitors

Lucila Sackmann-Sala; Aurélie Chiche; Nerea Mosquera-Garrote; Florence Boutillon; Corinne Cordier; Ivan Pourmir; Luz Pascual-Mathey; Karima Kessal; Natascha Pigat; Philippe Camparo; Vincent Goffin

Collaboration


Dive into the Florence Boutillon's collaboration.

Top Co-Authors

Avatar

Vincent Goffin

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel Pisera

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Jimena Ferraris

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

A. Chiche

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Je Guidotti

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

L. Sackmann Sala

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Natascha Pigat

Paris Descartes University

View shared research outputs
Researchain Logo
Decentralizing Knowledge