11 results on '"Martine Croset"'
Search Results
2. Data from Lysyl Oxidase Is a Strong Determinant of Tumor Cell Colonization in Bone
- Author
-
Philippe Clézardin, Marie Brevet, Antoine E. Karnoub, Géraldine Aimond, Clémence Thomas, Floriane Pez, Edith Bonnelye, Martine Croset, Casina Kan, Paola Di Mauro, Laura Ferreras, and Caroline Reynaud
- Abstract
Lysyl oxidase (LOX) is a secreted copper-dependent amine oxidase whose primary function is to drive collagen crosslinking and extracellular matrix stiffness. LOX in colorectal cancer synergizes with hypoxia-inducible factor-1 (HIF-1) to promote tumor progression. Here we investigated whether LOX/HIF1 endows colorectal cancer cells with full competence for aggressive colonization in bone. We show that a high LOX expression in primary tumors from patients with colorectal cancer was associated with poor clinical outcome, irrespective of HIF-1. In addition, LOX was expressed by tumor cells in the bone marrow from colorectal cancer patients with bone metastases. In vivo experimental studies show that LOX overexpression in colorectal cancer cells or systemic delivery of the conditioned medium from LOX-overexpressing colorectal cancer cells promoted tumor cell dissemination in the bone marrow and enhanced osteolytic lesion formation, irrespective of HIF-1. Conversely, silencing or pharmacologic inhibition of LOX activity blocked dissemination of colorectal cancer cells in the bone marrow and tumor-driven osteolytic lesion formation. In vitro, tumor-secreted LOX supported the attachment and survival of colorectal cancer cells to and in the bone matrix, and inhibited osteoblast differentiation. LOX overexpression in colorectal cancer cells also induced a robust production of IL6. In turn, both LOX and IL6 were acting in concert to promote RANKL-dependent osteoclast differentiation, thereby creating an imbalance between bone resorption and bone formation. Collectively, our findings show that LOX supports colorectal cancer cell dissemination in the bone marrow and they reveal a novel mechanism through which LOX-driven IL6 production by colorectal cancer cells impairs bone homeostasis. Cancer Res; 77(2); 268–78. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
3. Figures S1 to S11 from miRNA-30 Family Members Inhibit Breast Cancer Invasion, Osteomimicry, and Bone Destruction by Directly Targeting Multiple Bone Metastasis–Associated Genes
- Author
-
Philippe Clézardin, Klaus Pantel, Kai Bartkowiak, Saw-See Hong, Nathalie Allioli, Richard Bachelier, Charles-Henri Lecellier, Catherine Alix-Panabières, Françoise Descotes, Edith Bonnelye, Casina W.S. Kan, Francesco Pantano, and Martine Croset
- Abstract
Figure S1 shows Mesenchymal traits of human MDA-B02 and BC-M1 breast cancer cells. Figure S2 shows Quantitative RT-PCR measurements of miR-30 expression levels. Figure S3 shows Quantification of absolute amounts of miR-30s. Figure S4 shows Stable overexpression of miR-30 family members in human MDA-B02 breast cancer cells. Figure S5 shows miR-30s expression in MDA-B02 and BC-M1 breast cancer cells. Figure S6 shows Western blot analyses. Figure S7 shows that CDH11 is a target for repression by miR-30s. Figure S8 shows Interaction of MDA-MB-231 and Hs-578T silenced for CDH11 with MC3T3-E1 osteoblasts. Figure S9 shows ITGB3 and CTGF are direct targets for repression by miR-30s. Figure S10 shows Putative binding sites between miR-30s and the ITGA5 3'UTR. Figure S11 shows effects of miR-30s on MCF-7 tumor cell colonization of the bone marrow ex vivo.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure Legends from miRNA-30 Family Members Inhibit Breast Cancer Invasion, Osteomimicry, and Bone Destruction by Directly Targeting Multiple Bone Metastasis–Associated Genes
- Author
-
Philippe Clézardin, Klaus Pantel, Kai Bartkowiak, Saw-See Hong, Nathalie Allioli, Richard Bachelier, Charles-Henri Lecellier, Catherine Alix-Panabières, Françoise Descotes, Edith Bonnelye, Casina W.S. Kan, Francesco Pantano, and Martine Croset
- Abstract
Legends to Supplementary Figures S1 to S11.
- Published
- 2023
- Full Text
- View/download PDF
5. Data from miRNA-30 Family Members Inhibit Breast Cancer Invasion, Osteomimicry, and Bone Destruction by Directly Targeting Multiple Bone Metastasis–Associated Genes
- Author
-
Philippe Clézardin, Klaus Pantel, Kai Bartkowiak, Saw-See Hong, Nathalie Allioli, Richard Bachelier, Charles-Henri Lecellier, Catherine Alix-Panabières, Françoise Descotes, Edith Bonnelye, Casina W.S. Kan, Francesco Pantano, and Martine Croset
- Abstract
miRNAs are master regulators of gene expression that play key roles in cancer metastasis. During bone metastasis, metastatic tumor cells must rewire their biology and express genes that are normally expressed by bone cells (a process called osteomimicry), which endow tumor cells with full competence for outgrowth in the bone marrow. Here, we establish miR-30 family members miR-30a, miR-30b, miR-30c, miR-30d, and miR-30e as suppressors of breast cancer bone metastasis that regulate multiple pathways, including osteomimicry. Low expression of miR-30 in primary tumors from patients with breast cancer were associated with poor relapse-free survival. In addition, estrogen receptor (ER)-negative/progesterone receptor (PR)-negative breast cancer cells expressed lower miR-30 levels than their ER/PR-positive counterparts. Overexpression of miR-30 in ER/PR-negative breast cancer cells resulted in the reduction of bone metastasis burden in vivo. In vitro, miR-30 did not affect tumor cell proliferation, but did inhibit tumor cell invasion. Furthermore, overexpression of miR-30 restored bone homeostasis by reversing the effects of tumor cell–conditioned medium on osteoclastogenesis and osteoblastogenesis. A number of genes associated with osteoclastogenesis stimulation (IL8, IL11), osteoblastogenesis inhibition (DKK-1), tumor cell osteomimicry (RUNX2, CDH11), and invasiveness (CTGF, ITGA5, ITGB3) were identified as targets for repression by miR-30. Among these genes, silencing CDH11 or ITGA5 in ER-/PR-negative breast cancer cells recapitulated inhibitory effects of miR-30 on skeletal tumor burden in vivo. Overall, our findings provide evidence that miR-30 family members employ multiple mechanisms to impede breast cancer bone metastasis and may represent attractive targets for therapeutic intervention.Significance: These findings suggest miR-30 family members may serve as an effective means to therapeutically attenuate metastasis in triple-negative breast cancer. Cancer Res; 78(18); 5259–73. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplemental Figure legends from Lysyl Oxidase Is a Strong Determinant of Tumor Cell Colonization in Bone
- Author
-
Philippe Clézardin, Marie Brevet, Antoine E. Karnoub, Géraldine Aimond, Clémence Thomas, Floriane Pez, Edith Bonnelye, Martine Croset, Casina Kan, Paola Di Mauro, Laura Ferreras, and Caroline Reynaud
- Abstract
Supplementary figure legends from S1 to S7
- Published
- 2023
- Full Text
- View/download PDF
7. supplementary methods from miRNA-30 Family Members Inhibit Breast Cancer Invasion, Osteomimicry, and Bone Destruction by Directly Targeting Multiple Bone Metastasis–Associated Genes
- Author
-
Philippe Clézardin, Klaus Pantel, Kai Bartkowiak, Saw-See Hong, Nathalie Allioli, Richard Bachelier, Charles-Henri Lecellier, Catherine Alix-Panabières, Françoise Descotes, Edith Bonnelye, Casina W.S. Kan, Francesco Pantano, and Martine Croset
- Abstract
Description of (1) retroviral vector constructs, (2) ShRNA design sequences, (3) psiCHECK-2 vector constructs, (4) co-culture of breast tumor cells with osteoblasts, (5) breast cancer cohort of patients, (6) miRNA quantification in primary tumor of breast cancer patients and (7) miRNA profiling of breast cancer cell lines.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Figures 1 through 7 from Lysyl Oxidase Is a Strong Determinant of Tumor Cell Colonization in Bone
- Author
-
Philippe Clézardin, Marie Brevet, Antoine E. Karnoub, Géraldine Aimond, Clémence Thomas, Floriane Pez, Edith Bonnelye, Martine Croset, Casina Kan, Paola Di Mauro, Laura Ferreras, and Caroline Reynaud
- Abstract
Figure S1: LOX expression and clinical outcomes. Figure S2:The LOX inhibitor βAPN inhibits osteolytic lesion formation in vivo. Figure S3: Measurement of LOX enzymatic activity. Figure S4: Correlation analysis of the expression intensity of LOX and IL-6. Figure S5: Effects of LOX expression on EMT in Hct116 colorectal cancer cells. Figure S6: Effect of LOX on attachment of colorectal cancer cells to fibronectin. Figure S7: LOX enhances survival of Hct116 colorectal cancer cells.
- Published
- 2023
- Full Text
- View/download PDF
9. Abstract 486: MicroRNA-30 family members inhibit breast cancer invasion, osteomimicry, and bone destruction by directly targeting multiple bone metastasis-associated genes
- Author
-
Martine Croset, Francesco Pantano, Casina Kan, Edith Bonnelye, Francoise Descotes, Catherine Alix-Panabières, Charles Lecellier, Richard Bachelier, Nathalie Allioli, Saw See Hong, Kai Bartkowiak, Klaus Pantel, and Philippe Clézardin
- Subjects
Cancer Research ,Oncology - Abstract
MicroRNAs (miRNAs), which are master regulators of gene expression, have emerged as key players in cancer metastasis. Here, we established miR-30 family members (miR-30a, miR-30b, miR-30c, miR-30d and miR-30e) as breast cancer bone metastasis suppressor genes. Low expression levels of miR-30s in primary tumors of breast cancer patients were associated with poor relapse-free survival. Additionally, miR-30s levels were significantly lower in estrogen receptor (ER)-negative/progesterone receptor (PR)-negative tumors than in their hormone-responsive counterparts. Overexpression of miR-30s in ER/PR-negative breast cancer cells resulted in the reduction of bone metastasis burden in vivo. MiR-30s overexpression also reduced tumor outgrowth, when tumor cells were implanted subcutaneously in animals. In vitro, miR-30s did not affect tumor cell proliferation, but inhibited tumor cell invasion. Furthermore, miR-30s restored bone homeostasis by reversing the in vitro effects of the tumor cell conditioned medium on stimulation of osteoclastogenesis and inhibition of osteoblastogenesis. We identified a number of genes associated with osteoclastogenesis stimulation (IL-8, IL-11), osteoblastogenesis inhibition (DKK-1), tumor cell osteomimicry (RUNX2, CDH11) and invasiveness (CTGF, ITGA5, ITGB3) that were direct and/or indirect targets for repression by miR-30s. Among these genes, integrin ITGA5 was a previously unknown miR-30 target. By silencing ITGA5 in ER-/PR-negative breast cancer cells, colonization of the bone marrow by tumor cells was drastically reduced in vivo. Overall, our data indicate that miR-30s employ multiple mechanisms to impede breast cancer bone metastasis. These findings may pave the way to a new field of therapeutic interventions in breast cancer patients with bone metastasis. Citation Format: Martine Croset, Francesco Pantano, Casina Kan, Edith Bonnelye, Francoise Descotes, Catherine Alix-Panabières, Charles Lecellier, Richard Bachelier, Nathalie Allioli, Saw See Hong, Kai Bartkowiak, Klaus Pantel, Philippe Clézardin. MicroRNA-30 family members inhibit breast cancer invasion, osteomimicry, and bone destruction by directly targeting multiple bone metastasis-associated genes [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 486.
- Published
- 2018
- Full Text
- View/download PDF
10. Abstract 29: RANK-RANKL signaling inhibition delays early breast cancer bone metastasis formation
- Author
-
Evelyne Gineyts, Sandra Geraci, Martine Croset, Philippe Clézardin, and Sofia Sousa
- Subjects
Cancer Research ,Osteolysis ,biology ,business.industry ,Mesenchymal stem cell ,Bone metastasis ,medicine.disease ,Breast cancer ,Denosumab ,medicine.anatomical_structure ,Oncology ,RANKL ,Tumor progression ,Cancer research ,medicine ,biology.protein ,Bone marrow ,business ,medicine.drug - Abstract
Denosumab is a fully human monoclonal antibody to human RANKL, currently approved in the management of established bone metastases, mainly due to its anti-resorptive activity which blocks tumor-induced osteoclastogenesis and osteolysis. Our aims are to determine the role of RANK expression in breast cancer (BC) bone metastasis and the efficacy of RANKL blocking by AMG161 (IgG1 equivalent to Denosumab). AMG161 does not recognize murine RANKL, so we use a BalB/c nude knock-in (KI) mouse model, which expresses a chimeric murine/human RANKL. Moreover, non-invasive in vivo imaging techniques such as bioluminescence and µCT were used to monitor metastatic tumor burden and bone degradation, respectively. To evaluate bone marrow early colonization by BC cells, DiD labelling and ex vivo laser scanning multiphoton confocal microscopy were used. Long-term studies showed that intra-arterial (i.a.) injection of MDA-MB231 RANK versus MDA-MB231 leads to a poorer overall survival (OS), with the most striking differences seen in the earlier time-points. In this respect, the extent of radiographic osteolytic lesions was higher in the RANK overexpressing group than in the control group. Similar findings were observed following intra-tibial inoculation of RANK-expressing MCF-7 cells, when compared to that observed with parental MCF-7 cells. However, at later time points, bone and lung metastatic burden was similar between the two groups, suggesting that the poor OS of the RANK overexpressing group was due to a faster tumor progression owing to enhanced bone homing and colonization. Indeed, using short-term studies (5 and 14 days), i.a. injection of MDA-MB231 RANK led to higher incidence and extension of bone marrow micro-metastases than MDA-MB231, whereas there was no difference for lung micro-metastases. Daily subcutaneous injections of 1,5mg/kg AMG161 antibody to MDA-MB231RANK tumor-bearing animals, decreased bone micro-metastases and early bone marrow colonization without affecting lung micro-metastasis. We therefore hypothesized that RANK overexpressing BC cells interact with RANKL produced by osteocytes, osteoblasts and other cells in the osteogenic niche, favoring survival and colonization of BC cells in bone. To test this hypothesis, we made use of heterotypic mammosphere assays in which BC cells were cultured with or without cells of the osteogenic niche (e.g. pre-osteoblasts, mesenchymal stem cells), recombinant RANKL and anti-RANK antibody or AMG161. Co-culture with recombinant RANKL or a cellular source of RANKL increased BC cell mammosphere formation, and blocking RANK/RANKL signaling impaired it. In conclusion, RANK/RANKL signaling promotes the early engraftment of BC cells in the bone marrow. Blocking this signaling with AMG161 decreases bone marrow micro-metastasis formation in vivo, suggesting that adjuvant treatment of early-stage breast cancer patients with denosumab may prevent bone relapse. Citation Format: Sofia Sousa, Evelyne Gineyts, Sandra Geraci, Martine Croset, Philippe Clézardin. RANK-RANKL signaling inhibition delays early breast cancer bone metastasis formation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 29.
- Published
- 2018
- Full Text
- View/download PDF
11. Abstract P5-04-13: Antibody-based therapy targeting integrin a5 is an effective strategy to treat experimental breast cancer bone metastasis
- Author
-
Giuseppe Tonini, Philippe Clézardin, Keltouma Driouch, Daniele Santini, Francesco Pantano, and Martine Croset
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,biology ,medicine.drug_class ,business.industry ,Cell ,Micrometastasis ,Bone metastasis ,Cancer ,medicine.disease ,Monoclonal antibody ,medicine.anatomical_structure ,Breast cancer ,Oncology ,medicine ,biology.protein ,Bone marrow ,Antibody ,business - Abstract
Background: Integrin a5β1 is a specific fibronectin receptor that is often upregulated in breast cancer cells undergoing epithelial-to-mesenchymal transition. Preclinical evidence showed that α5β1-fibronectin interaction promotes in vitro the survival of growth-arrested breast cancer cells in the bone marrow. This is in line with the observation that disseminated tumor cells (DTCs) in the bone marrow, that are the earliest sign of development of metastatic disease in patients, express α5β1 integrin. However, the role of α5β1 in bone marrow micrometastasis formation remains unknown. Here, we investigated its role in experimental breast cancer bone metastasis, using an antibody-based strategy targeting the α5 subunit. Methods: Integrin α5 mRNA expression levels were quantified by qRT-PCR, using radically resected primary tumors of 427 breast cancer patients. Comparison between Kaplan-Meier curves was performed using the log-rank test. The Cox model was used for multivariate analysis. For experimental metastasis, female BALB/c immunodeficient mice were treated with a chimeric IgG4 monoclonal antibody that specifically binds to human integrin subunit α5 (M200;15mg/kg) or the vehicle. Treatment was performed 3 times per week starting the day before intra-arterial inoculation with luciferase-expressing human MDA-MB-231/B02 breast cancer cells, which selectively metastasize to bone. Vehicle and M200-treated mice were analyzed by radiography and bioluminescence. On day 28 after tumor cell inoculation, animals were sacrificed and histomorphometric analysis of metastatic legs was performed. Alternatively, animals were culled on day 7 after tumor cell inoculation, and the bone marrow was flushed for DTC colony assay. In-vitro cell-based assays were conducted to explore the functions of α5β1 on adhesion, invasion and survival. Statistical analysis was carried out by performing a Mann-Whitney U test. Results: Median durations of metastasis-free survival were 6.2 (high α5 expression) and 9.5 years (low α5 expression) (P = 0.0008). Additionally, compared with low expression, a high α5 expression was associated with shorter bone metastasis-free survival, both in univariate (P = 0.024) and multivariate analysis (HR = 1.65,95%CI = 1.02 to 2.67; P = 0.04).The treatment of tumor-bearing animals with M200 antibody statistically significantly delayed the onset of skeletal lesions (P = 0.02) and caused a 50% reduction in the extent of osteolytic lesions (P = 0.038), compared with vehicle. This difference was accompanied with a sharp reduction of tumor burden (P = 0.02), as determined by bioluminescence. Histomorphometric analysis of metastatic legs showed that M200 treatment decreased skeletal tumor burden (P = 0.027) and increased the bone volume (P = 0.02), compared with vehicle. Additionally, the number of DTC colonies in the bone marrow from M200-treated mice was dramatically decreased compared with vehicle (P Conclusion: Our results suggest that α5β1 integrin expression in breast cancer cells facilitates bone marrow micrometastasis formation and the subsequent development of osteolytic lesions. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P5-04-13.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.