20 results on '"Brunella Costanza"'
Search Results
2. Correction: Human colon cancer cells highly express myoferlin to maintain a fit mitochondrial network and escape p53-driven apoptosis
- Author
-
Gilles Rademaker, Brunella Costanza, Justine Bellier, Michael Herfs, Raphaël Peiffer, Ferman Agirman, Naïma Maloujahmoum, Yvette Habraken, Philippe Delvenne, Akeila Bellahcène, Vincent Castronovo, and Olivier Peulen
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. Paladin, overexpressed in colon cancer, is required for actin polymerisation and liver metastasis dissemination
- Author
-
Gilles Rademaker, Brunella Costanza, Sébastien Pyr dit Ruys, Raphaël Peiffer, Ferman Agirman, Naïma Maloujahmoum, Didier Vertommen, Andrei Turtoi, Akeila Bellahcène, Vincent Castronovo, and Olivier Peulen
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Introduction Colorectal cancer remains a public health issue and most colon cancer patients succumb to the development of metastases. Using a specific protocol of pressure-assisted interstitial fluid extrusion to recover soluble biomarkers, we identified paladin as a potential colon cancer liver metastases biomarker. Methods Using shRNA gene knockdown, we explored the biological function of paladin in colon cancer cells and investigated the phospho-proteome within colon cancer cells. We successively applied in vitro migration assays, in vivo metastasis models and co-immunoprecipitation experiments. Results We discovered that paladin is required for colon cancer cell migration and metastasis, and that paladin depletion altered the phospho-proteome within colon cancer cells. Data are available via ProteomeXchange with identifier PXD030803. Thanks to immunoprecipitation experiments, we demonstrated that paladin, was interacting with SSH1, a phosphatase involved in colon cancer metastasis. Finally, we showed that paladin depletion in cancer cells results in a less dynamic actin cytoskeleton. Conclusions Paladin is an undervalued protein in oncology. This study highlights for the first time that, paladin is participating in actin cytoskeleton remodelling and is required for efficient cancer cell migration.
- Published
- 2022
- Full Text
- View/download PDF
4. Methylglyoxal Scavengers Resensitize KRAS-Mutated Colorectal Tumors to Cetuximab
- Author
-
Justine Bellier, Marie-Julie Nokin, Maurine Caprasse, Assia Tiamiou, Arnaud Blomme, Jean L. Scheijen, Benjamin Koopmansch, Gillian M. MacKay, Barbara Chiavarina, Brunella Costanza, Gilles Rademaker, Florence Durieux, Ferman Agirman, Naïma Maloujahmoum, Pino G. Cusumano, Pierre Lovinfosse, Hing Y. Leung, Frédéric Lambert, Vincent Bours, Casper G. Schalkwijk, Roland Hustinx, Olivier Peulen, Vincent Castronovo, and Akeila Bellahcène
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: The use of cetuximab anti-epidermal growth factor receptor (anti-EGFR) antibodies has opened the era of targeted and personalized therapy in colorectal cancer (CRC). Poor response rates have been unequivocally shown in mutant KRAS and are even observed in a majority of wild-type KRAS tumors. Therefore, patient selection based on mutational profiling remains problematic. We previously identified methylglyoxal (MGO), a by-product of glycolysis, as a metabolite promoting tumor growth and metastasis. Mutant KRAS cells under MGO stress show AKT-dependent survival when compared with wild-type KRAS isogenic CRC cells. MGO induces AKT activation through phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin 2 (mTORC2) and Hsp27 regulation. Importantly, the sole induction of MGO stress in sensitive wild-type KRAS cells renders them resistant to cetuximab. MGO scavengers inhibit AKT and resensitize KRAS-mutated CRC cells to cetuximab in vivo. This study establishes a link between MGO and AKT activation and pinpoints this oncometabolite as a potential target to tackle EGFR-targeted therapy resistance in CRC. : Bellier et al. demonstrate that MGO stress is a constant feature of KRAS-mutated CRC tumors. MGO induces a key survival pathway implicated in resistance to EGFR-targeted therapy in CRC. The scavenging of this oncometabolite could be beneficial in the treatment of both wild-type and mutant KRAS CRC tumors. Keywords: methylglyoxal, colorectal cancer, KRAS mutation, EGFR-targeted therapy, Hsp27, carnosine, aminoguanidine, cetuximab, AKT signaling
- Published
- 2020
- Full Text
- View/download PDF
5. Methylglyoxal, a glycolysis side-product, induces Hsp90 glycation and YAP-mediated tumor growth and metastasis
- Author
-
Marie-Julie Nokin, Florence Durieux, Paul Peixoto, Barbara Chiavarina, Olivier Peulen, Arnaud Blomme, Andrei Turtoi, Brunella Costanza, Nicolas Smargiasso, Dominique Baiwir, Jean L Scheijen, Casper G Schalkwijk, Justine Leenders, Pascal De Tullio, Elettra Bianchi, Marc Thiry, Koji Uchida, David A Spiegel, James R Cochrane, Craig A Hutton, Edwin De Pauw, Philippe Delvenne, Dominique Belpomme, Vincent Castronovo, and Akeila Bellahcène
- Subjects
carbonyl stress ,glyoxalase 1 ,LATS1 ,breast cancer ,methylglyoxal ,YAP ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Metabolic reprogramming toward aerobic glycolysis unavoidably induces methylglyoxal (MG) formation in cancer cells. MG mediates the glycation of proteins to form advanced glycation end products (AGEs). We have recently demonstrated that MG-induced AGEs are a common feature of breast cancer. Little is known regarding the impact of MG-mediated carbonyl stress on tumor progression. Breast tumors with MG stress presented with high nuclear YAP, a key transcriptional co-activator regulating tumor growth and invasion. Elevated MG levels resulted in sustained YAP nuclear localization/activity that could be reverted using Carnosine, a scavenger for MG. MG treatment affected Hsp90 chaperone activity and decreased its binding to LATS1, a key kinase of the Hippo pathway. Cancer cells with high MG stress showed enhanced growth and metastatic potential in vivo. These findings reinforce the cumulative evidence pointing to hyperglycemia as a risk factor for cancer incidence and bring renewed interest in MG scavengers for cancer treatment.
- Published
- 2016
- Full Text
- View/download PDF
6. Asporin Is a Fibroblast-Derived TGF-β1 Inhibitor and a Tumor Suppressor Associated with Good Prognosis in Breast Cancer.
- Author
-
Pamela Maris, Arnaud Blomme, Ana Perez Palacios, Brunella Costanza, Akeila Bellahcène, Elettra Bianchi, Stephanie Gofflot, Pierre Drion, Giovanna Elvi Trombino, Emmanuel Di Valentin, Pino G Cusumano, Sylvie Maweja, Guy Jerusalem, Philippe Delvenne, Eric Lifrange, Vincent Castronovo, and Andrei Turtoi
- Subjects
Medicine - Abstract
BackgroundBreast cancer is a leading malignancy affecting the female population worldwide. Most morbidity is caused by metastases that remain incurable to date. TGF-β1 has been identified as a key driving force behind metastatic breast cancer, with promising therapeutic implications.Methods and findingsEmploying immunohistochemistry (IHC) analysis, we report, to our knowledge for the first time, that asporin is overexpressed in the stroma of most human breast cancers and is not expressed in normal breast tissue. In vitro, asporin is secreted by breast fibroblasts upon exposure to conditioned medium from some but not all human breast cancer cells. While hormone receptor (HR) positive cells cause strong asporin expression, triple-negative breast cancer (TNBC) cells suppress it. Further, our findings show that soluble IL-1β, secreted by TNBC cells, is responsible for inhibiting asporin in normal and cancer-associated fibroblasts. Using recombinant protein, as well as a synthetic peptide fragment, we demonstrate the ability of asporin to inhibit TGF-β1-mediated SMAD2 phosphorylation, epithelial to mesenchymal transition, and stemness in breast cancer cells. In two in vivo murine models of TNBC, we observed that tumors expressing asporin exhibit significantly reduced growth (2-fold; p = 0.01) and metastatic properties (3-fold; p = 0.045). A retrospective IHC study performed on human breast carcinoma (n = 180) demonstrates that asporin expression is lowest in TNBC and HER2+ tumors, while HR+ tumors have significantly higher asporin expression (4-fold; p = 0.001). Assessment of asporin expression and patient outcome (n = 60; 10-y follow-up) shows that low protein levels in the primary breast lesion significantly delineate patients with bad outcome regardless of the tumor HR status (area under the curve = 0.87; 95% CI 0.78-0.96; p = 0.0001). Survival analysis, based on gene expression (n = 375; 25-y follow-up), confirmed that low asporin levels are associated with a reduced likelihood of survival (hazard ratio = 0.58; 95% CI 0.37-0.91; p = 0.017). Although these data highlight the potential of asporin to serve as a prognostic marker, confirmation of the clinical value would require a prospective study on a much larger patient cohort.ConclusionsOur data show that asporin is a stroma-derived inhibitor of TGF-β1 and a tumor suppressor in breast cancer. High asporin expression is significantly associated with less aggressive tumors, stratifying patients according to the clinical outcome. Future pre-clinical studies should consider options for increasing asporin expression in TNBC as a promising strategy for targeted therapy.
- Published
- 2015
- Full Text
- View/download PDF
7. Metastatic colorectal cancer cells maintain the TGFβ program and use TGFBI to fuel angiogenesis
- Author
-
Akeila Bellahcene, Roberto Ronca, Guillaume Belthier, Patrick Balaguer, Simon Lacroix, Paola Chiodelli, Gaetan Van Simaeys, Andrei Turtoi, Barbara Chiavarina, Julie Pannequin, Olivier Detry, Guy Jerusalem, Serge Goldman, Stéphanie Gofflot, Arnaud Blomme, Eric Fabbrizio, Sara Rezzola, Gilles Doumont, Ambre Giguelay, Bilguun Erkhem-Ochir, Takehiko Yokobori, Vincent Castronovo, Brunella Costanza, Philippe Delvenne, Vincent Cavaillès, Emmanuel Di Valentin, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), GIGA [Université Liège], Université de Liège, University of Brescia, Institut de Génomique Fonctionnelle (IGF), Université de Montpellier (UM)-Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS), Center for Microscopy and Molecular Imaging (IBMM - CMMI), Université libre de Bruxelles (ULB), Hôpital Erasme [Bruxelles] (ULB), Faculté de Médecine [Bruxelles] (ULB), Université libre de Bruxelles (ULB)-Université libre de Bruxelles (ULB), Gunma University, Centre Hospitalier Universitaire de Liège (CHU-Liège), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), and UM, Faculté de Médecine
- Subjects
0301 basic medicine ,Colorectal Neoplasms -- blood supply -- metabolism -- pathology ,Colorectal cancer ,Angiogenesis ,Endothelial cells ,Cell ,Medicine (miscellaneous) ,Apoptosis ,alternative TGFβ signaling ,Liver Neoplasms -- blood supply -- metabolism -- secondary ,Liver metastases ,Mice ,0302 clinical medicine ,Circulating tumor cell ,Cell Movement ,Transforming Growth Factor beta ,Tumor Cells, Cultured ,Medicine ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Extracellular Matrix Proteins ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,Neovascularization, Pathologic ,Liver Neoplasms ,Sciences bio-médicales et agricoles ,Prognosis ,endothelial cells ,3. Good health ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Neovascularization, Pathologic -- metabolism -- pathology ,030220 oncology & carcinogenesis ,Signal transduction ,Colorectal Neoplasms ,Signal Transduction ,Research Paper ,Extracellular Matrix Proteins -- genetics -- metabolism ,Alternative TGFβ signaling ,Stromal cell ,03 medical and health sciences ,Settore MED/04 - PATOLOGIA GENERALE ,Biomarkers, Tumor ,Animals ,Humans ,Cell Proliferation ,business.industry ,Transforming Growth Factor beta -- genetics -- metabolism ,medicine.disease ,Xenograft Model Antitumor Assays ,eye diseases ,030104 developmental biology ,Cancer cell ,Cancer research ,Sciences pharmaceutiques ,business ,liver metastases ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,Biomarkers, Tumor -- genetics -- metabolism ,TGFBI - Abstract
Colorectal cancer (CRC) cells are traditionally considered unresponsive to TGFβ due to mutations in the receptors and/or downstream signaling molecules. TGFβ influences CRC cells only indirectly via stromal cells, such as cancer-associated fibroblasts. However, CRC cell ability to directly respond to TGFβ currently remains unexplored. This represents a missed opportunity for diagnostic and therapeutic interventions. Methods: We examined whether cancer cells from primary CRC and liver metastases respond to TGFβ by inducing TGFβ-induced protein ig-h3 (TGFBI) expression, and the contribution of canonical and non-canonical TGFβ signaling pathways to this effect. We then investigated in vitro and in vivo TGFBI impact on metastasis formation and angiogenesis. Using patient serum samples and an orthotopic mouse model of CRC liver metastases we assessed the diagnostic/tumor targeting value of novel antibodies against TGFBI. Results: Metastatic CRC cells, such as circulating tumor cells, directly respond to TGFβ. These cells were characterized by the absence of TGFβ receptor mutations and the frequent presence of p53 mutations. The pro-tumorigenic program orchestrated by TGFβ in CRC cells was mediated through TGFBI, the expression of which was positively regulated by non-canonical TGFβ signaling cascades. TGFBI inhibition was sufficient to significantly reduce liver metastasis formation in vivo. Moreover, TGFBI pro-tumorigenic function was linked to its ability to stimulate angiogenesis. TGFBI levels were higher in serum samples from untreated patients with CRC than in patients who were receiving chemotherapy. A radiolabeled anti-TGFBI antibody selectively targeted metastatic lesions in vivo, underscoring its diagnostic and therapeutic potential. Conclusions: TGFβ signaling in CRC cells directly contributes to their metastatic potential and stromal cell-independence. Proteins downstream of activated TGFβ, such as TGFBI, represent novel diagnostic and therapeutic targets for more specific anti-metastatic therapies., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2021
- Full Text
- View/download PDF
8. LSD1-directed therapy affects glioblastoma tumorigenicity by deregulating the protective ATF4-dependent integrated stress response
- Author
-
Stefania Faletti, Daniela Osti, Elena Ceccacci, Cristina Richichi, Brunella Costanza, Luciano Nicosia, Roberta Noberini, Giulia Marotta, Laura Furia, Mario R. Faretta, Silvia Brambillasca, Micaela Quarto, Luca Bertero, Renzo Boldorini, Bianca Pollo, Sara Gandini, Davide Cora, Saverio Minucci, Ciro Mercurio, Mario Varasi, Tiziana Bonaldi, and Giuliana Pelicci
- Subjects
Histone Demethylases ,animal structures ,Tumor ,Brain Neoplasms ,General Medicine ,Activating Transcription Factor 4 ,Cell Line ,Cell Line, Tumor ,Cell Proliferation ,Humans ,Neoplasm Recurrence, Local ,Neoplastic Stem Cells ,Glioblastoma ,Neoplasm Recurrence ,Local - Abstract
Glioblastoma (GBM) is a fatal tumor whose aggressiveness, heterogeneity, poor blood-brain barrier penetration, and resistance to therapy highlight the need for new targets and clinical treatments. A step toward clinical translation includes the eradication of GBM tumor-initiating cells (TICs), responsible for GBM heterogeneity and relapse. By using patient-derived TICs and xenograft orthotopic models, we demonstrated that the selective lysine-specific histone demethylase 1 inhibitor DDP_38003 (LSD1i) is able to penetrate the brain parenchyma in vivo in preclinical models, is well tolerated, and exerts antitumor activity in molecularly different GBMs. LSD1 genetic targeting further strengthens the role of LSD1 in GBM TIC maintenance. GBM TIC plasticity supports their adaptation and survival under a plethora of environmental stresses, including nutrient deficiency and proteostasis perturbation. By mimicking these stresses in vitro, we found that LSD1 inhibition hampers the induction of the activating transcription factor 4 (ATF4), the master regulator of the integrated stress response (ISR). The resulting aberrant ISR sensitizes GBM TICs to stress-induced cell death, hampering tumor aggressiveness. Functionally, LSD1i interferes with LSD1 scaffolding function and prevents its interaction with CREBBP, a critical ATF4 activator. By disrupting the interaction between CREBBP and LSD1-ATF4 axis, LSD1 inhibition prevents GBM TICs from overcoming stress and sustaining GBM progression. The effectiveness of the LSD1 inhibition in preclinical models shown here places a strong rationale toward its clinical translation for GBM treatment.
- Published
- 2021
9. Innovative methods for biomarker discovery in the evaluation and development of cancer precision therapies
- Author
-
Ijeoma Adaku Umelo, Vincenzo Castronovo, and Brunella Costanza
- Subjects
Proteomics ,0301 basic medicine ,Cancer Research ,Response to therapy ,Computational biology ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Biomarkers, Tumor ,Animals ,Humans ,Metabolomics ,Medicine ,Precision Medicine ,Biomarker discovery ,Clinical Oncology ,business.industry ,Cancer ,Genomics ,medicine.disease ,Precision medicine ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer biomarkers ,business - Abstract
The discovery of biomarkers able to detect cancer at an early stage, to evaluate its aggressiveness, and to predict the response to therapy remains a major challenge in clinical oncology and precision medicine. In this review, we summarize recent achievements in the discovery and development of cancer biomarkers. We also highlight emerging innovative methods in biomarker discovery and provide insights into the challenges faced in their evaluation and validation.
- Published
- 2018
- Full Text
- View/download PDF
10. Methylglyoxal Scavengers Resensitize KRAS-Mutated Colorectal Tumors to Cetuximab
- Author
-
Justine Bellier, Marie-Julie Nokin, Maurine Caprasse, Assia Tiamiou, Arnaud Blomme, Jean L. Scheijen, Benjamin Koopmansch, Gillian M. MacKay, Barbara Chiavarina, Brunella Costanza, Gilles Rademaker, Florence Durieux, Ferman Agirman, Naïma Maloujahmoum, Pino G. Cusumano, Pierre Lovinfosse, Hing Y. Leung, Frédéric Lambert, Vincent Bours, Casper G. Schalkwijk, Roland Hustinx, Olivier Peulen, Vincent Castronovo, Akeila Bellahcène, MUMC+: MA Alg Onderzoek Interne Geneeskunde (9), Interne Geneeskunde, and RS: Carim - V01 Vascular complications of diabetes and metabolic syndrome
- Subjects
Adult ,Male ,Glycosylation ,akt ,growth ,HSP27 Heat-Shock Proteins ,heat-shock-protein ,Cetuximab ,Mechanistic Target of Rapamycin Complex 2 ,Mice, SCID ,resistance ,Proto-Oncogene Proteins p21(ras) ,Phosphatidylinositol 3-Kinases ,Mice, Inbred NOD ,Stress, Physiological ,Cell Line, Tumor ,Animals ,Humans ,neoplasms ,lcsh:QH301-705.5 ,ras ,Aged ,Cell Proliferation ,Aged, 80 and over ,Carnosine ,Free Radical Scavengers ,Middle Aged ,Pyruvaldehyde ,digestive system diseases ,heat-shock-protein-27 ,targeted therapies ,Clone Cells ,Enzyme Activation ,lcsh:Biology (General) ,Mutation ,cancer cells ,hsp27 ,Colorectal Neoplasms ,metabolism ,Glycolysis ,Proto-Oncogene Proteins c-akt - Abstract
Summary: The use of cetuximab anti-epidermal growth factor receptor (anti-EGFR) antibodies has opened the era of targeted and personalized therapy in colorectal cancer (CRC). Poor response rates have been unequivocally shown in mutant KRAS and are even observed in a majority of wild-type KRAS tumors. Therefore, patient selection based on mutational profiling remains problematic. We previously identified methylglyoxal (MGO), a by-product of glycolysis, as a metabolite promoting tumor growth and metastasis. Mutant KRAS cells under MGO stress show AKT-dependent survival when compared with wild-type KRAS isogenic CRC cells. MGO induces AKT activation through phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin 2 (mTORC2) and Hsp27 regulation. Importantly, the sole induction of MGO stress in sensitive wild-type KRAS cells renders them resistant to cetuximab. MGO scavengers inhibit AKT and resensitize KRAS-mutated CRC cells to cetuximab in vivo. This study establishes a link between MGO and AKT activation and pinpoints this oncometabolite as a potential target to tackle EGFR-targeted therapy resistance in CRC. : Bellier et al. demonstrate that MGO stress is a constant feature of KRAS-mutated CRC tumors. MGO induces a key survival pathway implicated in resistance to EGFR-targeted therapy in CRC. The scavenging of this oncometabolite could be beneficial in the treatment of both wild-type and mutant KRAS CRC tumors. Keywords: methylglyoxal, colorectal cancer, KRAS mutation, EGFR-targeted therapy, Hsp27, carnosine, aminoguanidine, cetuximab, AKT signaling
- Published
- 2019
11. Myoferlin regulates cellular lipid metabolism and promotes metastases in triple-negative breast cancer
- Author
-
Philippe Delvenne, Sébastien Boutry, Etienne Cavalier, Masahiko Nishiyama, Olivier Peulen, Serge Goldman, Stéphanie Gofflot, Félicie Sherer, Marc Thiry, Arnaud Blomme, Robert N. Muller, Brunella Costanza, François Jouret, Vincenzo Castronovo, Touko Hirano, C. Le Goff, Ange Mouithys-Mickalad, Gilles Doumont, Andrei Turtoi, E Di Valentin, Takehiko Yokobori, Eric Lifrange, G. Van Simaeys, P. de Tullio, E. De Pauw, Akeila Bellahcene, P. Cusumano, Université de Liège, Center for Microscopy and Molecular Imaging (IBMM - CMMI), Université libre de Bruxelles (ULB), University of Mons [Belgium] (UMONS), Biothèque Wallonia-Bruxelles, Centre Hospitalier Universitaire de Liège (CHU-Liège), Mass Spectrometry Laboratory (MS LAB), Université de Liège-Centre d'Analyse des Résidus en Traces-Groupe Interdisciplinaire de Génoprotéomique Appliquée, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), and CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
0301 basic medicine ,Cancer Research ,[SDV]Life Sciences [q-bio] ,Muscle Proteins ,Triple Negative Breast Neoplasms ,Mice, SCID ,Biology ,Molecular oncology ,Oxidative Phosphorylation ,Receptor tyrosine kinase ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Growth factor receptor ,Mice, Inbred NOD ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Molecular Biology ,Triple-negative breast cancer ,Calcium-Binding Proteins ,Cytoplasmic Vesicles ,Membrane Proteins ,Cancer ,Cell cycle ,Lipid Metabolism ,medicine.disease ,3. Good health ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Heterografts ,Female ,Glycolysis - Abstract
International audience; Myoferlin is a multiple C2-domain-containing protein that regulates membrane repair, tyrosine kinase receptor function and endocytosis in myoblasts and endothelial cells. Recently it has been reported as overexpressed in several cancers and shown to contribute to proliferation, migration and invasion of cancer cells. We have previously demonstrated that myoferlin regulates epidermal growth factor receptor activity in breast cancer. In the current study, we report a consistent overexpression of myoferlin in triple-negative breast cancer cells (TNBC) over cells originating from other breast cancer subtypes. Using a combination of proteomics, metabolomics and electron microscopy, we demonstrate that myoferlin depletion results in marked alteration of endosomal system and metabolism. Mechanistically, myoferlin depletion caused impaired vesicle traffic that led to a misbalance of saturated/unsaturated fatty acids. This provoked mitochondrial dysfunction in TNBC cells. As a consequence of the major metabolic stress, TNBC cells rapidly triggered AMP activated protein kinase-mediated metabolic reprogramming to glycolysis. This reduced their ability to balance between oxidative phosphorylation and glycolysis, rendering TNBC cells metabolically inflexible, and more sensitive to metabolic drug targeting in vitro. In line with this, our in vivo findings demonstrated a significantly reduced capacity of myoferlin-deficient TNBC cells to metastasise to lungs. The significance of this observation was further supported by clinical data, showing that TNBC patients whose tumors overexpress myoferlin have worst distant metastasis-free and overall survivals. This novel insight into myoferlin function establishes an important link between vesicle traffic, cancer metabolism and progression, offering new diagnostic and therapeutic concepts to develop treatments for TNBC patients.
- Published
- 2016
- Full Text
- View/download PDF
12. Transforming growth factor beta-induced, an extracellular matrix interacting protein, enhances glycolysis and promotes pancreatic cancer cell migration
- Author
-
Justine Leenders, Assia Tiamiou, Andrei Turtoi, Pascal de Tullio, Arnaud Blomme, Vincenzo Castronovo, Gilles Rademaker, Justine Bellier, Brunella Costanza, Olivier Peulen, Akeila Bellahcene, Philippe Delvenne, Elettra Bianchi, Université de Liège, Cancer Research UK Beatson Institute [Glasgow], Centre Hospitalier Universitaire de Liège (CHU-Liège), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), and CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
Cancer Research ,Stromal cell ,endocrine system diseases ,integrin ,extracellular matrix ,[SDV]Life Sciences [q-bio] ,Chick Embryo ,Focal adhesion ,Transforming Growth Factor beta1 ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Gene silencing ,Animals ,Humans ,Receptors, Vitronectin ,pancreas ,Gene Silencing ,Extracellular Matrix Proteins ,biology ,Chemistry ,Transforming growth factor beta ,medicine.disease ,Hypoxia-Inducible Factor 1, alpha Subunit ,Survival Analysis ,eye diseases ,digestive system diseases ,3. Good health ,Pancreatic Neoplasms ,Oncology ,030220 oncology & carcinogenesis ,Focal Adhesion Protein-Tyrosine Kinases ,Cancer research ,biology.protein ,Signal transduction ,Glycolysis ,TGFBI ,Transforming growth factor ,Carcinoma, Pancreatic Ductal ,Signal Transduction ,Subcellular Fractions - Abstract
International audience; Pancreatic ductal adenocarcinoma (PDAC) remains a deadly malignancy with no efficient therapy available up-to-date. Glycolysis is the main provider of energetic substrates to sustain cancer dissemination of PDAC. Accordingly, altering the glycolytic pathway is foreseen as a sound approach to trigger pancreatic cancer regression. Here, we show for the first time that high transforming growth factor beta-induced (TGFBI) expression in PDAC patients is associated with a poor outcome. We demonstrate that, although usually secreted by stromal cells, PDAC cells synthesize and secrete TGFBI in quantity correlated with their migratory capacity. Mechanistically, we show that TGFBI activates focal adhesion kinase signaling pathway through its binding to integrin αVβ5, leading to a significant enhancement of glycolysis and to the acquisition of an invasive phenotype. Finally, we show that TGFBI silencing significantly inhibits PDAC tumor development in a chick chorioallantoic membrane assay model. Our study highlights TGFBI as an oncogenic extracellular matrix interacting protein that bears the potential to serve as a target for new anti-PDAC therapeutic strategies.
- Published
- 2018
- Full Text
- View/download PDF
13. Myoferlin controls mitochondrial structure and activity in pancreatic ductal adenocarcinoma, and affects tumor aggressiveness
- Author
-
Gilles, Rademaker, Vincent, Hennequière, Laura, Brohée, Marie-Julie, Nokin, Pierre, Lovinfosse, Florence, Durieux, Stéphanie, Gofflot, Justine, Bellier, Brunella, Costanza, Michael, Herfs, Raphael, Peiffer, Lucien, Bettendorff, Christophe, Deroanne, Marc, Thiry, Philippe, Delvenne, Roland, Hustinx, Akeila, Bellahcène, Vincent, Castronovo, and Olivier, Peulen
- Subjects
endocrine system diseases ,Calcium-Binding Proteins ,Membrane Proteins ,Muscle Proteins ,Adenocarcinoma ,digestive system diseases ,Oxidative Phosphorylation ,Article ,Mitochondria ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,Adenosine Triphosphate ,Cell Line, Tumor ,Autophagy ,Humans ,RNA, Small Interfering ,Energy Metabolism ,Glycolysis ,Carcinoma, Pancreatic Ductal ,Cell Proliferation - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related death. Therapeutic options remain very limited and are based on classical chemotherapies. Energy metabolism reprogramming appears as an emerging hallmark of cancer and is considered a therapeutic target with considerable potential. Myoferlin, a ferlin family member protein overexpressed in PDAC, is involved in plasma membrane biology and has a tumor-promoting function. In the continuity of our previous studies, we investigated the role of myoferlin in the context of energy metabolism in PDAC. We used selected PDAC tumor samples and PDAC cell lines together with small interfering RNA technology to study the role of myoferlin in energetic metabolism. In PDAC patients, we showed that myoferlin expression is negatively correlated with overall survival and with glycolytic activity evaluated by 18F-deoxyglucose positron emission tomography. We found out that myoferlin is more abundant in lipogenic pancreatic cancer cell lines and is required to maintain a branched mitochondrial structure and a high oxidative phosphorylation activity. The observed mitochondrial fission induced by myoferlin depletion led to a decrease of cell proliferation, ATP production, and autophagy induction, thus indicating an essential role of myoferlin for PDAC cell fitness. The metabolic phenotype switch generated by myoferlin silencing could open up a new perspective in the development of therapeutic strategies, especially in the context of energy metabolism.
- Published
- 2017
14. Murine stroma adopts a human-like metabolic phenotype in the PDX model of colorectal cancer and liver metastases
- Author
-
Pierre Lovinfosse, Roland Hustinx, Ana Palacios, Gaetan Van Simaeys, Edwin De Pauw, Serge Goldman, Sébastien Boutry, Félicie Sherer, Arnaud Blomme, Brunella Costanza, Touko Hirano, Akeila Bellahcene, Andrei Turtoi, Olivier Detry, Masahiko Nishiyama, Gilles Doumont, Takehiko Yokobori, Vincent Castronovo, Justine Bellier, Philippe Delvenne, Yukihiro Otaka, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), and CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Stromal cell ,Colorectal cancer ,Glucose uptake ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Mice, SCID ,Biology ,Metastasis ,Cohort Studies ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Stroma ,Cancer-Associated Fibroblasts ,In vivo ,Mice, Inbred NOD ,Genetics ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,Molecular Biology ,Liver Neoplasms ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,Disease Models, Animal ,030104 developmental biology ,Glucose ,Phenotype ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Metabolome ,Female ,Stromal Cells ,Colorectal Neoplasms ,Ex vivo ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
International audience; Cancer research is increasingly dependent of patient-derived xenograft model (PDX). However, a major point of concern regarding the PDX model remains the replacement of the human stroma with murine counterpart. In the present work we aimed at clarifying the significance of the human-to-murine stromal replacement for the fidelity of colorectal cancer (CRC) and liver metastasis (CRC-LM) PDX model. We have conducted a comparative metabolic analysis between 6 patient tumors and corresponding PDX across 4 generations. Metabolic signatures of cancer cells and stroma were measured separately by MALDI-imaging, while metabolite changes in entire tumors were quantified using mass spectrometry approach. Measurement of glucose metabolism was also conducted in vivo using [18F]-fluorodeoxyglucose (FDG) and positron emission tomography (PET). In CRC/CRC-LM PDX model, human stroma was entirely replaced at the second generation. Despite this change, MALDI-imaging demonstrated that the metabolic profiles of both stromal and cancer cells remained stable for at least four generations in comparison to the original patient material. On the tumor level, profiles of 86 water-soluble metabolites as well as 93 lipid mediators underlined the functional stability of the PDX model. In vivo PET measurement of glucose uptake (reflecting tumor glucose metabolism) supported the ex vivo observations. Our data show for the first time that CRC/CRC-LM PDX model maintains the functional stability at the metabolic level despite the early replacement of the human stroma by murine cells. The findings demonstrate that human cancer cells actively educate murine stromal cells during PDX development to adopt the human-like phenotype.
- Published
- 2017
- Full Text
- View/download PDF
15. Stromal Modulators of TGF-β in Cancer
- Author
-
Andrei Turtoi, Justine Bellier, Brunella Costanza, Ijeoma Adaku Umelo, Vincent Castronovo, Université de Liège, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), and CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
0301 basic medicine ,TGF-β ,Stromal cell ,Angiogenesis ,lcsh:Medicine ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Review ,Biology ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Mediator ,medicine ,stroma ,Tumor microenvironment ,R-SMAD ,lcsh:R ,General Medicine ,Endoglin ,medicine.disease ,3. Good health ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,proteoglycans ,proteases ,cancer-associated fibroblasts ,Transforming growth factor - Abstract
International audience; Transforming growth factor-β (TGF-β) is an intriguing cytokine exhibiting dual activities in malignant disease. It is an important mediator of cancer invasion, metastasis and angiogenesis, on the one hand, while it exhibits anti-tumor functions on the other hand. Elucidating the precise role of TGF-β in malignant development and progression requires a better understanding of the molecular mechanisms involved in its tumor suppressor to tumor promoter switch. One important aspect of TGF-β function is its interaction with proteins within the tumor microenvironment. Several stromal proteins have the natural ability to interact and modulate TGF-β function. Understanding the complex interplay between the TGF-β signaling network and these stromal proteins may provide greater insight into the development of novel therapeutic strategies that target the TGF-β axis. The present review highlights our present understanding of how stroma modulates TGF-β activity in human cancers.
- Published
- 2017
- Full Text
- View/download PDF
16. Methylglyoxal, a glycolysis side-product, induces Hsp90 glycation and YAP-mediated tumor growth and metastasis
- Author
-
Barbara Chiavarina, Brunella Costanza, Justine Leenders, Philippe Delvenne, Elettra Bianchi, James R. Cochrane, Koji Uchida, Pascal de Tullio, Paul Peixoto, Akeila Bellahcene, Craig A. Hutton, Olivier Peulen, Dominique Baiwir, Marc Thiry, Casper G. Schalkwijk, Vincent Castronovo, Andrei Turtoi, Dominique Belpomme, Arnaud Blomme, Florence Durieux, Jean L.J.M. Scheijen, Nicolas Smargiasso, Marie-Julie Nokin, Edwin De Pauw, David Spiegel, Promovendi CD, Interne Geneeskunde, MUMC+: MA Alg Interne Geneeskunde (9), RS: CARIM - R3.01 - Vascular complications of diabetes and the metabolic syndrome, Université de Liège, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Maastricht University [Maastricht], Cardiovascular Research Institute Maastricht (CARIM), Centre Hospitalier Universitaire de Liège (CHU-Liège), Nagoya University, Yale University [New Haven], University of Melbourne, and Association for Research and Treatments Against Cancer (ARTAC)
- Subjects
Glycation End Products, Advanced ,0301 basic medicine ,Glycosylation ,Metastasis ,chemistry.chemical_compound ,Lactoylglutathione lyase ,Glycation ,cell biology ,methylglyoxal ,Neoplasm Metastasis ,Biology (General) ,cancer biology ,General Neuroscience ,MESH: Glycation End Products, Advanced ,Methylglyoxal ,General Medicine ,Pyruvaldehyde ,MESH: Glycosylation ,Aerobiosis ,LATS1 ,3. Good health ,carbonyl stress ,MESH: Glycolysis ,Medicine ,YAP ,Glycolysis ,Research Article ,medicine.medical_specialty ,MESH: Pyruvaldehyde ,MESH: Cell Line, Tumor ,QH301-705.5 ,Science ,chicken ,Breast Neoplasms ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,MESH: Phosphoproteins ,glyoxalase 1 ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,breast cancer ,Cell Line, Tumor ,MESH: Aerobiosis ,MESH: Cell Proliferation ,Internal medicine ,MESH: HSP90 Heat-Shock Proteins ,medicine ,Humans ,HSP90 Heat-Shock Proteins ,human ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,mouse ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,MESH: Adaptor Proteins, Signal Transducing ,Hippo signaling pathway ,MESH: Humans ,General Immunology and Microbiology ,YAP-Signaling Proteins ,Phosphoproteins ,medicine.disease ,MESH: Neoplasm Metastasis ,030104 developmental biology ,Endocrinology ,chemistry ,Tumor progression ,Anaerobic glycolysis ,MESH: Protein Processing, Post-Translational ,Cancer cell ,biology.protein ,Cancer research ,Protein Processing, Post-Translational ,MESH: Breast Neoplasms ,Transcription Factors - Abstract
Metabolic reprogramming toward aerobic glycolysis unavoidably induces methylglyoxal (MG) formation in cancer cells. MG mediates the glycation of proteins to form advanced glycation end products (AGEs). We have recently demonstrated that MG-induced AGEs are a common feature of breast cancer. Little is known regarding the impact of MG-mediated carbonyl stress on tumor progression. Breast tumors with MG stress presented with high nuclear YAP, a key transcriptional co-activator regulating tumor growth and invasion. Elevated MG levels resulted in sustained YAP nuclear localization/activity that could be reverted using Carnosine, a scavenger for MG. MG treatment affected Hsp90 chaperone activity and decreased its binding to LATS1, a key kinase of the Hippo pathway. Cancer cells with high MG stress showed enhanced growth and metastatic potential in vivo. These findings reinforce the cumulative evidence pointing to hyperglycemia as a risk factor for cancer incidence and bring renewed interest in MG scavengers for cancer treatment. DOI: http://dx.doi.org/10.7554/eLife.19375.001
- Published
- 2016
- Full Text
- View/download PDF
17. Author response: Methylglyoxal, a glycolysis side-product, induces Hsp90 glycation and YAP-mediated tumor growth and metastasis
- Author
-
Jean L.J.M. Scheijen, Andrei Turtoi, James R. Cochrane, Florence Durieux, Dominique Baiwir, Paul Peixoto, Marie-Julie Nokin, Dominique Belpomme, Akeila Bellahcene, Pascal De Tullio, Edwin De Pauw, David Spiegel, Elettra Bianchi, Vincent Castronovo, Marc Thiry, Nicolas Smargiasso, Arnaud Blomme, Brunella Costanza, Craig A. Hutton, Olivier Peulen, Philippe Delvenne, Koji Uchida, Barbara Chiavarina, Casper G. Schalkwijk, and Justine Leenders
- Subjects
0301 basic medicine ,biology ,Methylglyoxal ,medicine.disease ,Hsp90 ,Metastasis ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,Glycation ,030220 oncology & carcinogenesis ,Side product ,Cancer research ,biology.protein ,medicine ,Glycolysis ,Tumor growth - Published
- 2016
- Full Text
- View/download PDF
18. PO-504 EXPEL: a novel non-destructive method for mining soluble tumour biomarkers
- Author
-
Brunella Costanza, Andrei Turtoi, Arnaud Blomme, Olivier Peulen, Akeila Bellahcene, and Vincenzo Castronovo
- Subjects
Cancer Research ,Tissue fluid ,business.industry ,Omics ,medicine.disease ,Bioinformatics ,Clinical routine ,Experimental research ,Metastasis ,Oncology ,Non destructive ,Biological fluids ,Medicine ,business ,Omics technologies - Abstract
Introduction The search for biomarkers able to detect and evaluate disease such as cancer at an early stage, or to predict resistance and response to therapies, has been and remains a major challenge. Despite very important progresses in all fields of omics technologies, the success of discovery of clinically valuable biomarkers is surprisingly disappointing. Difficult mining of secreted proteins in biological fluids poses the first major hurdle, mainly because the concentration of interesting proteins in serum or urine is generally very low. The second key limitation in the field is the inaccessibility of tissue specimens from early lesions. Those are routinely required in their integrity for the complete histological evaluation in the clinical routine, leaving no residual material for research. Material and methods We have developed a simple and original proximal tissue fluid mining method we named EXPEL. It enables efficient extraction of soluble biomarkers while conserving the tissue intact for subsequent pathological analysis. Importantly, the EXPEL method will not only allow the researchers to access human tissues that are very difficult to obtain, but for the first time, scientists and clinicians can share the same material for both experimental research and routine clinical analysis. Results and discussions We hypothesised that subjecting tissue biopsies to cycles of low-pressure pulses under mild hypertonic conditions would allow a rapid extrusion of interstitial fluid containing the biomarkers of interest, while preserving the morphology and antigenicity of the sample for subsequent pathological investigation. To test the value of the EXPEL method we have applied our procedure to a series of primary colorectal tumours (CRC) and liver metastasis samples (CRC-LM). This proof-of-principle study demonstrates the validity of EXPEL-extruded fluid as unique starting material for the most advanced OMICs methodologies such as proteomic, genomic, metabolic, while showing no disadvantage for routine clinical and pathological investigations. Conclusion Our method enables, for the first time, both clinicians and scientists to explore identical clinical material regardless of its origin and size, which has a major positive impact on translation to the clinic.
- Published
- 2018
- Full Text
- View/download PDF
19. Asporin Is a Fibroblast-Derived TGF-β1 Inhibitor and a Tumor Suppressor Associated with Good Prognosis in Breast Cancer
- Author
-
Sylvie Maweja, P. Cusumano, Guy Jerusalem, Philippe Delvenne, Pamela Maris, Vincent Castronovo, Ana Palacios, Emmanuel Di Valentin, Stéphanie Gofflot, Brunella Costanza, Elettra Bianchi, Arnaud Blomme, Eric Lifrange, Akeila Bellahcene, Pierre Drion, Giovanna Elvi Trombino, Andrei Turtoi, Université de Liège, Università della Calabria [Arcavacata di Rende] (Unical), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), and CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
Oncology ,CA15-3 ,medicine.medical_specialty ,Blotting, Western ,Interleukin-1beta ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Breast Neoplasms ,Enzyme-Linked Immunosorbent Assay ,Malignancy ,Real-Time Polymerase Chain Reaction ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Breast cancer ,Transforming Growth Factor beta ,Internal medicine ,medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Animals ,Humans ,skin and connective tissue diseases ,030304 developmental biology ,Retrospective Studies ,0303 health sciences ,Extracellular Matrix Proteins ,biology ,Asporin ,Cancer ,General Medicine ,Transforming growth factor beta ,Fibroblasts ,Middle Aged ,medicine.disease ,Prognosis ,Metastatic breast cancer ,Survival Analysis ,3. Good health ,Fibroblast Growth Factors ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,biology.protein ,[SDV.SP.PHARMA]Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Medicine ,Heterografts ,Female ,Research Article - Abstract
Background Breast cancer is a leading malignancy affecting the female population worldwide. Most morbidity is caused by metastases that remain incurable to date. TGF-β1 has been identified as a key driving force behind metastatic breast cancer, with promising therapeutic implications. Methods and Findings Employing immunohistochemistry (IHC) analysis, we report, to our knowledge for the first time, that asporin is overexpressed in the stroma of most human breast cancers and is not expressed in normal breast tissue. In vitro, asporin is secreted by breast fibroblasts upon exposure to conditioned medium from some but not all human breast cancer cells. While hormone receptor (HR) positive cells cause strong asporin expression, triple-negative breast cancer (TNBC) cells suppress it. Further, our findings show that soluble IL-1β, secreted by TNBC cells, is responsible for inhibiting asporin in normal and cancer-associated fibroblasts. Using recombinant protein, as well as a synthetic peptide fragment, we demonstrate the ability of asporin to inhibit TGF-β1-mediated SMAD2 phosphorylation, epithelial to mesenchymal transition, and stemness in breast cancer cells. In two in vivo murine models of TNBC, we observed that tumors expressing asporin exhibit significantly reduced growth (2-fold; p = 0.01) and metastatic properties (3-fold; p = 0.045). A retrospective IHC study performed on human breast carcinoma (n = 180) demonstrates that asporin expression is lowest in TNBC and HER2+ tumors, while HR+ tumors have significantly higher asporin expression (4-fold; p = 0.001). Assessment of asporin expression and patient outcome (n = 60; 10-y follow-up) shows that low protein levels in the primary breast lesion significantly delineate patients with bad outcome regardless of the tumor HR status (area under the curve = 0.87; 95% CI 0.78–0.96; p = 0.0001). Survival analysis, based on gene expression (n = 375; 25-y follow-up), confirmed that low asporin levels are associated with a reduced likelihood of survival (hazard ratio = 0.58; 95% CI 0.37–0.91; p = 0.017). Although these data highlight the potential of asporin to serve as a prognostic marker, confirmation of the clinical value would require a prospective study on a much larger patient cohort. Conclusions Our data show that asporin is a stroma-derived inhibitor of TGF-β1 and a tumor suppressor in breast cancer. High asporin expression is significantly associated with less aggressive tumors, stratifying patients according to the clinical outcome. Future pre-clinical studies should consider options for increasing asporin expression in TNBC as a promising strategy for targeted therapy., Andrei Turtoi and colleagues describe a mechanistic role for stroma-derived asporin in breast cancer development., Editors' Summary Background Breast cancer is the most common cancer in women worldwide. Nearly 1.7 million new cases were diagnosed in 2012, and half a million women died from the disease. Breast cancer begins when cells in the breast that normally make milk (epithelial cells) acquire genetic changes that allow them to divide uncontrollably and to move around the body (metastasize). Uncontrolled cell division leads to the formation of a lump that can be detected by mammography (a breast X-ray) or by manual breast examination. Breast cancer is treated by surgical removal of the lump or, if the cancer has started to spread, by removal of the whole breast (mastectomy). After surgery, women often receive chemotherapy or radiotherapy to kill any remaining cancer cells, and women whose tumors express receptors for the female sex hormones estrogen and progesterone or for HER2, a growth factor receptor, are treated with drugs that block these receptors; estrogen, progesterone, and HER2 all control breast cell growth. Nowadays, the prognosis (outlook) for women living in high-income countries who develop breast cancer is generally good—nearly 90% of such women are still alive five years after diagnosis. Why Was This Study Done? The cells surrounding cancer cells—cancer-associated fibroblasts and other components of the stroma—support cancer growth and metastasis and are good targets for new cancer therapies. But, although there is mounting evidence that cancer cells actively adapt the stroma so that it produces various factors the tumor needs to grow and spread, very few molecules produced by the stroma that might serve as targets for drug development have been identified. Here, the researchers investigate whether a molecule called asporin might represent one such target. Asporin, which is highly expressed in the stroma of breast tumors, inhibits a growth factor called TGF-β1. TGF-β1 is involved in maintaining healthy joints, but is also a key molecule in the development of metastatic breast cancer. Most particularly, it modulates an important step in metastasis called the epithelial to mesenchymal transition and it regulates “stemness” in cancer cells. Stem cells are a special type of cell that can multiply indefinitely; tumor cells often look and behave very much like stem cells. What Did the Researchers Do and Find? Using a technique called immunohistochemistry, the researchers first showed that asporin is highly expressed in the stroma of most human breast cancers but not in normal breast tissue. Next, they showed that breast fibroblasts secrete asporin when exposed to conditioned medium from some human breast cancer cell lines (breast cancer cells adapted to grow continuously in the laboratory; conditioned medium is the solution in which cells have been grown). Specifically, conditioned medium from hormone receptor positive cells induced strong asporin expression by breast fibroblasts, whereas medium from breast cancer cells not expressing estrogen or progesterone receptors or HER2 receptors (triple-negative breast cancer cells) suppressed asporin expression. Other experiments showed that TGF-β1 secreted by breast cancer cells induces asporin expression in breast fibroblasts, and that asporin, in turn, inhibits TGF-β1-mediated induction of the epithelial to mesenchymal transition and stemness in breast cancer cells. Triple negative breast cancers appear to inhibit stromal expression of asporin at least in part via expression of the soluble signaling protein interleukin-1β. Notably, in mouse models of triple-negative breast cancer, tumors engineered to express asporin grew slower and metastasized less than tumors not expressing asporin. Finally, among women with breast cancer, asporin expression was low in triple-negative and HER2-positive tumors but significantly higher in hormone receptor positive tumors, and low asporin levels in primary breast lesions were associated with a reduced likelihood of survival independent of hormone receptor and HER2 expression. What Do These Findings Mean? These findings suggest that asporin is a stroma-derived inhibitor of TGF-β1 and a tumor suppressor in breast cancer. Importantly, they also provide preliminary evidence that high asporin expression is associated with less aggressive tumors (hormone receptor positive tumors), whereas low asporin expression is associated with more aggressive tumors (triple negative tumors and HER2-positive tumors). Thus, asporin expression might provide a new prognostic marker for breast cancer. However, before asporin can be used as a biomarker to predict outcomes in women with breast cancer and to identify those women in need of more aggressive treatment, these findings need to be confirmed in large prospective clinical studies. If these findings are confirmed, methods for increasing asporin expression in the stromal tissues of triple negative breast cancer could be a promising strategy for targeted therapy for this group of breast cancers, which currently have a poor prognosis. Additional Information This list of resources contains links that can be accessed when viewing the PDF on a device or via the online version of the article at http://dx.doi.org/10.1371/journal.pmed.1001871. The US National Cancer Institute provides comprehensive information about cancer (in English and Spanish), including detailed information for patients and professionals about breast cancer and an online booklet for patients Cancer Research UK, a not-for-profit organization, provides information about cancer; its detailed information about breast cancer includes sections on tests for hormone receptors and HER2, on treatments that target hormone receptors and treatments that target HER2, and on triple negative breast cancer Breastcancer.org is a not-for-profit organization that provides up-to-date information about breast cancer (in English and Spanish), including information on hormone receptor status, HER2 status, and triple negative breast cancer The UK National Health Service Choices website has information and personal stories about breast cancer; the not-for-profit organization Healthtalk.org also provides personal stories about dealing with breast cancer
- Published
- 2015
- Full Text
- View/download PDF
20. HDAC7 inhibition resets STAT3 tumorigenic activity in human glioblastoma independently of EGFR and PTEN: new opportunities for selected targeted therapies
- Author
-
Brunella Costanza, P Meunier, Philippe Delvenne, Akeila Bellahcene, Vincent Hennequière, Pamela Maris, Sara Rezzola, Ana Palacios, Roberto Ronca, Paul Peixoto, Laurent Schoysman, Andrei Turtoi, Arnaud Blomme, Vincenzo Castronovo, Nicolas Goffart, Elettra Bianchi, Aurélie Henry, Robert N. Muller, Bernard Rogister, Sébastien Boutry, E Di Valentin, Olivier Peulen, Université de Liège, University of Brescia, Centre Hospitalier Universitaire de Liège (CHU-Liège), Université de Mons (UMons), Center for Microscopy and Molecular Imaging (IBMM - CMMI), Université libre de Bruxelles (ULB), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), and CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Angiogenesis ,A Kinase Anchor Proteins ,MESH: Janus Kinase 1 ,Cell Cycle Proteins ,Molecular oncology ,MESH: ErbB Receptors ,Mice ,MESH: Animals ,MESH: Histone Deacetylase Inhibitors ,Janus kinase 1 ,biology ,Neovascularization, Pathologic ,MESH: A Kinase Anchor Proteins ,MESH: Glioblastoma ,MESH: STAT3 Transcription Factor ,Brain ,Cell cycle ,ErbB Receptors ,Tyrosine kinase ,STAT3 Transcription Factor ,MESH: Cell Line, Tumor ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,MESH: PTEN Phosphohydrolase ,Histone Deacetylases ,03 medical and health sciences ,MESH: Brain ,MESH: Cell Cycle Proteins ,Molecular Biology ,Genetics ,Growth factor receptor ,Cell Line, Tumor ,Gene silencing ,PTEN ,Animals ,Humans ,MESH: Mice ,MESH: Humans ,PTEN Phosphohydrolase ,Janus Kinase 1 ,MESH: Male ,MESH: Histone Deacetylases ,Histone Deacetylase Inhibitors ,030104 developmental biology ,Cancer research ,biology.protein ,MESH: Neovascularization, Pathologic ,Glioblastoma ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
International audience; To date, the mutational status of EGFR and PTEN has been shown as relevant for favoring pro- or anti-tumor functions of STAT3 in human glioblastoma multiforme (GBM). We have screened genomic data from 154 patients and have identified a strong positive correlation between STAT3 and HDAC7 expression. In the current work we show the existence of a subpopulation of patients overexpressing HDAC7 and STAT3 that has particularly poor clinical outcome. Surprisingly, the somatic mutation rate of both STAT3 and HDAC7 was insignificant in GBM comparing with EGFR, PTEN or TP53. Depletion of HDAC7 in a range of GBM cells induced the expression of tyrosine kinase JAK1 and the tumor suppressor AKAP12. Both proteins synergistically sustained the activity of STAT3 by inducing its phosphorylation (JAK1) and protein expression (AKAP12). In absence of HDAC7, activated STAT3 was responsible for significant imbalance of secreted pro-/anti-angiogenic factors. This inhibited the migration and sprouting of endothelial cells in paracrine fashion in vitro as well as angiogenesis in vivo. In a murine model of GBM, induced HDAC7-silencing decreased the tumor burden by threefold. The current data show for the first time that silencing HDAC7 can reset the tumor suppressor activity of STAT3, independently of the EGFR/PTEN/TP53 background of the GBM. This effect could be exploited to overcome tumor heterogeneity and provide a new rationale behind the development of specific HDAC7 inhibitors for clinical use.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.