213 research outputs found

    Role of myoferlin in mitochondrial dynamics and metabolic fitness of Pancreatic Ductal Adenocarcinoma

    Full text link
    Pancreatic cancer is the 7th most common cause of cancer mortality in the world. It is predicted to become the 2nd leading cause of cancer-related death in 2030. In the majority of cases, due to a late diagnosis, the tumor is not resectable and already disseminated. Therefore, new specific biomarkers providing early diagnosis for pancreatic cancer are needed. In addition to the lack of specific and early biomarkers, chemotherapies (gemcitabine and folfirinox) poorly improve the overall survival of Pancreatic Ductal Adenocarcinoma (PDAC) patients. Hence, a better understanding of physiopathological processes underlying PDAC is required in order to offer more effective treatments. Myoferlin is a 230 kDa protein with multiple C2 domains known to interact, through calcium binding, with negatively charged phospholipids. This protein was first described in myoblast fusion. Interestingly, Myoferlin is also overexpressed in several cancers, including pancreatic cancer, where it plays a role in endocytosis, exocytosis, and has been located in exosomes. Recently, our team showed a fragmentation of the mitochondrial network in PDAC cells when myoferlin was depleted using siRNA. Understanding the mechanism underlying this mitochondrial disruption would be of great interest as mitochondria are major actors in cancer development, progression and resistance. Owing to the known role of myoferlin in membrane fusion, we assessed its direct involvement in the mitochondrial fusion machinery. Indeed, if myoferlin is a part of the mitochondrial fusion machinery, its silencing together with an unopposed fission would lead to mitochondrial fragmentation. First, we performed immunofluorescence to colocalize myoferlin and a mitochondrial outer membrane 65kDa protein. Colocalization studies showed no significant colocalization. We then performed immunofluorescence to stained myoferlin and the main factor of mitochondrial fusion mitofusin-1/2 (MFN1/2). Colocalization image analysis revealed a 60% colocalization between both proteins. Those results were further confirmed by PLA (Proximity Ligation Assay). Finally, to evaluate a direct protein-protein interaction, we performed a co-immunoprecipitation assay. The main isoform of myoferlin appeared to coimmunoprecipitate with MFN1/2, suggesting a direct interaction between these proteins.Role of myoferlin in mitochondrial dynamics and metabolic fitness of pancreatic cance

    Development of a serum-free co-culture of human intestinal epithelium cell-lines (Caco-2/HT29-5M21)

    Get PDF
    BACKGROUND: The absorptive and goblet cells are the main cellular types encountered in the intestine epithelium. The cell lineage Caco-2 is a model commonly used to reproduce the features of the bowel epithelium. However, there is a strong debate regarding the value of Caco-2 cell culture to mimick in vivo situation. Indeed, some authors report in Caco-2 a low paracellular permeability and an ease of access of highly diffusible small molecules to the microvilli, due to an almost complete lack of mucus. The HT29-5M21 intestinal cell lineage is a mucin-secreting cellular population. A co-culture system carried out in a serum-free medium and comprising both Caco-2 and HT29-5M21 cells was developed. The systematic use of a co-culture system requires the characterization of the monolayer under a given experimental procedure. RESULTS: In this study, we investigated the activity and localization of the alkaline phosphatase and the expression of IAP and MUC5AC genes to determine a correlation between these markers and the cellular composition of a differentiated monolayer obtained from a mixture of Caco-2 and HT29-5M21 cells. We observed that the culture conditions used (serum-free medium) did not change the phenotype of each cell type, and produced a reproducible model. The alkaline phosphatase expression characterizing Caco-2 cells was influenced by the presence of HT29-5M21 cells. CONCLUSION: The culture formed by 75% Caco-2 and 25% HT29-5M21 produce a monolayer containing the two main cell types of human intestinal epithelium and characterized by a reduced permeability to macromolecules

    Myoferlin: an indispensable component in VEGFA secretion by pancreas cancer cells

    Full text link
    In this poster, our laboratory showed the importance of myoferlin, a biomarker of pancreas cancer, in the controle of VEGF-A mediated angiogenesis. Our laboratory showed that silencing myoferlin in pancreas cancer cells, BxPC-3, provoques a decrease in cell prolifération in vitro and a decrease in tumor volumes in animal model. Myoferlin silencing also provokes a decrease in VEGF-A secretion in the conditioned medium and that decrease was abserved in the animal model as a decrease in microvessels dencity. It appeared that this decrease in secretion is due to a a blockage in the exocytosis. Our data also showed a significate correlation between myoferlin expression and microvessels density in patients section

    Myoferlin controls mitochondrial structure and activity in pancreatic ductal adenocarcinoma, and affects tumor aggressiveness

    Full text link
    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

    Myoferlin contributes to the metastatic phenotype of pancreatic cancer cells by enhancing their migratory capacity through the control of oxidative phosphorylation

    Full text link
    Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies with an overall survival of 5%, and is the second cause of death by cancer, mainly linked to its high metastatic aggressiveness. Accordingly, understanding the mechanisms sustaining the PDAC metastatic phenotype remains a priority. In this study, we have generated and used a murine in vivo model to select clones from the human PANC-1 PDAC cell line that exhibit a high propensity to seed and metastasized into the liver. We showed that myoferlin, a protein previously reported to be overexpressed in PDAC, is significantly involved in the migratory abilities of the selected cells. We first report that highly PANC-1 metastatic clones expressed significantly higher myoferlin level than the corresponding low metastatic ones. Using scratch wound and Boyden’s chamber assays, we show that cells expressing high myoferlin level have higher migratory potential than cells characterized by a low myoferlin abundance. Moreover, we demonstrate that myoferlin silencing leads to a migration decrease associated to a reduction of mitochondrial respiration. Since mitochondrial oxidative phosphorylation has been shown to be implicated in the tumor progression and dissemination, our data identify myoferlin as a valid potential therapeutic target in PDAC

    Methylglyoxal: a novel upstream regulator of DNA methylation.

    Full text link
    peer reviewed[en] BACKGROUND: Aerobic glycolysis, also known as the Warburg effect, is predominantly upregulated in a variety of solid tumors, including breast cancer. We have previously reported that methylglyoxal (MG), a very reactive by-product of glycolysis, unexpectedly enhanced the metastatic potential in triple negative breast cancer (TNBC) cells. MG and MG-derived glycation products have been associated with various diseases, such as diabetes, neurodegenerative disorders, and cancer. Glyoxalase 1 (GLO1) exerts an anti-glycation defense by detoxifying MG to D-lactate. METHODS: Here, we used our validated model consisting of stable GLO1 depletion to induce MG stress in TNBC cells. Using genome-scale DNA methylation analysis, we report that this condition resulted in DNA hypermethylation in TNBC cells and xenografts. RESULTS: GLO1-depleted breast cancer cells showed elevated expression of DNMT3B methyltransferase and significant loss of metastasis-related tumor suppressor genes, as assessed using integrated analysis of methylome and transcriptome data. Interestingly, MG scavengers revealed to be as potent as typical DNA demethylating agents at triggering the re-expression of representative silenced genes. Importantly, we delineated an epigenomic MG signature that effectively stratified TNBC patients based on survival. CONCLUSION: This study emphasizes the importance of MG oncometabolite, occurring downstream of the Warburg effect, as a novel epigenetic regulator and proposes MG scavengers to reverse altered patterns of gene expression in TNBC
    • …
    corecore