51 results on '"Fondation ARC"'
Search Results
2. Postcancer rehabilitation: multidisciplinary exercise - programme organisation and feasibility.
- Author
-
Drozd C, Jacquinot Q, Paget-Bailly S, Mansi L, Paillard MJ, Bazan F, Chaigneau L, Dobi E, Viot J, Meynard G, Goujon M, Dalens L, Pereira V, Robin E, Farret J, Gagnepain C, Simon O, Fagnoni-Legat C, Mougin F, Meneveau N, and Curtit E
- Abstract
Background: Although the benefit of supportive care in the postcancer period is now well demonstrated, its implementation in the patient journey remains challenging. This article describes the development, since 2015 and in routine care, of supportive postcancer care comprising a multidisciplinary rehabilitation programme (MRP) based on exercise for patients with early breast cancer., Methods: As part of quality control, we reviewed all patient files since the programme was implemented. Patient data regarding the type of cancer, clinical and pathological factors, and treatment were recorded in a computerised database., Results: From April 2015 to January 2024, 655 patients participated in the MRP. The programme lasts for 14 weeks, totalling 126 hours of face-to-face programme, with a maximum of 8 patients per group, in 5 different centres. A multidisciplinary professional team provide supportive care. The MRP is mainly based on supervised physical exercise and patients also participate in social, psychological, dietary support and educational sessions. Supervised physical exercise includes cardiorespiratory endurance work through specific sessions on ergometers or outdoor walking and adapted physical activity sessions to improve muscular capacities (endurance, strength and flexibility)., Conclusion: We describe here the practical implementation of a routine multidisciplinary supportive care programme, based mainly on physical activity, for post-treatment breast cancer patients. We report almost 9 years of experience with the programme. We show that offering this programme in the postcancer setting and in clinical routine practice is feasible and can be maintained in the long term., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ Group.)
- Published
- 2024
- Full Text
- View/download PDF
3. Botulinum toxin intoxication requires retrograde transport and membrane translocation at the ER in RenVM neurons.
- Author
-
Yeo JC, Tay FP, Bennion R, Loss O, Maignel J, Pons L, Foster K, Beard M, and Bard F
- Subjects
- Animals, Botulinum Toxins, Type A metabolism, Botulinum Toxins, Type A toxicity, Botulinum Toxins, Type A genetics, Rats, Golgi Apparatus metabolism, Cell Line, Cytosol metabolism, Neurons metabolism, Neurons drug effects, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum drug effects, Protein Transport
- Abstract
Botulinum neurotoxin A (BoNT/A) is a highly potent proteolytic toxin specific for neurons with numerous clinical and cosmetic uses. After uptake at the synapse, the protein is proposed to translocate from synaptic vesicles to the cytosol through a self-formed channel. Surprisingly, we found that after intoxication proteolysis of a fluorescent reporter occurs in the neuron soma first and then centrifugally in neurites. To investigate the molecular mechanisms at play, we use a genome-wide siRNA screen in genetically engineered neurons and identify over three hundred genes. An organelle-specific split-mNG complementation indicates BoNT/A traffic from the synapse to the soma-localized Golgi in a retromer-dependent fashion. The toxin then moves to the ER and appears to require the Sec61 complex for retro-translocation to the cytosol. Our study identifies genes and trafficking processes hijacked by the toxin, revealing a new pathway mediating BoNT/A cellular toxicity., Competing Interests: JY, FT, RB No competing interests declared, OL, KF former employee of Ipsen, JM, LP, MB employee of Ipsen, FB Reviewing editor, eLife, (© 2023, Yeo et al.)
- Published
- 2024
- Full Text
- View/download PDF
4. [A role of astral microtubules in the orientation of cell division: when length counts… too!]
- Author
-
Smolen P, Ruiz L, Barai A, Minc N, and Delacour D
- Subjects
- Humans, Animals, Spindle Apparatus physiology, Cell Polarity physiology, Microtubules physiology, Cell Division physiology
- Published
- 2024
- Full Text
- View/download PDF
5. PD-L1 at the crossroad between RNA metabolism and immunosuppression.
- Author
-
Arthur A, Nejmi S, Franchini DM, Espinos E, and Millevoi S
- Subjects
- Humans, Animals, Immune Tolerance, Immunosuppression Therapy, RNA Processing, Post-Transcriptional, Gene Expression Regulation, Neoplastic, G-Quadruplexes, B7-H1 Antigen metabolism, RNA metabolism, RNA genetics, Neoplasms metabolism, Neoplasms genetics, Neoplasms immunology
- Abstract
Programmed death ligand-1 (PD-L1) is a key component of tumor immunosuppression. The uneven therapeutic results of PD-L1 therapy have stimulated intensive studies to better understand the mechanisms underlying altered PD-L1 expression in cancer cells, and to determine whether, beyond its immune function, PD-L1 might have intracellular functions promoting tumor progression and resistance to treatments. In this Opinion, we focus on paradigmatic examples highlighting the central role of PD-L1 in post-transcriptional regulation, with PD-L1 being both a target and an effector of molecular mechanisms featured prominently in RNA research, such as RNA methylation, phase separation and RNA G-quadruplex structures, in order to highlight vulnerabilities on which future anti-PD-L1 therapies could be built., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024. Published by Elsevier Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
6. Trpv6 channel targeting using monoclonal antibody induces prostate cancer cell apoptosis and tumor regression.
- Author
-
Haustrate A, Cordier C, Shapovalov G, Mihalache A, Desruelles E, Soret B, Essonghé NC, Spriet C, Yassine M, Barras A, Marines J, Alcaraz LB, Szunerits S, Robin G, Gosset P, Prevarskaya N, and Lehen'kyi V
- Subjects
- Male, Humans, Animals, Mice, Cell Line, Tumor, Xenograft Model Antitumor Assays, Calpain metabolism, Calcium metabolism, TRPV Cation Channels metabolism, Prostatic Neoplasms pathology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms metabolism, Apoptosis drug effects, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Calcium Channels metabolism
- Abstract
TRPV6 calcium channel is a prospective target in prostate cancer (PCa) since it is not expressed in healthy prostate while its expression increases during cancer progression. Despite the role of TRPV6 in PCa cell survival and apoptotic resistance has been already established, no reliable tool to target TRPV6 channel in vivo and thus to reduce tumor burden is known to date. Here we report the generation of mouse monoclonal antibody mAb82 raised against extracellular epitope of the pore region of the channel. mAb82 inhibited TRPV6 currents by 90% at 24 µg/ml in a dose-dependent manner while decreasing store-operated calcium entry to 56% at only 2.4 µg/ml. mAb82 decreased PCa survival rate in vitro by 71% at 12 µg/ml via inducing cell death through the apoptosis cascade via activation of the protease calpain, following bax activation, mitochondria enlargement, and loss of cristae, Cyt C release, pro-caspase 9 cleavage with the subsequent activation of caspases 3/7. In vivo, mice bearing either PC3M
trpv6+/+ or PC3Mtrpv6-/- +pTRPV6 tumors were successfully treated with mAb82 at the dose as low as 100 µg/kg resulting in a significant reduction tumor growth by 31% and 90%, respectively. The survival rate was markedly improved by 3.5 times in mice treated with mAb82 in PC3Mtrpv6+/+ tumor group and completely restored in PC3Mtrpv6-/- +pTRPV6 tumor group. mAb82 showed a TRPV6-expression dependent organ distribution and virtually no toxicity in the same way as mAbAU1, a control antibody of the same Ig2a isotype. Overall, our data demonstrate for the first time the use of an anti-TRPV6 monoclonal antibody in vitro and in vivo in the treatment of the TRPV6-expressing PCa tumors., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
7. The shaping of mRNA translation plasticity by RNA G-quadruplexes in cancer progression and therapy resistance.
- Author
-
Cammas A, Desprairies A, Dassi E, and Millevoi S
- Abstract
Translational reprogramming in response to oncogenic signaling or microenvironmental stress factors shapes the proteome of cancer cells, enabling adaptation and phenotypic changes underlying cell plasticity, tumor progression and response to cancer therapy. Among the mechanisms regulating translation are RNA G-quadruplexes (RG4s), non-canonical four-stranded structures whose conformational modulation by small molecule ligands and RNA-binding proteins affects the expression of cancer proteins. Here, we discuss the role of RG4s in the regulation of mRNA translation by focusing on paradigmatic examples showing their contribution to adaptive mechanisms of mRNA translation in cancer., (© The Author(s) 2024. Published by Oxford University Press on behalf of NAR Cancer.)
- Published
- 2024
- Full Text
- View/download PDF
8. A fluorogenic substrate for the detection of lipid amidases in intact cells.
- Author
-
Casasampere M, Ung J, Iñáñez A, Dufau C, Tsuboi K, Casas J, Tan SF, Feith DJ, Andrieu-Abadie N, Segui B, Loughran TP Jr, Abad JL, and Fabrias G
- Subjects
- Ethanolamines chemistry, Lipids, Fluorescent Dyes, Amidohydrolases
- Abstract
Lipid amidases of therapeutic relevance include acid ceramidase (AC), N-acylethanolamine-hydrolyzing acid amidase, and fatty acid amide hydrolase (FAAH). Although fluorogenic substrates have been developed for the three enzymes and high-throughput methods for screening have been reported, a platform for the specific detection of these enzyme activities in intact cells is lacking. In this article, we report on the coumarinic 1-deoxydihydroceramide RBM1-151, a 1-deoxy derivative and vinilog of RBM14-C12, as a novel substrate of amidases. This compound is hydrolyzed by AC (
app Km = 7.0 μM;app Vmax = 99.3 nM/min), N-acylethanolamine-hydrolyzing acid amidase (app Km = 0.73 μM;app Vmax = 0.24 nM/min), and FAAH (app Km = 3.6 μM;app Vmax = 7.6 nM/min) but not by other ceramidases. We provide proof of concept that the use of RBM1-151 in combination with reported irreversible inhibitors of AC and FAAH allows the determination in parallel of the three amidase activities in single experiments in intact cells., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
9. Minute virus of mice shows oncolytic activity against pancreatic cancer cells exhibiting a mesenchymal phenotype.
- Author
-
Vienne M, Lopez C, Lulka H, Nevot A, Labrousse G, Dusetti N, Buscail L, and Cordelier P
- Abstract
Pancreatic cancer will soon become the second cause of death by cancer in Western countries. The main barrier to increase the survival of patients with this disease requires the development of novel and efficient therapeutic strategies that better consider tumor biology. In this context, oncolytic viruses emerge as promising therapeutics. Among them, the fibrotropic minute virus of mice prototype (MVMp) preferentially infects migrating and undifferentiated cells that highly resemble poorly differentiated, basal-like pancreatic tumors showing the worst clinical outcome. We report here that MVMp specifically infects, replicates in, and kills pancreatic cancer cells from murine and human origin with a mesenchymal, basal-like profile, while sparing cancer cells with an epithelial phenotype. Remarkably, MVMp infection, at a dose that does not provoke tumor growth inhibition in athymic mice, shows significant antitumoral effect in immune-competent models; extended mouse survival; and promoted the massive infiltration of tumors by innate, myeloid, and cytotoxic T cells that exhibit a less terminally exhausted phenotype. Collectively, we demonstrate herein for the first time that MVMp is specific and oncolytic for pancreatic tumors with mesenchymal, basal-like profile, paving the way for precision-medicine opportunities for the management of the most aggressive and lethal form of this disease., Competing Interests: The authors declare no competing interests., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. Super-enhancer-driven expression of BAHCC1 promotes melanoma cell proliferation and genome stability.
- Author
-
Berico P, Nogaret M, Cigrang M, Lallement A, Vand-Rajabpour F, Flores-Yanke A, Gambi G, Davidson G, Seno L, Obid J, Vokshi BH, Le Gras S, Mengus G, Ye T, Cordero CF, Dalmasso M, Compe E, Bertolotto C, Hernando E, Davidson I, and Coin F
- Subjects
- Humans, Cell Line, Tumor, Regulatory Sequences, Nucleic Acid, Genomic Instability, Cell Proliferation genetics, Gene Expression Regulation, Neoplastic, Enhancer Elements, Genetic, Proteins metabolism, Melanoma pathology
- Abstract
Super-enhancers (SEs) are stretches of enhancers ensuring a high level of expression of key genes associated with cell function. The identification of cancer-specific SE-driven genes is a powerful means for the development of innovative therapeutic strategies. Here, we identify a MITF/SOX10/TFIIH-dependent SE promoting the expression of BAHCC1 in a broad panel of melanoma cells. BAHCC1 is highly expressed in metastatic melanoma and is required for tumor engraftment, growth, and dissemination. Integrative genomics analyses reveal that BAHCC1 is a transcriptional regulator controlling expression of E2F/KLF-dependent cell-cycle and DNA-repair genes. BAHCC1 associates with BRG1-containing remodeling complexes at the promoters of these genes. BAHCC1 silencing leads to decreased cell proliferation and delayed DNA repair. Consequently, BAHCC1 deficiency cooperates with PARP inhibition to induce melanoma cell death. Our study identifies BAHCC1 as an SE-driven gene expressed in melanoma and demonstrates how its inhibition can be exploited as a therapeutic target., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. Tonic repression of collagen I by the bradykinin receptor 2 in skin fibroblasts.
- Author
-
Wong HH, Seet SH, Bascom CC, Isfort RJ, and Bard FA
- Subjects
- Humans, Collagen metabolism, Transforming Growth Factor beta metabolism, Cells, Cultured, Fibroblasts metabolism, Receptors, Bradykinin metabolism, Collagen Type I genetics, Collagen Type I metabolism, Skin metabolism
- Abstract
Imbalance of collagen I expression results in severe pathologies. Apart from activation by the TGFβ-receptor/Smad pathway, control of collagen I expression remains poorly understood. Here, we used human dermal fibroblasts expressing a mCherry fluorescent protein driven by endogenous COL1A1 promoter to functionally screen the kinome and phosphatome. We identify 8 negative regulators, revealing that collagen is under tonic repression. The cell surface receptor BDKRB2 represses collagen I and other pro-fibrotic genes. Interestingly, it also promotes other basal membrane ECM genes. This function is independent of the natural ligand, bradykinin, and of SMAD2/3 factors, instead requiring constant ERK1/2 repression. TGFβ stimulation induces rapid BDKRB2 transcriptional downregulation. Human fibrotic fibroblasts have reduced BDKRB2 levels and enhancing its expression in keloid fibroblasts represses COL1A1. We propose that tonic signalling by BDKRB2 prevents collagen overproduction in skin fibroblasts., Competing Interests: Declaration of Competing Interest None., (Copyright © 2023. Published by Elsevier B.V.)
- Published
- 2023
- Full Text
- View/download PDF
12. TRPV6 Calcium Channel Targeting by Antibodies Raised against Extracellular Epitopes Induces Prostate Cancer Cell Apoptosis.
- Author
-
Haustrate A, Shapovalov G, Spriet C, Cordier C, Kondratskyi A, Noyer L, Foulquier F, Prevarskaya N, and Lehen'kyi V
- Abstract
The TRPV6 calcium channel is known to be up-regulated in various tumors. The efforts to target the TRPV6 channel in vivo are still ongoing to propose an effective therapy against cancer. Here, we report the generation of two antibodies raised against extracellular epitopes corresponding to the extracellular loop between S1 and S2 (rb79) and the pore region (rb82). These antibodies generated a complex biphasic response with the transient activation of the TRPV6 channel. Store-operated calcium entry was consequently potentiated in the prostate cancer cell line LNCaP upon the treatment. Both rb79 and rb82 antibodies significantly decreased cell survival rate in a dose-dependent manner as compared to the control antibodies of the same isotype. This decrease was due to the enhanced cell death via apoptosis revealed using a sub-G1 peak in a cell cycle assay, TUNEL assay, and a Hoechst staining, having no effects in the PC3M
trpv6-/- cell line. Moreover, all TUNEL-positive cells had TRPV6 membrane staining as compared to the control antibody treatment where TRPV6-positive cells were all TUNEL negative. These data clearly demonstrate that TRPV6 channel targeting using rb79 and rb82 antibodies is fatal and may be successfully used in the anticancer therapies.- Published
- 2023
- Full Text
- View/download PDF
13. A Novel Anti-TRPV6 Antibody and Its Application in Cancer Diagnosis In Vitro.
- Author
-
Haustrate A, Mihalache A, Cordier C, Gosset P, Prevarskaya N, and Lehen'kyi V
- Subjects
- Humans, Male, Animals, Rabbits, Immunohistochemistry, Calcium metabolism, TRPV Cation Channels metabolism, Prostatic Neoplasms diagnosis, Prostatic Neoplasms metabolism
- Abstract
Though the first discovery of TRPV6 channel expression in various tissues took place in the early 2000s, reliable tools for its protein detection in various cells and tissues are still missing. Here we show the generation and validation of rabbit polyclonal anti-TRPV6 channel antibodies (rb79-82) against four epitopes of 15 amino acids. Among them, only one antibody, rb79, was capable of detecting the full-length glycosylated form of the TRPV6 channel at around 100 kDa. The generated antibody was shown to be suitable for all in vitro applications, such as immunoblotting, immunoprecipitation, immunocytochemistry, immunofluorescence, etc. One of the most important applications is immunohistochemistry using the paraffin-embedded sections from cancer resection specimens. Using prostate cancer resection specimens, we have confirmed the absence of the TRPV6 protein in both healthy and benign hyperplasia, as well as its expression and correlation to the prostate cancer grades. Thus, the generated rabbit polyclonal anti-TRPV6 channel antibody rb79 is suitable for all in vitro diagnostic applications and particularly for the diagnosis in clinics using paraffin-embedded sections from patients suffering from various diseases and disorders involving the TRPV6 channel.
- Published
- 2022
- Full Text
- View/download PDF
14. Spatial Positioning and Matrix Programs of Cancer-Associated Fibroblasts Promote T-cell Exclusion in Human Lung Tumors.
- Author
-
Grout JA, Sirven P, Leader AM, Maskey S, Hector E, Puisieux I, Steffan F, Cheng E, Tung N, Maurin M, Vaineau R, Karpf L, Plaud M, Begue AL, Ganesh K, Mesple J, Casanova-Acebes M, Tabachnikova A, Keerthivasan S, Lansky A, Berichel JL, Walker L, Rahman AH, Gnjatic S, Girard N, Lefevre M, Damotte D, Adam J, Martin JC, Wolf A, Flores RM, Beasley MB, Pradhan R, Muller S, Marron TU, Turley SJ, Merad M, Kenigsberg E, and Salmon H
- Subjects
- Humans, T-Lymphocytes, Tumor Microenvironment, Immunotherapy methods, Fibroblasts, Cancer-Associated Fibroblasts pathology, Lung Neoplasms pathology
- Abstract
It is currently accepted that cancer-associated fibroblasts (CAF) participate in T-cell exclusion from tumor nests. To unbiasedly test this, we used single-cell RNA sequencing coupled with multiplex imaging on a large cohort of lung tumors. We identified four main CAF populations, two of which are associated with T-cell exclusion: (i) MYH11+αSMA+ CAF, which are present in early-stage tumors and form a single cell layer lining cancer aggregates, and (ii) FAP+αSMA+ CAF, which appear in more advanced tumors and organize in patches within the stroma or in multiple layers around tumor nests. Both populations orchestrate a particular structural tissue organization through dense and aligned fiber deposition compared with T cell-permissive CAF. Yet they produce distinct matrix molecules, including collagen IV (MYH11+αSMA+ CAF) and collagen XI/XII (FAP+αSMA+ CAF). Hereby, we uncovered unique molecular programs of CAF driving T-cell marginalization, whose targeting should increase immunotherapy efficacy in patients bearing T cell-excluded tumors., Significance: The cellular and molecular programs driving T-cell marginalization in solid tumors remain unclear. Here, we describe two CAF populations associated with T-cell exclusion in human lung tumors. We demonstrate the importance of pairing molecular and spatial analysis of the tumor microenvironment, a prerequisite to developing new strategies targeting T cell-excluding CAF. See related commentary by Sherman, p. 2501. This article is highlighted in the In This Issue feature, p. 2483., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
15. Potential Role of Sphingolipidoses-Associated Lysosphingolipids in Cancer.
- Author
-
Dubot P, Astudillo L, Therville N, Carrié L, Pettazzoni M, Cheillan D, Stirnemann J, Levade T, Andrieu-Abadie N, and Sabourdy F
- Abstract
Sphingolipids play a key structural role in cellular membranes and/or act as signaling molecules. Inherited defects of their catabolism lead to lysosomal storage diseases called sphingolipidoses. Although progress has been made toward a better understanding of their pathophysiology, several issues still remain unsolved. In particular, whether lysosphingolipids, the deacylated form of sphingolipids, both of which accumulate in these diseases, are simple biomarkers or play an instrumental role is unclear. In the meanwhile, evidence has been provided for a high risk of developing malignancies in patients affected with Gaucher disease, the most common sphingolipidosis. This article aims at analyzing the potential involvement of lysosphingolipids in cancer. Knowledge about lysosphingolipids in the context of lysosomal storage diseases is summarized. Available data on the nature and prevalence of cancers in patients affected with sphingolipidoses are also reviewed. Then, studies investigating the biological effects of lysosphingolipids toward pro or antitumor pathways are discussed. Finally, original findings exploring the role of glucosylsphingosine in the development of melanoma are presented. While this lysosphingolipid may behave like a protumorigenic agent, further investigations in appropriate models are needed to elucidate the role of these peculiar lipids, not only in sphingolipidoses but also in malignant diseases in general.
- Published
- 2022
- Full Text
- View/download PDF
16. Harsh intertidal environment enhances metabolism and immunity in oyster (Crassostrea gigas) spat.
- Author
-
Corporeau C, Petton S, Vilaça R, Delisle L, Quéré C, Le Roy V, Dubreuil C, Lacas-Gervais S, Guitton Y, Artigaud S, Bernay B, Pichereau V, Huvet A, Petton B, Pernet F, Fleury E, Madec S, Brigaudeau C, Brenner C, and Mazure NM
- Subjects
- Animals, Host-Pathogen Interactions, Immunity, Innate, Proteomics, Crassostrea, Herpesviridae
- Abstract
The Pacific oyster Crassostrea gigas is established in the marine intertidal zone, experiencing rapid and highly dynamic environmental changes throughout the tidal cycle. Depending on the bathymetry, oysters face oxygen deprivation, lack of nutrients, and high changes in temperature during alternation of the cycles of emersion/immersion. Here we showed that intertidal oysters at a bathymetry level of 3 and 5 m delayed by ten days the onset of mortality associated with Pacific Oyster Mortality Syndrome (POMS) as compared to subtidal oysters. Intertidal oysters presented a lower growth but similar energetic reserves to subtidal oysters but induced proteomic changes indicative of a boost in metabolism, inflammation, and innate immunity that may have improved their resistance during infection with the Ostreid herpes virus. Our work highlights that intertidal harsh environmental conditions modify host-pathogen interaction and improve oyster health. This study opens new perspectives on oyster farming for mitigation strategies based on tidal height., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Corporeau Charlotte reports financial support was provided by Ifremer Brittany Centre. Corporeau charlotte reports a relationship with IFREMER that includes: employment., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
17. Combining TNF blockade with immune checkpoint inhibitors in patients with cancer.
- Author
-
Montfort A, Virazels M, Colacios C, Meyer N, and Ségui B
- Subjects
- Humans, Immunotherapy, Immune Checkpoint Inhibitors, Neoplasms drug therapy
- Published
- 2021
- Full Text
- View/download PDF
18. Thrombospondin-1 Silencing Improves Lymphocyte Infiltration in Tumors and Response to Anti-PD-1 in Triple-Negative Breast Cancer.
- Author
-
Marcheteau E, Farge T, Pérès M, Labrousse G, Tenet J, Delmas S, Chusseau M, Duprez-Paumier R, Franchet C, Dalenc F, Imbert C, Noujarède J, Colacios C, Prats H, Cabon F, and Ségui B
- Abstract
Triple-negative breast cancer (TNBC) is notoriously aggressive with a high metastatic potential, and targeted therapies are lacking. Using transcriptomic and histologic analysis of TNBC samples, we found that a high expression of thrombospondin-1 (TSP1), a potent endogenous inhibitor of angiogenesis and an activator of latent transforming growth factor beta (TGF-β), is associated with (i) gene signatures of epithelial-mesenchymal transition and TGF-β signaling, (ii) metastasis and (iii) a reduced survival in TNBC patients. In contrast, in tumors expressing low levels of TSP1, gene signatures of interferon gamma (IFN-γ) signaling and lymphocyte activation were enriched. In TNBC biopsies, TSP1 expression inversely correlated with the CD8+ tumor-infiltrating lymphocytes (TILs) content. In the 4T1 metastatic mouse model of TNBC, TSP1 silencing did not affect primary tumor development but, strikingly, impaired metastasis in immunocompetent but not in immunodeficient nude mice. Moreover, TSP1 knockdown increased tumor vascularization and T lymphocyte infiltration and decreased TGF-β activation in immunocompetent mice. Noteworthy was the finding that TSP1 knockdown increased CD8+ TILs and their programmed cell death 1 (PD-1) expression and sensitized 4T1 tumors to anti-PD-1 therapy. TSP1 inhibition might thus represent an innovative targeted approach to impair TGF-β activation and breast cancer cell metastasis and improve lymphocyte infiltration in tumors, and immunotherapy efficacy in TNBC.
- Published
- 2021
- Full Text
- View/download PDF
19. Neutral Sphingomyelinase 2 Heightens Anti-Melanoma Immune Responses and Anti-PD-1 Therapy Efficacy.
- Author
-
Montfort A, Bertrand F, Rochotte J, Gilhodes J, Filleron T, Milhès J, Dufau C, Imbert C, Riond J, Tosolini M, Clarke CJ, Dufour F, Constantinescu AA, Junior NF, Garcia V, Record M, Cordelier P, Brousset P, Rochaix P, Silvente-Poirot S, Therville N, Andrieu-Abadie N, Levade T, Hannun YA, Benoist H, Meyer N, Micheau O, Colacios C, and Ségui B
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Line, Tumor, Female, Humans, Immunity, Immunotherapy, Melanoma drug therapy, Melanoma pathology, Mice, Mice, Inbred C57BL, Programmed Cell Death 1 Receptor antagonists & inhibitors, Sphingomyelin Phosphodiesterase genetics, Th1 Cells immunology, CD8-Positive T-Lymphocytes immunology, Melanoma immunology, Melanoma metabolism, Sphingomyelin Phosphodiesterase metabolism
- Abstract
Dysregulation of lipid metabolism affects the behavior of cancer cells, but how this happens is not completely understood. Neutral sphingomyelinase 2 (nSMase2), encoded by SMPD3 , catalyzes the breakdown of sphingomyelin to produce the anti-oncometabolite ceramide. We found that this enzyme was often downregulated in human metastatic melanoma, likely contributing to immune escape. Overexpression of nSMase2 in mouse melanoma reduced tumor growth in syngeneic wild-type but not CD8-deficient mice. In wild-type mice, nSMase2-overexpressing tumors showed accumulation of both ceramide and CD8
+ tumor-infiltrating lymphocytes, and this was associated with increased level of transcripts encoding IFNγ and CXCL9. Overexpressing the catalytically inactive nSMase2 failed to alter tumor growth, indicating that the deleterious effect nSMase2 has on melanoma growth depends on its enzymatic activity. In vitro , small extracellular vesicles from melanoma cells overexpressing wild-type nSMase2 augmented the expression of IL12, CXCL9, and CCL19 by bone marrow-derived dendritic cells, suggesting that melanoma nSMase2 triggers T helper 1 (Th1) polarization in the earliest stages of the immune response. Most importantly, overexpression of wild-type nSMase2 increased anti-PD-1 efficacy in murine models of melanoma and breast cancer, and this was associated with an enhanced Th1 response. Therefore, increasing SMPD3 expression in melanoma may serve as an original therapeutic strategy to potentiate Th1 polarization and CD8+ T-cell-dependent immune responses and overcome resistance to anti-PD-1., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
20. An Update on the Role of Ubiquitination in Melanoma Development and Therapies.
- Author
-
Soysouvanh F, Giuliano S, Habel N, El-Hachem N, Pisibon C, Bertolotto C, and Ballotti R
- Abstract
The ubiquitination system plays a critical role in regulation of large array of biological processes and its alteration has been involved in the pathogenesis of cancers, among them cutaneous melanoma, which is responsible for the most deaths from skin cancers. Over the last decades, targeted therapies and immunotherapies became the standard therapeutic strategies for advanced melanomas. However, despite these breakthroughs, the prognosis of metastatic melanoma patients remains unoptimistic, mainly due to intrinsic or acquired resistances. Many avenues of research have been investigated to find new therapeutic targets for improving patient outcomes. Because of the pleiotropic functions of ubiquitination, and because each step of ubiquitination is amenable to pharmacological targeting, much attention has been paid to the role of this process in melanoma development and resistance to therapies. In this review, we summarize the latest data on ubiquitination and discuss the possible impacts on melanoma treatments.
- Published
- 2021
- Full Text
- View/download PDF
21. Combining Nivolumab and Ipilimumab with Infliximab or Certolizumab in Patients with Advanced Melanoma: First Results of a Phase Ib Clinical Trial.
- Author
-
Montfort A, Filleron T, Virazels M, Dufau C, Milhès J, Pagès C, Olivier P, Ayyoub M, Mounier M, Lusque A, Brayer S, Delord JP, Andrieu-Abadie N, Levade T, Colacios C, Ségui B, and Meyer N
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Certolizumab Pegol administration & dosage, Certolizumab Pegol adverse effects, Female, Follow-Up Studies, Humans, Infliximab administration & dosage, Infliximab adverse effects, Ipilimumab administration & dosage, Ipilimumab adverse effects, Male, Melanoma diagnosis, Melanoma mortality, Melanoma secondary, Middle Aged, Nivolumab administration & dosage, Nivolumab adverse effects, Progression-Free Survival, Skin Neoplasms diagnosis, Skin Neoplasms mortality, Skin Neoplasms pathology, Treatment Outcome, Tumor Necrosis Factor-alpha antagonists & inhibitors, Antineoplastic Combined Chemotherapy Protocols adverse effects, Melanoma drug therapy, Skin Neoplasms drug therapy
- Abstract
Purpose: TNF blockers can be used to manage gastrointestinal inflammatory side effects following nivolumab and/or ipilimumab treatment in patients with advanced melanoma. Our preclinical data showed that anti-TNF could promote the efficacy of immune checkpoint inhibitors., Patients and Methods: TICIMEL (NTC03293784) is an open-label, two-arm phase Ib clinical trial. Fourteen patients with advanced and/or metastatic melanoma (stage IIIc/IV) were enrolled. Patients were treated with nivolumab (1 mg/kg) and ipilimumab (3 mg/kg) combined to infliximab (5 mg/kg, N = 6) or certolizumab (400/200 mg, N = 8). The primary endpoint was safety and the secondary endpoint was antitumor activity. Adverse events (AEs) were graded according to the NCI Common Terminology Criteria for Adverse Events and response was assessed following RECIST 1.1., Results: Only one dose-limiting toxicity was observed in the infliximab cohort. The two different combinations were found to be safe. We observed lower treatment-related AEs with infliximab as compared with certolizumab. In the certolizumab cohort, one patient was not evaluable for response. In this cohort, four of eight patients exhibited hepatobiliary disorders and seven of seven evaluable patients achieved objective response including four complete responses (CRs) and three partial responses (PRs). In the infliximab cohort, we observed one CR, two PRs, and three progressive diseases. Signs of activation and maturation of systemic T-cell responses were seen in patients from both cohorts., Conclusions: Our results show that both combinations are safe in human and provide clinical and biological activities. The high response rate in the certolizumab-treated patient cohort deserves further investigations., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
22. Lipid metabolic Reprogramming: Role in Melanoma Progression and Therapeutic Perspectives.
- Author
-
Pellerin L, Carrié L, Dufau C, Nieto L, Ségui B, Levade T, Riond J, and Andrieu-Abadie N
- Abstract
Metabolic reprogramming contributes to the pathogenesis and heterogeneity of melanoma. It is driven both by oncogenic events and the constraints imposed by a nutrient- and oxygen-scarce microenvironment. Among the most prominent metabolic reprogramming features is an increased rate of lipid synthesis. Lipids serve as a source of energy and form the structural foundation of all membranes, but have also emerged as mediators that not only impact classical oncogenic signaling pathways, but also contribute to melanoma progression. Various alterations in fatty acid metabolism have been reported and can contribute to melanoma cell aggressiveness. Elevated expression of the key lipogenic fatty acid synthase is associated with tumor cell invasion and poor prognosis. Fatty acid uptake from the surrounding microenvironment, fatty acid β-oxidation and storage also appear to play an essential role in tumor cell migration. The aim of this review is ( i ) to focus on the major alterations affecting lipid storage organelles and lipid metabolism. A particular attention has been paid to glycerophospholipids, sphingolipids, sterols and eicosanoids, ( ii ) to discuss how these metabolic dysregulations contribute to the phenotype plasticity of melanoma cells and/or melanoma aggressiveness, and ( iii ) to highlight therapeutic approaches targeting lipid metabolism that could be applicable for melanoma treatment.
- Published
- 2020
- Full Text
- View/download PDF
23. Retraction Note: LAMP2 expression dictates azacytidine response and prognosis in MDS/AML.
- Author
-
Dubois A, Furstoss N, Calleja A, Zerhouni M, Cluzeau T, Savy C, Marchetti S, Hamouda MA, Boulakirba S, Orange F, Lacas-Gervais S, Karsenti JM, Mounier N, Tamburini J, Puissant A, Luciano F, Jacquel A, Auberger P, and Robert G
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
24. New Insights into the Role of Sphingolipid Metabolism in Melanoma.
- Author
-
Carrié L, Virazels M, Dufau C, Montfort A, Levade T, Ségui B, and Andrieu-Abadie N
- Subjects
- Humans, Melanoma, Cutaneous Malignant, Melanoma drug therapy, Skin Neoplasms drug therapy, Sphingolipids metabolism
- Abstract
Cutaneous melanoma is a deadly skin cancer whose aggressiveness is directly linked to its metastatic potency. Despite remarkable breakthroughs in term of treatments with the emergence of targeted therapy and immunotherapy, the prognosis for metastatic patients remains uncertain mainly because of resistances. Better understanding the mechanisms responsible for melanoma progression is therefore essential to uncover new therapeutic targets. Interestingly, the sphingolipid metabolism is dysregulated in melanoma and is associated with melanoma progression and resistance to treatment. This review summarises the impact of the sphingolipid metabolism on melanoma from the initiation to metastatic dissemination with emphasis on melanoma plasticity, immune responses and resistance to treatments.
- Published
- 2020
- Full Text
- View/download PDF
25. Towards a cancer mission in Horizon Europe: recommendations.
- Author
-
Berns A, Ringborg U, Celis JE, Heitor M, Aaronson NK, Abou-Zeid N, Adami HO, Apostolidis K, Baumann M, Bardelli A, Bernards R, Brandberg Y, Caldas C, Calvo F, Dive C, Eggert A, Eggermont A, Espina C, Falkenburg F, Foucaud J, Hanahan D, Helbig U, Jönsson B, Kalager M, Karjalainen S, Kásler M, Kearns P, Kärre K, Lacombe D, de Lorenzo F, Meunier F, Nettekoven G, Oberst S, Nagy P, Philip T, Price R, Schüz J, Solary E, Strang P, Tabernero J, and Voest E
- Subjects
- Cancer Survivors, Clinical Trials as Topic, Europe, Humans, Neoplasms prevention & control, Neoplasms psychology, Neoplasms rehabilitation, Organizational Innovation, Palliative Care, Patient Participation, Specialization, Translational Research, Biomedical, Neoplasms therapy
- Abstract
A comprehensive translational cancer research approach focused on personalized and precision medicine, and covering the entire cancer research-care-prevention continuum has the potential to achieve in 2030 a 10-year cancer-specific survival for 75% of patients diagnosed in European Union (EU) member states with a well-developed healthcare system. Concerted actions across this continuum that spans from basic and preclinical research through clinical and prevention research to outcomes research, along with the establishment of interconnected high-quality infrastructures for translational research, clinical and prevention trials and outcomes research, will ensure that science-driven and social innovations benefit patients and individuals at risk across the EU. European infrastructures involving comprehensive cancer centres (CCCs) and CCC-like entities will provide researchers with access to the required critical mass of patients, biological materials and technological resources and can bridge research with healthcare systems. Here, we prioritize research areas to ensure a balanced research portfolio and provide recommendations for achieving key targets. Meeting these targets will require harmonization of EU and national priorities and policies, improved research coordination at the national, regional and EU level and increasingly efficient and flexible funding mechanisms. Long-term support by the EU and commitment of Member States to specialized schemes are also needed for the establishment and sustainability of trans-border infrastructures and networks. In addition to effectively engaging policymakers, all relevant stakeholders within the entire continuum should consensually inform policy through evidence-based advice., (© 2020 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
26. Conditional Gene Targeting Reveals Cell Type-Specific Roles of the Lysosomal Protease Cathepsin L in Mammary Tumor Progression.
- Author
-
Parigiani MA, Ketscher A, Timme S, Bronsert P, Schlimpert M, Kammerer B, Jacquel A, Chaintreuil P, and Reinheckel T
- Abstract
Background: Cathepsin L (Ctsl) is a cysteine protease mainly located within the endosomal/lysosomal cell compartment. High expression of Ctsl indicates poor prognosis in human breast cancer. However, the cell type-specific Ctsl functions responsible for this association remain elusive. Methods: Because constitutive Ctsl
-/- mice develop a complex phenotype, we developed a conditional model allowing for cell type-specific inactivation of Ctsl in mammary epithelium or myeloid cells in the transgenic mouse mammary tumor virus (MMTV)-polyoma middle T (PyMT) breast cancer model. Results: Ctsl ablation in mammary epithelial cells resulted in delayed initiation and end-stage of cancers. The latter displayed large dead cell areas. Inducible in vitro deletion of Ctsl in MMTV-PyMT-derived breast cancer cells revealed expansion of the acidic cell compartment, alteration of intracellular amino acid levels, and impaired mTOR signaling. In consequence, Ctsl-deficient cells exhibited slow growth rates and high apoptosis susceptibility. In contrast to Ctsl-deficient mammary epithelium, selective knockout of Ctsl in myeloid cells had no effects on primary tumors, but promoted lung metastasis formation. Conclusions: Our cell type-specific in vivo analysis provides strong evidence for a cancer cell-intrinsic, tumor-promoting role of Ctsl in primary breast cancer, whereas metastasis is negatively regulated by Ctsl expressed by bone marrow-derived cells.- Published
- 2020
- Full Text
- View/download PDF
27. Retraction notice to "TRPV6 calcium channel regulation, downstream pathways, and therapeutic targeting in cancer" [Cell Calcium (2019) 117-124].
- Author
-
Haustrate A, Hantute-Ghesquier A, Prevarskaya N, and Lehen'kyi V
- Published
- 2020
- Full Text
- View/download PDF
28. Monoclonal Antibodies Targeting Ion Channels and Their Therapeutic Potential.
- Author
-
Haustrate A, Hantute-Ghesquier A, Prevarskaya N, and Lehen'kyi V
- Abstract
Monoclonal antibodies (mAbs) represent a rapidly growing pharmaceutical class of protein drugs that becomes an important part of the precision therapy. mAbs are characterized by their high specificity and affinity for the target antigen, which is mostly present on the cell surface. Ion channels are a large family of transmembrane proteins that control ion transport across the cell membrane. They are involved in almost all biological processes in both health and disease and are widely considered as prospective targets. However, no antibody-based drug targeting ion channel has been developed so far that has progressed to clinical use. Thus, we provide a comprehensive review of the elaborated mAbs against ion channels, describe their mechanisms of action, and discuss their therapeutic potential.
- Published
- 2019
- Full Text
- View/download PDF
29. LAMP2 expression dictates azacytidine response and prognosis in MDS/AML.
- Author
-
Dubois A, Furstoss N, Calleja A, Zerhouni M, Cluzeau T, Savy C, Marchetti S, Hamouda MA, Boulakirba S, Orange F, Lacas-Gervais S, Karsenti JM, Mounier N, Tamburini J, Puissant A, Luciano F, Jacquel A, Auberger P, and Robert G
- Subjects
- Aged, Aged, 80 and over, Apoptosis, Biomarkers, Tumor genetics, Cell Proliferation, Female, Follow-Up Studies, Humans, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute metabolism, Lysosomal-Associated Membrane Protein 2 genetics, Male, Middle Aged, Prognosis, Survival Rate, Tumor Cells, Cultured, Antimetabolites, Antineoplastic pharmacology, Azacitidine pharmacology, Biomarkers, Tumor metabolism, Gene Expression Regulation, Neoplastic drug effects, Leukemia, Myeloid, Acute pathology, Lysosomal-Associated Membrane Protein 2 metabolism
- Abstract
Chaperone-mediated autophagy (CMA) is a highly selective form of autophagy. During CMA, the HSC70 chaperone carries target proteins endowed with a KFERQ-like motif to the lysosomal receptor LAMP2A, which then translocate them into lysosomes for degradation. In the present study, we scrutinized the mechanisms underlying the response and resistance to Azacytidine (Aza) in MDS/AML cell lines and bone marrow CD34
+ blasts from MDS/AML patients. In engineered Aza-resistant MDS cell lines and some AML cell lines, we identified a profound defect in CMA linked to the absence of LAMP2A. LAMP2 deficiency was responsible for Aza resistance and hypersensitivity to lysosome and autophagy inhibitors. Accordingly, gain of function of LAMP2 in deficient cells or loss of function in LAMP2-expressing cells rendered them sensitive or resistant to Aza, respectively. A strict correlation was observed between the absence of LAMP2, resistance to Aza and sensitivity to lysosome inhibitors. Low levels of LAMP2 expression in CD34+ blasts from MDS/AML patients correlated with lack of sensitivity to Aza and were predictive of poor overall survival. We propose that CD34+ /LAMP2Low patients at diagnosis or who become CD34+ /LAMP2Low during the course of treatment with Aza might benefit from a lysosome inhibitor already used in the clinic.- Published
- 2019
- Full Text
- View/download PDF
30. RETRACTED: TRPV6 calcium channel regulation, downstream pathways, and therapeutic targeting in cancer.
- Author
-
Haustrate A, Hantute-Ghesquier A, Prevarskaya N, and Lehen'kyi V
- Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editors in Chief. This article was retracted because of inappropriate use of confidential material and text available to one of the authors through the review of “TRPV6 As A Target For Cancer Therapy”, John M Stewart, J. Cancer, online date 2019-5-13; doi:10.7150/jca.31640., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
31. [The oyster Crassostrea gigas, a new model against cancer].
- Author
-
Corporeau C, Huvet A, Pichereau V, Delisle L, Quéré C, Dubreuil C, Artigaud S, Brenner C, Meyenberg Cunha-De Padua M, and Mazure N
- Subjects
- Animals, Apoptosis, Cell Proliferation, Cellular Microenvironment, Cellular Reprogramming, Glycolysis, Crassostrea physiology, Disease Models, Animal, Neoplasms
- Abstract
The Warburg effect is one of the hallmarks of cancer cells in humans. It is a true metabolic reprogramming to aerobic glycolysis, allowing cancer cells to meet their particular energy needs for growth, proliferation, and resistance to apoptosis, depending on the microenvironment they encounter within the tumor. We have recently discovered that the Crassostrea gigas oyster can naturally reprogram its metabolism to the Warburg effect. Thus, the oyster becomes a new invertebrate model useful for cancer research. Due to its lifestyle, the oyster C. gigas has special abilities to adapt its metabolism to the extreme changes in the environment in which it is located. The oyster C. gigas is therefore a model of interest to study how the environment can control the Warburg effect under conditions that could not be explored in vertebrate model species., (© 2019 médecine/sciences – Inserm.)
- Published
- 2019
- Full Text
- View/download PDF
32. [Precision medicine in oncology: Challenges, stakes and new paradigms].
- Author
-
Cox S, Rousseau-Tsangaris M, Abou-Zeid N, Dalle S, Leurent P, Cutivet A, Le HH, Kotb S, Bogaert B, Gardette R, Baran Y, Holder JM, Lerner L, Blay JY, Cambrosio A, Tredan O, and Denèfle P
- Subjects
- Humans, Neoplasms genetics, Clinical Trials as Topic, Medical Oncology, Neoplasms therapy, Precision Medicine
- Published
- 2019
- Full Text
- View/download PDF
33. Author Correction: Necroptosis microenvironment directs lineage commitment in liver cancer.
- Author
-
Seehawer M, Heinzmann F, D'Artista L, Harbig J, Roux PF, Hoenicke L, Dang H, Klotz S, Robinson L, Doré G, Rozenblum N, Kang TW, Chawla R, Buch T, Vucur M, Roth M, Zuber J, Luedde T, Sipos B, Longerich T, Heikenwälder M, Wang XW, Bischof O, and Zender L
- Abstract
In this Article, the pCaMIN construct consisted of 'mouse MYC and mouse Nras
G12V ' instead of 'mouse Myc and human NRASG12V ; and the pCAMIA construct consisted of 'mouse Myc and human AKT1' instead of 'mouse Myc and Akt1' this has been corrected online.- Published
- 2018
- Full Text
- View/download PDF
34. Necroptosis microenvironment directs lineage commitment in liver cancer.
- Author
-
Seehawer M, Heinzmann F, D'Artista L, Harbig J, Roux PF, Hoenicke L, Dang H, Klotz S, Robinson L, Doré G, Rozenblum N, Kang TW, Chawla R, Buch T, Vucur M, Roth M, Zuber J, Luedde T, Sipos B, Longerich T, Heikenwälder M, Wang XW, Bischof O, and Zender L
- Subjects
- Animals, Carcinogenesis genetics, Carcinoma, Hepatocellular genetics, Cell Differentiation, Cholangiocarcinoma genetics, Cyclin-Dependent Kinase Inhibitor p16 deficiency, Cytokines metabolism, DNA Transposable Elements genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Disease Models, Animal, Epigenesis, Genetic genetics, Female, Gene Expression Profiling, Genes, myc, Genes, ras, Hepatocytes metabolism, Hepatocytes pathology, Humans, Liver Neoplasms genetics, Male, Mice, Mosaicism, Proto-Oncogene Proteins c-akt genetics, T-Box Domain Proteins genetics, T-Box Domain Proteins metabolism, Transcription Factors genetics, Transcription Factors metabolism, Apoptosis genetics, Carcinoma, Hepatocellular pathology, Cell Lineage genetics, Cholangiocarcinoma pathology, Liver Neoplasms pathology, Necrosis genetics, Tumor Microenvironment
- Abstract
Primary liver cancer represents a major health problem. It comprises hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC), which differ markedly with regards to their morphology, metastatic potential and responses to therapy. However, the regulatory molecules and tissue context that commit transformed hepatic cells towards HCC or ICC are largely unknown. Here we show that the hepatic microenvironment epigenetically shapes lineage commitment in mosaic mouse models of liver tumorigenesis. Whereas a necroptosis-associated hepatic cytokine microenvironment determines ICC outgrowth from oncogenically transformed hepatocytes, hepatocytes containing identical oncogenic drivers give rise to HCC if they are surrounded by apoptotic hepatocytes. Epigenome and transcriptome profiling of mouse HCC and ICC singled out Tbx3 and Prdm5 as major microenvironment-dependent and epigenetically regulated lineage-commitment factors, a function that is conserved in humans. Together, our results provide insight into lineage commitment in liver tumorigenesis, and explain molecularly why common liver-damaging risk factors can lead to either HCC or ICC.
- Published
- 2018
- Full Text
- View/download PDF
35. Implication and Regulation of AMPK during Physiological and Pathological Myeloid Differentiation.
- Author
-
Jacquel A, Luciano F, Robert G, and Auberger P
- Subjects
- AMP-Activated Protein Kinases chemistry, Animals, Enzyme Activation, Hematologic Neoplasms pathology, Hematopoietic Stem Cells pathology, Humans, AMP-Activated Protein Kinases metabolism, Cell Differentiation, Myeloid Cells enzymology, Myeloid Cells pathology
- Abstract
AMP-activated protein kinase (AMPK) is a heterotrimeric serine/threonine kinase consisting of the arrangement of various α β, and γisoforms that are expressed differently depending on the tissue or the cell lineage. AMPK is one of the major sensors of energy status in mammalian cells and as such plays essential roles in the regulation of cellular homeostasis, metabolism, cell growth, differentiation, apoptosis, and autophagy. AMPK is activated by two upstream kinases, the tumor suppressor liver kinase B1 (LKB1) and the calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) through phosphorylation of the kinase on Thr172, leading to its activation. In addition, AMPK inhibits the mTOR pathway through phosphorylation and activation of tuberous sclerosis protein 2 (TSC2) and causes direct activation of unc-51-like autophagy activating kinase 1 (ULK1) via phosphorylation of Ser555, thus promoting initiation of autophagy. Although it is well established that AMPK can control the differentiation of different cell lineages, including hematopoietic stem cells (HSCs), progenitors, and mature hematopoietic cells, the role of AMPK regarding myeloid cell differentiation is less documented. The differentiation of monocytes into macrophages triggered by colony stimulating factor 1 (CSF-1), a process during which both caspase activation (independently of apoptosis induction) and AMPK-dependent stimulation of autophagy are necessary, is one noticeable example of the involvement of AMPK in the physiological differentiation of myeloid cells. The present review focuses on the role of AMPK in the regulation of the physiological and pathological differentiation of myeloid cells. The mechanisms of autophagy induction by AMPK will also be addressed, as autophagy has been shown to be important for differentiation of hematopoietic cells. In addition, myeloid malignancies (myeloid leukemia or dysplasia) are characterized by profound defects in the establishment of proper differentiation programs. Reinduction of a normal differentiation process in myeloid malignancies has thus emerged as a valuable and promising therapeutic strategy. As AMPK seems to exert a key role in the differentiation of myeloid cells, notably through induction of autophagy, we will also discuss the potential to target this pathway as a pro-differentiating and anti-leukemic strategy in myeloid malignancies.
- Published
- 2018
- Full Text
- View/download PDF
36. The IGF2/IR/IGF1R Pathway in Tumor Cells and Myofibroblasts Mediates Resistance to EGFR Inhibition in Cholangiocarcinoma.
- Author
-
Vaquero J, Lobe C, Tahraoui S, Clapéron A, Mergey M, Merabtene F, Wendum D, Coulouarn C, Housset C, Desbois-Mouthon C, Praz F, and Fouassier L
- Subjects
- Animals, Cancer-Associated Fibroblasts drug effects, Cancer-Associated Fibroblasts pathology, Cell Line, Tumor, Cell Proliferation drug effects, Cholangiocarcinoma genetics, Cholangiocarcinoma pathology, Drug Resistance, Neoplasm genetics, Epithelial-Mesenchymal Transition drug effects, ErbB Receptors antagonists & inhibitors, ErbB Receptors genetics, Gene Expression Regulation, Neoplastic drug effects, Heterografts, Humans, Mice, Myofibroblasts drug effects, Protein Kinase Inhibitors pharmacology, Receptor, IGF Type 1, Signal Transduction drug effects, Cholangiocarcinoma drug therapy, Insulin-Like Growth Factor II genetics, Receptor, Insulin genetics, Receptors, Somatomedin genetics
- Abstract
Purpose: Cholangiocarcinoma (CCA) is a desmoplastic tumor of the biliary tree in which epidermal growth factor receptor (EGFR) is overexpressed and contributes to cancer progression. Although EGFR has been envisaged as a target for therapy, treatment with tyrosine kinase inhibitors (TKI) such as erlotinib did not provide therapeutic benefit in patients with CCA, emphasizing the need to investigate resistance mechanisms against EGFR inhibition. Experimental Design: Resistant CCA cells to EGFR inhibition were obtained upon long-time exposure of cells with erlotinib. Cell signaling, viability, migration, and spheroid growth were determined in vitro , and tumor growth was evaluated in CCA xenograft models. Results: Erlotinib-resistant CCA cells displayed metastasis-associated signatures that correlated with a marked change in cell plasticity associated with an epithelial-mesenchymal transition (EMT) and a cancer stem cell (CSC)-like phenotype. Resistant cells exhibited an upregulation of insulin receptor (IR) and insulin-like growth factor (IGF) 1 receptor (IGF1R), along with an increase in IGF2 expression. IR/IGF1R inhibition reduced EMT and CSC-like traits in resistant cells. In vivo , tumors developed from resistant CCA cells were larger and exhibited a more prominent stromal compartment, enriched in cancer-associated fibroblasts (CAF). Pharmacological coinhibition of EGFR and IR/IGF1R reduced tumor growth and stromal compartment in resistant tumors. Modeling of CCA-CAF crosstalk showed that IGF2 expressed by fibroblasts boosted IR/IGF1R signaling in resistant cells. Furthermore, IR/IGF1R signaling positively regulated fibroblast proliferation and activation. Conclusions: To escape EGFR-TKI treatment, CCA tumor cells develop an adaptive mechanism by undergoing an IR/IGF1R-dependent phenotypic switch, involving a contribution of stromal cells. Clin Cancer Res; 24(17); 4282-96. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
37. TRPM Family Channels in Cancer.
- Author
-
Hantute-Ghesquier A, Haustrate A, Prevarskaya N, and Lehen'kyi V
- Abstract
Members of the TRPM ("Melastatin") family fall into the subclass of the TRP channels having varying permeability to Ca
2+ and Mg2+ , with three members of the TRPM family being chanzymes, which contain C-terminal enzyme domains. The role of different TRPM members has been shown in various cancers such as prostate cancer for mostly TRPM8 and TRPM2, breast cancer for mostly TRPM2 and TRPM7, and pancreatic cancer for TRPM2/7/8 channels. The role of TRPM5 channels has been shown in lung cancer, TRPM1 in melanoma, and TRPM4 channel in prostate cancer as well. Thus, the TRPM family of channels may represent an appealing target for the anticancer therapy.- Published
- 2018
- Full Text
- View/download PDF
38. MicroRNA-506 promotes primary biliary cholangitis-like features in cholangiocytes and immune activation.
- Author
-
Erice O, Munoz-Garrido P, Vaquero J, Perugorria MJ, Fernandez-Barrena MG, Saez E, Santos-Laso A, Arbelaiz A, Jimenez-Agüero R, Fernandez-Irigoyen J, Santamaria E, Torrano V, Carracedo A, Ananthanarayanan M, Marzioni M, Prieto J, Beuers U, Oude Elferink RP, LaRusso NF, Bujanda L, Marin JJG, and Banales JM
- Subjects
- Apoptosis, Bile Ducts, Intrahepatic metabolism, Cell Culture Techniques, Cell Migration Assays, Cell Proliferation, Cytokines metabolism, Fluorescent Antibody Technique, Gene Expression Regulation genetics, Humans, Immunoblotting, Mass Spectrometry, Oxidative Stress, Proteomics, Signal Transduction genetics, Bile Ducts, Intrahepatic pathology, Epithelial Cells metabolism, Liver Cirrhosis, Biliary metabolism, MicroRNAs metabolism
- Abstract
Primary biliary cholangitis (PBC) is a chronic cholestatic liver disease associated with autoimmune phenomena targeting intrahepatic bile duct cells (cholangiocytes). Although its etiopathogenesis remains obscure, development of antimitochondrial autoantibodies against pyruvate dehydrogenase complex E2 is a common feature. MicroRNA (miR) dysregulation occurs in liver and immune cells of PBC patients, but its functional relevance is largely unknown. We previously reported that miR-506 is overexpressed in PBC cholangiocytes and directly targets both Cl
- / HCO3- anion exchanger 2 and type III inositol 1,4,5-trisphosphate receptor, leading to cholestasis. Here, the regulation of miR-506 gene expression and its role in cholangiocyte pathophysiology and immune activation was studied. Several proinflammatory cytokines overexpressed in PBC livers (such as interleukin-8 [IL8], IL12, IL17, IL18, and tumor necrosis factor alpha) stimulated miR-506 promoter activity in human cholangiocytes, as revealed by luciferase reporter assays. Experimental overexpression of miR-506 in cholangiocytes dysregulated the cell proteomic profile (by mass spectrometry), affecting proteins involved in different biological processes including mitochondrial metabolism. In cholangiocytes, miR-506 (1) induced dedifferentiation with down-regulation of biliary and epithelial markers together with up-regulation of mesenchymal, proinflammatory, and profibrotic markers; (2) impaired cell proliferation and adhesion; (3) increased oxidative and endoplasmic reticulum stress; (4) caused DNA damage; and (5) sensitized to caspase-3-dependent apoptosis induced by cytotoxic bile acids. These events were also associated with impaired energy metabolism in mitochondria (proton leak and less adenosine triphosphate production) and pyruvate dehydrogenase complex E2 overexpression. Coculture of miR-506 overexpressing cholangiocytes with PBC immunocytes induced activation and proliferation of PBC immunocytes., Conclusion: Different proinflammatory cytokines enhance the expression of miR-506 in biliary epithelial cells; miR-506 induces PBC-like features in cholangiocytes and promotes immune activation, representing a potential therapeutic target for PBC patients. (Hepatology 2018;67:1420-1440)., (© 2017 by the American Association for the Study of Liver Diseases.)- Published
- 2018
- Full Text
- View/download PDF
39. Role of ErbB/HER family of receptor tyrosine kinases in cholangiocyte biology.
- Author
-
Pellat A, Vaquero J, and Fouassier L
- Subjects
- Animals, Bile Duct Diseases etiology, Bile Ducts cytology, ErbB Receptors antagonists & inhibitors, ErbB Receptors genetics, Humans, Liver Regeneration, Receptor, ErbB-2 physiology, Receptor, ErbB-3 physiology, Receptor, ErbB-4 physiology, Signal Transduction physiology, Tumor Microenvironment, Bile Ducts physiology, Epithelial Cells physiology, ErbB Receptors physiology
- Abstract
The ErbB/HER family comprises four distinct tyrosine kinase receptors, EGFR/ErbB1/HER1, ErbB2/HER2, ErbB3/HER3, and ErbB4/HER4, which trigger intracellular signals at the origin of essential cellular functions, including differentiation, proliferation, survival, and migration. Epithelial cells, named cholangiocytes, that line intrahepatic and extrahepatic bile ducts, contribute substantially to biliary secretory functions and bile transport. Although ErbB receptors have been widely studied in cholangiocarcinoma (CCA), a malignancy of the biliary tract, knowledge of these receptors in biliary epithelium physiology and in non-malignant cholangiopathies is far from complete. Current knowledge suggests a role for epidermal growth factor receptor (EGFR) in cholangiocyte specification and proliferation, and in hepatocyte transdifferentiation into cholangiocytes during liver regeneration to restore biliary epithelium integrity. High expression and activation of EGFR and/or ErbB2 were recently demonstrated in biliary lithiasis and primary sclerosing cholangitis, two cholangiopathies regarded as risk factors for CCA. In CCA, ErbB receptors are frequently overexpressed, leading to tumor progression and low prognosis. Anti-ErbB therapies were efficient only in preclinical trials and have suggested the existence of resistance mechanisms with the need to identify predictive factors of therapy response. This review aims to compile the current knowledge on the functions of ErbB receptors in physiology and physiopathology of the biliary epithelium. (Hepatology 2018;67:762-773)., (© 2017 by the American Association for the Study of Liver Diseases.)
- Published
- 2018
- Full Text
- View/download PDF
40. Nuclear Translocation of Argonaute 2 in Cytokine-Induced Senescence.
- Author
-
Rentschler M, Chen Y, Pahl J, Soria-Martinez L, Braumüller H, Brenner E, Bischof O, Röcken M, and Wieder T
- Subjects
- Active Transport, Cell Nucleus, Cell Proliferation, Cell Survival, Humans, Interferon-gamma metabolism, MCF-7 Cells, Tumor Necrosis Factors metabolism, Argonaute Proteins metabolism, Cellular Senescence, Cytokines metabolism, Neoplasms metabolism
- Abstract
Background/aims: Cellular senescence, or permanent growth arrest, is known as an effective tumor suppressor mechanism that can be induced by different stressors, such as oncogenes, chemotherapeutics or cytokine cocktails. Previous studies demonstrated that the growth-repressing state of oncogene-induced senescent cells depends on argonaute protein 2 (Ago2)-mediated transcriptional gene silencing and Ago2/Rb corepression of E2F-dependent cell cycle genes. Cytokine-induced senescence (CIS) likewise depends on activation of the p16Ink4a/Rb pathway, and consecutive inactivation of the E2F family of transcription factors. In the present study, we therefore analyzed the role of Ago2 in CIS., Methods: Human cancer cell lines were treated with interferon-gamma (IFN-γ) and tumor necrosis factor (TNF) to induce senescence. Senescence was determined by growth assays and measurement of senescence-associated β-galactosidase (SA-β-gal) activity, Ago2 translocation by Ago2/ Ki67 immunofluorescence staining and western blot analysis, and gene transcription by quantitative polymerase chain reaction (qPCR)., Results: IFN-γ and TNF permanently stopped cell proliferation and time-dependently increased SA-β-gal activity. After 24 - 48 h of cytokine treatment, Ago2 translocated from the cytoplasm into the nucleus of Ki67-negative cells, an effect which was shown to be reversible. Importantly, the proinflammatory cytokine cocktail suppressed Ago2-regulated cell cycle control genes, and siRNA-mediated depletion of Ago2 interfered with cytokine-induced growth inhibition., Conclusion: IFN-γ and TNF induce a stable cell cycle arrest of cancer cells that is accompanied by a fast nuclear Ago2 translocation and repression of Ago2-regulated cell cycle control genes. As Ago2 downregulation impairs cytokine-induced growth regulation, Ago2 may contribute to tissue homeostasis in human cancers., (© 2018 The Author(s). Published by S. Karger AG, Basel.)
- Published
- 2018
- Full Text
- View/download PDF
41. ATP-competitive Plk1 inhibitors induce caspase 3-mediated Plk1 cleavage and activation in hematopoietic cell lines.
- Author
-
Dufies M, Ambrosetti D, Boulakirba S, Calleja A, Savy C, Furstoss N, Zerhouni M, Parola J, Aira-Diaz L, Marchetti S, Orange F, Lacas-Gervais S, Luciano F, Jacquel A, Robert G, Pagès G, and Auberger P
- Abstract
Polo-like kinases (Plks) define a highly conserved family of Ser/Thr kinases with crucial roles in the regulation of cell division. Here we show that Plk1 is cleaved by caspase 3, but not by other caspases in different hematopoietic cell lines treated with competitive inhibitors of the ATP-binding pocket of Plk1. Intriguingly, Plk1 was not cleaved in cells treated with Rigosertib, a non-competitive inhibitor of Plk1, suggesting that binding of the inhibitor to the ATP binding pocket of Plk1 triggers a conformational change and unmasks a cryptic caspase 3 cleavage site on the protein. Cleavage occurs after Asp-404 in a DYSD/K sequence and separates the kinase domain from the two PBDs of Plk1. All Plk1 inhibitors triggered G2/M arrest, activation of caspases 2 and 3, polyploidy, multiple nuclei and mitotic catastrophe, albeit at higher concentrations in the case of Rigosertib. Upon BI-2536 treatment, Plk1 cleavage occurred only in the cytosolic fraction and cleaved Plk1 accumulated in this subcellular compartment. Importantly, the cleaved N-Terminal fragment of Plk1 exhibited a higher enzymatic activity than its non-cleaved counterpart and accumulated into the cytoplasm conversely to the full length and the C-Terminal Plk1 fragments that were found essentially into the nucleus. Finally, the DYSD/K cleavage site was highly conserved during evolution from c. elegans to human. In conclusion, we described herein for the first time a specific cleavage of Plk1 by caspase 3 following treatment of cancer cells with ATP-competitive inhibitors of Plk1., Competing Interests: CONFLICTS OF INTEREST The authors declare no conflicts of interest.
- Published
- 2017
- Full Text
- View/download PDF
42. miRNA displacement as a promising approach for cancer therapy.
- Author
-
Gilot D and Galibert MD
- Abstract
microRNA (miRNA) are critical post-transcriptional regulators and key players in diseases development. We demonstrated that non-canonical microRNA Responsive Elements (here MRE-16) could sequester miR-16, dampening miR-16 tumor suppressor function. We developed small oligonucleotides, masking specifically these unusual miR-16 binding sites, that restored miR-16 function. This constitutes a promising targeted approach.
- Published
- 2017
- Full Text
- View/download PDF
43. SnapShot: Cellular Senescence in Pathophysiology.
- Author
-
Martínez-Zamudio RI, Robinson L, Roux PF, and Bischof O
- Subjects
- Aging pathology, Animals, Body Patterning, Cell Plasticity, Humans, Neoplasms pathology, Cellular Senescence
- Abstract
Cellular senescence is a fundamental cell fate, important both in physiological and pathophysiological processes. This SnapShot focuses on the role of cellular senescence in health, disease, and aging., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
44. SnapShot: Cellular Senescence Pathways.
- Author
-
Martínez-Zamudio RI, Robinson L, Roux PF, and Bischof O
- Subjects
- Animals, Cell Cycle, Humans, Inflammation immunology, Neoplasms drug therapy, Cellular Senescence
- Abstract
Cellular senescence is a fundamental cell fate, playing important physiological and pathophysiological roles. This SnapShot focuses on major signaling pathways and transcriptional control mechanisms that consolidate the senescence phenotype., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
45. Role of the PDZ-scaffold protein NHERF1/EBP50 in cancer biology: from signaling regulation to clinical relevance.
- Author
-
Vaquero J, Nguyen Ho-Bouldoires TH, Clapéron A, and Fouassier L
- Subjects
- Humans, Neoplasms, Signal Transduction, PDZ Domains genetics, Phosphoproteins genetics, Phosphoproteins metabolism, Sodium-Hydrogen Exchangers genetics, Sodium-Hydrogen Exchangers metabolism
- Abstract
The transmission of cellular information requires fine and subtle regulation of proteins that need to interact in a coordinated and specific way to form efficient signaling networks. The spatial and temporal coordination relies on scaffold proteins. Thanks to protein interaction domains such as PDZ domains, scaffold proteins organize multiprotein complexes enabling the proper transmission of cellular information through intracellular networks. NHERF1/EBP50 is a PDZ-scaffold protein that was initially identified as an organizer and regulator of transporters and channels at the apical side of epithelia through actin-binding ezrin-moesin-radixin proteins. Since, NHERF1/EBP50 has emerged as a major regulator of cancer signaling network by assembling cancer-related proteins. The PDZ-scaffold EBP50 carries either anti-tumor or pro-tumor functions, two antinomic functions dictated by EBP50 expression or subcellular localization. The dual function of NHERF1/EBP50 encompasses the regulation of several major signaling pathways engaged in cancer, including the receptor tyrosine kinases PDGFR and EGFR, PI3K/PTEN/AKT and Wnt-β-catenin pathways.
- Published
- 2017
- Full Text
- View/download PDF
46. Biomaterial-enabled delivery of SDF-1α at the ventral side of breast cancer cells reveals a crosstalk between cell receptors to promote the invasive phenotype.
- Author
-
Liu XQ, Fourel L, Dalonneau F, Sadir R, Leal S, Lortat-Jacob H, Weidenhaupt M, Albiges-Rizo C, and Picart C
- Subjects
- Animals, Cell Adhesion drug effects, Cell Line, Tumor, Cell Movement drug effects, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Hyaluronan Receptors metabolism, Integrin beta1 metabolism, Mice, Neoplasm Invasiveness, Phenotype, Phosphorylation drug effects, RNA, Small Interfering metabolism, Receptors, CXCR4 metabolism, rac1 GTP-Binding Protein metabolism, Biocompatible Materials chemistry, Breast Neoplasms pathology, Chemokine CXCL12 pharmacology, Drug Delivery Systems, Receptor Cross-Talk
- Abstract
The SDF-1α chemokine (CXCL12) is a potent bioactive chemoattractant known to be involved in hematopoietic stem cell homing and cancer progression. The associated SDF-1α/CXCR4 receptor signaling is a hallmark of aggressive tumors, which can metastasize to distant sites such as lymph nodes, lung and bone. Here, we engineered a biomimetic tumoral niche made of a thin and soft polyelectrolyte film that can retain SDF-1α to present it, in a spatially-controlled manner, at the ventral side of the breast cancer cells. Matrix-bound SDF-1α but not soluble SDF-1α induced a striking increase in cell spreading and migration in a serum-containing medium, which was associated with the formation of lamellipodia and filopodia in MDA-MB231 cells and specifically mediated by CXCR4. Other Knockdown and inhibition experiments revealed that CD44, the major hyaluronan receptor, acted in concert, via a spatial coincidence, to drive a specific matrix-bound SDFα-induced cell response associated with ERK signaling. In contrast, the β1 integrin adhesion receptor played only a minor role on cell polarity. The CXCR4/CD44 mediated cellular response to matrix-bound SDF-1α involved the Rac1 RhoGTPase and was sustained solely in the presence of matrix-bound SDFα, in contrast with the transient signaling observed in response to soluble SDF-1α. Our results highlight that a biomimetic tumoral niche enables to reveal potent cellular effects and so far hidden molecular mechanisms underlying the breast cancer response to chemokines. These results open new insights for the design of future innovative therapies in metastatic cancers, by inhibiting CXCR4-mediated signaling in the tumoral niche via dual targeting of receptors (CXCR4 and CD44) or of associated signaling molecules (CXCR4 and Rac1)., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
47. Epithelial-mesenchymal transition in cholangiocarcinoma: From clinical evidence to regulatory networks.
- Author
-
Vaquero J, Guedj N, Clapéron A, Nguyen Ho-Bouldoires TH, Paradis V, and Fouassier L
- Subjects
- Humans, Prognosis, Bile Duct Neoplasms diagnosis, Bile Duct Neoplasms pathology, Cholangiocarcinoma diagnosis, Cholangiocarcinoma pathology, Epithelial-Mesenchymal Transition physiology
- Abstract
Cholangiocarcinoma (CCA) is an aggressive tumor with a poor prognosis due to its late clinical presentation and the lack of effective non-surgical therapies. Unfortunately, most of the patients are not eligible for curative surgery owing to the presence of metastases at the time of diagnosis. Therefore, it is important to understand the steps leading to cell dissemination in patients with CCA. To metastasize from the primary site, cancer cells must acquire migratory and invasive properties by a cell plasticity-promoting phenomenon known as epithelial-mesenchymal transition (EMT). EMT is a reversible dynamic process by which epithelial cells gradually adopt structural and functional characteristics of mesenchymal cells, and has lately become a centre of attention in the field of metastatic dissemination. In the present review, we aim to provide an extensive overview of the current clinical data and the prognostic value of different EMT markers that have been analysed in CCA. We summarize all the regulatory networks implicated in EMT from the membrane receptors to the main EMT-inducing transcription factors (SNAIL, TWIST and ZEB). Furthermore, since a tumor is a complex structure not exclusively formed by tumor cells, we also address the prominent role of the main cell types of the desmoplastic stroma that characterizes CCA in the regulation of EMT. Finally, we discuss the therapeutic considerations and difficulties faced to develop an effective anti-EMT treatment due to the redundancies and bypasses among the pathways regulating EMT., (Copyright © 2016 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
48. Differentiation inducing factor 3 mediates its anti-leukemic effect through ROS-dependent DRP1-mediated mitochondrial fission and induction of caspase-independent cell death.
- Author
-
Dubois A, Ginet C, Furstoss N, Belaid A, Hamouda MA, El Manaa W, Cluzeau T, Marchetti S, Ricci JE, Jacquel A, Luciano F, Driowya M, Benhida R, Auberger P, and Robert G
- Subjects
- Animals, Biomarkers, Tumor metabolism, Caspases metabolism, Cell Differentiation drug effects, Cell Proliferation drug effects, Dynamins, Female, Humans, K562 Cells, Leukemia drug therapy, Leukemia metabolism, Mice, Mice, Nude, Mitochondria drug effects, Mitochondria metabolism, Mitochondria pathology, Phosphorylation drug effects, Xenograft Model Antitumor Assays, Apoptosis drug effects, GTP Phosphohydrolases metabolism, Gene Expression Regulation, Leukemic drug effects, Hexanones pharmacology, Leukemia pathology, Microtubule-Associated Proteins metabolism, Mitochondrial Dynamics drug effects, Mitochondrial Proteins metabolism, Reactive Oxygen Species metabolism
- Abstract
Differentiation-inducing factor (DIF) defines a group of chlorinated hexaphenones that orchestrate stalk-cell differentiation in the slime mold Dictyostelium discoideum (DD). DIF-1 and 3 have also been reported to have tumor inhibiting properties; however, the mechanisms that underlie the effects of these compounds remain poorly defined. Herein, we show that DIF-3 rapidly triggers Ca2+ release and a loss of mitochondrial membrane potential (MMP) in the absence of cytochrome c and Smac release and without caspase activation. Consistently with these findings, we also detected no evidence of apoptosis in cells treated with DIF-3 but instead found that this compound induced autophagy. In addition, DIF-3 promoted mitochondrial fission in K562 and HeLa cells, as assessed by electron and confocal microscopy analysis. Importantly, DIF-3 mediated the phosphorylation and redistribution of dynamin-related protein 1 (DRP1) from the cytoplasmic to the microsomal fraction of K562 cells. Pharmacological inhibition or siRNA silencing of DRP1 not only inhibited mitochondrial fission but also protected K562 cells from DIF-3-mediated cell death. Furthermore, DIF-3 potently inhibited the growth of imatinib-sensitive and imatinib-resistant K562 cells. It also inhibited tumor formation in athymic mice engrafted with an imatinib-resistant CML cell line. Finally, DIF-3 exhibited a clear selectivity toward CD34+ leukemic cells from CML patients, compared with CD34- cells. In conclusion, we show that the potent anti-leukemic effect of DIF-3 is mediated through the induction of mitochondrial fission and caspase-independent cell death. Our findings may have important therapeutic implications, especially in the treatment of tumors that exhibit defects in apoptosis regulation., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2016
- Full Text
- View/download PDF
49. Expert consensus document: Cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA).
- Author
-
Banales JM, Cardinale V, Carpino G, Marzioni M, Andersen JB, Invernizzi P, Lind GE, Folseraas T, Forbes SJ, Fouassier L, Geier A, Calvisi DF, Mertens JC, Trauner M, Benedetti A, Maroni L, Vaquero J, Macias RI, Raggi C, Perugorria MJ, Gaudio E, Boberg KM, Marin JJ, and Alvaro D
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bile Duct Neoplasms genetics, Bile Duct Neoplasms therapy, Cancer-Associated Fibroblasts physiology, Cell Proliferation physiology, Cell Transformation, Neoplastic genetics, Cholangiocarcinoma genetics, Cholangiocarcinoma therapy, Cytokines metabolism, Drug Resistance, Neoplasm genetics, Early Detection of Cancer, Epigenesis, Genetic genetics, Forecasting, Gene Fusion genetics, Genetic Heterogeneity, Humans, Intercellular Signaling Peptides and Proteins metabolism, Liver Transplantation, Macrophages physiology, Molecular Targeted Therapy methods, Neoplasm Proteins physiology, Neoplasm Staging, Neoplastic Stem Cells pathology, Receptor, Fibroblast Growth Factor, Type 2 genetics, Risk Factors, Signal Transduction genetics, Stents, Tumor Microenvironment genetics, Bile Duct Neoplasms pathology, Cholangiocarcinoma pathology
- Abstract
Cholangiocarcinoma (CCA) is a heterogeneous group of malignancies with features of biliary tract differentiation. CCA is the second most common primary liver tumour and the incidence is increasing worldwide. CCA has high mortality owing to its aggressiveness, late diagnosis and refractory nature. In May 2015, the "European Network for the Study of Cholangiocarcinoma" (ENS-CCA: www.enscca.org or www.cholangiocarcinoma.eu) was created to promote and boost international research collaboration on the study of CCA at basic, translational and clinical level. In this Consensus Statement, we aim to provide valuable information on classifications, pathological features, risk factors, cells of origin, genetic and epigenetic modifications and current therapies available for this cancer. Moreover, future directions on basic and clinical investigations and plans for the ENS-CCA are highlighted.
- Published
- 2016
- Full Text
- View/download PDF
50. Molecular Basis of the Rapamycin Insensitivity of Target Of Rapamycin Complex 2.
- Author
-
Gaubitz C, Oliveira TM, Prouteau M, Leitner A, Karuppasamy M, Konstantinidou G, Rispal D, Eltschinger S, Robinson GC, Thore S, Aebersold R, Schaffitzel C, and Loewith R
- Subjects
- Antifungal Agents metabolism, Antifungal Agents pharmacology, Binding Sites genetics, Biocatalysis drug effects, Blotting, Western, Carrier Proteins chemistry, Carrier Proteins genetics, Carrier Proteins metabolism, Cell Cycle drug effects, Cell Cycle genetics, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Drug Resistance genetics, Mass Spectrometry methods, Mechanistic Target of Rapamycin Complex 2, Microscopy, Electron, Multiprotein Complexes genetics, Multiprotein Complexes metabolism, Mutation, Phosphatidylinositol 3-Kinases chemistry, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Protein Structure, Tertiary, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Saccharomyces cerevisiae Proteins ultrastructure, Sirolimus metabolism, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Multiprotein Complexes chemistry, Saccharomyces cerevisiae Proteins chemistry, Sirolimus pharmacology, TOR Serine-Threonine Kinases chemistry
- Abstract
Target of Rapamycin (TOR) plays central roles in the regulation of eukaryote growth as the hub of two essential multiprotein complexes: TORC1, which is rapamycin-sensitive, and the lesser characterized TORC2, which is not. TORC2 is a key regulator of lipid biosynthesis and Akt-mediated survival signaling. In spite of its importance, its structure and the molecular basis of its rapamycin insensitivity are unknown. Using crosslinking-mass spectrometry and electron microscopy, we determined the architecture of TORC2. TORC2 displays a rhomboid shape with pseudo-2-fold symmetry and a prominent central cavity. Our data indicate that the C-terminal part of Avo3, a subunit unique to TORC2, is close to the FKBP12-rapamycin-binding domain of Tor2. Removal of this sequence generated a FKBP12-rapamycin-sensitive TORC2 variant, which provides a powerful tool for deciphering TORC2 function in vivo. Using this variant, we demonstrate a role for TORC2 in G2/M cell-cycle progression., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- 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.