Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Cell Death Dis ; 15(5): 311, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38697987

ABSTRACT

Cancer cells are highly dependent on bioenergetic processes to support their growth and survival. Disruption of metabolic pathways, particularly by targeting the mitochondrial electron transport chain complexes (ETC-I to V) has become an attractive therapeutic strategy. As a result, the search for clinically effective new respiratory chain inhibitors with minimized adverse effects is a major goal. Here, we characterize a new OXPHOS inhibitor compound called MS-L6, which behaves as an inhibitor of ETC-I, combining inhibition of NADH oxidation and uncoupling effect. MS-L6 is effective on both intact and sub-mitochondrial particles, indicating that its efficacy does not depend on its accumulation within the mitochondria. MS-L6 reduces ATP synthesis and induces a metabolic shift with increased glucose consumption and lactate production in cancer cell lines. MS-L6 either dose-dependently inhibits cell proliferation or induces cell death in a variety of cancer cell lines, including B-cell and T-cell lymphomas as well as pediatric sarcoma. Ectopic expression of Saccharomyces cerevisiae NADH dehydrogenase (NDI-1) partially restores the viability of B-lymphoma cells treated with MS-L6, demonstrating that the inhibition of NADH oxidation is functionally linked to its cytotoxic effect. Furthermore, MS-L6 administration induces robust inhibition of lymphoma tumor growth in two murine xenograft models without toxicity. Thus, our data present MS-L6 as an inhibitor of OXPHOS, with a dual mechanism of action on the respiratory chain and with potent antitumor properties in preclinical models, positioning it as the pioneering member of a promising drug class to be evaluated for cancer therapy. MS-L6 exerts dual mitochondrial effects: ETC-I inhibition and uncoupling of OXPHOS. In cancer cells, MS-L6 inhibited ETC-I at least 5 times more than in isolated rat hepatocytes. These mitochondrial effects lead to energy collapse in cancer cells, resulting in proliferation arrest and cell death. In contrast, hepatocytes which completely and rapidly inactivated this molecule, restored their energy status and survived exposure to MS-L6 without apparent toxicity.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Electron Transport Complex I , Mitochondria , Saccharomyces cerevisiae Proteins , Animals , Humans , Electron Transport Complex I/metabolism , Electron Transport Complex I/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Mice , Cell Line, Tumor , Mitochondria/metabolism , Mitochondria/drug effects , Cell Proliferation/drug effects , Uncoupling Agents/pharmacology , Oxidative Phosphorylation/drug effects , Xenograft Model Antitumor Assays , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/drug effects , Rats , NADH Dehydrogenase/metabolism , NADH Dehydrogenase/antagonists & inhibitors
2.
Cell Death Discov ; 9(1): 230, 2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37414800

ABSTRACT

Toll-like receptor 3 (TLR3) is a pattern recognition receptor mainly known for its role in innate immune response to infection. Indeed, binding of double-stranded RNA (dsRNA) to TLR3 triggers a pro-inflammatory cascade leading to cytokine release and immune cell activation. Its anti-tumoral potential has emerged progressively, associated with a direct impact on tumor cell death induction and with an indirect action on immune system reactivation. Accordingly, TLR3 agonists are currently being tested in clinical trials for several adult cancers. Meanwhile, TLR3 variants have been linked to auto-immune disorders, and as risk factors of viral infection and cancers. However, aside from neuroblastoma, TLR3 role in childhood cancers has not been evaluated. Here, by integrating public transcriptomic data of pediatric tumors, we unveil that high TLR3 expression is largely associated with a better prognosis in childhood sarcomas. Using osteosarcomas and rhabdomyosarcomas as models, we show that TLR3 efficiently drives tumor cell death in vitro and induces tumor regression in vivo. Interestingly, this anti-tumoral effect was lost in cells expressing the homozygous TLR3 L412F polymorphism, which is enriched in a rhabdomyosarcomas cohort. Thus, our results demonstrate the therapeutic potential associated with the targeting of TLR3 in pediatric sarcomas, but also the need to stratify patients eligible for this clinical approach with respect to the TLR3 variants expressed.

3.
Med Sci (Paris) ; 39(5): 452-457, 2023 May.
Article in French | MEDLINE | ID: mdl-37219350

ABSTRACT

In France, part of 40 % of preventable cancers can be attributed to lifestyle habits. Epidemiological data show that occupational exposures are a major cause of these cancers. However, despite this evidence, the prevention actions promoted by public authorities are focused on changing individual behaviors. In this article, we seek to understand the reasons of the erasure of the role of socio-environmental factors in cancer prevention discourse.


Title: Cancers évitables - Suffit-il de changer nos comportements ? Abstract: En France, une partie des 40 % de cancers évitables peut être attribuée aux habitudes de vie. Les données épidémiologiques révèlent que les expositions professionnelles et domestiques à des substances cancérogènes sont aussi responsables d'une fraction significative de ces cancers. Pourtant, en dépit de ces évidences, les actions de prévention promues par les pouvoirs publics se focalisent sur le changement des comportements individuels. Dans cet article, nous cherchons à comprendre les raisons de l'effacement de la place des facteurs socio-environnementaux des discours portant sur la prévention des cancers.


Subject(s)
Neoplasms , Occupational Exposure , Humans , Neoplasms/etiology , Life Style , France , Risk Factors
4.
Dev Dyn ; 252(8): 1077-1095, 2023 08.
Article in English | MEDLINE | ID: mdl-36880501

ABSTRACT

Neural crest cells (NCCs) are highly motile, multipotent, embryonic cells that delaminate from the dorsal edges of the neural tube. NCCs follow stereotypical long-range migratory pathways to reach target organs during development, where they give rise to multiple derivatives. The identification of reservoirs of neural crest stem cells that persist to adulthood has recently aroused renewed interest in the biology of NCCs. In this context, several recent studies have demonstrated the essential role of the metabolic kinase LKB1 in NCC establishment. This review surveys how LKB1 governs the formation and maintenance of several neural crest derivatives, including facial bones, melanocytes, Schwann cells, and the enteric nervous system. We also detail the underlying molecular mechanisms that involve downstream effectors of LKB1, in particular the contribution of the AMPK-mTOR signaling pathway to both polarity and metabolic processes. Collectively, these recent discoveries open promising perspectives for new therapeutic applications for the treatment of neural crest disorders.


Subject(s)
Neural Crest , Neural Stem Cells , Neural Crest/metabolism , Signal Transduction , Neural Tube , Schwann Cells , Cell Movement/physiology , Cell Differentiation
5.
Blood ; 141(10): 1209-1220, 2023 03 09.
Article in English | MEDLINE | ID: mdl-36375119

ABSTRACT

Understanding the functional role of mutated genes in cancer is required to translate the findings of cancer genomics into therapeutic improvement. BTG1 is recurrently mutated in the MCD/C5 subtype of diffuse large B-cell lymphoma (DLBCL), which is associated with extranodal dissemination. Here, we provide evidence that Btg1 knock out accelerates the development of a lethal lymphoproliferative disease driven by Bcl2 overexpression. Furthermore, we show that the scaffolding protein BCAR1 is a BTG1 partner. Moreover, after BTG1 deletion or expression of BTG1 mutations observed in patients with DLBCL, the overactivation of the BCAR1-RAC1 pathway confers increased migration ability in vitro and in vivo. These modifications are targetable with the SRC inhibitor dasatinib, which opens novel therapeutic opportunities in BTG1 mutated DLBCL.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Mutation , Genes, cdc , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Crk-Associated Substrate Protein/genetics , Crk-Associated Substrate Protein/metabolism
6.
EMBO Rep ; 21(4): e48938, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32052574

ABSTRACT

Intestinal stem cells (ISCs) fuel the lifelong self-renewal of the intestinal tract and are paramount for epithelial repair. In this context, the Wnt pathway component LGR5 is the most consensual ISC marker to date. Still, the effort to better understand ISC identity and regulation remains a challenge. We have generated a Mex3a knockout mouse model and show that this RNA-binding protein is crucial for the maintenance of the Lgr5+ ISC pool, as its absence disrupts epithelial turnover during postnatal development and stereotypical organoid maturation ex vivo. Transcriptomic profiling of intestinal crypts reveals that Mex3a deletion induces the peroxisome proliferator-activated receptor (PPAR) pathway, along with a decrease in Wnt signalling and loss of the Lgr5+ stem cell signature. Furthermore, we identify PPARγ activity as a molecular intermediate of MEX3A-mediated regulation. We also show that high PPARγ signalling impairs Lgr5+ ISC function, thus uncovering a new layer of post-transcriptional regulation that critically contributes to intestinal homeostasis.


Subject(s)
Intestinal Mucosa , Stem Cells , Animals , Intestines , Mice , Organoids , Receptors, G-Protein-Coupled/genetics , Wnt Signaling Pathway
7.
Sci Adv ; 5(7): eaau5106, 2019 07.
Article in English | MEDLINE | ID: mdl-31328154

ABSTRACT

Metabolic processes underlying the development of the neural crest, an embryonic population of multipotent migratory cells, are poorly understood. Here, we report that conditional ablation of the Lkb1 tumor suppressor kinase in mouse neural crest stem cells led to intestinal pseudo-obstruction and hind limb paralysis. This phenotype originated from a postnatal degeneration of the enteric nervous ganglia and from a defective differentiation of Schwann cells. Metabolomic profiling revealed that pyruvate-alanine conversion is enhanced in the absence of Lkb1. Mechanistically, inhibition of alanine transaminases restored glial differentiation in an mTOR-dependent manner, while increased alanine level directly inhibited the glial commitment of neural crest cells. Treatment with the metabolic modulator AICAR suppressed mTOR signaling and prevented Schwann cell and enteric defects of Lkb1 mutant mice. These data uncover a link between pyruvate-alanine cycling and the specification of glial cell fate with potential implications in the understanding of the molecular pathogenesis of neural crest diseases.


Subject(s)
Alanine/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Protein Serine-Threonine Kinases/genetics , Pyruvic Acid/metabolism , AMP-Activated Protein Kinases , Animals , Cell Differentiation/genetics , Energy Metabolism , Enteric Nervous System , Gene Silencing , Melanocytes/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neuroglia/cytology , Neuroglia/metabolism , Peripheral Nervous System Diseases/etiology , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Phenotype , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
8.
Med Sci (Paris) ; 34(8-9): 701-708, 2018.
Article in French | MEDLINE | ID: mdl-30230466

ABSTRACT

During tumor development, malignant cells rewire their metabolism to meet the biosynthetic needs required to increase their biomass and to overcome their microenvironment constraints. The sustained activation of aerobic glycolysis, also called Warburg effect, is one of these adaptative mechanisms. The progresses in this area of research have revealed the flexibility of cancer cells that alternate between glycolytic and oxidative metabolism to cope with their conditions of development while sharing their energetic resources. In this survey, we review these recent breakthroughs and discuss a model that likens tumor to an evolutive metabolic ecosystem. We further emphasize the ensuing therapeutic applications that target metabolic weaknesses of neoplastic cells.


Subject(s)
Energy Metabolism/physiology , Glycolysis/physiology , Neoplasms/metabolism , Neoplasms/pathology , Tumor Microenvironment/physiology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , Mitochondria/metabolism , Oxidation-Reduction
9.
Trends Cancer ; 3(7): 506-528, 2017 07.
Article in English | MEDLINE | ID: mdl-28718405

ABSTRACT

RNA-binding proteins (RBPs) are key players in post-transcriptional events. The combination of versatility of their RNA-binding domains with structural flexibility enables RBPs to control the metabolism of a large array of transcripts. Perturbations in RBP-RNA networks activity have been causally associated with cancer development, but the rational framework describing these contributions remains fragmented. We review here the evidence that RBPs modulate multiple cancer traits, emphasize their functional diversity, and assess future trends in the study of RBPs in cancer.


Subject(s)
Neoplasms/pathology , RNA Processing, Post-Transcriptional/drug effects , RNA-Binding Motifs , RNA-Binding Proteins/metabolism , RNA/metabolism , Alternative Splicing , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/drug effects , Carcinogenesis/pathology , Disease Progression , Gene Regulatory Networks , Humans , Molecular Targeted Therapy/methods , Neoplasm Invasiveness/pathology , Neoplasms/drug therapy , Neoplasms/genetics , RNA-Binding Proteins/genetics
10.
Article in English | MEDLINE | ID: mdl-28674522

ABSTRACT

Circadian clocks are innate oscillators that drive daily rhythms in metabolism, physiology, and behavior. 24-h rhythms in gene expression, driven by core clock transcription factors, reflect the epigenetic state of the cell, which in turn is dictated by the metabolic environment. Cancer cells alter their metabolic state and gene expression and therefore are likely to tweak circadian clock function in their favor. Over the past decade, we have witnessed an extraordinary increase in systems-level studies that suggest intricate mechanistic links between the cellular metabolome and the circadian epigenome. In parallel, reprogramming of cellular clock function in cancers is increasingly evident and the role of clock genes in the development of hematological tumors, as well as their pathophysiological effects on tissues distal to the tumor, has been described. Furthermore, the interplay between components of the circadian clock, metabolic enzymes, and oncogenes is starting to be better understood, such as the close association between overexpression of the Myc oncogene and perturbation of circadian and metabolic rhythms, thus opening new avenues to treat cancers. This review article explores current knowledge on the circadian metabolome and the molecular pathways they control, with a focus on their involvement in the development of hematopoietic malignancies.

11.
Dev Biol ; 418(2): 283-96, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27527806

ABSTRACT

Head development in vertebrates proceeds through a series of elaborate patterning mechanisms and cell-cell interactions involving cephalic neural crest cells (CNCC). These cells undergo extensive migration along stereotypical paths after their separation from the dorsal margins of the neural tube and they give rise to most of the craniofacial skeleton. Here, we report that the silencing of the LKB1 tumor suppressor affects the delamination of pre-migratory CNCC from the neural primordium as well as their polarization and survival, thus resulting in severe facial and brain defects. We further show that LKB1-mediated effects on the development of CNCC involve the sequential activation of the AMP-activated protein kinase (AMPK), the Rho-dependent kinase (ROCK) and the actin-based motor protein myosin II. Collectively, these results establish that the complex morphogenetic processes governing head formation critically depends on the activation of the LKB1 signaling network in CNCC.


Subject(s)
Avian Proteins/physiology , Neural Crest/physiology , Protein Serine-Threonine Kinases/physiology , AMP-Activated Protein Kinases/physiology , Animals , Avian Proteins/antagonists & inhibitors , Avian Proteins/genetics , Chick Embryo , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/genetics , Gene Expression Regulation, Developmental , Gene Silencing , Head/embryology , Mice , Mice, Knockout , Myosin Light Chains/physiology , Neural Crest/cytology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Signal Transduction/physiology , rho-Associated Kinases/physiology
12.
Cancer Res ; 76(12): 3541-52, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27216191

ABSTRACT

LIM kinases (LIMK) are emerging targets for cancer therapy, and they function as network hubs to coordinate actin and microtubule dynamics. When LIMKs are inhibited, actin microfilaments are disorganized and microtubules are stabilized. Owing to their stabilizing effect on microtubules, LIMK inhibitors may provide a therapeutic strategy to treat taxane-resistant cancers. In this study, we investigated the effect of LIMK inhibition on breast tumor development and on paclitaxel-resistant tumors, using a novel selective LIMK inhibitor termed Pyr1. Treatment of breast cancer cells, including paclitaxel-resistant cells, blocked their invasion and proliferation in vitro and their growth in vivo in tumor xenograft assays. The tumor-invasive properties of Pyr1 were investigated in vivo by intravital microscopy of tumor xenografts. A striking change of cell morphology was observed with a rounded phenotype arising in a subpopulation of cells, while other cells remained elongated. Notably, although Pyr1 decreased the motility of elongated cells, it increased the motility of rounded cells in the tumor. Pyr1 administration prevented the growth of metastasis but not their spread. Overall, our results provided a preclinical proof of concept concerning how a small-molecule inhibitor of LIMK may offer a strategy to treat taxane-resistant breast tumors and metastases. Cancer Res; 76(12); 3541-52. ©2016 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Carbazoles/pharmacology , Lim Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis
14.
Med Sci (Paris) ; 31(8-9): 797-803, 2015.
Article in French | MEDLINE | ID: mdl-26340841

ABSTRACT

The idea of personalized medicine raises a series of questions. If one considers that the physician takes into account the uniqueness of his patient in the frame of the medical consultation, is the definition of medicine as "personalized" not a pleonasm? If not, why has this ambiguous denomination been adopted? In addition, is this form of medicine a novel discipline capable of revolutionizing therapeutic approaches as claimed in its accompanying discourses or is it in continuity with the molecular conception of biomedicine? Rather than attempting to directly answer these questions, we focused our attention on the organizing concepts, the technological breakthroughs and the transformations in medical practices that characterize this medicine. Following this brief analysis, it appears that the choice of a term as equivocal as personalized medicine and the emphasis on the antagonistic notions of revolution and continuity in medicine are the signs of reshuffling that is emerging between actors in the health care system, in academia and in pharmaceutical companies.


Subject(s)
Precision Medicine , Terminology as Topic , Utopias
15.
PLoS One ; 10(6): e0128364, 2015.
Article in English | MEDLINE | ID: mdl-26046350

ABSTRACT

Oncogenic mutation of the RET receptor tyrosine kinase is observed in several human malignancies. Here, we describe three novel type II RET tyrosine kinase inhibitors (TKI), ALW-II-41-27, XMD15-44 and HG-6-63-01, that inhibit the cellular activity of oncogenic RET mutants at two digit nanomolar concentration. These three compounds shared a 3-trifluoromethyl-4-methylpiperazinephenyl pharmacophore that stabilizes the 'DFG-out' inactive conformation of RET activation loop. They blocked RET-mediated signaling and proliferation with an IC50 in the nM range in fibroblasts transformed by the RET/C634R and RET/M918T oncogenes. They also inhibited autophosphorylation of several additional oncogenic RET-derived point mutants and chimeric oncogenes. At a concentration of 10 nM, ALW-II-41-27, XMD15-44 and HG-6-63-01 inhibited RET kinase and signaling in human thyroid cancer cell lines carrying oncogenic RET alleles; they also inhibited proliferation of cancer, but not non-tumoral Nthy-ori-3-1, thyroid cells, with an IC50 in the nM range. The three compounds were capable of inhibiting the 'gatekeeper' V804M mutant which confers substantial resistance to established RET inhibitors. In conclusion, we have identified a type II TKI scaffold, shared by ALW-II-41-27, XMD15-44 and HG-6-63-01, that may be used as novel lead for the development of novel agents for the treatment of cancers harboring oncogenic activation of RET.


Subject(s)
Benzamides/chemistry , Niacinamide/analogs & derivatives , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Pyridines/chemistry , Small Molecule Libraries/chemistry , Animals , Benzamides/metabolism , Benzamides/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Molecular Docking Simulation , Mutation , NIH 3T3 Cells , Niacinamide/chemistry , Niacinamide/metabolism , Niacinamide/toxicity , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/toxicity , Protein Structure, Tertiary , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Pyridines/metabolism , Pyridines/toxicity , Small Molecule Libraries/metabolism , Small Molecule Libraries/toxicity , Transfection
16.
Int J Dev Biol ; 58(5): 379-84, 2014.
Article in English | MEDLINE | ID: mdl-25354459

ABSTRACT

Several human diseases are associated with the NUAK1 and NUAK2 genes. These genes encode kinases, members of the AMPK-related kinases (ARK) gene family. Both NUAK1 and NUAK2 are known targets of the serine threonine kinase LKB1, a tumor suppressor involved in regulating cell polarity. While much is known about their functions in disease, their expression pattern in normal development has not been extensively studied. Here, we present the expression patterns for NUAK1 and NUAK2 in the chick during early-stage embryogenesis, until day 3 (Hamburger and Hamilton stage HH20). Several embryonic structures, in particular the nascent head, showed distinct expression levels. NUAK1 expression was first detected at stage HH6 in the rostral neural folds. It was then expressed (HH7-11) throughout the encephalalon, predominantly in the telencephalon and mesencephalon. NUAK1 expression was also detected in the splanchnic endoderm area at HH8-10, and in the vitellin vein derived from this area, but not in the heart. NUAK2 expression was first detected at stage HH6 in the neural folds. It was then found throughout the encephalon at stage HH20. Particular attention was paid in this study to the dorsal ectoderm at stages HH7 and HH8, where a local deficit or accumulation of NUAK2 mRNA were found to correlate with the direction of curvature of the neural plate. This is the first description of NUAK1 and NUAK2 expression patterns in the chick during early development; it reveals non-identical expression profiles for both genes in neural development.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Developmental , Head/embryology , Protein Kinases/genetics , Repressor Proteins/genetics , Animals , Brain/embryology , Chick Embryo , Ectoderm/metabolism , Embryonic Development/genetics , Neural Crest/embryology , Neural Crest/metabolism , Phosphorylation , Protein Kinases/metabolism , Repressor Proteins/metabolism
17.
Development ; 141(10): 2096-107, 2014 May.
Article in English | MEDLINE | ID: mdl-24803656

ABSTRACT

The four related mammalian MEX-3 RNA-binding proteins are evolutionarily conserved molecules for which the in vivo functions have not yet been fully characterized. Here, we report that male mice deficient for the gene encoding Mex3b are subfertile. Seminiferous tubules of Mex3b-deficient mice are obstructed as a consequence of the disrupted phagocytic capacity of somatic Sertoli cells. In addition, both the formation and the integrity of the blood-testis barrier are compromised owing to mislocalization of N-cadherin and connexin 43 at the surface of Sertoli cells. We further establish that Mex3b acts to regulate the cortical level of activated Rap1, a small G protein controlling phagocytosis and cell-cell interaction, through the activation and transport of Rap1GAP. The active form of Rap1 (Rap1-GTP) is abnormally increased at the membrane cortex and chemically restoring Rap1-GTP to physiological levels rescues the phagocytic and adhesion abilities of Sertoli cells. Overall, these findings implicate Mex3b in the spatial organization of the Rap1 pathway that orchestrates Sertoli cell functions.


Subject(s)
RNA-Binding Proteins/physiology , Sertoli Cells/physiology , rap1 GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Embryo, Mammalian , Female , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mice , Mice, Knockout , RNA-Binding Proteins/genetics , Seminiferous Epithelium/metabolism , Sertoli Cells/metabolism , Signal Transduction , Tissue Distribution/genetics , rap1 GTP-Binding Proteins/genetics
18.
Int J Cancer ; 135(6): 1307-18, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24615515

ABSTRACT

Although the presence of nuclear estrogen receptor is widely used to guide breast cancer therapy, less attention has been paid to the receptor cytoplasmic signaling. Recently, we have shown that this pathway is operative in vivo and is activated in aggressive tumors representing a new potential target for breast cancer therapy. Here, we identified LKB1 as a partner of ERα and we explored its potential role in estrogen nongenomic signaling. The associations between LKB1 expression and the actors of this pathway, namely the methylated form of ERα (metERα), Src and PI3K, have been analyzed both in cultured cells and in 154 primary breast tumor samples. We found that LKB1 is a component of the cytoplasmic signaling complex in breast cell lines as well as in primary breast tumors. Moreover, an inverse correlation between the localization of LKB1 in nuclear and cytoplasmic compartments is observed. Importantly, high expression of cytoplasmic LKB1 is an independent marker of poor prognosis, associated with reduced overall survival (OS) and disease free survival (DFS). Conversely, the presence of nuclear LKB1 associates with increased OS and DFS. In conclusion, our results highlight that LKB1 expression in breast cancer appears to have opposite effects depending on its subcellular localization and may be used as a new prognostic biomarker.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Line, Tumor , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Female , Humans , Methylation , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Transfection , src-Family Kinases/metabolism
19.
Cancer Res ; 73(22): 6621-31, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24078802

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is a transdifferentiation process that converts epithelial cells into highly motile mesenchymal cells. This physiologic process occurs largely during embryonic development but is aberrantly reactivated in different pathologic situations, including fibrosis and cancer. We conducted a siRNA screening targeted to the human kinome with the aim of discovering new EMT effectors. With this approach, we have identified mTOR complex 1 (mTORC1), a nutrient sensor that controls protein and lipid synthesis, as a key regulator of epithelial integrity. Using a combination of RNAi and pharmacologic approaches, we report here that inhibition of either mTOR or RPTOR triggers EMT in mammary epithelial cells. This EMT was characterized by the induction of the mesenchymal markers such as fibronectin, vimentin, and PAI-1, together with the repression of epithelial markers such as E-cadherin and ZO-3. In addition, mTORC1 blockade enhanced in vivo migratory properties of mammary cells and induced EMT independent of the TGF-ß pathway. Finally, among the transcription factors known to activate EMT, both ZEB1 and ZEB2 were upregulated following mTOR repression. Their increased expression correlated with a marked reduction in miR-200b and miR-200c mRNA levels, two microRNAs known to downregulate ZEB1 and ZEB2 expression. Taken together, our findings unravel a novel function for mTORC1 in maintaining the epithelial phenotype and further indicate that this effect is mediated through the opposite regulation of ZEB1/ZEB2 and miR-200b and miR-200c. Furthermore, these results suggest a plausible etiologic explanation for the progressive pulmonary fibrosis, a frequent adverse condition associated with the therapeutic use of mTOR inhibitors.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Chick Embryo , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1 , MicroRNAs/metabolism , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Neoplasm Invasiveness , Protein Kinase Inhibitors/pharmacology , RNA Interference , Repressor Proteins/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/physiology , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
20.
Trends Biochem Sci ; 38(10): 477-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23999169

ABSTRACT

RNA-binding proteins of the evolutionarily-conserved MEX-3 family are mediators of post-transcriptional regulation in different organisms. Recent studies highlight their involvement in diverse physiological settings, including the maintenance of a balance between stem cell self-renewal and differentiation. Here, we draw attention to their putative role in tissue homeostasis and disease, particularly cancer.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Protein Biosynthesis , RNA-Binding Proteins/metabolism , RNA/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/immunology , Humans , RNA/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL