Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Dis Model Mech ; 17(3)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38506114

ABSTRACT

Breast cancer stands as the most prevalent malignancy afflicting women. Despite significant advancements in its diagnosis and treatment, breast cancer metastasis continues to be a leading cause of mortality among women. To metastasize, cancer cells face numerous challenges: breaking away from the primary tumor, surviving in the circulation, establishing in a distant location, evading immune detection and, finally, thriving to initiate a new tumor. Each of these sequential steps requires cancer cells to adapt to a myriad of stressors and develop survival mechanisms. In addition, most patients with breast cancer undergo surgical removal of their primary tumor and have various therapeutic interventions designed to eradicate cancer cells. Despite this plethora of attacks and stresses, certain cancer cells not only manage to persist but also proliferate robustly, giving rise to substantial tumors that frequently culminate in the patient's demise. To enhance patient outcomes, there is an imperative need for a deeper understanding of the molecular and cellular mechanisms that empower cancer cells to not only survive but also expand. Herein, we delve into the intrinsic stresses that cancer cells encounter throughout the metastatic journey and the additional stresses induced by therapeutic interventions. We focus on elucidating the remarkable strategies adopted by cancer cells, such as cell-cell clustering and intricate cell-cell communication mechanisms, to ensure their survival.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Cell Communication
2.
Front Immunol ; 14: 1196731, 2023.
Article in English | MEDLINE | ID: mdl-37539056

ABSTRACT

Introduction: Tumor Associated Macrophages (TAM) are a major component of the tumor environment and their accumulation often correlates with poor prognosis by contributing to local inflammation, inhibition of anti-tumor immune response and resistance to anticancer treatments. In this study, we thus investigated the anti-cancer therapeutic interest to target ChemR23, a receptor of the resolution of inflammation expressed by macrophages, using an agonist monoclonal antibody, αChemR23. Methods: Human GM-CSF, M-CSF and Tumor Associated Macrophage (TAM)-like macrophages were obtained by incubation of monocytes from healthy donors with GM-CSF, M-CSF or tumor cell supernatants (Breast cancer (BC) or malignant pleural mesothelioma (MPM) cells). The effects of αChemR23 on macrophages were studied at the transcriptomic, protein and functional level. Datasets from The Cancer Genome Atlas (TCGA) were used to study CMKLR1 expression, coding for ChemR23, in BC and MPM tumors. In vivo, αChemR23 was evaluated on overall survival, metastasis development and transcriptomic modification of the metastatic niche using a model of resected triple negative breast cancer. Results: We show that ChemR23 is expressed at higher levels in M-CSF and tumor cell supernatant differentiated macrophages (TAM-like) than in GM-CSF-differentiated macrophages. ChemR23 activation triggered by αChemR23 deeply modulates M-CSF and TAM-like macrophages including profile of cell surface markers, cytokine secretion, gene mRNA expression and immune functions. The expression of ChemR23 coding gene (CMKLR1) strongly correlates to TAM markers in human BC tumors and MPM and its histological detection in these tumors mainly corresponds to TAM expression. In vivo, treatment with αChemR23 agonist increased mouse survival and decreased metastasis occurrence in a model of triple-negative BC in correlation with modulation of TAM phenotype in the metastatic niche. Conclusion: These results open an attractive opportunity to target TAM and the resolution of inflammation pathways through ChemR23 to circumvent TAM pro-tumoral effects.


Subject(s)
Breast Neoplasms , Carcinoma , Receptors, Chemokine , Animals , Female , Humans , Mice , Carcinoma/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammation/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages , Phenotype , Receptors, Chemokine/metabolism
3.
Front Immunol ; 12: 702785, 2021.
Article in English | MEDLINE | ID: mdl-34276698

ABSTRACT

Inflammation is a fundamental physiological response orchestrated by innate immune cells to restore tissue homeostasis. Specialized pro-resolving mediators (SPMs) are involved in active resolution of inflammation but when inflammation is incomplete, chronic inflammation creates a favorable environment that fuels carcinogenesis and cancer progression. Conventional cancer therapy also strengthens cancer-related inflammation by inducing massive tumor cell death that activate surrounding immune-infiltrating cells such as tumor-associated macrophages (TAMs). Macrophages are key actors of both inflammation and its active resolution due to their plastic phenotype. In line with this high plasticity, macrophages can be hijacked by cancer cells to support tumor progression and immune escape, or therapy resistance. Impaired resolution of cancer-associated inflammation supported by TAMs may thus reinforces tumor progression. From this perspective, recent evidence suggests that stimulating macrophage's pro-resolving functions using SPMs can promote inflammation resolution in cancer and improve anticancer treatments. Thus, TAMs' re-education toward an antitumor phenotype by using SPMs opens a new line of attack in cancer treatment. Here, we review SPMs' anticancer capacities with special attention regarding their effects on TAMs. We further discuss how this new therapeutic approach could be envisioned in cancer therapy.


Subject(s)
Inflammation Mediators/immunology , Inflammation/immunology , Neoplasms/immunology , Tumor-Associated Macrophages/immunology , Animals , Humans
4.
Metabolites ; 11(6)2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34207741

ABSTRACT

The natural abundance of heavy stable isotopes (13C, 15N, 18O, etc.) is now of considerable importance in many research fields, including human physiology. In fact, it varies between tissues and metabolites due to isotope effects in biological processes, that is, isotope discriminations between heavy and light isotopic forms during enzyme or transporter activity. The metabolic deregulation associated with many diseases leads to alterations in metabolic fluxes, resulting in changes in isotope abundance that can be identified easily with current isotope ratio technologies. In this review, we summarize the current knowledge on changes in natural isotope composition in samples (including various tissues, hair, plasma, saliva) found in patients compared to controls, caused by human diseases. We discuss the metabolic origin of such isotope fractionations and highlight the potential of using isotopes at natural abundance for medical diagnosis and/or prognostic.

5.
Biomolecules ; 10(8)2020 07 25.
Article in English | MEDLINE | ID: mdl-32722518

ABSTRACT

The imbalance between BCL-2 homologues and pro-death counterparts frequently noted in cancer cells endows them with a cell autonomous survival advantage. To eradicate ectopic cells, inhibitors of these homologues (BH3 mimetics) were developed to trigger, during anticancer treatment, full activation of the canonical mitochondrial apoptotic pathway and related caspases. Despite efficiency in some clinical settings, these compounds do not completely fulfill their initial promise. We herein put forth that a growing body of evidence indicates that mitochondrial integrity, controlled by BCL-2 family proteins, and downstream caspases regulate other cell death modes and influence extracellular signaling by committed cells. Moreover, intercellular communications play a key role in spreading therapeutic response across cancer cell populations and in engaging an immune response. We thus advocate that BH3 mimetics administration would be more efficient in the long term if it did not induce apoptosis in all sensitive cells at the same time, but if it could instead allow (or trigger) death signal production by non-terminally committed dying cell populations. The development of such a trade-off strategy requires to unravel the effects of BH3 mimetics not only on each individual cancer cell but also on homotypic and heterotypic cell interactions in dynamic tumor ecosystems.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Drug Discovery , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Molecular Targeted Therapy , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism
6.
Mol Cell Oncol ; 7(3): 1735912, 2020.
Article in English | MEDLINE | ID: mdl-32391429

ABSTRACT

We recently identified a previously unappreciated ability of antimitotics to propagate apoptotic priming across cancer cell populations. The underlying paracrine cytotoxic signal, fueled by undead cells activating the cGAS/STING pathway, is required for in vivo antitumor response and it can be further exploited by delayed, but not synchronous, BCL-xL inhibition.

7.
Nat Commun ; 11(1): 259, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31937780

ABSTRACT

A fascinating but uncharacterized action of antimitotic chemotherapy is to collectively prime cancer cells to apoptotic mitochondrial outer membrane permeabilization (MOMP), while impacting only on cycling cell subsets. Here, we show that a proapoptotic secretory phenotype is induced by activation of cGAS/STING in cancer cells that are hit by antimitotic treatment, accumulate micronuclei and maintain mitochondrial integrity despite intrinsic apoptotic pressure. Organotypic cultures of primary human breast tumors and patient-derived xenografts sensitive to paclitaxel exhibit gene expression signatures typical of type I IFN and TNFα exposure. These cytokines induced by cGAS/STING activation trigger NOXA expression in neighboring cells and render them acutely sensitive to BCL-xL inhibition. cGAS/STING-dependent apoptotic effects are required for paclitaxel response in vivo, and they are amplified by sequential, but not synchronous, administration of BH3 mimetics. Thus anti-mitotic agents propagate apoptotic priming across heterogeneously sensitive cancer cells through cytosolic DNA sensing pathway-dependent extracellular signals, exploitable by delayed MOMP targeting.


Subject(s)
Antimitotic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Membrane Proteins/metabolism , Paracrine Communication/drug effects , Animals , Breast Neoplasms/metabolism , Cell Line , Female , Gene Knockout Techniques , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Membrane Proteins/genetics , Mice , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/metabolism
8.
Int J Mol Sci ; 20(22)2019 Nov 11.
Article in English | MEDLINE | ID: mdl-31717935

ABSTRACT

BACKGROUND: Considering the positive or negative potential effects of adipocytes, depending on their lipid composition, on breast tumor progression, it is important to evaluate whether adipose tissue (AT) harvesting procedures, including epinephrine infiltration, may influence breast cancer progression. METHODS: Culture medium conditioned with epinephrine-infiltrated adipose tissue was tested on human Michigan Cancer Foundation-7 (MCF7) breast cancer cells, cultured in monolayer or in oncospheres. Lipid composition was evaluated depending on epinephrine-infiltration for five patients. Epinephrine-infiltrated adipose tissue (EI-AT) or corresponding conditioned medium (EI-CM) were injected into orthotopic breast carcinoma induced in athymic mouse. RESULTS: EI-CM significantly increased the proliferation rate of MCF7 cells Moreover EI-CM induced an output of the quiescent state of MCF7 cells, but it could be either an activator or inhibitor of the epithelial mesenchymal transition as indicated by gene expression changes. EI-CM presented a significantly higher lipid total weight compared with the conditioned medium obtained from non-infiltrated-AT of paired-patients. In vivo, neither the EI-CM or EI-AT injection significantly promoted MCF7-induced tumor growth. CONCLUSIONS: Even though conditioned media are widely used to mimic the secretome of cells or tissues, they may produce different effects on tumor progression, which may explain some of the discrepancy observed between in vitro, preclinical and clinical data using AT samples.


Subject(s)
Adipose Tissue/metabolism , Cell Proliferation/drug effects , Epinephrine/pharmacology , Lipid Metabolism , Mammary Neoplasms, Experimental/metabolism , Xenograft Model Antitumor Assays/methods , Animals , Culture Media, Conditioned/pharmacology , Female , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Xenograft Model Antitumor Assays/standards
9.
Eur J Med Chem ; 162: 334-347, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30453244

ABSTRACT

We describe the first examples of small molecules able to disrupt the nanomolar interaction between the pro-apoptotic protein PUMA and its anti-apoptotic counterpart BcL-xL in malignant cells. Based on molecular modelling studies, we propose a rationale to this result, through a new "bottle-opener"-type strategy which could be of general use in the area of protein-protein interaction studies.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Models, Molecular , Proto-Oncogene Proteins/metabolism , bcl-X Protein/metabolism , Animals , Apoptosis/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Protein Interaction Domains and Motifs/drug effects
10.
Blood Adv ; 2(23): 3492-3505, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30530776

ABSTRACT

In this study, we assessed the sensitivity of myeloma cells to the oncolytic measles virus (MV) in relation to p53 using 37 cell lines and 23 primary samples. We showed that infection and cell death were correlated with CD46 expression, which was associated with TP53 status; TP53 abn cell lines highly expressed CD46 and were preferentially infected by MV when compared with the TP53 wt cell lines (P = .046 and P = .045, respectively). Infection of myeloma cells was fully dependent on CD46 expression in both cell lines and primary cells. In the TP53 wt cell lines, but not the TP53 abn cell lines, activation of the p53 pathway with nutlin3a inhibited both CD46 expression and MV infection, while TP53 silencing reciprocally increased CD46 expression and MV infection. We showed using a p53 chromatin immunoprecipitation assay and microRNA assessment that CD46 gene expression was directly and indirectly regulated by p53. Primary myeloma cells overexpressed CD46 as compared with normal cells and were highly infected and killed by MV. CD46 expression and MV infection were inhibited by nutlin3a in primary p53-competent myeloma cells, but not in p53-deficient myeloma cells, and the latter were highly sensitive to MV infection. In summary, myeloma cells were highly sensitive to MV and infection inhibition by the p53 pathway was abrogated in p53-deficient myeloma cells. These results argue for an MV-based clinical trial for patients with p53 deficiency.


Subject(s)
Measles virus/physiology , Membrane Cofactor Protein/metabolism , Multiple Myeloma/pathology , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Humans , Membrane Cofactor Protein/antagonists & inhibitors , Membrane Cofactor Protein/genetics , MicroRNAs/metabolism , Multiple Myeloma/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
11.
EMBO Rep ; 19(2): 234-243, 2018 02.
Article in English | MEDLINE | ID: mdl-29233828

ABSTRACT

E2F1 is the main pro-apoptotic effector of the pRB-regulated tumor suppressor pathway by promoting the transcription of various pro-apoptotic proteins. We report here that E2F1 partly localizes to mitochondria, where it favors mitochondrial outer membrane permeabilization. E2F1 interacts with BCL-xL independently from its BH3 binding interface and induces a stabilization of BCL-xL at mitochondrial membranes. This prevents efficient control of BCL-xL over its binding partners, in particular over BAK resulting in the induction of cell death. We thus identify a new, non-BH3-binding regulator of BCL-xL localization dynamics that influences its anti-apoptotic activity.


Subject(s)
Cell Death , E2F1 Transcription Factor/metabolism , bcl-X Protein/metabolism , Apoptosis , Cell Line, Tumor , E2F1 Transcription Factor/chemistry , Extracellular Space/metabolism , Gene Expression Regulation/drug effects , Humans , Mitochondria/metabolism , Protein Binding , Protein Transport , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription, Genetic , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/chemistry
12.
Nat Commun ; 8(1): 1123, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29066722

ABSTRACT

In tumours, accumulation of chemoresistant cells that express high levels of anti-apoptotic proteins such as BCL-XL is thought to result from the counter selection of sensitive, low expresser clones during progression and/or initial treatment. We herein show that BCL-XL expression is selectively advantageous to cancer cell populations even in the absence of pro-apoptotic pressure. In transformed human mammary epithelial cells BCL-XL favours full activation of signalling downstream of constitutively active RAS with which it interacts in a BH4-dependent manner. Comparative proteomic analysis and functional assays indicate that this is critical for RAS-induced expression of stemness regulators and maintenance of a cancer initiating cell (CIC) phenotype. Resistant cancer cells thus arise from a positive selection driven by BCL-XL modulation of RAS-induced self-renewal, and during which apoptotic resistance is not necessarily the directly selected trait.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/cytology , Signal Transduction , bcl-X Protein/metabolism , ras Proteins/metabolism , Animals , Apoptosis , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , HMGA2 Protein/metabolism , Humans , MCF-7 Cells , Mass Spectrometry , Mice , Mice, Nude , Neoplasm Recurrence, Local , Phenotype , Plasmids/metabolism , Proteomics , Proto-Oncogene Proteins c-fos/metabolism
13.
Sci Rep ; 6: 34251, 2016 Sep 28.
Article in English | MEDLINE | ID: mdl-27678172

ABSTRACT

Breast cancer is the most common cancer in women worldwide. Despite the information provided by anatomopathological assessment and molecular markers (such as receptor expression ER, PR, HER2), breast cancer therapies and prognostics depend on the metabolic properties of tumor cells. However, metabolomics have not provided a robust and congruent biomarker yet, likely because individual metabolite contents are insufficient to encapsulate all of the alterations in metabolic fluxes. Here, we took advantage of natural 13C and 15N isotope abundance to show there are isotopic differences between healthy and cancer biopsy tissues or between healthy and malignant cultured cell lines. Isotope mass balance further suggests that these differences are mostly related to lipid metabolism, anaplerosis and urea cycle, three pathways known to be impacted in malignant cells. Our results demonstrate that the isotope signature is a good descriptor of metabolism since it integrates modifications in C partitioning and N excretion altogether. Our present study is thus a starting point to possible clinical applications such as patient screening and biopsy characterization in every cancer that is associated with metabolic changes.

14.
Eur J Med Chem ; 119: 17-33, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27153346

ABSTRACT

Hsp90 is an ATP-dependent chaperone known to be overexpressed in many cancers. This way, Hsp90 is an important target for drug discovery. Novobiocin, an aminocoumarin antibiotic, was reported to inhibit Hsp90 targeting C-terminal domain, and showed anti-proliferative properties, leading to the development of new and more active compounds. Consequently, a new set of novobiocin analogs derived from 1,6-naphthyridin-2(1H)-one scaffold was designed, synthesized and evaluated against two breast cancer cell lines. Subsequently, cell cycle progression and apoptosis were conducted on best candidates, finally Western Blot analysis was performed to measure their ability to induce degradation of Hsp90 client proteins.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , HSP90 Heat-Shock Proteins/metabolism , Molecular Targeted Therapy , Naphthyridines/chemistry , Naphthyridines/pharmacology , Amines/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Hydrophobic and Hydrophilic Interactions
15.
Planta Med ; 82(11-12): 967-72, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27224268

ABSTRACT

The seeds of Carapa procera are exploited extensively in West African ethnopharmacy for the treatment of several pathologies, including inflammation. They also are effective as insect antifeedants and as a mosquito repellent. With the aim of identifying bioactive principles, an ethyl acetate extract of the defatted seeds was made and fractionated. Two principle compounds were isolated. One of these, 5,6-dehydro-7-deacetoxy-7-oxogedunin (1), while known from another genus of the Meliaceae, is newly identified from the genus Carapa and its X-ray structure is described for the first time. In addition, 1 displayed strong anti-clonogenic activity at 10 µM. The other compound, mexicanolide (2), is known from this species and showed neither cytotoxicity nor anti-clonogenicity. These differences in efficacy are discussed in relation to known structure-activity relationships of limonoids.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Limonins/isolation & purification , Meliaceae/chemistry , Plant Extracts/isolation & purification , Triterpenes/isolation & purification , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Limonins/chemistry , Limonins/pharmacology , Molecular Structure , Plant Extracts/chemistry , Plant Extracts/pharmacology , Seeds/chemistry , Triterpenes/chemistry , Triterpenes/pharmacology
16.
Breast Cancer Res ; 18(1): 40, 2016 Apr 02.
Article in English | MEDLINE | ID: mdl-27039296

ABSTRACT

BACKGROUND: ADAM8 (a disintegrin and metalloproteinase 8) protein promotes the invasive and metastatic phenotype of triple-negative breast cancer (TNBC) cells. High ADAM8 expression in breast cancer patients is an independent predictor of poor prognosis. Here, we investigated whether ADAM8 regulates specific miRNAs, their roles in aggressive phenotype, and potential use as biomarkers of disease. METHODS: Microarray analysis was performed on RNA from MDA-MB-231 cells after transient ADAM8 knockdown using TaqMan miRNA cards. Changes in miRNA levels were confirmed using two ADAM8 siRNAs in TNBC cell lines. Kinase inhibitors, ß1-integrin antagonist antibody, and different forms of ADAM8 were employed to elucidate the signaling pathway required for miR-720 expression. miR-720 levels were modulated using a specific antagomiR or a mimic, and effects on aggressive phenotype of TNBC cells were determined using Boyden chamber and 3D-Matrigel outgrowth assays. Plasma was isolated from mice before and after implantation of MDA-MB-231 cells and analyzed for miR-720 levels. Serum samples of TNBC patients were evaluated for their ADAM8 and miR-720 levels. RESULTS: We identified 68 miRNAs differentially regulated upon ADAM8 knockdown, including decreased levels of secreted miR-720. Ectopic overexpression of wild-type ADAM8 or forms that lack metalloproteinase activity similarly induced miR-720 levels. The disintegrin and cysteine-rich domains of ADAM8 were shown to induce miR-720 via activation of a ß1-integrin to ERK signaling cascade. Knockdown of miR-720 led to a significant decrease in migratory and invasive abilities of TNBC cells. Conversely, miR-720 overexpression rescued these properties. A profound increase in plasma levels of miR-720 was detected 7 days after TNBC cell inoculation into mouse mammary fat pads when tumors were barely palpable. Concordantly, miR-720 levels were found to be significantly higher in serum samples of TNBC patients with high ADAM8 expression. CONCLUSIONS: We have shown for the first time that miR-720 is induced by ADAM8 signaling via ERK and plays an essential role in promoting the aggressive phenotype of TNBCs. miR-720 is elevated in serum of patients with ADAM8-high TNBC and, in a group with other miRNAs downstream of ADAM8, holds promise as a biomarker for early detection of or treatment response of ADAM8-positive TNBCs.


Subject(s)
ADAM Proteins/biosynthesis , Membrane Proteins/biosynthesis , MicroRNAs/biosynthesis , Neoplasm Invasiveness/genetics , Triple Negative Breast Neoplasms/genetics , ADAM Proteins/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System , Membrane Proteins/genetics , Mice , MicroRNAs/genetics , RNA, Small Interfering , Triple Negative Breast Neoplasms/pathology
17.
Oncotarget ; 7(12): 14064-82, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26908442

ABSTRACT

Terminal fucosylated motifs of glycoproteins and glycolipid chains are often altered in cancer cells. We investigated the link between fucosylation changes and critical steps in cancer progression: epithelial-to-mesenchymal transition (EMT) and lymph node metastasis.Using mammary cell lines, we demonstrate that during EMT, expression of some fucosylated antigens (e.g.: Lewis Y) is decreased as a result of repression of the fucosyltransferase genes FUT1 and FUT3. Moreover, we identify the fucose-binding bacterial lectin BC2L-C-Nt as a specific probe for the epithelial state.Prolectin (CLEC17A), a human lectin found on lymph node B cells, shares ligand specificities with BC2L-C-Nt. It binds preferentially to epithelial rather than to mesenchymal cells, and microfluidic experiments showed that prolectin behaves as a cell adhesion molecule for epithelial cells. Comparison of paired primary tumors/lymph node metastases revealed an increase of prolectin staining in metastasis and high FUT1 and FUT3 mRNA expression was associated with poor prognosis. Our data suggest that tumor cells invading the lymph nodes and expressing fucosylated motifs associated with the epithelial state could use prolectin as a colonization factor.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Cell Adhesion , Fucosyltransferases/metabolism , Lectins, C-Type/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Fucosyltransferases/genetics , Humans , Lectins, C-Type/genetics , Lymphatic Metastasis , Tumor Cells, Cultured , Galactoside 2-alpha-L-fucosyltransferase
18.
Med Chem ; 12(5): 419-25, 2016.
Article in English | MEDLINE | ID: mdl-26825069

ABSTRACT

The Bcl-2 family includes 26 proteins involved in apoptosis. Cancer cells can develop the ability to avoid apoptosis through the upregulation and/or down regulation of such proteins Bax, Bcl-xL or Mcl-1, especially during chemoresistance progress. These proteins engaged in a network of dynamic interactions that control apoptosis triggering have become attractive therapeutic targets in cancers including melanoma. Among them, the Bax/Bcl-xL interaction appears critical in maintaining mitochondria integrity. Therefore a series of mixed polyphenol-heterocyclic molecules, were rationally designed by molecular docking as Bax/Bcl-xL inhibitors. It has been screened against B16-F10 melanoma cancer cells for a preliminary investigation of their cytotoxicity. All these compounds exhibited a significant cytotoxicity against these cancer cells, in the 0.3-6 .M range. A pyrazole-type molecule, which had a submicromolar IC50 value with an excellent selectivity index (14), is the most promising derivative for further development.


Subject(s)
Antineoplastic Agents/pharmacology , Catechols/pharmacology , Pyrazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Bioluminescence Resonance Energy Transfer Techniques , Catechols/chemical synthesis , Cell Line, Tumor , HeLa Cells , Humans , Melanoma, Experimental , Molecular Docking Simulation , Pyrazoles/chemical synthesis , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-X Protein/antagonists & inhibitors
19.
Breast Cancer Res Treat ; 155(1): 53-63, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26679694

ABSTRACT

Survivin overexpression, frequently found in breast cancers and others, is associated with poor prognosis. Its dual regulation of cell division and apoptosis makes it an attractive therapeutic target but its exact functions that are required for tumor maintenance are still elusive. Survivin protects cancer cells from genotoxic agents and this ability is generally assigned to a universal anti-apoptotic function. However, a specific role in cancer cell protection from DNA damage has been overlooked so far. We assessed DNA damage occurrence in Survivin-depleted breast cancer cells using γH2AX staining and comete assay. QPCR data and a gene conversion assay indicated that homologous recombination (HR) was impaired upon Survivin depletion. We conducted the analysis of Survivin and HR genes' expression in breast tumors. We revealed BRCAness phenotype of Survivin-depleted cells using cell death assays combined to PARP targeting. Survivin silencing leads to DNA double-strand breaks in breast cancer cells and functionally reduces HR. Survivin depletion decreases the transcription of a set of genes involved in HR, decreases RAD51 protein expression and impairs the endonuclease complex MUS81/EME1 involved in the resolution of Holliday junctions. Clinically, EME1, RAD51, EXO1, BLM expressions correlate with that of BIRC5 (coding for Survivin) and are of prognostic value. Functionally, Survivin depletion triggers p53 activation and sensitizes cancer cells to of PARP inhibition. We defined Survivin as a constitutive actor of HR in breast cancers, and implies that its inhibition would enhance cell vulnerability upon PARP inhibition.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , DNA Repair , Homologous Recombination , Inhibitor of Apoptosis Proteins/metabolism , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Cdc20 Proteins/genetics , Cdc20 Proteins/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , Drug Resistance, Neoplasm , Female , Gene Expression , Gene Knockdown Techniques , Gene Silencing , Histones/metabolism , Humans , Inhibitor of Apoptosis Proteins/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Prognosis , Survivin , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Oncotarget ; 6(15): 13476-86, 2015 May 30.
Article in English | MEDLINE | ID: mdl-25974963

ABSTRACT

Specific overexpression in cancer cells and evidence of oncogenic functions make Survivin an attractive target in cancer therapy. The small molecule compound YM155 has been described as the first "Survivin suppressant" but molecular mechanisms involved in its biological activity and its clinical potential remain obscure. We herein show that YM155 exerts single agent toxicity on primary breast cancer cells grown in an ex vivo assay preserving tumor microenvironment. In vitro assays indicate that YM155 more efficiently triggers cell death in breast cancer cells (including these with stem-cell like properties) than in non tumorigenic mammary cells. YM155-induced cell death is critically dependent on autophagy and NF-kB but independent of p53 and it coïncides with DNA damage and a DNA damage response in p53-proficient cells. Our results point out a crosstalk between NF-kB and autophagy controlling YM155-induced death in breast cancer cells and argue for the potential use of YM155 as a genotoxic agent in breast cancer therapy.


Subject(s)
Breast Neoplasms/drug therapy , Imidazoles/pharmacology , NF-kappa B/metabolism , Naphthoquinones/pharmacology , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage , Female , Humans , MCF-7 Cells , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...