Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cancers (Basel) ; 15(2)2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36672396

ABSTRACT

Hepatocellular carcinoma (HCC) is a frequent and deadly cancer in need of new treatments. Immunotherapy has shown promising results in several solid tumors. The TIGIT/DNAM-1 axis gathers targets for new immune checkpoint inhibitors (ICIs). Here, we aimed at highlighting the potential of this axis as a new therapeutic option for HCC. For this, we built a large transcriptomic database of 683 HCC samples, clinically annotated, and 319 normal liver tissues. We interrogated this database for the transcriptomic expression of each member of the TIGIT/DNAM-1 axis and tested their prognostic value for survival. We then focused on the most discriminant one for these criteria, i.e., PVRIG, and analyzed the clinical characteristics, the disease-free and overall survivals, and biological pathways associated with PVRIG High tumors. Among all members of the TIGIT/DNAM-1 axis, PVRIG expression was higher in tumors than in normal liver, was heterogeneous across tumors, and was the only member with independent prognostic value for better survival. PVRIG High tumors were characterized by a higher lymphocytic infiltrate and enriched for signatures associated with tertiary lymphoid structures and better anti-tumor immune response. These results suggest that patients with PVRIG High tumors might be good candidates for immune therapy involving ICIs, notably ICIs targeting the TIGIT/DNAM-1 axis. Further functional and clinical validation is urgently required.

3.
Mol Cancer Ther ; 21(7): 1227-1235, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35534238

ABSTRACT

Antibody-drug conjugates (ADC) represent a fast-growing drug class in oncology. However, ADCs are associated with resistance, and therapies able to overcome it are of utmost importance. Recently, enfortumab vedotin-ejfv (EV) was approved in nectin-4+ metastatic urothelial cancer. We previously described PVRL4/nectin-4 as a new therapeutic target in breast cancer and produced an efficient EV-like ADC comprising a human anti-nectin-4 mAb conjugated to monomethyl auristatin-E (MMAE) named N41mab-vcMMAE. To study the consequence of the long-term treatment with this ADC, we developed a preclinical breast cancer model in mice, and report a mechanism of resistance to N41mab-vcMMAE after 9-month treatment and a way to reverse it. RNA-sequencing pointed to an upregulation in resistant tumors of ABCB1 expression, encoding the multidrug resistance protein MDR-1/P-glycoprotein (P-gp), associated with focal gene amplification and high protein expression. Sensitivity to N41mab-vcMMAE of the resistant model was restored in vitro by P-gp pharmacologic inhibitors, like tariquidar. P-gp is expressed in a variety of normal tissues. By delivering the drug to the tumor more specifically than classical chemotherapy, we hypothesized that the combined use of ADC with P-gp inhibitors might reverse resistance in vivo without toxicity. Indeed, we showed that the tariquidar/N41mab-vcMMAE combination was well tolerated and induced a rapid regression of ADC-resistant tumors in mice. In contrast, the tariquidar/docetaxel combination was toxic and poorly efficient. These results show that ABC transporter inhibitors can be safely used with ADC to reverse ADC-induced resistance and open new opportunities in the fight against multidrug resistance.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Carcinoma, Transitional Cell , Immunoconjugates , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carcinoma, Transitional Cell/drug therapy , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Female , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Mice
4.
Cancers (Basel) ; 14(4)2022 Feb 13.
Article in English | MEDLINE | ID: mdl-35205679

ABSTRACT

Circulating tumor cells have a strong potential as a quasi-non-invasive tool for setting up a precision medicine strategy for cancer patients. Using a second-generation "filtration-based" technology to isolate CTCs, the Screencell™ technology (Sarcelles, France), we performed a large and simultaneous analysis of all atypical circulating tumor cells (aCTCs) isolated from the blood of metastatic breast cancer (mBC) patients. We correlated their presence with clinicopathological and survival data. We included 91 mBC patients from the PERMED-01 study. The median number of aCTCs was 8.3 per mL of blood. Three subsets of aCTCs, absent from controls, were observed in patients: single (s-aCTCs), circulating tumor micro-emboli (CTM), and giant-aCTCs (g-aCTCs). The presence of g-aCTCs was associated with shorter progression free survival and overall survival. This study highlights the heterogeneity of aCTCs in mBC patients both at the cytomorphological and molecular levels. In addition, it suggests the usefulness of the g-aCTC subset as a prognostic factor and a potential stratification tool to treat late-stage mBC patients and improve their chances of benefiting from early clinical trials.

5.
Nat Commun ; 12(1): 2198, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33850160

ABSTRACT

Cancer is initiated by somatic mutations in oncogenes or tumor suppressor genes. However, additional alterations provide selective advantages to the tumor cells to resist treatment and develop metastases. Their identification is of paramount importance. Reduced expression of EFA6B (Exchange Factor for ARF6, B) is associated with breast cancer of poor prognosis. Here, we report that loss of EFA6B triggers a transcriptional reprogramming of the cell-to-ECM interaction machinery and unleashes CDC42-dependent collective invasion in collagen. In xenograft experiments, MCF10 DCIS.com cells, a DCIS-to-IDC transition model, invades faster when knocked-out for EFA6B. In addition, invasive and metastatic tumors isolated from patients have lower expression of EFA6B and display gene ontology signatures identical to those of EFA6B knock-out cells. Thus, we reveal an EFA6B-regulated molecular mechanism that controls the invasive potential of mammary cells; this finding opens up avenues for the treatment of invasive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Animals , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Knockout Techniques , Humans , Mice , Mice, Nude , Transcriptome , cdc42 GTP-Binding Protein
6.
Hum Mol Genet ; 27(19): 3377-3391, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29982567

ABSTRACT

Skeletal dysplasias are a clinically and genetically heterogeneous group of bone and cartilage disorders. A total of 436 skeletal dysplasias are listed in the 2015 revised version of the nosology and classification of genetic skeletal disorders, of which nearly 20% are still genetically and molecularly uncharacterized. We report the clinical and molecular characterization of a lethal skeletal dysplasia of the short-rib group caused by mutation of the mouse Fop gene. Fop encodes a centrosomal and centriolar satellite (CS) protein. We show that Fop mutation perturbs ciliogenesis in vivo and that this leads to the alteration of the Hedgehog signaling pathway. Fop mutation reduces CSs movements and affects pericentriolar material composition, which probably participates to the ciliogenesis defect. This study highlights the role of a centrosome and CSs protein producing phenotypes in mice that recapitulate a short rib-polydactyly syndrome when mutated.


Subject(s)
Ciliopathies/genetics , Proto-Oncogene Proteins/genetics , Short Rib-Polydactyly Syndrome/genetics , Transcription Factors/genetics , Animals , Centrioles/genetics , Centrosome/metabolism , Centrosome/pathology , Cilia/genetics , Cilia/pathology , Ciliopathies/physiopathology , Humans , Mice , Mutation , Short Rib-Polydactyly Syndrome/physiopathology
7.
Nat Chem ; 9(10): 1025-1033, 2017 10.
Article in English | MEDLINE | ID: mdl-28937680

ABSTRACT

Cancer stem cells (CSCs) represent a subset of cells within tumours that exhibit self-renewal properties and the capacity to seed tumours. CSCs are typically refractory to conventional treatments and have been associated to metastasis and relapse. Salinomycin operates as a selective agent against CSCs through mechanisms that remain elusive. Here, we provide evidence that a synthetic derivative of salinomycin, which we named ironomycin (AM5), exhibits a more potent and selective activity against breast CSCs in vitro and in vivo, by accumulating and sequestering iron in lysosomes. In response to the ensuing cytoplasmic depletion of iron, cells triggered the degradation of ferritin in lysosomes, leading to further iron loading in this organelle. Iron-mediated production of reactive oxygen species promoted lysosomal membrane permeabilization, activating a cell death pathway consistent with ferroptosis. These findings reveal the prevalence of iron homeostasis in breast CSCs, pointing towards iron and iron-mediated processes as potential targets against these cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Iron/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Pyrans/pharmacology , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , Homeostasis/drug effects , Humans , Lysosomes/chemistry , Molecular Conformation , Neoplastic Stem Cells/metabolism , Pyrans/chemistry , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism
8.
Nat Med ; 23(5): 568-578, 2017 May.
Article in English | MEDLINE | ID: mdl-28394329

ABSTRACT

Chromosomal instability (CIN), a feature of most adult neoplasms from their early stages onward, is a driver of tumorigenesis. However, several malignancy subtypes, including some triple-negative breast cancers, display a paucity of genomic aberrations, thus suggesting that tumor development may occur in the absence of CIN. Here we show that the differentiation status of normal human mammary epithelial cells dictates cell behavior after an oncogenic event and predetermines the genetic routes toward malignancy. Whereas oncogene induction in differentiated cells induces massive DNA damage, mammary stem cells are resistant, owing to a preemptive program driven by the transcription factor ZEB1 and the methionine sulfoxide reductase MSRB3. The prevention of oncogene-induced DNA damage precludes induction of the oncosuppressive p53-dependent DNA-damage response, thereby increasing stem cells' intrinsic susceptibility to malignant transformation. In accord with this model, a subclass of breast neoplasms exhibit unique pathological features, including high ZEB1 expression, a low frequency of TP53 mutations and low CIN.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , Cell Differentiation/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Genomic Instability/genetics , Methionine Sulfoxide Reductases/genetics , Stem Cells/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Chromatin Immunoprecipitation , DNA Damage , Epithelial Cells/cytology , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Immunoblotting , Mammary Glands, Human/cytology , Methionine Sulfoxide Reductases/metabolism , Mice, Inbred NOD , Middle Aged , Reactive Oxygen Species , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Stem Cells/cytology , Tissue Array Analysis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Young Adult , Zinc Finger E-box-Binding Homeobox 1/metabolism
9.
Cell Rep ; 18(9): 2256-2268, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28249169

ABSTRACT

Breast cancer stem cells (bCSCs) have been implicated in tumor progression and therapeutic resistance; however, the molecular mechanisms that define this state are unclear. We have performed two microRNA (miRNA) gain- and loss-of-function screens to identify miRNAs that regulate the choice between bCSC self-renewal and differentiation. We find that micro-RNA (miR)-600 silencing results in bCSC expansion, while its overexpression reduces bCSC self-renewal, leading to decreased in vivo tumorigenicity. miR-600 targets stearoyl desaturase 1 (SCD1), an enzyme required to produce active, lipid-modified WNT proteins. In the absence of miR-600, WNT signaling is active and promotes self-renewal, whereas overexpression of miR-600 inhibits the production of active WNT and promotes bCSC differentiation. In a series of 120 breast tumors, we found that a low level of miR-600 is correlated with active WNT signaling and a poor prognosis. These findings highlight a miR-600-centered signaling network that governs bCSC-fate decisions and influences tumor progression.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , MicroRNAs/genetics , Neoplastic Stem Cells/pathology , Signal Transduction/physiology , Wnt Proteins/genetics , Wnt Signaling Pathway/physiology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Stearoyl-CoA Desaturase/genetics
10.
Cancer Res ; 74(19): 5493-506, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25115298

ABSTRACT

One of the earliest events in epithelial carcinogenesis is the dissolution of tight junctions and cell polarity signals that are essential for normal epithelial barrier function. Here, we report that EFA6B, a guanine nucleotide exchange factor for the Ras superfamily protein Arf6 that helps assemble and stabilize tight junction, is required to maintain apico-basal cell polarity and mesenchymal phenotypes in mammary epithelial cells. In organotypic three-dimensional cell cultures, endogenous levels of EFA6B were critical to determine epithelial-mesenchymal status. EFA6B downregulation correlated with a mesenchymal phenotype and ectopic expression of EFA6B hampered TGFß-induced epithelial-to-mesenchymal transition (EMT). Transcriptomic and immunohistochemical analyses of human breast tumors revealed that the reduced expression of EFA6B was associated with loss of tight junction components and with increased signatures of EMT, cancer stemness, and poor prognosis. Accordingly, tumors with low levels of EFA6B were enriched in the aggressive triple-negative and claudin-low breast cancer subtypes. Our results identify EFA6B as a novel antagonist in breast cancer and they point to its regulatory and signaling pathways as rational therapeutic targets in aggressive forms of this disease.


Subject(s)
Breast Neoplasms/physiopathology , Guanine Nucleotide Exchange Factors/physiology , Breast Neoplasms/pathology , Cell Line, Tumor , Claudin-3/metabolism , Epithelial-Mesenchymal Transition , Female , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Middle Aged , RNA, Messenger/genetics , Tight Junctions/physiology
11.
Clin Cancer Res ; 19(23): 6520-31, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24141629

ABSTRACT

PURPOSE: Cancer stem cells (CSC) are the tumorigenic cell population that has been shown to sustain tumor growth and to resist conventional therapies. The purpose of this study was to evaluate the potential of histone deacetylase inhibitors (HDACi) as anti-CSC therapies. EXPERIMENTAL DESIGN: We evaluated the effect of the HDACi compound abexinostat on CSCs from 16 breast cancer cell lines (BCL) using ALDEFLUOR assay and tumorsphere formation. We performed gene expression profiling to identify biomarkers predicting drug response to abexinostat. Then, we used patient-derived xenograft (PDX) to confirm, in vivo, abexinostat treatment effect on breast CSCs according to the identified biomarkers. RESULTS: We identified two drug-response profiles to abexinostat in BCLs. Abexinostat induced CSC differentiation in low-dose sensitive BCLs, whereas it did not have any effect on the CSC population from high-dose sensitive BCLs. Using gene expression profiling, we identified the long noncoding RNA Xist (X-inactive specific transcript) as a biomarker predicting BCL response to HDACi. We validated that low Xist expression predicts drug response in PDXs associated with a significant reduction of the breast CSC population. CONCLUSIONS: Our study opens promising perspectives for the use of HDACi as a differentiation therapy targeting the breast CSCs and identified a biomarker to select patients with breast cancer susceptible to responding to this treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , Breast Neoplasms/pathology , Cell Differentiation/drug effects , Hydroxamic Acids/pharmacology , Neoplastic Stem Cells/physiology , RNA, Long Noncoding/metabolism , Animals , Antineoplastic Agents/therapeutic use , Benzofurans/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/therapeutic use , Inhibitory Concentration 50 , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , RNA, Long Noncoding/genetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Cancer Res ; 73(24): 7290-300, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24142344

ABSTRACT

Cancer stem-like cells (CSC) have been widely studied, but their clinical relevance has yet to be established in breast cancer. Here, we report the establishment of primary breast tumor-derived xenografts (PDX) that encompass the main diversity of human breast cancer and retain the major clinicopathologic features of primary tumors. Successful engraftment was correlated with the presence of ALDH1-positive CSCs, which predicted prognosis in patients. The xenografts we developed showed a hierarchical cell organization of breast cancer with the ALDH1-positive CSCs constituting the tumorigenic cell population. Analysis of gene expression from functionally validated CSCs yielded a breast CSC signature and identified a core transcriptional program of 19 genes shared with murine embryonic, hematopoietic, and neural stem cells. This generalized stem cell program allowed the identification of potential CSC regulators, which were related mainly to metabolic processes. Using an siRNA genetic screen designed to target the 19 genes, we validated the functional role of this stem cell program in the regulation of breast CSC biology. Our work offers a proof of the functional importance of CSCs in breast cancer, and it establishes the reliability of PDXs for use in developing personalized CSC therapies for patients with breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Isoenzymes/metabolism , Neoplastic Stem Cells/enzymology , Retinal Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Differentiation/genetics , Female , Gene Expression Profiling , Heterografts , Humans , Isoenzymes/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/pathology , Nucleic Acid Hybridization , Prognosis , Prospective Studies , Retinal Dehydrogenase/genetics , Transfection
13.
PLoS One ; 7(9): e43409, 2012.
Article in English | MEDLINE | ID: mdl-23028451

ABSTRACT

BACKGROUND: Targeted therapies, associated with standard chemotherapies, have improved breast cancer care. However, primary and acquired resistances are frequently observed and the development of new concepts is needed. High-throughput approaches to identify new active and safe molecules with or without an "a priori" are currently developed. Also, repositioning already-approved drugs in cancer therapy is of growing interest. The thiomorpholine hydroxamate compound TMI-1 has been previously designed to inhibit metalloproteinase activity for the treatment of rheumatoid arthritis. We present here the repositioning of TMI-1 drug in breast cancer. METHODOLOGY/PRINCIPAL FINDINGS: We tested the effect of TMI-1 on luminal, basal and ERBB2-overexpressing breast tumor cell lines and on MMTV-ERBB2/neu tumor evolution. We measured the effects on i) cell survival, ii) cell cycle, iii) extrinsic and intrinsic apoptotic pathways, iv) association with doxorubicin, docetaxel and lapatinib, v) cancer stem cells compartment. In contrast with conventional cytotoxic drugs, TMI-1 was highly selective for tumor cells and cancer stem cells at submicromolar range. All non-malignant cells tested were resistant even at high concentration. TMI-1 was active on triple negative (TN) and ERBB2-overexpressing breast tumor cell lines, and was also highly efficient on human and murine "primary" ERBB2-overexpressing cells. Treatment of transgenic MMTV-ERBB2/neu mice with 100 mg/kg/day TMI-1 alone induced tumor apoptosis, inhibiting mammary gland tumor occurrence and development. No adverse effects were noticed during the treatment. This compound had a strong synergistic effect in association with docetaxel, doxorubicin and lapatinib. We showed that TMI-1 mediates its selective effects by caspase-dependent apoptosis. TMI-1 was efficient in 34/40 tumor cell lines of various origins (ED50: 0.6 µM to 12.5 µM). CONCLUSIONS/SIGNIFICANCE: This is the first demonstration of the tumor selective cytotoxic action of a thiomorpholin hydroxamate compound. TMI-1 is a novel repositionable drug not only for the treatment of adverse prognosis breast cancers but also for other neoplasms.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Morpholines/pharmacology , Animals , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Humans , Metalloproteases/antagonists & inhibitors , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Morpholines/toxicity , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Spheroids, Cellular/drug effects , Tumor Cells, Cultured
14.
Stem Cells ; 30(7): 1327-37, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22605458

ABSTRACT

There is increasing evidence that breast tumors are organized in a hierarchy, with a subpopulation of tumorigenic cancer cells, the cancer stem cells (CSCs), which sustain tumor growth. The characterization of protein networks that govern CSC behavior is paramount to design new therapeutic strategies targeting this subpopulation of cells. We have sought to identify specific molecular pathways of CSCs isolated from 13 different breast cancer cell lines of luminal or basal/mesenchymal subtypes. We compared the gene expression profiling of cancer cells grown in adherent conditions to those of matched tumorsphere cultures. No specific pathway was identified to be commonly regulated in luminal tumorspheres, resulting from a minor CSC enrichment in tumorsphere passages from luminal cell lines. However, in basal/mesenchymal tumorspheres, the enzymes of the mevalonate metabolic pathway were overexpressed compared to those in cognate adherent cells. Inhibition of this pathway with hydroxy-3-methylglutaryl CoA reductase blockers resulted in a reduction of breast CSC independent of inhibition of cholesterol biosynthesis and of protein farnesylation. Further modulation of this metabolic pathway demonstrated that protein geranylgeranylation (GG) is critical to breast CSC maintenance. A small molecule inhibitor of the geranylgeranyl transferase I (GGTI) enzyme reduced the breast CSC subpopulation both in vitro and in primary breast cancer xenografts. We found that the GGTI effect on the CSC subpopulation is mediated by inactivation of Ras homolog family member A (RHOA) and increased accumulation of P27(kip1) in the nucleus. The identification of protein GG as a major contributor to CSC maintenance opens promising perspectives for CSC targeted therapy in basal breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Mevalonic Acid/metabolism , Neoplasms, Basal Cell/metabolism , Neoplastic Stem Cells/metabolism , Animals , Antineoplastic Agents/therapeutic use , Benzamides , Blotting, Western , Breast Neoplasms/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel , Female , Gene Expression Profiling , Humans , Mice , Mice, SCID , Neoplasms, Basal Cell/drug therapy , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Taxoids/therapeutic use
15.
J Clin Invest ; 120(2): 485-97, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20051626

ABSTRACT

Recent evidence suggests that breast cancer and other solid tumors possess a rare population of cells capable of extensive self-renewal that contribute to metastasis and treatment resistance. We report here the development of a strategy to target these breast cancer stem cells (CSCs) through blockade of the IL-8 receptor CXCR1. CXCR1 blockade using either a CXCR1-specific blocking antibody or repertaxin, a small-molecule CXCR1 inhibitor, selectively depleted the CSC population in 2 human breast cancer cell lines in vitro. Furthermore, this was followed by the induction of massive apoptosis in the bulk tumor population via FASL/FAS signaling. The effects of CXCR1 blockade on CSC viability and on FASL production were mediated by the FAK/AKT/FOXO3A pathway. In addition, repertaxin was able to specifically target the CSC population in human breast cancer xenografts, retarding tumor growth and reducing metastasis. Our data therefore suggest that CXCR1 blockade may provide a novel means of targeting and eliminating breast CSCs.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/transplantation , Receptors, Interleukin-8A/antagonists & inhibitors , Stem Cells/pathology , Animals , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Fas Ligand Protein/genetics , Female , Humans , Mice , RNA, Messenger/genetics , Receptors, Interleukin-8A/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Transplantation, Heterologous/pathology
16.
J Immunol ; 183(12): 7939-48, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19923469

ABSTRACT

The TCRbeta gene enhancer (Ebeta) commands TCRbeta gene expression through the lifespan of T lymphocytes. Genetic and molecular studies have implied that in early thymocytes, Ebeta directs chromatin opening over the Dbeta-Jbeta-Cbeta domains and triggers initial Dbeta-Jbeta recombination. In mature T cells, Ebeta is required for expression of the assembled TCRbeta gene. Whether these separate activities rely on distinct Ebeta regulatory sequences and involve differing modes of activation is unclear. Using gene targeting in mouse embryonic stem cells, we replaced Ebeta by a conserved core fragment (Ebeta169). We found that Ebeta169-carrying alleles were capable of sustaining beta gene expression and the development of mature T cells in homozygous knockin mice. Surprisingly, these procedures and underlying molecular transactions were affected to a wide range of degrees depending on the developmental stage. Early thymocytes barely achieved Dbeta-Jbeta germline transcription and recombination. In contrast, T cells displayed substantial though heterogeneous levels of VDJ-rearranged TCRbeta gene expression. Our results have implications regarding enhancer function in cells of the adaptive immune system and, potentially, TCRbeta gene recombination and allelic exclusion.


Subject(s)
Enhancer Elements, Genetic/immunology , Gene Knock-In Techniques , Models, Immunological , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Alleles , Animals , Binding Sites/genetics , Binding Sites/immunology , Enhancer Elements, Genetic/genetics , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , Hybridomas , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/deficiency
18.
PLoS Biol ; 5(3): e43, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17298184

ABSTRACT

It has long been thought that signal joints, the byproducts of V(D)J recombination, are not involved in the dynamics of the rearrangement process. Evidence has now started to accumulate that this is not the case, and that signal joints play unsuspected roles in events that might compromise genomic integrity. Here we show both ex vivo and in vivo that the episomal circles excised during the normal process of receptor gene rearrangement may be reintegrated into the genome through trans-V(D)J recombination occurring between the episomal signal joint and an immunoglobulin/T-cell receptor target. We further demonstrate that cryptic recombination sites involved in T-cell acute lymphoblastic leukemia-associated chromosomal translocations constitute hotspots of insertion. Eventually, the identification of two in vivo cases associating episomal reintegration and chromosomal translocation suggests that reintegration events are linked to genomic instability. Altogether, our data suggest that V(D)J-mediated reintegration of episomal circles, an event likely eluding classical cytogenetic screenings, might represent an additional potent source of genomic instability and lymphoid cancer.


Subject(s)
Genomic Instability , VDJ Recombinases/metabolism , Animals , Cells, Cultured , Leukemia-Lymphoma, Adult T-Cell/genetics , Mice , Polymerase Chain Reaction , Recombination, Genetic , Translocation, Genetic
19.
Immunology ; 109(4): 510-4, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12871217

ABSTRACT

V(D)J recombination and expression of the T-cell receptor beta (TCRbeta) gene are required for the development of the alphabeta T lymphocyte lineage. These processes depend on a transcriptional enhancer (Ebeta) which acts preferentially on adjacent upstream sequences, and has little impact on the 5' distal and 3' proximal regions of the TCRbeta locus. Using knock-in mice, we show that alphabeta T-cell differentiation and TCRbeta gene recombination and expression are not sensitive to the orientation of Ebeta sequences. We discuss the implication of these results regarding the mode of enhancer function at this locus during T lymphocyte development.


Subject(s)
Enhancer Elements, Genetic/genetics , Genes, T-Cell Receptor beta/genetics , T-Lymphocyte Subsets/physiology , Animals , Antigens, CD/genetics , Cell Differentiation/genetics , Flow Cytometry/methods , Gene Expression , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription, Genetic
20.
J Biol Chem ; 278(20): 18101-9, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12639959

ABSTRACT

To assess the role of the T cell receptor (TCR) beta gene enhancer (Ebeta) in regulating the processing of VDJ recombinase-generated coding ends, we assayed TCRbeta rearrangement of Ebeta-deleted (DeltaEbeta) thymocytes in which cell death is inhibited via expression of a Bcl-2 transgene. Compared with DeltaEbeta, DeltaEbeta Bcl-2 thymocytes show a small accumulation of TCRbeta standard recombination products, including coding ends, that involves the proximal Dbeta-Jbeta and Vbeta14 loci but not the distal 5' Vbeta genes. These effects are detectable in double negative pro-T cells, predominate in double positive pre-T cells, and correlate with regional changes in chromosomal structure during double negative-to-double positive differentiation. We propose that Ebeta, by driving long range nucleoprotein interactions and the control of locus expression and chromatin structure, indirectly contributes to the stabilization of coding ends within the recombination processing complexes. The results also illustrate Ebeta-dependent and -independent changes in chromosomal structure, suggesting distinct modes of regulation of TCRbeta allelic exclusion depending on the position within the locus.


Subject(s)
DNA Nucleotidyltransferases/metabolism , Enhancer Elements, Genetic , Genes, T-Cell Receptor beta/genetics , Alleles , Animals , Chromatin/metabolism , DNA Repair , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Genetic , Polymerase Chain Reaction , Recombination, Genetic , Thymus Gland/cytology , VDJ Recombinases
SELECTION OF CITATIONS
SEARCH DETAIL