Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
Curr Issues Mol Biol ; 46(1): 710-728, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38248348

ABSTRACT

The catalytically inactive caspase-8-homologous protein, c-FLIP, is a potent antiapoptotic protein highly expressed in various types of cancers. c-FLIP competes with caspase-8 for binding to the adaptor protein FADD (Fas-Associated Death Domain) following death receptors' (DRs) activation via the ligands of the TNF-R family. As a consequence, the extrinsic apoptotic signaling pathway involving DRs is inhibited. The inhibition of c-FLIP activity in tumor cells might enhance DR-mediated apoptosis and overcome immune and anticancer drug resistance. Based on an in silico approach, the aim of this work was to identify new small inhibitory molecules able to bind selectively to c-FLIP and block its anti-apoptotic activity. Using a homology 3D model of c-FLIP, an in silico screening of 1880 compounds from the NCI database (National Cancer Institute) was performed. Nine molecules were selected for in vitro assays, based on their binding affinity to c-FLIP and their high selectivity compared to caspase-8. These molecules selectively bind to the Death Effector Domain 2 (DED2) of c-FLIP. We have tested in vitro the inhibitory effect of these nine molecules using the human lung cancer cell line H1703, overexpressing c-FLIP. Our results showed that six of these newly identified compounds efficiently prevent FADD/c-FLIP interactions in a molecular pull-down assay, as well as in a DISC immunoprecipitation assay. The overexpression of c-FLIP in H1703 prevents TRAIL-mediated apoptosis; however, a combination of TRAIL with these selected molecules significantly restored TRAIL-induced cell death by rescuing caspase cleavage and activation. Altogether, our findings indicate that new inhibitory chemical molecules efficiently prevent c-FLIP recruitment into the DISC complex, thus restoring the caspase-8-dependent apoptotic cascade. These results pave the way to design new c-FLIP inhibitory molecules that may serve as anticancer agents in tumors overexpressing c-FLIP.

3.
Front Immunol ; 14: 1311658, 2023.
Article in English | MEDLINE | ID: mdl-38152397

ABSTRACT

Background: Immune checkpoint therapies have led to significant breakthroughs in cancer patient treatment in recent years. However, their efficiency is variable, and resistance to immunotherapies is common. VISTA is an immune-suppressive checkpoint inhibitor of T cell response belonging to the B7 family and a promising novel therapeutic target. VISTA is expressed in the immuno-suppressive tumor microenvironment, primarily by myeloid lineage cells, and its genetic knockout or antibody blockade restores an efficient antitumor immune response. Methods: Fully human monoclonal antibodies directed against VISTA were produced after immunizing humanized Trianni mice and single B cell sequencing. Anti-VISTA antibodies were evaluated for specificity, cross-reactivity, monocyte and T cell activation, Fc-effector functions, and antitumor efficacy using in vitro and in vivo models to select the KVA12123 antibody lead candidate. The pharmacokinetics and safety profiles of KVA12123 were evaluated in cynomolgus monkeys. Results: Here, we report the development of a clinical candidate anti-VISTA monoclonal antibody, KVA12123. KVA12123 showed high affinity binding to VISTA through a unique epitope distinct from other clinical-stage anti-VISTA monoclonal antibodies. This clinical candidate demonstrated high specificity against VISTA with no cross-reactivity detected against other members of the B7 family. KVA12123 blocked VISTA binding to its binding partners. KVA12123 induced T cell activation and demonstrated NK-mediated monocyte activation. KVA12123 treatment mediated strong single-agent antitumor activity in several syngeneic tumor models and showed enhanced efficacy in combination with anti-PD-1 treatment. This clinical candidate was engineered to improve its pharmacokinetic characteristics and reduce Fc-effector functions. It was well-tolerated in preclinical toxicology studies in cynomolgus monkeys, where hematology, clinical chemistry evaluations, and clinical observations revealed no indicators of toxicity. No cytokines associated with cytokine release syndrome were elevated. Conclusion: These results establish that KVA12123 is a promising drug candidate with a distinct but complementary mechanism of action of the first generation of immune checkpoint inhibitors. This antibody is currently evaluated alone and in combination with pembrolizumab in a Phase 1/2 open-label clinical trial in patients with advanced solid tumors.


Subject(s)
Antibodies, Monoclonal , Immune Checkpoint Inhibitors , Neoplasms , Animals , Humans , Mice , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Macaca fascicularis , Neoplasms/therapy , T-Lymphocytes , Tumor Microenvironment
4.
Front Oncol ; 13: 1225081, 2023.
Article in English | MEDLINE | ID: mdl-37795437

ABSTRACT

Immune checkpoints limit the activation of the immune system and serve an important homeostatic function but can also restrict immune responses against tumors. Inhibition of specific immune checkpoint proteins such as the B7:CD28 family members programmed cell death protein-1 (PD-1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) has transformed the treatment of various cancers by promoting the anti-tumor activation of immune cells. In contrast to these effects, the V-domain immunoglobulin suppressor of T-cell activation (VISTA) regulates the steady state of the resting immune system and promotes homeostasis by mechanisms distinct from PD-1 and CTLA-4. The effects of VISTA blockade have been shown to include a decrease in myeloid suppression coupled with proinflammatory changes by mechanisms that are separate and distinct from other immune checkpoint proteins; in some preclinical studies these immune effects appear synergistic. Given the potential benefits of VISTA blockade in the context of cancer therapy, the second Annual VISTA Symposium was convened virtually on September 23, 2022, to review new research from investigators and immuno-oncology experts. Discussions in the meeting extended the knowledge of VISTA biology and the effects of VISTA inhibition, particularly on cells of the myeloid lineage and resting T cells, as three candidate anti-VISTA antibodies are in, or nearing, clinical development.

5.
Nanomedicine ; 40: 102499, 2022 02.
Article in English | MEDLINE | ID: mdl-34843982

ABSTRACT

Glioblastoma remains a cancer for which the effectiveness of treatments has shown little improvement over the last decades. For this pathology, multiple therapies combining resection, chemotherapy and radiotherapy remain the norm. In this context, the use of high-Z nanoparticles such as gold or hafnium to potentiate radiotherapy is attracting more and more attention. Here, we evaluate the potentiating effect of hollow shells made of gold and iron oxide nanoparticles (hybridosomes®) on the radiotherapy of glioblastoma, using murine GL261-Luc+ brain tumor model. While iron oxide seems to have no beneficial effect for radiotherapy, we observe a real effect of gold nanoparticles-despite their low amount-with a median survival increase of almost 20% compared to radiotherapy only and even 33% compared to the control group. Cellular and in vivo studies show that a molecule of interest nano-precipitated in the core of the hybridosomes® is released and internalized by the surrounding brain cells. Finally, in vivo studies show that hybridosomes® injected intra-tumorally are still present in the vicinity of the brain tumor more than 5 days after injection (duration of the Stupp protocol's radiation treatment). Interestingly, one mouse treated with radiotherapy in the presence of gold-containing hybridosomes® survived 78 days. Monitoring of the tumoral growth of this long-term survivor using both MRI and bioluminescence revealed a decrease of the tumor size after treatment. These very encouraging results are a proof-of-concept that hybridosomes® are really effective tools for the development of combined therapies (chemo-radiotherapy).


Subject(s)
Brain Neoplasms , Glioblastoma , Metal Nanoparticles , Nanocapsules , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Gold/therapeutic use , Metal Nanoparticles/therapeutic use , Mice , Nanocapsules/therapeutic use
6.
BMC Cancer ; 21(1): 530, 2021 May 10.
Article in English | MEDLINE | ID: mdl-33971844

ABSTRACT

BACKGROUND: Osimertinib is a third generation tyrosine kinase inhibitor (TKI) that targets the epidermal growth factor receptor (EGFR) in lung cancer. However, although this molecule is not subject to some of the resistance mechanisms observed in response to first generation TKIs, ultimately, patients relapse because of unknown resistance mechanisms. New relevant non-small cell lung cancer (NSCLC) mice models are therefore required to allow the analysis of these resistance mechanisms and to evaluate the efficacy of new therapeutic strategies. METHODS: Briefly, PC-9 cells, previously modified for luciferase expression, were injected into the tail vein of mice. Tumor implantation and longitudinal growth, almost exclusively localized in the lung, were evaluated by bioluminescence. Once established, the tumor was treated with osimertinib until tumor escape and development of bone metastases. RESULTS: Micro-metastases were detected by bioluminescence and collected for further analysis. CONCLUSION: We describe an orthotopic model of NSCLC protocol that led to lung primary tumor nesting and, after osimertinib treatment, by metastases dissemination, and that allow the isolation of these small osimertinib-resistant micro-metastases. This model provides new biological tools to study tumor progression from the establishment of a lung tumor to the generation of drug-resistant micro-metastases, mimicking the natural course of the disease in human NSCLC patients.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Bone Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Neoplasm Micrometastasis , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , ErbB Receptors/genetics , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mutation , Xenograft Model Antitumor Assays
7.
Oncogene ; 40(24): 4111-4123, 2021 06.
Article in English | MEDLINE | ID: mdl-34017078

ABSTRACT

ING2 (Inhibitor of Growth 2) is a tumor suppressor gene that has been implicated in critical biological functions (cell-cycle regulation, replicative senescence, DNA repair and DNA replication), most of which are recognized hallmarks of tumorigenesis occurring in the cell nucleus. As its close homolog ING1 has been recently observed in the mitochondrial compartment, we hypothesized that ING2 could also translocate into the mitochondria and be involved in new biological functions. In the present study, we demonstrate that ING2 is imported in the inner mitochondrial fraction in a redox-sensitive manner in human cells and that this mechanism is modulated by 14-3-3η protein expression. Remarkably, ING2 is necessary to maintain mitochondrial ultrastructure integrity without interfering with mitochondrial networks or polarization. We observed an interaction between ING2 and mtDNA under basal conditions. This interaction appears to be mediated by TFAM, a critical regulator of mtDNA integrity. The loss of mitochondrial ING2 does not impair mtDNA repair, replication or transcription but leads to a decrease in mitochondrial ROS production, suggesting a detrimental impact on OXPHOS activity. We finally show using multiple models that ING2 is involved in mitochondrial respiration and that its loss confers a protection against mitochondrial respiratory chain inhibition in vitro. Consequently, we propose a new tumor suppressor role for ING2 protein in the mitochondria as a metabolic shift gatekeeper during tumorigenesis.


Subject(s)
Homeodomain Proteins/genetics , Homeostasis/genetics , Mitochondria/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Tumor Suppressor Proteins/genetics , A549 Cells , Cell Line, Tumor , DNA Repair/genetics , DNA Replication/genetics , DNA, Mitochondrial/genetics , DNA-Binding Proteins/genetics , Humans , Transcription, Genetic/genetics
9.
Cancers (Basel) ; 13(7)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33916038

ABSTRACT

Liquid biopsies are increasingly used for the diagnosis and follow-up of cancer patients. Urine is a body fluid that can be used to detect cancers and others diseases. It is noninvasive and easy to collect. To detect Bladder Cancer (BC), cytology is the first assay used. It is an effective way to detect high grade BC but has a high rate of equivocal results, especially for low grade BC. Furthermore, cystoscopy is used to confirm cytology results and to determine cancer status. Cystoscopy is also effective but highly invasive, and not well accepted by patients, especially for BC follow-up. In this review we survey the numerous assays recently developed in order to diagnose BC at an early stage, and to facilitate the follow-up of patients. We discuss their effectiveness, ease of use, and applications. Finally, we discuss assays that, in the future, could improve the diagnosis and management of BC patients.

10.
Cell Death Differ ; 26(11): 2344-2357, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30804473

ABSTRACT

Inhibitor of Growth 3 (ING3) is a candidate tumor suppressor gene whose expression is lost in tumors such as hepatocellular carcinoma, head and neck squamous cell carcinoma and melanoma. In the present study, we show that ING3-depleted human cells and yeast cells deleted for its ortholog YNG2 are sensitive to DNA damage suggesting a conserved role in response to such stress. In human cells, ING3 is recruited to DNA double strand breaks and is required for ATM activation. Remarkably, in response to doxorubicin, ATM activation is dependent on ING3 but not on TIP60, whose recruitment to DNA breaks also depends on ING3. These events lead to ATM-mediated phosphorylation of NBS1 and the subsequent recruitment of RNF8, RNF168, 53BP1, and BRCA1, which are major mediators of the DNA damage response. Accordingly, upon genotoxic stress, DNA repair by non-homologous end joining (NHEJ) or homologous recombination (HR) were impaired in absence of ING3. Finally, immunoglobulin class switch recombination (CSR), a physiological mechanism requiring NHEJ repair, was impaired in the absence of ING3. Since deregulation of DNA double strand break repair is associated with genomic instability, we propose a novel function of ING3 as a caretaker tumor suppressor involved in the DNA damage signaling and repair.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA End-Joining Repair/genetics , Genomic Instability/genetics , Homeodomain Proteins/metabolism , Tumor Suppressor Proteins/genetics , A549 Cells , Acetyltransferases/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , BRCA1 Protein/metabolism , Cell Line, Tumor , DNA/genetics , DNA/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Doxorubicin/pharmacology , Enzyme Activation/genetics , Homeodomain Proteins/genetics , Humans , Immunoglobulin Class Switching/genetics , Lysine Acetyltransferase 5/genetics , Mice , RNA Interference , RNA, Small Interfering/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin-Protein Ligases/metabolism
11.
Development ; 145(12)2018 06 21.
Article in English | MEDLINE | ID: mdl-29802150

ABSTRACT

Using electron microscopy to localize rare cellular events or structures in complex tissue is challenging. Correlative light and electron microscopy procedures have been developed to link fluorescent protein expression with ultrastructural resolution. Here, we present an optimized scanning electron microscopy (SEM) workflow for volumetric array tomography for asymmetric samples and model organisms (Caenorhabditis elegans, Drosophila melanogaster, Danio rerio). We modified a diamond knife to simplify serial section array acquisition with minimal artifacts. After array acquisition, the arrays were transferred to a glass coverslip or silicon wafer support. Using light microscopy, the arrays were screened rapidly for initial recognition of global anatomical features (organs or body traits). Then, using SEM, an in-depth study of the cells and/or organs of interest was performed. Our manual and automatic data acquisition strategies make 3D data acquisition and correlation simpler and more precise than alternative methods. This method can be used to address questions in cell and developmental biology that require the efficient identification of a labeled cell or organelle.


Subject(s)
Imaging, Three-Dimensional , Microscopy, Electron, Scanning , Tomography , Animals , Caenorhabditis elegans/cytology , Drosophila melanogaster/cytology , Drosophila melanogaster/ultrastructure , Microscopy, Fluorescence , Models, Biological
12.
Cancer Res ; 77(21): 5721-5727, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28883003

ABSTRACT

Canine cancers represent a tremendous natural resource due to their incidence and striking similarities to human cancers, sharing similar clinical and pathologic features as well as oncogenic events, including identical somatic mutations. Considering the importance of gene fusions as driver alterations, we explored their relevance in canine cancers. We focused on three distinct human-comparable canine cancers representing different tissues and embryonic origins. Through RNA-Seq, we discovered similar gene fusions as those found in their human counterparts: IGK-CCND3 in B-cell lymphoma, MPB-BRAF in glioma, and COL3A1-PDGFB in dermatofibrosarcoma protuberans-like. We showed not only similar partner genes but also identical breakpoints leading to oncogene overexpression. This study demonstrates similar gene fusion partners and mechanisms in human-dog corresponding tumors and allows for selection of targeted therapies in preclinical and clinical trials with pet dogs prior to human trials, within the framework of personalized medicine. Cancer Res; 77(21); 5721-7. ©2017 AACR.


Subject(s)
Dog Diseases/genetics , Neoplasms/genetics , Neoplasms/veterinary , Oncogene Proteins, Fusion/genetics , Animals , Base Sequence , Blotting, Western , Chromosome Breakpoints , Dog Diseases/metabolism , Dogs , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Glioma/veterinary , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/veterinary , Neoplasms/metabolism , Oncogene Fusion , Oncogene Proteins, Fusion/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Translocation, Genetic
13.
Bioorg Med Chem ; 25(6): 1817-1829, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28214231

ABSTRACT

22 Flavokawain derivatives (FKd) were obtained by one step syntheses in order to conduct a SAR study to understand the structural requirements for optimum and selective cytotoxicity. FKd and natural flavokawains A and B found into kava, a South Pacific traditional beverage, were evaluated against nine cancer and one healthy cell lines. The targeted cell cycle phases as well as the effects on the induction of apoptosis and cell cycle protein levels were investigated. Therapeutic improvements (more activity and selectivity) were achieved with FKd compared to natural flavokawains and notably with the 2',3,4',6'-tetramethoxychalcone (FKd 19). FKd induced a G1/S arrest on p53 wild-type cells and an M arrest on p53 mutant-type, via the up-regulation of p21 and cyclin B1 proteins, followed by apoptosis. Moreover, FKd exhibited a 24h-effect on Akt/mTor normal cells versus a 48h-effect on Akt/mTor up-regulated cells. The SAR study resulted in the conclusion that trimethoxy A-ring allowed the best compromise between cytotoxicity and selectivity, as well as the substitution of the meta position on the B-ring and the use of halogens substituents.


Subject(s)
Chalcone/analogs & derivatives , Flavonoids/pharmacology , Carbon-13 Magnetic Resonance Spectroscopy , Cell Line , Cell Line, Tumor , Chalcone/chemistry , Chalcone/pharmacology , Drug Screening Assays, Antitumor , Flavonoids/chemistry , Humans , Proton Magnetic Resonance Spectroscopy , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship
14.
Cancer Lett ; 378(2): 150-9, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27224890

ABSTRACT

Apoptosis is a gene-directed program that is engaged to efficiently eliminate dysfunctional cells. Evasion of apoptosis may be an important gate to tumor initiation and therapy resistance. Like any other developmental program, apoptosis can be disrupted by several genetic aberrations driving malignant cells into an uncontrolled progression and survival. For its sustained growth, cancer develops in a complex environment, which provides survival signals and rescues malignant cells from apoptosis. Recent studies have clearly shown a wide interaction between tumor cells and their microenvironment, confirming the influence of the surrounding cells on tumor expansion and invasion. These non-malignant cells not only intensify tumor cells growth but also upgrade the process of metastasis. The strong crosstalk between malignant cells and a reactive microenvironment is mediated by soluble chemokines and cytokines, which act on tumor cells through surface receptors. Disturbing the microenvironment signaling might be an encouraging approach for patient's treatment. Therefore, the ultimate knowledge of "tumor-microenvironment" interactions facilitates the identification of novel therapeutic procedures that mobilize cancer cells from their supportive cells. This review focuses on cancer progression mediated by the dysfunction of apoptosis and by the fundamental relationship between tumor and reactive cells. New insights and valuable targets for cancer prevention and therapy are also presented.


Subject(s)
Apoptosis , Neoplasms/pathology , Tumor Microenvironment , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Proliferation , Disease Progression , Humans , Inflammation Mediators/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Tumor Escape
15.
Oncoimmunology ; 4(8): e1026503, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26405582

ABSTRACT

Non-Hodgkin's lymphomas (NHLs) are malignant neoplasms which are clinically and biologically diverse. Their incidence is constantly increasing and despite treatment advances, there is a need for novel targeted therapies. Here, we identified Lectin-like transcript 1 (LLT1) as a biomarker of germinal center (GC)-derived B-cell NHLs. LLT1 identifies GC B cells in reactive tonsils and lymph nodes and its expression is maintained in B-cell NHLs which derive from GC, including Burkitt lymphoma (BL), follicular lymphoma (FL), and GC-derived diffuse large B-cell lymphoma (DLBCL). We further show that LLT1 expression by tumors dampens natural killer (NK) cell functions following interaction with its receptor CD161, uncovering a potential immune escape mechanism. Our results pinpoint LLT1 as a novel biomarker of GC-derived B-cell NHLs and as a candidate target for innovative immunotherapies.

16.
Clin Cancer Res ; 20(10): 2663-73, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24637636

ABSTRACT

PURPOSE: Despite therapeutic advances, non-Hodgkin lymphomas (NHL) remain incurable. They form a group of neoplasms strongly dependent on their inflammatory microenvironment, which plays an important supportive role in tumor B-cell survival and in the resistance to antitumor immune response. New therapies must consider both tumor cells and their surrounding microenvironment EXPERIMENTAL DESIGN: Stromal cells, derived from bone marrow or lymph nodes, and B cells from follicular lymphoma patients were cocultured or cultured alone with celecoxib treatment, a nonsteroidal anti-inflammatory drug, and/or TRAIL, a promising cytotoxic molecule for cancer therapy. RESULTS: In this study, we show that follicular lymphoma stromal cells produce large amounts of PGE2. This production is abrogated after celecoxib treatment, targeting the COX-2 isoenzyme involved in PGE2 synthesis. Furthermore, we demonstrate that celecoxib increases apoptosis in NHL B-cell lines and in primary follicular lymphoma B cells cocultured with stromal cells, but independently of the PGE2/COX-2 axis. Finally, celecoxib increases the apoptotic activity of TRAIL. We provide evidence that celecoxib affects proliferation and sensitizes NHL B-cell lines to apoptosis through COX-2-independent effects by slowing down the cell cycle and decreasing the expression of survival proteins, such as Mcl-1. CONCLUSIONS: These data suggest new potent strategies for NHL therapy combining drugs targeting both tumor B cells and survival signals provided by the tumor microenvironment.


Subject(s)
Apoptosis/drug effects , Cyclooxygenase 2/metabolism , Pyrazoles/pharmacology , Sulfonamides/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Blotting, Western , Celecoxib , Cell Cycle/drug effects , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/metabolism , Drug Synergism , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Cells, Cultured , bcl-2-Associated X Protein/metabolism
17.
Haematologica ; 97(1): 38-46, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21933852

ABSTRACT

BACKGROUND: Non-Hodgkin's B-cell lymphomas account for approximately 70% of B-cell lymphomas. While its incidence is dramatically increasing worldwide, the disease is still associated with high morbidity due to ineffectiveness of conventional therapies, creating an urgent need for novel therapeutic approaches. Unconventional compounds, including polyphenols and the cytokine TRAIL, are being extensively studied for their capacity to restore apoptosis in a large number of tumors, including lymphomas. DESIGN AND METHODS: Molecular mechanisms of TRAIL-resistance and reactivation of the apoptotic machinery by quercetin in non-Hodgkin's lymphoma cell lines were determined by Hoescht, flow cytometry, Western blot, qPCR, by use of siRNA or pharmacological inhibitors of the mitochondrial pathway and by immunoprecipitation followed by post-translational modification analysis. RESULTS: Results demonstrate that quercetin, a natural flavonoid, restores TRAIL-induced cell death in resistant transformed follicular lymphoma B-cell lines, despite high Bcl-2 expression levels due to the chromosomal translocation t(14;18). Quercetin rescues mitochondrial activation by inducing the proteasomal degradation of Mcl-1 and by inhibiting survivin expression at the mRNA level, irrespective of p53. Restoration of the TRAIL pathway requires Bax and Bak but is independent of enhanced TRAIL DISC formation. CONCLUSIONS: We demonstrate that inactivation of survivin and Mcl-1 expression by quercetin is sufficient to restore TRAIL sensitivity in resistant non-Hodgkin's lymphoma B cells. Our results suggest, therefore, that combining quercetin with TRAIL treatments may be useful in the treatment of non-Hodgkin's lymphoma.


Subject(s)
Antioxidants/pharmacology , Apoptosis , Inhibitor of Apoptosis Proteins/metabolism , Lymphoma, B-Cell/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Quercetin/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Caspase 10/metabolism , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Drug Resistance, Neoplasm/genetics , Humans , Inhibitor of Apoptosis Proteins/genetics , Lymphoma, B-Cell/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/drug effects , Survivin , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
18.
J Immunol ; 181(2): 1001-11, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606651

ABSTRACT

The TNF family member TRAIL is emerging as a promising cytotoxic molecule for antitumor therapy. However, its mechanism of action and the possible modulation of its effect by the microenvironment in follicular lymphomas (FL) remain unknown. We show here that TRAIL is cytotoxic only against FL B cells and not against normal B cells, and that DR4 is the main receptor involved in the initiation of the apoptotic cascade. However, the engagement of CD40 by its ligand, mainly expressed on a specific germinal center CD4(+) T cell subpopulation, counteracts TRAIL-induced apoptosis in FL B cells. CD40 induces a rapid RNA and protein up-regulation of c-FLIP and Bcl-x(L). The induction of these antiapoptotic molecules as well as the inhibition of TRAIL-induced apoptosis by CD40 is partially abolished when NF-kappaB activity is inhibited by a selective inhibitor, BAY 117085. Thus, the antiapoptotic signaling of CD40, which interferes with TRAIL-induced apoptosis in FL B cells, involves NF-kappaB-mediated induction of c-FLIP and Bcl-x(L) which can respectively interfere with caspase 8 activation or mitochondrial-mediated apoptosis. These findings suggest that a cotreatment with TRAIL and an inhibitor of NF-kappaB signaling or a blocking anti-CD40 Ab could be of great interest in FL therapy.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , CD40 Antigens/immunology , CD40 Ligand/metabolism , Lymphoma, Follicular/immunology , NF-kappa B/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/drug effects , B-Lymphocytes/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/immunology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD40 Ligand/immunology , Caspase 8/metabolism , Cell Line, Tumor , Cells, Cultured , Humans , Lymphoma, Follicular/metabolism , Lymphoma, Follicular/pathology , NF-kappa B/drug effects , NF-kappa B/immunology , Nitriles/pharmacology , Palatine Tonsil/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Sulfones/pharmacology , Up-Regulation , bcl-X Protein/immunology , bcl-X Protein/metabolism
19.
Blood ; 109(2): 693-702, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-16985173

ABSTRACT

Accumulating evidence indicates that the cellular microenvironment plays a key role in follicular lymphoma (FL) pathogenesis, both within tumor lymph nodes (LNs) and in infiltrated bone marrow where ectopic LN-like reticular cells are integrated within malignant B-cell nodular aggregates. In normal secondary lymphoid organs, specific stromal cell subsets provide a highly specialized microenvironment that supports immune response. In particular, fibroblastic reticular cells (FRCs) mediate immune cell migration, adhesion, and reciprocal interactions. The role of FRCs and their postulated progenitors, that is, bone marrow mesenchymal stem cells (MSCs), in FL remains unexplored. In this study, we investigated the relationships between FRCs and MSCs and their capacity to sustain malignant B-cell growth. Our findings strongly suggest that secondary lymphoid organs contain MSCs able to give rise to adipocytes, chondrocytes, osteoblasts, as well as fully functional B-cell supportive FRCs. In vitro, bone marrow-derived MSCs acquire a complete FRC phenotype in response to a combination of tumor necrosis factor-alpha and lymphotoxin-alpha1beta2. Moreover, MSCs recruit primary FL cells that, in turn, trigger their differentiation into FRCs, making them able to support malignant B-cell survival. Altogether, these new insights into the cross talk between lymphoma cells and their microenvironment could offer original therapeutic strategies.


Subject(s)
Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, Follicular/immunology , Lymphoma, Follicular/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Stromal Cells/physiology , Bone Marrow Cells/cytology , Cell Separation/methods , Cells, Cultured , Humans , Lymphoid Tissue/cytology , Palatine Tonsil/cytology , Stromal Cells/cytology
20.
Prostate ; 66(9): 987-95, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16541419

ABSTRACT

BACKGROUND: As advanced prostate cancers are resistant to currently available chemotherapies, we evaluated the cytotoxic effect of TNF-related apoptosis-inducing ligand (TRAIL) and characterized the involvement of its five receptors DR4, DR5, DcR1, DcR2, and osteoprotegerin (OPG) and of the death-inducing signaling complex (DISC)-forming proteins caspase 8 and c-FLIP in prostate cell lines. METHODS: We used six prostate cell lines, each corresponding to a particular stage in prostate tumorigenesis, and analyzed TRAIL sensitivity in relation to TRAIL receptors' expression. RESULTS: TRAIL sensitivity was correlated with tumor progression and DR5 expression levels and apoptosis was exclusively mediated by DR5. DcR2 was significantly more abundant in tumor cells than in non-neoplastic ones and may contribute to partial resistance to TRAIL in some prostate tumor cells. Conversely, non-tumoral cells secreted high levels of OPG, which can protect them from apoptosis. Finally, caspase 8 expression levels were as DR5 directly correlated to TRAIL sensitivity in prostate tumor cells. CONCLUSION: TRAIL-induced apoptosis is closely related to the balanced expression of its different receptors in prostate cancer cells and their modulation could be of potential clinical value for advanced tumor treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Apoptosis/drug effects , Caspases/physiology , Membrane Glycoproteins/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/pharmacology , Androgens/pharmacology , Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 8 , Caspases/genetics , Cell Line , Cell Line, Tumor , Death Domain Receptor Signaling Adaptor Proteins , Disease Progression , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Glycoproteins/physiology , Humans , Intracellular Signaling Peptides and Proteins/analysis , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Male , Neoplasm Staging , Osteoprotegerin , Prostate/chemistry , Prostate/drug effects , Prostate/physiology , Prostatic Neoplasms/physiopathology , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor Decoy Receptors , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/analysis , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...