Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 6881, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32327691

ABSTRACT

Triphenylamines (TPAs) were previously shown to trigger cell death under prolonged one- or two-photon illumination. Their initial subcellular localization, before prolonged illumination, is exclusively cytoplasmic and they translocate to the nucleus upon photoactivation. However, depending on their structure, they display significant differences in terms of precise initial localization and subsequent photoinduced cell death mechanism. Here, we investigated the structural features of TPAs that influence cell death by studying a series of molecules differing by the number and chemical nature of vinyl branches. All compounds triggered cell death upon one-photon excitation, however to different extents, the nature of the electron acceptor group being determinant for the overall cell death efficiency. Photobleaching susceptibility was also an important parameter for discriminating efficient/inefficient compounds in two-photon experiments. Furthermore, the number of branches, but not their chemical nature, was crucial for determining the cellular uptake mechanism of TPAs and their intracellular fate. The uptake of all TPAs is an active endocytic process but two- and three-branch compounds are taken up via distinct endocytosis pathways, clathrin-dependent or -independent (predominantly caveolae-dependent), respectively. Two-branch TPAs preferentially target mitochondria and photoinduce both apoptosis and a proper necrotic process, whereas three-branch TPAs preferentially target late endosomes and photoinduce apoptosis only.


Subject(s)
Amines/toxicity , Endocytosis/drug effects , Endocytosis/radiation effects , Intracellular Space/metabolism , Light , Amines/chemistry , Cell Death/drug effects , Cell Death/radiation effects , Cell Survival/drug effects , HeLa Cells , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/radiation effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Reactive Oxygen Species/metabolism , Spectrometry, Fluorescence
2.
Oncotarget ; 8(59): 99950-99965, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29245952

ABSTRACT

Müllerian inhibiting substance, also called anti-Müllerian hormone (AMH), inhibits proliferation and induces apoptosis of AMH type II receptor-positive tumor cells, such as human ovarian cancers (OCs). On this basis, a humanized glyco-engineered monoclonal antibody (3C23K) has been developed. The aim of this study was therefore to experimentally confirm the therapeutic potential of 3C23K in human OCs. We first determined by immunofluorescence, immunohistochemistry and cytofluorometry analyses the expression of AMHRII in patient's tumors and found that a majority (60 to 80% depending on the detection technique) of OCs were positive for this marker. We then provided evidence that the tumor stroma of OC is enriched in tumor-associated macrophages and that these cells are responsible for 3C23K-induced killing of tumor cells through ADCP and ADCC mechanisms. In addition, we showed that 3C23K reduced macrophages induced-T cells immunosuppression. Finally, we evaluated the therapeutic efficacy of 3C23K alone and in combination with a carboplatin-paclitaxel chemotherapy in a panel of OC Patient-Derived Xenografts. In those experiments, we showed that 3C23K significantly increased the proportion and the quality of chemotherapy-based in vivo responses. Altogether, our data support the potential interest of AMHRII targeting in human ovarian cancers and the evaluation of 3C23K in further clinical trials.

3.
J Vis Exp ; (130)2017 12 27.
Article in English | MEDLINE | ID: mdl-29364247

ABSTRACT

CD8 T cell are key players in the fight against cancer. In order for CD8 T cells to kill tumor cells they need to enter into the tumor, migrate within the tumor microenvironment and respond adequately to tumor antigens. The recent development of improved imaging approaches, such as 2-photon microscopy, and the use of powerful mouse tumor models have shed light on some of the mechanisms that regulate anti-tumor T cell activities. Whereas such systems have provided valuable insights, they do not always predict human responses. In human, our knowledge in the field mainly comes from a description of fixed tumor samples from human patients, as well as in vitro studies. However, in vitro models lack the complex three-dimensional tumor milieu and, therefore, are incomplete approximations of in vivo T cell activities. Fresh slices made from explanted tissue represent a complex multi-cellular tumor environment that can act as an important link between co-cultured studies and animal models. Originally set up in murine lymph nodes1 and previously described in a JoVE article2, this approach has now been transposed to human tumors to examine the dynamics of both plated3 as well as resident T cells4. Here, a protocol for the preparation of human lung tumor slices, immunostaining of resident CD8 T and tumor cells, and tracking of CD8 T lymphocytes within the tumor microenvironment using confocal microscopy is described. This system is uniquely placed to screen for novel immunotherapy agents favoring T cell migration in tumors.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Lung Neoplasms/diagnostic imaging , Microscopy, Confocal/methods , CD8-Positive T-Lymphocytes/immunology , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology
4.
Sci Rep ; 6: 21458, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26947258

ABSTRACT

Photodynamic therapy (PDT) leads to cell death by using a combination of a photosensitizer and an external light source for the production of lethal doses of reactive oxygen species (ROS). Since a major limitation of PDT is the poor penetration of UV-visible light in tissues, there is a strong need for organic compounds whose activation is compatible with near-infrared excitation. Triphenylamines (TPAs) are fluorescent compounds, recently shown to efficiently trigger cell death upon visible light irradiation (458 nm), however outside the so-called optical/therapeutic window. Here, we report that TPAs target cytosolic organelles of living cells, mainly mitochondria, triggering a fast apoptosis upon two-photon excitation, thanks to their large two-photon absorption cross-sections in the 760-860 nm range. Direct ROS imaging in the cell context upon multiphoton excitation of TPA and three-color flow cytometric analysis showing phosphatidylserine externalization indicate that TPA photoactivation is primarily related to the mitochondrial apoptotic pathway via ROS production, although significant differences in the time courses of cell death-related events were observed, depending on the compound. TPAs represent a new class of water-soluble organic photosensitizers compatible with direct two-photon excitation, enabling simultaneous multiphoton fluorescence imaging of cell death since a concomitant subcellular TPA re-distribution occurs in apoptotic cells.


Subject(s)
Aniline Compounds/metabolism , Apoptosis/drug effects , Mitochondria/drug effects , Optical Imaging/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/metabolism , Cell Death , Flow Cytometry , HeLa Cells , Humans , Light , Reactive Oxygen Species/analysis
5.
Front Immunol ; 6: 500, 2015.
Article in English | MEDLINE | ID: mdl-26528284

ABSTRACT

T cells play a key role in the battle against cancer. To perform their antitumor activities, T cells need to adequately respond to tumor antigens by establishing contacts with either malignant cells or antigen-presenting cells. These latter functions rely on a series of migratory steps that go from entry of T cells into the tumor followed by their locomotion in the tumor stroma. Our knowledge of how T cells migrate within tumors mainly comes from experiments performed in mouse models. Whereas such systems have greatly advanced our understanding, they do not always faithfully recapitulate the disease observed in cancer patients. We previously described a technique based on tissue slices that enables to track with real-time imaging microscopy the motile behavior of fluorescent T cells plated onto fresh sections of human lung tumors. We have now refined this approach to monitor the locomotion of resident tumor-infiltrating CD8 T cells labeled with fluorescently coupled antibodies. Using this approach, our findings reveal that CD8 T cells accumulate in the stroma of ovarian and lung carcinomas but move slowly in this compartment. Conversely, even though less populated, tumors islets were found to be zones of faster migration for resident CD8 T cells. We also confirm the key role played by collagen fibers, which, by their orientation, spacing and density, control the distribution and migration of resident CD8 T cells within the tumor stroma. We have subsequently demonstrated that, under some physical tissue constraints, CD8 T cells exhibited a mode of migration characterized by alternate forward and backward movements. In sum, using an ex vivo assay to track CD8 T cells in fresh human tumor tissues, we have identified the extracellular matrix as a major stromal component in influencing T cell migration, thereby impacting the control of tumor growth. This approach will aid in the development and testing of novel immunotherapy strategies to promote T cell migration in tumors.

6.
Cell Mol Life Sci ; 70(23): 4431-48, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23649148

ABSTRACT

The migration of T cells and access to tumor antigens is of utmost importance for the induction of protective anti-tumor immunity. Once having entered a malignant site, T cells encounter a complex environment composed of non-tumor cells along with the extracellular matrix (ECM). It is now well accepted that a deregulated ECM favors tumor progression and metastasis. Recent progress in imaging technologies has also highlighted the impact of the matrix architecture found in solid tumor on immune cells and especially T cells. In this review, we argue that the ability of T cells to mount an antitumor response is dependent on the matrix structure, more precisely on the balance between pro-migratory reticular fiber networks and unfavorable migration zones composed of dense and aligned ECM structures. Thus, the matrix architecture, that has long been considered to merely provide the structural framework of connective tissues, can play a key role in facilitating or suppressing the antitumor immune surveillance. A new challenge in cancer therapy will be to develop approaches aimed at altering the architecture of the tumor stroma, rendering it more permissive to antitumor T cells.


Subject(s)
Extracellular Matrix/immunology , Immunologic Surveillance/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Cell Movement/immunology , Disease Progression , Extracellular Matrix/metabolism , Humans , Models, Immunological , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
7.
Clin Lymphoma Myeloma Leuk ; 13(1): 62-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23127495

ABSTRACT

BACKGROUND: Systemic mastocytosis (SM) is a heterogeneous disease that displays variable aggressivity. Adults with SM frequently have a D816V mutation in the tyrosine kinase (TK) receptor gene KIT. We previously reported that, in a Chinese hamster ovarian cell model expressing exogenous KIT variants, constitutive activating KIT mutations induced intracellular mislocalization of KIT reversed by inhibition of KIT TK activity. Hence, we hypothesized that inhibition of KIT kinase activity by the TK inhibitor dasatinib could be useful to increase KIT detection sensitivity in samples from patients with SM. PATIENTS AND METHODS: We tested this hypothesis on a BaF/3 cell line modified to express either KIT wild-type (WT) or KIT D816V, on the human mastocytoma cell line HMC1.2, and among 28 patients with proven SM who did (n = 24) or did not (n = 4) carry the D816V KIT mutation and displayed various SM subtypes by using a simple flow cytometry assay to quantify KIT relocalization upon dasatinib treatment. RESULTS: We confirm KIT cell surface increase upon dasatinib treatment on BaF/3 KIT D816V and HMC1.2 cell lines but not on BaF/3 KIT WT cell line. The analysis of bone marrow and peripheral blood samples of patients with SM showed KIT surface level increase for patients with the KIT D816V mutation but not for patients who had no KIT mutation. Interestingly, the extent of KIT level relocalization correlates with SM severity, with a higher relocalization for patients with aggressive forms compared with indolent forms. CONCLUSIONS: Overall, results of this study suggests that treating the peripheral blood sample with dasatinib of a patient with SM before analysis by flow cytometry could contribute to narrowing the SM diagnosis.


Subject(s)
Cell Membrane/metabolism , Mastocytosis, Systemic/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyrimidines/therapeutic use , Thiazoles/therapeutic use , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Dasatinib , Female , Humans , Male , Mastocytosis, Systemic/blood , Mastocytosis, Systemic/enzymology , Mastocytosis, Systemic/genetics , Middle Aged , Mutation , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Young Adult
8.
Cancer Res ; 70(13): 5497-506, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20530676

ABSTRACT

The major route of iron uptake by cells occurs through transferrin receptor (TfR)-mediated endocytosis of diferric-charged plasma transferrin (holo-Tf). In this work, we pursued TfR antibodies as potential cancer therapeutics, characterizing human single-chain variable antibody fragments (scFv) specific for the human TfR isolated from a phage display library. We hypothesized that many of these antibodies would function as ligand mimetics because scFvs from the library were selected for binding and internalization into living cells. In support of this hypothesis, the anti-TfR scFvs identified were antagonists of TfR binding to holo-Tf, particularly two of the most potent antibodies, 3TF12 and 3GH7, which blocked the in vitro proliferation of a number of hematopoietic cancer cell lines. We optimized this activity of 3TF12 and 3GH7 by engineering 55-kDa bivalent antibody formats, namely, F12CH and H7CH, which could block cell proliferation with an IC(50) of 0.1 microg/mL. We found that the mechanism of the scFv antibody cytotoxicity was unique compared with cytotoxic anti-TfR monoclonal antibodies that have been described, causing cell surface upregulation of TfR along with the inhibition of holo-Tf cell uptake and induction of cell death. In a nude mouse model of erythroleukemia, administration of F12CH reduced tumor growth. Together, our findings define a new class of fully human anti-TfR antibodies suitable for immunotherapy against tumors whose proliferation relies on high levels of TfR and iron uptake, such as acute lymphoid and myeloid leukemias.


Subject(s)
Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/pharmacology , Leukemia/therapy , Lymphoma/therapy , Receptors, Transferrin/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Growth Processes , Female , Ferric Compounds/metabolism , Humans , Leukemia/immunology , Leukemia/pathology , Leukemia, Erythroblastic, Acute/immunology , Leukemia, Erythroblastic, Acute/therapy , Lymphoma/immunology , Lymphoma/pathology , Mice , Mice, Nude , Receptors, Transferrin/metabolism , Transferrin/immunology , Transferrin/metabolism , Xenograft Model Antitumor Assays
9.
Mol Cancer Res ; 7(9): 1525-33, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19737976

ABSTRACT

Kit is a cell surface type III tyrosine kinase (TK) receptor implicated in cell transformation through overexpression or oncogenic mutation. Two categories of Kit mutants displaying mutations either in the juxtamembrane intracellular domain (regulatory mutants) or in the catalytic domain (catalytic mutants) have been described. To explore the effect of Kit oncogenic mutations on its subcellular localization, we constructed enhanced green fluorescent protein (EGFP)-tagged human Kit chimeras harboring mutations either in the regulatory (V560G) or in the catalytic (D816V) domain. When expressed in Chinese hamster ovary cells, EGFP-tagged wild-type Kit was activated on stem cell factor stimulation, whereas both EGFP-tagged Kit mutants displayed a constitutive TK activity. Constitutively activated mutants exhibited a high-mannose-type N-glycosylation pattern and an intracellular localization, suggesting that these mutants induce downstream oncogenic signaling without the need to reach the cell surface. Inhibition of constitutive Kit TK activity with dasatinib induced a complex, mature N-glycosylation pattern identical to unstimulated wild-type Kit and resulted in the redistribution of the mutants to the plasma membrane. This relocalization was clearly correlated to the inhibition of TK activity because imatinib, a specific inhibitor of the V560G mutant, inactive on the catalytic D816V mutant, induced only the relocalization of the V560G mutant. These data show that on TK inhibition, the aberrant localization of Kit mutants can be fully reversed. Kit mutants are then exported and/or stabilized at the cell surface as inactive and fully N-glycosylated isoforms.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Animals , Benzamides , CHO Cells , Cloning, Molecular , Cricetinae , Cricetulus , Dasatinib , Flow Cytometry , Glycosylation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Imatinib Mesylate , Immunohistochemistry , Intracellular Space/metabolism , Mutation , Piperazines/pharmacology , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyrimidines/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Thiazoles/pharmacology
10.
Mol Immunol ; 44(15): 3777-88, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17498801

ABSTRACT

To generate a panel of antibodies binding human breast cancers, a human single chain Fv phage display library was selected for rapid internalization into the SK-BR-3 breast cancer cell line. Thirteen unique antibodies were identified within the 55 cell binding antibodies studied, all of them showing specific staining of tumor cells compare to normal epithelial cells. Two of the antibodies bound the ErbB2 oncogene while 6 bound the tumor marker transferrin receptor (TfR). By developing a scFv immunoprecipitation method, we were able to use LC-MS/MS to identify the antigen bound by one of the antibodies (3GA5) as FPRP (prostaglandin F2alpha receptor-regulatory protein)/EWI-F/CD9P-1 (CD9 partner 1) an Ig superfamily member that has been described to interact directly with CD9 and CD81 tetraspanins and to be overexpressed in adherent cancer cell lines. Although the 3GA5 scFv had no direct anti-proliferative effect, intracellular expression of the scFv was able to knockdown CD9P-1 expression and could be used to further define the role of the tetraspanin system in proliferation and metastasis. Moreover, the 3GA5 scFv was rapidly internalized into breast tumor cells and could have potential for the targeted delivery of cytotoxic agents to breast cancers. This study is the proof of principle that the direct selection of phage antibody libraries on tumor cells can effectively lead to the identification and functional characterization of relevant tumor markers.


Subject(s)
Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/isolation & purification , Peptide Library , Amino Acid Sequence , Animals , Antibodies, Neoplasm/genetics , Antibody Specificity/immunology , Antigens, Neoplasm/chemistry , CHO Cells , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Chromatography, Liquid , Cricetinae , Cricetulus , Down-Regulation , Endocytosis , Epitopes/immunology , Humans , Immunoglobulin Variable Region/immunology , Mass Spectrometry , Molecular Sequence Data , Neoplasm Proteins/metabolism , Phenotype , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...