Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Pharmaceutics ; 14(4)2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35456626

ABSTRACT

Daunorubicin pharmacokinetics (PK) are characterised by an important inter-individual variability, which raises questions about the optimal dose regimen in patients with acute myeloid leukaemia. The aim of the study is to assess the joint daunorubicin/daunorubicinol PK profile and to define an optimal population PK study design. Fourteen patients were enrolled in the PK ancillary study of the BIG-1 trial and 6-8 samples were taken up to 24 h after administration of the first dose of daunorubicin (90 mg/m2/day). Daunorubicin and daunorubicinol quantifications were assessed using a validated liquid chromatography technique coupled with a fluorescence detector method. Data were analysed using a non-compartmental approach and non-linear mixed effects modelling. Optimal sampling strategy was proposed using the R function PFIM. The median daunorubicin and daunorubicinol AUC0-tlast were 577 ng/mL·hr (Range: 375-1167) and 2200 ng/mL·hr (range: 933-4683), respectively. The median metabolic ratio was 0.32 (range: 0.1-0.44). Daunorubicin PK was best described by a three-compartment parent, two-compartment metabolite model, with a double first-order transformation of daunorubicin to metabolite. Body surface area and plasma creatinine had a significant impact on the daunorubicin and daunorubicinol PK. A practical optimal population design has been derived from this model with five sampling times per subject (0.5, 0.75, 2, 9, 24 h) and this can be used for a future population PK study.

2.
Int J Mol Sci ; 21(22)2020 Nov 14.
Article in English | MEDLINE | ID: mdl-33202543

ABSTRACT

The bone marrow (BM) microenvironment plays a crucial role in the development and progression of leukemia (AML). Intracellular reactive oxygen species (ROS) are involved in the regulation of the biology of leukemia-initiating cells, where the antioxidant enzyme GPx-3 could be involved as a determinant of cellular self-renewal. Little is known however about the role of the microenvironment in the control of the oxidative metabolism of AML cells. In the present study, a coculture model of BM mesenchymal stromal cells (MSCs) and AML cells (KG1a cell-line and primary BM blasts) was used to explore this metabolic pathway. MSC-contact, rather than culture with MSC-conditioned medium, decreases ROS levels and inhibits the Nrf-2 pathway through overexpression of GPx3 in AML cells. The decrease of ROS levels also inactivates p38MAPK and reduces the proliferation of AML cells. Conversely, contact with AML cells modifies MSCs in that they display an increased oxidative stress and Nrf-2 activation, together with a concomitant lowered expression of GPx-3. Altogether, these experiments suggest that a reciprocal control of oxidative metabolism is initiated by direct cell-cell contact between MSCs and AML cells. GPx-3 expression appears to play a crucial role in this cross-talk and could be involved in the regulation of leukemogenesis.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glutathione Peroxidase/biosynthesis , Leukemia, Myeloid, Acute/enzymology , Neoplasm Proteins/biosynthesis , Tumor Microenvironment , Cell Line, Tumor , Coculture Techniques , Humans , Leukemia, Myeloid, Acute/pathology , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/pathology , Oxidation-Reduction
3.
Cancer Chemother Pharmacol ; 86(5): 687-691, 2020 11.
Article in English | MEDLINE | ID: mdl-32990804

ABSTRACT

PURPOSE: Cytarabine, a key chemotherapy agent for acute myeloid leukemia (AML) treatment, is deaminated into inactive uracil-arabinoside by cytidine deaminase. This deamination leads to samples stability issues with respect to clinical pharmacokinetic trials. The aim of our study was to study in vitro cytarabine stability in blood samples obtained from AML patients. METHODS: Cytarabine quantification was performed using a fully validated LC/MS/MS method. In vitro cytarabine stability was assessed at room temperature over 24 h in samples coming from 14 AML patients and 7 control patients (CTRL) with no hematological malignancy. In vitro concentrations versus time data were analyzed using a noncompartmental approach. RESULTS: Cytarabine in vitro area under the curve (AUCIVlast) was 22-fold higher in AML samples as compared to CTRL samples (AML mean (standard deviation (SD)), 51,829 (27,004) h ng/mL; CTRL mean (SD), 2356 (1250) h ng/mL, p = 0.00019). This increase was associated with a prolonged in vitro degradation half-life (t1/2IVdeg AML mean (SD), 15 (11.8) h; CTRL mean (SD), 0.36 (0.37) h, p = 0.0033). Multiple linear regression analysis showed that AML diagnosis significantly influenced t1/2IVdeg and AUCIVlas relationship. CONCLUSION: Cytarabine stability is higher in AML than in CTRL samples. The absence of correlation between t1/2IVdeg and AUCIVlast in AML samples suggests that in vitro cytarabine degradation in AML is complex. These results open perspectives including the evaluation of the clinical relevance and the involved molecular mechanisms.


Subject(s)
Antimetabolites, Antineoplastic/blood , Cytarabine/blood , Cytidine Deaminase/metabolism , Leukemia, Myeloid, Acute/drug therapy , Adolescent , Adult , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/isolation & purification , Case-Control Studies , Chromatography, High Pressure Liquid , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Cytarabine/administration & dosage , Cytarabine/chemistry , Cytarabine/isolation & purification , Cytidine Deaminase/isolation & purification , Deamination , Drug Stability , Female , Half-Life , Humans , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Randomized Controlled Trials as Topic , Specimen Handling , Tandem Mass Spectrometry , Time Factors , Young Adult
4.
Mol Diagn Ther ; 24(4): 443-449, 2020 08.
Article in English | MEDLINE | ID: mdl-32524539

ABSTRACT

BACKGROUND: The incidence of pancreatic ductal adenocarcinoma (PDAC) is increasing sharply. The survival of patients with metastases is usually about a year. However, the occurrence of isolated lung metastases after resection of the primary tumor, although rare, seems to indicate a better prognosis, with an average survival ranging from 40 to 80 months. KRAS, TP53, CDK2NA, and SMAD4 are the most common driver genes in pancreatic adenocarcinoma. OBJECTIVE: Our objectives were to determine whether a link exists between survival and mutations of driver genes in patients with isolated pulmonary metastases. METHODS: All patients who underwent curative surgery in our institution between 2010 and 2018 were included in the study. From these, we identified patients for whom recurrence was only pulmonary and those with metastases at other sites. KRAS, TP53, CDK2NA, and SMAD4 were analyzed on the primary tumor of patients with pulmonary metastases. RESULTS: Among 233 patients diagnosed with PDAC in our institution over 8 years, 41 (17.5%) underwent curative surgery. Of these, seven (3%) developed isolated pulmonary metastases, 32 developed other metastases, and two did not recur. Median survival was 59 months for patients with isolated lung metastases and 25.3 months for patients with metastases at other sites. An absence of mutations of two driver genes in primary tumors (CDK2NA and SMAD4) was observed in patients with isolated pulmonary metastases. CONCLUSIONS: The absence of mutations in the CDK2NA and SMAD4 tumor-suppressor genes in patients with isolated pulmonary metastases contrasts with the commonly observed high rates of driver gene mutations and suggests a link with overall survival.


Subject(s)
Adenocarcinoma/genetics , Lung Neoplasms/genetics , Mutation/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Female , Follow-Up Studies , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Pancreatic Neoplasms/pathology , Prognosis , Retrospective Studies , Smad4 Protein/genetics
5.
Sci Rep ; 10(1): 5900, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32246006

ABSTRACT

Peripheral CD4+CD8+ double positive (DP) T cells are a phenotypically and functionally heterogeneous population depending on their origin and pathologic context. We previously identified among tumour infiltrating lymphocytes in melanoma, a tumour-reactive MHC class-I restricted CD4lowCD8high DP αß T-cell subpopulation with CD4-like function. In this study, we used an in-depth comparative transriptomic analysis of intra-melanoma DP T cells and CD4 and CD8 single positive (SP) T cells, to better comprehend the origin of this DP phenotype, and define the transcriptomic signature of activated DP T cells. We observed that intra-melanoma DP T cells were transcriptome-wise closer to their CD8 SP T-cell counterparts in terms of number of genes differentially expressed (97 in common with CD8 SP T cells and 15 with CD4 SP T cells) but presented hallmarks of a transition to a CD4-like functional profile (CD40LG) with a decreased cytotoxic signature (KLRC1) in favour of an increased cytokine-receptor interaction signature (IL4, IL24, IL17A…). This unleashed CD4-like program could be the results of the observed unbalanced expression of the THPOK/Runx3 transcription factors in DP T cells. Overall, this study allow us to speculate that intra-melanoma DP T cells arise from CD8 SP T cells being reprogrammed to a helper function.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Skin Neoplasms/immunology , T-Lymphocyte Subsets/immunology , Transcriptome/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Plasticity/genetics , Cell Plasticity/immunology , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Core Binding Factor Alpha 3 Subunit/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/immunology , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma/genetics , Melanoma/secondary , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/pathology , T-Lymphocyte Subsets/metabolism , Transcription Factors/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
7.
Front Genet ; 10: 1292, 2019.
Article in English | MEDLINE | ID: mdl-31998361

ABSTRACT

Intrauterine Growth Restriction (IUGR) affects 8% of newborns and increases morbidity and mortality for the offspring even during later stages of life. Single omics studies have evidenced epigenetic, genetic, and metabolic alterations in IUGR, but pathogenic mechanisms as a whole are not being fully understood. An in-depth strategy combining methylomics and transcriptomics analyses was performed on 36 placenta samples in a case-control study. Data-mining algorithms were used to combine the analysis of more than 1,200 genes found to be significantly expressed and/or methylated. We used an automated text-mining approach, using the bulk textual gene annotations of the discriminant genes. Machine learning models were then used to explore the phenotypic subgroups (premature birth, birth weight, and head circumference) associated with IUGR. Gene annotation clustering highlighted the alteration of cell signaling and proliferation, cytoskeleton and cellular structures, oxidative stress, protein turnover, muscle development, energy, and lipid metabolism with insulin resistance. Machine learning models showed a high capacity for predicting the sub-phenotypes associated with IUGR, allowing a better description of the IUGR pathophysiology as well as key genes involved.

8.
Oncotarget ; 9(89): 36017-36028, 2018 Nov 13.
Article in English | MEDLINE | ID: mdl-30542515

ABSTRACT

Glioblastoma, the most frequent and lethal form of glioma, displays chromosome instability and recurrent somatic copy number alterations (SCNA). Chromothripsis and whole genome duplication (WGD) have been recently identified in cancer. In the present study, we analyzed SCNA and determine the ploidy pattern in 123 IDH-wild-type glioblastomas, using SNP array data. WGD and chromothripsis events were validated using, respectively, FISH and CTLPScanner. WGD was detected in 11.4% glioblastomas (14/123) and was associated with TP53 mutation (p = 0.0068). It was an early event occurring after the recurrent SCNA observed in diffuse high-grade gliomas. Glioblastomas with WGD were more aneuploid compared to glioblastomas without WGD (p < 0.0001). Chromothripsis occurred in 29.3% glioblastomas (36/123) and mostly affected chromosomes 7, 9 and 12, with amplification of oncogenes (EGFR, MDM2/CDK4), and homozygous deletion of tumor suppressor genes (CDKN2A). There was a significant association between chromothripsis and gene rearrangement at a given locus. WGD is an early genetic event significantly associated to TP53 mutation and leading to chromosome instability and aneuploidy in IDH-wild-type glioblastoma. Chromothripsis recurrently targets oncogenes and tumor suppressor genes that are key players in gliomagenesis and tumor progression. The occurrence of chromothripsis points to underlying gene rearrangements (including gene fusions), potential therapeutic targets in glioblastoma.

9.
Eur J Haematol ; 101(4): 570-574, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30007088

ABSTRACT

Donor cell leukemia (DCL) is an infrequent complication after allogeneic hematopoietic stem cell transplantation (HSCT). Its true incidence is difficult to assess, although improvements in chimerism studies contributed to a better diagnosis of DCL. We report two rare cases of donor cell-derived acute promyelocytic leukemia (APL). To our knowledge, only two cases have been described in the literature. Here, we report one male and one female patients with acute myeloid leukemia (AML), who developed an APL in donor cells after HSCT. The latency between HSCT and DCL was 279 and 43 months, respectively. Fluorescent in situ hybridation and chimerism monitoring analysis proved the donor origin of APL. Surprisingly, donor lymphocyte infusion provided a hematological response during 19 months in the female patient. The mechanisms associated with pathogenesis of DCL are unclear and seem to be multifactorial. Increasing worldwide allogeneic hematopoietic stem cell transplantation activity and potentially the age of donor could explain the increasing incidence of DCL in the future. It is highlighted that long-term follow up of recipients will allow to report all cases of DCL, to clarify the genetic landscape and factors which contribute to DCL, to understand the response to DLI.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Leukemia, Promyelocytic, Acute/diagnosis , Leukemia, Promyelocytic, Acute/etiology , Tissue Donors , Adult , Biomarkers , Biopsy , Bone Marrow/pathology , Female , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Middle Aged , Transplantation Conditioning/adverse effects , Transplantation Conditioning/methods , Transplantation, Homologous
10.
PLoS One ; 13(4): e0196021, 2018.
Article in English | MEDLINE | ID: mdl-29677202

ABSTRACT

Osteoblast differentiation is a highly regulated process that requires coordinated information from both soluble factors and the extracellular matrix. Among these extracellular stimuli, chemical and physical properties of the matrix are sensed through cell surface receptors such as integrins and transmitted into the nucleus to drive specific gene expression. Here, we showed that the conditional deletion of ß1 integrins in the osteo-precursor population severely impacts bone formation and homeostasis both in vivo and in vitro. Mutant mice displayed a severe bone deficit characterized by bone fragility and reduced bone mass. We showed that ß1 integrins are required for proper BMP2 dependent signaling at the pre-osteoblastic stage, by positively modulating Smad1/5-dependent transcriptional activity at the nuclear level. The lack of ß1 integrins results in a transcription modulation that relies on a cooperative defect with other transcription factors rather than a plain blunted BMP2 response. Our results point to a nuclear modulation of Smad1/5 transcriptional activity by ß1 integrins, allowing a tight control of osteoblast differentiation.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Integrin beta1/genetics , Osteoblasts/cytology , Osteogenesis , Smad1 Protein/genetics , Smad5 Protein/genetics , Animals , Cell Differentiation , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Gene Expression Regulation , Gene Knockout Techniques , Homeostasis , Mice , Osteoblasts/metabolism , Signal Transduction , Transcription, Genetic
12.
J Biol Chem ; 292(47): 19179-19197, 2017 11 24.
Article in English | MEDLINE | ID: mdl-28972170

ABSTRACT

Cell adhesion to the extracellular matrix or to surrounding cells plays a key role in cell proliferation and differentiation and is critical for proper tissue homeostasis. An important pathway in adhesion-dependent cell proliferation is the Hippo signaling cascade, which is coregulated by the transcription factors Yes-associated protein 1 (YAP1) and transcriptional coactivator with PDZ-binding motif (TAZ). However, how cells integrate extracellular information at the molecular level to regulate YAP1's nuclear localization is still puzzling. Herein, we investigated the role of ß1 integrins in regulating this process. We found that ß1 integrin-dependent cell adhesion is critical for supporting cell proliferation in mesenchymal cells both in vivo and in vitro ß1 integrin-dependent cell adhesion relied on the relocation of YAP1 to the nucleus after the down-regulation of its phosphorylated state mediated by large tumor suppressor gene 1 and 2 (LATS1/2). We also found that this phenotype relies on ß1 integrin-dependent local activation of the small GTPase RAC1 at the plasma membrane to control the activity of P21 (RAC1)-activated kinase (PAK) of group 1. We further report that the regulatory protein merlin (neurofibromin 2, NF2) interacts with both YAP1 and LATS1/2 via its C-terminal moiety and FERM domain, respectively. PAK1-mediated merlin phosphorylation on Ser-518 reduced merlin's interactions with both LATS1/2 and YAP1, resulting in YAP1 dephosphorylation and nuclear shuttling. Our results highlight RAC/PAK1 as major players in YAP1 regulation triggered by cell adhesion.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Genes, Neurofibromatosis 2/physiology , Integrin beta1/physiology , Neurofibromin 2/metabolism , Phosphoproteins/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Adhesion , Cell Cycle Proteins , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Knockout , Neurofibromin 2/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins , p21-Activated Kinases/genetics , rac1 GTP-Binding Protein/genetics
13.
J Clin Oncol ; 35(4): 387-393, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28129526

ABSTRACT

Purpose Elderly patients with acute myeloid leukemia (AML) have a poor prognosis, and innovative maintenance therapy could improve their outcomes. Androgens, used in the treatment of aplastic anemia, have been reported to block proliferation of and initiate differentiation in AML cells. We report the results of a multicenter, phase III, randomized open-label trial exploring the benefit of adding androgens to maintenance therapy in patients 60 years of age or older. Patients and Methods A total of 330 patients with AML de novo or secondary to chemotherapy or radiotherapy were enrolled in the study. Induction therapy included idarubicin 8 mg/m2 on days 1 to 5, cytarabine 100 mg/m2 on days 1 to 7, and lomustine 200 mg/m2 on day 1. Patients in complete remission or partial remission received six reinduction courses, alternating idarubicin 8 mg/m2 on day 1, cytarabine 100 mg/m2 on days 1 to 5, and a regimen of methotrexate and mercaptopurine. Patients were randomly assigned to receive norethandrolone 10 or 20 mg/day, according to body weight, or no norethandrolone for a 2-year maintenance therapy regimen. The primary end point was disease-free survival by intention to treat. Secondary end points were event-free survival, overall survival, and safety. This trial was registered at www.ClinicalTrials.gov identifier NCT00700544. Results Random assignment allotted 165 patients to each arm; arm A received norethandrolone, and arm B did not receive norethandrolone. Complete remission or partial remission was achieved in 247 patients (76%). The Schoenfeld time-dependent model showed that norethandrolone significantly improved survival for patients still in remission at 1 year after induction. In arms A and B, respectively, 5-year disease-free survival was 31.2% and 16.2%, event-free survival was 21.5% and 12.9%, and overall survival was 26.3% and 17.2%. Norethandrolone improved outcomes irrelevant to all prognosis factors. Only patients with baseline leukocytes > 30 × 109/L did not benefit from norethandrolone. Conclusion This study demonstrates that maintenance therapy with norethandrolone significantly improves survival in elderly patients with AML without increasing toxicity.


Subject(s)
Androgens/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Age Factors , Aged , Female , Humans , Male , Middle Aged
14.
Transplantation ; 101(2): 437-444, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26950729

ABSTRACT

BACKGROUND: Admission of allogeneic stem cell transplantation (SCT) recipients to the intensive care unit (ICU) remains controversial, especially when graft-versus-host disease (GVHD) is present. METHODS: We performed a retrospective study to assess prognostic factors of survival in all allogeneic SCT recipients admitted to the ICU between 2002 and 2013 in our center which has flexible admission criteria, especially regarding GVHD. RESULTS: Of 349 patients who underwent allogeneic SCT during the study period, 92 patients (26%) were admitted to the ICU. Intensive care unit and hospital discharge rates were 66% and 46%, respectively, whereas 1 year survival was 24%. Acute GVHD, either grade III to IV (30 patients, 33%) or refractory (12 patients, 13%) had a nonsignificant impact on hospital mortality (odds ratio [OR], 2.1; P = 0.1; OR, 5, P = 0.05, respectively). Fifty percent of patients required invasive mechanical ventilation, 30% required vasopressors, 17% required renal replacement therapy, and 28% had liver impairment (bilirubin >34 µmol/L), each of these parameters defining organ failure. Mortality was closely associated with the number of organ failures as hospital discharge rates were 69%, 50%, 42%, and 0% among patients with 0 (26 patients), 1 (26 patients), 2 (26 patients), and 3 to 4 (14 patients) organ failures, respectively (OR, 2.7; 95% confidence interval, 1.6-4.6; P < 0.001 according to the number of organ failures). CONCLUSIONS: Early mortality of allogeneic SCT recipients admitted to the ICU is especially influenced by the number of organ failures and therefore patients with 0 to 2 organ failures should be considered if required. Refractory GVHD affects survival but not within the confined ICU admission.


Subject(s)
Critical Care , Graft vs Host Disease/therapy , Multiple Organ Failure/therapy , Stem Cell Transplantation/adverse effects , Adult , Chi-Square Distribution , Female , France , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , Graft vs Host Disease/mortality , Hospital Mortality , Humans , Intensive Care Units , Kaplan-Meier Estimate , Logistic Models , Male , Middle Aged , Multiple Organ Failure/diagnosis , Multiple Organ Failure/etiology , Multiple Organ Failure/mortality , Multivariate Analysis , Odds Ratio , Patient Admission , Patient Discharge , Proportional Hazards Models , Retrospective Studies , Risk Assessment , Risk Factors , Stem Cell Transplantation/mortality , Time Factors , Transplantation, Homologous , Treatment Outcome
15.
Eur J Immunol ; 46(7): 1770-82, 2016 07.
Article in English | MEDLINE | ID: mdl-27094152

ABSTRACT

We previously demonstrated an accumulation of tumor-reactive CD4(+) CD8(+) double positive (DP) T cells within melanoma-infiltrating lymphocytes, supporting their role in the regulation of anti-tumor immune responses. Similarly to their CD8(+) counterparts, intra-tumor DP T cells are MHC class-I restricted but differed by a limited lytic activity against autologous melanoma cells. Based on these observations and to further characterize DP T cells, both populations were compared at the transcriptional level. Our results revealed the overexpression of the IL-9 receptor (IL-9R) by DP T cells and prompted us to investigate the impact of IL-9 on their biology. We show that IL-9 favors DP T-cell survival by protecting them from apoptosis and by promoting their proliferation. In addition, IL-9 enhances their ability to produce cytokines and increased their levels of granzyme B/perforin as well as degranulation capacity, leading to a strengthened cytotoxic activity against melanoma cells. Taken together, the IL-9R(high) DP T-cell population could be a new preferential target for IL-9, which could take part in their retention within the melanoma infiltrate while also favoring their anti-tumor activity. More generally, our results extend the pleiotropic effects of IL-9 to IL-9R-expressing intra-tumor T cells, which could further potentiate anti-tumor immune responses.


Subject(s)
Interleukin-9/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma/immunology , Melanoma/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Apoptosis/drug effects , Apoptosis/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Survival/immunology , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Gene Expression , Humans , Interleukin-9/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma/genetics , Melanoma/pathology , Receptors, Interleukin-9/genetics , Receptors, Interleukin-9/metabolism , T-Lymphocyte Subsets/drug effects
16.
Oncoimmunology ; 5(12): e1250991, 2016.
Article in English | MEDLINE | ID: mdl-28123891

ABSTRACT

Although CD4+CD8+ double positive (DP) T cells represent a small fraction of peripheral T lymphocytes in healthy human donors, their frequency is often increased under pathological conditions (in blood and targeted tissues). In solid cancers such as melanoma, we previously demonstrated an enrichment of tumor reactive CD4lowCD8highαß DP T cells among tumor-infiltrating lymphocytes of unknown function. Similarly to their single positive (SP) CD8+ counterparts, intra-melanoma DP T cells recognized melanoma cell lines in an HLA-class-I restricted context. However, they presented a poor cytotoxic activity but a strong production of diverse Th1 and Th2 cytokines. The aim of this study was to clearly define the role of intra-melanoma CD4lowCD8highαß DP T cells in the antitumor immune response. Based on a comparative transcriptome analysis between intra-melanoma SP CD4+, SP CD8+ and DP autologous melanoma-infiltrating T-cell compartments, we evidenced an overexpression of the CD40L co-stimulatory molecule on activated DP T cells. We showed that, like SP CD4+ T cells, and through CD40L involvement, DP T cells are able to induce both proliferation and differentiation of B lymphocytes and maturation of functional DCs able to efficiently prime cytotoxic melanoma-specific CD8 T-cell responses. Taken together, these results highlight the helper potential of atypical DP T cells and their role in potentiating antitumor response.

17.
J Mol Cell Biol ; 8(4): 349-62, 2016 08.
Article in English | MEDLINE | ID: mdl-26459632

ABSTRACT

Although the conserved AAA ATPase and bromodomain factor, ATAD2, has been described as a transcriptional co-activator upregulated in many cancers, its function remains poorly understood. Here, using a combination of ChIP-seq, ChIP-proteomics, and RNA-seq experiments in embryonic stem cells where Atad2 is normally highly expressed, we found that Atad2 is an abundant nucleosome-bound protein present on active genes, associated with chromatin remodelling, DNA replication, and DNA repair factors. A structural analysis of its bromodomain and subsequent investigations demonstrate that histone acetylation guides ATAD2 to chromatin, resulting in an overall increase of chromatin accessibility and histone dynamics, which is required for the proper activity of the highly expressed gene fraction of the genome. While in exponentially growing cells Atad2 appears dispensable for cell growth, in differentiating ES cells Atad2 becomes critical in sustaining specific gene expression programmes, controlling proliferation and differentiation. Altogether, this work defines Atad2 as a facilitator of general chromatin-templated activities such as transcription.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , ATPases Associated with Diverse Cellular Activities , Acetylation , Cell Differentiation , Cell Proliferation , Chromatin Immunoprecipitation , Embryonic Stem Cells/cytology , Genome , Germ Cells/metabolism , Humans , Male , Nucleosomes/metabolism , Protein Binding , Proteomics
18.
PLoS One ; 10(6): e0129222, 2015.
Article in English | MEDLINE | ID: mdl-26053431

ABSTRACT

Human acute promyelocytic leukemia (APL) is characterized by a specific balanced translocation t(15;17)(q22;q21) involving the PML and RARA genes. In both de novo and therapy-related APL, the most frequent PML breakpoints are located within intron 6, and less frequently in intron 3; the precise mechanisms by which these breakpoints arise and preferentially in PML intron 6 remain unsolved. To investigate the intrinsic properties of the PML intron sequences in vivo, we designed Saccharomyces cerevisiae strains containing human PML intron 6 or intron 3 sequences inserted in yeast chromosome V and measured gross chromosomal rearrangements (GCR). This approach provided evidence that intron 6 had a superior instability over intron 3 due to an intrinsic property of the sequence and identified the 3' end of intron 6 as the most susceptible to break. Using yeast strains invalidated for genes that control DNA replication, we show that this differential instability depended at least upon Rrm3, a DNA helicase, and Mrc1, the human claspin homolog. GCR induction by hydrogen peroxide, a general genotoxic agent, was also dependent on genetic context. We conclude that: 1) this yeast system provides an alternative approach to study in detail the properties of human sequences in a genetically controlled situation and 2) the different susceptibility to produce DNA breaks in intron 6 versus intron 3 of the human PML gene is likely due to an intrinsic property of the sequence and is under replication fork genetic control.


Subject(s)
DNA Replication , DNA-Binding Proteins/metabolism , Genomic Instability , Introns , Nuclear Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Chromosome Breakpoints , Chromosome Mapping , DNA Breaks/drug effects , Gene Order , Genetic Loci , Humans , Hydrogen Peroxide/pharmacology , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Promyelocytic Leukemia Protein , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Translocation, Genetic
19.
BMC Cancer ; 15: 262, 2015 Apr 11.
Article in English | MEDLINE | ID: mdl-25881299

ABSTRACT

BACKGROUND: Cyclin D1 and its kinase partners control cell cycle progression. Cyclin D1 is frequently deregulated in various cancers, including malignant hemopathies, and tumor cells display uncontrolled cell proliferation. Cyclin D1 is not expressed in the B-cell lineage but is found in multiple myeloma (MM) cells in almost 50% of patients with this condition. Paradoxically, cyclin D1 expression is associated with a good prognosis and longer overall survival in MM patients. METHODS: We used two independent MM cell lines (RPMI 8226 and LP1) to generate several clones stably expressing either the green fluorescent protein (GFP) or a GFP-cyclin D1 fusion protein, and we analyzed the properties acquired following cyclin D1 expression. RESULTS: Whole-genome expression analysis in the cell clones indicated that cyclin D1 profoundly modified several cellular functions, including the regulation of apoptotic cell death. We studied the apoptotic response of GFP- and GFP-cyclin D1-expressing clones to bortezomib-treatment. We found that the apoptotic response occurred faster and was of a greater amplitude in cyclin D1-expressing cells. Cyclin D1 expression enhanced the caspase-dependent apoptosis mediated by the intrinsic mitochondrial pathway. More importantly, cyclin D1 also activated the unfolded protein response (UPR) and induced endoplasmic reticulum (ER) stress-mediated apoptosis. CONCLUSION: The ER is well known to be a crucial regulator of plasma cell death and it plays the same role in their malignant counterparts, myeloma cells. This role involves activation of the UPR controlled at least in part by cyclin D1.


Subject(s)
Cyclin D1/biosynthesis , Multiple Myeloma/genetics , Neoplasm Proteins/biosynthesis , Unfolded Protein Response/genetics , Apoptosis/genetics , Cell Line, Tumor , Cell Lineage/genetics , Cell Proliferation , Cyclin D1/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genome, Human , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Neoplasm Proteins/genetics , Prognosis , Signal Transduction/drug effects
20.
Antioxid Redox Signal ; 23(1): 1-14, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-24512221

ABSTRACT

AIMS: Ectopic activation of tissue-specific genes accompanies malignant transformation in many cancers. Prolactin (PRL) aberrant activation in lung cancer was investigated here to highlight its value as a biomarker. RESULTS: PRL is ectopically activated in a subset of very aggressive lung tumors, associated with a rapid fatal outcome, in our cohort of 293 lung tumor patients and in an external independent series of patients. Surprisingly PRL receptor expression was not detected in the vast majority of PRL-expressing lung tumors. Additionally, the analysis of the PRL transcripts in lung tumors and cell lines revealed systematic truncations of their 5' regions, including the signal peptide-encoding portions. PRL expression was found to sustain cancer-specific gene expression circuits encompassing genes that are normally responsive to hypoxia. Interestingly, this analysis also indicated that histone deacetylase (HDAC) inhibitors could counteract the PRL-associated transcriptional activity. INNOVATION AND CONCLUSION: Altogether, this work not only unravels a yet unknown oncogenic mechanism but also indicates that the specific category of PRL-expressing aggressive lung cancers could be particularly responsive to an HDAC inhibitor-based treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Histone Deacetylase Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Prolactin/genetics , Adult , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/diagnosis , Cell Line, Tumor , Cohort Studies , Female , Histone Deacetylase Inhibitors/metabolism , Humans , Male , Middle Aged , Pregnancy , Prognosis , Prolactin/metabolism , RNA, Messenger/metabolism , Receptors, Prolactin/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...