Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
1.
Exp Ther Med ; 23(1): 47, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-34934426

RÉSUMÉ

Treatment of resistant or recurrent acute lymphoblastic leukemia (ALL) remains a challenge. It was previously demonstrated that the adhesion molecule integrin α4, referred to hereafter as α4, mediates the cell adhesion-mediated drug resistance (CAM-DR) of B-cell ALL by binding to vascular cell adhesion molecule-1 (VCAM-1) on bone marrow stroma. In addition, it was previously observed that the blockade of α4 with natalizumab or inhibition using the small molecule antagonist TBC3486 sensitized relapsed ALL cells to chemotherapy. However, α4-targeted therapy is not clinically available for the treatment of leukemia to date. In the present study, the use of a novel non-peptidic small molecule integrin α4 antagonist, AVA4746, as a potential new approach to combat drug-resistant B-ALL was explored. An in vitro co-culture = model of primary B-ALL cells and an in vivo xenograft model of patient-derived B-ALL cells were utilized for evaluation of AVA4746. VLA-4 conformation activation, cell adhesion/de-adhesion, endothelial tube formation, in vivo leukemia cell mobilization and survival assays were performed. AVA4746 exhibited high affinity for binding to B-ALL cells, where it also efficiently blocked ligand-binding to VCAM-1. In addition, AVA4746 caused the functional de-adhesion of primary B-ALL cells from VCAM-1. Inhibition of α4 using AVA4746 also prevented angiogenesis in vitro and when applied in combination with chemotherapy consisting of Vincristine, Dexamethasone and L-asparaginase, it prolonged the survival of ~33% of the mice in an in vivo xenograft model of B-ALL. These data implicate the potential of targeting the α4-VCAM-1 interaction using AVA4746 for the treatment of drug-resistant B-lineage ALL.

2.
Front Oncol ; 11: 766888, 2021.
Article de Anglais | MEDLINE | ID: mdl-34926269

RÉSUMÉ

The PI3K/Akt pathway-and in particular PI3Kδ-is known for its role in drug resistant B-cell acute lymphoblastic leukemia (B-ALL) and it is often upregulated in refractory or relapsed B-ALL. Myc proteins are transcription factors responsible for transcribing pro-proliferative genes and c-Myc is often overexpressed in cancers. The chromatin regulator BRD4 is required for expression of c-Myc in hematologic malignancies including B-ALL. Previously, combination of BRD4 and PI3K inhibition with SF2523 was shown to successfully decrease Myc expression. However, the underlying mechanism and effect of dual inhibition of PI3Kδ/BRD4 in B-ALL remains unknown. To study this, we utilized SF2535, a novel small molecule dual inhibitor which can specifically target the PI3Kδ isoform and BRD4. We treated primary B-ALL cells with various concentrations of SF2535 and studied its effect on specific pharmacological on-target mechanisms such as apoptosis, cell cycle, cell proliferation, and adhesion molecules expression usingin vitro and in vivo models. SF2535 significantly downregulates both c-Myc mRNA and protein expression through inhibition of BRD4 at the c-Myc promoter site and decreases p-AKT expression through inhibition of the PI3Kδ/AKT pathway. SF2535 induced apoptosis in B-ALL by downregulation of BCL-2 and increased cleavage of caspase-3, caspase-7, and PARP. Moreover, SF2535 induced cell cycle arrest and decreased cell counts in B-ALL. Interestingly, SF2535 decreased the mean fluorescence intensity (MFI) of integrin α4, α5, α6, and ß1 while increasing MFI of CXCR4, indicating that SF2535 may work through inside-out signaling of integrins. Taken together, our data provide a rationale for the clinical evaluation of targeting PI3Kδ/BRD4 in refractory or relapsed B-ALL using SF2535.

4.
Cells ; 9(6)2020 06 16.
Article de Anglais | MEDLINE | ID: mdl-32560076

RÉSUMÉ

Drug resistance is an obstacle in the therapy of acute lymphoblastic leukemia (ALL). Whether the physical properties such as the motility of the cells contribute to the survival of ALL cells after drug treatment has recently been of increasing interest, as they could potentially allow the metastasis of solid tumor cells and the migration of leukemia cells. We hypothesized that chemotherapeutic treatment may alter these physical cellular properties. To investigate the motility of chemotherapeutics-treated B-cell ALL (B-ALL) cells, patient-derived B-ALL cells were treated with chemotherapy for 7 days and left for 12 h without chemotherapeutic treatment. Two parameters of motility were studied, velocity and migration distance, using a time-lapse imaging system. The study revealed that compared to non-chemotherapeutically treated B-ALL cells, B-ALL cells that survived chemotherapy treatment after 7 days showed reduced motility. We had previously shown that Tysabri and P5G10, antibodies against the adhesion molecules integrins α4 and α6, respectively, may overcome drug resistance mediated through leukemia cell adhesion to bone marrow stromal cells. Therefore, we tested the effect of integrin α4 or α6 blockade on the motility of chemotherapeutics-treated ALL cells. Only integrin α4 blockade decreased the motility and velocity of two chemotherapeutics-treated ALL cell lines. Interestingly, integrin α6 blockade did not affect the velocity of chemoresistant ALL cells. This study explores the physical properties of the movements of chemoresistant B-ALL cells and highlights a potential link to integrins. Further studies to investigate the underlying mechanism are warranted.


Sujet(s)
Adhérence cellulaire/physiologie , Mouvement cellulaire/physiologie , Leucémie-lymphome lymphoblastique à précurseurs B et T/imagerie diagnostique , Imagerie accélérée , Cellules de la moelle osseuse/cytologie , Adhérence cellulaire/effets des médicaments et des substances chimiques , Molécules d'adhérence cellulaire/métabolisme , Humains , Intégrine alpha4/pharmacologie , Cellules stromales/cytologie , Imagerie accélérée/méthodes
5.
Blood ; 136(2): 210-223, 2020 07 09.
Article de Anglais | MEDLINE | ID: mdl-32219444

RÉSUMÉ

Resistance to multimodal chemotherapy continues to limit the prognosis of acute lymphoblastic leukemia (ALL). This occurs in part through a process called adhesion-mediated drug resistance, which depends on ALL cell adhesion to the stroma through adhesion molecules, including integrins. Integrin α6 has been implicated in minimal residual disease in ALL and in the migration of ALL cells to the central nervous system. However, it has not been evaluated in the context of chemotherapeutic resistance. Here, we show that the anti-human α6-blocking Ab P5G10 induces apoptosis in primary ALL cells in vitro and sensitizes primary ALL cells to chemotherapy or tyrosine kinase inhibition in vitro and in vivo. We further analyzed the underlying mechanism of α6-associated apoptosis using a conditional knockout model of α6 in murine BCR-ABL1+ B-cell ALL cells and showed that α6-deficient ALL cells underwent apoptosis. In vivo deletion of α6 in combination with tyrosine kinase inhibitor (TKI) treatment was more effective in eradicating ALL than treatment with a TKI (nilotinib) alone. Proteomic analysis revealed that α6 deletion in murine ALL was associated with changes in Src signaling, including the upregulation of phosphorylated Lyn (pTyr507) and Fyn (pTyr530). Thus, our data support α6 as a novel therapeutic target for ALL.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Délétion de gène , Intégrine alpha6 , Protéines tumorales , Leucémie-lymphome lymphoblastique à précurseurs B , Pyrimidines/pharmacologie , Animaux , Anticorps antitumoraux/pharmacologie , Anticorps neutralisants/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Humains , Intégrine alpha6/génétique , Intégrine alpha6/métabolisme , Mâle , Souris , Souris knockout , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B/thérapie
6.
Int J Mol Sci ; 21(3)2020 Feb 05.
Article de Anglais | MEDLINE | ID: mdl-32033444

RÉSUMÉ

Minimal residual disease (MRD) refers to a chemotherapy/radiotherapy-surviving leukemia cell population that gives rise to relapse of the disease. The detection of MRD is critical for predicting the outcome and for selecting the intensity of further treatment strategies. The development of various new diagnostic platforms, including next-generation sequencing (NGS), has introduced significant advances in the sensitivity of MRD diagnostics. Here, we review current methods to diagnose MRD through phenotypic marker patterns or differential gene patterns through analysis by flow cytometry (FCM), polymerase chain reaction (PCR), real-time quantitative polymerase chain reaction (RQ-PCR), reverse transcription polymerase chain reaction (RT-PCR) or NGS. Future advances in clinical procedures will be molded by practical feasibility and patient needs regarding greater diagnostic sensitivity.


Sujet(s)
Récidive tumorale locale/diagnostic , Récidive tumorale locale/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/diagnostic , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Cytométrie en flux , Séquençage nucléotidique à haut débit , Humains , Maladie résiduelle/diagnostic , Maladie résiduelle/génétique , Cellules tumorales circulantes/anatomopathologie , Réaction de polymérisation en chaîne , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie
7.
J Ultrasound Med ; 39(3): 589-595, 2020 Mar.
Article de Anglais | MEDLINE | ID: mdl-31633840

RÉSUMÉ

OBJECTIVES: This article reports a study of cell mechanics in patient-derived (primary) B-cell acute lymphocytic leukemia (ALL) cells treated with antibodies against integrins. Leukemia cell adhesion to stromal cells mediates chemotherapeutic drug resistance, also known as cell adhesion-mediated chemotherapeutic drug resistance. We have previously shown that antibodies against integrin α4 and α6 adhesion molecules can de-adhere ALL cells from stromal cells or counter-receptors. Because drug-resistant cells are more deformable, as evaluated by single-beam acoustic tweezers, we hypothesized that changes in cell mechanics might contribute to the de-adhesive effect of integrin-targeting antibodies. METHODS: In this study, the deformability of primary pre-B ALL cells was evaluated by single-beam acoustic tweezers after treatments with the de-adhering antibody Tysabri or P5G10 against integrin α4 and α6 adhesion molecules. RESULTS: We demonstrated that primary ALL cells treated with P5G10 expressed decreased deformability compared with immunoglobulin G1 -treated control cells (P < .05). Tysabri did not show an effect on deformability (P > .05). CONCLUSIONS: These results suggest that decreased deformability is associated with an integrin α6 blockade. Further assessments of the functional roles of deformability and integrin blockades in B-ALL cell drug resistance and deformability, respectively, are necessary.


Sujet(s)
Adhérence cellulaire/effets des médicaments et des substances chimiques , Facteurs immunologiques/usage thérapeutique , Intégrines/effets des médicaments et des substances chimiques , Natalizumab/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B et T/imagerie diagnostique , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Acoustique , Cellules cultivées , Humains , Immunoglobuline G/administration et posologie , Échographie/méthodes
8.
Int J Mol Sci ; 20(2)2019 Jan 18.
Article de Anglais | MEDLINE | ID: mdl-30669372

RÉSUMÉ

Adhesion of acute lymphoblastic leukemia (ALL) cells to bone marrow stroma cells triggers intracellular signals regulating cell-adhesion-mediated drug resistance (CAM-DR). Stromal cell protection of ALL cells has been shown to require active AKT. In chronic lymphocytic leukemia (CLL), adhesion-mediated activation of the PI3K/AKT pathway is reported. A novel FDA-approved PI3Kδ inhibitor, CAL-101/idelalisib, leads to downregulation of p-AKT and increased apoptosis of CLL cells. Recently, two additional PI3K inhibitors have received FDA approval. As the PI3K/AKT pathway is also implicated in adhesion-mediated survival of ALL cells, PI3K inhibitors have been evaluated preclinically in ALL. However, PI3K inhibition has yet to be approved for clinical use in ALL. Here, we review the role of PI3K in normal hematopoietic cells, and in ALL. We focus on summarizing targeting strategies of PI3K in ALL.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Thérapie moléculaire ciblée , Phosphatidylinositol 3-kinases/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Antinéoplasiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lymphocytes B/effets des médicaments et des substances chimiques , Lymphocytes B/métabolisme , Essais cliniques comme sujet , Évaluation préclinique de médicament , Résistance aux médicaments antinéoplasiques , Humains , Isoenzymes , Thérapie moléculaire ciblée/effets indésirables , Thérapie moléculaire ciblée/méthodes , Leucémie-lymphome lymphoblastique à précurseurs B et T/diagnostic , Leucémie-lymphome lymphoblastique à précurseurs B et T/mortalité , Protéines proto-oncogènes c-akt/métabolisme , Résultat thérapeutique
9.
Sci Rep ; 8(1): 15708, 2018 10 24.
Article de Anglais | MEDLINE | ID: mdl-30356155

RÉSUMÉ

The role of cell mechanics in cancer cells is a novel research area that has resulted in the identification of new mechanisms of therapy resistance. Single beam acoustic (SBA) tweezers are a promising technology for the quantification of the mechanical phenotype of cells. Our previous study showed that SBA tweezers can be used to quantify the deformability of adherent breast cancer cell lines. The physical properties of patient-derived (primary) pre-B acute lymphoblastic leukemia (ALL) cells involved in chemotherapeutic resistance have not been widely investigated. Here, we demonstrate the feasibility of analyzing primary pre-B ALL cells from four cases using SBA tweezers. ALL cells showed increased deformability with increasing acoustic pressure of the SBA tweezers. Moreover, ALL cells that are resistant to chemotherapeutic drugs were more deformable than were untreated ALL cells. We demonstrated that SBA tweezers can quantify the deformability of nonadherent leukemia cells and discriminate this mechanical phenotype in chemotherapy-resistant leukemia cells in a contact- and label-free manner.


Sujet(s)
Phénomènes biomécaniques , Résistance aux médicaments antinéoplasiques , Pinces optiques , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Leucémie-lymphome lymphoblastique à précurseurs B et T/anatomopathologie , Acoustique , Lignée cellulaire tumorale , Forme de la cellule , Élasticité , Humains , Analyse sur cellule unique
10.
PLoS One ; 12(11): e0187684, 2017.
Article de Anglais | MEDLINE | ID: mdl-29117236

RÉSUMÉ

We recently demonstrated the effectiveness of blocking CD49d with anti-functional antibodies or small molecule inhibitors as a rational targeted approach to the treatment of acute leukemia in combination with chemotherapy. Antisense oligonucleotide promises to be no less specific than antibodies and inhibitors, but more interesting for pharmacokinetics and pharmacodynamics. We addressed this using the published CD49d antisense drug ATL1102. In vitro, we incubated/nucleofected the ALL cell line Kasumi-2 with ATL1102. In vivo, immunodeficient hosts were engrafted with primary ALL cells and treated with ATL1102. Changes in expression of CD49d mRNA and CD49d protein, and of cooperating gene products, including ß1 integrin and CXCR4, as well as survival in the mouse experiments were quantified. We observed dose-dependent down-regulation of CD49d mRNA and protein levels and its partner integrin ß1 cell surface protein level and, up-regulation of CXCR4 surface expression. The suppression was more pronounced after nucleofection than after incubation, where down-regulation was significant only at the higher doses. In vivo effects of ATL1102 were not sufficient to translate into "clinical" benefit in the leukemia model. In summary, antisense oligonucleotides are successful tools for specifically modulating gene expression but sufficient delivery to down-regulate CD49d in vivo may be difficult to achieve.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Intégrine alpha4/antagonistes et inhibiteurs , Oligonucléotides antisens/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie , Animaux , Transport biologique , Lignée cellulaire tumorale , Femelle , Humains , Injections veineuses , Injections sous-cutanées , Intégrine alpha4/génétique , Intégrine alpha4/métabolisme , Antigènes CD29/génétique , Antigènes CD29/métabolisme , Souris , Souris de lignée NOD , Oligonucléotides antisens/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/mortalité , Leucémie-lymphome lymphoblastique à précurseurs B et T/anatomopathologie , Récepteurs CXCR4/génétique , Récepteurs CXCR4/métabolisme , Transduction du signal , Analyse de survie , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Cancers (Basel) ; 9(9)2017 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-28891959

RÉSUMÉ

The quest continues for targeted therapies to reduce the morbidity of chemotherapy and to improve the response of resistant leukemia. Adhesion of acute lymphoblastic leukemia (ALL) cells to bone marrow stromal cells triggers intracellular signals that promote cell-adhesion-mediated drug resistance (CAM-DR). Idelalisib, an U.S. Food and Drug Administration (FDA)-approved PI3Kδ-specific inhibitor has been shown to be effective in CLL in down-regulating p-Akt and prolonging survival in combination with Rituximab; herein we explore the possibility of its use in B ALL and probe the mechanism of action. Primary B ALL in contact with OP9 stromal cells showed increased p-Aktser473. Idelalisib decreased p-Akt in patient samples of ALL with diverse genetic lesions. Addition of idelalisib to vincristine inhibited proliferation when compared to vincristine monotherapy in a subset of samples tested. Idelalisib inhibited ALL migration to SDF-1α in vitro and blocked homing of ALL cells to the bone marrow in vivo. This report tests PI3Kδ inhibitors in a more diverse group of ALL than has been previously reported and is the first published report of idelalisib inhibiting homing of ALL cells to bone marrow. Our data support further pre-clinical evaluation of idelalisib for the therapy of B ALL.

12.
Exp Hematol ; 52: 1-11, 2017 08.
Article de Anglais | MEDLINE | ID: mdl-28479420

RÉSUMÉ

Cancer stem cells (CSCs), including leukemia stem cells (LSCs), exhibit self-renewal capacity and differentiation potential and have the capacity to maintain or renew and propagate a tumor/leukemia. The initial isolation of CSCs/LSCs was in adult myelogenous leukemia, although more recently, the existence of CSCs in a wide variety of other cancers has been reported. CSCs, in general, and LSCs, specifically with respect to this review, are responsible for initiation of disease, therapeutic resistance and ultimately disease relapse. One key focus in cancer research over the past decade has been the development of therapies that safely eliminate the LSC/CSC population. One major obstacle to this goal is the identification of key mechanisms that distinguish LSCs from normal endogenous hematopoietic stem cells. An additional daunting feature that has recently come to light with advances in next-generation sequencing and single-cell sequencing is the heterogeneity within leukemias/tumors, with multiple combinations of mutations, gain and loss of function of genes, and so on being capable of driving disease, even within the CSC/LSC population. The focus of this review/perspective is on our work in identifying and validating, in both chronic myelogenous leukemia and acute lymphoblastic leukemia, a safe and efficacious mechanism to target an evolutionarily conserved signaling nexus, which constitutes a common "Achilles heel" for LSCs/CSCs, using small molecule-specific CBP/catenin antagonists.


Sujet(s)
Composés hétérocycliques bicycliques/pharmacologie , Protéine de liaison à l'élément de réponse à l'AMP cyclique/antagonistes et inhibiteurs , Cellules souches tumorales/effets des médicaments et des substances chimiques , Pyrimidinones/pharmacologie , bêta-Caténine/antagonistes et inhibiteurs , gamma-Caténine/antagonistes et inhibiteurs , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/métabolisme , Humains , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Leucémie-lymphome lymphoblastique à précurseurs B et T/anatomopathologie , Liaison aux protéines/effets des médicaments et des substances chimiques , bêta-Caténine/métabolisme , gamma-Caténine/métabolisme
13.
Blood ; 121(10): 1814-8, 2013 Mar 07.
Article de Anglais | MEDLINE | ID: mdl-23319569

RÉSUMÉ

Bone marrow (BM) provides chemoprotection for acute lymphoblastic leukemia (ALL) cells, contributing to lack of efficacy of current therapies. Integrin alpha4 (alpha4) mediates stromal adhesion of normal and malignant B-cell precursors, and according to gene expression analyses from 207 children with minimal residual disease, is highly associated with poorest outcome. We tested whether interference with alpha4-mediated stromal adhesion might be a new ALL treatment. Two models of leukemia were used, one genetic (conditional alpha4 ablation of BCR-ABL1 [p210(+)] leukemia) and one pharmacological (anti-functional alpha4 antibody treatment of primary ALL). Conditional deletion of alpha4 sensitized leukemia cell to nilotinib. Adhesion of primary pre-B ALL cells was alpha4-dependent; alpha4 blockade sensitized primary ALL cells toward chemotherapy. Chemotherapy combined with Natalizumab prolonged survival of NOD/SCID recipients of primary ALL, suggesting adjuvant alpha4 inhibition as a novel strategy for pre-B ALL.


Sujet(s)
Anticorps monoclonaux humanisés/pharmacologie , Résistance aux médicaments antinéoplasiques , Protéines de fusion bcr-abl/physiologie , Intégrine alpha4/composition chimique , Maladie résiduelle/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Animaux , Moelle osseuse/effets des médicaments et des substances chimiques , Moelle osseuse/métabolisme , Moelle osseuse/anatomopathologie , Adhérence cellulaire , Enfant , Cytométrie en flux , Humains , Integrases/métabolisme , Intégrine alpha4/génétique , Intégrine alpha4/métabolisme , Souris , Souris de lignée NOD , Souris knockout , Souris SCID , Natalizumab , Maladie résiduelle/métabolisme , Maladie résiduelle/mortalité , Leucémie-lymphome lymphoblastique à précurseurs B/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B/mortalité , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , RT-PCR , Cellules stromales/effets des médicaments et des substances chimiques , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie
14.
Blood ; 118(8): 2191-9, 2011 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-21715311

RÉSUMÉ

Relapse of drug-resistant acute lymphoblastic leukemia (ALL) has been associated with increased expression of survivin/BIRC5, an inhibitor of apoptosis protein, suggesting a survival advantage for ALL cells. In the present study, we report that inhibition of survivin in patient-derived ALL can eradicate leukemia. Targeting survivin with shRNA in combination with chemotherapy resulted in no detectable minimal residual disease in a xenograft model of primary ALL. Similarly, pharmacologic knock-down of survivin using EZN-3042, a novel locked nucleic acid antisense oligonucleotide, in combination with chemotherapy eliminated drug-resistant ALL cells. These findings show the importance of survivin expression in drug resistance and demonstrate that survivin inhibition may represent a powerful approach to overcoming drug resistance and preventing relapse in patients with ALL.


Sujet(s)
Protéines IAP/antagonistes et inhibiteurs , Protéines IAP/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie , Animaux , Association thérapeutique , Résistance aux médicaments antinéoplasiques/génétique , Expression des gènes , Ciblage de gène , Humains , Protéines IAP/déficit , Souris , Souris de lignée NOD , Souris knockout , Maladie résiduelle , Oligonucléotides/génétique , Oligonucléotides/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Petit ARN interférent/génétique , Protéines de répression/déficit , Protéines de répression/génétique , Survivine , Test clonogénique de cellules souches tumorales , Tests d'activité antitumorale sur modèle de xénogreffe
15.
Curr Cancer Drug Targets ; 11(2): 213-25, 2011 Feb.
Article de Anglais | MEDLINE | ID: mdl-21158719

RÉSUMÉ

Previously, we demonstrated that survivin expression is CBP/ß-catenin/TCF-dependent. Now, using NCI-H28 cells, which harbor a homozygous deletion of ß-catenin, we demonstrate that survivin transcription can similarly be mediated by nuclear γ-catenin. ICG-001, a specific inhibitor of binding to the N-terminus of CBP, effectively attenuates survivin expression. We demonstrate that γ-catenin by binding to TCF family members and specifically recruiting the coactivator CBP drives survivin transcription particularly in ß-catenin-deficient cells. We also examined the relative expression of γ-catenin and ß-catenin in 90 cases of chronic myeloid leukemia (CML) in a published gene expression microarray data base. A statistically significant negative correlation between γ-catenin and ß-catenin was found in AP/BC cases (-0.389, P = 0.006). Furthermore, in subsequent independent validation studies by qPCR in 28 CP and BC patients increased γ-catenin expression predominated in BC cases and was associated with concomitantly increased survivin expression. Gene expression was 3- and 6-fold greater in BC patients as compared to CP patients, for γ-catenin and survivin, respectively. Consistent with this observation, nuclear γ-catenin accumulation was evident in this population consistent with a potential transcriptional role. Combined treatment with imatinib mesylate (IM) and ICG-001 significantly inhibited colony formation in sorted CD34(+) CML progenitors (survivin(+)/γ-catenin(high)/ß-catenin(low)) isolated from one BC and one AP patient resistant to IM. Therefore, we believe that the ability of ICG-001 to block both the CBP/γ-catenin interaction and the CBP/ß-catenin interaction may have clinical significance in cancers in which γ-catenin plays a significant transcriptional role.


Sujet(s)
Protéine CBP/métabolisme , Protéines IAP/métabolisme , Transcription génétique , bêta-Caténine/métabolisme , gamma-Caténine/métabolisme , Animaux , Antigènes CD34/métabolisme , Antinéoplasiques/pharmacologie , Composés hétérocycliques bicycliques/pharmacologie , Protéine CBP/antagonistes et inhibiteurs , Protéine CBP/génétique , Lignée cellulaire tumorale , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/métabolisme , Humains , Protéines IAP/génétique , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Souris , Pyrimidinones/pharmacologie , ARN messager/métabolisme , Petit ARN interférent , Protéines de fusion recombinantes/métabolisme , Survivine , Facteurs de transcription TCF/métabolisme , Transcription génétique/effets des médicaments et des substances chimiques , bêta-Caténine/génétique , gamma-Caténine/génétique , Facteurs de transcription CBP-p300/génétique , Facteurs de transcription CBP-p300/métabolisme
16.
Exp Cell Res ; 315(15): 2624-36, 2009 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-19460366

RÉSUMÉ

Mesenchymal stem cell preparations have been proposed for muscle regeneration in musculoskeletal disorders. Although MSCs have great in vitro expansion potential and possess the ability to differentiate into several mesenchymal lineages, myogenesis has proven to be much more difficult to induce. We have recently demonstrated that Pax3, the master regulator of the embryonic myogenic program, enables the in vitro differentiation of a murine mesenchymal stem cell line (MSCB9-Pax3) into myogenic progenitors. Here we show that injection of these cells into cardiotoxin-injured muscles of immunodeficient mice leads to the development of muscle tumors, resembling rhabdomyosarcomas. We then extended these studies to primary human mesenchymal stem cells (hMSCs) isolated from bone marrow. Upon genetic modification with a lentiviral vector encoding PAX3, hMSCs activated the myogenic program as demonstrated by expression of myogenic regulatory factors. Upon transplantation, the PAX3-modified MSCs did not generate rhabdomyosarcomas but rather, resulted in donor-derived myofibers. These were found at higher frequency in PAX3-transduced hMSCs than in mock-transduced MSCs. Nonetheless, neither engraftment of PAX3-modified or unmodified MSCs resulted in improved contractility. Thus these findings suggest that limitations remain to be overcome before MSC preparations result in effective treatment for muscular dystrophies.


Sujet(s)
Différenciation cellulaire/physiologie , Dystrophine , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses/physiologie , Facteurs de transcription PAX/métabolisme , Récupération fonctionnelle , Animaux , Antigènes CD/métabolisme , Marqueurs biologiques/métabolisme , Cellules cultivées , Dystrophine/génétique , Dystrophine/métabolisme , Humains , Mâle , Cellules souches mésenchymateuses/cytologie , Souris , Souris knockout , Développement musculaire/physiologie , Muscles squelettiques/anatomopathologie , Muscles squelettiques/physiologie , Dystrophies musculaires/génétique , Dystrophies musculaires/anatomopathologie , Dystrophies musculaires/thérapie , Neuréguline-1/métabolisme , Facteur de transcription PAX3 , Facteurs de transcription PAX/génétique , Rhabdomyosarcome/métabolisme , Rhabdomyosarcome/anatomopathologie
17.
EMBO J ; 27(20): 2766-79, 2008 Oct 22.
Article de Anglais | MEDLINE | ID: mdl-18833193

RÉSUMÉ

Facioscapulohumeral muscular dystrophy (FSHD) is caused by an unusual deletion with neomorphic activity. This deletion derepresses genes in cis; however which candidate gene causes the FSHD phenotype, and through what mechanism, is unknown. We describe a novel genetic tool, inducible cassette exchange, enabling rapid generation of isogenetically modified cells with conditional and variable transgene expression. We compare the effects of expressing variable levels of each FSHD candidate gene on myoblasts. This screen identified only one gene with overt toxicity: DUX4 (double homeobox, chromosome 4), a protein with two homeodomains, each similar in sequence to Pax3 and Pax7. DUX4 expression recapitulates key features of the FSHD molecular phenotype, including repression of MyoD and its target genes, diminished myogenic differentiation, repression of glutathione redox pathway components, and sensitivity to oxidative stress. We further demonstrate competition between DUX4 and Pax3/Pax7: when either Pax3 or Pax7 is expressed at high levels, DUX4 is no longer toxic. We propose a hypothesis for FSHD in which DUX4 expression interferes with Pax7 in satellite cells, and inappropriately regulates Pax targets, including myogenic regulatory factors, during regeneration.


Sujet(s)
Régulation de l'expression des gènes , Protéines à homéodomaine/physiologie , Dystrophie musculaire facio-scapulo-humérale/anatomopathologie , Myoblastes/métabolisme , Animaux , Différenciation cellulaire , Clonage moléculaire , Délétion de gène , Glutathion/métabolisme , Protéines à homéodomaine/métabolisme , Souris , Dystrophie musculaire facio-scapulo-humérale/métabolisme , Oxydoréduction , Facteur de transcription PAX3 , Facteur de transcription PAX7/métabolisme , Facteurs de transcription PAX/métabolisme , Phénotype , Transgènes
18.
Exp Cell Res ; 314(8): 1721-33, 2008 May 01.
Article de Anglais | MEDLINE | ID: mdl-18395202

RÉSUMÉ

Mesenchymal stem cells (MSCs) residing within the bone marrow (BM) differentiate into multiple lineages, including fat, bone, and cartilage. Because MSCs are multipotent and have a great capacity to be expanded in vitro, these cells are an attractive candidate for clinical applications to repair or regenerate damaged tissues of mesenchymal origin. However, application of MSCs to muscle degenerative diseases has been hampered by the poor differentiation of MSCs into the muscle lineage. To date most methods require the presence of strong non-physiological agents, such as azacytidine. In the present study we explored the potential of Pax3, the master regulator of the embryonic myogenic program, to promote myogenic differentiation from MSCs. Our results clearly demonstrate that Pax3 promotes the differentiation of MSCs towards the myogenic lineage, which occurs at the expense of other mesenchymal lineages including fat, bone, and cartilage. This effect is cell type-selective since Pax3 overexpression in endothelial cells fails to promote myogenesis. These results highlight the potential of regulating transcriptional pathways to direct differentiation of adult stem cells.


Sujet(s)
Cellules souches mésenchymateuses/cytologie , Cellules musculaires/cytologie , Facteurs de transcription PAX/métabolisme , Adipogenèse , Animaux , Différenciation cellulaire , Lignée cellulaire , Lignage cellulaire , Chondrogenèse , Cellules souches mésenchymateuses/métabolisme , Souris , Ostéogenèse , Facteur de transcription PAX3
19.
Biochem Biophys Res Commun ; 330(4): 1153-61, 2005 May 20.
Article de Anglais | MEDLINE | ID: mdl-15823564

RÉSUMÉ

In addition to long-term self-renewal capability, human mesenchymal stem cells (MSCs) possess versatile differentiation potential ranging from mesenchyme-related multipotency to neuroectodermal and endodermal competency. Of particular concern is hepatogenic potential that can be used for liver-directed stem cell therapy and transplantation. In this study, we have investigated whether human umbilical cord blood (UCB)-derived MSCs are also able to differentiate into hepatocyte-like cells. MSCs isolated from UCB were cultured under the pro-hepatogenic condition similar to that for bone marrow (BM)-derived MSCs. Expression of a variety of hepatic lineage markers was analyzed by flow cytometry, RT-PCR, Western blot, and immunofluorescence. The functionality of differentiated cells was assessed by their ability to incorporate DiI-acetylated low-density lipoprotein (DiI-Ac-LDL). As the cells were morphologically transformed into hepatocyte-like cells, they expressed Thy-1, c-Kit, and Flt-3 at the cell surface, as well as albumin, alpha-fetoprotein, and cytokeratin-18 and 19 in the interior. Moreover, about a half of the cells were found to acquire the capability to transport DiI-Ac-LDL. Based on these observations, and taking into account immense advantages of UCB over other stem cell sources, we conclude that UCB-derived MSCs retain hepatogenic potential suitable for cell therapy and transplantation against intractable liver diseases.


Sujet(s)
Différenciation cellulaire , Sang foetal/cytologie , Hépatocytes/cytologie , Cellules souches mésenchymateuses/cytologie , Acétylation , Marqueurs biologiques/métabolisme , Cellules cultivées , Sang foetal/métabolisme , Cytométrie en flux , Hépatocytes/métabolisme , Humains , Lipoprotéines LDL/métabolisme , Cellules souches mésenchymateuses/métabolisme , RT-PCR
20.
Stem Cells ; 23(4): 584-93, 2005 Apr.
Article de Anglais | MEDLINE | ID: mdl-15790779

RÉSUMÉ

Mesenchymal stem cells (MSCs) retain both self-renewal and multilineage differentiation capabilities. Despite wide therapeutic potential, many aspects of human MSCs, particularly the molecular parameters to define the stemness, remain largely unknown. Using high-density oligonucleotide micro-arrays, we obtained the differential gene expression profile between a fraction of mononuclear cells of human umbilical cord blood (UCB) and its MSC subpopulation. Of particular interest was a subset of 47 genes preferentially expressed at 50-fold or higher in MSCs, which could be regarded as a molecular foundation of human MSCs. This subset contains numerous genes encoding collagens, other extracellular matrix or related proteins, cytokines or growth factors, and cytoskeleton-associated proteins but very few genes for membrane and nuclear proteins. In addition, a direct comparison of this microarray-generated transcriptome with the published serial analysis of gene expression data suggests that a molecular context of UCB-derived MSCs is more or less similar to that of bone marrow-derived cells. Altogether, our results will provide a basis for studies on molecular mechanisms controlling core properties of human MSCs.


Sujet(s)
Sang foetal/cytologie , Analyse de profil d'expression de gènes , Cellules souches mésenchymateuses/cytologie , Cellules de la moelle osseuse/cytologie , Cellules de la moelle osseuse/métabolisme , Différenciation cellulaire , Lignage cellulaire , Cellules cultivées , Sang foetal/métabolisme , Régulation de l'expression des gènes au cours du développement , Humains , Cellules souches mésenchymateuses/métabolisme , Séquençage par oligonucléotides en batterie , RT-PCR
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...