Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.037
Filtrer
1.
Naunyn Schmiedebergs Arch Pharmacol ; 397(4): 2389-2400, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-37837474

RÉSUMÉ

BACKGROUND: 7,12-Dimethylbenzanthracene (DMBA) is a member of the polycyclic aromatic hydrocarbon family. It is a member of the polycyclic aromatic hydrocarbon family. It is a mutagenic, carcinogenic, and immunosuppressor agent. Cannabidiol (CBD) is a phytocannabinoid. It has anticonvulsant, anti-inflammatory, anti-anxiety, antioxidant, and anti-cancer properties. The purpose of this study was to investigate the possible protective and therapeutic benefits of CBD oil in DMBA-induced leukemia in rats. METHOD: Experimental animals were divided into six groups of five rats each. Group 1 (normal control) included healthy rats. Group 2 included normal rats that received olive oil. Group 3 included normal rats that received CBD. Group 4 included the DMBA-induced leukemic group. Group 5 (prophylactic group) included rats that received CBD as a prophylaxis before IV injection with DMBA. Group 6 (treated group) included DMBA-induced leukemic rats that received CBD as treatment. Liver functions (total, direct and indirect bilirubin, alkaline phosphatase (ALP), alanine transaminase (ALT), aspartate aminotransferase (AST), albumin, globulin, and albumin globulin ratio) were measured. Superoxide dismutase (SOD) and catalase (CAT) were also measured. Total RNA extraction followed by-real time qRT-PCR gene expression of LC3-II, Beclin, mTOR, and P62 was performed. Histopathological examination of liver and spleen tissues was performed. RESULTS: Administration of CBD in groups 5 and 6 resulted in a significant improvement of the levels of liver functions compared to the leukemic untreated rats. Also, the levels of catalase and SOD significantly increased after treatment with CBD compared to the leukemic group. After treatment with CBD in groups 5 and 6, there were downregulations in the expression of all studied genes compared to leukemic untreated rats. Treatment with CBD was more statistically effective than prophylactic use. CONCLUSION: Administration of CBD resulted in a significant improvement in the biochemical, antioxidant status, morphological, and molecular measures in DMBA-induced leukemia in adult male rats. The therapeutic use was more effective than the prophylactic one.


Sujet(s)
Cannabidiol , Globulines , Leucémie expérimentale , Rats , Mâle , Animaux , Antioxydants/pharmacologie , Catalase/métabolisme , 7,12-Diméthyl-benzo[a]anthracène/métabolisme , 7,12-Diméthyl-benzo[a]anthracène/pharmacologie , Leucémie expérimentale/traitement médicamenteux , Leucémie expérimentale/métabolisme , Leucémie expérimentale/anatomopathologie , Foie , Globulines/métabolisme , Globulines/pharmacologie , Superoxide dismutase/métabolisme , Albumines/métabolisme
2.
Virol Sin ; 38(3): 429-439, 2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-37172825

RÉSUMÉ

Erythroleukemia belongs to acute myeloid leukemia (AML) type 6 (M6), and treatment remains difficult due to the poor prognosis of the disease. Friend virus (FV) is a complex of two viruses: Friend murine leukemia virus (F-MuLV) strain along with a defective spleen focus-forming virus (SFFV), which can induce acute erythroleukemia in mice. We have previously reported that activation of vagal α7 nicotinic acetylcholine receptor (nAChR) signaling promotes HIV-1 transcription. Whether vagal muscarinic signaling mediates FV-induced erythroleukemia and the underlying mechanisms remain unclear. In this study, sham and vagotomized mice were intraperitoneally injected with FV. FV infection caused anemia in sham mice, and vagotomy reversed this change. FV infection increased erythroblasts ProE, EryA, and EryB cells in the spleen, and these changes were blocked by vagotomy. In bone marrow, FV infection reduced EryC cells in sham mice, an effect that was counteracted by vagotomy. FV infection increased choline acetyltransferase (ChAT) expression in splenic CD4+ and CD8+ T cells, and this change was reversed by vagotomy. Furthermore, the increase of EryA and EryB cells in spleen of FV-infected wild-type mice was reversed after deletion of ChAT in CD4+ T cells. In bone marrow, FV infection reduced EryB and EryC cells in sham mice, whereas lack of ChAT in CD4+ T cells did not affect this change. Activation of muscarinic acetylcholine receptor 4 (mAChR4) by clozapine N-oxide (CNO) significantly increased EryB in the spleen but decreased the EryC cell population in the bone marrow of FV-infected mice. Thus, vagal-mAChR4 signaling in the spleen and bone marrow synergistically promotes the pathogenesis of acute erythroleukemia. We uncover an unrecognized mechanism of neuromodulation in erythroleukemia.


Sujet(s)
Leucémie érythroblastique aigüe , Leucémie expérimentale , Souris , Animaux , Virus de la leucémie murine de Friend/physiologie , Lymphocytes T CD8+ , Transduction du signal , Leucémie expérimentale/anatomopathologie
3.
Leukemia ; 36(1): 68-79, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34321607

RÉSUMÉ

Despite recent advances in therapeutic approaches, patients with MLL-rearranged leukemia still have poor outcomes. Here, we find that the RNA-binding protein IGF2BP3, which is overexpressed in MLL-translocated leukemia, strongly amplifies MLL-Af4-mediated leukemogenesis. Deletion of Igf2bp3 significantly increases the survival of mice with MLL-Af4-driven leukemia and greatly attenuates disease, with a minimal impact on baseline hematopoiesis. At the cellular level, MLL-Af4 leukemia-initiating cells require Igf2bp3 for their function in leukemogenesis. At the molecular level, IGF2BP3 regulates a complex posttranscriptional operon governing leukemia cell survival and proliferation. IGF2BP3-targeted mRNA transcripts include important MLL-Af4-induced genes, such as those in the Hoxa locus, and the Ras signaling pathway. Targeting of transcripts by IGF2BP3 regulates both steady-state mRNA levels and, unexpectedly, pre-mRNA splicing. Together, our findings show that IGF2BP3 represents an attractive therapeutic target in this disease, providing important insights into mechanisms of posttranscriptional regulation in leukemia.


Sujet(s)
Carcinogenèse/anatomopathologie , Protéines de liaison à l'ADN/génétique , Régulation de l'expression des gènes dans la leucémie , Histone-lysine N-methyltransferase/génétique , Leucémie expérimentale/anatomopathologie , Protéine de la leucémie myéloïde-lymphoïde/génétique , Protéines nucléaires/génétique , Protéines de fusion oncogènes/génétique , Protéines de liaison à l'ARN/physiologie , Animaux , Apoptose , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Prolifération cellulaire , Femelle , Leucémie expérimentale/étiologie , Leucémie expérimentale/métabolisme , Souris , Souris de lignée C57BL , Souris knockout
5.
Cell Rep ; 37(6): 109991, 2021 11 09.
Article de Anglais | MEDLINE | ID: mdl-34758311

RÉSUMÉ

The existence of a dysfunctional CD8+ T cell state in cancer is well established. However, the degree to which CD8+ T cell fates are influenced by the context in which they encounter cognate tumor antigen is less clear. We previously demonstrated that CD8+ T cells reactive to a model leukemia antigen were deleted by antigen cross-presenting type 1 conventional dendritic cells (cDC1s). Here, through a study of T cell receptor (TCR) transgenic CD8+ T cells (TCRTg101) reactive to a native C1498 leukemia cell antigen, we uncover a different mode of T cell tolerance in which TCRTg101 undergo progressive expansion and differentiation into an exhausted state. Antigen encounter by TCRTg101 requires leukemia cell major histocompatibility complex (MHC)-I expression and is independent of DCs, implying that leukemia cells directly mediate the exhausted TCRTg101 phenotype. Collectively, our data reveal that leukemia antigens are presented to CD8+ T cells via discrete pathways, leading to distinct tolerant states.


Sujet(s)
Présentation d'antigène/immunologie , Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Tolérance immunitaire , Leucémie expérimentale/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Animaux , Cellules cultivées , Leucémie expérimentale/métabolisme , Leucémie expérimentale/anatomopathologie , Souris , Souris de lignée C57BL , Souris transgéniques
6.
Sci Rep ; 11(1): 9103, 2021 04 27.
Article de Anglais | MEDLINE | ID: mdl-33907248

RÉSUMÉ

(-)-Epigallocatechin-3-gallate (EGCG), the major active polyphenol extracted from green tea, has been shown to induce apoptosis and inhibit cell proliferation, cell invasion, angiogenesis and metastasis. Herein, we evaluated the in vivo effects of EGCG in acute myeloid leukaemia (AML) using an acute promyelocytic leukaemia (APL) experimental model (PML/RARα). Haematological analysis revealed that EGCG treatment reversed leucocytosis, anaemia and thrombocytopenia, and prolonged survival of PML/RARα mice. Notably, EGCG reduced leukaemia immature cells and promyelocytes in the bone marrow while increasing mature myeloid cells, possibly due to apoptosis increase and cell differentiation. The reduction of promyelocytes and neutrophils/monocytes increase detected in the peripheral blood, in addition to the increased percentage of bone marrow cells with aggregated promyelocytic leukaemia (PML) bodies staining and decreased expression of PML-RAR oncoprotein corroborates our results. In addition, EGCG increased expression of neutrophil differentiation markers such as CD11b, CD14, CD15 and CD66 in NB4 cells; and the combination of all-trans retinoic acid (ATRA) plus EGCG yield higher increase the expression of CD15 marker. These findings could be explained by a decrease of peptidyl-prolyl isomerase NIMA-interacting 1 (PIN1) expression and reactive oxygen species (ROS) increase. EGCG also decreased expression of substrate oncoproteins for PIN1 (including cyclin D1, NF-κB p65, c-MYC, and AKT) and 67 kDa laminin receptor (67LR) in the bone marrow cells. Moreover, EGCG showed inhibition of ROS production in NB4 cells in the presence of N-acetyl-L-cysteine (NAC), as well as a partial blockage of neutrophil differentiation and apoptosis, indicating that EGCG-activities involve/or are in response of oxidative stress. Furthermore, apoptosis of spleen cells was supported by increasing expression of BAD and BAX, parallel to BCL-2 and c-MYC decrease. The reduction of spleen weights of PML/RARα mice, as well as apoptosis induced by EGCG in NB4 cells in a dose-dependent manner confirms this assumption. Our results support further evaluation of EGCG in clinical trials for AML, since EGCG could represent a promising option for AML patient ineligible for current mainstay treatments.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Catéchine/analogues et dérivés , Leucémie aiguë promyélocytaire/traitement médicamenteux , NIMA-interacting peptidylprolyl isomerase/métabolisme , Espèces réactives de l'oxygène/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Catéchine/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Humains , Leucémie expérimentale/traitement médicamenteux , Leucémie expérimentale/mortalité , Leucémie expérimentale/anatomopathologie , Leucémie aiguë promyélocytaire/métabolisme , Leucémie aiguë promyélocytaire/anatomopathologie , Souris transgéniques , Récepteur alpha de l'acide rétinoïque/génétique , Rate/effets des médicaments et des substances chimiques , Rate/anatomopathologie , Protéine Bax/métabolisme , Protéine Bad/métabolisme
7.
Leukemia ; 35(8): 2274-2284, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33526858

RÉSUMÉ

Despite significant progress over the last few decades in the treatment of acute myeloid leukemia (AML), there still remains a major unmet medical need for this disease. Immunotherapy approaches for redirecting pan CD3+ T cells to target leukemia blasts have shown limited efficacy in clinical trials and often accompanied with severe toxicity in AML patients. We designed an alternative engager molecule (Anti-TRGV9/anti-CD123), a bispecific antibody that can simultaneously bind to the Vγ9 chain of the Vγ9Vδ2+ γδ T cell receptor and to AML target antigen, CD123, to selectively recruit Vγ9+ γδ T cells rather than pan T cells to target AML blasts. Our results suggest that prototypic bispecific antibodies (a) selectively activate Vγ9+ γδ T cells as judged by CD69 and CD25 surface expression, and intracellular Granzyme B expression, (b) selectively recruit Vγ9+ γδ T cells into cell-cell conjugate formation of γδ T cells with tumor cells indicating selective and effective engagement of effector and target tumor cells, and (c) mediate γδ T cell cytotoxicity (in vitro and in vivo) against tumor antigen-expressing cells. Collectively, these findings suggest that selectively redirecting Vγ9+ γδ T cells to target AML blasts has a potential for immunotherapy for AML patients and favors further exploration of this concept.


Sujet(s)
Anticorps bispécifiques/immunologie , Antinéoplasiques immunologiques/pharmacologie , Immunothérapie/méthodes , Leucémie expérimentale/traitement médicamenteux , Leucémie aigüe myéloïde/traitement médicamenteux , Récepteur lymphocytaire T antigène, gamma-delta/immunologie , Animaux , Cytotoxicité immunologique , Humains , Leucémie expérimentale/immunologie , Leucémie expérimentale/anatomopathologie , Leucémie aigüe myéloïde/immunologie , Leucémie aigüe myéloïde/anatomopathologie , Souris , Souris de lignée NOD , Souris SCID , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Leukemia ; 35(8): 2243-2257, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33414484

RÉSUMÉ

Targeted T cell therapy is highly effective in disease settings where tumor antigens are uniformly expressed on malignant cells and where off-tumor on-target-associated toxicity is manageable. Although acute myeloid leukemia (AML) has in principle been shown to be a T cell-sensitive disease by the graft-versus-leukemia activity of allogeneic stem cell transplantation, T cell therapy has so far failed in this setting. This is largely due to the lack of target structures both sufficiently selective and uniformly expressed on AML, causing unacceptable myeloid cell toxicity. To address this, we developed a modular and controllable MHC-unrestricted adoptive T cell therapy platform tailored to AML. This platform combines synthetic agonistic receptor (SAR) -transduced T cells with AML-targeting tandem single chain variable fragment (scFv) constructs. Construct exchange allows SAR T cells to be redirected toward alternative targets, a process enabled by the short half-life and controllability of these antibody fragments. Combining SAR-transduced T cells with the scFv constructs resulted in selective killing of CD33+ and CD123+ AML cell lines, as well as of patient-derived AML blasts. Durable responses and persistence of SAR-transduced T cells could also be demonstrated in AML xenograft models. Together these results warrant further translation of this novel platform for AML treatment.


Sujet(s)
Immunothérapie adoptive/méthodes , Leucémie expérimentale/thérapie , Leucémie aigüe myéloïde/thérapie , Récepteurs aux antigènes des cellules T/immunologie , Lymphocytes T/transplantation , Animaux , Femelle , Humains , Leucémie expérimentale/immunologie , Leucémie expérimentale/anatomopathologie , Leucémie aigüe myéloïde/immunologie , Leucémie aigüe myéloïde/anatomopathologie , Souris , Souris de lignée NOD , Souris SCID , Lymphocytes T/immunologie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
10.
J Ethnopharmacol ; 267: 113519, 2021 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-33137432

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Astragalus glycyphyllos L. has been extensively used in Bulgarian folk medicine as an antihypertensive, diuretic, anti-inflammatory, anti-tumour, in cases of cardiac insufficiency, renal inflammation, calculosis, etc. AIM OF THE STUDY: To evaluate the possible in vitro/in vivo anti-proliferative/anti-tumour activity of a purified saponins' mixture (PSM) obtained from the plant. MATERIALS AND METHODS: Viability and proliferative activity of the Graffi myeloid tumour cells was assessed by MTT test. The morphological alterations were visualized and analysed by fluorescent microscopy after intravital double staining. An in vivo model of Graffi tumour bearing hamsters was used to examine the influence of PSM on transplantability, tumour growth, survival and mortality as well as to observe pathomorphological changes. RESULTS: Graffi tumour cells were sensitive to purified saponins' mixture after 24 and 48 h treatment. The treatment induced a statistically significant decrease of the viability/proliferation of the Graffi tumour cells. These effects were concentration- and time-dependent. Fluorescent microscopy studies showed that these antiproliferative effects were connected to the induction of apoptosis. The in vivo study showed the presence of a stromal component, single mononuclear cells in the stroma. Multiple incorrect mitotic figures were observed in the tumour tissue from the control group. Well-formed stroma with accumulation of mononuclear cells and mitotic cells were found in the group, treated with PSM. The tumour weight was decreased in the group, treated with PMS. CONCLUSION: The results indicate that PSM exhibited in vitro/in vivo antiproliferative/anti-tumour effects.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Astragalus , Prolifération cellulaire/effets des médicaments et des substances chimiques , Leucémie expérimentale/traitement médicamenteux , Extraits de plantes/pharmacologie , Saponines/pharmacologie , Animaux , Antinéoplasiques d'origine végétale/isolement et purification , Apoptose/effets des médicaments et des substances chimiques , Astragalus/composition chimique , Cricetinae , Femelle , Virus de la leucémie murine/pathogénicité , Leucémie expérimentale/anatomopathologie , Leucémie expérimentale/virologie , Mâle , Mesocricetus , Extraits de plantes/isolement et purification , Culture de cellules primaires , Infections à Retroviridae/virologie , Saponines/isolement et purification , Charge tumorale/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture , Infections à virus oncogènes/virologie
11.
Anticancer Drugs ; 32(1): 61-65, 2021 01 01.
Article de Anglais | MEDLINE | ID: mdl-32701559

RÉSUMÉ

The pharmacologically active metabolite of cyclophosphamide is aldophosphamide. With cysteine, aldophosphamide forms stable aldophosphamide-thiazolidine which under physiological pH and temperature conditions hydrolyzes to aldophosphamide and cysteine. Aldophosphamide-thiazolidine was synthesized and tested for its ability as a cytostatic. The LD50 after a single intraperitoneal injection in mice was determined to be 2162 mg/kg, but after intravenous bolus administration of 500 mg/kg or in chronic toxicity tests with daily intraperitoneal injections, neurological side effects were observed. Antitumor activity was determined in therapy experiments in CD2F1 mice bearing subcutaneously transplanted P388 mouse leukemia cells. Administration of 100 mg/kg (less than 5% LD50) days 1-5 after tumor transplantation yielded an ILS of 100%. Organ distribution studies showed that aldophosphamide-thiazolidine is evenly distributed in all tissues examined, including brain tissue. The possibilities to increase the antitumor activity of aldophosphamide-thiazolidine by modulating the alkylating function are discussed.


Sujet(s)
Barrière hémato-encéphalique/métabolisme , Cytostatiques/pharmacologie , Leucémie expérimentale/traitement médicamenteux , Moutardes à l'azote/pharmacologie , Thiazolidines/pharmacologie , Animaux , Apoptose , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Prolifération cellulaire , Cytostatiques/pharmacocinétique , Femelle , Leucémie expérimentale/métabolisme , Leucémie expérimentale/anatomopathologie , Souris , Moutardes à l'azote/pharmacocinétique , Thiazolidines/pharmacocinétique , Distribution tissulaire , Cellules cancéreuses en culture
13.
Exp Cell Res ; 397(2): 112368, 2020 12 15.
Article de Anglais | MEDLINE | ID: mdl-33220260

RÉSUMÉ

Mixed lineage leukemia (MLL) arises from several KMT2A-gene chromosomal translocations. Shb gene deficiency has been found to exhibit pleiotropic effects in different models of leukemia, and consequently, this study aimed to investigate MLL-AF9-induced leukemia in Shb deficiency. Bone marrow cells from wild type and Shb knockout (KO) mice were transduced with the MLL-AF9 gene. Shb KO MLL-AF9 cells proliferated at an increased rate, exhibited altered expression of certain cytokine genes (Kitl, Csf3, IL6, IL1b) and higher expression of cell cycle genes (Ccnd2, Ccne1). Mice receiving Shb KO MLL-AF9 cells showed longer latency without displaying any difference in rates of leukemic cell proliferation, indicating a dichotomy between the in vitro and in vivo phenotypes. The mice with Shb deficient MLL-AF9 cells had a lower content of leukemic bone marrow cells allowing elevated normal hematopoiesis, explaining the longer latency. Finally, Shb knockout GFP-positive bone marrow cells showed a higher percentage of cells expressing myeloid markers. The result suggests a role of Shb in the progression of leukemia and that the relevance of the Shb gene is context-dependent as inferred from the differences between the in vivo and in vitro responses. These findings help to obtain an increased understanding of human MLL-AF9 leukemia.


Sujet(s)
Prolifération cellulaire , Transformation cellulaire néoplasique/anatomopathologie , Régulation de l'expression des gènes dans la leucémie , Leucémie expérimentale/anatomopathologie , Protéine de la leucémie myéloïde-lymphoïde/génétique , Protéines de fusion oncogènes/génétique , Protéines proto-oncogènes/physiologie , Animaux , Apoptose , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Humains , Leucémie expérimentale/génétique , Leucémie expérimentale/métabolisme , Souris , Souris de lignée BALB C , Souris knockout , Cellules cancéreuses en culture
14.
Blood ; 136(18): 2003-2017, 2020 10 29.
Article de Anglais | MEDLINE | ID: mdl-32911536

RÉSUMÉ

The majority of childhood leukemias are precursor B-cell acute lymphoblastic leukemias (pB-ALLs) caused by a combination of prenatal genetic predispositions and oncogenic events occurring after birth. Although genetic predispositions are frequent in children (>1% to 5%), fewer than 1% of genetically predisposed carriers will develop pB-ALL. Although infectious stimuli are believed to play a major role in leukemogenesis, the critical determinants are not well defined. Here, by using murine models of pB-ALL, we show that microbiome disturbances incurred by antibiotic treatment early in life were sufficient to induce leukemia in genetically predisposed mice, even in the absence of infectious stimuli and independent of T cells. By using V4 and full-length 16S ribosomal RNA sequencing of a series of fecal samples, we found that genetic predisposition to pB-ALL (Pax5 heterozygosity or ETV6-RUNX1 fusion) shaped a distinct gut microbiome. Machine learning accurately (96.8%) predicted genetic predisposition using 40 of 3983 amplicon sequence variants as proxies for bacterial species. Transplantation of either wild-type (WT) or Pax5+/- hematopoietic bone marrow cells into WT recipient mice revealed that the microbiome is shaped and determined in a donor genotype-specific manner. Gas chromatography-mass spectrometry (GC-MS) analyses of sera from WT and Pax5+/- mice demonstrated the presence of a genotype-specific distinct metabolomic profile. Taken together, our data indicate that it is a lack of commensal microbiota rather than the presence of specific bacteria that promotes leukemia in genetically predisposed mice. Future large-scale longitudinal studies are required to determine whether targeted microbiome modification in children predisposed to pB-ALL could become a successful prevention strategy.


Sujet(s)
Prédisposition aux maladies , Dysbiose/complications , Fèces/microbiologie , Microbiome gastro-intestinal , Leucémie expérimentale/prévention et contrôle , Protéine activatrice spécifique des lymphocytes B/physiologie , Leucémie-lymphome lymphoblastique à précurseurs B/prévention et contrôle , Animaux , Femelle , Leucémie expérimentale/génétique , Leucémie expérimentale/microbiologie , Leucémie expérimentale/anatomopathologie , Souris , Souris de lignée C57BL , Souris knockout , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/microbiologie , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie
15.
FASEB J ; 34(7): 8843-8857, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32433826

RÉSUMÉ

Drug resistance is a common obstacle in leukemia treatment and failing to eradicate leukemia stem cells is the main cause of leukemia relapse. Previous studies have demonstrated that telomerase activity is associated with deregulated self-renewal of leukemia stem cells (LSCs). Here, we identified a novel compound IX, an imatinib derivative with a replacement fragment of a telomerase inhibitor, which can effectively eradicate LSCs but had no influence on normal hematopoietic stem cells (HSCs) survival. We showed that compound IX can decrease the viability of drug-resistant K562/G cells and blast crisis CML primary patient cells. Besides, IX can affect LSC survival, inhibit the colony-forming ability, and reduce LSC frequency. In vivo results showed that IX can relieve the tumor burden in patient-derived xenograft (PDX) model and prolong the lifespan. We observed that compound IX can not only decrease telomerase activity, but also affect the alternative lengthening of telomeres. In addition, IX can inhibit both the canonical and non-canonical Wnt pathways. Our data suggested this novel compound IX as a promising candidate for drug-resistant leukemia therapy.


Sujet(s)
Carcinogenèse/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques , Leucémie expérimentale/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie aigüe myéloïde/traitement médicamenteux , Bibliothèques de petites molécules/pharmacologie , Télomère/effets des médicaments et des substances chimiques , Apoptose , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Cycle cellulaire , Mouvement cellulaire , Prolifération cellulaire , Humains , Leucémie expérimentale/métabolisme , Leucémie expérimentale/anatomopathologie , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Préparations pharmaceutiques/administration et posologie , Télomère/métabolisme , Cellules cancéreuses en culture
16.
Methods Mol Biol ; 2041: 183-195, 2020.
Article de Anglais | MEDLINE | ID: mdl-31646489

RÉSUMÉ

ATP is one of the main components of the tumor microenvironment, where it affects cell growth, tumor progression and antitumor immune response. The development of the pmeLUC probe, a luciferase engineered to be expressed on the outer facet of the plasma membrane, allowed real-time measurement of extracellular ATP in vitro and in vivo systems, among which the tumor microenvironment. Here we describe the experimental procedures to measure extracellular ATP levels in the tumor microenvironment of three different cancer models generated by the implant of pmeLUC-expressing tumor cells into the appropriate mice strain: ACN human neuroblastoma (nude/nude mice host), WEHI-3B murine leukemia (BALB/c host), and B16F10 murine melanoma (C57Bl/6 host). The procedure to obtain stable expression of pmeLUC in different cell types and methods for the measurement of extracellular ATP with pmeLUC in vitro are also described.


Sujet(s)
Adénosine triphosphate/métabolisme , Techniques de biocapteur/méthodes , Leucémie expérimentale/métabolisme , Luciferases/métabolisme , Mélanome expérimental/métabolisme , Neuroblastome/métabolisme , Microenvironnement tumoral/physiologie , Animaux , Membrane cellulaire/métabolisme , Humains , Leucémie expérimentale/anatomopathologie , Mélanome expérimental/anatomopathologie , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris nude , Neuroblastome/anatomopathologie , Cellules cancéreuses en culture
17.
Sci Adv ; 5(7): eaaw5096, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-31281894

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell therapy for hematologic malignancies is fraught with several unknowns, including number of functional T cells that engage target tumor, durability and subsequent expansion and contraction of that engagement, and whether toxicity can be managed. Non-invasive, serial imaging of CAR T cell therapy using a reporter transgene can address those issues quantitatively. We have transduced anti-CD19 CAR T cells with the prostate-specific membrane antigen (PSMA) because it is a human protein with restricted normal tissue expression and has an expanding array of positron emission tomography (PET) and therapeutic radioligands. We demonstrate that CD19-tPSMA(N9del) CAR T cells can be tracked with [18F]DCFPyL PET in a Nalm6 model of acute lymphoblastic leukemia. Divergence between the number of CD19-tPSMA(N9del) CAR T cells in peripheral blood and bone marrow and those in tumor was evident. These findings underscore the need for non-invasive repeatable monitoring of CAR T cell disposition clinically.


Sujet(s)
Antigènes de surface/métabolisme , Glutamate carboxypeptidase II/métabolisme , Immunothérapie adoptive , Tomographie par émission de positons/méthodes , Leucémie-lymphome lymphoblastique à précurseurs B et T/imagerie diagnostique , Animaux , Antigènes CD19/métabolisme , Antigènes de surface/génétique , Glutamate carboxypeptidase II/génétique , Humains , Leucémie expérimentale/imagerie diagnostique , Leucémie expérimentale/anatomopathologie , Lysine/analogues et dérivés , Souris de lignée NOD , Récepteurs aux antigènes des cellules T/métabolisme , Lymphocytes T/physiologie , Urée/analogues et dérivés
18.
Nat Commun ; 10(1): 2189, 2019 05 16.
Article de Anglais | MEDLINE | ID: mdl-31097698

RÉSUMÉ

Improvement in survival has been achieved for children and adolescents with AML but is largely attributed to enhanced supportive care as opposed to the development of better treatment regimens. High risk subtypes continue to have poor outcomes with event free survival rates <40% despite the use of high intensity chemotherapy in combination with hematopoietic stem cell transplant. Here we combine high-throughput screening, intracellular accumulation assays, and in vivo efficacy studies to identify therapeutic strategies for pediatric AML. We report therapeutics not currently used to treat AML, gemcitabine and cabazitaxel, have broad anti-leukemic activity across subtypes and are more effective relative to the AML standard of care, cytarabine, both in vitro and in vivo. JAK inhibitors are selective for acute megakaryoblastic leukemia and significantly prolong survival in multiple preclinical models. Our approach provides advances in the development of treatment strategies for pediatric AML.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Désoxycytidine/analogues et dérivés , Inhibiteurs des Janus kinases/pharmacologie , Leucémie expérimentale/traitement médicamenteux , Leucémie aigüe myéloïde/traitement médicamenteux , Adulte , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Moelle osseuse/anatomopathologie , Moelle osseuse/effets des radiations , Transplantation de moelle osseuse , Lignée cellulaire tumorale , Enfant , Enfant d'âge préscolaire , Cytarabine/pharmacologie , Cytarabine/usage thérapeutique , Désoxycytidine/pharmacologie , Désoxycytidine/usage thérapeutique , Survie sans rechute , Femelle , Tests de criblage à haut débit/méthodes , Humains , Nourrisson , Inhibiteurs des Janus kinases/usage thérapeutique , Leucémie expérimentale/étiologie , Leucémie expérimentale/mortalité , Leucémie expérimentale/anatomopathologie , Leucémie aigüe myéloïde/mortalité , Leucémie aigüe myéloïde/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Taxoïdes/pharmacologie , Taxoïdes/usage thérapeutique , Irradiation corporelle totale/effets indésirables , Tests d'activité antitumorale sur modèle de xénogreffe , Jeune adulte , Gemcitabine
19.
Cancer ; 125(16): 2818-2828, 2019 08 15.
Article de Anglais | MEDLINE | ID: mdl-31034603

RÉSUMÉ

BACKGROUND: Tyrosine kinase inhibitors (TKIs) are the standard treatment for chronic myeloid leukemia (CML). Despite their clinical success, TKIs are faced with challenges such as treatment resistance, which may be driven by kinase domain mutations, and frequent disease relapse upon the cessation of treatment. The combination of arsenic trioxide (ATO) and interferon-α (IFN) was previously demonstrated to inhibit proliferation and induce apoptosis in CML cell lines, prolong the survival of primary wild-type CML mice, and dramatically decrease the activity of leukemia-initiating cells (LICs). METHODS: The ATO/IFN combination was tested in vitro on imatinib (IMN)-resistant K562-R and Ar230-R cells. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling assays were used to evaluate proliferation and apoptosis, respectively. The acridine orange assay was used to assess autophagy, and quantitative reverse transcription-polymerase chain reaction was used to assess the involvement of the hedgehog (Hh) pathway. In vivo, a retroviral transduction/transplantation T315I BCR-ABL CML mouse model was used to assay the effect of the treatment on survival, tumor burden (histopathology and blood counts), and LIC activity (secondary transplantation). RESULTS: In vitro, ATO/IFN synergized to inhibit proliferation and induce apoptosis of IMN-resistant cells with variant modes of resistance. Furthermore, the preclinical effects of ATO/IFN were associated with induction of autophagy along with inhibition of the Hh pathway. Most remarkably, ATO/IFN significantly prolonged the survival of primary T315I-CML mice and displayed a dramatic impairment of disease engraftment in secondary mice, which reflected decreased LIC activity. CONCLUSIONS: Collectively, the ATO/IFN strategy has been demonstrated to have the potential to lead to durable remissions in TKI-resistant CML preclinical models and to overcome various TKI-specific mechanisms of resistance.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Leucémie expérimentale/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Trioxyde d'arsenic/administration et posologie , Autophagie/effets des médicaments et des substances chimiques , Protéines de fusion bcr-abl/métabolisme , Protéines Hedgehog/métabolisme , Humains , Mésilate d'imatinib/pharmacologie , Interféron alpha/administration et posologie , Leucémie expérimentale/anatomopathologie , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Souris de lignée BALB C , Souris transgéniques
20.
Neoplasia ; 21(5): 469-481, 2019 05.
Article de Anglais | MEDLINE | ID: mdl-30974389

RÉSUMÉ

Leukemogenic potential of MLL fusion with the coiled-coil domain-containing partner genes and the downstream target genes of this type of MLL fusion have not been clearly investigated. In this study, we demonstrated that the coiled-coil-four-helix bundle structure of EB1 that participated in the MLL/EB1 was required for immortalizing mouse bone marrow (BM) cells and producing myeloid, but not lymphoid, cell lines. Compared to MLL/AF10, MLL/EB1 had low leukemogenic ability. The MLL/EB1 cells grew more slowly owing to increased apoptosis in vitro and induced acute monocytic leukemia with an incomplete penetrance and longer survival in vivo. A comparative analysis of transcriptome profiling between MLL/EB1 and MLL/AF10 cell lines revealed that there was an at least two-fold difference in the induction of 318 genes; overall, 51.3% (163/318) of the genes were known to be bound by MLL, while 15.4% (49/318) were bound by both MLL and MLL/AF9. Analysis of the 318 genes using Gene Ontology-PANTHER overrepresentation test revealed significant differences in several biological processes, including cell differentiation, proliferation/programmed cell death, and cell homing/recruitment. The Ets1 gene, bound by MLL and MLL/AF9, was involved in several biological processes. We demonstrated that Ets1 was selectively upregulated by MLL/EB1. Short hairpin RNA knockdown of Ets1 in MLL/EB1 cells reduced the expression of CD115, apoptosis rate, competitive engraftment to BM and spleen, and incidence of leukemia and prolonged the survival of the diseased mice. Our results demonstrated that MLL/EB1 upregulated Ets1, which controlled the balance of leukemia cells between apoptosis and BM engraftment/clonal expansion. Novelty and impact of this study The leukemogenic potential of MLL fusion with cytoplasmic proteins containing coiled-coil dimerization domains and the downstream target genes of this type of MLL fusion remain largely unknown. Using a retroviral transduction/transplantation mouse model, we demonstrated that MLL fusion with the coiled-coil-four-helix bundle structure of EB1 has low leukemogenic ability; Ets1, which is upregulated by MLL/EB1, plays a critical role in leukemic transformation by balance between apoptosis and BM engraftment/clonal expansion.


Sujet(s)
Transplantation de moelle osseuse , Transformation cellulaire néoplasique/anatomopathologie , Histone-lysine N-methyltransferase/métabolisme , Leucémie expérimentale/anatomopathologie , Leucémie aigüe monoblastique/anatomopathologie , Protéines associées aux microtubules/métabolisme , Protéine de la leucémie myéloïde-lymphoïde/métabolisme , Protéine proto-oncogène c-ets-1/métabolisme , Animaux , Apoptose , Différenciation cellulaire , Prolifération cellulaire , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Modèles animaux de maladie humaine , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes dans la leucémie , Histone-lysine N-methyltransferase/génétique , Leucémie expérimentale/génétique , Leucémie expérimentale/métabolisme , Leucémie aigüe monoblastique/génétique , Leucémie aigüe monoblastique/métabolisme , Souris , Souris de lignée C57BL , Protéines associées aux microtubules/génétique , Protéine de la leucémie myéloïde-lymphoïde/génétique , Cellules NIH 3T3 , Protéines de fusion oncogènes , Protéine proto-oncogène c-ets-1/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE