Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.615
Filtrer
1.
Molecules ; 29(17)2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39274835

RÉSUMÉ

The cell signaling pathways involved in the antiproliferative activities of T. rosea inner bark remain unexplored. This study evaluated the apoptotic effects of two iridoids from the inner bark of T. rosea and apicidin on THP-1 cells. The cytotoxic effects of the extract and the pure compounds on THP-1 and Jurkat cells were also evaluated using the MTT assay. The apoptotic effect was determined by measuring the mitochondrial membrane potential. The expression of mRNA and MAPK kinase, Bax, and Bcl-2 proteins was detected by Western blotting and RT-qPCR, respectively. The extract and the compounds evaluated increased the percentage of apoptotic cells. Depolarization of the mitochondrial membrane was observed, and the number of cells in the G0/G1 phase increased. Catalposide and specioside significantly increased p38 protein expression, mostly in cells pretreated with apicidin. The p38 MAPK signaling pathway is at least one of the pathways by which the n-butanol extract obtained from Tabebuia rosea, catalposide, and specioside exerts its apoptotic effect on THP-1 cells, and this effect generates a response in the G0/G1 phase and subsequent cell death. In addition, there was depolarization of the mitochondrial membrane, an effect that was related to the participation of the proapoptotic protein Bax.


Sujet(s)
Apoptose , Potentiel de membrane mitochondriale , Écorce , Extraits de plantes , Tabebuia , Humains , Apoptose/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Écorce/composition chimique , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Tabebuia/composition chimique , Peptides cycliques/pharmacologie , Peptides cycliques/composition chimique , Peptides cycliques/isolement et purification , Cellules Jurkat , Leucémies/traitement médicamenteux , Leucémies/métabolisme , Leucémies/anatomopathologie , Butan-1-ol/composition chimique , p38 Mitogen-Activated Protein Kinases/métabolisme , Cellules THP-1 , Antinéoplasiques d'origine végétale/pharmacologie , Antinéoplasiques d'origine végétale/composition chimique , Antinéoplasiques d'origine végétale/isolement et purification , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques
2.
Trends Pharmacol Sci ; 45(10): 919-930, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39306527

RÉSUMÉ

Hematopoietic stem cells (HSCs) and leukemic stem cells (LSCs) are crucial for ensuring hematopoietic homeostasis and driving leukemia progression, respectively. Recent research has revealed that metabolic adaptations significantly regulate the function and survival of these stem cells. In this review, we provide an overview of how metabolic pathways regulate oxidative and proteostatic stresses in HSCs during homeostasis and aging. Furthermore, we highlight targetable metabolic pathways and explore their interactions with epigenetics and the microenvironment in addressing the chemoresistance and immune evasion capacities of LSCs. The metabolic differences between HSCs and LSCs have profound implications for therapeutic strategies.


Sujet(s)
Cellules souches hématopoïétiques , Leucémies , Cellules souches tumorales , Humains , Cellules souches tumorales/métabolisme , Animaux , Leucémies/métabolisme , Leucémies/anatomopathologie , Leucémies/traitement médicamenteux , Cellules souches hématopoïétiques/métabolisme , Épigenèse génétique , Stress oxydatif , Voies et réseaux métaboliques
3.
Nat Commun ; 15(1): 8070, 2024 Sep 14.
Article de Anglais | MEDLINE | ID: mdl-39277592

RÉSUMÉ

Recent advances in high-resolution mapping of spatial interactions among regulatory elements support the existence of complex topological assemblies of enhancers and promoters known as enhancer-promoter hubs or cliques. Yet, organization principles of these multi-interacting enhancer-promoter hubs and their potential role in regulating gene expression in cancer remain unclear. Here, we systematically identify enhancer-promoter hubs in breast cancer, lymphoma, and leukemia. We find that highly interacting enhancer-promoter hubs form at key oncogenes and lineage-associated transcription factors potentially promoting oncogenesis of these diverse cancer types. Genomic and optical mapping of interactions among enhancer and promoter elements further show that topological alterations in hubs coincide with transcriptional changes underlying acquired resistance to targeted therapy in T cell leukemia and B cell lymphoma. Together, our findings suggest that enhancer-promoter hubs are dynamic and heterogeneous topological assemblies with the potential to control gene expression circuits promoting oncogenesis and drug resistance.


Sujet(s)
Carcinogenèse , Résistance aux médicaments antinéoplasiques , Éléments activateurs (génétique) , Régulation de l'expression des gènes tumoraux , Réseaux de régulation génique , Régions promotrices (génétique) , Humains , Régions promotrices (génétique)/génétique , Éléments activateurs (génétique)/génétique , Résistance aux médicaments antinéoplasiques/génétique , Carcinogenèse/génétique , Femelle , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Oncogènes/génétique , Lignée cellulaire tumorale , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Leucémies/génétique , Leucémies/métabolisme , Lymphomes/génétique , Lymphomes/métabolisme
4.
Mol Biol Rep ; 51(1): 997, 2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39297923

RÉSUMÉ

BACKGROUND: Thiazole derivatives are gaining prominence in cancer research due to their potent anti-cancer effects and multifaceted biological activities. In leukemia research, these compounds are particularly studied for their ability to induce apoptosis, disrupt mitochondrial membrane potential (MMP), and modulate cell signaling pathways. METHODS AND RESULTS: This study investigates the efficacy of 4-Methylthiazole in inducing apoptosis in HL-60 leukemia cells. Apoptosis was quantified via flow cytometry using FITC Annexin V and propidium iodide staining. Mitochondrial disruption was evaluated through alterations in mitochondrial membrane potential (MMP) as measured by the JC-1 assay. The compound significantly disrupted MMP, activated Caspase-3, and induced the release of Cytochrome C, all of which are critical markers of apoptosis (****p < 0.0001, ***p < 0.001, **p < 0.01, *p < 0.05). Additionally, treatment with 4-Methylthiazole markedly reduced CD45 and CD123 surface markers, indicating significant phenotypic alterations in leukemia cells (****p < 0.0001). High-dose treatment with 4-Methylthiazole significantly increased ROS levels, suggesting elevated oxidative stress and the presence of intracellular free radicals, contributing to its cytotoxic effects (*p < 0.05). A significant rise in TNF-α levels was observed post-treatment, indicating a pro-inflammatory response that may further inhibit leukemia cell viability. While IL-6 levels remained unchanged, a dose-dependent decrease in IL-10 levels was noted, suggesting a reduction in immunosuppressive conditions within the tumor microenvironment (*p < 0.05). CONCLUSIONS: Overall, 4-Methylthiazole targets leukemia cells through multiple apoptotic mechanisms and modifies the immune landscape of the tumor microenvironment, enhancing its therapeutic potential. This study highlights the need for further clinical investigation to fully exploit the potential of thiazole derivatives in leukemia treatment.


Sujet(s)
Apoptose , Potentiel de membrane mitochondriale , Mitochondries , Thiazoles , Humains , Apoptose/effets des médicaments et des substances chimiques , Cellules HL-60 , Thiazoles/pharmacologie , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Cytochromes c/métabolisme , Leucémies/traitement médicamenteux , Leucémies/métabolisme , Leucémies/anatomopathologie , Caspase-3/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
5.
Int J Mol Sci ; 25(17)2024 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-39273157

RÉSUMÉ

In the last decade, geopolitical instability across the globe has increased the risk of a large-scale radiological event, when radiation biomarkers would be needed for an effective triage of an irradiated population. Ionizing radiation elicits a complex response in the proteome, genome, and metabolome and hence can be leveraged as rapid and sensitive indicators of irradiation-induced damage. We analyzed the plasma of total-body irradiated (TBI) leukemia patients (n = 24) and nonhuman primates (NHPs; n = 10) before and 24 h after irradiation, and we performed a global metabolomic study aiming to provide plasma metabolites as candidate radiation biomarkers for biological dosimetry. Peripheral blood samples were collected according to the appropriate ethical approvals, and metabolites were extracted and analyzed by liquid chromatography mass spectrometry. We identified an array of metabolites significantly altered by irradiation, including bilirubin, cholesterol, and 18-hydroxycorticosterone, which were detected in leukemia patients and NHPs. Pathway analysis showed overlapping perturbations in steroidogenesis, porphyrin metabolism, and steroid hormone biosynthesis and metabolism. Additionally, we observed dysregulation in bile acid biosynthesis and tyrosine metabolism in the TBI patient cohort. This investigation is, to our best knowledge, among the first to provide valuable insights into a comparison between human and NHP irradiation models. The findings from this study could be leveraged for translational biological dosimetry.


Sujet(s)
Métabolome , Irradiation corporelle totale , Animaux , Humains , Mâle , Femelle , Adulte , Marqueurs biologiques/sang , Adulte d'âge moyen , Leucémies/sang , Leucémies/métabolisme , Macaca mulatta , Rayonnement ionisant , Métabolomique/méthodes
6.
Sci Adv ; 10(35): eado1432, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39196923

RÉSUMÉ

The histone acylation reader eleven-nineteen leukemia (ENL) plays a pivotal role in sustaining oncogenesis in acute leukemias, particularly in mixed-lineage leukemia-rearranged (MLL-r) leukemia. ENL relies on its reader domain to recognize histone lysine acylation promoting oncogenic gene expression and leukemia progression. Here, we report the development of MS41, a highly potent and selective von Hippel-Lindau-recruiting ENL degrader that effectively inhibits the growth of ENL-dependent leukemia cells. MS41-induced ENL degradation reduces the chromatin occupancy of ENL-associated transcription elongation machinery, resulting in the suppression of key oncogenic gene expression programs and the activation of differentiation genes. MS41 is well-tolerated in vivo and substantially suppresses leukemia progression in a xenograft mouse model of MLL-r leukemia. Notably, MS41 also induces the degradation of mutant ENL proteins identified in Wilms' tumors. Our findings emphasize the therapeutic potential of pharmacological ENL degradation for treating ENL-dependent cancers, making MS41 not only a valuable chemical probe but also potential anticancer therapeutic for further development.


Sujet(s)
Évolution de la maladie , Leucémies , Humains , Animaux , Souris , Lignée cellulaire tumorale , Leucémies/génétique , Leucémies/anatomopathologie , Leucémies/traitement médicamenteux , Leucémies/métabolisme , Facteurs d'élongation transcriptionnelle/métabolisme , Facteurs d'élongation transcriptionnelle/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Régulation de l'expression des gènes dans la leucémie/effets des médicaments et des substances chimiques , Protéolyse/effets des médicaments et des substances chimiques , Protéine Von Hippel-Lindau supresseur de tumeur/génétique , Protéine Von Hippel-Lindau supresseur de tumeur/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques
7.
J Med Chem ; 67(17): 15494-15508, 2024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-39196554

RÉSUMÉ

From previous studies, it is evident that metal-organic gold(I) complexes have antiproliferative activities. The aim of this study is not only to find new anticancer agents but also to overcome existing cytostatic resistance in cancer cells. The synthesis and medicinal evaluation of two cationic 1,3-disubstituted gold(I) bis-tetrazolylidene complexes 1 and 2 are reported. To determine apoptosis-inducing properties of the complexes, DNA fragmentation was measured using propidium iodide staining followed by flow cytometry. Gold(I) complex 1 targets explicitly malignant cells, effectively inhibiting their growth and selectively inducing apoptosis without signs of necrosis. Even in cells resistant to common treatments such as doxorubicin, it overcomes multidrug resistance and sensitizes existing drug-resistant cells to common cytostatic drugs. It is assumed that gold(I) complex 1 involves the mitochondrial pathway in apoptosis and targets members of the BCL-2 family, enhancing its potential as a therapeutic agent in cancer treatment.


Sujet(s)
Antinéoplasiques , Apoptose , Multirésistance aux médicaments , Résistance aux médicaments antinéoplasiques , Or , Protéines proto-oncogènes c-bcl-2 , Humains , Apoptose/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-bcl-2/métabolisme , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Multirésistance aux médicaments/effets des médicaments et des substances chimiques , Or/composition chimique , Or/pharmacologie , Lignée cellulaire tumorale , Composés hétérocycliques/pharmacologie , Composés hétérocycliques/composition chimique , Composés hétérocycliques/synthèse chimique , Leucémies/traitement médicamenteux , Leucémies/anatomopathologie , Leucémies/métabolisme , Méthane/analogues et dérivés , Méthane/pharmacologie , Méthane/composition chimique , Complexes de coordination/pharmacologie , Complexes de coordination/composition chimique , Complexes de coordination/synthèse chimique , Doxorubicine/pharmacologie , Tests de criblage d'agents antitumoraux , Relation structure-activité , Prolifération cellulaire/effets des médicaments et des substances chimiques
8.
Eur J Med Chem ; 277: 116734, 2024 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-39094275

RÉSUMÉ

Proteolysis targeting chimeras (PROTAC) are bifunctional chimeric molecules capable of directly degrading binding proteins through the ubiquitin-proteasome pathway. PROTACs have demonstrated significant potential in overcoming drug resistance and targeting previously untreatable targets. However, several limitations still need to be addressed, including their high molecular weight resulting in poor membrane permeability and bioavailability. In this study, we proposed that cancer-targeted penetrating peptides could enhance the cell permeability of PROTACs. We developed 26 novel targeted penetrating peptides for leukemia and lymphoma cells, among which C9C-f(3Bta) and Cyclo-C9C-R exhibited superior membrane permeability, targetability, and stability. By combining C9C-f(3Bta) and Cyclo-C9C-R with IMA-PROTAC, we effectively enhanced the anti-proliferative activity of IMA-PROTAC, facilitated degradation of Bcr-Abl protein in K562 cells, and reduced downstream STAT5 phosphorylation. Furthermore, the combined application promoted cell apoptosis while blocking G1 phase progression. HPLC-MRM-MS revealed that the combination of C9C-f(3Bta) or Cyclo-C9C-R with IMA-PROTAC significantly enhanced intracellular IMA-PROTAC content. In summary, our proof-of-concept study validated the hypothesis that combining PROTACs with targeted penetrating peptides can improve protein degradation efficiency as well as anti-proliferative capabilities.


Sujet(s)
Antinéoplasiques , Apoptose , Prolifération cellulaire , Leucémies , Lymphomes , Protéolyse , Humains , Leucémies/traitement médicamenteux , Leucémies/anatomopathologie , Leucémies/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Protéolyse/effets des médicaments et des substances chimiques , Lymphomes/traitement médicamenteux , Lymphomes/anatomopathologie , Lymphomes/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Découverte de médicament , Tests de criblage d'agents antitumoraux , Relation structure-activité , Peptides de pénétration cellulaire/composition chimique , Peptides de pénétration cellulaire/pharmacologie , Peptides de pénétration cellulaire/synthèse chimique , Structure moléculaire , Cellules K562 , Relation dose-effet des médicaments , Systèmes de délivrance de médicaments
9.
Amino Acids ; 56(1): 47, 2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39060524

RÉSUMÉ

sulfur-containing amino acids have been reported to patriciate in gene regulation, DNA methylation, protein synthesis and other physiological or pathological processes. In recent years, metabolism-related molecules of sulfur-containing amino acids affecting the occurrence, development and treatment of tumors have been implicated in various disorders, especially in leukemia. Here, we summarize current knowledge on the sulfur-containing amino acid metabolism pathway in leukemia and examine ongoing efforts to target this pathway, including treatment strategies targeting (a) sulfur-containing amino acids, (b) metabolites of sulfur-containing amino acids, and (c) enzymes and cofactors related to sulfur-containing amino acid metabolism in leukemia. Future leukemia therapy will likely involve innovative strategies targeting the sulfur-containing amino acid metabolism pathway.


Sujet(s)
Leucémies , Humains , Leucémies/métabolisme , Leucémies/traitement médicamenteux , Leucémies/génétique , Soufre/métabolisme , Animaux , Acides aminés/métabolisme , Acides aminés soufrés/métabolisme , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie
10.
Biomolecules ; 14(7)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-39062461

RÉSUMÉ

Leukemias are cancers of the blood-forming system, representing a significant challenge in medical science. The development of leukemia cells involves substantial disturbances within the cellular machinery, offering hope in the search for effective selective treatments that could improve the 5-year survival rate. Consequently, the pathophysiological processes within leukemia cells are the focus of critical research. Enzymes such as cystathionine beta-synthase and sulfurtransferases like thiosulfate sulfurtransferase, 3-mercaptopyruvate sulfurtransferase, and cystathionine gamma-lyase play a vital role in cellular sulfur metabolism. These enzymes are essential to maintaining cellular homeostasis, providing robust antioxidant defenses, and supporting cell division. Numerous studies have demonstrated that cancerous processes can alter the expression and activity of these enzymes, uncovering potential vulnerabilities or molecular targets for cancer therapy. Recent laboratory research has indicated that certain leukemia cell lines may exhibit significant changes in the expression patterns of these enzymes. Analysis of the scientific literature and online datasets has confirmed variations in sulfur enzyme function in specific leukemic cell lines compared to normal leukocytes. This comprehensive review collects and analyzes available information on sulfur enzymes in normal and leukemic cell lines, providing valuable insights and identifying new research pathways in this field.


Sujet(s)
Cystéine , Sulfure d'hydrogène , Leucémies , Soufre , Sulfurtransferases , Humains , Sulfure d'hydrogène/métabolisme , Leucémies/métabolisme , Leucémies/anatomopathologie , Cystéine/métabolisme , Soufre/métabolisme , Sulfurtransferases/métabolisme , Animaux
12.
Mol Biol Rep ; 51(1): 807, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39002036

RÉSUMÉ

BACKGROUND: Acute Myeloid Leukemia (AML) is a fast-developing invading cancer that impacts the blood and bone marrow, marked by the rapid proliferation of abnormal white blood cells. Chemotherapeutic agents, a primary treatment for AML, encounter clinical limitations such as poor solubility and low bioavailability. Previous studies have highlighted antibiotics as effective in inducing cancer cell death and potentially preventing metastasis. Besides, insulin is known to activate the PI3K/Akt pathway, often disrupted in cancers, leading to enhanced cell survival and resistance to apoptosis. In light of the above-mentioned points, we examined the anti-cancer impact of antibiotics Ciprofloxacin (CP) and Salinomycin (SAL) and their combination on KG1-a cells in the presence and absence of insulin. METHODS: This was accomplished by exposing KG1-a cells to different doses of CP and SAL alone, in combination, and with or without insulin for 24-72 h. Cell viability was evaluated using the MTT assay. Besides, apoptotic effects were examined using Hoechst staining and Annexin-V/PI flow cytometry. The expression levels of Bax, p53, BIRC5, Akt, PTEN, and FOXO1 were analyzed through Real-Time PCR. RESULTS: CP and SAL demonstrated cytotoxic and notable pro-apoptotic impact on KG1-a cells by upregulating Bax and p53 and downregulating BIRC5, leading to G0/G1 cell cycle arrest and prevention of the PI3K-Akt signaling pathway. Our findings demonstrated that combination of CP and SAL promote apoptosis in the KG1-a cell line by down-regulating BIRC5 and Akt, as well as up-regulating Bax, p53, PTEN, and FOXO1. Additionally, the findings strongly indicated that insulin effectively mitigates apoptosis by enhancing Akt expression and reducing FOXO1 and PTEN gene expression in the cells treated with CP and SAL. CONCLUSION: Our findings showed that the combined treatment of CP and SAL exhibit a strong anti-cancer effect on leukemia KG1-a cells. Moreover, it was discovered that the PI3K-Akt signaling can be a promising target in leukemia treatment particularly in hyperinsulinemia condition.


Sujet(s)
Apoptose , Survie cellulaire , Ciprofloxacine , Insuline , Pyrannes , Humains , Ciprofloxacine/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Pyrannes/pharmacologie , Lignée cellulaire tumorale , Insuline/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéine O1 à motif en tête de fourche/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Leucémies/traitement médicamenteux , Leucémies/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Polyether Polyketides
13.
BMC Cancer ; 24(1): 900, 2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39060972

RÉSUMÉ

Leukemia is a type of blood cell cancer that is in the bone marrow's blood-forming cells. Two types of Leukemia are acute and chronic; acute enhances fast and chronic growth gradually which are further classified into lymphocytic and myeloid leukemias. This work evaluates a unique deep convolutional neural network (CNN) classifier that improves identification precision by carefully examining concatenated peptide patterns. The study uses leukemia protein expression for experiments supporting two different techniques including independence and applied cross-validation. In addition to CNN, multilayer perceptron (MLP), gated recurrent unit (GRU), and recurrent neural network (RNN) are applied. The experimental results show that the CNN model surpasses competitors with its outstanding predictability in independent and cross-validation testing applied on different features extracted from protein expressions such as amino acid composition (AAC) with a group of AAC (GAAC), tripeptide composition (TPC) with a group of TPC (GTPC), and dipeptide composition (DPC) for calculating its accuracies with their receiver operating characteristic (ROC) curve. In independence testing, a feature expression of AAC and a group of GAAC are applied using MLP and CNN modules, and ROC curves are achieved with overall 100% accuracy for the detection of protein patterns. In cross-validation testing, a feature expression on a group of AAC and GAAC patterns achieved 98.33% accuracy which is the highest for the CNN module. Furthermore, ROC curves show a 0.965% extraordinary result for the GRU module. The findings show that the CNN model is excellent at figuring out leukemia illnesses from protein expressions with higher accuracy.


Sujet(s)
Leucémies , 29935 , Humains , Leucémies/métabolisme , Leucémies/anatomopathologie , Courbe ROC , Peptides/analyse
14.
Sci Rep ; 14(1): 16636, 2024 07 18.
Article de Anglais | MEDLINE | ID: mdl-39025941

RÉSUMÉ

In therapies, curcumin is now commonly formulated in liposomal form, administered through injections or creams. This enhances its concentration at the cellular level compared to its natural form ingestion. Due to its hydrophobic nature, curcumin is situated in the lipid part of the membrane, thereby modifying its properties and influencing processes The aim of the research was to investigate whether the toxicity of specific concentrations of curcumin, assessed through biochemical tests for the SK-N-SH and H-60 cell lines, is related to structural changes in the membranes of these cells, caused by the localization of curcumin in their hydrophobic regions. Biochemical tests were performed using spectrophotometric methods. Langmuir technique were used to evaluate the interaction of the curcumin with the studied lipids. Direct introduction of curcumin into the membranes alters their physicochemical parameters. The extent of these changes depends on the initial properties of the membrane. In the conducted research, it has been demonstrated that curcumin may exhibit toxicity to human cells. The mechanism of this toxicity is related to its localization in cell membranes, leading to their dysfunction. The sensitivity of cells to curcumin presence depends on the saturation level of their membranes; the more rigid the membrane, the lower the concentration of curcumin causes its disruption.


Sujet(s)
Membrane cellulaire , Curcumine , Neuroblastome , Curcumine/pharmacologie , Curcumine/composition chimique , Humains , Membrane cellulaire/effets des médicaments et des substances chimiques , Membrane cellulaire/métabolisme , Neuroblastome/traitement médicamenteux , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Lignée cellulaire tumorale , Leucémies/traitement médicamenteux , Leucémies/métabolisme , Leucémies/anatomopathologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Interactions hydrophobes et hydrophiles
15.
Leukemia ; 38(9): 1918-1928, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38987275

RÉSUMÉ

Selinexor, a first-in-class exportin1 (XPO1) inhibitor, is an attractive anti-tumor agent because of its unique mechanisms of action; however, its dose-dependent toxicity and lack of biomarkers preclude its wide use in clinical applications. To identify key molecules/pathways regulating selinexor sensitivity, we performed genome-wide CRISPR/Cas9 dropout screens using two B-ALL lines. We identified, for the first time, that paralogous DDX19A and DDX19B RNA helicases modulate selinexor sensitivity by regulating MCL1 mRNA nuclear export. While single depletion of either DDX19A or DDX19B barely altered MCL1 protein levels, depletion of both significantly attenuated MCL1 mRNA nuclear export, reducing MCL1 protein levels. Importantly, combining selinexor treatment with depletion of either DDX19A or DDX19B markedly induced intrinsic apoptosis of leukemia cells, an effect rescued by MCL1 overexpression. Analysis of Depmap datasets indicated that a subset of T-ALL lines expresses minimal DDX19B mRNA levels. Moreover, we found that either selinexor treatment or DDX19A depletion effectively induced apoptosis of T-ALL lines expressing low DDX19B levels. We conclude that XPO1 and DDX19A/B coordinately regulate cellular MCL1 levels and propose that DDX19A/B could serve as biomarkers for selinexor treatment. Moreover, pharmacological targeting of DDX19 paralogs may represent a potential strategy to induce intrinsic apoptosis in leukemia cells.


Sujet(s)
DEAD-box RNA helicases , Hydrazines , Protéine Mcl-1 , ARN messager , Triazoles , Triazoles/pharmacologie , Humains , Protéine Mcl-1/métabolisme , Protéine Mcl-1/génétique , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/génétique , Hydrazines/pharmacologie , ARN messager/génétique , Leucémies/métabolisme , Leucémies/traitement médicamenteux , Leucémies/génétique , Leucémies/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Transport nucléaire actif/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/génétique , Antinéoplasiques/pharmacologie
16.
Adv Exp Med Biol ; 1459: 359-378, 2024.
Article de Anglais | MEDLINE | ID: mdl-39017852

RÉSUMÉ

ETS proto-oncogene 1 (ETS1) is a transcription factor (TF) critically involved in lymphoid cell development and function. ETS1 expression is tightly regulated throughout differentiation and activation in T-cells, natural killer (NK) cells, and B-cells. It has also been described as an oncogene in a range of solid and hematologic cancer types. Among hematologic malignancies, its role has been best studied in T-cell acute lymphoblastic leukemia (T-ALL), adult T-cell leukemia/lymphoma (ATLL), and diffuse large B-cell lymphoma (DLBCL). Aberrant expression of ETS1 in these malignancies is driven primarily by chromosomal amplification and enhancer-driven transcriptional regulation, promoting the ETS1 transcriptional program. ETS1 also facilitates aberrantly expressed or activated transcriptional complexes to drive oncogenic pathways. Collectively, ETS1 functions to regulate cell growth, differentiation, signaling, response to stimuli, and viral interactions in these malignancies. A tumor suppressor role has also been indicated for ETS1 in select lymphoma types, emphasizing the importance of cellular context in ETS1 function. Research is ongoing to further characterize the clinical implications of ETS1 dysregulation in hematologic malignancies, to further resolve binding complexes and transcriptional targets, and to identify effective therapeutic targeting approaches.


Sujet(s)
Proto-oncogène Mas , Protéine proto-oncogène c-ets-1 , Humains , Protéine proto-oncogène c-ets-1/métabolisme , Protéine proto-oncogène c-ets-1/génétique , Animaux , Lymphomes/génétique , Lymphomes/métabolisme , Lymphomes/anatomopathologie , Transduction du signal , Régulation de l'expression des gènes dans la leucémie , Régulation de l'expression des gènes tumoraux , Leucémies/génétique , Leucémies/métabolisme , Leucémies/anatomopathologie
18.
Adv Exp Med Biol ; 1459: 405-430, 2024.
Article de Anglais | MEDLINE | ID: mdl-39017854

RÉSUMÉ

HOXA9, an important transcription factor (TF) in hematopoiesis, is aberrantly expressed in numerous cases of both acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) and is a strong indicator of poor prognosis in patients. HOXA9 is a proto-oncogene which is both sufficient and necessary for leukemia transformation. HOXA9 expression in leukemia correlates with patient survival outcomes and response to therapy. Chromosomal transformations (such as NUP98-HOXA9), mutations, epigenetic dysregulation (e.g., MLL- MENIN -LEDGF complex or DOT1L/KMT4), transcription factors (such as USF1/USF2), and noncoding RNA (such as HOTTIP and HOTAIR) regulate HOXA9 mRNA and protein during leukemia. HOXA9 regulates survival, self-renewal, and progenitor cell cycle through several of its downstream target TFs including LMO2, antiapoptotic BCL2, SOX4, and receptor tyrosine kinase FLT3 and STAT5. This dynamic and multilayered HOXA9 regulome provides new therapeutic opportunities, including inhibitors targeting DOT1L/KMT4, MENIN, NPM1, and ENL proteins. Recent findings also suggest that HOXA9 maintains leukemia by actively repressing myeloid differentiation genes. This chapter summarizes the recent advances understanding biochemical mechanisms underlying HOXA9-mediated leukemogenesis, the clinical significance of its abnormal expression, and pharmacological approaches to treat HOXA9-driven leukemia.


Sujet(s)
Régulation de l'expression des gènes dans la leucémie , Protéines à homéodomaine , Nucléophosmine , Proto-oncogène Mas , Humains , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Régulation de l'expression des gènes dans la leucémie/effets des médicaments et des substances chimiques , Animaux , Leucémies/génétique , Leucémies/métabolisme , Leucémies/traitement médicamenteux , Leucémies/anatomopathologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie
19.
Nucleic Acids Res ; 52(16): 9463-9480, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-38989615

RÉSUMÉ

The H3K4 methyltransferase SETD1A plays an essential role in both development and cancer. However, essential components involved in SETD1A chromatin binding remain unclear. Here, we discovered that BOD1L exhibits the highest correlated SETD1A co-dependency in human cancer cell lines. BOD1L knockout reduces leukemia cells in vitro and in vivo, and mimics the transcriptional profiles observed in SETD1A knockout cells. The loss of BOD1L immediately reduced SETD1A distribution at transcriptional start sites (TSS), induced transcriptional elongation defect, and increased the RNA polymerase II content at TSS; however, it did not reduce H3K4me3. The Shg1 domain of BOD1L has a DNA binding ability, and a tryptophan residue (W104) in the domain recruits SETD1A to chromatin through the association with SETD1A FLOS domain. In addition, the BOD1L-SETD1A complex associates with transcriptional regulators, including E2Fs. These results reveal that BOD1L mediates chromatin and SETD1A, and regulates the non-canonical function of SETD1A in transcription.


Sujet(s)
Chromatine , Histone-lysine N-methyltransferase , Histone , Animaux , Humains , Souris , Lignée cellulaire tumorale , Chromatine/métabolisme , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , Histone/métabolisme , Leucémies/génétique , Leucémies/métabolisme , Liaison aux protéines , Domaines protéiques , RNA polymerase II/métabolisme , Site d'initiation de la transcription , Transcription génétique
20.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38928344

RÉSUMÉ

The association between leukemic stem cells (LSCs) and leukemia development has been widely established in the context of genetic alterations, epigenetic pathways, and signaling pathway regulation. Hematopoietic stem cells are at the top of the bone marrow hierarchy and can self-renew and progressively generate blood and immune cells. The microenvironment, niche cells, and complex signaling pathways that regulate them acquire genetic mutations and epigenetic alterations due to aging, a chronic inflammatory environment, stress, and cancer, resulting in hematopoietic stem cell dysregulation and the production of abnormal blood and immune cells, leading to hematological malignancies and blood cancer. Cells that acquire these mutations grow at a faster rate than other cells and induce clone expansion. Excessive growth leads to the development of blood cancers. Standard therapy targets blast cells, which proliferate rapidly; however, LSCs that can induce disease recurrence remain after treatment, leading to recurrence and poor prognosis. To overcome these limitations, researchers have focused on the characteristics and signaling systems of LSCs and therapies that target them to block LSCs. This review aims to provide a comprehensive understanding of the types of hematopoietic malignancies, the characteristics of leukemic stem cells that cause them, the mechanisms by which these cells acquire chemotherapy resistance, and the therapies targeting these mechanisms.


Sujet(s)
Tumeurs hématologiques , Cellules souches tumorales , Humains , Tumeurs hématologiques/génétique , Tumeurs hématologiques/anatomopathologie , Tumeurs hématologiques/métabolisme , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Cellules souches hématopoïétiques/métabolisme , Leucémies/anatomopathologie , Leucémies/génétique , Leucémies/métabolisme , Transduction du signal , Animaux , Microenvironnement tumoral/génétique , Résistance aux médicaments antinéoplasiques/génétique , Épigenèse génétique , Mutation
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE