Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Oncol Rep ; 50(3)2023 Sep.
Article in English | MEDLINE | ID: mdl-37477144

ABSTRACT

Neuroblastoma (NB), the most frequent solid extracranial tumor in children, is not always cured by current aggressive therapies that have notable adverse effects; therefore, novel treatments are necessary. Phosphoinositide 3­kinase (PI3K) and fibroblast growth factor receptor inhibitors exhibit synergistic effect in NB cell lines. In the present study, mono­ and combination therapy of the United States Food and Drug Administration­approved PI3K, cyclin­dependent kinase­4/6 (CDK4/6), poly­ADP­ribose­polymerase (PARP) and WEE1 G2 checkpoint kinase (WEE1) inhibitors (BYL719, PD­0332991, BMN673 and MK­1775, respectively), were used to treat NB cell lines SK­N­AS, SK­N­BE(2)­C, SK­N­DZ, SK­N­FI and SK­N­SH and viability (assessed by WST­1 assay), proliferation (incucyte analysis) and cell cycle (FACS) changes were assessed. Treatments with all single drugs presented dose­-dependent responses with decreased viability and proliferation and combining BYL719 with PD­0332991 or BMN673 with MK­1775 resulted in additive or synergistic effects in most cell lines., except for SK­N­SH for the former and for SK­N­AS for the latter. Moreover, combining MK­1775 and BMN673 decreased the numbers of cells in S phase to a greater extent than either drug alone, while when combining PD­0332991 and BYL719 the observed effect was close to that of PD­0332991 alone. To summarize, PI3K and CDK4/6 or PARP and WEE1 exhibited synergistic anti­NB effects and lower doses of the inhibitors could be utilized, thereby potentially reducing adverse side effects.


Subject(s)
Neuroblastoma , Phosphatidylinositol 3-Kinases , Child , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Phosphatidylinositol 3-Kinase , Cell Line, Tumor , Cell Proliferation , Neuroblastoma/drug therapy , Cell Cycle Proteins/metabolism , Protein-Tyrosine Kinases , Cyclin-Dependent Kinase 4
2.
Blood ; 142(1): 90-105, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37146239

ABSTRACT

RNA-binding proteins (RBPs) form a large and diverse class of factors, many members of which are overexpressed in hematologic malignancies. RBPs participate in various processes of messenger RNA (mRNA) metabolism and prevent harmful DNA:RNA hybrids or R-loops. Here, we report that PIWIL4, a germ stem cell-associated RBP belonging to the RNase H-like superfamily, is overexpressed in patients with acute myeloid leukemia (AML) and is essential for leukemic stem cell function and AML growth, but dispensable for healthy human hematopoietic stem cells. In AML cells, PIWIL4 binds to a small number of known piwi-interacting RNA. Instead, it largely interacts with mRNA annotated to protein-coding genic regions and enhancers that are enriched for genes associated with cancer and human myeloid progenitor gene signatures. PIWIL4 depletion in AML cells downregulates the human myeloid progenitor signature and leukemia stem cell (LSC)-associated genes and upregulates DNA damage signaling. We demonstrate that PIWIL4 is an R-loop resolving enzyme that prevents R-loop accumulation on a subset of AML and LSC-associated genes and maintains their expression. It also prevents DNA damage, replication stress, and activation of the ATR pathway in AML cells. PIWIL4 depletion potentiates sensitivity to pharmacological inhibition of the ATR pathway and creates a pharmacologically actionable dependency in AML cells.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/pathology , Hematopoietic Stem Cells/metabolism , Cell Proliferation , Genomics , RNA, Messenger/metabolism , Neoplastic Stem Cells/pathology
3.
Leukemia ; 36(2): 416-425, 2022 02.
Article in English | MEDLINE | ID: mdl-34462525

ABSTRACT

Acute myeloid leukemia (AML) is considered a poor prognosis malignancy where patients exhibit altered glucose metabolism and stem cell signatures that contribute to AML growth and maintenance. Here, we report that the epigenetic factor, Ten-Eleven Translocation 3 (TET3) dioxygenase is overexpressed in AML patients and functionally validated human leukemic stem cells (LSCs), is required for leukemic growth by virtue of its regulation of glucose metabolism in AML cells. In human AML cells, TET3 maintains 5-hydroxymethylcytosine (5hmC) epigenetic marks and expression of early myeloid progenitor program, critical glucose metabolism and STAT5A signaling pathway genes, which also positively correlate with TET3 expression in AML patients. Consequently, TET3 depletion impedes hexokinase activity and L-Lactate production in AML cells. Conversely, overexpression of TET3 in healthy human hematopoietic stem progenitors (HSPCs) upregulates the expression of glucose metabolism, STAT5A signaling and AML associated genes, and impairs normal HSPC lineage differentiation in vitro. Finally, TET3 depletion renders AML cells highly sensitive to blockage of the TET3 downstream pathways glycolysis and STAT5 signaling via the combination of 2-Deoxy-D-glucose and STAT5 inhibitor which preferentially targets AML cells but spares healthy CD34+ HSPCs.


Subject(s)
Dioxygenases/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Glucose/metabolism , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Animals , Apoptosis , Cell Proliferation , Dioxygenases/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Blood Adv ; 3(22): 3729-3739, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31770439

ABSTRACT

Acute erythroid leukemia (AEL) is a rare and aggressive form of acute leukemia, the biology of which remains poorly understood. Here we demonstrate that the ParaHox gene CDX4 is expressed in patients with acute erythroid leukemia, and that aberrant expression of Cdx4 induced homogenously a transplantable acute erythroid leukemia in mice. Gene expression analyses demonstrated upregulation of genes involved in stemness and leukemogenesis, with parallel downregulation of target genes of Gata1 and Gata2 responsible for erythroid differentiation. Cdx4 induced a proteomic profile that overlapped with a cluster of proteins previously defined to represent the most primitive human erythroid progenitors. Whole-exome sequencing of diseased mice identified recurrent mutations significantly enriched for transcription factors involved in erythroid lineage specification, as well as TP53 target genes partly identical to the ones reported in patients with AEL. In summary, our data indicate that Cdx4 is able to induce stemness and inhibit terminal erythroid differentiation, leading to the development of AEL in association with co-occurring mutations.


Subject(s)
Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/pathology , Adult , Aged , Animals , Biomarkers, Tumor , Cell Differentiation/genetics , Disease Models, Animal , Female , Gene Expression Regulation , Genetic Association Studies , Hematopoiesis/genetics , Humans , Immunophenotyping , Male , Mice , Middle Aged , Mutation , Whole Genome Sequencing
5.
Sci Rep ; 8(1): 989, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29343865

ABSTRACT

Acute leukemia is initiated and maintained by leukemia stem cells (LSCs) and therefore there is great interest to develop innovative therapeutic approaches which target LSCs. Here we show that mesoporous silica nanoparticles (MSNs) functionalized with succinic anhydride, tagged with an anti-B220 antibody and loaded with the anthracycline daunorubicin are efficiently incorporated into murine B220-positive AML LSCs and preferentially kill these cells in comparison to B220-negative AML LSCs in vitro. Furthermore, short - term treatment of the AML LSCs with these MSNs before transplant significantly delayed leukemia development in recipient mice. These data demonstrate that targeting of AML LSCs can be improved by using functionalized and antigen directed MSNs as carriers for anti-leukemic drugs.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antibodies, Monoclonal/chemistry , Daunorubicin/pharmacology , Leukemia, Myeloid, Acute/therapy , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Animals , Antibiotics, Antineoplastic/chemistry , Cell Line, Tumor , Daunorubicin/chemistry , Drug Compounding/methods , Gene Expression , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Nanoparticles/ultrastructure , Neoplasms, Experimental , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Binding , Succinic Anhydrides/chemistry , Tumor Cells, Cultured
6.
Small ; 13(41)2017 11.
Article in English | MEDLINE | ID: mdl-28922574

ABSTRACT

The adsorption of serum proteins, leading to the formation of a biomolecular corona, is a key determinant of the biological identity of nanoparticles in vivo. Therefore, gaining knowledge on the formation, composition, and temporal evolution of the corona is of utmost importance for the development of nanoparticle-based therapies. Here, it is shown that the use of super-resolution optical microscopy enables the imaging of the protein corona on mesoporous silica nanoparticles with single protein sensitivity. Particle-by-particle quantification reveals a significant heterogeneity in protein absorption under native conditions. Moreover, the diversity of the corona evolves over time depending on the surface chemistry and degradability of the particles. This paper investigates the consequences of protein adsorption for specific cell targeting by antibody-functionalized nanoparticles providing a detailed understanding of corona-activity relations. The methodology is widely applicable to a variety of nanostructures and complements the existing ensemble approaches for protein corona study.


Subject(s)
Microscopy/methods , Nanoparticles/chemistry , Protein Corona/chemistry , Adsorption , Animals , Cattle , Kinetics , Porosity , Serum Albumin, Bovine/chemistry , Silicon Dioxide/chemistry , Surface Properties , Time Factors
7.
ACS Appl Mater Interfaces ; 9(22): 18566-18574, 2017 Jun 07.
Article in English | MEDLINE | ID: mdl-28525262

ABSTRACT

The functionalization of nanoparticles with a ligand targeting receptors overexpressed by the target cells is a commonly used strategy when aiming at nanoparticle-based, cell type-specific drug delivery.1-4 However, the influence of particle surface chemistry on the targetability has received much less attention. The surface charge is known to directly or indirectly affect the nanoparticle cellular uptake kinetics by influencing serum protein adsorption.5-7 Thus, it is fair to assume that both the specificity and cellular uptake kinetics of targeted nanoparticles are influenced by the nanoparticle charge, both of which are important parameters for controlling cell-specific drug delivery efficiency. We therefore studied the influence of the surface chemistry of mesoporous silica nanoparticles (MSNs) carrying identical amounts of a specific antibody (anti-B220) on the selectivity toward B220-positive leukemia stem cells. The uptake by these cells was higher compared to the nanoparticle uptake by B220-negative leukemia stem cells, demonstrating uptake specificity. In addition, the adsorption of serum proteins onto the differently charged MSNs was studied by SDS-PAGE. Interestingly, the highest selectivity was not observed for the MSNs with the lowest level of serum protein adsorption, which suggests that proteins present in the protein corona of the MSNs may positively influence the selective uptake of targeted nanoparticles. For the particles exhibiting the highest selectivity, successful selective delivery of cargo to the B220-positive cells was demonstrated. Taken together, our results indicate that nanoparticle surface charge and adsorption of serum proteins is an important factor for enhancing selectivity in targeted delivery of drugs using nanoparticulate vectors, an observation tentatively attributed to enhanced cellular internalization kinetics in the presence of adsorbed serum proteins on the nanoparticles.


Subject(s)
Neoplastic Stem Cells , Adsorption , Blood Proteins , Humans , Leukemia , Nanoparticles , Porosity , Silicon Dioxide
8.
Oncotarget ; 7(52): 86889-86901, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27888632

ABSTRACT

Homeobox genes are key regulators in normal and malignant hematopoiesis. The human Vent-like homeobox gene VENTX, a putative homolog of the Xenopus laevis Xvent-2 gene, was shown to be highly expressed in normal myeloid cells and in patients with acute myeloid leukemia. We now demonstrate that constitutive expression of VENTX suppresses expression of genes responsible for terminal erythroid differentiation in normal CD34+ stem and progenitor cells. Transplantation of bone marrow progenitor cells retrovirally engineered to express VENTX caused massive expansion of primitive erythroid cells and partly acute erythroleukemia in transplanted mice. The leukemogenic potential of VENTX was confirmed in the AML1-ETO transplantation model, as in contrast to AML1-ETO alone co-expression of AML1-ETO and VENTX induced acute myeloid leukemia, partly expressing erythroid markers, in all transplanted mice. VENTX was highly expressed in patients with primary human erythroleukemias and knockdown of VENTX in the erythroleukemic HEL cell line significantly blocked cell growth. In summary, these data indicate that VENTX is able to perturb erythroid differentiation and to contribute to myeloid leukemogenesis when co-expressed with appropriate AML oncogenes and point to its potential significance as a novel therapeutic target in AML.


Subject(s)
Cell Proliferation/genetics , Erythroid Cells/metabolism , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Adult , Aged , Aged, 80 and over , Animals , Cell Differentiation/genetics , Female , Gene Expression Regulation, Leukemic , Homeodomain Proteins/metabolism , Humans , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Myeloid, Acute/metabolism , Male , Mice, Inbred C3H , Mice, Inbred C57BL , Middle Aged , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , RNA Interference
9.
Cell Rep ; 16(2): 498-507, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27346355

ABSTRACT

Homeobox genes are known to be key factors in leukemogenesis. Although the TALE family homeodomain factor Meis1 has been linked to malignancy, a role for MEIS2 is less clear. Here, we demonstrate that MEIS2 is expressed at high levels in patients with AML1-ETO-positive acute myeloid leukemia and that growth of AML1-ETO-positive leukemia depends on MEIS2 expression. In mice, MEIS2 collaborates with AML1-ETO to induce acute myeloid leukemia. MEIS2 binds strongly to the Runt domain of AML1-ETO, indicating a direct interaction between these transcription factors. High expression of MEIS2 impairs repressive DNA binding of AML1-ETO, inducing increased expression of genes such as the druggable proto-oncogene YES1. Collectively, these data describe a pivotal role for MEIS2 in AML1-ETO-induced leukemia.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein/genetics , Transcription Factors/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression , Gene Expression Regulation, Leukemic , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Neoplasm Transplantation , Oncogene Proteins, Fusion/metabolism , Oncogenes , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Mas , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , Transcription Factors/metabolism
10.
BMC Cancer ; 14: 785, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25348805

ABSTRACT

BACKGROUND: The Bmi1 polycomb ring finger oncogene, a transcriptional repressor belonging to the Polycomb group of proteins plays an important role in the regulation of stem cell self-renewal and is elevated in several cancers. In the current study, we have explored the role of Bmi1 in regulating the stemness and drug resistance of breast cancer cells. METHODS: Using real time PCR and immunohistochemistry primary breast tissues were analyzed. Retro- and lentiviruses were utilized to overexpress and knockdown Bmi1, RT-PCR and Western blot was performed to evaluate mRNA and protein expression. Stemness properties were analyzed by flow cytometry and sphere-formation and tumor formation was determined by mouse xenograft experiments. Dual luciferase assay was employed to assess promoter activity and MTT assay was used to analyze drug response. RESULTS: We found Bmi1 overexpression in 64% of grade III invasive ductal breast adenocarcinomas compared to normal breast tissues. Bmi1 overexpression in immortalized and transformed breast epithelial cells increased their sphere-forming efficiency, induced epithelial to mesenchymal transition (EMT) with an increase in the expression of stemness-related genes. Knockdown of Bmi1 in tumorigenic breast cells induced epithelial morphology, reduced expression of stemness-related genes, decreased the IC50 values of doxorubicin and abrogated tumor-formation. Bmi1-high tumors showed elevated Nanog expression whereas the tumors with lower Bmi1 showed reduced Nanog levels. Overexpression of Bmi1 increased Nanog levels whereas knockdown of Bmi1 reduced its expression. Dual luciferase promoter-reporter assay revealed Bmi1 positively regulated the Nanog and NFκB promoter activity. RT-PCR analysis showed that Bmi1 overexpression activated the NFκB pathway whereas Bmi1 knockdown reduced the expression of NFκB target genes, suggesting that Bmi1 might regulate Nanog expression through the NFκB pathway. CONCLUSIONS: Our study showed that Bmi1 is overexpressed in several high-grade, invasive ductal breast adenocarcinomas, thus supporting its role as a prognostic marker. While Bmi1 overexpression increased self-renewal and promoted EMT, its knockdown reversed EMT, reduced stemness, and rendered cells drug sensitive, thus highlighting a crucial role for Bmi1 in regulating the stemness and drug response of breast cancer cells. Bmi1 may control self-renewal through the regulation of Nanog expression via the NFκB pathway.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Homeodomain Proteins/genetics , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Heterografts , Homeodomain Proteins/metabolism , Humans , Mice , Nanog Homeobox Protein , Neoplasm Grading , Polycomb Repressive Complex 1/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects , Tumor Burden/genetics
11.
Food Chem Toxicol ; 48(12): 3449-57, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20854871

ABSTRACT

Antimycin A (AMA) treatment of cells blocks mitochondrial electron transport chain, and leads to elevated ROS generation, thereby causing damage to mtDNA, proteins and lipids, along with mitochondrial membrane depolarization, release of pro-apoptotic proteins into the cytoplasm, and induction of apoptosis. Prevention of such oxidative cellular damage by the aqueous extract of Phyllanthus amarus has been investigated in this study. The extract demonstrated significant potential in mitigating H(2)O(2)-induced membrane damage along with considerable recession in AMA-governed mitochondrial protein and lipid degradation in Hep3B cells. 8-OHdG analysis of mtDNA damage revealed substantial protective potential of the extract against mtDNA damage. SQ-PCR of selected mtDNA sequences confirmed the potential of the extract to alleviate levels of mtDNA damage. FACS analysis with JC-1 fluorescent dye established significant escalation of mitochondrial membrane potential by the extract in AMA-treated cells. Extract treatment resulted in a distinct decline in the degrees of AMA-induced release of cytochrome c and AIF into the cytoplasm along with consequent pacification of apoptosis. All protective efficiencies of the extract reported in this study were found to hold strong and significant (P<0.05) positive correlation to its total phenolic contents, thereby proving that polyphenolic constituents of P. amarus aqueous extract mitigate oxidative stress-induced cellular degeneration and aging.


Subject(s)
Anti-Bacterial Agents/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Antimycin A/antagonists & inhibitors , Antimycin A/pharmacology , Apoptosis/drug effects , Mitochondria/drug effects , Phyllanthus/chemistry , 8-Hydroxy-2'-Deoxyguanosine , Apoptosis Inducing Factor/pharmacology , Bromodeoxyuridine , Cell Line , Coloring Agents , Cytochromes c/metabolism , DNA, Mitochondrial/antagonists & inhibitors , DNA, Mitochondrial/isolation & purification , Deoxyguanosine/analogs & derivatives , Flow Cytometry , Humans , In Vitro Techniques , Indicators and Reagents , Lipid Peroxidation/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/ultrastructure , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Phenols/analysis , Plant Extracts/chemistry , Plant Extracts/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tetrazolium Salts , Thiazoles
SELECTION OF CITATIONS
SEARCH DETAIL
...