Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cell Mol Life Sci ; 78(6): 2987-3003, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33206203

ABSTRACT

The pathogenesis of obesity-related metabolic diseases has been linked to the inflammation of white adipose tissue (WAT), but the molecular interconnections are still not fully understood. MiR-146a controls inflammatory processes by suppressing pro-inflammatory signaling pathways. The aim of this study was to characterize the role of miR-146a in obesity and insulin resistance. MiR-146a-/- mice were subjected to a high-fat diet followed by metabolic tests and WAT transcriptomics. Gain- and loss-of-function studies were performed using human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes. Compared to controls, miR-146a-/- mice gained significantly more body weight on a high-fat diet with increased fat mass and adipocyte hypertrophy. This was accompanied by exacerbated liver steatosis, insulin resistance, and glucose intolerance. Likewise, adipocytes transfected with an inhibitor of miR-146a displayed a decrease in insulin-stimulated glucose uptake, while transfecting miR-146a mimics caused the opposite effect. Natriuretic peptide receptor 3 (NPR3) was identified as a direct target gene of miR-146a in adipocytes and CRISPR/Cas9-mediated knockout of NPR3 increased insulin-stimulated glucose uptake and enhanced de novo lipogenesis. In summary, miR-146a regulates systemic and adipocyte insulin sensitivity via downregulation of NPR3.


Subject(s)
Insulin Resistance , MicroRNAs/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Adipocytes/cytology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Antagomirs/metabolism , Body Weight , Diet, High-Fat , Fatty Liver/pathology , Glucose Tolerance Test , Humans , Insulin Resistance/genetics , Lipogenesis , Liver/metabolism , Mice , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Triglycerides/metabolism
2.
Int J Mol Sci ; 21(21)2020 Oct 24.
Article in English | MEDLINE | ID: mdl-33114405

ABSTRACT

Brown adipose tissue (BAT) is a thermogenic organ in rodents and humans. In mice, the transplantation of BAT has been successfully used to combat obesity and its comorbidities. While such beneficial properties of BAT are now evident, the developmental and cellular origins of brown, beige, and white adipocytes have remained only poorly understood, especially in humans. We recently discovered that CD90 is highly expressed in stromal cells isolated from human white adipose tissue (WAT) compared to BAT. Here, we studied whether CD90 interferes with brown or white adipogenesis or white adipocyte beiging. We applied flow cytometric sorting of human adipose tissue stromal cells (ASCs), a CRISPR/Cas9 knockout strategy in the human Simpson-Golabi-Behmel syndrome (SGBS) adipocyte model system, as well as a siRNA approach in human approaches supports the hypothesis that CD90 affects brown or white adipogenesis or white adipocyte beiging in humans. Taken together, our findings call the conclusions drawn from previous studies, which claimed a central role of CD90 in adipocyte differentiation, into question.


Subject(s)
Adipose Tissue, Beige/cytology , Adipose Tissue, Brown/cytology , Arrhythmias, Cardiac/genetics , Genetic Diseases, X-Linked/genetics , Gigantism/genetics , Heart Defects, Congenital/genetics , Intellectual Disability/genetics , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Adult , Arrhythmias, Cardiac/metabolism , CRISPR-Cas Systems , Cell Differentiation , Cells, Cultured , Female , Flow Cytometry , Gene Knockout Techniques , Genetic Diseases, X-Linked/metabolism , Gigantism/metabolism , Heart Defects, Congenital/metabolism , Humans , Intellectual Disability/metabolism , Male , Middle Aged , Stromal Cells/metabolism , Thermogenesis , Up-Regulation
3.
Haematologica ; 99(6): 1050-61, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24700491

ABSTRACT

Novel therapies are needed for pediatric acute lymphoblastic leukemia resistant to conventional therapy. While emerging data suggest leukemias as possible targets of oncolytic attenuated measles virus, it is unknown whether measles virus can eradicate disseminated leukemia, in particular pediatric acute lymphoblastic leukemia. We evaluated the efficacy of attenuated measles virus against a large panel of pediatric xenografted and native primary acute lymphoblastic leukemias ex vivo, and against four different acute lymphoblastic leukemia xenografts of B-lineage in non-obese diabetic/severe combined immunodeficient mice. Ex vivo, attenuated measles virus readily spread among and effectively killed leukemia cells while sparing normal human blood cells and their progenitors. In immunodeficient mice with disseminated acute lymphoblastic leukemia a few intravenous injections of attenuated measles virus sufficed to eradicate leukemic blasts in the hematopoietic system and to control central nervous system disease resulting in long-term survival in three of the four xenografted B-lineage leukemias. Differential sensitivity of leukemia cells did not require increased expression of the measles entry receptors CD150 or CD46 nor absence of the anti-viral retinoic acid-inducible gene I/melanoma differentiation associated gene-5 /interferon pathway. Attenuated oncolytic measles virus is dramatically effective against pediatric B-lineage acute lymphoblastic leukemia in the pre-clinical setting warranting further investigations towards clinical translation.


Subject(s)
Genetic Vectors/genetics , Measles virus/genetics , Oncolytic Viruses/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Antigens, CD/metabolism , Cell Line, Tumor , Cell Survival , Child , Cytopathogenic Effect, Viral , Disease Models, Animal , Genetic Vectors/administration & dosage , Humans , Membrane Cofactor Protein/metabolism , Membrane Proteins/metabolism , Mice, Inbred NOD , Mice, SCID , Nerve Tissue Proteins/metabolism , Oncolytic Virotherapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Transcription, Genetic , Virus Replication , Xenograft Model Antitumor Assays
4.
Ann Hematol ; 92(2): 173-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23161387

ABSTRACT

NPM1 mutations, the most frequent molecular alterations in acute myeloid leukemia (AML), have become important for risk stratification and treatment decisions for patients with normal karyotype AML. Rapid screening for NPM1 mutations should be available shortly after diagnosis. Several methods for detecting NPM1 mutations have been described, most of which are technically challenging and require additional laboratory equipment. We developed and validated an assay that allows specific, rapid, and simple screening for NPM1 mutations. FAST PCR spanning exons 8 to 12 of the NPM1 gene was performed on 284 diagnostic AML samples. PCR products were visualized on a 2 % agarose E-gel and verified by direct sequencing. The FAST PCR screening method showed a specificity and sensitivity of 100 %, i.e., all mutated cases were detected, and none of negative cases carried mutations. The limit of detection was at 5-10 % of mutant alleles. We conclude that the FAST PCR assay is a highly specific, rapid (less than 2 h), and sensitive screening method for the detection of NPM1 mutations. Moreover, this method is inexpensive and can easily be integrated in the routine molecular diagnostic work-up of established risk factors in AML using standard laboratory equipment.


Subject(s)
DNA Mutational Analysis/methods , DNA, Complementary/genetics , DNA, Neoplasm/genetics , Electrophoresis, Agar Gel/methods , Frameshift Mutation , Genetic Testing/methods , Leukemia, Myeloid, Acute/genetics , Leukemia, Myelomonocytic, Acute/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction/methods , Adolescent , Adult , Aged , Aged, 80 and over , Exons/genetics , Female , Humans , Male , Middle Aged , Mutagenesis, Insertional , Nucleophosmin , Oncogene Proteins, Fusion/genetics , RNA, Neoplasm/genetics , Time Factors , Young Adult
5.
Front Oncol ; 12: 744984, 2022.
Article in English | MEDLINE | ID: mdl-35814385

ABSTRACT

Neuroblastoma (NB) is the most common extracranial solid tumor in childhood and has a poor prognosis in high-risk cases, requiring novel therapies. Pathways that depend on phospho-signaling maintain the aggressiveness of NB. Protein phosphatase 2 (PP2A) with its catalytic subunit PPP2CA is a major phosphatase in cancer cells, including NB. We show that reduction of PPP2CA by knock-down decreased growth of NB cells and that complete ablation of PPP2CA by knock-out was not tolerated. Thus, NB cells are addicted to PPP2CA, an addiction augmented by MYCN activation. SET, a crucial endogenous inhibitor of PP2A, was overexpressed in poor-prognosis NB. The SET inhibitor OP449 effectively decreased the viability of NB cells, independent of their molecular alterations and in line with a tumor suppressor function of PPP2CA. The contrasting concentration-dependent functions of PPP2CA as an essential survival gene at low expression levels and a tumor suppressor at high levels are reminiscent of other genes showing this so-called Goldilocks phenomenon. PP2A reactivated by OP449 decreased activating phosphorylation of serine/threonine residues in the AKT pathway. Conversely, induced activation of AKT led to partial rescue of OP449-mediated viability inhibition. Dasatinib, a kinase inhibitor used in relapsed/refractory NB, and OP449 synergized, decreasing activating AKT phosphorylations. In summary, concomitantly reactivating phosphatases and inhibiting kinases with a combination of OP449 and dasatinib are promising novel therapeutic approaches to NB.

6.
BMC Cancer ; 10: 560, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20950443

ABSTRACT

BACKGROUND: Targeted therapy approaches have been successfully introduced into the treatment of several cancers. The multikinase inhibitor Sorafenib has antitumor activity in solid tumors and its effects on acute lymphoblastic leukemia (ALL) cells are still unclear. METHODS: ALL cell lines (SEM, RS4;11 and Jurkat) were treated with Sorafenib alone or in combination with cytarabine, doxorubicin or RAD001. Cell count, apoptosis and necrosis rates, cell cycle distribution, protein phosphorylation and metabolic activity were determined. RESULTS: Sorafenib inhibited the proliferation of ALL cells by cell cycle arrest accompanied by down-regulation of CyclinD3 and CDK4. Furthermore, Sorafenib initiated apoptosis by cleavage of caspases 3, 7 and PARP. Apoptosis and necrosis rates increased significantly with most pronounced effects after 96 h. Antiproliferative effects of Sorafenib were associated with a decreased phosphorylation of Akt (Ser473 and Thr308), FoxO3A (Thr32) and 4EBP-1 (Ser65 and Thr70) as early as 0.5 h after treatment. Synergistic effects were seen when Sorafenib was combined with other cytotoxic drugs or a mTOR inhibitor emphasizing the Sorafenib effect. CONCLUSION: Sorafenib displays significant antileukemic activity in vitro by inducing cell cycle arrest and apoptosis. Furthermore, it influences PI3K/Akt/mTOR signaling in ALL cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , B-Lymphocytes/pathology , Benzenesulfonates/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Pyridines/pharmacology , T-Lymphocytes/pathology , Cell Line, Tumor , Cell Proliferation , Humans , Jurkat Cells , Niacinamide/analogs & derivatives , Phenylurea Compounds , Phosphorylation , Sorafenib
7.
Sci Rep ; 8(1): 5527, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29615749

ABSTRACT

Due to the highly invasive nature of Glioblastoma (GB), complete surgical resection is not feasible, while motile tumour cells are often associated with several specific brain structures that enhance treatment-resistance. Here, we investigate the therapeutic potential of Disulfiram and Carbenoxolone, that inhibit two distinct interactions between GB and the brain tissue microenvironment: stress-induced cell-matrix adhesion and gap junction mediated cell-cell communication, respectively. Increase in cell numbers of tumour-initiating cells, which are cultured in suspension as cell clusters, and adherent differentiated cells can be blocked to a similar extent by Carbenoxolone, as both cell populations form gap junctions, but the adherent differentiated cells are much more sensitive to Disulfiram treatment, which - via modulation of NF-κB signalling - interferes with cell-substrate adhesion. Interestingly, inducing adhesion in tumour-initiating cells without differentiating them does not sensitize for Disulfiram. Importantly, combining Disulfiram, Carbenoxolone and the standard chemotherapeutic drug Temozolomide reduces tumour size in an orthotopic mouse model. Isolating GB cells from their direct environment within the brain represents an important addition to current therapeutic approaches. The blockage of cellular interactions via the clinically relevant substances Disulfiram and Carbenoxolone, has distinct effects on different cell populations within a tumour, potentially reducing motility and/or resistance to apoptosis.


Subject(s)
Brain Neoplasms/drug therapy , Carbenoxolone/pharmacology , Disulfiram/pharmacology , Glioblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Tumor Microenvironment/drug effects , Acetaldehyde Dehydrogenase Inhibitors/pharmacology , Animals , Anti-Ulcer Agents/pharmacology , Apoptosis , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Adhesion , Cell Proliferation , Drug Therapy, Combination , Gene Expression Profiling , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Sci Rep ; 7(1): 5691, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28720906

ABSTRACT

High serum concentrations of TNF-related apoptosis-inducing ligand (TRAIL), a member of the tumor necrosis factor protein family, are found in patients with increased BMI and serum lipid levels. In a model of murine obesity, both the expression of TRAIL and its receptor (TRAIL-R) is elevated in adipose tissue. Accordingly, TRAIL has been proposed as an important mediator of adipose tissue inflammation and obesity-associated diseases. The aim of this study was to investigate if TRAIL regulates inflammatory processes at the level of the adipocyte. Using human Simpson-Golabi-Behmel syndrome (SGBS) cells as a model system, we found that TRAIL induces an inflammatory response in both preadipocytes and adipocytes. It stimulates the expression of interleukin 6 (IL-6), interleukin 8 (IL-8) as well as the chemokines monocyte chemoattractant protein-1 (MCP-1) and chemokine C-C motif ligand 20 (CCL-20) in a time- and dose-dependent manner. By using small molecule inhibitors, we found that both the NFκB and the ERK1/2 pathway are crucial for mediating the effect of TRAIL. Taken together, we identified a novel pro-inflammatory function of TRAIL in human adipocytes. Our findings suggest that targeting the TRAIL/TRAIL-R system might be a useful strategy to tackle obesity-associated adipose tissue inflammation.


Subject(s)
Adipocytes/drug effects , Inflammation/physiopathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Adult , Arrhythmias, Cardiac , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CCL20 , Genetic Diseases, X-Linked , Gigantism , Heart Defects, Congenital , Humans , Intellectual Disability , Interleukin-6/metabolism , Interleukin-8/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Signal Transduction
9.
Cancer Lett ; 371(1): 79-89, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26616283

ABSTRACT

Aurora Kinase A (AURKA) is often overexpressed in neuroblastoma (NB) with poor outcome. The causes of AURKA overexpression in NB are unknown. Here, we describe a gene regulatory network consisting of core regulators of AURKA protein expression and activation during mitosis to identify potential causes. This network was transformed to a dynamic Boolean model. Simulated activation of the serine/threonine protein kinase Greatwall (GWL, encoded by MASTL) that attenuates the pivotal AURKA inhibitor PP2A, predicted stabilization of AURKA. Consistent with this notion, gene set enrichment analysis showed enrichment of mitotic spindle assembly genes and MYCN target genes in NB with high GWL/MASTL expression. In line with the prediction of GWL/MASTL enhancing AURKA, elevated expression of GWL/MASTL was associated with NB risk factors and poor survival of patients. These results establish Boolean network modeling of oncogenic pathways in NB as a useful means for guided discovery in this enigmatic cancer.


Subject(s)
Aurora Kinase A/genetics , Computer Simulation , Microtubule-Associated Proteins/genetics , Models, Genetic , Neuroblastoma/genetics , Protein Serine-Threonine Kinases/genetics , Adolescent , Aurora Kinase A/metabolism , Child , Child, Preschool , Databases, Genetic , Enzyme Stability , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Infant , Infant, Newborn , Male , Microtubule-Associated Proteins/metabolism , N-Myc Proto-Oncogene Protein , Neuroblastoma/enzymology , Neuroblastoma/mortality , Neuroblastoma/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Survival Analysis , Young Adult
10.
Oncotarget ; 7(6): 6620-5, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26735174

ABSTRACT

Little is known about changes within the mitochondrial (mt) genome during tumor progression in general and during initiation and progression of neuroblastoma (NB) in particular. Whole exome sequencing of corresponding healthy tissue, primary tumor and relapsed tumor from 16 patients with NB revealed that most NB harbor tumor-specific mitochondrial variants. In relapsed tumors, the status of mt variants changed in parallel to the status of nuclear variants, as shown by increased number and spatio-temporal differences of tumor-specific variants, and by a concomitant decrease of germline variants. As mt variants are present in most NB patients, change during relapse and have a higher copy number compared to nuclear variants, they represent a promising new source of biomarkers for monitoring and phylogenetic analysis of NB.


Subject(s)
Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Mitochondrial Proteins/genetics , Mutation/genetics , Neoplasm Recurrence, Local/genetics , Neuroblastoma/genetics , Disease Progression , High-Throughput Nucleotide Sequencing/methods , Humans
11.
Oncotarget ; 5(11): 3907-18, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-25004098

ABSTRACT

Although apoptotic phenomena have been observed in malignant cells infected by measles virus vaccine strain Edmonston B (MV-Edm), the precise oncolytic mechanisms are poorly defined. In this study we found that MV-Edm induced autophagy and sequestosome 1-mediated mitophagy leading to decreased cytochrome c release, which blocked the pro-apoptotic cascade in non-small cell lung cancer cells (NSCLCs). The decrease of apoptosis by mitophagy favored viral replication. Persistent viral replication sustained by autophagy ultimately resulted in necrotic cell death due to ATP depletion. Importantly, when autophagy was impaired in NSCLCs MV-Edm-induced cell death was significantly abrogated despite of increased apoptosis. Taken together, our results define a novel oncolytic mechanism by which mitophagy switches cell death from apoptosis to more efficient necrosis in NSCLCs following MV-Edm infection. This provides a foundation for future improvement of oncolytic virotherapy or antiviral therapy.


Subject(s)
Adenocarcinoma/therapy , Lung Neoplasms/therapy , Measles virus/physiology , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/virology , Adenocarcinoma of Lung , Adenosine Triphosphate/metabolism , Animals , Apoptosis/physiology , Cell Death/physiology , Cell Line, Tumor , Chlorocebus aethiops , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/virology , Mitophagy/physiology , Necrosis , Vero Cells
12.
J Clin Invest ; 124(4): 1810-20, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569453

ABSTRACT

Alteration of the surface glycosylation pattern on malignant cells potentially affects tumor immunity by directly influencing interactions with glycan-binding proteins (lectins) on the surface of immunomodulatory cells. The sialic acid-binding Ig-like lectins Siglec-7 and -9 are MHC class I-independent inhibitory receptors on human NK cells that recognize sialic acid-containing carbohydrates. Here, we found that the presence of Siglec-9 defined a subset of cytotoxic NK cells with a mature phenotype and enhanced chemotactic potential. Interestingly, this Siglec-9+ NK cell population was reduced in the peripheral blood of cancer patients. Broad analysis of primary tumor samples revealed that ligands of Siglec-7 and -9 were expressed on human cancer cells of different histological types. Expression of Siglec-7 and -9 ligands was associated with susceptibility of NK cell-sensitive tumor cells and, unexpectedly, of presumably NK cell-resistant tumor cells to NK cell-mediated cytotoxicity. Together, these observations have direct implications for NK cell-based therapies and highlight the requirement to consider both MHC class I haplotype and tumor-specific glycosylation.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Killer Cells, Natural/immunology , Lectins/metabolism , Monitoring, Immunologic , Neoplasms/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Glycosylation , HeLa Cells , Humans , Immunity, Innate , K562 Cells , Killer Cells, Natural/classification , Ligands , Male , Mice , Mice, Inbred NOD , Mice, SCID
13.
Anticancer Res ; 33(2): 445-52, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23393335

ABSTRACT

Dysregulation of microRNAs (miRs) has been linked to several types of cancer. In the present study, we investigated the expression of miR-181a in different leukemia cell lines and healthy hematopoietic cells, as well as its influence on cell proliferation, metabolic activity and potential targets. Expression of miR-181a differed between various leukemia cell lines and mature blood cells. Inhibition of miR 181a expression in T- and B-Acute Lymphoblastic Leukemia (ALL) cells revealed an influence on the potential targets High-Mobility-Group-Protein B1 (HMGB1) and Cluster of Differentiation 4 (CD4). Overexpression of miR-181a in AML cells led to a significant decrease in cell proliferation and metabolic activity. The present data indicate a possible role of this specific miRNA in immunogenicity.


Subject(s)
CD4 Antigens/biosynthesis , Gene Expression Regulation/genetics , HMGB1 Protein/biosynthesis , Leukemia/genetics , Leukemia/metabolism , MicroRNAs/genetics , Blotting, Western , CD4 Antigens/genetics , Cell Line, Tumor , Cell Separation , Electroporation , Flow Cytometry , Gene Expression Profiling , HMGB1 Protein/genetics , Humans , Real-Time Polymerase Chain Reaction
14.
Anticancer Res ; 32(2): 463-74, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22287733

ABSTRACT

BACKGROUND: Inhibition of signal transduction pathways has been successfully introduced into cancer treatment. The dual phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR) inhibitor NVP-BEZ235 has antitumor activity in vitro against solid tumors. Here, we examined the activity of NVP-BEZ235 in acute lymphoblastic leukemia (ALL) cells and the best modalities for combination approaches. MATERIALS AND METHODS: ALL cell lines (SEM, RS4;11, Jurkat and MOLT4) were treated with NVP-BEZ235 alone, or in combination with cytarabine (AraC), doxorubicin (Doxo) or dexamethasone (Dexa). RESULTS: NVP-BEZ235 potently inhibited the proliferation and metabolic activity of ALL cells. Antiproliferative effects were associated with G(0)/G(1) arrest and reduced levels of cyclin-dependent kinase 4 (CDK4) and cyclin D3. Inhibition of PI3K and mTOR activity was detected at 10 and 100 nM. NVP-BEZ235 combined with AraC, Doxo or Dexa synergistically enhanced the cytotoxicity compared to single-drug treatment, even in glucocorticoid-resistant cells. CONCLUSION: NVP-BEZ235 displays pronounced antiproliferative effects in ALL cells and might therefore be a useful drug in the treatment of ALL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Imidazoles/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Quinolines/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cytarabine/administration & dosage , Dexamethasone/administration & dosage , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm , Drug Synergism , Humans , Imidazoles/administration & dosage , Jurkat Cells , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/enzymology , Leukemia, B-Cell/metabolism , Leukemia, B-Cell/pathology , Oncogene Protein v-akt/antagonists & inhibitors , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Quinolines/administration & dosage , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
15.
Mol Endocrinol ; 24(12): 2331-42, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20943817

ABSTRACT

The transcriptional transactivator Pax6 binds the pancreatic islet cell-specific enhancer sequence (PISCES) of the rat insulin I gene. However the human, mouse, and rat insulin gene II promoters do not contain a PISCES element. To analyze the role of Pax6 in those PISCES-less promoters, we investigated its influence on rat insulin gene II expression and included in our studies the main activators: pancreatic and duodenal homeobox protein-1 (PDX-1) and BETA2/E47. Luciferase assays, Northern blots, and RIA were used to study effects of Pax6 overexpression, gel shift and chromatin precipitation assays to study its binding to the DNA, and yeast two-hybrid assays and glutathione S transferase capture assays to investigate its interactions with PDX-1 and BETA2. Finally, glucose-dependent intracellular transport of Pax6 was demonstrated by fluorescence microscopy. Overexpression of Pax6 prevents activation of the rat insulin II gene by BETA2 and PDX-1 and hence suppresses insulin synthesis and secretion. In vitro, Pax6 binds to the A-boxes, thereby blocking binding of PDX-1, and at the same time, its paired domain interacts with BETA2. Fluorescence microscopy demonstrated that the nuclear-cytoplasmic localization of Pax6 and PDX-1 are oppositely regulated by glucose. From the results, it is suggested that at low concentrations of glucose, Pax6 is localized in the nucleus and prevents the activation of the insulin gene by occupying the PDX-1 binding site and by interacting with BETA2.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Eye Proteins/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin/genetics , Insulinoma/genetics , Paired Box Transcription Factors/genetics , Pancreatic Neoplasms/genetics , Repressor Proteins/genetics , Trans-Activators/metabolism , Active Transport, Cell Nucleus , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Down-Regulation , Eye Proteins/biosynthesis , Eye Proteins/metabolism , Glucose/pharmacology , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/biosynthesis , Humans , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Insulinoma/metabolism , Mice , PAX6 Transcription Factor , Paired Box Transcription Factors/biosynthesis , Paired Box Transcription Factors/metabolism , Pancreatic Neoplasms/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Transport , Rats , Repressor Proteins/biosynthesis , Repressor Proteins/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL