Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Leukemia ; 31(4): 861-871, 2017 04.
Article in English | MEDLINE | ID: mdl-27694924

ABSTRACT

Somatic inactivating mutations in epigenetic regulators are frequently found in combination in myelodysplastic syndrome (MDS). However, the mechanisms by which combinatory mutations in epigenetic regulators promote the development of MDS remain unknown. Here we performed epigenomic profiling of hematopoietic progenitors in MDS mice hypomorphic for Tet2 following the loss of the polycomb-group gene Ezh2 (Tet2KD/KDEzh2Δ/Δ). Aberrant DNA methylation propagated in a sequential manner from a Tet2-insufficient state to advanced MDS with deletion of Ezh2. Hyper-differentially methylated regions (hyper-DMRs) in Tet2KD/KDEzh2Δ/Δ MDS hematopoietic stem/progenitor cells were largely distinct from those in each single mutant and correlated with transcriptional repression. Although Tet2 hypomorph was responsible for enhancer hypermethylation, the loss of Ezh2 induced hyper-DMRs that were enriched for CpG islands of polycomb targets. Notably, Ezh2 targets largely lost the H3K27me3 mark while acquiring a significantly higher level of DNA methylation than Ezh1 targets that retained the mark. These findings indicate that Ezh2 targets are the major targets of the epigenetic switch in MDS with Ezh2 insufficiency. Our results provide a detailed trail for the epigenetic drift in a well-defined MDS model and demonstrate that the combined dysfunction of epigenetic regulators cooperatively remodels the epigenome in the pathogenesis of MDS.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Base Sequence , Binding Sites , CpG Islands , DNA Methylation , DNA-Binding Proteins/genetics , Dioxygenases , Disease Models, Animal , Enhancer Elements, Genetic , Enhancer of Zeste Homolog 2 Protein/genetics , Hematopoiesis/genetics , Mice , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Nucleotide Motifs , Protein Binding , Proto-Oncogene Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism
2.
Rev Gastroenterol Mex ; 74(4): 314-20, 2009.
Article in Spanish | MEDLINE | ID: mdl-20423760

ABSTRACT

BACKGROUND: Laparoscopic cholecystectomy is the gold standard in the management of symptomatic gallstone disease. Postoperative abdominal and shoulder-tip pain are the main adverse side effects. AIM: To determine whether the low-pressure pneumoperitoneum (7 mmHg) decreases postoperative pain in patients undergoing laparoscopic cholecystectomy compared with standard pressure pneumoperitoneum (12-15 mmHg). MATERIAL AND METHODS: Double-blind clinical trial that included 68 patients divided into two groups: low-pressure and standard pressure pneumoperitoneum. Main variables assessed were abdominal pain at 6, 12 and 24 hours (by visual analogue scale), the incidence of shoulder- tip pain, time and quality of exposure of the surgical field. Variables were compared using Chi square and T-Student, considering significance at p<0.05. RESULTS: The demographic characteristics of patients were similar in both groups. Abdominal pain was significantly less at 12 and 24 hours in the group with low-pressure pneumoperitoneum (p=0.02). The presence of shoulder-tip pain occurred more frequently in the group with standard- pressure pneumoperitoneum (p=0.007). CONCLUSIONS: Low-pressure pneumoperitoneum significantly reduces abdominal and shoulder tip pain. Key words: pneumoperitoneum, laparoscopic cholecystectomy, postoperative pain, shoulder pain, complications, Mexico.


Subject(s)
Cholecystectomy, Laparoscopic , Pain, Postoperative/prevention & control , Pneumoperitoneum, Artificial/methods , Adult , Double-Blind Method , Elective Surgical Procedures , Female , Humans , Male , Pressure
3.
Oncogene ; 26(28): 4179-88, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17213809

ABSTRACT

Dasatinib is an ATP-competitive, multi-targeted SRC and ABL kinase inhibitor that can bind BCR-ABL in both the active and inactive conformations. From a clinical standpoint, dasatinib is particularly attractive because it has been shown to induce hematologic and cytogenetic responses in imatinib-resistant chronic myeloid leukemia patients. The fact because the combination of imatinib and dasatinib shows the additive/synergistic growth inhibition on wild-type p210 BCR-ABL-expressing cells, we reasoned that these ABL kinase inhibitors might induce the different molecular pathways. To address this question, we used DNA microarrays to identify genes whose transcription was altered by imatinib and dasatinib. K562 cells were cultured with imatinib or dasatinib for 16 h, and gene expression data were obtained from three independent microarray hybridizations. Almost all of the imatinib- and dasatinib-responsive genes appeared to be similarly increased or decreased in K562 cells; however, small subsets of genes were identified as selectively altered expression by either imatinib or dasatinib. The distinct genes that are selectively modulated by dasatinib are cyclin-dependent kinase 2 (CDK2) and CDK8, which had a maximal reduction of <5-fold in microarray screen. To assess the functional importance of dasatinib regulated genes, we used RNA interference to determine whether reduction of CDK2 and CDK8 affected the growth inhibition. K562 and TF-1BCR-ABL cells, pretreated with CDK2 or CDK8 small interfering RNA, showed additive growth inhibition with imatinib, but not with dasatinib. These findings demonstrate that the additive/synergistic growth inhibition by imatinib and dasatinib may be mediated in part by CDK2 and CDK8.


Subject(s)
Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Oncogene Proteins v-abl/antagonists & inhibitors , Oncogenes , Piperazines/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Benzamides , Cell Proliferation , DNA Damage , DNA Repair/genetics , Dasatinib , Humans , Imatinib Mesylate , K562 Cells , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering
4.
Oncogene ; 25(42): 5719-25, 2006 Sep 21.
Article in English | MEDLINE | ID: mdl-16652154

ABSTRACT

The telomerase complex is responsible for telomere maintenance and represents a promising neoplasia therapeutic target. Recently, we have demonstrated that treatment with a G-quadruplex-interactive agent, telomestatin reproducibly inhibited telomerase activity in the BCR-ABL-positive leukemic cell lines. In the present study, we investigated the mechanisms of apoptosis induced by telomerase inhibition in acute leukemia. We have found the activation of caspase-3 and poly-(ADP-ribose) polymerase in telomestatin-treated U937 cells (PD20) and dominant-negative DN-hTERT-expressing U937 cells (PD25). Activation of p38 mitogen-activated protein (MAP) kinase and MKK3/6 was also found in telomestatin-treated U937 cells (PD20) and dominant-negative DN-hTERT-expressing U937 cells (PD25); however, activation of JNK and ASK1 was not detected in these cells. To examine the effect of p38 MAP kinase inhibition on growth properties and apoptosis in telomerase-inhibited cells, we cultured DN-hTERT-expressing U937 cells with or without SB203580. Dominant-negative-hTERT-expressing U937 cells stopped proliferation on PD25; however, a significant increase in growth rate was observed in the presence of SB203580. Treatment of SB203580 also reduced the induction of apoptosis in DN-hTERT-expressing U937 cells (PD25). These results suggest that p38 MAP kinase has a critical role for the induction of apoptosis in telomerase-inhibited leukemia cells. Further, we evaluated the effect of telomestatin on the growth of U937 cells in xenograft mouse model. Systemic intraperitoneal administration of telomestatin in U937 xenografts decreased tumor telomerase levels and reduced tumor volumes. Tumor tissue from telomestatin-treated animals exhibited marked apoptosis. None of the mice treated with telomestatin displayed any signs of toxicity. Taken together, these results lay the foundations for a program of drug development to achieve the dual aims of efficacy and selectivity in vivo.


Subject(s)
Leukemia/drug therapy , Oxazoles/pharmacology , Telomerase/antagonists & inhibitors , Acute Disease , Animals , Antineoplastic Agents/pharmacology , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , Humans , Kinetics , Transplantation, Heterologous , U937 Cells
6.
Br J Cancer ; 92(10): 1942-7, 2005 May 23.
Article in English | MEDLINE | ID: mdl-15827550

ABSTRACT

Functionally active telomerase is affected at various steps including transcriptional and post-transcriptional levels of major telomerase components (hTR and human telomerase reverse transcriptase (hTERT)). We therefore developed a rapid and sensitive method to quantify hTERT and its splicing variants as well as the hTR by a Taqman real-time reverse transcriptase-polymerase chain reaction to determine whether their altered expression may contribute to telomere attrition in vivo or not. Fresh leukaemia cells obtained from 38 consecutive patients were used in this study. The enzymatic level of telomerase activity measured by TRAP assay was generally associated with the copy numbers of full-length hTERT+alpha+beta mRNA (P=0.0024), but did not correlate with hTR expression (P=0.6753). In spite of high copy numbers of full-length hTERT mRNA, telomerase activity was low in some cases correlating with low copy numbers of hTR, raising the possibility that alteration of the hTR : hTERT ratio may affect functionally active telomerase activity in vivo. The spliced nonactive hTERT mRNA tends to be lower in patients with high telomerase activity, suggesting that this epiphenomenon may play some role in telomerase regulation. An understanding of the complexities of telomerase gene regulation in biologically heterogeneous leukaemia cells may offer new therapeutic approaches to the treatment of acute leukaemia.


Subject(s)
Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Telomerase/analysis , Telomerase/pharmacology , Adolescent , Adult , Aged , Child , DNA-Binding Proteins , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/biosynthesis , Tumor Cells, Cultured
7.
Int J Oncol ; 24(6): 1481-7, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15138591

ABSTRACT

Telomerase, the ribonucleoprotein enzyme maintaining the telomeres of eukaryotic chromosomes, is up-regulated in the vast majority of human neoplasias but not in normal somatic tissues. Therefore, the telomerase complex represents a promising universal therapeutic target in cancer. Telomeric G-rich single-stranded DNA can adopt in vitro an intramolecular quadruplex structure, which has been shown to inhibit telomerase activity. We examined G-quadruplex interactive agent, telomestatin (SOT-095), for its ability to inhibit the proliferation of human leukemia cells, including freshly obtained leukemia cells. Telomere length was determined by either the terminal restriction fragment method or flow-FISH, and apoptosis was assessed by flow cytometry. Moreover, chemosensitivity was examined in telomestatin-treated U937 cells before ultimate telomere shortening. Treatment with telomestatin reproducibly inhibited telomerase activity in U937 and NB4 cells followed by telomere shortening. Enhanced chemosensitivity toward daunorubicin and cytosine-arabinoside was observed in telomestatin-treated U937 cells, before ultimate telomere shortening. Telomere shortening associated with apoptosis by telomestatin was evident in some freshly obtained leukemia cells from acute myeloid leukemia patients, regardless of sub-types of AML and post-myelodysplasia AML. These results suggest that disruption of telomere maintenance by telomestatin limits the cellular lifespan of AML cells, as well. However, in a minority of AML patients apoptosis was not evident, thus indicating that resistant mechanism might exist in some freshly obtained AML cells. Therefore, further investigation of telomestatin as a therapeutic agent is warranted.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Neoplasm , Leukemia, Myeloid/drug therapy , Oxazoles/pharmacology , Telomerase/antagonists & inhibitors , Telomere/genetics , Acute Disease , Aged , Antibiotics, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Cytarabine/pharmacology , DNA-Binding Proteins , Daunorubicin/pharmacology , Female , Humans , Leukemia, Myeloid/enzymology , Male , Middle Aged , Telomere/metabolism , U937 Cells/drug effects , U937 Cells/metabolism , U937 Cells/pathology
8.
Leukemia ; 17(3): 560-7, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12646945

ABSTRACT

Telomerase is a ribonucleoprotein enzyme that maintains protective structures at the ends of eukaryotic chromosomes. We examined the impact of telomerase inhibition by the dominant-negative human catalytic subunit of telomerase (DN-hTERT) on the biological features of acute leukemia. We introduced vectors encoding dominant- negative (DN)-hTERT, wild-type (WT)-hTERT, or a control vector expressing only a drug-resistant marker into a telomerase-positive human acute lymphoblastic leukemia cell line, HAL-01. Expression of DN-hTERT dramatically inhibited telomerase activity, leading to apoptotic cell death. Mutant telomerase expression also enhanced daunorubicin-induced apoptosis. Nude mice (n=5 per group) received subcutanous implants of HAL-01 cells expressing the control vector or DN-hTERT or WT-hTERT. Implantation of HAL-01 cells expressing control vector (n=5) rapidly produced tumors, whereas implantation of those expressing DN-hTERT (n=5) did not. Thus, telomerase inhibition both growth of HAL-01 cells in vitro and tumorigenic capacity in vivo. Furthermore, the G-quadruplex-interactive telomerase-specific inhibitor, telomestatin, shortened the telomere length and induced apoptosis in freshly isolated primary acute leukemia cells. These results suggest that antitelomerase therapy may be useful in some acute leukemias in combination with antileukemic agents such as daunorubicin.


Subject(s)
Apoptosis/drug effects , Leukemia/pathology , Telomerase/antagonists & inhibitors , Telomerase/genetics , Telomerase/pharmacology , Acute Disease , Animals , DNA-Binding Proteins , Daunorubicin/pharmacology , Drug Synergism , Genes, Dominant , Genetic Therapy , Humans , Leukemia/therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Telomerase/administration & dosage , Telomere/drug effects , Telomere/ultrastructure , Transfection , Tumor Cells, Cultured
9.
Haematologica ; 86(8): E15, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11524265

ABSTRACT

We report here a case of adult T-cell leukemia (ATL) who presented acute renal failure and skin eruption. Renal and skin biopsies showed diffuse invasion of ATL cells. Furthermore, the surface phenotype of tumor cells taken from the bone marrow (BM) or peripheral blood (PB)(CD4-CD8-) differed from that of cells taken from the kidney or skin (CD4+CD8-). These findings suggested that CD4-CD8-ATL cells in the BM and PB had differentiated to CD4+CD8- cells in the kidney and skin.


Subject(s)
Immunophenotyping , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukemic Infiltration/immunology , Acute Kidney Injury/etiology , Blood Cells/immunology , Blood Cells/pathology , Bone Marrow/immunology , Bone Marrow/pathology , Female , Humans , Kidney/immunology , Kidney/pathology , Leukemia-Lymphoma, Adult T-Cell/complications , Leukemia-Lymphoma, Adult T-Cell/immunology , Middle Aged , Skin/immunology , Skin/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...