Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 314
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Br J Anaesth ; 123(2): 246-254, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31160064

ABSTRACT

BACKGROUND: Electrical impedance tomography (EIT) with indicator dilution may be clinically useful to measure relative lung perfusion, but there is limited information on the performance of this technique. METHODS: Thirteen pigs (50-66 kg) were anaesthetised and mechanically ventilated. Sequential changes in ventilation were made: (i) right-lung ventilation with left-lung collapse, (ii) two-lung ventilation with optimised PEEP, (iii) two-lung ventilation with zero PEEP after saline lung lavage, (iv) two-lung ventilation with maximum PEEP (20/25 cm H2O to achieve peak airway pressure 45 cm H2O), and (v) two-lung ventilation under unilateral pulmonary artery occlusion. Relative lung perfusion was assessed with EIT and central venous injection of saline 3%, 5%, and 10% (10 ml) during breath holds. Relative perfusion was determined by positron emission tomography (PET) using 68Gallium-labelled microspheres. EIT and PET were compared in eight regions of equal ventro-dorsal height (right, left, ventral, mid-ventral, mid-dorsal, and dorsal), and directional changes in regional perfusion were determined. RESULTS: Differences between methods were relatively small (95% of values differed by less than 8.7%, 8.9%, and 9.5% for saline 10%, 5%, and 3%, respectively). Compared with PET, EIT underestimated relative perfusion in dependent, and overestimated it in non-dependent, regions. EIT and PET detected the same direction of change in relative lung perfusion in 68.9-95.9% of measurements. CONCLUSIONS: The agreement between EIT and PET for measuring and tracking changes of relative lung perfusion was satisfactory for clinical purposes. Indicator-based EIT may prove useful for measuring pulmonary perfusion at bedside.


Subject(s)
Lung/diagnostic imaging , Lung/physiopathology , Positron-Emission Tomography , Pulmonary Ventilation/physiology , Respiration, Artificial , Animals , Disease Models, Animal , Electric Impedance , Swine
2.
Apoptosis ; 19(4): 698-707, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24337870

ABSTRACT

The apoptosis repressor with caspase recruitment domain (ARC) protein is known to suppress both intrinsic and extrinsic apoptosis. We previously reported that ARC expression is a strong, independent adverse prognostic factor in acute myeloid leukemia (AML). Here, we investigated the regulation and role of ARC in AML. ARC expression is upregulated in AML cells co-cultured with bone marrow-derived mesenchymal stromal cells (MSCs) and suppressed by inhibition of MAPK and PI3K signaling. AML patient samples with RAS mutations (N = 64) expressed significantly higher levels of ARC than samples without RAS mutations (N = 371) (P = 0.016). ARC overexpression protected and ARC knockdown sensitized AML cells to cytarabine and to agents that selectively induce intrinsic (ABT-737) or extrinsic (TNF-related apoptosis inducing ligand) apoptosis. NOD-SCID mice harboring ARC-overexpressing KG-1 cells had significantly shorter survival than mice injected with control cells (median 84 vs 111 days) and significantly fewer leukemia cells were present when NOD/SCID IL2Rγ null mice were injected with ARC knockdown as compared to control Molm13 cells (P = 0.005 and 0.03 at 2 and 3 weeks, respectively). Together, these findings demonstrate that MSCs regulate ARC in AML through activation of MAPK and PI3K signaling pathways. ARC confers drug resistance and survival advantage to AML in vitro and in vivo, suggesting ARC as a novel target in AML therapy.


Subject(s)
Caspases/metabolism , Cytoskeletal Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cytarabine/pharmacology , Drug Resistance, Neoplasm , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Signal Transduction
3.
Br J Cancer ; 104(1): 91-100, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21139584

ABSTRACT

BACKGROUND: Mantle cell lymphoma (MCL) is an aggressive B-cell lymphoma with poor prognosis, requiring novel anticancer strategies. METHODS: Mantle cell lymphoma cell lines with known p53 status were treated with GUT-70, a tricyclic coumarin derived from Calophyllum brasiliense, and the biological and biochemical consequences of GUT-70 were studied. RESULTS: GUT-70 markedly reduced cell proliferation/viability through G(1) cell cycle arrest and increased apoptosis, with greater sensitivity in mutant (mt)-p53-expressing MCL cells than in wild-type (wt)-p53-bearing cells. Mechanistically, GUT-70 showed binding affinity to heat-shock protein 90 (Hsp90) and ubiquitin-dependent proteasomal degradation of Hsp90 client proteins, including cyclin D1, Raf-1, Akt, and mt-p53. Depletion of constitutively overexpressed cyclin D1 by GUT-70 was accompanied by p27 accumulation and decreased Rb phosphorylation. GUT-70 induced mitochondrial apoptosis with Noxa upregulation and Mcl-1 downregulation in mt-p53 cells, but Mcl-1 accumulation in wt-p53 cells. Noxa and Mcl-1 were coimmunoprecipitated, and activated BAK. Treatment with a combination of GUT-70 and bortezomib or doxorubicin had synergistic antiproliferative effects in MCL cells that were independent of p53 status. CONCLUSION: GUT-70 has pronounced antiproliferative effects in MCL with mt-p53, a known negative prognostic factor for MCL, through Hsp90 inhibition. These findings suggest that GUT-70 has potential utility for the treatment of MCL.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Coumarins/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lymphoma, Mantle-Cell/drug therapy , Antineoplastic Agents/therapeutic use , Blotting, Western , Boronic Acids/therapeutic use , Bortezomib , Cell Cycle/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Drug Synergism , Drug Therapy, Combination , Flow Cytometry , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Immunoenzyme Techniques , Immunoprecipitation , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Mutation/genetics , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Pyrazines/therapeutic use , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism
4.
J Exp Med ; 155(5): 1357-69, 1982 May 01.
Article in English | MEDLINE | ID: mdl-6175721

ABSTRACT

Human leukemic cells were induced to proliferate and mature to macrophage-like cells in primary cultures supplemented with conditioned medium (CM) from phytohemagglutinin and alloantigen-stimulated normal T lymphocytes. Blast and promyelocyte-enriched preparations, isolated after depletion of adherent phagocytic cells and lymphoid cells from samples of myelogenous leukemia patients, were suspended in liquid cultures with 30% CM. Cell cycle analysis was performed throughout the course of induced cellular maturation. Within 24 h of exposure to CM, cells with macrophage-like morphology were identified among the developing adherent cells. Approximately 15-30% of the cells in culture suspensions also developed macrophage-like morphology and esterase reactivity with alpha-napthyl acetate after incubation for 2 d. The number of these nonproliferating cells increased and became predominant in the later culture period. Flow cytometric measurement of DNA content showed that these mature cells had the same aneuploid stemline as the undifferentiated leukemic cells, indicating that genetically abnormal leukemic cells can be induced to differentiate. Reduction in the total RNA content of the macrophage-like cells was also determined by flow cytometry. Reduction in RNA and development of adherent cells served as early markers of maturation, in addition to the later acquisition of complement receptors and phagocytic capacity. Cell cycle analysis showed that CM stimulated the proliferation of immature cells. This initial proliferation may precede intertwined events of proliferation and concurrent maturation of immature cells. Later in the culture period, cellular proliferation decreased, leading to termination of the cultures.


Subject(s)
Aneuploidy , Cell Transformation, Neoplastic/pathology , Leukemia, Myeloid, Acute/blood , Adult , Aged , Cell Adhesion , Cell Count , Cell Cycle/drug effects , Cell Transformation, Neoplastic/drug effects , Culture Media/pharmacology , DNA/blood , Female , Humans , Leukemia, Myeloid, Acute/immunology , Lymphocyte Activation , Macrophages , Male , Monocytes , Phagocytosis/drug effects , RNA/blood , Receptors, Complement/drug effects , T-Lymphocytes/metabolism
5.
J Exp Med ; 160(5): 1390-403, 1984 Nov 01.
Article in English | MEDLINE | ID: mdl-6092510

ABSTRACT

We investigated the effect of OKT3 antibody and interleukin 2 (IL-2) on Tac antigen expression and the proliferation of human peripheral blood mononuclear leukocytes. OKT3 monoclonal antibody at low, nonmitogenic concentrations (25 pg/ml) or IL-2 alone at optimal concentrations (20 U/ml) did not induce IL-2 receptor expression, as measured by Tac antibody or by T cell proliferation. However, costimulation with these concentrations of OKT3 antibody and IL-2 led to Tac antigen expression and T cell proliferation. These data suggest that the T cells are activated in two steps: OKT3 antibody at 25 pg/ml does not induce Tac antigen expression, but preactivates T cells to become responsive to IL-2. The addition of exogenous IL-2 then leads to expression of the IL-2 receptor, as recognized by Tac antibody, and to subsequent proliferation.


Subject(s)
Antigens, Surface/analysis , Interleukin-2/physiology , T-Lymphocytes/immunology , Antibodies, Monoclonal/physiology , Antigens, Surface/immunology , DNA/analysis , DNA/biosynthesis , Fluorescent Antibody Technique , Humans , Interferon-gamma/physiology , Interleukin-2/biosynthesis , Interleukin-2/metabolism , Kinetics , Lymphocyte Activation , Macrophage Activation , Mitomycin , Mitomycins/pharmacology , Protein Biosynthesis , T-Lymphocytes/analysis , Tumor Necrosis Factor Receptor Superfamily, Member 7
6.
Nuklearmedizin ; 49(6): 203-8, 2010.
Article in German | MEDLINE | ID: mdl-21057723

ABSTRACT

AIM: Imaging of lung perfusion with positron emission tomography (PET) is already possible with 68Ga labeled denaturized albumin. The purpose of our study was to produce and test a 68Ga labeled aerosol (Galligas®) for ventilation and 68Ga labeled albumin particles (microspheres) for perfusion imaging with PET. PATIENTS, METHODS: Galligas was produced by simmering and burning generator eluted 68Ga solution (100 MBq/0.1 ml) in an ordinary technegas generator. Fifteen patients with suspicion on pulmonary embolism underwent PET/CT (Biograph 16) after inhalation of Galligas and application of 68Ga labeled microspheres. A low dose CT was acquired for attenuation correction (AC). Images were reconstructed with and without AC. The inhaled activity was calculated compared to the activity injected. RESULTS: Inhaled radioaerosol Galligas demonstrated typical distribution as known from 99mTc-labeled technegas with homogeneous distribution in lung without hilar deposits. Attenuation corrected images resulted in artefacts in the lung base. Therefore, non-corrected images were used for making the results. Three out of fifteen patients showed a deficient perfusion whereas ventilation was normal corresponding to pulmonary embolism. CONCLUSION: Lung scintigraphy with PET is feasible. Galligas is simple to produce (analogously to technegas). 68Ga labeled microspheres are available. The method is applicable to daily routine and rendered clinically relevant informations.


Subject(s)
Gallium Radioisotopes , Lung/diagnostic imaging , Pulmonary Embolism/diagnostic imaging , Aerosols , Aged , Female , Gallium Radioisotopes/administration & dosage , Humans , Male , Microspheres , Middle Aged , Positron-Emission Tomography/methods
7.
Science ; 250(4980): 559-62, 1990 Oct 26.
Article in English | MEDLINE | ID: mdl-2237408

ABSTRACT

Chronic myelogeneous leukemia (CML) is genetically characterized by fusion of the bcr and abl genes on chromosomes 22 and 9, respectively. In most cases, the fusion involves a reciprocal translocation t(9;22)(q34;q11), which produces the cytogenetically distinctive Philadelphia chromosome (Ph1). Fusion can be detected by Southern (DNA) analysis or by in vitro amplification of the messenger RNA from the fusion gene with polymerase chain reaction (PCR). These techniques are sensitive but cannot be applied to single cells. Two-color fluorescence in situ hybridization (FISH) was used with probes from portions of the bcr and abl genes to detect the bcr-abl fusion in individual blood and bone marrow cells from six patients. The fusion event was detected in all samples analyzed, of which three were cytogenetically Ph1-negative. One of the Ph1-negative samples was also PCR-negative. This approach is fast and sensitive, and provides potential for determining the frequency of the abnormality in different cell lineages.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Protein-Tyrosine Kinases , Chromosomes, Human, Pair 22 , Chromosomes, Human, Pair 9 , Genes, abl , Humans , Interphase , Metaphase , Nucleic Acid Hybridization , Philadelphia Chromosome , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcr , Translocation, Genetic
8.
Nuklearmedizin ; 48(6): 227-32, 2009.
Article in German | MEDLINE | ID: mdl-19862463

ABSTRACT

AIM: The absorbed dose is an important parameter in experiments involving irradiation of cells in vitro with unsealed radionuclides. Typically, this is estimated with a model calculation, although the results thus obtained cannot be verified. Generally used real-time measurement methods are not applicable in this setting. A new detector material with in vitro suitability is the subject of this work. METHODS: Optically-stimulated luminescence (OSL) dosimeters based on beryllium oxide (BeO) were used for dose measurement in cell cultures exposed to unsealed radionuclides. Their qualitative properties (e. g. energy-dependent count rate sensitivity, fading, contamination by radioactive liquids) were determined and compared to the results of a Monte Carlo simulation (using AMOS software). OSL dosimeters were tested in common cell culture setups with a known geometry. RESULTS: Dose reproducibility of the OSL dosimeters was +/-1.5%. Fading at room temperature was 0.07% per day. Dose loss (optically-stimulated deletion) under ambient lighting conditions was 0.5% per minute. The Monte Carlo simulation for the relative sensitivity at different beta energies provided corresponding results to those obtained with the OSL dosimeters. Dose profile measurements using a 6 well plate and 14 ml PP tube showed that the geometry of the cell culture vessel has a marked influence on dose distribution with 188Re. CONCLUSION: A new dosimeter system was calibrated with beta-emitters of different energy. It turned out as suitable for measuring dose in liquids. The dose profile measurements obtained are suitably precise to be used as a check against theoretical dose calculations.


Subject(s)
Beryllium/radiation effects , Cell Culture Techniques/instrumentation , Monitoring, Physiologic/instrumentation , Radiometry/instrumentation , Equipment Design , Equipment Failure Analysis , Radiation Dosage , Reproducibility of Results , Sensitivity and Specificity
9.
Nuklearmedizin ; 48(5): 201-7, 2009.
Article in German | MEDLINE | ID: mdl-19639164

ABSTRACT

AIM: Investigation of the biodistribution and calculation of dosimetry of Ga-68-DOTATOC- for patients imaged in the routine clinical setting for diagnosis or exclusion of neuroendocrine tumours. PATIENTS, METHODS: Dynamic PET/CT-imaging (Biograph 16) was performed over 20 min in 14 patients (8 men, 6 women) after injection of (112+/-22) MBq 68Ga-DOTATOC followed by whole body 3D-acquisition (8 bed positions, 3 or 4 min each) 30 min p.i. and 120 min p.i.. Urinary tracer elimination was measured and blood activity was derived non-invasively from the blood pool of the heart. The relevant organs for dosimetry were spleen, kidneys, liver, adrenals, urinary bladder and pituitary gland. Dosimetry was performed using OLINDA/EXM 1.0 software and specific organ uptake was expressed as standardized uptake values (SUVs). RESULTS: Rapid physiological uptake of the radiotracer could be demonstrated in liver, spleen and kidneys, adrenals and pituitary gland (mean SUVs were 6, 20, 16, 10, and 4, respectively). Radiotracer elimination was exclusively via urine (16% of injected dose within 2h); no redistribution could be observed. The spleen and the kidneys received the highest radiation exposure (0.24 mSv/MBq, 0.22 mSv/MBq resp.), mean effective dose yielded 0.023 mSv/MBq. CONCLUSION: 68Ga-DOTATOC is used extensively for diagnosis of somatostatin receptor positive tumours because it has several advantages over the 111In-labelled ligand. The derived dosimetric values are lower than first approximations from the biological data of OctreoScan. The use of CT for transmission correction of the PET data delivers radiation exposure up to 1 mSv (low dose).


Subject(s)
Gallium Radioisotopes , Octreotide/analogs & derivatives , Adult , Aged , Bone and Bones/radiation effects , Environmental Exposure , Female , Gallium Radioisotopes/pharmacokinetics , Humans , Kidney/radiation effects , Liver/radiation effects , Male , Middle Aged , Neoplasm Staging , Octreotide/pharmacokinetics , Positron-Emission Tomography/methods , Tomography, X-Ray Computed/methods
11.
Oncogene ; 26(10): 1439-48, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-16936778

ABSTRACT

To investigate if the tumor suppressor properties of p57KIP2 are dependent on its DNA methylation status, we studied the impact of several stress stimuli in leukemic cell lines with different p57KIP2 promoter DNA methylation levels. p57KIP2 reactivation was observed after stimulation with transforming growth factor-beta, other cytokines, high-density culture or serum withdrawal in p57KIP2 promoter unmethylated cells but not in methylated cells. In these cells, p57KIP2 reactivation required the use of a hypomethylating agent or a histone deacetylase inhibitor. Overexpression of p57KIP2 in p57KIP2 promoter methylated leukemic cell lines resulted in cell growth arrest and the induction of apoptosis. In contrast, overexpression of p57KIP2 in partially methylated cells only resulted in a moderate inhibition of cell growth and had no impact on apoptosis. Transduction of unmethylated cells expressing high levels of p57KIP2 with p57KIP2 short hairpin RNA resulted in increased cell proliferation. These results suggest that the tumor suppressive properties of p57KIP2 in leukemia may depend on the intrinsic promoter DNA methylation status of the gene.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/genetics , DNA Methylation , Genes, Tumor Suppressor , Leukemia/genetics , Promoter Regions, Genetic , Transforming Growth Factor beta/pharmacology , Apoptosis , Cell Culture Techniques/methods , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Jurkat Cells
12.
Oncogene ; 26(28): 4148-57, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17260024

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs of 19-25 nucleotides that are involved in the regulation of critical cell processes such as apoptosis, cell proliferation and differentiation. However, little is known about the role of miRNAs in granulopoiesis. Here, we report the expression of miRNAs in acute promyelocytic leukemia patients and cell lines during all-trans-retinoic acid (ATRA) treatment by using a miRNA microarrays platform and quantitative real time-polymerase chain reaction (qRT-PCR). We found upregulation of miR-15a, miR-15b, miR-16-1, let-7a-3, let-7c, let-7d, miR-223, miR-342 and miR-107, whereas miR-181b was downregulated. Among the upregulated miRNAs, miR-107 is predicted to target NFI-A, a gene that has been involved in a regulatory loop involving miR-223 and C/EBPa during granulocytic differentiation. Indeed, we have confirmed that miR-107 targets NF1-A. To get insights about ATRA regulation of miRNAs, we searched for ATRA-modulated transcription factors binding sites in the upstream genomic region of the let-7a-3/let-7b cluster and identified several putative nuclear factor-kappa B (NF-kappaB) consensus elements. The use of reporter gene assays, chromatin immunoprecipitation and site-directed mutagenesis revealed that one proximal NF-kappaB binding site is essential for the transactivation of the let-7a-3/let-7b cluster. Finally, we show that ATRA downregulation of RAS and Bcl2 correlate with the activation of known miRNA regulators of those proteins, let-7a and miR-15a/miR-16-1, respectively.


Subject(s)
Cell Differentiation/drug effects , Gene Expression/drug effects , Leukemia, Promyelocytic, Acute/pathology , MicroRNAs/genetics , Tretinoin/pharmacology , Base Sequence , Blotting, Northern , Blotting, Western , DNA Primers , Humans , Luciferases/genetics , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction
13.
Gene Ther ; 15(10): 730-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18401438

ABSTRACT

Mesenchymal stem cells (MSC) exhibit tropism for sites of tissue damage as well as the tumor microenvironment. Many of the same inflammatory mediators that are secreted by wounds are found in the tumor microenvironment and are thought to be involved in attracting MSC to these sites. Cell migration is dependent on a multitude of signals ranging from growth factors to chemokines secreted by injured cells and/or respondent immune cells. MSC are likely to have chemotactic properties similar to other immune cells that respond to injury and sites of inflammation. Thus, the well-described model of leukocyte migration can serve as a reasonable example to facilitate the identification of factors involved in MSC migration. Understanding the factors involved in regulating MSC migration to tumors is essential to ultimately develop novel clinical strategies aimed at using MSC as vehicles to deliver antitumor proteins or suppress MSC migration to reduce tumor growth. For example, radiation enhances inflammatory signaling in the tumor microenvironment and may be used to potentiate site-specific MSC migration. Alternatively, restricting the migration of the MSC to the tumor microenvironment may prevent competent tumor-stroma formation, thereby hindering the growth of the tumor. In this review, we will discuss the role of inflammatory signaling in attracting MSC to tumors.


Subject(s)
Genetic Therapy/methods , Mesenchymal Stem Cell Transplantation/methods , Neoplasms/therapy , Animals , Cell Movement , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Humans , Inflammation Mediators/physiology , Mesenchymal Stem Cells/physiology , Neoplasms/immunology
14.
Cell Death Differ ; 14(8): 1443-56, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17464329

ABSTRACT

The chimeric fusion protein AML1-ETO, created by the t(8;21) translocation, recruits histone deacetylase (HDAC) to AML1-dependent promoters, resulting in transcriptional repression of the target genes. We analyzed the transcriptional changes in t(8;21) Kasumi-1 AML cells in response to the HDAC inhibitors, depsipeptide (FK228) and suberoylanilide hydroxamic acid (SAHA), which induced marked growth inhibition and apoptosis. Using cDNA array, annexin A1 (ANXA1) was identified as one of the FK228-induced genes. Induction of ANXA1 mRNA was associated with histone acetylation in ANXA1 promoter and reversal of the HDAC-dependent suppression of C/EBPalpha by AML1-ETO with direct recruitment of C/EBPalpha to ANXA1 promoter. This led to increase in the N-terminal cleaved isoform of ANXA1 protein and accumulation of ANXA1 on cell membrane. Neutralization with anti-ANXA1 antibody or gene silencing with ANXA1 siRNA inhibited FK228-induced apoptosis, suggesting that the upregulation of endogenous ANXA1 promotes cell death. FK228-induced ANXA1 expression was associated with massive increase in cell attachment and engulfment of Kasumi-1 cells by human THP-1-derived macrophages, which was completely abrogated with ANXA1 knockdown via siRNA transfection or ANXA1 neutralization. These findings identify a novel mechanism of action of HDAC inhibitors, which induce the expression and externalization of ANXA1 in leukemic cells, which in turn mediates the phagocytic clearance of apoptotic cells by macrophages.


Subject(s)
Annexin A1/biosynthesis , Core Binding Factor Alpha 2 Subunit/metabolism , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Oncogene Proteins, Fusion/metabolism , Acetylation , Annexin A1/genetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Base Sequence , Cell Line, Tumor , Cell Proliferation/drug effects , DNA, Complementary/genetics , Depsipeptides/pharmacology , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Macrophages/physiology , Phagocytosis/drug effects , RUNX1 Translocation Partner 1 Protein , Up-Regulation/drug effects , Vorinostat
15.
Cell Death Differ ; 14(9): 1667-77, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17510658

ABSTRACT

Bcr-Abl is the cause of Philadelphia-positive (Ph(+)) leukemias and also constitutes their principal therapeutic target, as exemplified by dramatic effects of imatinib mesylate. However, mono-targeting of Bcr-Abl does not always achieve complete leukemia eradication, and additional strategies those enable complete elimination of leukemic cells are desired to develop. Here we demonstrate that INNO-406, a much more active Bcr-Abl tyrosine kinase inhibitor than imatinib, augments the activities of several proapoptotic Bcl-2 homology (BH)3-only proteins (Bim, Bad, Bmf and Bik) and induces apoptosis in Ph(+) leukemia cells via Bcl-2 family-regulated intrinsic apoptosis pathway. ABT-737, an inhibitor of antiapoptotic Bcl-2 and Bcl-X(L), greatly enhanced the apoptosis by INNO-406, even in INNO-406-less sensitive cells with Bcr-Abl point mutations except T315I mutation. In contrast, co-treatment with INNO-406 and other pharmacologic inducers of those BH3-only proteins, such as 17-allylaminogeldanamycin, an heat shock protein-90 inhibitor, or PS-341, a proteasome inhibitor, did not further increase the BH3-only protein levels or sensitize leukemic cells to INNO-406-induced apoptosis, suggesting a limit to how much expression levels of BH3-only proteins can be increased by anticancer agents. Thus, double-barrelled molecular targeting for Bcr-Abl-driven oncogenic signaling and the cell protection by antiapoptotic Bcl-2 family proteins may be the rational therapeutic approach for eradicating Ph(+) leukemic cells.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Nitrophenols/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/metabolism , Apoptosis/physiology , Benzamides , Benzoquinones/pharmacology , Biphenyl Compounds/metabolism , Boronic Acids/metabolism , Boronic Acids/pharmacology , Bortezomib , Cell Line, Transformed , Cell Line, Tumor , Humans , Imatinib Mesylate , Lactams, Macrocyclic/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Nitrophenols/metabolism , Piperazines/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcr/metabolism , Pyrazines/metabolism , Pyrazines/pharmacology , Pyrimidines/metabolism , Sulfonamides/metabolism
16.
Cytotherapy ; 10(7): 657-67, 2008.
Article in English | MEDLINE | ID: mdl-18985472

ABSTRACT

Recent progress in the research of mesenchymal stromal cells/multipotent stromal cells (MSC) has revealed numerous beneficial innate characteristics, suggesting potential value in an array of cellular therapies. MSC are easily isolated from bone marrow (BM), fat and other tissues, and are readily propagated in vitro. Transplanted/injected MSC have been shown to migrate to a variety of organs and tissues; however, sites of inflammation and pathology elicit enhanced MSC homing for tissue remodeling and repair. Tumors utilize many of the same inflammatory mediators uncovered in wound healing and likewise provide a site for preferential MSC homing. Although incorporation into the tumor microenvironment is apparent, the role of recruited MSC in the tumor microenvironment remains unclear. Some published studies have shown enhancement of tumor growth and development, perhaps through immunomodulatory and pro-angiogenic properties, while others have shown no apparent effect or have demonstrated inhibition of tumor growth and extended survival. This controversy remains at the forefront as clinical applications of MSC commence in anti-tumor therapies as well as as adjuncts to stem cell transplantation and in ameliorating graft-versus-host disease. Careful analysis of past studies and thoughtful design of future experiments will help to resolve the discrepancies in the field and lead to clinical utility of MSC in disease treatment. This review highlights the current theories of the role of MSC in tumors and explores current controversies.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Neoplasms/therapy , Animals , Humans , Mice , Neoplasms/immunology , Stromal Cells/immunology , Stromal Cells/transplantation , Wound Healing/immunology
17.
Oncogene ; 25(32): 4483-90, 2006 Jul 27.
Article in English | MEDLINE | ID: mdl-16547503

ABSTRACT

The fundamental basis for immunotherapy of leukemia is that leukemic cells express specific antigens that are not expressed by normal hematopoietic cells. However, the host immune system appears to be tolerant to leukemia cells. To overcome this tolerance, we vaccinated immunocompetent mice with murine leukemia cells (WEHI-3B and BCR-ABL+ 32D cells) transduced with a specifically constructed transmembrane form of granulocyte-macrophage colony-stimulating factor (tmGM-CSF). The transduced cells expressed tmGM-CSF on the cell-surface. To determine whether tmGM-CSF-expressing WEHI-3B leukemia cells would prevent leukemia formation as a vaccine, immunocompetent mice (BALB/c and C3H/HEJ) were immunized with lethally irradiated murine leukemia cells expressing cell-surface tmGM-CSF before challenging mice with murine leukemia cells. Two immunocompetent mouse models were investigated, either WEHI-3B cells in BALB/c mice or BCR-ABL+ 32D cells in C3H/HEJ mouse. The results showed that 100% of WEHI-3B/tmGM-CSF-vaccinated BALB/c mice and about 65% of 32D+ BCR-ABL/tmGM-CSF-vaccinated C3H/HEJ mice were protected from leukemia after leukemia cell challenge, whereas all non-vaccinated mice succumbed to leukemia. Spleen and marrow cell suspensions from vaccinated mice challenged with WEHI-3B cells lacked detectable GFP+ WEHI-3B cells at 82 days post-challenge. A significant delay of death was observed in C3H/HEJ mice challenged with the very aggressive DA-1 cell line expressing BCR-ABL. Vaccination of mice with WEHI-3B/CD40L cells protected 80% of the mice from the WEHI-3B challenge. Notably, 60% of the WEHI-3B/BALB/c mice were also protected from leukemia when WEHI-3B/tmGM-CSF vaccination was carried out after the leukemia challenge. In order to determine whether cellular immunity is involved in this vaccine-mediated protection, either CD4+ or CD8+ T cells were depleted from mice after the WEHI-3B/tmGM-CSF vaccination. The results indicate that CD8+ T-cells mediated the protective immune response provided by the irradiated tmGM-CSF-expressing WEHI-3B cells. In addition, vaccination of nude mice did not provide protection from WEHI-3B leukemia induction. Importantly, 80% of non-vaccinated mice were also protected from a WEHI-3B cell challenge after receiving spleen cells from vaccinated mice 1 day before challenge with leukemia cells. These results indicate that overexpression of tmGM-CSF on the leukemia cell-surface can enhance the recognition of leukemic cells by CD8+ T cells, and can either prevent or significantly delay leukemia induction. These findings suggest that injection of irradiated leukemia cells expressing cell-surface-bound GM-CSF has the potential as an immunological approach to treat leukemia.


Subject(s)
Cancer Vaccines/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunotherapy, Adoptive , Leukemia, Myelomonocytic, Acute/prevention & control , Animals , CD40 Ligand/biosynthesis , CD40 Ligand/genetics , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line , Cell Line, Tumor , Disease Models, Animal , Female , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Leukemia, Myelomonocytic, Acute/immunology , Leukemia, Myelomonocytic, Acute/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Nude , Transduction, Genetic
18.
J Clin Invest ; 108(6): 851-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11560954

ABSTRACT

The mitogen-activated protein kinase (MAPK) pathway regulates growth and survival of many cell types, and its constitutive activation has been implicated in the pathogenesis of a variety of malignancies. In this study we demonstrate that small-molecule MEK inhibitors (PD98059 and PD184352) profoundly impair cell growth and survival of acute myeloid leukemia (AML) cell lines and primary samples with constitutive MAPK activation. These agents abrogate the clonogenicity of leukemic cells but have minimal effects on normal hematopoietic progenitors. MEK blockade also results in sensitization to spontaneous and drug-induced apoptosis. At a molecular level, these effects correlate with modulation of the expression of cyclin-dependent kinase inhibitors (p27(Kip1) and p21(Waf1/CIP1)) and antiapoptotic proteins of the inhibitor of apoptosis proteins (IAP) and Bcl-2 families. Interruption of constitutive MEK/MAPK signaling therefore represents a promising therapeutic strategy in AML.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Tumor Suppressor Proteins , Apoptosis/drug effects , Benzamides/pharmacology , Cell Cycle/drug effects , Cell Cycle Proteins/genetics , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/genetics , Cytarabine/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Leukemia, Myeloid, Acute/pathology , Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction , Tumor Cells, Cultured , X-Linked Inhibitor of Apoptosis Protein
19.
J Clin Invest ; 101(5): 1012-9, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9486971

ABSTRACT

Retinoids, including retinol and retinoic acid derivatives, maintain the normal growth and differentiation of human bronchial epithelial (HBE) cells and are under investigation as agents for lung cancer prevention. In this study, we examined the biologic effects of retinoids on normal HBE cells and the molecular mechanisms of retinoid actions. At a dose of 10(-6) M, all-trans retinoic acid (t-RA) suppressed the proliferation of normal HBE cells, which accumulated in the G0 phase. No evidence of programmed cell death was observed. The class of retinoid nuclear receptor that mediated the growth arrest was explored. Normal HBE cell growth was suppressed by a retinoid that selectively activates retinoic acid receptors but not by one that activates retinoid X receptors. The E2F transcription factor has demonstrated a role in G0 entry through transcriptional suppression of genes that induce cell cycle progression. To investigate the role of E2F in retinoid signaling, transient transfection assays were performed using reporter plasmids containing E2F-binding sites. Findings from these experiments suggested that t-RA treatment converted E2F into a transcriptional suppressor. Supporting this possibility, t-RA inhibited the expression of the E2F target genes B-myb, cyclin A, and cyclin E. Further, t-RA increased the levels of nuclear E2F-4, p107, and p130 and enhanced the binding of E2F-4 to p107, which have been associated with the conversion of E2F into a transcriptional suppressor in other cells. These findings point to retinoic acid receptor- and E2F-dependent pathways as potential mediators of retinoid-induced growth arrest in normal HBE cells and have implications for the use of retinoids in clinical trials on the prevention of lung cancer.


Subject(s)
Bronchi/growth & development , Bronchi/metabolism , Carrier Proteins , Cell Cycle Proteins , Epithelial Cells/metabolism , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Tretinoin/metabolism , Apoptosis , Blotting, Northern , Blotting, Western , Bronchi/cytology , Cells, Cultured , DNA-Binding Proteins/metabolism , E2F Transcription Factors , E2F4 Transcription Factor , Epithelial Cells/cytology , Epithelial Cells/drug effects , Genes, Reporter , Humans , Plasmids , Precipitin Tests , Resting Phase, Cell Cycle , Retinoblastoma-Binding Protein 1 , Retinoid X Receptors , Rhodamines/metabolism , Signal Transduction/genetics , Transcription Factor DP1 , Transcription Factors/immunology , Transcription, Genetic , Transfection , Tretinoin/immunology , Tretinoin/pharmacology
20.
Leukemia ; 20(4): 610-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16482209

ABSTRACT

17-Allylamino-17-demethoxygeldanamycin (17-AAG) is a new anticancer agent currently in clinical trials. The ability of 17-AAG to abrogate the function of heat-shock protein Hsp90 and modulate cellular sensitivity to anticancer agents has prompted recent research to use this compound in drug combination therapy. Here we report that 17-AAG has striking opposite effects on the activity of arsenic trioxide (ATO) and ara-C. Combination of 17-AAG with ATO exhibited a synergistic effect in leukemia cells, whereas coincubation of 17-AAG and ara-C showed antagonistic activity. Mechanistic studies revealed that ATO exerted cytotoxic action by reactive oxygen species generation, and activated Akt survival pathway. 17-AAG abrogated Akt activation and enhanced the activity of ATO. In contrast, treatment of leukemia cells with 17-AAG caused a G1 arrest, a decrease in DNA synthesis and reduced ara-C incorporation into DNA, leading to antagonism. The ability of 17-AAG to enhance the antileukemia activity of ATO was further demonstrated in primary leukemia cells isolated from patients with acute myeloid leukemia and chronic lymphocytic leukemia, including cells from refractory patients. Our data suggest that combination of 17-AAG and ATO may be an effective therapeutic regimen. Caution should be exercised in using 17-AAG together with ara-C, as their combination effects are schedule dependent.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Benzoquinones/pharmacology , Cytarabine/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Myeloid/metabolism , Oxides/pharmacology , Acute Disease , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals/antagonists & inhibitors , Cytarabine/antagonists & inhibitors , DNA/biosynthesis , DNA/drug effects , DNA/metabolism , Drug Administration Schedule , Drug Antagonism , Drug Screening Assays, Antitumor , Drug Synergism , Drug Therapy, Combination , G1 Phase/drug effects , HSP90 Heat-Shock Proteins/physiology , HeLa Cells , Humans , Jurkat Cells , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Myeloid/drug therapy , Oxides/antagonists & inhibitors , Sensitivity and Specificity , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL