Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Invest New Drugs ; 31(4): 1023-34, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23397500

ABSTRACT

Survivin is expressed in tumor cells, including acute myeloid leukemia (AML), regulates mitosis, and prevents tumor cell death. The antisense oligonucleotide sodium LY2181308 (LY2181308) inhibits survivin expression and may cause cell cycle arrest and restore apoptosis in AML. In this study, the safety, pharmacokinetics, and pharmacodynamics/efficacy of LY2181308 was examined in AML patients, first in a cohort with monotherapy (n = 8) and then post-amendment in a cohort with the combination of cytarabine and idarubicin treatment (n = 16). LY2181308 was administered with a loading dosage of three consecutive daily infusions of 750 mg followed by weekly intravenous (IV) maintenance doses of 750 mg. Cytarabine 1.5 g/m(2) was administered as a 4-hour IV infusion on Days 3, 4, and 5 of Cycle 1, and idarubicin 12 mg/m(2) was administered as a 30-minute IV infusion on Days 3, 4, and 5 of Cycle 1. Cytarabine and idarubicin were administered on Days 1, 2, and 3 of each subsequent 28-day cycle. Reduction of survivin was evaluated in peripheral blasts and bone marrow. Single-agent LY2181308 was well tolerated and survivin was reduced only in patients with a high survivin expression. In combination with chemotherapy, 4/16 patients had complete responses, 1/16 patients had incomplete responses, and 4/16 patients had cytoreduction. Nine patients died on study: 6 (monotherapy), 3 (combination). LY2181308 alone is well tolerated in patients with AML. In combination with cytarabine and idarubicin, LY2181308 does not appear to cause additional toxicity, and has shown some clinical benefit needing confirmation in future clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytarabine/therapeutic use , Idarubicin/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Oligonucleotides/adverse effects , Oligonucleotides/pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/blood , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cytarabine/adverse effects , Cytarabine/blood , Cytarabine/pharmacokinetics , Demography , Female , Humans , Idarubicin/adverse effects , Idarubicin/blood , Idarubicin/pharmacokinetics , Inhibitor of Apoptosis Proteins/metabolism , Leukemia, Myeloid, Acute/blood , Male , Middle Aged , Oligonucleotides/blood , Oligonucleotides/therapeutic use , Oligonucleotides, Antisense/adverse effects , Oligonucleotides, Antisense/blood , Oligonucleotides, Antisense/pharmacokinetics , Oligonucleotides, Antisense/therapeutic use , Recurrence , Survivin , Treatment Outcome
2.
Cancer Manag Res ; 3: 157-75, 2011.
Article in English | MEDLINE | ID: mdl-21625399

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) is a life-threatening malignancy with limited treatment options in chemotherapy-refractory patients. A first-in-human dose study was designed to investigate a safe and biologically effective dose range for LY2457546, a novel multikinase inhibitor, in patients with relapsed AML. METHODS: In this nonrandomized, open-label, dose escalation Phase I study, LY2457546 was administered orally once a day. Safety, pharmacokinetics, changes in phosphorylation of target kinases in AML blasts, and risk of drug-drug interactions (DDI) were assessed. RESULTS: Five patients were treated at the starting and predicted minimal biologically effective dose of 50 mg/day. The most commonly observed adverse events were febrile neutropenia, epistaxis, petechiae, and headache. The majority of adverse events (81%) were Grade 1 or 2. One patient had generalized muscle weakness (Grade 3), which was deemed to be a dose-limiting toxicity. Notably, the pharmacokinetic profile of LY2457546 showed virtually no elimination of LY2457546 within 24 hours, and thus prevented further dose escalation. No significant DDI were observed. Ex vivo flow cytometry studies showed downregulation of the phosphoproteins, pcKIT, pFLT3, and pS6, in AML blasts after LY2457546 administration. No medically relevant responses were observed in the five treated patients. CONCLUSION: No biologically effective dose could be established for LY2457546 in chemotherapy-resistant AML patients. Lack of drug clearance prevented safe dose escalation, and the study was terminated early. Future efforts should be made to develop derivatives with a more favorable pharmacokinetic profile.

3.
J Biomed Opt ; 14(6): 064036, 2009.
Article in English | MEDLINE | ID: mdl-20059274

ABSTRACT

We characterize T- and B-lymphocytes from several donors, determining cell diameter, ratio of nucleus to cell diameter, and refractive index of the nucleus and cytoplasm for each individual cell. We measure light-scattering profiles with a scanning flow cytometer and invert the signals using a coated sphere as an optical model of the cell and by relying on a global optimization technique. The main difference in morphology of T- and B-lymphocytes is found to be the larger mean diameters of the latter. However, the difference is smaller than the natural biological variability of a single cell. We propose nuclear inhomogeneity as a possible reason for the deviation of measured light-scattering profiles from real lymphocytes from those obtained from the coated sphere model.


Subject(s)
B-Lymphocytes/cytology , Light , Models, Biological , Scattering, Radiation , T-Lymphocytes/cytology , Algorithms , Bayes Theorem , Cell Nucleus , Cell Shape , Cell Size , Flow Cytometry , Humans , Microscopy, Confocal , Refractometry
4.
J Immunol Methods ; 332(1-2): 31-40, 2008 Mar 20.
Article in English | MEDLINE | ID: mdl-18255093

ABSTRACT

OBJECTIVES: The validity of endothelial progenitor cells as biomarkers and their therapeutic potential depend on the accuracy of techniques used for enumeration. This study assessed the agreement between 6 flow cytometric methods and a CFU assay used for EPC quantification. METHODS: Two blood samples were obtained from 30 healthy volunteers (60 samples). CD34+/VEGFR2+ cells were analyzed with flow cytometry, starting from whole blood (A-C) or PBMC (D-F), using different gating strategies: A: lymphocyte gating; B and D: exclusion of autofluorescent cells (CD3 negative selection); C and E: exclusion of autofluorescence and cell aggregates (pulse shape analysis by FSCarea/FSCpeak); F: exclusion of autofluorescence, cell aggregates and non-nucleated cells (Draq 5). PBMC were cultured under endothelial cell conditions to assess CFU numbers. RESULTS: Moderate agreement was found between methods B-C and D-E (ICC 0.647 and 0.530). Comparison of methods B-D and C-E showed poor agreement (ICC 0.178 and 0.249). This was also the case for techniques that considerably differed with regard to gating strategies (A-B, A-F, B-F). CFU numbers did not correlate with flow cytometric quantification (all p>0.05). CONCLUSIONS: Agreement between methods for EPC quantification is moderate to poor, which may explain apparent controversies in literature. Although each protocol is highly reproducible, this study cautions against comparing study results gathered with different enumeration techniques.


Subject(s)
Colony-Forming Units Assay/methods , Endothelial Cells/cytology , Flow Cytometry/methods , Stem Cells/cytology , Adult , Cell Count , Female , Humans , Male , Reproducibility of Results
5.
J Biomed Opt ; 12(4): 044017, 2007.
Article in English | MEDLINE | ID: mdl-17867821

ABSTRACT

Quantitative data on cell structure, shape, and size distribution are obtained by optical measurement of normal peripheral blood granulocytes and lymphocytes in a cell suspension. The cell nuclei are measured in situ. The distribution laws of the cell and nuclei sizes are estimated. The data gained are synthesized to construct morphometric models of a segmented neutrophilic granulocyte and a lymphocyte. Models of interrelation between the cell and nucleus metric characteristics for granulocyte and lymphocyte are obtained. The discovered interrelation decreases the amount of cell-nucleus size combinations that have to be considered under simulation of cell scattering patterns. It allows faster analysis of light scattering to discriminate cells in a real-time scale. Our morphometric data meet the requirements of scanning flow cytometry dealing with the high rate analysis of cells in suspension. Our findings can be used as input parameters for the solution of the direct and inverse light-scattering problems in scanning flow cytometry, dispensing with a costly and time-consuming immunophenotyping of the cells, as well as in turbidimetry and nephelometry. The cell models developed can ensure better interpretations of scattering patterns for an improvement of discriminating capabilities of immunophenotyping-free scanning flow cytometry.


Subject(s)
Cell Count/methods , Flow Cytometry/methods , Granulocytes/cytology , Granulocytes/physiology , Lymphocytes/cytology , Lymphocytes/physiology , Models, Biological , Nephelometry and Turbidimetry/methods , Cell Size , Computer Simulation , Humans , Light , Scattering, Radiation
6.
Cell Biol Int ; 31(11): 1323-30, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17606388

ABSTRACT

The C-type lectin family is a group of animal proteins which can be distinguished from other lectins by the presence of a Ca2+-dependent carbohydrate recognition domain (CRD) in their protein sequence. They are classified into 17 groups according to their domain architecture and have a wide variety of functions. The human chondrolectin gene encodes transmembrane (CHODL, CHODLf) and soluble proteins (CHODLDeltaE, CHODLfDeltaE) belonging to the family of C-type lectins because of the presence of one CRD domain in their N-terminal region. The CHODL splice variants (CHODLf, CHODLDeltaE and CHODLfDeltaE) are differentially expressed in T lymphocytes. The transmembrane-containing isoform CHODLf is localized in the ER-Golgi apparatus. CHODLDeltaE and CHODLfDeltaE are devoid of the transmembrane domain and terminate in QDEL, an ER retention signal. In this paper we have investigated the expression of the CHODLDeltaE/CHODLfDeltaE protein. This variant localizes in the late endoplasmic reticulum. We detected the protein in spleen and tonsils in a small population of lymphocytes. Moreover, the isoform seems to be differentially expressed in thymocytes and lymphocytes suggesting an important biological function during T cell development.


Subject(s)
Lectins, C-Type/metabolism , Membrane Proteins/metabolism , T-Lymphocyte Subsets/metabolism , Animals , Cell Line , Gene Expression , Humans , Lectins, C-Type/chemistry , Lectins, C-Type/isolation & purification , Membrane Proteins/chemistry , Membrane Proteins/isolation & purification , Palatine Tonsil/cytology , Palatine Tonsil/metabolism , Protein Isoforms/metabolism , Spleen/cytology , Spleen/metabolism , T-Lymphocyte Subsets/cytology , Thymus Gland/cytology , Thymus Gland/metabolism
7.
New Phytol ; 175(1): 140-154, 2007.
Article in English | MEDLINE | ID: mdl-17547674

ABSTRACT

* To characterize plant cell cycle activation following Rhodococcus fascians infection, bacterial impact on cell cycle progression of tobacco BY-2 cells was investigated. * S-phase-synchronized BY-2 cells were cocultivated with R. fascians and cell cycle progression was monitored by measuring mitotic index, cell cycle gene expression and flow cytometry parameters. Cell cycle alteration was further investigated by cDNA-AFLP (amplified fragment length polymorphism). * It was shown that cell cycle progression of BY-2 cells was accelerated only upon infection with bacteria whose virulence gene expression was induced by a leafy gall extract. Thirty-eight BY-2 genes showed a differential expression within 6 h post-infection. Among these, seven were previously associated with specific plant cell cycle phases (in particular S and G2/M phases). Several genes also showed a differential expression during leafy gall formation. * R. fascians-infected BY-2 cells provide a simple model to identify plant genes related to leafy gall development. R. fascians can also be regarded as a useful biotic agent to alter cell cycle progression and, thereby, gain a better understanding of cell cycle regulation in plants.


Subject(s)
Gene Expression Regulation, Plant , Nicotiana/genetics , Nicotiana/microbiology , Plant Diseases/microbiology , Rhodococcus/pathogenicity , Amino Acid Sequence , Aphidicolin/metabolism , Cell Cycle/genetics , Cell Division , Cell Line , DNA, Complementary/genetics , DNA, Plant/genetics , Kinetics , Mitosis , Mitotic Index , Molecular Sequence Data , Plant Proteins/genetics , Polymorphism, Genetic , RNA, Messenger/genetics , RNA, Plant/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Nicotiana/cytology , Nicotiana/growth & development
8.
Neurosci Lett ; 410(2): 146-51, 2006 Dec 20.
Article in English | MEDLINE | ID: mdl-17095159

ABSTRACT

Although reactive oxygen species (ROS) at physiological concentrations are required for normal cell function, excessive production of ROS is detrimental to cells. Neuroglobin and cytoglobin are two globins, whose functions are still a matter of debate. A potential role in the detoxification of ROS is suggested. The influence of neuroglobin and cytoglobin on cell death after oxidative stress in human neuroblastoma SH-SY5Y cells was evaluated. Exposure of SH-SY5Y cells to paraquat or H(2)O(2) resulted in a concentration- and time-dependent induction of apoptotic and necrotic cell death. H(2)O(2) was 16 times more potent to induce cell death as compared to paraquat. SH-SY5Y cells transfected with plasmid DNA containing the neuroglobin or cytoglobin sequence showed enhanced survival after exposure to 300 microM H(2)O(2) for 24h as compared to untransfected controls. This finding suggests that neuroglobin and cytoglobin protect SH-SY5Y cells against oxidative stress-induced cell death.


Subject(s)
Gene Expression/physiology , Globins/metabolism , Nerve Tissue Proteins/metabolism , Oxidative Stress/physiology , Blotting, Western/methods , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Cytoglobin , Dose-Response Relationship, Drug , Flow Cytometry/methods , Gene Expression/drug effects , Herbicides/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Neuroblastoma , Neuroglobin , Oxidants/pharmacology , Oxidative Stress/drug effects , Paraquat/pharmacology , Time Factors
9.
Mol Cancer ; 5: 49, 2006 Oct 26.
Article in English | MEDLINE | ID: mdl-17067378

ABSTRACT

BACKGROUND: Cervical cancer is the second most common gynecological cancer amongst women world-wide. Despite optimized protocols, standard treatments still face several disadvantages. Therefore, research aims at the development of immune-based strategies using tumor antigen-loaded dendritic cells for the induction of cellular anti-tumor immunity. RESULTS: In this study, we used dendritic cells loaded with the HLA-A2-restricted HPV type 16 E711-20 peptide in order to induce an in vitro CD8+ T cell response. For this purpose, peptide-pulsed dendritic cells were co-cultured with autologous CD8+ T cells. After 5 weekly stimulations with peptide-pulsed mature dendritic cells, cultured T cells were analyzed for antigen specificity by an IFN-gamma ELISPOT assay. Using this ELISPOT assay, we were able to detect E7-specific IFN-gamma-secreting CD8+ T cells in 5/5 healthy donors. CONCLUSION: We show that peptide-pulsed mature dendritic cells are able to stimulate a HPV type 16 E7 peptide-specific immune response in vitro. These experiments describe an efficient culture protocol for antigen-specific T cells for use in pre-clinical vaccination research and confirm the need for sensitive T cell assays for detection of tumor-specific immune responses in vitro.


Subject(s)
Antigens, Viral, Tumor/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay/methods , Oncogene Proteins, Viral/immunology , Coculture Techniques , Female , Humans , Immunophenotyping , Interferon-gamma/biosynthesis , K562 Cells , Oligopeptides/immunology , Papillomavirus E7 Proteins , Papillomavirus Infections/immunology , Sensitivity and Specificity , Uterine Cervical Neoplasms/virology
10.
J Immunother ; 29(5): 512-23, 2006.
Article in English | MEDLINE | ID: mdl-16971807

ABSTRACT

Recently, it has become obvious that not only CD8 T-cells, but also CD4 T-helper cells are required for the induction of an effective, long-lasting cellular immune response. In view of the clinical importance of cytomegalovirus (CMV) and human immunodeficiency virus (HIV) infection, we developed 2 strategies to simultaneously reactivate viral antigen-specific memory CD4 and CD8 T-cells of CMV-seropositive and HIV-seropositive subjects using mRNA-electroporated autologous CD40-activated B cells. In the setting of HIV, we provide evidence that CD40-activated B cells can be cultured from HAART-naive HIV-1 seropositive patients. These cells not only express and secrete the HIV p24 antigen after electroporation with codon-optimized HIV-1 gag mRNA, but can also be used to in vitro reactivate Gag antigen-specific interferon-gamma-producing CD4 and CD8 autologous T-cells. For the CMV-specific approach, we applied mRNA coding for the pp65 protein coupled to the lysosomal-associated membrane protein-1 to transfect CD40-activated B cells to induce CMV antigen-specific CD4 and CD8 T-cells. More detailed analysis of the activated interferon-gamma-producing CMV pp65 tetramer positive CD8 T-cells revealed an effector memory phenotype with the capacity to produce interleukin-2. Our findings clearly show that the concomitant activation of both CD4 and CD8 (memory) T-cells using mRNA-electroporated CD40-B cells is feasible in CMV and HIV-1-seropositive persons, which indicates the potential value of this approach for application in cellular immunotherapy of infectious diseases.


Subject(s)
Antigens, Viral/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , HIV Infections/immunology , RNA, Messenger/immunology , Adult , Cells, Cultured , Electroporation , Feasibility Studies , Female , Gene Products, gag/immunology , HIV-1/immunology , Humans , Interleukin-2/metabolism , Lymphocyte Activation , Lysosomal-Associated Membrane Protein 1/immunology , Lysosomal Membrane Proteins/immunology , Male , Middle Aged , Phenotype , Phosphoproteins/immunology , RNA, Viral/immunology , Viral Matrix Proteins/immunology
11.
Semin Thromb Hemost ; 32(4 Pt 2): 307-40, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16810609

ABSTRACT

The clinical criteria according to the Polycythemia Vera Study Group (PVSG) do not distinguish between essential thrombocythemia (ET), thrombocythemia associated with early-stage polycythemia vera (PV) and prefibrotic chronic idiopathic myelofibrosis (CIMF). The criteria only classify the advanced stage of PV with increased red cell mass. The classification of myeloproliferative disorders (MPDs), proposed by the World Health Organization (WHO) in 2001, is a compromise of the clinical PVSG and WHO bone marrow criteria, and excludes early stages of ET and PV. The updated European clinical and pathological criteria combine the WHO bone marrow criteria with established and new clinical, laboratory, biological, and molecular MPD markers. This allows clinicians and pathologists to diagnose early-stage MPD and to differentiate ET, PV, and prefibrotic chronic idiopathic myelofibrosis (CIMF). Depending on laboratory tests and diagnostic criteria used, the population of the MPD patients defined as ET, PV, and CIMF are heterogeneous at the clinical, laboratory, and biological and pathological levels. The recent discovery of the JAK2 V617F mutation, which is the cause of a distinct trilinear MPD in its manifold clinical manifestations during long-term follow-up, increases the specificity of a positive JAK2 V617F polymerase chain reaction (PCR) test for the diagnosis of MPD (near 100%), but only half of the ET and CIMF patients according to the PVSG (sensitivity 50%) and the majority of PV patients (sensitivity 95%) are JAK2 V617F positive. A comparison of the laboratory features of JAK2 V617-positive and JAK2 wild-type ET patients clearly showed that JAK2 V617-positive ET is characterized by higher values for hemoglobin, hematocrit, and neutrophil counts; lower values for serum erythropoietin (EPO) levels, serum ferritin, and mean corpuscular volume; and by increased cellularity of the bone marrow in biopsy material. This indicates that JAK2 V617-positive ET patients, diagnosed according to the PVSG criteria, represent a "forme fruste of PV" consistent with early PV mimicking ET (JAK2 V617F trilinear MPD). In contrast, the JAK2 wild-type ET patients had significantly higher platelet counts and usually had a clinical picture of ET with normal serum EPO levels, PRV-1 expression, and leukocyte alkaline phosphatase score, and a typical WHO ET bone marrow picture. The clinical and pathological data on JAK2 V617F-positive MPD patients suggest that the JAK2 V617F mutation defines one disease entity with several sequential steps of ET, PV, and secondary myelofibrosis during long-term follow-up, and that the wild-type JAK2 MPDs may represent another distinct entity with a related but different molecular etiology. MPD-specific markers such as serum EPO, endogenous erythroid colony formation (EEC), and JAK2 V617F have high specificities, but the sensitivities are not high enough to detect the early stages of the MPDs, ET, PV, and prefibrotic CIMF. Bone marrow histopathology in addition to clinical, laboratory, biological, and molecular markers, including the JAK2 V617 PCR test, serum EPO, PRV-1, EEC, LAP score, peripheral blood parameters, and spleen size on echogram will detect the early stages of MPD and allows diagnostic differentiation of the three primary MPDs (ET, PV, and CIMF) in both JAK2 V617F-positive and JAK2 wild-type MPD patients.


Subject(s)
Myeloproliferative Disorders/diagnosis , Philadelphia Chromosome , World Health Organization , Biomarkers/blood , Bone Marrow/pathology , Chronic Disease , Diagnosis, Differential , Europe , History, 21st Century , Humans , Janus Kinase 2 , Myeloproliferative Disorders/blood , Myeloproliferative Disorders/classification , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/history , Myeloproliferative Disorders/pathology , Point Mutation , Practice Guidelines as Topic , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , World Health Organization/history
12.
Coron Artery Dis ; 17(3): 243-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16728874

ABSTRACT

BACKGROUND: Dendritic cells are potent antigen-presenting and immune modulating cells that have been implicated in the development of atherosclerosis. In human blood, two distinct lineages are distinguished: plasmacytoid dendritic cells and myeloid dendritic cells. Although dendritic cells have been described in atherosclerotic plaques, no information exists concerning circulating blood dendritic cells in atherosclerosis. This study aims to evaluate the number of circulating dendritic cells in patients with coronary artery disease. The relation with the extent of coronary artery disease, the clinical syndrome and with a marker of inflammation will be documented. METHODS: Patients with angiographically proven coronary artery disease (n=18) and age and sex-matched controls (n=18) were included. Myeloid dendritic cells and plasmacytoid dendritic cells were detected with the specific blood dendritic cell antigens, blood dendritic cell antigen-1 and blood dendritic cell antigen-2, respectively. RESULTS: Absolute and relative numbers of circulating plasmacytoid dendritic cells were significantly lower in patients with coronary artery disease (5722+/-601/ml and 0.08+/-0.01%) than in controls (12,640+/-1289/ml and 0.21+/-0.02%). Plasmacytoid dendritic cells were more decreased in patients with troponin-positive unstable coronary syndromes than in patients with low troponin values, and tended to be lower in more extensive coronary artery disease. Absolute myeloid dendritic cells numbers tended to be reduced in patients, whereas relative numbers were significantly decreased: 11,857+/-1895/ml versus 15,226+/-928/ml and 0.17+/-0.03% versus 0.26+/-0.01% in controls. CONCLUSIONS: The present study shows a significant decrease of circulating blood dendritic cell antigen-2 positive plasmacytoid dendritic cells in patients with coronary artery disease. The decrease tended to be more pronounced in unstable coronary syndromes and extensive coronary artery disease, suggesting a possible role of dendritic cells in plaque progression and rupture.


Subject(s)
Coronary Artery Disease/blood , Coronary Stenosis/blood , Dendritic Cells/metabolism , Adult , Aged , Biomarkers/blood , C-Reactive Protein/metabolism , Case-Control Studies , Coronary Angiography , Coronary Artery Disease/diagnostic imaging , Coronary Stenosis/diagnostic imaging , Female , Humans , Immunophenotyping , Inflammation Mediators/blood , Male , Middle Aged , Research Design , Troponin I/blood
13.
Semin Thromb Hemost ; 32(3): 174-207, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16673274

ABSTRACT

Microvascular disturbances in essential thrombocythemia (ET) and polycythemia vera (PV), including erythromelalgia, and atypical and typical transient cerebral, ocular, and coronary ischemic attacks, are caused by platelet-mediated transient and occlusive thrombosis in the end-arterial circulation. ET patients with microvascular disturbances have shortened platelet survival, increased beta-thromboglobulin (beta-TG), platelet factor 4 (PF4), and thrombomodulin (TM) levels, and increased urinary thromboxane B2 (TXB2) excretion, indicating platelet-mediated thrombotic processes. Inhibition of platelet cyclooxygenase-1 by aspirin is followed by relief of microvascular disturbances; correction of shortened platelet survival; correction of increased plasma beta-TG, PF4, and TM levels; and correction of increased TXB2 excretion to normal. In PV associated with thrombocythemia, increased hematocrit and whole blood viscosity aggravate the platelet-mediated microvascular syndrome of thrombocythemia to produce major arterial and venous thrombotic complications. Correction of hematocrit to normal by phlebotomy will reduce the major arterial and venous thrombotic complications, but fails to prevent the platelet-mediated microvascular circulation disturbances in PV patients because thrombocythemia persists. Complete relief and prevention of microvascular and major thrombosis in ET and PV patients, in addition to phlebotomy, are obtained by treatment with aspirin and not with coumarin. The discovery of JAK2 V617F gain of function mutation in patients with myeloproliferative disorders (MPDs) expands our insights into the molecular etiology and biological features of ET, PV, and chronic idiopathic myelofibrosis (CIMF). The current concept is that heterozygous JAK2 V617F mutation with increased kinase activity is enough for megakaryocyte proliferation and increased hypersensitive platelets with no or slightly increased erythropoiesis in ET and in early PV mimicking ET. Homozygous JAK2 mutation with pronounced kinase activity is associated with trilinear megakaryocyte, erythroid, and granulocytic myeloproliferation, myeloid metaplasia, and secondary myelofibrosis (MF), with the most frequent clinical picture of classical PV complicated by major thrombosis in addition to the platelet-mediated microvascular thrombotic syndrome of thrombocythemia. The positive predictive value of a JAK2 V617F polymerase chain reaction test for the diagnosis of MPDs is high (near to 100%), but only half of ET and MF (sensitivity 50%) and the majority of PV (sensitivity 85 to 97%) are JAK2 V617F positive. Bone marrow histopathology, when used in combination with specific markers such as serum erythropoietin, PRV-1, endogenous erythroid colony formation, peripheral blood parameters and red cell mass, has a high sensitivity and specificity (near 100%) to detect the early and overt stages of the MPDs and to differentiate between ET, PV, and CIMF in both JAK2 V617F-positive and -negative MPDs.


Subject(s)
Hemorrhage/etiology , Polycythemia Vera/etiology , Thrombocythemia, Essential/etiology , Thrombosis/etiology , Blood Platelets/physiology , Erythropoiesis , Hemorrhage/drug therapy , Humans , Polycythemia Vera/drug therapy , Thrombocythemia, Essential/complications , Thrombocythemia, Essential/drug therapy , Thrombosis/complications , Thrombosis/drug therapy
14.
J Immunother ; 29(2): 107-21, 2006.
Article in English | MEDLINE | ID: mdl-16531812

ABSTRACT

Current antiviral drugs do not fully reconstitute the specific antiviral immune control in chronically human immunodeficiency virus (HIV)-1-infected patients or in cytomegalovirus (CMV)-infected patients after hematopoietic stem cell transplantation. Therefore, immunotherapy in which the patient's immune system is manipulated to enhance antiviral immune responses has become a promising area of viral immunology research. In this review, an overview is provided on the cellular immunotherapy strategies that have been developed for HIV infection and CMV reactivation in immunocompromised patients. As an introduction, the mechanisms behind the cellular immune system and their importance for the development of a workable immunotherapy approach are discussed. Next, the focus is shifted to the immunopathogenesis of CMV and HIV-1 infections to correlate these findings with the concepts and ideas behind the viral-specific immunotherapies discussed. Current and future perspectives of active and passive cellular immunotherapy for the treatment of CMV and HIV-1 infections are reviewed. Finally, pitfalls and key issues with regard to the development of immunotherapy protocols that can be applied in a clinical setting are addressed.


Subject(s)
Cytomegalovirus Infections/therapy , HIV Infections/therapy , Immunotherapy , Adoptive Transfer , Cytomegalovirus Infections/immunology , Dendritic Cells/immunology , HIV Infections/immunology , Humans , Immunity, Cellular , Immunosuppression Therapy , Organ Transplantation , T-Lymphocyte Subsets/immunology , Transplantation Immunology
15.
Blood ; 107(5): 1818-27, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16263796

ABSTRACT

Infection with human immunodeficiency virus type 1 (HIV-1) is characterized by dysfunction of HIV-1-specific T cells. To control the virus, antigen-loaded dendritic cells (DCs) might be useful to boost and broaden HIV-specific T-cell responses. In the present study, monocyte-derived DCs from nontreated HIV-1-seropositive patients were electroporated with codon-optimized ("humanized") mRNA encoding consensus HxB-2 (hHXB-2) Gag protein. These DCs elicited a strong HIV-1 Gag-specific interferon-gamma (IFN-gamma) response by an HLA-A2-restricted CD8+ T-cell line. Moreover, hHXB-2 gag mRNA-electroporated DCs also triggered IFN-gamma secretion by autologous peripheral blood mononuclear cells (PBMCs), CD4+ T cells, and CD8+ T cells from all patients tested. Next, a novel strategy was developed using autologous virus sequences. Significant specific IFN-gamma T-cell responses were induced in all patients tested by DCs electroporated with patients' autologous polymerase chain reaction (PCR)-amplified and in vitro-transcribed proviral and plasma viral mRNA encoding either Gag or Env. The stimulatory effect was seen on PBMCs, CD8+ T cells, and CD4+ T cells, demonstrating both major histocompatibility complex (MHC) class I and MHC class II antigen presentation. Moreover, a significant interleukin-2 (IL-2) T-cell response was induced by DCs electroporated with hHxB-2 or proviral gag mRNA. These findings open a major perspective for the development of patient-specific immunotherapy for HIV-1 disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Gene Products, gag/immunology , Glycoproteins/immunology , HIV Seropositivity/immunology , HIV-1/immunology , Viral Envelope Proteins/immunology , Adoptive Transfer/methods , Adult , Cell Line , Dendritic Cells/transplantation , Electroporation , Female , Gene Products, gag/genetics , Glycoproteins/genetics , HIV Seropositivity/therapy , HIV-1/genetics , HLA-A2 Antigen/immunology , Humans , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Male , Middle Aged , Monocytes/immunology , RNA, Viral/genetics , RNA, Viral/immunology , Transplantation, Autologous , Viral Envelope Proteins/genetics
16.
Mol Cell Neurosci ; 30(3): 316-25, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16139508

ABSTRACT

We performed differential gene expression profiling in the peripheral nervous system by comparing the transcriptome of sensory neurons with the transcriptome of lower motor neurons. Using suppression subtractive cDNA hybridization, we identified 5 anonymous transcripts with a predominant expression in sensory neurons. We determined the gene structures and predicted the functional protein domains. The 4930579P15Rik gene encodes for a novel inhibitor of protein phosphatase-1 and 9030217H17Rik was found to be the mouse gene synaptopodin. We performed in situ hybridization for all genes in mouse embryos, and found expression predominantly in the primary class of sensory neurons. Expression of 4930579P15Rik and synaptopodin was restricted to craniospinal sensory ganglia. Neither synaptopodin, nor any known family member of 4930579P15Rik, has ever been described in sensory neurons. The identification of protein domains and expression patterns allows further functional analysis of these novel genes in relation to the development and biology of sensory neurons.


Subject(s)
Ganglia, Spinal/embryology , Ganglia, Spinal/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurons, Afferent/metabolism , Animals , Chromosome Mapping , DNA, Complementary/analysis , DNA, Complementary/genetics , Ganglia, Spinal/cytology , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Genetic Markers/genetics , Genomic Library , Mice , Nerve Tissue Proteins/isolation & purification , Nerve Tissue Proteins/metabolism , Neurons, Afferent/cytology , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 1
17.
Ann Hematol ; 84(10): 627-39, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16041532

ABSTRACT

Apoptosis or programmed cell death is a process with typical morphological characteristics including plasma membrane blebbing, cell shrinkage, chromatin condensation and fragmentation. A family of cystein-dependent aspartate-directed proteases, called caspases, is responsible for the proteolytic cleavage of cellular proteins leading to the characteristic apoptotic features, e.g. cleavage of caspase-activated DNase resulting in internucleosomal DNA fragmentation. Currently, two pathways for activating caspases have been studied in detail. One starts with ligation of a death ligand to its transmembrane death receptor, followed by recruitment and activation of caspases in the death-inducing signalling complex. The second pathway involves the participation of mitochondria, which release caspase-activating proteins into the cytosol, thereby forming the apoptosome where caspases will bind and become activated. In addition, two other apoptotic pathways are emerging: endoplasmic reticulum stress-induced apoptosis and caspase-independent apoptosis. Naturally occurring cell death plays a critical role in many normal processes like foetal development and tissue homeostasis. Dysregulation of apoptosis contributes to many diseases, including cancer. On the other hand, apoptosis-regulating proteins also provide targets for drug discovery and new approaches to the treatment of cancer.


Subject(s)
Apoptosis , Caspases/metabolism , DNA Fragmentation , Neoplasms/metabolism , Signal Transduction , Animals , Apoptosis/physiology , Chromatin Assembly and Disassembly/physiology , DNA Fragmentation/physiology , Enzyme Activation/physiology , Fetal Development/physiology , Homeostasis/physiology , Humans , Mitochondria/metabolism , Neoplasms/therapy , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/physiology , fas Receptor/metabolism
18.
Curr Biol ; 15(9): 824-7, 2005 May 10.
Article in English | MEDLINE | ID: mdl-15886099

ABSTRACT

In most plants and animals, a consistent relationship exists between the DNA content of a cell and its metabolic activity. The male-haploid sex determination of Hymenoptera and other arthropods may therefore impose a particular selective pressure upon males, which must evolve adaptations to cope with a genomic DNA reduced by half compared with that of females. Here, we show that a nuclear DNA content similar to that of females is restored in muscles of males in all hymenopteran lineages tested except the most basal one (Xyelidae). This doubling of DNA content in males does not occur in other haplodiploid insects, such as thrips (Thysanoptera) and whiteflies (Sternorrhyncha). These results indicate that this adaptation probably occurred early in hymenopteran history, possibly because males acquired strong flying and dispersal abilities.


Subject(s)
Adaptation, Physiological/genetics , Cell Nucleus/genetics , DNA/metabolism , Hymenoptera/genetics , Muscles/cytology , Ploidies , Animals , Flow Cytometry , Fluorescence , Male , Phylogeny
19.
Environ Pollut ; 132(3): 385-94, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15325454

ABSTRACT

We carried out an immunotoxicological field study of wood mice in three populations along a heavy metal pollution gradient. Heavy metal concentrations in liver tissue indicated that exposure to silver, arsenic, cadmium, cobalt and lead decreased with increasing distance from a non-ferrous smelter. Host resistance to the endoparasite Heligmosomoides polygyrus decreased with increasing exposure, while the abundance of tick larvae and the nematode Syphacia stroma was unrelated to heavy metal exposure. Spleen mass was increased at the intermediate and the most polluted sites and was positively correlated with the number of H. polygyrus and tick larvae. Proportion of early apoptotic leukocytes increased towards the smelter and was positively related to cadmium exposure. Red and white blood cell counts and lysozyme activity showed no relationship with metal exposure. All together, our observations suggest negative effects of heavy metal exposure on the immune function of wood mice under field conditions.


Subject(s)
Environmental Exposure/adverse effects , Metallurgy , Metals, Heavy/analysis , Muridae , Animals , Apoptosis/immunology , Blood Cell Count , Larva , Leukocytes/immunology , Liver/chemistry , Liver/immunology , Metals, Heavy/toxicity , Muridae/immunology , Muridae/metabolism , Nematospiroides dubius , Oxyuroidea , Spleen/anatomy & histology , Spleen/chemistry , Spleen/immunology , Strongylida Infections/immunology , Ticks
20.
Int J Radiat Oncol Biol Phys ; 57(4): 1075-83, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14575839

ABSTRACT

PURPOSE: The mechanism of radiosensitization by gemcitabine is still unclear. It has been hypothesized that the accumulation of cells in early S phase may play a role in enhancing radiosensitivity. METHODS AND MATERIALS: The schedule dependency of the radiosensitizing effect was studied in ECV304, human bladder cancer cells, and H292, human lung cancer cells, by varying the incubation time and time interval between gemcitabine and radiation treatment. To determine the role of cell cycle perturbations in the radiosensitization, the influence of gemcitabine on the cell cycle at the moment of radiation was investigated by flow cytometry. RESULTS: The radiosensitizing effect increased with a longer incubation period: Dose enhancement factors varied from 1.30 to 2.82 in ECV304 and from 1.04 to 1.78 in H292 after treatment during 8-32 h, respectively. Radiosensitization decreased with an increasing interval: Dose enhancement factors varied from 2.26 to 1.49 in ECV304 and from 1.45 to 1.11 in H292 after an interval 0-24 h, respectively. Cells were blocked in the early S phase of the cell cycle by gemcitabine. The highest percentage S-phase cells was observed after treatment with the schedules that resulted in the highest radiosensitizing effect. CONCLUSIONS: We observed a clear schedule-dependent radiosensitization by gemcitabine. Our findings demonstrated a correlation between gemcitabine-induced early S-phase block and the radiosensitizing effect.


Subject(s)
Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Cell Survival/drug effects , Cell Survival/radiation effects , Flow Cytometry , Humans , S Phase , Time Factors , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...