Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
JCI Insight ; 6(8)2021 04 22.
Article in English | MEDLINE | ID: mdl-33724956

ABSTRACT

After 9/11, threat of nuclear attack on American urban centers prompted government agencies to develop medical radiation countermeasures to mitigate hematopoietic acute radiation syndrome (H-ARS) and higher-dose gastrointestinal acute radiation syndrome (GI-ARS) lethality. While repurposing leukemia drugs that enhance bone marrow repopulation successfully treats H-ARS in preclinical models, no mitigator potentially deliverable under mass casualty conditions preserves GI tract. Here, we report generation of an anti-ceramide 6B5 single-chain variable fragment (scFv) and show that s.c. 6B5 scFv delivery at 24 hours after a 90% lethal GI-ARS dose of 15 Gy mitigated mouse lethality, despite administration after DNA repair was complete. We defined an alternate target to DNA repair, an evolving pattern of ceramide-mediated endothelial apoptosis after radiation, which when disrupted by 6B5 scFv, initiates a durable program of tissue repair, permitting crypt, organ, and mouse survival. We posit that successful preclinical development will render anti-ceramide 6B5 scFv a candidate for inclusion in the Strategic National Stockpile for distribution after a radiation catastrophe.


Subject(s)
Acute Radiation Syndrome/drug therapy , Ceramides/immunology , Gastrointestinal Diseases/drug therapy , Intestine, Small/drug effects , Intestine, Small/radiation effects , Single-Chain Antibodies/pharmacology , Acute Radiation Syndrome/mortality , Animals , DNA Repair , Gastrointestinal Diseases/mortality , Humans , Injections, Subcutaneous , Intestine, Small/pathology , Jurkat Cells/drug effects , Jurkat Cells/radiation effects , Mice , Single-Chain Antibodies/therapeutic use
2.
Front Immunol ; 9: 922, 2018.
Article in English | MEDLINE | ID: mdl-29760710

ABSTRACT

Impairment or stimulation of the immune system by ionizing radiation (IR) impacts on immune surveillance of tumor cells and non-malignant cells and can either foster therapy response or side effects/toxicities of radiation therapy. For a better understanding of the mechanisms by which IR modulates T-cell activation and alters functional properties of these immune cells, we exposed human immortalized Jurkat cells and peripheral blood lymphocytes (PBL) to X-ray doses between 0.1 and 5 Gy. This resulted in cellular responses, which are typically observed also in naïve T-lymphocytes in response of T-cell receptor immune stimulation or mitogens. These responses include oscillations of cytosolic Ca2+, an upregulation of CD25 surface expression, interleukin-2 and interferon-γ synthesis, elevated expression of Ca2+ sensitive K+ channels and an increase in cell diameter. The latter was sensitive to inhibition by the immunosuppressant cyclosporine A, Ca2+ buffer BAPTA-AM, and the CDK1-inhibitor RO3306, indicating the involvement of Ca2+-dependent immune activation and radiation-induced cell cycle arrest. Furthermore, on a functional level, Jurkat and PBL cell adhesion to endothelial cells was increased upon radiation exposure and was highly dependent on an upregulation of integrin beta-1 expression and clustering. In conclusion, we here report that IR impacts on immune activation and functional properties of T-lymphocytes that may have implications in both toxic effects and treatment response to combined radiation and immune therapy in cancer patients.


Subject(s)
Jurkat Cells/immunology , Jurkat Cells/radiation effects , Lymphocyte Activation/radiation effects , Radiation, Ionizing , Cell Adhesion/radiation effects , Humans , Integrin beta1/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2 Receptor alpha Subunit/genetics , Monocytes/radiation effects
3.
Sci Rep ; 7: 43345, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28266526

ABSTRACT

Extremely-low-frequency magnetic fields (ELF-MF) have been classified as "possibly carcinogenic" to humans on the grounds of an epidemiological association of ELF-MF exposure with an increased risk of childhood leukaemia. Yet, underlying mechanisms have remained obscure. Genome instability seems an unlikely reason as the energy transmitted by ELF-MF is too low to damage DNA and induce cancer-promoting mutations. ELF-MF, however, may perturb the epigenetic code of genomes, which is well-known to be sensitive to environmental conditions and generally deranged in cancers, including leukaemia. We examined the potential of ELF-MF to influence key epigenetic modifications in leukaemic Jurkat cells and in human CD34+ haematopoietic stem cells undergoing in vitro differentiation into the neutrophilic lineage. During granulopoiesis, sensitive genome-wide profiling of multiple replicate experiments did not reveal any statistically significant, ELF-MF-dependent alterations in the patterns of active (H3K4me2) and repressive (H3K27me3) histone marks nor in DNA methylation. However, ELF-MF exposure showed consistent effects on the reproducibility of these histone and DNA modification profiles (replicate variability), which appear to be of a stochastic nature but show preferences for the genomic context. The data indicate that ELF-MF exposure stabilizes active chromatin, particularly during the transition from a repressive to an active state during cell differentiation.


Subject(s)
Epigenesis, Genetic/radiation effects , Hematopoietic Stem Cells/radiation effects , Jurkat Cells/radiation effects , Magnetic Fields , Cell Differentiation/radiation effects , DNA/metabolism , Histones/metabolism , Humans , Methylation
4.
PLoS One ; 11(8): e0160575, 2016.
Article in English | MEDLINE | ID: mdl-27500529

ABSTRACT

The radioprotective capacity of a rationally-designed Mn2+-decapeptide complex (MDP), based on Mn antioxidants in the bacterium Deinococcus radiodurans, was investigated in a mouse model of radiation injury. MDP was previously reported to be extraordinarily radioprotective of proteins in the setting of vaccine development. The peptide-component (DEHGTAVMLK) of MDP applied here was selected from a group of synthetic peptides screened in vitro for their ability to protect cultured human cells and purified enzymes from extreme damage caused by ionizing radiation (IR). We show that the peptides accumulated in Jurkat T-cells and protected them from 100 Gy. MDP preserved the activity of T4 DNA ligase exposed to 60,000 Gy. In vivo, MDP was nontoxic and protected B6D2F1/J (female) mice from acute radiation syndrome. All irradiated mice treated with MDP survived exposure to 9.5 Gy (LD70/30) in comparison to the untreated mice, which displayed 63% lethality after 30 days. Our results show that MDP provides early protection of white blood cells, and attenuates IR-induced damage to bone marrow and hematopoietic stem cells via G-CSF and GM-CSF modulation. Moreover, MDP mediated the immunomodulation of several cytokine concentrations in serum including G-CSF, GM-CSF, IL-3 and IL-10 during early recovery. Our results present the necessary prelude for future efforts towards clinical application of MDP as a promising IR countermeasure. Further investigation of MDP as a pre-exposure prophylactic and post-exposure therapeutic in radiotherapy and radiation emergencies is warranted.


Subject(s)
Deinococcus/chemistry , Radiation-Protective Agents/chemistry , Radiation-Protective Agents/pharmacology , Animals , Antigens, CD34/metabolism , Antioxidants/chemistry , Bone Marrow/drug effects , Bone Marrow/radiation effects , Cytokines/blood , DNA Ligases/metabolism , Drug Design , Female , Humans , Jurkat Cells/drug effects , Jurkat Cells/radiation effects , Leukopenia/drug therapy , Manganese/chemistry , Mice, Inbred Strains , Peptides/chemistry , Radiation Injuries/prevention & control , Radiation, Ionizing , Radiation-Protective Agents/adverse effects , Splenomegaly/drug therapy
5.
Free Radic Res ; 49(10): 1218-32, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26021764

ABSTRACT

Ionizing radiation (IR)-induced oxidative stress in tumor cells is effectively managed by constitutive and inducible antioxidant defense systems. This study was initiated to understand the relative contribution of different redox regulatory systems in determining the tumor radio-resistance. In this study, human T-cell lymphoma (Jurkat) cells were exposed to IR (4 Gy) and monitored for the spatio-temporal changes in cellular redox regulatory parameters. We monitored the changes in the levels of reactive oxygen species (ROS) (total, mitochondrial, primary, and secondary), thiols (total, surface, and intracellular), GSH/GSSG ratio, antioxidant enzyme activity viz. thioredoxin (Trx), Trx reductase (TrxR), glutathione peroxidase, and glutathione reductase with respect to time. We have also measured protein glutathionylation. We observed that tumor cells mount a biphasic response after IR exposure which can be divided into early (0-6 h) and late (16-48 h) responses in terms of changes in cellular redox parameters. During early response, constitutively active GSH and Trx systems respond to restore cellular redox balance to pre-exposure levels and help in activation of redox-sensitive transcription factor Nrf-2. During late response, increase in the levels of antioxidants GSH and Trx rescue cells against IR-mediated damage. We observed that disruption of either glutathione or thioredoxin metabolism led to partial impairment of ability of cells to survive against IR-induced damage. But simultaneous disruption of both the pathways significantly increased radio sensitivity of Jurkat cells. This highlighted the importance of these two antioxidant pathways in regulating redox homeostasis under conditions of IR-induced oxidative stress.


Subject(s)
Gamma Rays/adverse effects , Glutathione/metabolism , Neoplasm Proteins/metabolism , Thioredoxins/metabolism , Active Transport, Cell Nucleus , Apoptosis/radiation effects , DNA Damage , DNA, Neoplasm/radiation effects , Glutathione/antagonists & inhibitors , Homeostasis , Humans , Jurkat Cells/radiation effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/radiation effects , Mitochondria/metabolism , Mitochondria/radiation effects , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/physiology , NF-E2-Related Factor 2/physiology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Oxidation-Reduction , Oxidative Stress , RNA Interference , RNA, Small Interfering/genetics , Radiation Tolerance , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism , Thioredoxins/antagonists & inhibitors , Thioredoxins/genetics , Time Factors , Transfection
6.
Eur Biophys J ; 40(8): 947-57, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21594746

ABSTRACT

The initial effect of nanosecond pulsed electric fields (nsPEFs) on cells is a change of charge distributions along membranes. This first response is observed as a sudden shift in the plasma transmembrane potential that is faster than can be attributed to any physiological event. These immediate, yet transient, effects are only measurable if the diagnostic is faster than the exposure, i.e., on a nanosecond time scale. In this study, we monitored changes in the plasma transmembrane potential of Jurkat cells exposed to nsPEFs of 60 ns and amplitudes from 5 to 90 kV/cm with a temporal resolution of 5 ns by means of the fast voltage-sensitive dye Annine-6. The measurements suggest the contribution of both dipole effects and asymmetric conduction currents across opposite sides of the cell to the charging. With the application of higher field strengths the membrane charges until a threshold voltage value of 1.4-1.6 V is attained at the anodic pole. This indicates when the ion exchange rates exceed charging currents, thus providing strong evidence for pore formation. Prior to reaching this threshold, the time for the charging of the membrane by conductive currents is qualitatively in agreement with accepted models of membrane charging, which predict longer charging times for lower field strengths. The comparison of the data with previous studies suggests that the sub-physiological induced ionic imbalances may trigger other intracellular signaling events leading to dramatic outcomes, such as apoptosis.


Subject(s)
Cell Membrane/radiation effects , Electromagnetic Fields , Jurkat Cells/radiation effects , Membrane Potentials/radiation effects , Pulse Radiolysis/methods , Voltage-Sensitive Dye Imaging/methods , Cell Culture Techniques , Dose-Response Relationship, Radiation , Electric Conductivity , Fluorescent Dyes , Humans , Spectrometry, Fluorescence
7.
Eur Biophys J ; 39(6): 929-34, 2010 May.
Article in English | MEDLINE | ID: mdl-19343334

ABSTRACT

We have made a preliminary analysis of the results about the effects on tumoral cell line (lymphoid T cell line Jurkat) induced by UVB radiation (dose of 310 mJ/cm(2)) with and without a vegetable mixture. In the present study, we have used two techniques: Fourier transform infrared spectroscopy (FTIR) and flow cytometry. FTIR spectroscopy has the potential to provide the identification of the vibrational modes of some of the major compounds (lipid, proteins and nucleic acids) without being invasive in the biomaterials. The second technique has allowed us to perform measurements of cytotoxicity and to assess the percentage of apoptosis. We already studied the induction of apoptotic process in the same cell line by UVB radiation; in particular, we looked for correspondences and correlations between FTIR spectroscopy and flow cytometry data finding three highly probable spectroscopic markers of apoptosis (Pozzi et al. in Radiat Res 168:698-705, 2007). In the present work, the results have shown significant changes in the absorbance and spectral pattern in the wavenumber protein and nucleic acids regions after the treatments.


Subject(s)
Apoptosis/drug effects , Jurkat Cells/radiation effects , Spectroscopy, Fourier Transform Infrared/methods , Ultraviolet Rays , Apoptosis/physiology , Cells, Cultured , Flow Cytometry/methods , Humans , Spectrophotometry, Infrared/methods
8.
Int J Radiat Oncol Biol Phys ; 73(4): 1211-8, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19251092

ABSTRACT

PURPOSE: To define which intracellular pools of sphingomyelin and ceramide are involved in the triggering of apoptosis of Jurkat leukemia cells in response to gamma-ray exposure. METHODS AND MATERIALS: We examined the kinetics of ceramide generation at the whole-cell level and in different subcellular compartments (plasma membrane rafts, mitochondria, and endoplasmic reticulum) after irradiation with photons. Ceramide was measured by high-performance liquid chromatography or after pulse labeling experiments, and the presence of sphingomyelinase within mitochondria was assessed by electron microscopy. RESULTS: Irradiation of Jurkat leukemia cells resulted in the sequential triggering of sphingomyelin hydrolysis, followed by de novo synthesis that led to a late ceramide response (from 24 h) correlated with the triggering of apoptosis. At the subcellular level, pulse-label experiments, using [(3)H]-palmitate as a precursor, strengthened the involvement of the radiation-induced sphingomyelin breakdown and revealed a very early peak (15 min) of ceramide in plasma membrane rafts. A second peak in mitochondria was measured 4 h after irradiation, resulting from an increase of the sphingomyelin content relating to the targeting of acid sphingomyelinase toward this organelle. CONCLUSION: These data confirm that ceramide is a major determinant in the triggering of radiation-induced apoptosis and highlight the complexity of the sequential compartment-specific ceramide-mediated response of Jurkat leukemia cells to gamma-rays.


Subject(s)
Apoptosis/physiology , Ceramides/biosynthesis , Jurkat Cells/radiation effects , Sphingomyelins/biosynthesis , Cell Membrane/metabolism , Cell Membrane/radiation effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/radiation effects , Gamma Rays , Humans , Hydrolysis , Jurkat Cells/metabolism , Mitochondria/metabolism , Mitochondria/radiation effects , Palmitates , Sphingomyelin Phosphodiesterase/metabolism , Time Factors
9.
J Biomed Opt ; 14(6): 064042, 2009.
Article in English | MEDLINE | ID: mdl-20059280

ABSTRACT

We report a photonic approach for selective inactivation of viruses with a near-infrared subpicosecond laser. We demonstrate that this method can selectively inactivate viral particles ranging from nonpathogenic viruses such as the M13 bacteriophage and the tobacco mosaic virus to pathogenic viruses such as the human papillomavirus and the human immunodeficiency virus (HIV). At the same time, sensitive materials such as human Jurkat T cells, human red blood cells, and mouse dendritic cells remain unharmed. The laser technology targets the global mechanical properties of the viral protein shell, making it relatively insensitive to the local genetic mutation in the target viruses. As a result, the approach can inactivate both the wild and mutated strains of viruses. This intriguing advantage is particularly important in the treatment of diseases involving rapidly mutating viral species such as HIV. Our photonic approach could be used for the disinfection of viral pathogens in blood products and for the treatment of blood-borne viral diseases in the clinic.


Subject(s)
Lasers , Optics and Photonics/methods , Spectroscopy, Near-Infrared/methods , Virus Inactivation/radiation effects , Viruses/radiation effects , Alphapapillomavirus/physiology , Alphapapillomavirus/radiation effects , Animals , Bacteriophage M13/physiology , Bacteriophage M13/radiation effects , Cells, Cultured , Dendritic Cells/radiation effects , Erythrocytes/radiation effects , HIV/physiology , HIV/radiation effects , Humans , Jurkat Cells/radiation effects , Mice , Microscopy, Atomic Force , Tobacco Mosaic Virus/physiology , Tobacco Mosaic Virus/radiation effects
10.
Georgian Med News ; (159): 58-61, 2008 Jun.
Article in Russian | MEDLINE | ID: mdl-18633154

ABSTRACT

Mechanisms of radio-inducible death of Jurkat cells were investigated. Human lymphoblastoid T-cell line Jurkat is widely established model for studying apoptosis mechanisms. The cell was radiated by "Teragam" (Czech Republic) by dose 2 g during 1 minute. After radiation cells were incubated at standard conditions during 24 hours. After gamma radiation in cell population amount of cells in gaplois (apoptotic G 0) stage was increased 8,2 folds, in diplois (G 0/G1) stage - by 17%, in synthetic (S) stage decreased by 35% and tetraploid (G2/M) stage by 73% in comparison to control group. It was revealed intensive production of free radicals of oxygen and nitric oxide and decreasing activity of antioxidant enzymes (superoxidismutasa, catalasa and glutathione peroxidase). Revealed dependence between intensification of apoptosis and radiation-induced arrest of cell cycle G2/M phase may be determined by excess amount of free oxygen and nitrogen radicals generated in Jurkat cells as a result of nondirect effects of low doses of gamma radiation.


Subject(s)
Apoptosis/radiation effects , Gamma Rays , Jurkat Cells/pathology , Radiation Injuries/pathology , Dose-Response Relationship, Radiation , Humans , Jurkat Cells/radiation effects
11.
Int J Radiat Oncol Biol Phys ; 70(2): 543-53, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17980509

ABSTRACT

PURPOSE: The ability of heat shock protein 27 (Hsp27) to protect cells from stressful stimuli and its increased levels in tumors resistant to anticancer therapeutics suggest that it may represent a target for sensitization to radiotherapy. In this study, we investigate the protective role of Hsp27 against radiation-induced apoptosis and the effect of its attenuation in highly expressing radioresistant cancer cell lines. METHODS AND MATERIALS: We examined clonogenic death and the kinetics of apoptotic events in different tumor cell lines overexpressing or underexpressing Hsp27 protein irradiated with photons. The radiosensitive Jurkat cell line, which does not express Hsp27 constitutively or in response to gamma-rays, was stably transfected with Hsp27 complementary DNA. Attenuation of Hsp27 expression was accomplished by antisense or RNAi (interfering RNA) strategies in SQ20B head-and-neck squamous carcinoma, PC3 prostate cancer, and U87 glioblastoma radioresistant cells. RESULTS: We measured concentration-dependent protection against the cytotoxic effects of radiation in Jurkat-Hsp27 cells, which led to a 50% decrease in apoptotic cells at 48 hours in the highest expressing cells. Underlying mechanisms leading to radiation resistance involved a significant increase in glutathione levels associated with detoxification of reactive oxygen species, a delay in mitochondrial collapse, and caspase activation. Conversely, attenuation of Hsp27 in SQ20B cells, characterized by their resistance to apoptosis, sensitizes cells to irradiation. This was emphasized by increased apoptosis, decreased glutathione basal level, and clonogenic cell death. Sensitization to irradiation was confirmed in PC3 and U87 radioresistant cells. CONCLUSION: Hsp27 gene therapy offers a potential adjuvant to radiation-based therapy of resistant tumors.


Subject(s)
Apoptosis/radiation effects , Gene Silencing/physiology , Heat-Shock Proteins/physiology , Neoplasm Proteins/physiology , Radiation Tolerance/physiology , Apoptosis/physiology , Caspases/metabolism , Cell Line, Tumor/radiation effects , Down-Regulation , Enzyme Activation/radiation effects , Gamma Rays , Glioblastoma/metabolism , Glioblastoma/radiotherapy , Glutathione/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Jurkat Cells/radiation effects , Male , Mitochondria/physiology , Mitochondria/radiation effects , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oxidative Stress/physiology , Oxidative Stress/radiation effects , Photons , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/radiotherapy , RNA, Antisense/therapeutic use , RNA, Small Interfering/therapeutic use , Radiation Tolerance/genetics , Reactive Oxygen Species/metabolism , Time Factors , Transfection/methods , Tumor Stem Cell Assay
12.
Bioconjug Chem ; 18(2): 494-9, 2007.
Article in English | MEDLINE | ID: mdl-17279724

ABSTRACT

Zn-protoporphyrin (ZnPP) is a promising candidate for cancer therapy. It is known to inhibit heme-oxygenase-1 (HO-1), resulting in suppressed biliverdin/bilirubin production accompanying lowered antioxidative capacity. As a consequence, a significant suppression of tumor growth in vivo was reported. Recent findings also showed that ZnPP efficiently generated reactive singlet oxygen under illumination of visible light. In the present report, we describe the photosensitizing capabilities of water-soluble polymer conjugates of ZnPP as novel compounds for photodynamic therapy against solid tumors. The polymer conjugation made ZnPP water-soluble, thus possible for injection for its aqueous solution. The cellular uptake and photobiological activity of ZnPP derivatives have been tested using a human T-cell leukemia cell line in vitro and demonstrated most potent phototoxic effects of SMA-ZnPP followed by PEG-ZnPP under aerobic conditions.


Subject(s)
Cell Survival/drug effects , Photochemotherapy , Polymers/pharmacology , Protoporphyrins/pharmacology , Water/chemistry , Caspase 3/metabolism , Caspase 7/metabolism , Cell Survival/radiation effects , Drug Delivery Systems , Heme Oxygenase-1/antagonists & inhibitors , Humans , Jurkat Cells/drug effects , Jurkat Cells/radiation effects , Polyethylene Glycols/chemistry , Polymers/administration & dosage , Polymers/chemical synthesis , Protoporphyrins/administration & dosage , Protoporphyrins/chemical synthesis , Solubility
13.
Ann Rheum Dis ; 65(2): 216-21, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16014673

ABSTRACT

BACKGROUND: It has been suggested that defective handling of apoptotic cells by macrophages plays a key role in the development of systemic lupus erythematosus (SLE). The relative contribution of intrinsic defects and serum factors remains controversial. OBJECTIVE: To compare monocytes from SLE patients, patients with rheumatoid arthritis, and healthy controls for their ability to differentiate in vitro into macrophages and to bind/engulf apoptotic cells. METHODS: Peripheral blood derived monocytes from healthy donors or from patients with SLE or rheumatoid arthritis were allowed to differentiate into macrophages. The in vitro uptake of apoptotic cells by macrophages was evaluated by a flow cytometry assay that allowed discrimination between binding and internalisation. RESULTS: Monocytes from SLE and rheumatoid patients showed a striking defect in adherence to plastic compared with healthy donors. Absence or heat inactivation of serum resulted in a reduction in the binding and engulfment of apoptotic cells by macrophages. Macrophages from rheumatoid and SLE patients had similar percentages of apoptotic cells bound to their surface compared with normal controls. However, macrophages from SLE patients showed a significant defect in the internalisation of apoptotic cells compared with those from healthy controls, even in the presence of normal human serum. CONCLUSIONS: Monocytes from patients with SLE and rheumatoid arthritis have a similar defect in their capacity to adhere to plastic. However, only macrophages from SLE patients showed an impaired ability to engulf apoptotic cells, which indicates that an intrinsic cellular defect may be responsible for this phenomenon.


Subject(s)
Arthritis, Rheumatoid/pathology , Lupus Erythematosus, Systemic/pathology , Macrophages/physiology , Adolescent , Adult , Aged , Analysis of Variance , Apoptosis , Case-Control Studies , Cell Adhesion , Cell Separation , Cells, Cultured , Female , Flow Cytometry , Humans , Jurkat Cells/pathology , Jurkat Cells/radiation effects , Male , Middle Aged , Phagocytosis , Ultraviolet Rays/adverse effects
14.
Photodermatol Photoimmunol Photomed ; 21(1): 23-31, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15634220

ABSTRACT

BACKGROUND: Ultraviolet-A1 (340-400 nm) (UVA1) radiation causes singlet-oxygen damage that depolarizes mitochondrial membranes triggering immediate apoptosis (T < or = 4 h), while it also causes oxidative damage to DNA inducing delayed apoptosis (T > or = 24 h). In this study, we examined some potential therapeutic endpoints associated with UVA1-mediated immediate and delayed apoptosis, such as receptor and cytokine changes. METHODS: We quantified the number of membrane-bound CD3 receptors on transformed T lymphocytes (Jurkat) and the number of membrane-bound CD19 receptors on transformed B lymphocytes (Daudi) using flow cytometry. We also quantified the release of the cytokines interferon gamma (IFN-gamma) and interleukin-2 (IL-2) using enzyme-linked immunosorbent assays. RESULTS: Out of the entire population of cells, only the apoptotic Daudi cells immediately decreased CD19 expression via capping, while only the apoptotic Jurkat cells increased CD3 receptor expression 24 h post-exposure. Both receptor changes occurred in a UVA1 dose-dependent manner. We also examined other T-cell receptors, such as CD4, CD25, and CD69, but they did not change for up to 24 h following exposure. During UVA1-triggered immediate apoptosis of Jurkat T cells, IFN-gamma levels increased in a dose-dependent manner at 4 h, but returned to baseline levels at 24 h post-exposure, whereas, there was no significant change in IL-2 at 4 or 24 h. CONCLUSION: Thus, UVA1-triggered immediate apoptosis causes a rapid decrease in the number of CD19 receptors on Daudi B cells and release of IFN-gamma from Jurkat T cells at 4 h, and UVA1-mediated delayed apoptosis causes an increase in the number of CD3 receptors on Jurkat T cells.


Subject(s)
Antigens, CD19/metabolism , Apoptosis/radiation effects , B-Lymphocytes/radiation effects , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , T-Lymphocytes/radiation effects , Ultraviolet Rays , Cell Line, Transformed , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Jurkat Cells/radiation effects , Microscopy, Electron
15.
Toxicology ; 207(3): 411-8, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15664269

ABSTRACT

DCB, 3,3'-dichlorobenzidine, is used primarily as an intermediate in the manufacture of diarylide yellow or azo red pigments for printing ink, textile, paint, and plastics. It is also used in tattoo inks. In this article, we investigate light-induced toxicity of DCB in both bacteria and human Jurkat T-cells. DCB itself is not toxic or mutagenic to Salmonella typhimurium TA102, but is photomutagenic at concentrations as low as 2 microM and phototoxic at concentrations >100 microM when bacteria are exposed to DCB and light at the same time (1.2 J/cm2 of UVA and 2.1 J/cm2 of visible light). Furthermore, DCB is both photocytotoxic and photogenotoxic to human Jurkat T-cells. Under a light irradiation dose of 2.3 J/cm2 of UVA and 4.2 J/cm2 of visible light, it causes the Jurkat T-cells to become nonviable in a DCB dose-dependent manner and the nonviable cells reaches 60% at DCB concentrations higher than 50 microM. At the same time, DNA fragmentation is observed for cells exposed to both DCB and light, determined by single cell gel electrophoresis (alkaline comet assay). As much as 5% (average) DNA fragmentation was observed when exposed to 200 microM DCB and light irradiation. This suggests that DCB can penetrate the cell membrane and enter the cell. Upon light activation, DCB in the cells can cause various cellular damages, leading to nonviable Jurkat T-cells. It appears, the nonviable cells are not caused solely by fragmentation of cellular DNA, but by other damages such as to proteins and cell membranes, or DNA alkylation. Therefore, persons exposed to DCB through environmental contamination or through tattoo piercing using DCB-containing inks must not only concern about its toxicity without exposing to light, but also its phototoxicity.


Subject(s)
3,3'-Dichlorobenzidine/toxicity , DNA Damage , Jurkat Cells , Mutagens/toxicity , Salmonella typhimurium , Ultraviolet Rays/adverse effects , 3,3'-Dichlorobenzidine/chemistry , Cell Survival/drug effects , Cell Survival/radiation effects , Coloring Agents/chemistry , Comet Assay , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Humans , Ink , Jurkat Cells/drug effects , Jurkat Cells/pathology , Jurkat Cells/radiation effects , Light , Mutagens/chemistry , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Salmonella typhimurium/radiation effects , Tattooing
16.
Toxicology ; 202(3): 237-47, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15337586

ABSTRACT

Nonylphenol polyethoxylates (NPEOs) are widely used as non-ionic surfactants and their biodegradation products such as 4-n-nonylphenol are stable and have been demonstrated to be cytotoxic. In the aquatic environment, these compounds are usually exposed to sunlight, and while the correlation between the biodegradation of NPEOs and changes in cytotoxicity has been reported, the relationship between the photodegradation of NPEOs and cytotoxicity has not. In this study, we investigated the degradation of NPEO by ultraviolet (UV) irradiation, especially UVB irradiation, and the effects on mammalian cell lines. NPEO with a smaller number of ethylene oxide (EO) units showed greater cytotoxicity. Although NPEO (10) completely inhibited the proliferation of the cells, NPEO (70) showed no toxicity. UVB irradiation significantly induced a shortening of the side chain, which was due to the production of ROS. The EO side chain of NPEO (10), was gradually degradated, but that of NPEO (70) was degradated near the benzene ring. Furthermore, the degradation of the benzene ring was more effective in NPEO (70) than NPEO (10). The toxicity of NPEO (10) in cultured cells decreased following UVB irradiation, whereas that of NPEO (70) was induced after UVB irradiation at 500 J/cm2 and disappeared at 1000 J/cm2. This might be due to the production of NPEO with a short side chain and 4-n-nonylphenol by the degradation of EO and due to the degradation of the benzene ring at higher doses of UVB irradiation. This study shows the significance of UV exposure to the degradation of alkylphenol polyethoxylates in the environment.


Subject(s)
Ethylene Glycols/radiation effects , Ethylene Glycols/toxicity , Animals , Cell Division/drug effects , Dose-Response Relationship, Radiation , Humans , Jurkat Cells/drug effects , Jurkat Cells/radiation effects , Mice , NIH 3T3 Cells/drug effects , NIH 3T3 Cells/radiation effects , Photolysis , Reactive Oxygen Species/metabolism , Ultraviolet Rays
17.
Int J Radiat Oncol Biol Phys ; 58(2): 386-96, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14751507

ABSTRACT

PURPOSE: Tumor hypoxia reduces the efficacy of radiotherapy, many types of chemotherapy, and tumor necrosis factor-alpha (TNF-alpha). TRAIL (TNF-alpha-related apoptosis-inducing ligand) is a ligand for death receptors of the TNF superfamily shown to be selectively toxic for tumor cells and thereby a promising antineoplastic tool. The impact of hypoxia on TRAIL-induced apoptosis was examined in this study. METHODS AND MATERIALS: Apoptosis induction and growth rates of various tumor cell lines under hypoxia were evaluated in vitro. Biologically effective induction of hypoxia was verified by determination of hypoxia-inducible factor-1 (HIF-1) activation. The efficacy of TRAIL- and radiation-induced apoptosis under different oxygen conditions was quantified in vitro. The impact of Bcl-2 on TRAIL-induced apoptosis under hypoxia or normoxia was evaluated by comparing cells expressing Bcl-2 with a vector control. RESULTS: Moderate hypoxia caused no growth retardation or apoptosis, but led to activation of HIF-1 as a prerequisite of hypoxic gene induction. Cellular responses to TRAIL differed considerably among the cell lines tested. Hypoxia reduced radiation-induced, but not TRAIL-induced, apoptosis in the tested cell lines. Hypoxia did not induce Bcl-2 expression. Bcl-2 had a minor impact on the efficacy of TRAIL-induced apoptosis. CONCLUSION: Taken together, the data indicate that TRAIL is clearly effective under conditions of proven hypoxia.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Cell Hypoxia/physiology , DNA-Binding Proteins/metabolism , Membrane Glycoproteins/pharmacology , Nuclear Proteins/metabolism , Transcription Factors , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , Caspase 8 , Caspases/metabolism , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Cell Nucleus/metabolism , DNA/metabolism , Drug Screening Assays, Antitumor , Enzyme Activation , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Jurkat Cells/drug effects , Jurkat Cells/radiation effects , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , TNF-Related Apoptosis-Inducing Ligand
18.
Int J Cancer ; 107(4): 520-7, 2003 Nov 20.
Article in English | MEDLINE | ID: mdl-14520687

ABSTRACT

The human leukemic T-cell line Jurkat was used to define the role of the cellular stress pathway with its key player kinase JNK in cancer therapy-induced apoptosis. JNK activity was inhibited by stable transfection with a dominant negative mutant of the upstream kinase JNKK/MKK4 or with the novel, potent and selective JNK1, -2 and -3 inhibitor SP600125. Inhibition of JNK activity delayed the onset of apoptosis induced by cisplatin, doxorubicin, gamma-irradiation and CD95-L but did not prevent apoptosis per se. Early events during apoptosis such as induction of CD95-L, activation of caspase-8 and exposure of phosphatidylserine on the cell surface were strongly inhibited. Also, at early time points of apoptosis, loss of the mitochondrial membrane potential and release of cytochrome c were markedly impaired. However, late signaling events during apoptosis such as cleavage of PARP and DNA fragmentation apoptosis were only marginally affected. These findings are in accordance with the activity of initiator and effector caspases. Whereas activity of the initiator caspase-8 was strongly inhibited early and late after induction, an inhibition of caspase-3 activity was only observed early after induction of apoptosis. We therefore suggest that cellular stress signaling contributes to the initiation of apoptosis, whereas it might be dispensable for the progression of apoptosis. Dysfunction of this pathway under pathological conditions might contribute to therapy resistance of cancer cells.


Subject(s)
Apoptosis , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Oxidative Stress , Signal Transduction/drug effects , Anthracenes/pharmacology , Antineoplastic Agents/pharmacology , Caspases/metabolism , Cisplatin/pharmacology , Cytochromes c , Doxorubicin/pharmacology , Enzyme Activation , Fas Ligand Protein , Genes, Dominant , Humans , Jurkat Cells/drug effects , Jurkat Cells/radiation effects , Membrane Glycoproteins/metabolism , Membrane Potentials/drug effects , Membrane Potentials/radiation effects , Mitochondria/drug effects , Mitochondria/radiation effects , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/pharmacology , Phosphatidylserines/metabolism , Poly(ADP-ribose) Polymerases , fas Receptor/pharmacology
19.
J Cell Biochem ; 89(5): 956-63, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12874830

ABSTRACT

Exposure of cells to ionizing radiation (IR) determines cellular lesions, such as DNA and membrane damage, which involve a coordinate network of signal transduction pathways responsible for resistance to or delay of apoptosis, depending on cell type and administered dose. Since, after IR exposure, the apoptotic profile appeared different in the two chosen cell lines K562 and Jurkat along with caspase-3 activation, we paid attention to the influence exerted by Protein kinase C delta on transcription factor NF-kappaB activation. Interestingly, K562 resist to IR carrying out a survival strategy which includes PKC delta/NF-kappaB pathway activation, probably mediated by novel IKKs and a role for PI-3-kinase in activating PKC delta at Thr 505 by PDK-1 phosphorylation is suggested. In addition, since caspase-3 is not activated in these cells upon ionizing radiation exposure, it could be supposed that NF-kappaB antagonizes apoptosis induction interfering with pathways which lead to caspase activation, may be by inducing expression of IAP, caspases 3, 7, 9, inhibitor. Thus NF-kappaB activation explains the resistance displayed by K562 to IR and drug potential interference directed to this protein could overcome apoptosis resistance in clinical settings.


Subject(s)
Apoptosis/physiology , Apoptosis/radiation effects , Leukemia/pathology , NF-kappa B/physiology , Caspase 3 , Caspases/metabolism , Caspases/radiation effects , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Cell Survival/physiology , Cell Survival/radiation effects , Cytoplasm/metabolism , Cytoplasm/radiation effects , Enzyme Activation/radiation effects , Humans , I-kappa B Proteins/metabolism , I-kappa B Proteins/radiation effects , In Situ Nick-End Labeling , Jurkat Cells/cytology , Jurkat Cells/radiation effects , K562 Cells , Leukemia/metabolism , Microscopy, Fluorescence , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/radiation effects , Phosphorylation , Protein Kinase C/metabolism , Protein Kinase C/radiation effects , Radiation, Ionizing
20.
Biomed Sci Instrum ; 39: 554-60, 2003.
Article in English | MEDLINE | ID: mdl-12724951

ABSTRACT

It was recently shown that antibodies catalyze a reaction between water and ultraviolet light (UV) creating singlet oxygen and ultimately H2O2. Although the in vivo relevance of these antibody reactions is unclear, it is interesting that among a wide variety of non-antibody proteins tested, the T cell receptor is the only protein with similar capabilities. In clinical settings UV is believed to exert therapeutic effects by eliminating inflammatory epidermal T cells and we hypothesized that UV-triggered H2O2 production is involved in this process. To test the hypothesis we developed tools to study production of H2O2 by T cell receptors with the long-term goal of understanding, and improving, UV phototherapy. Here, we report the development of an inexpensive, real time H2O2 monitoring system having broad applicability. The detector is a Clark oxygen electrode (Pt, Ag/AgCl) modified to detect UV-driven H2O2 production. Modifications include painting the electrode black to minimize UV effects on the Ag/AgCl electrode and the use of hydrophilic, large pore Gelnots electrode membranes. Electrode current was converted to voltage and then amplified and recorded using a digital multimeter coupled to a PC. A reaction vessel with a quartz window was developed to maintain constant temperature while permitting UV irradiation of the samples. The sensitivity and specificity of the system and its use in cell-free and cell-based assays will be presented. In a cellfree system, production of H2O2 by CD3 antibodies was confirmed using our real time H2O2 monitoring method. Additionally we report the finding that splenocytes and Jurkat T cells also produce H2O2 when exposed to UV light.


Subject(s)
Antibodies, Catalytic/metabolism , Hydrogen Peroxide/analysis , Hydrogen Peroxide/metabolism , Polarography/instrumentation , Water/metabolism , Animals , Antibodies/analysis , Antibodies/metabolism , Antibodies, Catalytic/chemistry , Antibodies, Catalytic/radiation effects , CD3 Complex/metabolism , CD3 Complex/radiation effects , Calibration , Cell-Free System/metabolism , Electrodes , Equipment Design , Humans , Jurkat Cells/metabolism , Jurkat Cells/radiation effects , Male , Membranes, Artificial , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/radiation effects , Sensitivity and Specificity , Spleen/chemistry , Spleen/metabolism , Spleen/radiation effects , Temperature , Ultraviolet Rays , Water/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...