Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Publication year range
1.
J Photochem Photobiol B ; 214: 112099, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33285486

ABSTRACT

Long-term illumination of the retina with blue-light-excited phosphor-converted light-emitting diodes (LEDs) may result in decreased retinal function, even if the levels of blue light emitted are low. New low-color-temperature dual-primary-color LEDs have been developed that are composed of only two LED chips: a red chip and a yellow chip. These LEDs are expected to become a new type of healthy lighting source because they do not emit blue light, they lack phosphor, and they solve the problem of low efficiency encountered with phosphor-converted low-color-temperature LEDs. Many studies have indicated that these new low-color-temperature LEDs are likely to have therapeutic effects. However, the biological safety of these LEDs needs to be explored before the therapeutic effects are explored. Therefore, this experiment was conducted to investigate the effects of the new low-color-temperature LEDs and fluorescent white LEDs on three types of retinal cells. We observed that the viability and numbers of retinal cells decreased gradually with increasing LED color temperature. The new low-color-temperature LEDs caused less death and adverse effects on proliferation than the fluorescent white LEDs. After irradiation with high-color-temperature LEDs, the expression of Zonula Occludens-1 (ZO-1) was decreased and discontinuous in ARPE-19 cells; the stress protein hemeoxygenase-1 (HO-1) was upregulated in R28 cells; and glial fibrillary acidic protein (GFAP) and vimentin were upregulated in rMC-1 cells. We therefore conclude that the new white LEDs cause almost no damage to retinal cells and reduce the potential human health risks of chronic exposure to fluorescent white LEDs.


Subject(s)
Retina/radiation effects , Apoptosis/radiation effects , Cell Line , Cell Proliferation/radiation effects , Color , Containment of Biohazards , Gene Expression Regulation/radiation effects , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/radiation effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/radiation effects , Humans , Light , Lighting , Radiation Exposure , Retina/cytology , Temperature , Time Factors , Vimentin/genetics , Vimentin/radiation effects
2.
Oncotarget ; 7(38): 62267-62279, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27556699

ABSTRACT

Alternating electric fields at an intermediate frequency (100~300 kHz), referred to as tumour-treating fields (TTF), are believed to interrupt the process of mitosis via apoptosis and to act as an inhibitor of cell proliferation. Although the existence of an antimitotic effect of TTF is widely known, the proposed apoptotic mechanism of TTF on cell function and the efficacy of TTF are controversial issues among medical experts. To resolve these controversial issues, a better understanding of the underlying molecular mechanisms of TTF on cell function and the differences between the effects of TTF alone and in combination with other treatment techniques is essential. Here, we report experimental evidence of TTF-induced apoptosis and the synergistic antimitotic effect of TTF in combination with ionizing radiation (IR). For these experiments, two human Glioblastoma multiforme (GBM) cells (U373 and U87) were treated either with TTF alone or with TTF followed by ionizing radiation (IR). Cell apoptosis, DNA damage, and mitotic abnormalities were quantified after the application of TTF, and their percentages were markedly increased when TTF was combined with IR. Our experimental results also suggested that TTF combined with IR synergistically suppressed both cell migration and invasion, based on the inhibition of MMP-9 and vimentin.


Subject(s)
Apoptosis/radiation effects , Brain Neoplasms/therapy , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Electric Stimulation Therapy/methods , Glioblastoma/therapy , Mitosis/radiation effects , Radiotherapy/methods , Cell Line, Tumor , Combined Modality Therapy/methods , Humans , Immunohistochemistry , Matrix Metalloproteinase 9/radiation effects , Vimentin/radiation effects
3.
Int J Oral Maxillofac Surg ; 42(8): 939-48, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23583647

ABSTRACT

The aim of this study was to evaluate the suitability of tissue-engineered mucosa (TEM) as a model for studying the acute effects of ionizing radiation (IR) on the oral mucosa. TEM and native non-keratinizing oral mucosa (NNOM) were exposed to a single dose of 16.5Gy and harvested at 1, 6, 24, 48, and 72h post-irradiation. DNA damage induced by IR was determined using p53 binding protein 1 (53BP1), and DNA repair was determined using Rad51. Various components of the epithelial layer, basement membrane, and underlying connective tissue were analyzed using immunohistochemistry. The expression of cytokines interleukin-1ß (IL-1ß) and transforming growth factor beta 1 (TGF-ß1) was analyzed using an enzyme-linked immunosorbent assay. The expression of DNA damage protein 53BP1 and repair protein Rad51 were increased post-irradiation. The expression of keratin 19, vimentin, collage type IV, desmoglein 3, and integrins α6 and ß4 was altered post-irradiation. Proliferation significantly decreased at 24, 48, and 72h post-irradiation in both NNOM and TEM. IR increased the secretion of IL-1ß, whereas TGF-ß1 secretion was not altered. All observed IR-induced alterations in TEM were also observed in NNOM. Based on the similar response of TEM and NNOM to IR we consider our TEM construct a suitable model to quantify the acute biological effects of IR.


Subject(s)
Mouth Mucosa/radiation effects , Tissue Engineering , Basement Membrane/radiation effects , Cell Adhesion/radiation effects , Cell Culture Techniques , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Collagen Type IV/analysis , Collagen Type IV/radiation effects , Connective Tissue/radiation effects , DNA Damage/radiation effects , DNA Repair/radiation effects , Desmoglein 3/analysis , Desmoglein 3/radiation effects , Epithelium/radiation effects , Female , Fibroblasts/radiation effects , Gamma Rays , Humans , Integrin alpha6/analysis , Integrin alpha6/radiation effects , Integrin beta4/analysis , Integrin beta4/radiation effects , Interleukin-1beta/analysis , Interleukin-1beta/radiation effects , Intracellular Signaling Peptides and Proteins/analysis , Intracellular Signaling Peptides and Proteins/radiation effects , Keratin-19/analysis , Keratin-19/radiation effects , Keratinocytes/radiation effects , Male , Middle Aged , Mouth Mucosa/cytology , Rad51 Recombinase/analysis , Rad51 Recombinase/radiation effects , Radiation Dosage , Transforming Growth Factor beta1/analysis , Transforming Growth Factor beta1/radiation effects , Tumor Suppressor p53-Binding Protein 1 , Vimentin/analysis , Vimentin/radiation effects
5.
J Invest Dermatol ; 107(6): 838-43, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8941671

ABSTRACT

Acute low-dose ultraviolet B radiation (UVR) impairs contact hypersensitivity (CH) induction in genetically defined strains of mice by a mechanism triggered by cis-urocanic acid (UCA) and dependent upon tumor necrosis factor-alpha (TNF-alpha). UVR, TNF-alpha, and cis-UCA cause similar morphologic changes among Langerhans cells, which spawns the speculation that UVR impairs CH induction in part by altering the Langerhans cell cytoskeleton. To examine this speculation, we studied the expression of vimentin in Langerhans cells after treatment with UVR, TNF-alpha, and cis-UCA. All treatments caused a reduction in expression of vimentin within the cytoplasm of Langerhans cells. Because partial loss of detectable vimentin may correlate with cytoskeletal disruption, we evaluated the effects of vinblastine, an agent that disrupts the cytoskeleton by disassembling microtubules, on Langerhans cell density and morphology. Epicutaneous treatment with vinblastine caused a reduction in Langerhans cell density, a loss of dendrites, and a reduction in vimentin expression. When dinitrofluorobenzene was painted on vinblastine-treated skin of BALB/c or C3H/HeN mice, only feeble CH was induced. Consequently, we propose that UVR prevents CH induction in susceptible mice by disrupting the cytoskeleton of Langerhans cells, thereby preventing them from carrying out their crucial role as antigen-presenting cells.


Subject(s)
Antigen Presentation/radiation effects , Dermatitis, Contact/immunology , Langerhans Cells/radiation effects , Vimentin/radiation effects , Animals , Antigen Presentation/drug effects , Antigen Presentation/physiology , Antineoplastic Agents, Phytogenic/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/radiation effects , Dinitrofluorobenzene , Langerhans Cells/drug effects , Langerhans Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Tumor Necrosis Factor-alpha/pharmacology , Ultraviolet Rays , Urocanic Acid/pharmacology , Vimentin/drug effects , Vimentin/metabolism , Vinblastine/pharmacology
6.
Mutagenesis ; 11(5): 419-24, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8921502

ABSTRACT

Unrepaired or misrepaired radiation damage in mammalian chromosomes can result in micronucleus formation at the first cell division. This represents loss of genomic information which may cause cell death. To improve our understanding of the mechanism of radiation-induced micronucleus formation, we characterized micronucleus ultrastructure and identified the origin of micronucleus DNA. Immunofluorescence microscopy showed that micronuclei were structurally similar to main nuclei since they contained nuclear lamins A and C and were encapsulated by a network of vimentin intermediate filaments. The contents of radiation-induced micronuclei were characterized using fluorescence in situ hybridization to probe for DNA originating from chromosomes 2, 7, 11 and 16. We postulated that if incorporation of DNA into micronuclei were random, then the probability of chromosomal DNA in micronuclei would be related to the target, i.e. chromosome size. Our results demonstrated that incorporation of DNA from smaller chromosomes (11 and 16) was not different from expected values but incorporation of DNA from the larger chromosomes (2 and 7) was significantly greater than expected. Not all chromosomes in the human genome, therefore, were equally susceptible to genomic loss by micronucleus encapsulation. In conclusion, radiation-induced micronuclei have similar structural characteristics to main nuclei, chromosome damage and/or repair after ionizing radiation may be non-random, and micronucleus formation may reflect this variability.


Subject(s)
Cell Nucleus/radiation effects , Cell Nucleus/ultrastructure , Chromosomes, Human/radiation effects , Cell Nucleus/genetics , DNA/analysis , DNA/radiation effects , Dose-Response Relationship, Radiation , Fibroblasts/radiation effects , Gamma Rays , Humans , In Situ Hybridization, Fluorescence , Lamins , Micronucleus Tests , Nuclear Proteins/radiation effects , Nuclear Proteins/ultrastructure , Vimentin/radiation effects , Vimentin/ultrastructure
7.
Acta Histochem ; 96(4): 365-77, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7536376

ABSTRACT

The immunohistochemical staining of vimentin in paraffin-embedded sections from adult cattle, rabbit, rat and mouse brain fixed in different fixatives (formaldehyde, methacarn, ethanol) was examined using two monoclonal antibodies and a polyclonal antiserum. In non-trypsinized formaldehyde-fixed tissue sections both monoclonal antibodies and the polyclonal antibodies failed to stain vimentin. Following trypsinization of formaldehyde-fixed sections of the four species the meninges, endothelial cells of blood vessels, ependymal cells and the stroma of the choroid plexus were labelled by the monoclonal and polyclonal antibodies used. Astrocytes and Bergmann glial fibers in pretrypsinized formaldehyde-fixed sections from cattle, rabbit and rat brain, however, showed only weak staining. Fixation of cattle and rat brain in methacarn markedly improved the vimentin immunoreactivity of astrocytes and Bergmann glial fibers. The best fixative for the preservation of immunoreactive determinants of vimentin in astrocytes and Bergmann glial fibers in cattle, rabbit and rat brain was ethanol. In brain tissue from mice both monoclonal antibodies labelled only mesoderm-derived tissue components, but did not recognize vimentin in astrocytes and Bergmann glial fibers. Pre-heating formaldehyde-fixed sections from cattle, rabbit and rat brain in a microwave oven prior to the immunohistochemical reaction resulted in an enormous enhancement of vimentin staining of mesoderm-derived tissues, of astrocytes and bergmann cell fibers.


Subject(s)
Acetic Acid , Antibodies, Monoclonal/metabolism , Antibodies/metabolism , Brain/metabolism , Fixatives/chemistry , Vimentin/metabolism , Acetates/chemistry , Acetates/metabolism , Animals , Antibodies/immunology , Antibodies, Monoclonal/immunology , Astrocytes/cytology , Astrocytes/metabolism , Brain/radiation effects , Cattle , Cerebellum/metabolism , Cerebral Cortex/metabolism , Chloroform/chemistry , Chloroform/metabolism , Ethanol/chemistry , Ethanol/metabolism , Female , Fixatives/metabolism , Formaldehyde/chemistry , Formaldehyde/metabolism , Immune Sera/immunology , Immunohistochemistry , Male , Methanol/chemistry , Methanol/metabolism , Mice , Mice, Inbred BALB C , Microwaves , Neuroglia/metabolism , Rabbits , Rats , Rats, Inbred Lew , Staining and Labeling , Tissue Fixation , Trypsin/metabolism , Vimentin/radiation effects
8.
Cell Motil Cytoskeleton ; 26(1): 40-8, 1993.
Article in English | MEDLINE | ID: mdl-8221906

ABSTRACT

Ultraviolet radiation in the near range (UVA) causes lens opacification and disrupts the actin cytoskeleton in rabbit and gray squirrel lenses. Changes were noted using transmission electron microscopy of tangential sections and rhodaminephalloidin fluorescence microscopy of epithelial whole mounts of irradiated and unirradiated lenses, and corresponded with gross cataract formation. Irradiated lenses lacked microfilament polygonal arrays at the inner surface of the apical plasma membrane (i.e., in the cell pole next to the lens fibers) in lens epithelia of both species; a condensed actin bundle was present instead. This bundle, and scattered small actin clumps in the cytoplasm, were identified by immunogold TEM, using a specific antibody and a secondary antibody conjugated with colloidal gold. Similar techniques showed breakdown of tubulin and vimentin, but after longer intervals than for the breakdown of actin. Generalized cytologic damage was also present in epithelial cells, but not in the underlying cortical lens fibers. Damage began to occur after 4 hr of irradiation and became more severe with increased exposure. Shielded controls remained clear, had normal cytology and polygonal arrays, and no clumping of actin filaments.


Subject(s)
Actins/analysis , Actins/radiation effects , Lens, Crystalline/chemistry , Lens, Crystalline/cytology , Ultraviolet Rays , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/radiation effects , Actin Cytoskeleton/ultrastructure , Actins/ultrastructure , Animals , Cytoskeleton/chemistry , Cytoskeleton/radiation effects , Cytoskeleton/ultrastructure , Epithelial Cells , Epithelium/chemistry , Epithelium/radiation effects , Lens, Crystalline/radiation effects , Microscopy, Electron , Sciuridae , Tubulin/analysis , Tubulin/radiation effects , Tubulin/ultrastructure , Vimentin/analysis , Vimentin/radiation effects , Vimentin/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...