Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Lasers Med Sci ; 35(5): 1047-1054, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31754908

ABSTRACT

Snakebites caused by the genus Bothrops are often associated with severe and complex local manifestations such as edema, pain, hemorrhage, and myonecrosis. Conventional treatment minimizes the systemic effects of venom; however, their local action is not neutralized. The purpose of this study was to evaluate the effect of photobiomodulation (PBM) on C2C12 muscle cells exposed to B. jararaca, B. jararacussu, and B. moojeni venoms on events involved in cell death and the release of inflammatory mediators. Cells were exposed to venoms and immediately irradiated with low-level laser (LLL) application in continuous wave at the wavelength of 660 nm, energy density of 4.4 J/cm2, power of 10 mW, area of 0.045 cm2, and time of 20 s. Cell integrity was analyzed by phase contrast microscope and cell death was performed by flow cytometry. In addition, interleukin IL1-ß, IL-6, and IL-10 levels were measured in the supernatant. Our results showed that the application of PBM increases cell viability and decreases cell death by apoptosis and necrosis. Moreover, the release of pro-inflammatory interleukins was also reduced. The data reported here indicate that PBM resulted in cytoprotection on myoblast C2C12 cells after venom exposure. This protection involves the modulation of cell death mechanism and decreased pro-inflammatory cytokine release.


Subject(s)
Apoptosis/drug effects , Bothrops/metabolism , Crotalid Venoms/toxicity , Cytokines/biosynthesis , Low-Level Light Therapy , Muscle Cells/pathology , Animals , Cell Line , Cell Shape/drug effects , Mice , Muscle Cells/drug effects , Muscle Cells/radiation effects
2.
J Neurosci ; 33(9): 3834-43, 2013 Feb 27.
Article in English | MEDLINE | ID: mdl-23447595

ABSTRACT

Nonvisual photosensation enables animals to sense light without sight. However, the cellular and molecular mechanisms of nonvisual photobehaviors are poorly understood, especially in vertebrate animals. Here, we describe the photomotor response (PMR), a robust and reproducible series of motor behaviors in zebrafish that is elicited by visual wavelengths of light but does not require the eyes, pineal gland, or other canonical deep-brain photoreceptive organs. Unlike the relatively slow effects of canonical nonvisual pathways, motor circuits are strongly and quickly (seconds) recruited during the PMR behavior. We find that the hindbrain is both necessary and sufficient to drive these behaviors. Using in vivo calcium imaging, we identify a discrete set of neurons within the hindbrain whose responses to light mirror the PMR behavior. Pharmacological inhibition of the visual cycle blocks PMR behaviors, suggesting that opsin-based photoreceptors control this behavior. These data represent the first known light-sensing circuit in the vertebrate hindbrain.


Subject(s)
Movement/physiology , Opsins/metabolism , Photoreceptor Cells, Vertebrate/physiology , Rhombencephalon/cytology , Stereotyped Behavior/physiology , Age Factors , Analysis of Variance , Animals , Biomechanical Phenomena , Biophysics , Calcium/metabolism , Embryo, Nonmammalian , Female , Male , Microscopy, Confocal , Morpholinos/pharmacology , Movement/drug effects , Movement/radiation effects , Muscle Cells/drug effects , Muscle Cells/radiation effects , Neural Pathways/drug effects , Neural Pathways/physiology , Neural Pathways/radiation effects , Opsins/chemistry , Photic Stimulation , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/radiation effects , Rhombencephalon/physiology , Stereotyped Behavior/drug effects , Stereotyped Behavior/radiation effects , Time Factors , Zebrafish
3.
Int J Biol Macromol ; 107(Pt B): 2020-2025, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29037876

ABSTRACT

This report describes the effect of photobiomodulation (PBM) on edema formation, leukocyte influx, prostaglandin E2 (PGE2) biosynthesis and cytotoxicity caused by bothropstoxin-I (BthTX-I), a phospholipase A2 (PLA2) homologue isolated from Bothrops jararacussu snake venom. Swiss mice or C2C12 cells were irradiated with low-level laser (LLL) at 685nm wavelength, an energy density of 4.6J/cm2 and an irradiation time of 13s. To evaluate the effect on edema formation and leukocyte influx, LLL was applied to the site of inoculation 30min and 3h post-injection. C2C12 cells were exposed to BthTX-I and immediately irradiated. PBM significantly reduced paw edema formation, peritoneal leukocyte influx and PGE2 synthesis, but increased the viability of C2C12 muscle cells after BthTX-I incubation. These findings demonstrate that PBM attenuated the inflammatory events induced by BthTX-I. The attenuation of PGE2 synthesis could be an important factor in the reduced inflammatory response caused by laser irradiation. The ability of LLL irradiation to protect muscle cells against the deleterious effects of BthTX-I may indicate preservation of the plasma membrane.


Subject(s)
Bothrops/metabolism , Low-Level Light Therapy , Phospholipases A2/pharmacology , Snake Venoms/pharmacology , Animals , Cell Line , Cell Survival/radiation effects , Dinoprostone/metabolism , Edema/pathology , Leukocytes/pathology , Male , Mice , Muscle Cells/radiation effects , Peritoneum/pathology
4.
Mol Cell Biol ; 24(14): 6350-61, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15226436

ABSTRACT

DNA damage induces cell cycle arrest and DNA repair or apoptosis in proliferating cells. Terminally differentiated cells are permanently withdrawn from the cell cycle and partly resistant to apoptosis. To investigate the effects of genotoxic agents in postmitotic cells, we compared DNA damage-activated responses in mouse and human proliferating myoblasts and their differentiated counterparts, the myotubes. DNA double-strand breaks caused by ionizing radiation (IR) induced rapid activating autophosphorylation of ataxia-teleangiectasia-mutated (ATM), phosphorylation of histone H2AX, recruitment of repair-associated proteins MRE11 and Nbs1, and activation of Chk2 in both myoblasts and myotubes. However, IR-activated, ATM-mediated phosphorylation of p53 at serine 15 (human) or 18 (mouse) [Ser15(h)/18(m)], and apoptosis occurred in myoblasts but was impaired in myotubes. This phosphorylation could be enforced in myotubes by the anthracycline derivative doxorubicin, leading to selective activation of proapoptotic genes. Unexpectedly, the abundance of autophosphorylated ATM was indistinguishable after exposure of myotubes to IR (10 Gy) or doxorubicin (1 microM/24 h) despite efficient phosphorylation of p53 Ser15(h)/18(m), and apoptosis occurred only in response to doxorubicin. These results suggest that radioresistance in myotubes might reflect a differentiation-associated, pathway-selective blockade of DNA damage signaling downstream of ATM. This mechanism appears to preserve IR-induced activation of the ATM-H2AX-MRE11/Rad50/Nbs1 lesion processing and repair pathway yet restrain ATM-p53-mediated apoptosis, thereby contributing to life-long maintenance of differentiated muscle tissues.


Subject(s)
Cell Differentiation/physiology , Muscle Cells/physiology , Muscle Cells/radiation effects , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/physiology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line , Checkpoint Kinase 2 , DNA Damage , DNA Repair , DNA Repair Enzymes , DNA-Binding Proteins/metabolism , Doxorubicin/pharmacology , Enzyme Activation , Enzyme Inhibitors/metabolism , Histones/metabolism , Humans , In Situ Nick-End Labeling , MRE11 Homologue Protein , Mice , Muscle Cells/cytology , Muscle Cells/drug effects , Nuclear Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Radiation, Ionizing , Serine/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins
5.
Phys Med Biol ; 62(4): 1417-1436, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28114113

ABSTRACT

This work investigates how doses to cellular targets depend on cell morphology, as well as relations between cellular doses and doses to bulk tissues and water. Multicellular models of five healthy and cancerous soft tissues are developed based on typical values of cell compartment sizes, elemental compositions and number densities found in the literature. Cells are modelled as two concentric spheres with nucleus and cytoplasm compartments. Monte Carlo simulations are used to calculate the absorbed dose to the nucleus and cytoplasm for incident photon energies of 20-370 keV, relevant for brachytherapy, diagnostic radiology, and out-of-field radiation in higher-energy external beam radiotherapy. Simulations involving cell clusters, single cells and single nuclear cavities are carried out for cell radii between 5 and [Formula: see text]m, and nuclear radii between 2 and [Formula: see text]m. Seven nucleus and cytoplasm elemental compositions representative of animal cells are considered. The presence of a cytoplasm, extracellular matrix and surrounding cells can affect the nuclear dose by up to [Formula: see text]. Differences in cell and nucleus size can affect dose to the nucleus (cytoplasm) of the central cell in a cluster of 13 cells by up to [Formula: see text] ([Formula: see text]). Furthermore, the results of this study demonstrate that neither water nor bulk tissue are reliable substitutes for subcellular targets for incident photon energies <50 keV: nuclear (cytoplasm) doses differ from dose-to-medium by up to [Formula: see text] ([Formula: see text]), and from dose-to-water by up to [Formula: see text] ([Formula: see text]). The largest differences between dose descriptors are seen for the lowest incident photon energies; differences are less than [Formula: see text] for energies [Formula: see text]90 keV. The sensitivity of results with regard to the parameters of the microscopic tissue structure model and cell model geometry, and the importance of the nucleus and cytoplasm as targets for radiation-induced cell death emphasize the importance of accurate models for cellular dosimetry studies.


Subject(s)
Brachytherapy/methods , Cell Nucleus/radiation effects , Cytoplasm/radiation effects , Radiation Dosage , Radiotherapy Planning, Computer-Assisted/methods , Connective Tissue Cells/radiation effects , Humans , Models, Theoretical , Monte Carlo Method , Muscle Cells/radiation effects , Photons , Radiation Dosimeters
6.
Photomed Laser Surg ; 34(11): 572-579, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27627137

ABSTRACT

OBJECTIVE: The purpose of this article was to review the molecular mechanisms of low-level laser irradiation (LLLI) preconditioning for heart cell therapy. BACKGROUND DATA: Stem cell transplantation appears to offer a better alternative to cardiac regenerative therapy. Previous studies have confirmed that the application of LLLI plays a positive role in regulating stem cell proliferation and in remodeling the hostile milieu of infarcted myocardium. Greater understanding of LLLI's underlying mechanisms would be helpful in translating cell transplantation therapy into the clinic. METHODS: Studies investigating LLLI preconditioning for cardiac regenerative therapy published up to 2015 were retrieved from library sources and Pubmed databases. RESULTS: LLLI preconditioning stimulates proliferation and differentiation of stem cells through activation of cell proliferation signaling pathways and alteration of microRNA expression. It also could stimulate paracrine secretion of stem cells and alter cardiac cytokine expression in infarcted myocardium. CONCLUSIONS: LLLI preconditioning provides a promising approach to maximize the efficacy of cardiac cell-based therapy. Although many studies have reported possible molecular mechanisms involved in LLLI preconditioning, the exact mechanisms are still not clearly understood.


Subject(s)
Ischemic Preconditioning, Myocardial/methods , Low-Level Light Therapy/methods , Muscle Cells/radiation effects , Myocardial Infarction/radiotherapy , Cell Differentiation/physiology , Cell Differentiation/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Female , Humans , Male , Mesenchymal Stem Cell Transplantation/methods , Muscle Cells/physiology , Regeneration/radiation effects , Ventricular Remodeling/radiation effects
7.
Photodiagnosis Photodyn Ther ; 13: 196-200, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26116282

ABSTRACT

BACKGROUND: We have proposed an application of photodynamic reaction for less-heated myocardial ablation which employs talaporfin sodium. Intracellular photodynamic reactions with ongoing uptake have the ability to induce apoptosis over time, raising the possibility of extending the lesion depth. The objective of this study was to understand how, in myocardial cells, the late cell survival levels change by incubation time with talaporfin sodium, and what dependence talaporfin sodium uptake has on the duration of incubation with talaporfin sodium in vitro. METHODS: Rat myocardial cells were incubated with talaporfin sodium for 5-360 min and intracellular concentrations measured using a fluorescence micro-plate reader after wash. Cell survival was measured using a water-soluble tetrazolium assay at 2 and 24 h after a photodynamic reaction using a red diode laser of 660 nm, following 15-180 min of incubation with talaporfin sodium. Cells were stained with Hoechst 33342 to observe nuclear changes. RESULTS: Intracellular talaporfin sodium concentration increased with incubation time, with a marked increase between 0 and 60 min. Cell survival at 24 h decreased by 20% when the duration of incubation with talaporfin sodium was extended from 15 to 30 min. Following incubation time of 30-180 min with talaporfin sodium, cell survival was decreased by approximately 30% between measurements at 2 and 24 h. The intracellular talaporfin sodium concentration that induced higher levels of late cell death with cell nuclei fragmentation in these cells was approximately 0.2 µg/mL. CONCLUSION: We obtained the characteristics of late cell death occurrence and talaporfin sodium uptake to myocardial cell with various incubation times with talaporfin sodium.


Subject(s)
Apoptosis/drug effects , Cell Survival/drug effects , Muscle Cells/drug effects , Muscle Cells/radiation effects , Photochemotherapy/methods , Porphyrins/administration & dosage , Animals , Apoptosis/radiation effects , Cell Survival/radiation effects , Cells, Cultured , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Light , Photosensitizing Agents/administration & dosage , Radiation Dosage , Rats
8.
J Exp Med ; 213(12): 2575-2589, 2016 11 14.
Article in English | MEDLINE | ID: mdl-27810924

ABSTRACT

Cell transplantation into immunodeficient mice has revolutionized our understanding of regeneration, stem cell self-renewal, and cancer; yet models for direct imaging of engrafted cells has been limited. Here, we characterize zebrafish with mutations in recombination activating gene 2 (rag2), DNA-dependent protein kinase (prkdc), and janus kinase 3 (jak3). Histology, RNA sequencing, and single-cell transcriptional profiling of blood showed that rag2 hypomorphic mutant zebrafish lack T cells, whereas prkdc deficiency results in loss of mature T and B cells and jak3 in T and putative Natural Killer cells. Although all mutant lines engraft fluorescently labeled normal and malignant cells, only the prkdc mutant fish reproduced as homozygotes and also survived injury after cell transplantation. Engraftment into optically clear casper, prkdc-mutant zebrafish facilitated dynamic live cell imaging of muscle regeneration, repopulation of muscle stem cells within their endogenous niche, and muscle fiber fusion at single-cell resolution. Serial imaging approaches also uncovered stochasticity in fluorescently labeled leukemia regrowth after competitive cell transplantation into prkdc mutant fish, providing refined models to assess clonal dominance and progression in the zebrafish. Our experiments provide an optimized and facile transplantation model, the casper, prkdc mutant zebrafish, for efficient engraftment and direct visualization of fluorescently labeled normal and malignant cells at single-cell resolution.


Subject(s)
DNA-Activated Protein Kinase/deficiency , Imaging, Three-Dimensional/methods , Neoplasm Transplantation , Optical Phenomena , Single-Cell Analysis/methods , Zebrafish/metabolism , Anemia/pathology , Animals , Base Sequence , Clone Cells , DNA-Activated Protein Kinase/metabolism , Disease Models, Animal , Gamma Rays , Homozygote , Humans , Immunocompromised Host/radiation effects , Luminescent Proteins/metabolism , Muscle Cells/pathology , Muscle Cells/radiation effects , Mutation/genetics , Proto-Oncogene Proteins c-myc/metabolism , Regeneration/radiation effects , Transplantation, Homologous , V(D)J Recombination/genetics , Zebrafish Proteins/metabolism , Red Fluorescent Protein
9.
Circulation ; 109(8): 1036-40, 2004 Mar 02.
Article in English | MEDLINE | ID: mdl-14967723

ABSTRACT

BACKGROUND: Hypoxic human coronary smooth muscle cells (HCSMCs) are possible targets for brachytherapy to prevent restenosis after percutaneous transluminal coronary angiography. It is unclear whether growth kinetics and gene expression of these cells undergoing gamma-irradiation are changed by reoxygenation. METHODS AND RESULTS: Hypoxic (H) and hypoxia-reoxygenated (H-R) HCSMCs were irradiated with gamma-radiation at single doses of 4, 8, and 16 Gy using a 60Co-source. Vascular endothelial growth factor gene expression of HCSMCs was dramatically suppressed in H-R versus H cells independent of the radiation dose (15+/-7% versus 2183+/-2023%, P<0.01, H-R versus H cells). An oxygen enhancement ratio of 1.8 was calculated after irradiation from the retarded growth of H-R versus hypoxic HCSMCs. Production of reactive oxygen species by HCSMCs after irradiation increased by 15+/-2% in H-R cells versus 7+/-1% in H cells (P<0.05). CONCLUSIONS: Reoxygenation of hypoxic HCSMCs markedly amplifies growth-retarding effects of ionizing irradiation. On the basis of these findings, oxygenating radiosensitizers should be analyzed with regard to suitability for coronary brachytherapy to prevent restenosis.


Subject(s)
Coronary Vessels/radiation effects , Gamma Rays , Muscle Cells/radiation effects , Muscle, Smooth, Vascular/radiation effects , Oxygen/metabolism , Brachytherapy , Cell Division/radiation effects , Cell Hypoxia , Cells, Cultured/metabolism , Cells, Cultured/radiation effects , Coronary Vessels/metabolism , Dose-Response Relationship, Radiation , Gene Expression Regulation/radiation effects , Humans , Muscle Cells/metabolism , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/biosynthesis , RNA, Ribosomal, 28S/biosynthesis , RNA, Ribosomal, 28S/genetics , Radiation Tolerance , Reactive Oxygen Species , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
10.
DNA Repair (Amst) ; 2(12): 1297-308, 2003 Dec 09.
Article in English | MEDLINE | ID: mdl-14642560

ABSTRACT

Nucleotide excision repair (NER) is the principal pathway for the removal of a wide range of DNA helix-distorting lesions. Two NER subpathways have been identified, i.e. global genome repair (GGR) and transcription-coupled repair (TCR). Little is known about the expression of NER pathways in differentiated cells. We assessed the repair of UV-induced cyclobutane pyrimidine dimers (CPD) and 6-4-photoproducts (6-4 PP) in terminally differentiated myocytes and proliferating fibroblasts isolated from the hearts of neonatal rats. Myocytes and fibroblasts were found to carry out efficient removal of 6-4 PP but display poor repair of CPD by GGR. Furthermore, both cell types were found to carry out TCR of CPD, thus mimicking the repair phenotype of established rodent cell lines. The inefficient repair of CPD at the genome overall level occurs in the absence of massive apoptosis, but goes along with an undetectable level of transcription of the p48 gene, known to be mutated in xeroderma pigmentosum group E (XP-E) patients and recently proposed to be essential for repair of CPD in nonexpressed DNA. Taken together, the results suggest that primary non-dividing cardiac myocytes and proliferating fibroblasts from rat heart selectively remove CPD from the transcribed strand of transcriptionally active genes. GGR of CPD is poor due to the absence of p48 expression.


Subject(s)
DNA Repair/genetics , DNA/radiation effects , Genome , Heart/radiation effects , Pyrimidine Dimers/genetics , Animals , Apoptosis/radiation effects , Caspase 3 , Caspases/metabolism , Cell Differentiation , Cell Division , Cells, Cultured , DNA Damage , Fibroblasts/radiation effects , Male , Muscle Cells/radiation effects , RNA/genetics , RNA/metabolism , RNA/radiation effects , Rats , Rats, Wistar , Ultraviolet Rays
11.
Int J Radiat Biol ; 79(11): 889-96, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14698957

ABSTRACT

PURPOSE: To determine the effect of daily fractionated irradiation on the expression of growth factors and cytokines in different cardiac and vascular cell types. MATERIALS AND METHODS: Cell cultures of rat cardiac myocytes, fibroblasts, a rat cardiac microvascular endothelial cell line and human artery endothelial cells were irradiated with doses of 2 Gy, given daily during 5 consecutive days. Twenty-four hours after each fraction, gene expression was determined by competitive or semiquantitative polymerase chain reaction. Protein secretion into culture media was determined by enzyme-linked immunoabsorbant assay. RESULTS: Of all investigated mRNA levels, transforming growth factor (TGF)-ss1 and fibroblast growth factor (FGF)-2 were slightly upregulated in the rat cardiac endothelial cell line after irradiation. TGF-ss1 protein secretion by these cells was slightly, but non-significantly, elevated. Interleukin 1ss protein levels in myocyte culture media were decreased in control cultures at days 3 and 4 compared with day 2. No significant changes were observed in expression of FGF-2 in either of the four cell types. Moreover, no changes were observed in gene expression of platelet-derived growth factors A, B and interleukin 8 in the human artery endothelial cells. CONCLUSIONS: Fractionated irradiation leads to minor changes in the expression of specific cytokines in cardiac myocytes, fibroblasts and endothelial cells.


Subject(s)
Cytokines/biosynthesis , Endothelium, Vascular/radiation effects , Muscle Cells/radiation effects , Radiation, Ionizing , Animals , Cell-Free System , Cells, Cultured , DNA, Complementary/metabolism , Dose Fractionation, Radiation , Dose-Response Relationship, Radiation , Endothelial Cells/radiation effects , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 2/metabolism , Fibroblasts/metabolism , Humans , Interleukin-8/metabolism , Platelet-Derived Growth Factor/metabolism , Polymerase Chain Reaction , Proto-Oncogene Proteins c-sis/metabolism , RNA, Messenger/metabolism , Rats , Time Factors , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
13.
Mitochondrion ; 14(1): 42-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24246911

ABSTRACT

Near-infrared (NIR) light is a complementary therapy used to treat musculoskeletal injuries but the underlying mechanisms are unclear. Acute NIR light treatment (~800-950 nm; 22.8 J/cm(2)) induced a dose-dependent increase in mitochondrial signaling (AMPK, p38 MAPK) in differentiated muscle cells. Repeated NIR light exposure (4 days) appeared to elevate oxidative stress and increase the upstream mitochondrial regulatory proteins AMPK (3.1-fold), p38 (2.8-fold), PGC-1α (19.7%), Sirt1 (26.8%), and reduced RIP140 (23.2%), but downstream mitochondrial regulation/content (Tfam, NRF-1, Sirt3, cytochrome c, ETC subunits) was unaltered. Our data indicates that NIR light alters mitochondrial biogenesis signaling and may represent a mechanistic link to the clinical benefits.


Subject(s)
Infrared Rays , Mitochondria/physiology , Mitochondria/radiation effects , Mitochondrial Turnover/radiation effects , Muscle Cells/physiology , Muscle Cells/radiation effects , Signal Transduction , Animals , Mice
14.
PLoS One ; 7(7): e40937, 2012.
Article in English | MEDLINE | ID: mdl-22815873

ABSTRACT

Essentially any behavior in simple and complex animals depends on neuronal network function. Currently, the best-defined system to study neuronal circuits is the nematode Caenorhabditis elegans, as the connectivity of its 302 neurons is exactly known. Individual neurons can be activated by photostimulation of Channelrhodopsin-2 (ChR2) using blue light, allowing to directly probe the importance of a particular neuron for the respective behavioral output of the network under study. In analogy, other excitable cells can be inhibited by expressing Halorhodopsin from Natronomonas pharaonis (NpHR) and subsequent illumination with yellow light. However, inhibiting C. elegans neurons using NpHR is difficult. Recently, proton pumps from various sources were established as valuable alternative hyperpolarizers. Here we show that archaerhodopsin-3 (Arch) from Halorubrum sodomense and a proton pump from the fungus Leptosphaeria maculans (Mac) can be utilized to effectively inhibit excitable cells in C. elegans. Arch is the most powerful hyperpolarizer when illuminated with yellow or green light while the action spectrum of Mac is more blue-shifted, as analyzed by light-evoked behaviors and electrophysiology. This allows these tools to be combined in various ways with ChR2 to analyze different subsets of neurons within a circuit. We exemplify this by means of the polymodal aversive sensory ASH neurons, and the downstream command interneurons to which ASH neurons signal to trigger a reversal followed by a directional turn. Photostimulating ASH and subsequently inhibiting command interneurons using two-color illumination of different body segments, allows investigating temporal aspects of signaling downstream of ASH.


Subject(s)
Caenorhabditis elegans/physiology , Light , Nerve Net/physiology , Nerve Net/radiation effects , Neurons/physiology , Proton Pumps/pharmacology , Proton Pumps/radiation effects , Animals , Archaeal Proteins/pharmacology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/radiation effects , Cholinergic Neurons/drug effects , Cholinergic Neurons/physiology , Fungal Proteins/pharmacology , Halorhodopsins/pharmacology , Locomotion/drug effects , Motor Neurons/drug effects , Motor Neurons/physiology , Muscle Cells/drug effects , Muscle Cells/physiology , Muscle Cells/radiation effects , Nerve Net/drug effects , Neurons/drug effects , Neurons/radiation effects , Nociception/drug effects , Optogenetics , Signal Transduction/drug effects , Touch/drug effects
15.
Genetics ; 189(3): 885-97, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21868609

ABSTRACT

The acquisition and maintenance of shape is critical for the normal function of most cells. Here we investigate the morphology of the pharyngeal glands of Caenorhabditis elegans. These unicellular glands have long cellular processes that extend discrete lengths through the pharyngeal musculature and terminate at ducts connected to the pharyngeal lumen. From a genetic screen we identified several mutants that affect pharyngeal gland morphology. The most severe such mutant is an allele of sma-1, which encodes a ß-spectrin required for embryonic elongation, including elongation of the pharynx. In sma-1 mutants, gland projections form normally but become increasingly abnormal over time, acquiring additional branches, outgrowths, and swelling, suggestive of hypertrophy. Rather than acting in pharyngeal glands, sma-1 functions in the surrounding musculature, suggesting that pharyngeal muscles play a critical role in maintenance of gland morphology by restricting their growth, and analysis of other mutants known to affect pharyngeal muscles supports this hypothesis. We suggest that gland morphology is maintained by a balance of forces from the muscles and the glands.


Subject(s)
Caenorhabditis elegans/cytology , Muscle Cells/cytology , Pharyngeal Muscles/cytology , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/radiation effects , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/genetics , DNA Mutational Analysis , Gene Expression Regulation/radiation effects , Hypertrophy/genetics , Lasers/adverse effects , Membrane Proteins/genetics , Models, Biological , Muscle Cells/metabolism , Muscle Cells/radiation effects , Muscle Cells/ultrastructure , Pharyngeal Muscles/metabolism , Pharyngeal Muscles/pathology , Pharyngeal Muscles/radiation effects
16.
Ukr Biokhim Zh (1999) ; 81(5): 66-73, 2009.
Article in Ukrainian | MEDLINE | ID: mdl-20387649

ABSTRACT

The effects of the N-stearoylethanolamine (NSE), N-oleoylethanolamine (OEA) and N-acylethanolamine (NAE) mixture on the cell survival, apoptosis and activity of mitochondrial succinate dehydrogenase (SDG) and glycerophosphate dehydrogenase (GFDG) in embryonic cell culture under normal conditions and irradiation were compared in the work. It was shown, that all investigated NAE were able to modulate the proliferative activity of intact cells as well as irradiation-exposed cells in concentration-depended manner. The most pronounced effect was observed under the NAE mixture action. It was established, that NAE prevented the damage effects of the irradiation and diminished the activation of apoptotic cell death. It was found that NSE and OEA decreased the activity of the SDG (42.3 and 44.14%, accordingly) and the GFDG activity (14.67 and 17.33%, accordingly) in embryonic cell culture, while addition of the NAE mixture decreased SDG activity by 20%, at the same time GFDG activity increased by 20%. Our findings suggested that antiproliferative effects of NAE depended on their influence on mitochondrial functioning.


Subject(s)
Antioxidants/pharmacology , Ethanolamines/pharmacology , Fibroblasts , Muscle Cells , Muscles , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Culture Techniques , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/radiation effects , Glycerolphosphate Dehydrogenase/metabolism , Mitochondria , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/enzymology , Mitochondria, Muscle/radiation effects , Muscle Cells/drug effects , Muscle Cells/radiation effects , Muscles/cytology , Muscles/embryology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Rats , Succinate Dehydrogenase/metabolism
18.
Bioelectromagnetics ; 25(5): 380-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15197763

ABSTRACT

A theoretical model is proposed for the evaluation of dielectric properties of the cell nucleus between 0.3 and 3 GHz, as a function of its nucleic acids (NA) concentration (CNA). It is based on literature data on dielectric properties of DNA solutions and nucleoplasm. In skeletal muscle cells, the specific absorption rate (SAR) ratio between nucleoplasm and cytoplasm is found to be larger than one for CNA above 30 mg/ml. A nearly linear relationship is found between CNA and this nucleocytoplasmic SAR ratio. Considering the nanoscale of the layer of condensed counterions and bound water molecules at the NA-solution interface, the power absorption per unit volume is evaluated at this precise location. It is found to be between one and two orders of magnitude above that in muscle tissue as a whole. Under realistic microwave (MW) exposure conditions, however, these SAR inhomogeneities do not generate any significant thermal gradient at the scale considered here. Nevertheless, the question arises of a possible biological relevance of nonnegligible and preferential heat production at the location of the cell nucleus and of the NA molecules.


Subject(s)
Microwaves , Models, Biological , Nucleic Acids/radiation effects , Absorption , Cell Nucleus/radiation effects , Cytoplasm/radiation effects , DNA/radiation effects , Electric Conductivity , Electrochemistry , Environmental Exposure , Humans , Intracellular Fluid/radiation effects , Intranuclear Space/radiation effects , Muscle Cells/radiation effects , Muscle, Skeletal/cytology , RNA/radiation effects
19.
Heart Vessels ; 18(4): 207-12, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14520490

ABSTRACT

We studied the effects of cobalt-60 Gamma-radiation on the gene expression and secretion of adrenomedullin (Adm) and endothelin (ET) in cultured rat vascular smooth muscle cells (VSMCs). Rat VSMCs cultured in Dulbecoo's modified Eagle's medium containing 10% FBS were radiated with cobalt-60 Gamma-radiation at doses of 1, 14, and 25 Gy, respectively. Then the mRNA of Adm and ET in VSMCs were detected by the reverse-transcriptative competitive polymerase chain reaction. Adm and ET levels in rat VSMCs were measured by radioimmunoassay. As compared with that of the control, the secretions of Adm in rat VSMCs radiated at doses of 14 and 25 Gy were increased by 270% (P < 0.05) and 233% (P < 0.05), respectively. The mRNA levels of Adm were increased by 82.4% (P < 0.01) and 101% (P <0.01), respectively. Meanwhile, the secretions of ET were decreased by 27.3% (P < 0.01) and 58.0% (P < 0.01) in VSMCs radiated at doses of 14 and 25 Gy, respectively. In parallel, the mRNA levels of ET were decreased by 47.1% (P < 0.01) and 40.2% (P < 0.01), respectively. Radiation at a dose of 1 Gy had no significant effect on Adm and ET at the gene and protein levels. As compared with the control, the Adm/ET ratios in VSMCs increased by 65% (P > 0.05), 409% (P < 0.01), and 693% (P < 0.01), respectively, with radiation at doses of 1,14 and 25 Gy, respectively. The balance of Adm/ET in VSMCs could be changed by cobalt-60 Gamma-radiation, which might play an important role in the use of radiotherapy for restenosis.


Subject(s)
Cobalt Radioisotopes , Endothelins/biosynthesis , Gamma Rays , Muscle Cells/radiation effects , Muscle, Smooth, Vascular/radiation effects , Peptides/metabolism , Adrenomedullin , Animals , Dose-Response Relationship, Radiation , Endothelins/genetics , Male , Muscle Cells/metabolism , Muscle, Smooth, Vascular/metabolism , Peptides/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL