Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Biotechnol J ; 12(1)2017 Jan.
Article in English | MEDLINE | ID: mdl-27440252

ABSTRACT

As a possible viable and non-invasive method to identify high producing cells, Confocal Raman Microscopy was shown to be able to differentiate CHO host cell lines and derivative production clones. Cluster analysis of spectra and their derivatives was able to differentiate between different producer cell lines and a host, and also distinguished between an intracellular region of high lipid and protein content that in structure resembles the Endoplasmic Reticulum. This ability to identify the ER may be a major contributor to the identification of high producers. PCA enabled the discrimination even of host cell lines and their subclones with inherently higher production capacity. The method is thus a promising option that may contribute to early, non-invasive identification of high potential candidates during cell line development and possibly could also be used for proof of identity of established production clones.


Subject(s)
CHO Cells/cytology , CHO Cells/ultrastructure , Microscopy, Confocal/methods , Protein Engineering/methods , Adalimumab/genetics , Adalimumab/metabolism , Amine Oxidase (Copper-Containing)/genetics , Amine Oxidase (Copper-Containing)/metabolism , Animals , Cluster Analysis , Cricetulus , Endoplasmic Reticulum/ultrastructure , Humans , Lipids/chemistry , Metals/chemistry , Molecular Imaging/methods , Principal Component Analysis , Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrum Analysis, Raman/methods
2.
Nanotoxicology ; 10(2): 204-15, 2016.
Article in English | MEDLINE | ID: mdl-26037905

ABSTRACT

Cellular organelles have been shown to shuttle between cells in co-culture. We hereby show that titanium dioxide (TiO2) nanoparticles (NPs) can be transferred in such a manner, between cells in direct contact, along with endosomes and lysosomes. A co-culture system was employed for this purpose and the NP transfer was observed in mammalian cells including normal rat kidney (NRK) and HeLa cells. We found that the small GTPase Arf6 facilitates the intercellular transfer of smaller NPs and agglomerates. Spherical, anatase nano-TiO2 with sizes of 5 (Ti5) and 40 nm (Ti40) were used in this study. Humans are increasingly exposed to TiO2 NPs from external sources such as constituents of foods, cosmetics, and pharmaceuticals, or from internal sources represented by Ti-based implants, which release NPs upon abrasion. Exposure to 5 mg/l of Ti5 and Ti40 for 24 h did not affect cellular viability but modified their ability to communicate with surrounding cells. Altogether, our results have important implications for the design of nanomedicines, drug delivery and toxicity.


Subject(s)
Cell Communication , Nanoparticles/metabolism , Titanium/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/physiology , Animals , CHO Cells/metabolism , CHO Cells/ultrastructure , Cell Survival/drug effects , Coculture Techniques , Cricetulus , HeLa Cells/metabolism , HeLa Cells/ultrastructure , Humans , Kidney/metabolism , Kidney/ultrastructure , Lysosomes/metabolism , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Particle Size , Titanium/chemistry , Titanium/toxicity
3.
J Acoust Soc Am ; 128(5): 3175-80, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21110612

ABSTRACT

A cell pellet biophantom technique is introduced, and applied to the ultrasonic backscatter coefficient (BSC) estimate using Chinese hamster ovary (CHO) cells. Also introduced is a concentric sphere scattering model because of its geometrical similarities to cells with a nucleus. BSC comparisons were made between the concentric sphere model and other well-understood models for mathematical verification purposes. BSC estimates from CHO cell pellet biophantoms of known number density were performed with 40 and 80 MHz focused transducers (overall bandwidth: 26-105 MHz). These biophantoms were histologically processed and then evaluated for cell viability. Cell pellet BSC estimates were in agreement with the concentric sphere model. Fitting the model to the BSC data yielded quantitative values for the outer sphere and inner sphere. The radius of the cell model was 6.8 ± 0.7 µm; the impedance of the cytoplasm model was 1.63 ± 0.03 Mrayl and the impedance of the nuclear model was 1.55 ± 0.09 Mrayl. The concentric sphere model appears as a new tool for providing quantitative information on cell structures and will tend to have a fundamental role in the classification of biological tissues.


Subject(s)
CHO Cells/diagnostic imaging , Models, Biological , Phantoms, Imaging , Ultrasonics/methods , Ultrasonography/methods , Animals , CHO Cells/ultrastructure , Cell Count , Cell Nucleus/diagnostic imaging , Cell Size , Cricetinae , Cricetulus , Cytoplasm/diagnostic imaging
4.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP, SESSP-IBACERVO | ID: biblio-1062325

ABSTRACT

Proline-rich peptides from Bothrops jararaca venom (Bj-PRO) were characterized based on the capability to inhibit the somatic angiotensin-converting enzyme. The pharmacologicalaction of these peptides resulted in the development of Captopril, one of thebest examples of a target-driven drug discovery for treatment of hypertension. However, biochemical and biological properties of Bj-PROs were not completely elucidated yet, and many recent studies have suggested that their activity relies on angiotensinconvertingenzyme-independent mechanisms. Here, we show that Bj-PRO-7a (

Subject(s)
Bothrops/physiology , Hypertension/therapy , Angiotensin-Converting Enzyme Inhibitors/analysis , Muscarinic Agonists/analysis , Muscarinic Agonists/therapeutic use , CHO Cells/ultrastructure , Microscopy, Confocal/methods , Proline/analogs & derivatives
5.
J Neurol ; 257(10): 1661-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20461396

ABSTRACT

Mutations in MPZ, which encodes myelin protein zero (P(0)), may lead to different subtypes of Charcot-Marie-Tooth disease (CMT). The aim of this study was to characterize the cellular manifestations of various MPZ mutations associated with CMT1, Dejerine-Sottas syndrome (DSS) and CMT2, and to correlate their cellular and clinical phenotypes. Nine P(0) mutants associated with CMT1 (P(0)S63F, R98H, R277S, and S233fs), DSS (P(0) I30T and R98C), and CMT2 (P(0)S44F, D75V, and T124M), were investigated. Wild-type and mutant P(0) fused with fluorescent proteins were expressed in vitro to monitor their intracellular localization. An adhesiveness assay was used to evaluate the adhesiveness of the transfected cells. Protein localization and cell adhesiveness of each mutant protein were compared and correlated with their clinical phenotypes. Three different intracellular localization patterns of the mutant P(0) were observed. Wild-type P(0), P(0)I30T, S44F, S63F, D75V, T124M, and R227S were mostly localized on the cell membrane, P(0)R98H, and R98C were found in the endoplasmic reticulum (ER) or Golgi apparatus, and P(0)S233fs formed aggregates within the ER. Cells expressing mutant P(0), as compared with those expressing wild-type P(0), demonstrated variable degrees of reduction in the cell adhesiveness. The molecular patho-mechanisms of MPZ mutations are likely very complex and the clinical phenotype must be influenced by many genetic or environmental factors. This complexity may contribute to the highly variable clinical manifestations resulting from different MPZ mutations.


Subject(s)
Mutation/genetics , Myelin P0 Protein/genetics , Myelin P0 Protein/metabolism , Phenotype , Animals , CHO Cells/metabolism , CHO Cells/ultrastructure , Cell Adhesion/genetics , Charcot-Marie-Tooth Disease/genetics , Cricetinae , Cricetulus , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Green Fluorescent Proteins/genetics , Hereditary Sensory and Motor Neuropathy , Humans , Models, Molecular , Myelin P0 Protein/chemistry , Time Factors , Transfection
6.
J Occup Health ; 52(3): 155-66, 2010.
Article in English | MEDLINE | ID: mdl-20379079

ABSTRACT

OBJECTIVES: The potential applications and industrial production of multi-wall carbon nanotubes (MWCNT) have raised serious concerns about their safety for human health and the environment. The present study was designed to examine the in vitro cytotoxicity and genotoxicity of MWCNT and UICC chrysotile A (chrysotile). METHODS: Cytotoxicity using both colony formation and lactate dehydrogenase (LDH) assays and genotoxicity including chromosome aberration, micronucleus induction and hgprt mutagenicity were examined by exposing cultured Chinese hamster lung (CHL/IU) cells to MWCNT or chrysotile at different concentrations. RESULTS: The in vitro cytotoxicity of MWCNT depended on the solvent used for suspension of MWCNT and ultrasonication duration of the MWCNT suspension. A combination of DMSO/culture medium and 3-minute ultrasonication resulted in a well-dispersed medium with dispersion and isolation of agglomerated MWCNT by ultrasonication which manifested the highest cytotoxicity. The cytotoxicity was more potent for chrysotile than MWCNT. The genotoxicity of MWCNT was characterized by the formation of polyploidy without structural chromosome aberration, and an increased number of bi- and multi-nucleated cells without micronucleus induction, as well as negative hgprt mutagenicity. Chrysotile exhibited essentially the same genotoxicity as MWCNT, except for marginal but significant induction of micronuclei. MWCNT and chrysotile were incompletely internalized in the cells and localized in the cytoplasm. CONCLUSIONS: MWCNT and chrysotile were cytotoxic and genotoxic in Chinese hamster lung cells, but might interact indirectly with DNA. The results suggest that both test substances interfere physically with biological processes during cytokinesis.


Subject(s)
Asbestos, Serpentine/toxicity , Nanotubes, Carbon/toxicity , Animals , CHO Cells/drug effects , CHO Cells/ultrastructure , Chromosome Aberrations/chemically induced , Cricetinae , Cricetulus , Cytokinesis/drug effects , Dose-Response Relationship, Drug , L-Lactate Dehydrogenase/biosynthesis , Micronucleus Tests , Microscopy, Electron, Scanning
7.
J Neurosci ; 29(41): 13042-52, 2009 Oct 14.
Article in English | MEDLINE | ID: mdl-19828817

ABSTRACT

Amyloid beta protein (Abeta), a pathogenic molecule associated with Alzheimer's disease, is produced by gamma-secretase, which cleaves the beta-carboxyl terminal fragment (betaCTF) of beta-amyloid precursor protein in the middle of its transmembrane domain. How the cleavage proceeds within the membrane has long been enigmatic. We hypothesized previously that betaCTF is cleaved first at the membrane-cytoplasm boundary, producing two long Abetas, Abeta(48) and Abeta(49), which are processed further by releasing three residues at each step to produce Abeta(42) and Abeta(40), respectively. To test this hypothesis, we used liquid chromatography tandem mass spectrometry (LC-MS/MS) to quantify the specific tripeptides that are postulated to be released. Using CHAPSO (3-[(3-cholamidopropyl)dimethylammonio]-2-hydroxyl-1-propanesulfonate)-reconstituted gamma-secretase system, we confirmed that Abeta(49) is converted to Abeta(43/40) by successively releasing two or three tripeptides and that Abeta(48) is converted to Abeta(42/38) by successively releasing two tripeptides or these plus an additional tetrapeptide. Most unexpectedly, LC-MS/MS quantification revealed an induction period, 3-4 min, in the generation of peptides. When extrapolated, each time line for each tripeptide appears to intercept the same point on the x-axis. According to numerical simulation based on the successive reaction kinetics, the induction period exists. These results strongly suggest that Abeta is generated through the stepwise processing of betaCTF by gamma-secretase.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Peptide Fragments/metabolism , Amyloid beta-Protein Precursor/chemistry , Analysis of Variance , Animals , CHO Cells/ultrastructure , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholic Acids/pharmacology , Chromatography, Liquid/methods , Cricetinae , Cricetulus , Detergents/pharmacology , Immunoprecipitation/methods , Models, Biological , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptide Fragments/analysis , Protein Structure, Tertiary/physiology , Substrate Specificity , Tandem Mass Spectrometry/methods , Time Factors
8.
Neurosci Lett ; 447(2-3): 172-4, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-18840504

ABSTRACT

Huntington's disease is an autosomal dominant neurodegenerative disorder caused by the expansion of a polyglutamine repeat tract in the huntingtin protein. Polyglutamine-expanded huntingtin forms intranuclear as well as perinuclear inclusion bodies. Perinuclear aggregates formed by polyglutamine-expanded proteins are associated with a characteristic indentation of the nuclear envelope. We examined the nuclear envelope in cells containing huntingtin aggregates using immunostaining for lamin B1, a major component of the nuclear lamina. Laser confocal microscopy analysis revealed that huntingtin aggregates in a juxtanuclear position were associated with a clear focal distortion in the nuclear envelope in cells transfected with polyglutamine-expanded huntingtin. Lamin B1 distribution was not altered by aggregates of polyglutamine-expanded ataxin-1, that are exclusively intranuclear. Thus lamin immunocytochemistry demonstrates clearly the depression of the nuclear envelope resulting from the formation of perinuclear aggregates by polyglutamine-expanded huntingtin. Lamin immunocytochemistry would be of value to monitor the state of the nuclear envelope in experimental paradigms aimed at establishing the significance of perinuclear aggregates of pathogenic proteins.


Subject(s)
Lamin Type B/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Animals , Ataxin-1 , Ataxins , CHO Cells/ultrastructure , Cricetinae , Cricetulus , Glutamine/metabolism , Green Fluorescent Proteins/genetics , Humans , Huntingtin Protein , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Microscopy, Confocal/methods , Nerve Tissue Proteins/genetics , Nuclear Envelope/genetics , Nuclear Proteins/genetics , Peptides/genetics , Transfection/methods
9.
J Microsc ; 230(Pt 2): 308-16, 2008 May.
Article in English | MEDLINE | ID: mdl-18445161

ABSTRACT

Cryo-electron microscopy of vitrified specimen is the method of choice to explore cellular ultrastructure at high resolution as close as possible to the native state and environment. In this study, we investigated the Golgi apparatus - the main organelle of the secretory pathway. Cultured mammalian cells were fixed by high-pressure freezing, sectioned in vitreous ice and subjected to cryo-electron microscopy and cryo-electron tomography. Although the overall morphology of Golgi stacks was comparable to well prepared and plastic-embedded samples, in detail we reached much higher resolution in terms of distinction between biological structures based on their native density. On cisternal buds and peri-Golgi vesicles--some associated with microtubules--we detected two different subtypes of COPI coats: (1) a homogenous coat and (2) an inhomogeneous spiky coat, providing an 8-9 nm regularity, clearly distinct from clathrin coat. Next, we monitored the secretion of cargo, namely, procollagen I, through the Golgi complex. Temporally correlated with fluorescence microscopy, we performed three-dimensional cryo-electron tomography analysis and detected Golgi cisternae enlarged to saccules, containing cargo and showing inter-cisternal connections. Our work provides a first step towards the high-resolution description of the secretory pathway in native vitrified samples and describes the challenges associated with this attempt.


Subject(s)
Cryoelectron Microscopy/methods , Cryopreservation/methods , Frozen Sections , Golgi Apparatus/ultrastructure , Animals , CHO Cells/ultrastructure , COP-Coated Vesicles/ultrastructure , Cricetinae , Cricetulus , Freezing , Hydrostatic Pressure , Microscopy, Fluorescence , Tomography
10.
Autoimmunity ; 41(2): 143-53, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18324484

ABSTRACT

Since GAD65 undergoes post-translational processing and targeting to subcellular compartments and membranes, it may exhibit different immunochemical properties in the cell context compared with the soluble protein expressed in the cell-free eukaryotic system used in the reference method for GADA assessment (radioligand binding assay (RBA)). In the present work, we detected and characterized GADA in 72 sera from patients with type 1 diabetes mellitus (DM) and 14 sera from adult-onset diabetes patients using analytical systems in which GAD65 is expressed in a cellular context: confocal indirect immunofluorescence (IIF) and electron microscopy after immunogold labeling on monolayers of transfected Chinese hamster ovary (CHO) cells, and immunoprecipitation (IP) of metabolically labeled GAD65. Eighteen serum samples, 16 from type 1 diabetes patients and two from adult-onset diabetes patients, were positive by confocal IIF but scored negative by RBA. All of these 18 sera immunoprecipitated a 65 kDa protein, supporting the existence of the GADA marker in such patients. It may be concluded that GADA negativity by the conventional RBA method using the soluble antigen, as well as negativity for other common markers measured by similar methods, is not enough to rule out the existence of the specific autoimmune component in childhood or adult-onset diabetes. Other analytical methods based in a more physiological presentation of the autoantigen structure, as confocal IIF and IP, bring an extra support to assess the complete repertoire of specific autoantibodies to native-like and membrane-bound, or denatured, beta-cell antigens.


Subject(s)
Autoantibodies/blood , Autoantigens/immunology , Diabetes Mellitus, Type 1/immunology , Glutamate Decarboxylase/immunology , Adolescent , Adult , Animals , Autoantibodies/chemistry , Autoantigens/genetics , Autoimmunity , Biomarkers/blood , Biomarkers/chemistry , CHO Cells/ultrastructure , Child , Cricetinae , Cricetulus , Diabetes Mellitus, Type 2/immunology , Female , Fluorescent Antibody Technique , Glutamate Decarboxylase/genetics , Humans , Immunoprecipitation , Male , Microscopy, Confocal , Microscopy, Electron , Radioligand Assay , Transfection
11.
Eur J Neurosci ; 26(11): 3126-40, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18005063

ABSTRACT

Several causal missense mutations in protein kinase C gamma (gamma PKC) gene have been found in spinocerebellar ataxia type 14 (SCA14), an autosomal dominant neurodegenerative disease. We previously demonstrated that mutant gamma PKC found in SCA14 is susceptible to two types of aggregation, cytoplasmic dot-like and perinuclear massive aggregation, and causes cell death in Chinese hamster ovary cells. Long-term time-lapse imaging revealed that firstly accumulated dot-like aggregation of mutant gamma PKC-green fluorescent protein (GFP) gradually formed perinuclear massive aggregations, followed by cell death. However, it remains unclear how aggregate formation of mutant gamma PKC causes cell death. In the present study, we examined whether these mutant aggregations affect the ubiquitin-proteasome system (UPS) and endoplasmic reticular (ER) stress. Two mutant gamma PKC-GFPs (S119P and G128D) were strongly ubiquitinated, and dot-like aggregations of these mutants were ubiquitin-positive and colocalized with proteasome 20S. Furthermore, proteasome activity in cells with aggregates, especially massive ones, was significantly decreased. Aggregate formation of mutant gamma PKC-GFP induced phosphorylation of PERK (PKR-like ER kinase) and nuclear expression of CHOP (C/EBP homologous protein), hallmarks of ER stress and subsequently activated caspase-3. These results indicate that aggregate formation of mutant gamma PKC found in SCA14 impairs UPS and induces ER stress, leading to apoptotic cell death.


Subject(s)
Endoplasmic Reticulum/pathology , Mutation/physiology , Proteasome Endopeptidase Complex/metabolism , Protein Kinase C/genetics , Stress, Physiological/pathology , Ubiquitin/physiology , Animals , Aspartic Acid/genetics , CHO Cells/metabolism , CHO Cells/ultrastructure , Caspases/metabolism , Cell Death/physiology , Cricetinae , Cricetulus , Glycine/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation/methods , Proline/genetics , Protein Processing, Post-Translational/physiology , Serine/genetics , Time Factors , Transfection/methods
12.
Brain Res ; 1184: 46-56, 2007 Dec 12.
Article in English | MEDLINE | ID: mdl-17980352

ABSTRACT

Lipid rafts are small cholesterol- and glycosphingolipid-enriched membrane subdomains. Here we compared the mu opioid receptor (MOR)-lipid rafts relationship in the rat brain, where neurons have non-caveolae rafts, and in CHO cells stably transfected with HA-rat MOR (CHO-HA-rMOR), which are enriched in caveolae. Membranes of rat caudate putamen (CPu) and thalamus or CHO-HA-rMOR cells were homogenized, sonicated in a detergent-free 0.5 M Na(2)CO(3) buffer and fractionated through sucrose density gradients. Western blot and [(3)H]diprenorphine binding showed that approximately 70% of MOR in CHO-HA-rMOR was present in low-density (5-20% sucrose) fractions enriched in cholesterol and/or ganglioside M1 (GM1) (lipid rafts) in plasma membranes, whereas about 70% and 45% of MOR in CPu and thalamus, respectively, were associated with lipid rafts. Incubation with a saturating concentration of etorphine or morphine at 37 degrees C for 30 min failed to change the MOR location in rafts in CHO-HA-rMOR, indicating that the internalized MOR does not move out of rafts, in contrast to the delta opioid receptor. In vivo, rafts association of MOR in CPu and thalamus was not affected significantly in rats implanted with two 75-mg morphine pellets for 72 h. In addition, cholesterol reduction by methyl-beta-cyclodextrin (MCD) disrupted rafts and shifted MOR to higher density fractions in both CHO-HA-rMOR and CPu membranes. However, MCD treatment had opposite impacts on MOR signaling in the two tissues: it attenuated MOR-mediated [(35)S]GTPgammaS binding in CPu but enhanced it in CHO-HA-rMOR.


Subject(s)
Brain/ultrastructure , Cholesterol/metabolism , Membrane Microdomains/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction/physiology , Animals , Binding, Competitive/drug effects , CHO Cells/drug effects , CHO Cells/ultrastructure , Caveolin 1/metabolism , Cricetinae , Cricetulus , Diprenorphine/metabolism , Dose-Response Relationship, Drug , Etorphine/pharmacology , Male , Membrane Microdomains/drug effects , Morphine/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/metabolism , Narcotic Antagonists/pharmacology , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transfection
13.
Glycobiology ; 17(10): 1084-93, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17681998

ABSTRACT

A glycosyl hydrolase family 38 enzyme, neutral alpha-mannosidase, has been proposed to be involved in hydrolysis of cytosolic free oligosaccharides originating either from ER-misfolded glycoproteins or the N-glycosylation process. Although this enzyme has been isolated from the cytosol, it has also been linked to the ER by subcellular fractionations. We have studied the subcellular localization of neutral alpha-mannosidase by immunofluorescence microscopy and characterized the human recombinant enzyme with natural substrates to elucidate the biological function of this enzyme. Immunofluorescence microscopy showed neutral alpha-mannosidase to be absent from the ER, lysosomes, and autophagosomes, and being granularly distributed in the cytosol. In experiments with fluorescent recovery after photo bleaching, neutral alpha-mannosidase had slower than expected two-phased diffusion in the cytosol. This result together with the granular appearance in immunostaining suggests that portion of the neutral alpha-mannosidase pool is somehow complexed. The purified recombinant enzyme is a tetramer and has a neutral pH optimum for activity. It hydrolyzed Man(9)GlcNAc to Man(5)GlcNAc in the presence of Fe(2+), Co(2+), and Mn(2+), and uniquely to neutral alpha-mannosidases from other organisms, the human enzyme was more activated by Fe(2+) than Co(2+). Without activating cations the main reaction product was Man(8)GlcNAc, and Cu(2+) completely inhibited neutral alpha-mannosidase. Our findings from enzyme-substrate characterizations and subcellular localization studies support the suggested role for neutral alpha-mannosidase in hydrolysis of soluble cytosolic oligomannosides.


Subject(s)
Cytosol/enzymology , Oligosaccharides/metabolism , alpha-Mannosidase/metabolism , Animals , Autophagy , CHO Cells/ultrastructure , Cricetinae , Cricetulus , Endoplasmic Reticulum/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Glycosylation , Humans , Hydrolysis , Immunization , Immunoglobulin G/immunology , Lysosomes/metabolism , Male , Microscopy, Confocal , Microscopy, Fluorescence , Pichia/growth & development , Pichia/metabolism , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Subcellular Fractions
14.
Anal Chem ; 78(1): 35-43, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16383308

ABSTRACT

G protein-coupled receptors (GPCRs) constitute important targets for drug discovery against a wide range of ailments including cancer, inflammatory, and cardiovascular diseases. Efforts are underway to screen selective modulators of GPCRs and also to deorphanize GPCRs with unidentified natural ligands. Most GPCR-based cellular screens depend on labeling or recombinant expression of receptor or reporter proteins, which may not capture the true physiology or pharmacology of the GPCRs. In this paper, we describe a noninvasive and label-free assay for GPCRs that can be used with both engineered and nonengineered cell lines. The assay is based on using cell-electrode impedance to measure minute changes in cellular morphology as a result of ligand-dependent GPCR activation. We have used this technology to assay the functional activation of GPCRs coupled to different signaling pathways and have compared it to standard assays. We have used pharmacological modulators of GPCR signaling pathways to demonstrate the specificity of impedance-based measurements. Our data indicate that cell-electrode impedance measurements offer a convenient, sensitive, and quantitative method for assessing GPCR function. Moreover, the noninvasive nature of the readout offers the added advantage of performing multiple treatments in the same well to study events such as desensitization and receptor cross-talk.


Subject(s)
Electronics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Blotting, Western , CHO Cells/metabolism , CHO Cells/ultrastructure , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Dopamine/metabolism , Electric Impedance , HeLa Cells/metabolism , HeLa Cells/ultrastructure , Histamine/metabolism , Humans , Immunoprecipitation , Inositol Phosphates/metabolism , Kinetics , Ligands , Microscopy, Fluorescence , Receptors, Dopamine/genetics , Receptors, Dopamine/metabolism , Receptors, Histamine/genetics , Receptors, Histamine/metabolism , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Spectrum Analysis , Vasopressins/metabolism
15.
Mol Biol Cell ; 16(11): 5087-93, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16107561

ABSTRACT

Polycystin-1 (PC-1) is the product of the PKD1 gene, which is mutated in autosomal dominant polycystic kidney disease. We show that the Na,K-ATPase alpha-subunit interacts in vitro and in vivo with the final 200 amino acids of the polycystin-1 protein, which constitute its cytoplasmic C-terminal tail. Functional studies suggest that this association may play a role in the regulation of the Na,K-ATPase activity. Chinese hamster ovary cells stably expressing the entire PC-1 protein exhibit a dramatic increase in Na,K-ATPase activity, although the kinetic properties of the enzyme remain unchanged. These data indicate that polycystin-1 may contribute to the regulation of Na,K-ATPase activity in kidneys in situ, thus modulating renal tubular fluid and electrolyte transport.


Subject(s)
Ouabain/pharmacology , Proteins , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium-Potassium-Exchanging ATPase/physiology , Sodium/pharmacology , Animals , CHO Cells/ultrastructure , Cell Line , Cricetinae , Cricetulus , Dogs , Enzyme Inhibitors , Escherichia , Mutation , Polycystic Kidney Diseases , Recombinant Proteins , Sodium-Potassium-Exchanging ATPase/biosynthesis , Sodium-Potassium-Exchanging ATPase/drug effects , TRPP Cation Channels , Transfection
16.
Cytogenet Genome Res ; 104(1-4): 137-41, 2004.
Article in English | MEDLINE | ID: mdl-15162027

ABSTRACT

Research on the subtelomeric region has considerably increased because this chromosome segment (1) keeps the chromosome number constant, (2) intervenes in cancer and cell senescence processes, (3) presents more crossovers than other regions of the genome and, (4) is the site of cryptic chromosome aberrations associated with mental retardation and congenital malformations. Quantitative microphotometrical scanning and computer graphic image analysis enables the detection of differentially distributed Giemsa-stained structures in T-banded subtelomeric segments of human and Chinese hamster ovary (CHO) chromosomes. The presence of high density stain patterns in the subtelomeric region was confirmed using endoreduplicated chromosomes as a model. Besides, prolonging the incubation in the T-buffer, specific holes were induced in subtelomeric segments. Hole specificity was confirmed inducing them in complex CHO chromosome aberrations obtained by AluI. The method was also used to detect minute sister chromatid exchanges in the T-banded subtelomeric area (t-SCEs). The presence of t-SCEs was suspected to reflect, at the microscope level, the high crossover activity prevailing in the region. Due to the fact that the fluorescent signals obtained with subtelomeric probes seem to be colocalized with subtelomeric high density areas, measurements on the position of both structures with respect to the diffraction and chromosome edges were carried out. Data obtained showed comparable values suggesting that the high density segments were located where telomeric probes usually fluoresce. The possible relationship of the high density patterns, the production of specific holes, the localization of fluorescent areas and the detection of minute SCEs in the subtelomeric segment observed in T-banded CHO and human chromosomes is briefly reviewed.


Subject(s)
Chromosomes/ultrastructure , Photometry/methods , Animals , CHO Cells/ultrastructure , Cells, Cultured/ultrastructure , Chromosomal Proteins, Non-Histone/physiology , Chromosomes, Human/ultrastructure , Cricetinae , Cricetulus , Humans , Intellectual Disability/genetics , Metaphase , Sister Chromatid Exchange , Telomere/ultrastructure
17.
Cytogenet Genome Res ; 104(1-4): 173-7, 2004.
Article in English | MEDLINE | ID: mdl-15162033

ABSTRACT

In a recent paper we reported the results of an experiment carried out by analysing chromosomal damage in Chinese hamster (CHO) cells exposed to low doses of X-rays. The present investigation was undertaken in order to validate those results using a different approach, the single cell gel electrophoresis assay (comet assay) immediately after irradiation. Cells were cultured during 14 cycles, irradiation treatment was performed once per cycle when the cells were at 90-95% of confluence. Doses of 2.5, 5.0 and 10.0 mSv were used. Sequential irradiation of CHO cells induced a decrease of cells without migration and an increase of cells showing DNA damage with the three doses employed. Significant increases of low-level damaged cells (p < 0.001) were found for the 14 exposures when compared to controls except for the first irradiations with 2.5 and 10 mSv, respectively. No significant increase of the frequency of cells with severe damage was observed in any case. These findings could be explained by assuming a complex interactive process of cell recovery, DNA damage and repair together with the induction of genomic instability, the incidence of bystander effects as well as some kind of radioadaptative response of the cells. If these phenomena are limited to the cell line employed deserves further investigation.


Subject(s)
CHO Cells/radiation effects , DNA Damage , Adaptation, Physiological , Animals , CHO Cells/ultrastructure , Chromatids/radiation effects , Chromatids/ultrastructure , Chromosome Aberrations , Chromosome Breakage , Chromosomes/radiation effects , Chromosomes/ultrastructure , Comet Assay , Cricetinae , Cricetulus , DNA/radiation effects , DNA Repair , Dose-Response Relationship, Radiation , Image Processing, Computer-Assisted , Linear Energy Transfer , Microscopy, Fluorescence , Radiation Tolerance
18.
Cytogenet Genome Res ; 104(1-4): 182-7, 2004.
Article in English | MEDLINE | ID: mdl-15162035

ABSTRACT

SORB (selected observed residual breakpoints) induced by ionizing radiation or endonucleases are often non-randomly distributed in mammalian chromosomes. However, the role played by chromatin structure in the localization of chromosome SORB is not well understood. Anti-topoisomerase drugs such as etoposide are potent clastogens and unlike endonucleases or ionizing radiation, induce DNA double-strand breaks (DSB) by an indirect mechanism. Topoisomerase II (Topo II) is a main component of the nuclear matrix and the chromosome scaffold. Since etoposide leads to DSB by influencing the activity of Topo II, this compound may be a useful tool to study the influence of the chromatin organization on the distribution of induced SORB in mammalian chromosomes. In the present work, we compared the distribution of SORB induced during S-phase by etoposide or X-rays in the short euchromatic and long heterochromatic arms of the CHO9 X chromosome. The S-phase stage (early, mid or late) at which CHO9 cells were exposed to etoposide or X-rays was marked by incorporation of BrdU during treatments and later determined by immunolabeling of metaphase chromosomes with an anti-BrdU FITC-coupled antibody. The majority of treated cells were in late S-phase during treatment either with etoposide or X-rays. SORB induced by etoposide mapped preferentially to Xq but random localization was observed for SORB produced by X-rays. Possible explanations for the uneven distribution of etoposide-induced breakpoints along Xq are discussed.


Subject(s)
CHO Cells/drug effects , CHO Cells/radiation effects , Chromosome Breakage , Enzyme Inhibitors/toxicity , Etoposide/toxicity , Topoisomerase II Inhibitors , X Chromosome/drug effects , X Chromosome/radiation effects , Animals , CHO Cells/ultrastructure , Chromatids/drug effects , Chromatids/radiation effects , Chromatids/ultrastructure , Chromosome Aberrations , Chromosome Mapping , Cricetinae , Cricetulus , DNA/drug effects , DNA/radiation effects , DNA Damage , Female , S Phase/drug effects , S Phase/radiation effects , X Chromosome/genetics , X Chromosome/ultrastructure
19.
Cytogenet Genome Res ; 104(1-4): 211-5, 2004.
Article in English | MEDLINE | ID: mdl-15162040

ABSTRACT

Radiation-induced chromosome damage can be measured in interphase using the Premature Chromosome Condensation (PCC) technique. With the introduction of a new PCC technique using the potent phosphatase inhibitor calyculin-A, chromosomes can be condensed within five minutes, and it is now possible to examine the early damage induced by radiation. Using this method, it has been shown that high-LET radiation induces a higher frequency of chromatid breaks and a much higher frequency of isochromatid breaks than low-LET radiation. The kinetics of chromatid break rejoining consists of two exponential components representing a rapid and a slow time constant, which appears to be similar for low- and high-LET radiations. However, after high-LET radiation exposures, the rejoining process for isochromatid breaks influences the repair kinetics of chromatid-type breaks, and this plays an important role in the assessment of chromatid break rejoining in the G2 phase of the cell cycle.


Subject(s)
Chromosome Breakage , Chromosomes, Human/radiation effects , DNA Repair/physiology , Animals , CHO Cells/radiation effects , CHO Cells/ultrastructure , Cells, Cultured/radiation effects , Cells, Cultured/ultrastructure , Chromatids/radiation effects , Chromatids/ultrastructure , Chromosome Aberrations , Chromosomes, Human/ultrastructure , Cricetinae , Cricetulus , Dose-Response Relationship, Radiation , Enzyme Inhibitors/pharmacology , Fibroblasts/radiation effects , Fibroblasts/ultrastructure , G2 Phase/radiation effects , Gamma Rays/adverse effects , Humans , Kinetics , Linear Energy Transfer , Lymphocytes/radiation effects , Lymphocytes/ultrastructure , Marine Toxins , Neutrons/adverse effects , Oxazoles/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors
20.
Cytogenet Genome Res ; 104(1-4): 304-9, 2004.
Article in English | MEDLINE | ID: mdl-15162056

ABSTRACT

The DNA lesions responsible for the formation of sister chromatid exchanges (SCEs) have been the object of research for a long time. SCEs can be visualized by growing cells for either two rounds of replication in the presence of 5-bromo-2'-deoxyuridine (BrdU) or for one round with BrdU and the next without. If BrdU is added after cells were treated with a DNA-damaging agent, the effect on SCEs can only be analyzed in the second post-treatment mitosis. If one wishes to analyze the first post-treatment mitosis, cells unifilarily labeled with BrdU must be treated. Due to the highly reactive bromine atom, BrdU interacts with such agents like ionizing and UV radiation enhancing the frequency of SCEs. However, its precise role in this process was difficult to assess for a long time, because no alternative technique existed that allowed differential staining of chromatids. We have recently developed a method to differentially label sister chromatids with biotin-16-2'-deoxyuridine-5'-triphosphate (biotin-dUTP) circumventing the disadvantage of BrdU. This technique was applied to study the SCEs induced by ionizing and UV radiation as well as by mitomycin C, DNaseI and AluI. This article is a review of the results and conclusions of our previous studies.


Subject(s)
Biotin/analogs & derivatives , Chromosomes/radiation effects , Sister Chromatid Exchange , Animals , Biotin/pharmacology , Bromodeoxyuridine/pharmacology , Bromodeoxyuridine/toxicity , CHO Cells/drug effects , CHO Cells/radiation effects , CHO Cells/ultrastructure , Chromosome Inversion , Chromosomes/drug effects , Cricetinae , Cricetulus , Cross-Linking Reagents/pharmacology , Cross-Linking Reagents/toxicity , DNA Damage , DNA Replication , Deoxyuracil Nucleotides/pharmacology , Free Radicals , G1 Phase , Humans , Mitomycin/pharmacology , Mitomycin/toxicity , Radiation-Sensitizing Agents/pharmacology , Sister Chromatid Exchange/drug effects , Sister Chromatid Exchange/physiology , Sister Chromatid Exchange/radiation effects , Staining and Labeling , Ultraviolet Rays/adverse effects , X-Rays/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...