Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-18557399

ABSTRACT

The paper discusses characteristics of a new modeling-based technique for determining dielectric properties of materials. Complex permittivity is found with an optimization algorithm designed to match complex S-parameters obtained from measurements and from 3D FDTD simulation. The method is developed on a two-port (waveguide-type) fixture and deals with complex reflection and transmission characteristics at the frequency of interest. A computational part is constructed as an inverse-RBF-network-based procedure that reconstructs dielectric constant and the loss factor of the sample from the FDTD modeling data sets and the measured reflection and transmission coefficients. As such, it is applicable to samples and cavities of arbitrary configurations provided that the geometry of the experimental setup is adequately represented by the FDTD model. The practical implementation of the method considered in this paper is a section of a WR975 waveguide containing a sample of a liquid in a cylindrical cutout of a rectangular Teflon cup. The method is run in two stages and employs two databases--first, built for a sparse grid on the complex permittivity plane, in order to locate a domain with an anticipated solution and, second, made as a denser grid covering the determined domain, for finding an exact location of the complex permittivity point. Numerical tests demonstrate that the computational part of the method is highly accurate even when the modeling data is represented by relatively small data sets. When working with reflection and transmission coefficients measured in an actual experimental fixture and reconstructing a low dielectric constant and the loss factor the technique may be less accurate. It is shown that the employed neural network is capable of finding complex permittivity of the sample when experimental data on the reflection and transmission coefficients are numerically dispersive (noise-contaminated). A special modeling test is proposed for validating the results; it confirms that the values of complex permittivity for several liquids (including salt water acetone and three types of alcohol) at 915 MHz are reconstructed with satisfactory accuracy.


Subject(s)
Microwaves , Models, Theoretical , Neural Networks, Computer , Computer Simulation , Electric Conductivity , Finite Element Analysis
2.
J Neurosci ; 21(14): 4996-5006, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11438575

ABSTRACT

Basic fibroblast growth factor (FGF2) has many roles in neuronal development and maintenance including effects on mitogenesis, survival, fate determination, differentiation, and migration. Using a conditionally immortalized rat hippocampal cell line, H19-7, and primary hippocampal cultures, we now demonstrate that FGF2 treatment differentially regulates members of the tumor necrosis factor (TNF) superfamily of death domain receptors and their ligands. H19-7 cells transferred from serum to defined (N2) medium undergo apoptosis by a Fas-dependent mechanism similar to primary neurons. In contrast, H19-7 cells treated with FGF undergo apoptosis by a Fas-independent mechanism. FGF suppresses the Fas death pathway but also induces apoptosis by activation of a TNFalpha death pathway in both H19-7 and hippocampal progenitor cells. Expression of the TNF receptor 1 (TNFR1) or TNFR2 in H19-7 cells is sufficient to sensitize the cells to TNFalpha, similar to the effects of FGF. Because TNFalpha can be either proapoptotic or antiapoptotic, these results provide an explanation for the divergent trophic effects of FGF2 treatment and the observation that multiple trophic inputs are required for the survival of specific neurons.


Subject(s)
Apoptosis , Fibroblast Growth Factor 2/pharmacology , Intracellular Signaling Peptides and Proteins , Neurons/drug effects , Neurons/metabolism , Signal Transduction/drug effects , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Caspase Inhibitors , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Fas Ligand Protein , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neurons/cytology , RNA, Messenger/metabolism , Rats , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Transfection , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/metabolism
3.
Mol Cell Biol ; 20(15): 5392-403, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10891480

ABSTRACT

Although mitogenic and differentiating factors often activate a number of common signaling pathways, the mechanisms leading to their distinct cellular outcomes have not been elucidated. In a previous report, we demonstrated that mitogen-activated protein (MAP) kinase (ERK) activation by the neurogenic agents fibroblast growth factor (FGF) and nerve growth factor is dependent on protein kinase Cdelta (PKCdelta), whereas MAP kinase activation in response to the mitogen epidermal growth factor (EGF) is independent of PKCdelta in rat hippocampal (H19-7) and pheochromocytoma (PC12) cells. We now show that EGF activates MAP kinase through a PKCzeta-dependent pathway involving phosphatidylinositol 3-kinase and PDK1 in H19-7 cells. PKCzeta, like PKCdelta, acts upstream of MEK, and PKCzeta can potentiate Raf-1 activation by EGF. Inhibition of PKCzeta also blocks EGF-induced DNA synthesis as monitored by bromodeoxyuridine incorporation in H19-7 cells. Finally, in embryonic rat brain hippocampal cell cultures, inhibitors of PKCzeta or PKCdelta suppress MAP kinase activation by EGF or FGF, respectively, indicating that these factors activate distinct signaling pathways in primary as well as immortalized neural cells. Taken together, these results implicate different PKC isoforms as determinants of growth factor signaling specificity within the same cell. Furthermore, these data provide a mechanism whereby different growth factors can differentially activate a common signaling intermediate and thereby generate biological diversity.


Subject(s)
Epidermal Growth Factor/metabolism , Isoenzymes/metabolism , Neurons/metabolism , Protein Kinase C/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Cells, Cultured , Epidermal Growth Factor/pharmacology , Hippocampus/cytology , Hippocampus/embryology , Isoenzymes/genetics , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Oligonucleotides, Antisense/pharmacology , PC12 Cells/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/genetics , Protein Kinase C-delta , Protein Kinase C-theta , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Rats , Rats, Sprague-Dawley
4.
Cancer Res ; 60(8): 2263-72, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10786694

ABSTRACT

The type I insulin-like growth factor receptor (IGF-IR) is known to send two seemingly contradictory signals inducing either cell proliferation or cell differentiation, depending on cell type and/or conditions. H19-7 cells are rat hippocampal neuronal cells immortalized by a temperature-sensitive SV40 large T antigen that grow at 34 degrees C in epidermal growth factor or serum but differentiate at 39 degrees C when induced by basic fibroblast growth factor. At 39 degrees C, expression of the human IGF-IR in H19-7 cells induces an insulin-like growth factor (IGF) I-dependent differentiation. We have investigated the domains of the IGF-IR required for differentiation of H19-7 cells. The tyrosine 950 residue and serines 1280-1283 in the COOH terminus of the receptor are required for IGF-I-induced differentiation at 39 degrees C, although they are dispensable for IGF-I-mediated growth at 34 degrees C. Both domains have to be mutated to inactivate the differentiating function. The inability of these mutant receptors to induce differentiation correlates with mitogen-activated protein kinase activation. In contrast, inhibitors of phosphatidylinositol 3'-kinase have no effect on IGF-I-mediated differentiation of H19-7 cells, although they do inhibit the mitogenic response.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Cell Differentiation , MAP Kinase Signaling System , Neurons/cytology , Receptor, IGF Type 1/metabolism , Amino Acid Substitution/genetics , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/metabolism , Hippocampus , Humans , Insulin Receptor Substrate Proteins , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Mutation/genetics , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphoproteins/metabolism , Phosphorylation/drug effects , Proteins/metabolism , Rats , Receptor, IGF Type 1/chemistry , Receptor, IGF Type 1/genetics , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Temperature
5.
J Neurooncol ; 45(1): 19-26, 1999.
Article in English | MEDLINE | ID: mdl-10728906

ABSTRACT

Dividing cells and non-dividing cells are distinct in their cell cycle kinetics, and react differently when facing cytotoxic stimuli. A protein synthesis inhibitor, cycloheximide (CHX), has recently been found to protect neuronal cells from oxidative stress. We investigated whether CHX exerts differential effects on dividing and non-dividing cells in the brain under cytotoxic stimuli. Mitotic C6 rat glioma cells and postmitotic neuronal cells were studied with a cytotoxic regimen combining gamma-irradiation (RT) and 1,3-bis,2-chloroethyl-1-nitrosourea (BCNU). Cells were exposed to BCNU (1 g/ml) for 15 h before gamma-irradiation and incubated with CHX (1 g/ml) from 30 min before and until 5 h after irradiation. Clonogenic assay was used to assess cytotoxic effects on C6 glioma cells. LDH assay was used for the viability of H19-7 postmitotic neuronal cells. A 2.27-3.75 fold enhancement of cytotoxicity was noticed with the addition of CHX to BCNU and 2-10 Gy of radiation. Our data demonstrated that CHX enhanced cytotoxicity of RT plus BCNU, while no additional toxicity was incurred to the postmitotic neuronal cells when CHX was added. We further studied whether the inhibition of DNA repair, assayed by single-cell DNA electrophoresis (comet assay), is a contributing factor for the enhanced cytotoxicity on C6 glioma cells. Interestingly, the initial DNA damage after RT plus BCNU was equivalent; whereas DNA repair was significantly less at 5 h after radiation in CHX-treated C6 glioma cells. Protecting non-dividing neuronal cells to avoid excessive functional deficit is an integral part of a successful brain tumor treatment regimen. Taking advantage of the differential effect of CHX on glioma and neuronal cells may improve tumor control without excessive neural toxicity.


Subject(s)
Cycloheximide/therapeutic use , Glioma/drug therapy , Glioma/radiotherapy , Neurons/drug effects , Protein Synthesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Carmustine/therapeutic use , Cell Death/drug effects , Cell Line, Transformed , DNA Repair/drug effects , Drug Therapy, Combination , Glioma/pathology , Rats , Tumor Cells, Cultured
6.
Mol Cell Biol ; 18(4): 2143-52, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9528786

ABSTRACT

Phosphatidylinositol (PI) 3-kinase has been suggested to mediate cell survival. Consistent with this possibility, apoptosis of conditionally (simian virus 40 Tts) immortalized rat hippocampal H19-7 neuronal cells was increased in response to wortmannin, an inhibitor of PI 3-kinase. Downstream effectors of PI 3-kinase include Rac1, protein kinase C, and the serine-threonine kinase Akt (protein kinase B). Here, we show that activation of Akt is one mechanism by which PI 3-kinase can mediate survival of H19-7 cells during serum deprivation or differentiation. While ectopic expression of wild-type Akt (c-Akt) does not significantly enhance survival in H19-7 cells, expression of activated forms of Akt (v-Akt or myristoylated Akt) results in enhanced survival which can be comparable to that conferred by Bcl-2. Conversely, expression of a dominant-negative mutant of Akt accelerates cell death upon serum deprivation or differentiation. Finally, the results indicate that Akt can transduce a survival signal for differentiating neuronal cells through a mechanism that is independent of induction of Bcl-2 or Bcl-XL or inhibition of Jun kinase activity.


Subject(s)
Apoptosis , Neurons/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Androstadienes/pharmacology , Animals , Cell Differentiation , Cell Line , Culture Media, Serum-Free , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genes, Dominant , Mutation , Neurons/enzymology , Oncogene Protein v-akt , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats , Retroviridae Proteins, Oncogenic/metabolism , Wortmannin , bcl-X Protein
7.
Regul Pept ; 69(1): 7-14, 1997 Mar 12.
Article in English | MEDLINE | ID: mdl-9163577

ABSTRACT

Employing reverse transcription-polymerase chain reaction and clonal cell lines derived by retroviral transduction of the temperature sensitive simian virus 40 large T-antigen into dispersed rat embryonic hippocampal cells, we detected the ancestral gene-insulin II mRNA in three progenitor subcloned cell lines. These cell lines upon differentiation are known to express markers indicative of commitment to either neuronal (H19-7; NF + , GFAP -), glial (H19-5; GFAP +, NF -), or bipotential (H583-5, NF +, GFAP + ) lineages. No duplicated, i.e., insulin I gene expression, was observed in any of the three cell lines. Induction of differentiation was associated with the persistence of insulin II mRNA and in the cells expressing a neuronal phenotype (H19-7; NF +, GFAP -) a relative doubling in insulin II mRNA level was present (P < 0.05). Minimal cellular insulin immunoreactivity was detected only in a subpopulation of cells with a differentiated neuronal phenotype. Radioimmunoassayable insulin peptide in the H19-7 cellular conditioned medium revealed a 5-fold increase in the differentiated state. In contrast, peripheral sympathetic PC-12 neuronal cells both in the undifferentiated and nerve growth factor-driven differentiated states, failed to express both insulin I and insulin II genes. We conclude that insulin II is expressed by cultured rat hippocampal clonal cell lines, and not by the peripheral sympathetic PC-12 neuronal cell line.


Subject(s)
Gene Expression , Hippocampus/metabolism , Insulin/genetics , Neurons/metabolism , Animals , Blotting, Southern , Cell Differentiation , Hippocampus/cytology , PC12 Cells , Polymerase Chain Reaction , RNA, Messenger/metabolism , Radioimmunoassay , Rats
9.
J Neurochem ; 67(5): 1908-20, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8863495

ABSTRACT

To characterize the nature of programmed cell death (PCD) induced in neuronal cells during development, three regulators of apoptosis were investigated: one, the bcl-2-related genes, modulate cell survival, and the other two, the interleukin-1 beta converting enzyme (ICE)-related enzymes and the tumor suppressor protein p53, have been implicated as mediators of apoptosis. These regulators were studied in H19-7 cells, an SV40 Tts-immortalized rat hippocampal neuronal cell line that can be differentiated with basic fibroblast growth factor at the nonpermissive temperature, resulting in a rapid attrition of cells by apoptosis. PCD occurred by two mechanisms in H19-7 cells: The first was initiated by removal of serum from undifferentiated cells, and the second was a consequence of neuronal differentiation. In differentiated H19-7 cells, the survival time was increased by both human bcl-2 and bcl-xL, and this could be reversed by bcl-xs. Addition of a peptide inhibitor of the ICE enzyme family to H19-7 cells resulted in a transient protection against differentiation-associated apoptosis, whereas no further protection was observed in the BCL-2- or BCL-XL-expressing cells. Shifting the differentiated cells to 33 degrees C to inactivate p53 did not significantly affect the apoptotic process, indicating that apoptosis induced by neuronal differentiation is not dependent on the continued presence of p53. By contrast, in undifferentiated cells, cell loss induced by transfer to serum-free media occurred more rapidly on inactivation of large T, consistent with p53 involvement. This medium-induced decrease in cell survival could not be rescued by the ICE inhibitor but was partially rescued by BCL-2 or BCL-XL. Furthermore, studies involving expression of BCL-2 and BCL-XL alone or together revealed differences in the survival dependent on the cellular environment. These results suggest that apoptosis of neuronal cells occurs by at least two processes: one in undifferentiated cells initiated by removal of serum and one linked to differentiation. The data implicate the ICE enzyme family but not p53 in apoptosis induced by differentiation and demonstrate that either BCL-2 or BCL-XL can prolong the survival of differentiated neuronal cells.


Subject(s)
Apoptosis , Hippocampus/physiology , Neurons/cytology , Neurons/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antigens, Viral, Tumor/biosynthesis , Biomarkers , Caspase 1 , Cell Differentiation , Cell Line, Transformed , Culture Media, Serum-Free , Cysteine Endopeptidases/biosynthesis , Embryo, Mammalian , Gene Expression , Hippocampus/cytology , Humans , Kinetics , Protease Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats , Simian virus 40/genetics , Transfection , Tumor Suppressor Protein p53/biosynthesis
10.
Eur J Neurosci ; 8(10): 2042-55, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8921294

ABSTRACT

A clonal cell line, AS583-8.E4.22, from the embryonic day 15 rat basal forebrain was established using retrovirus-mediated transduction of a temperature-sensitive mutant of the simian virus 40 (SV40) large tumour antigen. The cell line expresses cytoskeletal and neurotransmitter features indicative of neuronal commitment. In response to agents that increase intracellular cAMP, including forskolin and catecholamines, the cell line exhibits rapid process outgrowth and growth cone formation that does not require new gene expression or protein synthesis. The neurite outgrowth induced by catecholamines is mediated by beta 2-adrenergic receptors and is characterized by a rapid, reversible redistribution of filamentous actin. Neurons from primary cultures of embryonic day 15 basal forebrain were also found to respond to beta-adrenergic receptor agonists by enhancing growth cone formation. These results suggest that catecholamines provide cues that induce cytoskeletal rearrangements leading to neuronal process outgrowth and growth cone formation in the developing basal forebrain and possibly other neuronal progenitor cell populations. The neuronal basal forebrain cell line provides an ideal model to study the signalling mechanisms underlying the catecholamine-induced process outgrowth.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Antigens, Polyomavirus Transforming/genetics , Catecholamines/pharmacology , Neurons/drug effects , Prosencephalon/drug effects , Animals , Cell Line , Cellular Senescence , Cyclic AMP/metabolism , Cytoskeletal Proteins/biosynthesis , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Gestational Age , Mutation , Neurons/metabolism , Neurons/ultrastructure , Prosencephalon/cytology , Prosencephalon/embryology , Rats , Rats, Inbred ACI , Transduction, Genetic
11.
Brain Res ; 726(1-2): 141-52, 1996 Jul 08.
Article in English | MEDLINE | ID: mdl-8836554

ABSTRACT

A clonal cell line of rat embryonic hippocampal origin (H19-7) has been examined for the expression of glucocorticoid receptors (GR) and mineralocorticoid receptors (MR). H19-7 cells grown at 33 degrees C continue to divide, however when grown at 39 degrees C in reduced levels of serum the cells undergo morphological differentiation and express neuronal properties. Immunocytochemistry demonstrated that H19-7 cells express both MR and GR when grown at either 33 degrees C or 39 degrees C. GR mRNA is readily detected in H19-7 cells by RNase protection assay. MR mRNA levels in H19-7 cells are too low to detect by RNase protection, but can be detected by RT-PCR. RT-PCR also demonstrated that H19-7 cells express more GR mRNA than primary hippocampal neurons. Since previous studies have shown that the level of MR mRNA is higher than that of GR mRNA in hippocampal neurons, these studies suggest that H19-7 cells represent hippocampal neurons immortalized at an early stage when the MR system is not yet fully differentiated.


Subject(s)
Hippocampus/chemistry , Neurons/chemistry , Receptors, Glucocorticoid/analysis , Receptors, Mineralocorticoid/analysis , Animals , Cell Line , Cell Survival/physiology , Hippocampus/cytology , Hippocampus/embryology , Polymerase Chain Reaction/methods , Rats , Rats, Sprague-Dawley , Ribonucleases , Transcription, Genetic
12.
Mol Cell Biol ; 16(4): 1458-70, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8657119

ABSTRACT

To elucidate signal transduction pathways leading to neuronal differentiation, we have investigated a conditionally immortalized cell line from rat hippocampal neurons (H19-7) that express a temperature sensitive simian virus 40 large T antigen. Treatment of H19-7 cells with the differentiating agent basic fibroblast growth factor at 39 degrees C, the nonpermissive temperature for T function, resulted in the activation of c-Raf-1, MEK, and mitogen-activated protein (MAP) kinases (ERK1 and -2). To evaluate the role of Raf-1 in neuronal cell differentiation, we stably transfected H19-7 cells with v-raf or an oncogenic human Raf-1-estrogen receptor fusion gene (deltaRaf-1:ER). deltaRaf-1:ER transfectants in the presence of estradiol for 1 to 2 days expressed a differentiation phenotype only at the nonpermissive temperature. However, extended exposure of the deltaRaf-1:ER transfectants to estradiol or stable expression of the v-raf construct yielded cells that extended processes at the permissive as well as the nonpermissive temperature, suggesting that cells expressing the large T antigen are capable of responding to the Raf differentiation signal. deltaRaf-1:ER, MEK, and MAP kinase activities in the deltaRaf-1:ER cells were elevated constitutively for up to 36 h of estradiol treatment at the permissive temperature. At the nonpermissive temperature, MEK and ERKs were activated to a significantly lesser extent, suggesting that prolonged MAP kinase activation may not be sufficient for differentiation. To test this possibility, H19-7 cells were transfected or microinjected with constitutively activated MEK. The results indicate that prolonged activation of MEK or MAP kinases (ERK1 and -2) is not sufficient for differentiation of H19-7 neuronal cells and raise the possibility that an alternative signaling pathway is required for differentiation of H19-7 cells by Raf.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/genetics , Hippocampus/cytology , MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases , Neurons/cytology , Protein Serine-Threonine Kinases/genetics , Retroviridae Proteins, Oncogenic/genetics , Animals , Antigens, Polyomavirus Transforming/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Estradiol/pharmacology , Fibroblast Growth Factor 2/pharmacology , Hippocampus/metabolism , Humans , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Oncogene Proteins v-raf , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Rats , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Signal Transduction , Temperature , Transfection
13.
Brain Res ; 656(2): 396-404, 1994 Sep 12.
Article in English | MEDLINE | ID: mdl-7820600

ABSTRACT

To determine whether postmitotic neurons can be immortalized by oncogenic transduction, we used two approaches involving conditional expression of a temperature-sensitive SV40 large T antigen (Tts). Initially, Tts was introduced into E17 rat embryonal hippocampal cells that were then cultured at the non-permissive temperature to enrich for postmitotic pyramidal neurons, and subsequently cloned at the permissive temperature. One clonal line (HMR10-3) expressed neuron-specific proteins upon differentiation, was capable of generating action potentials, and formed synapses with primary rat neurons in co-culture. Replating of these postmitotic cells at the permissive temperature resulted in reversible loss of neurofilament expression. Conditionally immortalized cell lines were also generated from the brain of an adult mouse carrying an inducible Tts transgene. These lines proliferated in a T antigen-dependent manner and expressed neuron-specific proteins upon differentiation at the non-permissive temperature. These results suggest that postmitotic neurons can be induced to enter the cell cycle without losing their commitment to a neuronal lineage.


Subject(s)
Mitosis/physiology , Neurons/physiology , Pyramidal Cells/cytology , Animals , Antigens, Polyomavirus Transforming/biosynthesis , Cell Line, Transformed , DNA Replication/physiology , Electrophysiology , Glial Fibrillary Acidic Protein/metabolism , Mice , Mice, Transgenic , Microscopy, Electron , Pyramidal Cells/ultrastructure , Rats , Transfection , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
14.
J Neurochem ; 61(4): 1376-87, 1993 Oct.
Article in English | MEDLINE | ID: mdl-7690847

ABSTRACT

Epidermal growth factor (EGF) functions in a bimodal capacity in the nervous system, acting as a mitogen in neuronal stem cells and a neurotrophic factor in differentiated adult neurons. Thus, it is likely that EGF signal transduction, as well as receptor expression, differs among various cell types and possibly in the same cell type at different stages of development. We used hippocampal neuronal cell lines capable of terminal differentiation to investigate changes in EGF receptor expression, DNA synthesis, and stimulation of mitogen-activated protein (MAP) kinase by EGF before and after differentiation. H19-7, the line that was most representative of hippocampal neurons, was mitogenically responsive to EGF only before differentiation and increased in EGF binding after differentiation. MAP kinase was stimulated by EGF in both undifferentiated and differentiated cells, as well as in primary hippocampal cultures treated with either EGF or glutamate. These results indicate that the activation of MAP kinase by EGF is an early signaling event in both mitotic and postmitotic neuronal cells. Furthermore, these studies demonstrate the usefulness of hippocampal cell lines as a homogeneous neuronal system for studies of EGF signaling or other receptor signaling mechanisms in the brain.


Subject(s)
Epidermal Growth Factor/pharmacology , Hippocampus/metabolism , Mitogens/pharmacology , Neurons/metabolism , Protein Kinases/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases , Cell Differentiation , Cell Line , DNA/biosynthesis , Enzyme Activation , ErbB Receptors/metabolism , Glycogen Synthase Kinase 3 , Hippocampus/cytology , Hippocampus/drug effects , Immunohistochemistry/methods , Neurons/drug effects , Staining and Labeling
15.
Proc Natl Acad Sci U S A ; 89(10): 4373-7, 1992 May 15.
Article in English | MEDLINE | ID: mdl-1316607

ABSTRACT

Clonal cell lines of rat embryonic hippocampal origin have been developed by using retroviral transduction of temperature-sensitive simian virus 40 large tumor antigens. The cell lines undergo morphological differentiation at the nonpermissive temperature and in response to differentiating agents. Immunocytochemical analysis indicates that various lines are derived from progenitors of neuronal, glial, and bipotential lineages. Selected neuronal lines differentiate in response to diffusible factors released by primary glia, and one line of glial lineage supports the maturation of primary neurons in culture. Selected cell lines exhibit different patterns of neurotrophin gene expression that change after differentiation. In some lines, the relative levels of neurotrophin 3 and brain-derived neurotrophic factor message expression may reflect the developmental or regional differential expression seen for these genes in the hippocampus in situ. These hippocampal cell lines, which express markers indicative of commitment to neuronal or glial lineages, are valuable for studies of development and plasticity in these lineages, as well as for studies of the regulation of neural trophic interactions.


Subject(s)
Cell Transformation, Viral , Hippocampus/physiology , Nerve Tissue Proteins/genetics , Neuroglia/physiology , Neurons/physiology , Simian virus 40/genetics , 3T3 Cells , Animals , Antigens, Polyomavirus Transforming/genetics , Brain-Derived Neurotrophic Factor , Cell Line, Transformed , Cells, Cultured , Clone Cells , DNA Replication , Embryo, Mammalian , Gene Expression , Mice , Nerve Growth Factors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Thymidine/metabolism
16.
Proc Natl Acad Sci U S A ; 88(16): 7318-22, 1991 Aug 15.
Article in English | MEDLINE | ID: mdl-1871139

ABSTRACT

Developmental regulation of gamma-globin gene expression probably occurs through developmental-stage-specific trans-acting factors able to promote the interaction of enhancer elements located in the far upstream locus control region with regulatory elements in the gamma gene promoters and 3' A gamma enhancer located in close proximity to the genes. We have detected a nuclear protein in K562 and baboon fetal bone marrow nuclear extracts capable of binding to A+T-rich sequences in the locus control region, gamma gene promoter, and 3' A gamma enhancer. SDS/polyacrylamide gel analysis of the purified K562 binding activity revealed a single protein of 87 kDa. A K562 cDNA clone was isolated encoding a beta-galactosidase fusion protein with a DNA binding specificity identical to that of the K562/fetal bone marrow nuclear protein. The cDNA clone encodes a homeodomain homologous to the Drosophila antennapedia protein.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Globins/genetics , Homeodomain Proteins , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Transcription Factors , Animals , Antennapedia Homeodomain Protein , Base Sequence , Binding Sites , Cell Line , Cell Nucleus/physiology , Gene Library , HeLa Cells/physiology , Humans , Molecular Sequence Data , Oligonucleotide Probes , Papio , Protein Binding , Restriction Mapping
17.
Exp Hematol ; 17(2): 73-6, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2463934

ABSTRACT

Baboon species differ markedly in the proportions of fetal hemoglobin (HbF) they produce in response to hemopoietic stress. Bone marrow erythroid progenitor cells from untreated and stressed baboons of three species were cultured to determine if differences in number, size, or other characteristics of the colonies and bursts could be correlated with the HbF response. BFU-E from adult Papio cynocephalus produced high proportions of macroscopic, well-hemo-globinized bursts, whereas those from P. anubis and P. papio produced only small, moderately hemoglobinized bursts. The species-specific differences in burst characteristics did not correlate with the in vivo HbF response to stress and they were not altered by variations in culture conditions. However, bone marrow BFU-E from P. anubis and P. cynocephalus fetuses produced similar bursts, suggesting developmental regulation of progenitor cell growth patterns. The proportions of macroscopic bursts were reduced in P. cynocephalus animals undergoing hemopoietic stress, suggesting that culture is a more severe erythropoietic stress for P. anubis and P. papio BFU-E than for P. cynocephalus BFU-E.


Subject(s)
Erythroblasts/cytology , Erythropoiesis , Hematopoietic Stem Cells/cytology , Papio/blood , Animals , Azacitidine/administration & dosage , Bloodletting , Bone Marrow/metabolism , Bone Marrow Cells , Colony-Forming Units Assay , Erythroblasts/drug effects , Erythroblasts/metabolism , Erythrocyte Count , Erythropoiesis/drug effects , Erythropoietin/pharmacology , Fetal Hemoglobin/biosynthesis , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Species Specificity
18.
Genetics ; 107(4): 563-76, 1984 Aug.
Article in English | MEDLINE | ID: mdl-6589201

ABSTRACT

The transmission of two non-Mendelian drug resistance markers has been studied in crosses of Chlamydomonas reinhardtii involving diploids and aneuploids with different mating type genotypes. Under normal laboratory conditions for gametogenesis, mating and zygote maturation, the transmission pattern of the non-Mendelian markers sr-u-1 (resistance to streptomycin) and spr-u-1-27-3 (resistance to spectinomycin) is primarily determined by the mating type genotypes of the parental cells. Our results confirm and expand an earlier observation suggesting that an apparent codominant function of the female (mt+) allele in regulating chloroplast gene transmission in meiosis appears to be distinct and separate from its recessive function in regulating mating behavior. The chloroplast DNA complement (as indexed by the number of extranuclear DNA-containing bodies) may exert a secondary effect on the transmission of these markers. Within a mating type group (mt+/mt- or mt-/mt-) a cell line with more chloroplast DNA tended to transmit its non-Mendelian markers more frequently than a cell line with less chloroplast DNA.


Subject(s)
Chlamydomonas/genetics , Extrachromosomal Inheritance , Aneuploidy , Chlamydomonas/drug effects , Chloroplasts , Drug Resistance, Microbial , Genetic Markers
19.
Proc Natl Acad Sci U S A ; 81(10): 3153-7, 1984 May.
Article in English | MEDLINE | ID: mdl-6328502

ABSTRACT

We have used the eukaryotic-prokaryotic shuttle vector pSV2Neo to demonstrate that cultured mammalian somatic cells have the enzymatic machinery to mediate homologous recombination and that the frequency of this recombination can be enhanced by pretreatment of the input DNA. Two nonoverlapping deletion mutants of pSV2Neo were constructed, each affecting the bacterial aminoglycoside 3'-phosphorylase gene (the neo gene), which confers resistance to aminoglycoside antibiotics on bacteria and resistance to the antibiotic G418 on mammalian cells. Mammalian cells transfected with either deletion plasmid alone yield no G418 -resistant colonies. Cells cotransfected with both deletion plasmids yield G418 -resistant colonies with high frequency. We show that these resistant colonies result from recombination involving homologous crossing-over or gene conversion between the deletion plasmids by rescuing from the resistant cells both types of reciprocal recombinant, full-length plasmids, and doubly deleted plasmids. Cutting one of the input plasmids to generate a double-stranded gap in the neo gene considerably enhances the frequency of homologous recombination within the gene. This suggests that targeting exogenous DNA to specific sites in mammalian chromosomes could be facilitated by suitable pretreatment of the DNA.


Subject(s)
DNA, Neoplasm/genetics , DNA, Recombinant/metabolism , Plasmids , Animals , Cell Line , Chlorocebus aethiops , Chromosome Deletion , DNA Restriction Enzymes , Humans , Hybrid Cells/physiology , Kidney , Mutation , Transfection , Urinary Bladder Neoplasms
20.
Somatic Cell Genet ; 9(6): 771-8, 1983 Nov.
Article in English | MEDLINE | ID: mdl-6581543

ABSTRACT

Karyotypes of recessive mutants at the autosomal adenine phosphoribosyltransferase (Aprt) locus in a clone of the near-diploid mouse CAK cell line have been analyzed. The Aprt located on chromosome 8. One copy of chromosome 8 was morphologically abnormal in the parental clone (CAK-B3-Toyr13) from which Aprt- mutants were isolated. Among 22 mutants, there were ten in which one copy of chromosome 8 had been lost. Four of these were monosomic, and in the others duplication of the remaining homolog had occurred. These findings indicate that newly induced recessive mutations in cultured mammalian cells can be expressed as the result of loss of one chromosome carrying a wild-type allele with or without duplication of the homolog carrying the mutant allele. Loss and duplication would not be detected in cell lines lacking morphologically marked chromosomes.


Subject(s)
Adenine Phosphoribosyltransferase/genetics , Chromosome Aberrations , Gene Expression Regulation , Genes, Recessive , Pentosyltransferases/genetics , Aneuploidy , Animals , Cell Line , Chromosome Mapping , Genetic Markers , Karyotyping , Mice , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...