Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 18215, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31796776

ABSTRACT

Modifications to the rates of water flowing from the surface to groundwater (groundwater recharge) due to climate variability are the most difficult to assess because of the lack of direct long-term observations. Here, we analyze the chloride salt distribution below the surface soil on a plateau near Los Angeles to reconstruct the amount of recharge that occurred in the last five centuries. Over this time interval, periods of major high and low recharge with different duration follow each other and this cyclicity is consistent with long-term atmospheric forcing patterns, such as the Pacific Decadal Oscillation. This study determines the range and the natural variability of recharge to groundwater, which sustains local freshwater flow system, and helps forecast future availability of groundwater resource in southern California, where water scarcity is critical to both local and global populations.

2.
Article in English | MEDLINE | ID: mdl-23863866

ABSTRACT

Circadian rhythms can be entrained by a light-dark (LD) cycle and can also be reset pharmacologically, for example, by the CK1δ/ε inhibitor PF-670462. Here, we determine how these two independent signals affect circadian timekeeping from the molecular to the behavioral level. By developing a systems pharmacology model, we predict and experimentally validate that chronic CK1δ/ε inhibition during the earlier hours of a LD cycle can produce a constant stable delay of rhythm. However, chronic dosing later during the day, or in the presence of longer light intervals, is not predicted to yield an entrained rhythm. We also propose a simple method based on phase response curves (PRCs) that predicts the effects of a LD cycle and chronic dosing of a circadian drug. This work indicates that dosing timing and environmental signals must be carefully considered for accurate pharmacological manipulation of circadian phase.CPT: Pharmacometrics & Systems Pharmacology (2013) 2, e57; doi:10.1038/psp.2013.34; published online 17 July 2013.

3.
J Neurosci ; 20(1): 43-50, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10627579

ABSTRACT

We have studied the mechanisms of auditory hair cell death after insults in vitro and in vivo. We show DNA fragmentation of hair cell nuclei after ototoxic drug and intense noise trauma. By using phospho-specific c-Jun-N-terminal kinase (JNK) and c-Jun antibodies in immunohistochemistry, we show that the JNK pathway, associated with stress, injury, and apoptosis, is activated in hair cells after trauma. CEP-1347, a derivative of the indolocarbazole K252a, is a small molecule that has been shown to attenuate neurodegeneration by blocking the activation of JNK (). Subcutaneously delivered CEP-1347 attenuated noise-induced hearing loss. The protective effect was demonstrated by functional tests, which showed less hearing threshold shift in CEP-1347-treated than in nontreated guinea pigs, and by morphometric methods showing less hair cell death in CEP-1347-treated cochleas. In organotypic cochlear cultures, CEP-1347 prevented neomycin-induced hair cell death. In addition to hair cells, CEP-1347 promoted survival of dissociated cochlear neurons. These results suggest that therapeutic intervention in the JNK signaling cascade, possibly by using CEP-1347, may offer opportunities to treat inner ear injuries.


Subject(s)
Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Hair Cells, Auditory/cytology , Hearing Loss, Noise-Induced/drug therapy , Indoles/pharmacology , Neurons, Afferent/cytology , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Aminoglycosides/toxicity , Animals , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/physiology , Hearing Loss, Noise-Induced/chemically induced , Hearing Loss, Noise-Induced/pathology , Neomycin/toxicity , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Noise/adverse effects , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
4.
Mol Neurobiol ; 19(1): 43-59, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10321971

ABSTRACT

The identification of novel factors that promote neuronal survival could have profound effects on developing new therapeutics for neurodegenerative disorders. Glial cell line-derived neurotrophic factor (GDNF) is a novel protein purified and cloned based on its marked ability to promote dopaminergic neuronal function. GDNF, now known to be the first identified member of a family of factors, signals through the previously known receptor tyrosine kinase, Ret. Unlike most ligands for receptor tyrosine kinases, GDNF does not bind and activate Ret directly, but requires the presence of GPI-linked coreceptors. There are several coreceptors with differing affinities for the GDNF family members. The profile of coreceptors in a cell may determine which factor preferentially activates Ret. In vivo differences in localization of the GDNF family members, its coreceptors and Ret suggest this ligand/receptor interaction has extensive and multiple functions in the CNS as well as in peripheral tissues. GDNF promotes survival of several neuronal populations both in vitro and in vivo. Dopaminergic neuronal survival and function are preserved by GDNF in vivo when challenged by the toxins MPTP and 6-hydroxydopamine. Furthermore, GDNF improves the symptoms of pharmacologically induced Parkinson's disease in monkeys. Several motor neuron populations isolated in vitro are also rescued by GDNF. In vivo, GDNF protects these neurons from programmed cell death associated with development and death induced by neuronal transection. These experiments suggest that GDNF may provide significant therapeutic opportunities in several neurodegenerative disorders.


Subject(s)
Drosophila Proteins , Nerve Growth Factors , Nerve Tissue Proteins/pharmacology , Neurodegenerative Diseases/drug therapy , Neurons/drug effects , Animals , Cell Survival/drug effects , Dopamine/physiology , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/isolation & purification , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/therapeutic use , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/metabolism , Neurons/cytology , Neurons/enzymology , Neurons/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction
5.
J Comp Neurol ; 398(3): 373-92, 1998 Aug 31.
Article in English | MEDLINE | ID: mdl-9714150

ABSTRACT

Cell survival, death, and stress signals are transduced from the cell surface to the cytoplasm and nucleus via a cascade of phosphorylation events involving the mitogen-activated protein kinase (MAPK) family. We compared the distribution of p42 mitogen-activated protein kinase (p42MAPK) and its activator MAPK or ERK kinase (MEK1; involved in transduction of growth and differentiation signals), with c-Jun N-terminal kinase (JNK1) and its activator MEK4 (involved in transduction of stress and death signals) in the adult rat central nervous system. All four kinases were present in the cytoplasm, dendrites, and axons of neurons. The presence of p42MAPK and JNK1 in dendrites and axons, as well as in cell bodies, suggests a role for these kinases in phosphorylation and regulation of cytoplasmic targets. A high degree of correspondence was found between the regional distribution of MEK1 and p42MAPK. Immunostaining for MEK1 and p42MAPK was intense in olfactory structures, neocortex, hippocampus, striatum, midline, and interlaminar thalamic nuclei, hypothalamus, brainstem, Purkinje cells, and spinal cord. In addition to neurons, p42MAPK was also present in oligodendrocytes. Whereas MEK4 was ubiquitously distributed, JNK1 was more selective. Immunostaining for MEK4 and JNK1 was intense in the olfactory bulb, lower cortical layers, the cholinergic basal forebrain, most nuclei of the thalamus, medial habenula, and cranial motor nuclei. The distribution of MEK1 and p42MAPK proteins only partially overlapped with that of MEK4 and JNK1. This suggests that the growth/differentiation and death/stress pathways affected by these kinases may not necessarily act to counterbalance each other in response to extracellular stimuli. The differential distribution of these kinases may control the specificity of neuronal function to extracellular signals.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Central Nervous System/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Blotting, Western , Central Nervous System/enzymology , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Signal Transduction/physiology
6.
J Neurochem ; 70(4): 1764-7, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9523596

ABSTRACT

p38MAPK has been implicated in the regulation of proinflammatory cytokines and apoptosis in vitro. To understand its role in neurodegeneration, we determined the time course and localization of the dually phosphorylated active form of p38MAPK in hippocampus after global forebrain ischemia. Phosphorylated p38MAPK and mitogen-activated protein kinase-activated protein 2 activity increased over 4 days after ischemia. Phosphorylated p38MAPK immunoreactivity was observed in microglia in regions adjacent to, but not in, the dying CA1 neurons. In contrast, neither c-Jun N-terminal kinase 1 nor p42/p44MAPK activity was altered after ischemia. These results provide the first evidence for localization of activated p38MAPK in the CNS and support a role for p38MAPK in the microglial response to stress.


Subject(s)
Brain Ischemia/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Microglia/enzymology , Mitogen-Activated Protein Kinases , Animals , Blotting, Western , Brain Ischemia/physiopathology , Enzyme Activation/physiology , Gerbillinae , Hippocampus/enzymology , Immunohistochemistry , Male , Phosphorylation , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases
7.
J Neurochem ; 70(2): 558-71, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9453550

ABSTRACT

In vitro studies indicate that p42/p44MAPK phosphorylate both nuclear and cytoplasmic proteins. However, the functional targets of p42/p44MAPK activation in vivo remain unclear. To address this question, we localized activated p42/p44MAPK in hippocampus and cortex and determined their signaling effects after electroconvulsive shock treatment (ECT) in rats. Phosphorylated p42/p44MAPK content increased in the cytoplasm of hippocampal neurons in response to ECT. Consistent with this cytoplasmic localization, inhibition of ECT-induced p42/p44MAPK activation by the extracellular signal-regulated kinase kinase inhibitor PD098059 blocked phosphorylation of the cytoplasmic protein microtubule-associated protein 2c (MAP2c), but failed to inhibit the induction of the nuclear protein c-Fos in response to ECT. In contrast to hippocampal neurons, cortical neurons exhibited an increase in amount of phosphorylated p42/p44MAPK in both the nucleus and cytoplasm after ECT. Accordingly, PD098059 blocked the induction of Fos-like immunoreactivity in the nuclei of cortical neurons as well as MAP2c phosphorylation in the cytoplasm. Our data indicate that both nuclear and cytoplasmic substrates can be activated by p42/p44MAPK in vivo. However, the functional targets of p42/p44MAPK signaling depend on the precise location of p42/p44MAPK within different subcellular compartments of brain regions. These results indicate unique functional pathways of p42/p44MAPK-mediated signal transduction within different brain regions in vivo.


Subject(s)
Brain/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Signal Transduction/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Nucleus/physiology , Cerebral Cortex/physiology , Cytoplasm/metabolism , Electroshock , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Hippocampus/physiology , MAP Kinase Kinase 1 , Microtubule-Associated Proteins/metabolism , Mitogen-Activated Protein Kinase 3 , Organ Specificity , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Rats, Sprague-Dawley
8.
J Neurosci ; 18(1): 104-11, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9412490

ABSTRACT

Neurons undergoing apoptosis can be rescued by trophic factors that simultaneously increase the activity of extracellular signal-regulated kinase (ERK) and decrease c-Jun N-terminal kinase (JNK) and p38. We identified a molecule, CEP-1347 (KT7515), that rescues motoneurons undergoing apoptosis and investigated its effect on ERK1 and JNK1 activity. Cultured rat embryonic motoneurons, in the absence of trophic factor, began to die 24-48 hr after plating. During the first 24 hr ERK1 activity was unchanged, whereas JNK1 activity increased fourfold. CEP-1347 completely rescued motoneurons for at least 72 hr with an EC50 of 20 +/- 2 nM. CEP-1347 did not alter ERK1 activity but rapidly inhibited JNK1 activation. The IC50 of CEP-1347 for JNK1 activation was the same as the EC50 for motoneuron survival. Inhibition of JNK1 activation by CEP-1347 was not selective to motoneurons. CEP-1347 also inhibited JNK1 activity in Cos7 cells under conditions of ultraviolet irradiation, osmotic shock, and inhibition of glycosylation. Inhibition by CEP-1347 of the JNK1 signaling pathway appeared to be selective, because CEP-1347 did not inhibit p38-regulated mitogen-activated protein kinase-activated protein kinase-2 (MAPKAP2) activity in Cos7 cells subjected to osmotic shock. The direct molecular target of CEP-1347 was not JNK1, because CEP-1347 did not inhibit JNK1 activity in Cos7 cells cotransfected with MEKK1 and JNK1 cDNA constructs. This is the first demonstration of a small organic molecule that promotes motoneuron survival and that simultaneously inhibits the JNK1 signaling cascade.


Subject(s)
Apoptosis/drug effects , Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Motor Neurons/cytology , Protein Kinase Inhibitors , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carbazoles/chemical synthesis , Cell Survival/drug effects , Choline O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Fetus/cytology , Gene Expression , Imidazoles/pharmacology , Indole Alkaloids , MAP Kinase Kinase 4 , Motor Neurons/enzymology , Motor Neurons/ultrastructure , Neurites/physiology , Protein Kinases/genetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , p38 Mitogen-Activated Protein Kinases
9.
J Neurochem ; 68(5): 1820-9, 1997 May.
Article in English | MEDLINE | ID: mdl-9109506

ABSTRACT

A receptor-type protein tyrosine phosphatase, PTP NE-6, was identified from rat olfactory epithelial cDNA and cloned from a rat brain cDNA library. PTP NE-6 mRNA is abundant in brain and expressed at lower levels in olfactory tissue and adrenal gland. In situ hybridization demonstrates that PTP NE-6 mRNA is expressed throughout the brain, with highest levels in the medial habenula and at intermediate levels in layer IV of cortex, medial geniculate nucleus, inferior colliculus, hypothalamus, and thalamus. The predicted amino acid sequence demonstrates that PTP NE-6 contains a single catalytic domain that diverges from the consensus protein tyrosine phosphatase catalytic domain by expressing an aspartate instead of the conserved alanine residue in the catalytic site. Recombinantly expressed PTP NE-6 does not exhibit detectable phosphatase activity. Upon mutation of the aspartate to the consensus alanine, phosphatase activity toward p-nitrophenyl phosphate is observable with a k(cat) value of 3.7 s(-1) and a Km of 980 microM. These data demonstrate that the inactivity of native PTP NE-6 toward p-nitrophenyl phosphate is due to the divergent aspartate in the catalytic site and not to variant amino acids within the phosphatase domain.


Subject(s)
Brain/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Catalysis , Cloning, Molecular , Molecular Sequence Data , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 8 , Tissue Distribution
10.
Plant J ; 7(6): 981-8, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7599654

ABSTRACT

The first evidence for tyrosine phosphatase signalling pathways in plants is presented by characterizing a putative protein tyrosine phosphatase gene from the unicellular green alga Chlamydomonas eugametos. This cDNA, referred to as VH-PTP13, contains an open reading frame specifying a protein with a molecular weight of 30.3 kDa, that has significant homology with a distinct group of dual-specificity phosphatases. The highest homology is found with CL-100, a human stress-response gene that regulates MAPkinase activity. The purified VH-PTP13 protein expressed in E. coli had phosphatase activity and inactivated MAPkinases from alfalfa and tobacco. Nondividing C. eugametos gametes did not express the VH-PTP13 gene whereas synchronously dividing vegetative cells only expressed VH-PTP13 in the early G1-phase of the cycle, implying a function there. When vegetative cells were subjected to oxidative stress, expression of the VH-PTP13 gene was strongly induced, analogous to the human CL-100 gene. Its potential role in plant signalling pathways is discussed.


Subject(s)
Chlamydomonas/genetics , Gene Expression Regulation , Oxidative Stress , Protein Tyrosine Phosphatases/genetics , Signal Transduction , Tyrosine/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle , Chlamydomonas/enzymology , Cloning, Molecular , DNA, Complementary , Dual Specificity Phosphatase 3 , Escherichia coli , Medicago sativa/enzymology , Medicago sativa/genetics , Molecular Sequence Data , Phylogeny , Protein Kinase Inhibitors , Protein Tyrosine Phosphatases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
11.
Neuron ; 11(2): 387-400, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8352946

ABSTRACT

Tyrosine phosphorylation plays a central role in the control of neuronal cell development and function. Yet, few neuronal protein tyrosine phosphatases (PTPs) have been identified. We examined rat olfactory neuroepithelium for expression of novel PTPs potentially important in neuronal development and regeneration. Using the polymerase chain reaction with degenerate DNA oligomers directed to the conserved tyrosine phosphatase domain, we identified 6 novel tyrosine phosphatases. One of these, PTP NE-3, is a receptor-type PTP expressed selectively in both rat brain and olfactory neuroepithelium. In the olfactory neuroepithelium, PTP NE-3 expression is restricted to neurons and describes a novel pattern of expression with a high level in the immature neurons and a lower level in mature olfactory sensory neurons.


Subject(s)
Olfactory Pathways/growth & development , Olfactory Pathways/metabolism , Protein Tyrosine Phosphatases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Epithelium/growth & development , Epithelium/metabolism , Histocytochemistry , In Situ Hybridization , Male , Molecular Sequence Data , Protein Tyrosine Phosphatases/genetics , Rats , Rats, Sprague-Dawley , Tissue Distribution
13.
Mol Endocrinol ; 6(10): 1725-33, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1333055

ABSTRACT

The thyroid follicular cell requires elevated levels of cAMP for normal growth and optimal expression of the differentiated phenotype. The recent discovery of cAMP-regulated enhancer binding (CREB) proteins prompted us to analyze the possible role of these transcription factors in controlling thyroid cell growth and differentiated phenotype using the FRTL5 thyroid cell line as a model system. FRTL5 cells were stably transfected with an expression vector containing either the gene for wild type CREB (WTCREB) or a dominant negative mutant form of CREB, termed KCREB, which dimerizes with and inactivates endogenous CREB. Transfected clones were found to express the transfected KCREB and WTCREB mRNAs at higher levels than the endogenous CREB mRNA. Transient expression of a somatostatin-chloramphenicol acetyltransferase fusion gene in these clones demonstrated a 60% reduction of cAMP-regulated enhancer-dependent transcriptional activity in the KCREB transfected clones and wild type levels of activity in the WTCREB transfected clones. Parameters of growth (DNA synthesis and growth rate) and differentiation (iodide uptake and thyroglobulin mRNA levels) were then analyzed in the transfected clones. Transfection of WTCREB had no effect on any of the parameters examined in comparison to untransfected cells, presumably because CREB is already constitutively expressed at maximal levels in normal FRTL5 cells. However, cells expressing KCREB showed an 18-40% reduction in TSH-stimulated thymidine incorporation, a 31% increase in the length of the cell cycle, and a 4-fold reduction in TSH-stimulated iodide uptake in comparison with wild type cells or cells tranfected with wild type CREB.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cell Differentiation/physiology , Cell Division/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Thyroid Gland/physiology , 3T3 Cells , Animals , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , DNA/biosynthesis , Gene Expression , Kinetics , Mice , Mice, Inbred BALB C , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Recombinant Proteins/metabolism , Thymidine/metabolism , Thyroid Gland/cytology , Thyrotropin/pharmacology , Transfection
14.
Mol Endocrinol ; 6(4): 647-55, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1350057

ABSTRACT

The transactivation of genes through the cAMP-regulated enhancer (CRE) is proposed to occur by the binding and phosphorylation of the transcription factor CREB (CRE-binding protein). Originally believed to be a single protein, more than 10 different CREB proteins have been cloned. The contributions of each of these factors to gene regulation have yet to be determined unambiguously. We have isolated a CREB cDNA that contains a mutation of a single amino acid in the DNA-binding domain. In gel shift assays, this mutant, designated KCREB, is unable to bind to the somatostatin (SS) CRE. In addition, KCREB acts as a dominant repressor of the wild-type factor, blocking the ability of wild-type CREB to bind to the CRE when present as a KCREB:CREB heterodimer. The KCREB mutant also acts as a dominant repressor in vivo, completely blocking the ability of wild-type CREB to mediate induction by protein kinase-A of a SS CRE reporter gene in F9 teratocarcinoma cells. We have used this mutant to analyze the participation of CREB in the induction of the SS promoter in CA-77 cells, a medullary thyroid carcinoma cell line that produces high levels of SS. Although KCREB can block a portion of the cAMP induction of the SS promoter in CA-77 cells, approximately 45% of the induction remains insensitive to the mutant. These data support the paradigm that CREB is involved in the cAMP induction of SS in vivo. Furthermore, the inability of KCREB to completely block cAMP-mediated SS expression in CA-77 cells suggests that additional factors may contribute to the cAMP regulation of CRE function.


Subject(s)
Cyclic AMP/physiology , DNA-Binding Proteins/metabolism , Promoter Regions, Genetic , Somatostatin/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Cyclic AMP Response Element-Binding Protein , DNA-Binding Proteins/antagonists & inhibitors , Gene Library , Humans , Hypothalamus/physiology , Molecular Sequence Data , Phosphorylation , Plasmids , Polymerase Chain Reaction/methods , Transcription, Genetic , Transfection
15.
J Biol Chem ; 266(28): 18431-4, 1991 Oct 05.
Article in English | MEDLINE | ID: mdl-1655749

ABSTRACT

Many promoters respond transcriptionally to elevated levels of cAMP through the cAMP-responsive enhancer (CRE). Several proteins have been characterized which bind to the CRE and presumably modulate CRE-dependent transcription. Of these CRE-binding proteins, only CREB has been shown to be activated by cAMP-dependent protein kinase A (PKA), and as such, CREB represents the only basis for our understanding of cAMP-regulated transcriptional activity. In this report, we describe the complete cDNA sequence of another CRE-binding protein, ATF-1. This protein contains a consensus phosphorylation site for PKA and shares extensive homology with CREB in the region surrounding and carboxyl-terminal to the PKA site. ATF-1 does not contain sequences homologous to the glutamine-rich amino-terminal domain found in CREB, however. ATF-1, like CREB, is expressed in a wide variety of cell types, and ATF-1 is capable of dimerizing with CREB. Both ATF-1 homodimers and ATF-1/CREB heterodimers bind to the CRE but not to the related phorbol ester response element. ATF-1 is as active as CREB in its ability to mediate the transcriptional effects of PKA, and, because ATF-1 has a smaller effect on basal expression, it is actually more responsive than CREB to cAMP. These findings indicate that CREB is not unique in its ability to mediate cAMP-dependent transcriptional regulation.


Subject(s)
Blood Proteins/metabolism , Cyclic AMP/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Protein Kinases/metabolism , Transcription Factors/metabolism , Activating Transcription Factors , Amino Acid Sequence , Animals , Blood Proteins/genetics , Cell Line , Cyclic AMP Response Element-Binding Protein , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Regulatory Sequences, Nucleic Acid , Sequence Homology, Nucleic Acid , Transcription Factors/genetics , Transcription, Genetic
16.
Mol Neurobiol ; 4(3-4): 197-210, 1990.
Article in English | MEDLINE | ID: mdl-1966918

ABSTRACT

The ability of many genes to be induced by cAMP is dependent on the presence of enhancers located in the regions of DNA upstream of the start sites to the genes. The two best characterized enhancers are the CRE (5'-TGACGTCA-3') and the AP-2 site (5'-CCCCAGGC-3'). The activity of the CRE is modulated by sequences adjacent to the consensus sequence as well as by promoter context and cell type. The complex control of the CRE is reflected in the large number of cloned CRE binding proteins that arise both from unique genes and from splice variants. These factors are leucine zipper proteins that must dimerize before binding to DNA. Although all of the factors isolated can form active homodimers, many are also able to form heterodimers. The amino termini of these proteins contain consensus phosphorylation sites through which these factors trans-activate their cognate promoters. The diversity of the trans-acting factors and their cis-acting sequences reflects the precise control that cells require in the modulation of gene expression by cAMP.


Subject(s)
Cyclic AMP/physiology , Gene Expression Regulation , Transcription Factors/physiology , Animals , Base Sequence , Consensus Sequence , DNA-Binding Proteins/physiology , Enhancer Elements, Genetic , Humans , Leucine Zippers/physiology , Molecular Sequence Data , Promoter Regions, Genetic , RNA Splicing , Rats , Regulatory Sequences, Nucleic Acid , Signal Transduction , Transcription Factor AP-2 , Transcription, Genetic
17.
J Neurochem ; 52(5): 1537-44, 1989 May.
Article in English | MEDLINE | ID: mdl-2540274

ABSTRACT

The enzymatic basis for ganglioside regulation during differentiation of NG108-15 mouse neuroblastoma x rat glioma hybrid cells was studied. This cell line contains four gangliosides that lie along the same biosynthetic pathway: GM3, GM2, GM1, and GD1a. Chemically induced neuronal differentiation of NG108-15 cells led to an 80% drop in the steady-state level of their major ganglioside, GM3, a sixfold increase in the level of a minor ganglioside, GM2 (which became the predominant ganglioside of differentiated cells); and relatively little change in the levels of GM1 and GD1a, which lie further along the same biosynthetic pathway. The enzymatic basis for this selective change in ganglioside expression was investigated by measuring the activity of two glycosyltransferases involved in ganglioside biosynthesis. UDP-N-acetylgalactosamine: GM3 N-acetylgalactosaminyltransferase (GM2-synthetase) activity increased fivefold during butyrate-induced differentiation, whereas UDP-galactose: GM2 galactosyltransferase (GM1-synthetase) activity decreased to 10% of its control level. Coordinate regulation of these two glycosyltransferases appears to be primarily responsible for the selective increase of GM2 expression during NG108-15 differentiation.


Subject(s)
Galactosyltransferases/metabolism , Glioma/enzymology , Hybrid Cells/enzymology , N-Acetylgalactosaminyltransferases , Neuroblastoma/enzymology , Alprostadil/pharmacology , Animals , Bucladesine/pharmacology , Cell Differentiation , G(M1) Ganglioside/analysis , G(M1) Ganglioside/metabolism , G(M2) Ganglioside/analysis , G(M2) Ganglioside/metabolism , G(M3) Ganglioside/analysis , G(M3) Ganglioside/metabolism , Ganglioside Galactosyltransferase , Gangliosides/analysis , Gangliosides/metabolism , Kinetics , Mass Spectrometry , Mice , Rats , Theophylline/pharmacology , Tumor Cells, Cultured , Polypeptide N-acetylgalactosaminyltransferase
18.
J Biol Chem ; 263(4): 2055-63, 1988 Feb 05.
Article in English | MEDLINE | ID: mdl-3339002

ABSTRACT

Ganglioside expression and tetanus toxin binding were studied in the rat pheochromocytoma cell line PC12. Seven ganglioside species were readily detected in extracts of PC12 cells; two were identified as tri- and tetrasialogangliosides, which are common brain constituents but unusual components of neuronal cell lines. Carbohydrate composition, acid and enzyme hydrolyses, and mass spectral analysis revealed that the major species is GT 1b, a predominant mammalian brain ganglioside previously reported to support high affinity tetanus toxin binding (Rogers, T. B., and Snyder, S. H. (1981) J. Biol. Chem. 256, 2402-2407). Direct binding of 125I-tetanus toxin to PC12 gangliosides on TLC plates revealed selective binding to the tri- and tetrasialogangliosides. Radioiodinated toxin also bound with high affinity to intact PC12 cells or their isolated membranes. The binding affinity (Kd = 1.25 nM), density of receptors (Bmax = 238 pmol/mg of membrane protein), and dependence on pH, ionic strength, and temperature were similar to those previously reported for toxin binding to rat brain synaptic membranes. Differentiation of PC12 cells caused an increase in expression of the tri- and tetrasialogangliosides and a closely matched increase in tetanus toxin binding to cell membranes. These data provide evidence that complex gangliosides may act as tetanus toxin receptors, and demonstrate the utility of the PC12 cell line for studies of tetanus toxicity and complex ganglioside expression.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Gangliosides/biosynthesis , Membrane Proteins , Pheochromocytoma/metabolism , Tetanus Toxin/metabolism , Animals , Carbohydrates/analysis , Mass Spectrometry , Neuraminidase/metabolism , Rats , Receptors, Cholinergic/metabolism , Tumor Cells, Cultured/metabolism
19.
J Neurosci ; 6(5): 1443-51, 1986 May.
Article in English | MEDLINE | ID: mdl-3711989

ABSTRACT

Tetanus toxin is known to bind neuronal tissue selectively. To study the interactions of this potent neurotoxin in an intact cell system, the binding of 125I-tetanus toxin was characterized in a neuroblastoma retina hybrid cell line, N18-RE-105. The binding of 125I-tetanus toxin to membranes prepared from N18-RE-105 cells showed many similarities to the interactions of 125I-toxin with rat synaptic membranes. The binding was decreased with increasing temperature, ionic strength, and pH. 125I-Toxin bound to membranes with high affinity: KD = 0.62 +/- 0.05 nM; Bmax = 196 +/- 45 pmol/mg protein. Quantitative thin-layer chromatography and acid-degradation analysis revealed that N18-RE-105 cells contained polysialogangliosides GD1a and GT1b in high concentrations. An assay was developed to quantitate surface-bound and internalized 125I-tetanus toxin by exploiting the observation that surface-bound 125I-toxin is susceptible to pronase digestion. When cells were incubated with 125I-tetanus toxin at 0 degree C, all of the bound 125I-toxin could be degraded with pronase. In contrast, when the incubations were performed at 37 degrees C, within 10 min about 50% of the total cell-associated 125I-toxin was pronase-resistant. Temperature pulse experiments demonstrated that 125I-tetanus toxin that was bound to cells at 0 degree C rapidly disappeared from the surface when the cells were warmed to 37 degrees C, as revealed by the appearance of pronase-resistant radioactivity. This internalization was sensitive to metabolic inhibitors.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Hybrid Cells/metabolism , Membrane Proteins , Neuroblastoma/metabolism , Tetanus Toxin/metabolism , Animals , Binding, Competitive , Cell Line , Chromatography, Thin Layer , Drug Resistance , Gangliosides/analysis , Hybrid Cells/analysis , Mice , Neuroblastoma/analysis , Neuroblastoma/pathology , Oligomycins/pharmacology , Pronase/pharmacology , Rats , Receptors, Cholinergic/metabolism , Rotenone/pharmacology , Temperature , Tetanus Toxin/antagonists & inhibitors
20.
Anal Biochem ; 152(1): 154-9, 1986 Jan.
Article in English | MEDLINE | ID: mdl-3082240

ABSTRACT

A rapid method for determination of ganglioside glycosyltransferase activity has been developed employing ion-exchange chromatography. Using 13-day chick brain as a source of uridine diphospho-N-acetyl-D-galactosamine: ganglioside GM3 N-acetylgalactosaminyltransferase (ganglioside GM2 synthetase), we were able to accurately measure transfer of N-[3H]acetylgalactosamine (GalNAc) from UDP-[3H]GalNAc to GM3 by application of the reaction mixture to small columns of DEAE-Sepharose and elution of radiolabeled GM2 reaction product with 10 mM potassium acetate in methanol. This method proved to be as reliable and sensitive as previously published assays but requires less time and fewer manipulations.


Subject(s)
Chromatography, Ion Exchange , Galactosyltransferases/analysis , N-Acetylgalactosaminyltransferases , Animals , Brain/enzymology , Chick Embryo , Microsomes/enzymology , Polypeptide N-acetylgalactosaminyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL
...