Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Intern Med ; 290(5): 993-1009, 2021 11.
Article in English | MEDLINE | ID: mdl-34156133

ABSTRACT

Brain oscillations underlie the function of our brains, dictating how we both think and react to the world around us. The synchronous activity of neurons generates these rhythms, which allow different parts of the brain to communicate and orchestrate responses to internal and external stimuli. Perturbations of cognitive rhythms and the underlying oscillator neurons that synchronize different parts of the brain contribute to the pathophysiology of diseases including Alzheimer's disease, (AD), Parkinson's disease (PD), epilepsy and other diseases of rhythm that have been studied extensively by Gyorgy Buzsaki. In this review, we discuss how neurologists manipulate brain oscillations with neuromodulation to treat diseases and how this can be leveraged to improve cognition and pathology underlying AD. While multiple modalities of neuromodulation are currently clinically indicated for some disorders, nothing is yet approved for improving memory in AD. Recent investigations into novel methods of neuromodulation show potential for improving cognition in memory disorders. Here, we demonstrate that neuronal stimulation using audiovisual sensory stimulation that generated 40-HZ gamma waves reduced AD-specific pathology and improved performance in behavioural tests in mouse models of AD, making this new mode of neuromodulation a promising new avenue for developing a new therapeutic intervention for the treatment of dementia.


Subject(s)
Alzheimer Disease , Brain Waves , Acoustic Stimulation , Alzheimer Disease/therapy , Animals , Brain , Cognition , Mice , Neurons , Photic Stimulation
2.
Mol Psychiatry ; 20(2): 162-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25403839

ABSTRACT

Psychiatric disorders have clear heritable risk. Several large-scale genome-wide association studies have revealed a strong association between susceptibility for psychiatric disorders, including bipolar disease, schizophrenia and major depression, and a haplotype located in an intronic region of the L-type voltage-gated calcium channel (VGCC) subunit gene CACNA1C (peak associated SNP rs1006737), making it one of the most replicable and consistent associations in psychiatric genetics. In the current study, we used induced human neurons to reveal a functional phenotype associated with this psychiatric risk variant. We generated induced human neurons, or iN cells, from more than 20 individuals harboring homozygous risk genotypes, heterozygous or homozygous non-risk genotypes at the rs1006737 locus. Using these iNs, we performed electrophysiology and quantitative PCR experiments that demonstrated increased L-type VGCC current density as well as increased mRNA expression of CACNA1C in iNs homozygous for the risk genotype, compared with non-risk genotypes. These studies demonstrate that the risk genotype at rs1006737 is associated with significant functional alterations in human iNs, and may direct future efforts at developing novel therapeutics for the treatment of psychiatric disease.


Subject(s)
Calcium Channels, L-Type/metabolism , Membrane Potentials/physiology , Mental Disorders/genetics , Mental Disorders/pathology , Neurons/physiology , Adult , Aged , Astrocytes/drug effects , Calcium/metabolism , Calcium Channel Agonists/pharmacology , Calcium Channels, L-Type/genetics , Cell Differentiation/drug effects , Coculture Techniques , Female , Fibroblasts/drug effects , Humans , Intercellular Signaling Peptides and Proteins/therapeutic use , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Middle Aged , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Transduction, Genetic , Young Adult
3.
Mol Psychiatry ; 20(3): 388-97, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24821222

ABSTRACT

Ankyrin-G is a scaffolding protein required for the formation of the axon initial segment in neurons. Recent genome-wide association studies and whole-exome sequencing have identified ANK3, the gene coding for ankyrin-G, to be a risk gene for multiple neuropsychiatric disorders, such as bipolar disorder, schizophrenia and autism spectrum disorder. Here, we describe a novel role for ankyrin-G in neural progenitor proliferation in the developing cortex. We found that ankyrin-G regulates canonical Wnt signaling by altering the subcellular localization and availability of ß-catenin in proliferating cells. Ankyrin-G loss-of-function increases ß-catenin levels in the nucleus, thereby promoting neural progenitor proliferation. Importantly, abnormalities in proliferation can be rescued by reducing Wnt pathway signaling. Taken together, these results suggest that ankyrin-G is required for proper brain development.


Subject(s)
Actins/metabolism , Neurogenesis/genetics , Neurons/physiology , Subcellular Fractions/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , Actins/genetics , Animals , Ankyrins/deficiency , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Transgenic , Pregnancy
4.
Mol Psychiatry ; 18(8): 898-908, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23587879

ABSTRACT

Disrupted-In-Schizophrenia 1 (DISC1), a risk factor for major mental illnesses, has been studied extensively in the context of neurodevelopment. However, the role of DISC1 in neuronal signaling, particularly in conjunction with intracellular cascades that occur in response to dopamine, a neurotransmitter implicated in numerous psychiatric disorders, remains elusive. Previous data suggest that DISC1 interacts with numerous proteins that impact neuronal function, including activating transcription factor 4 (ATF4). In this study, we identify a novel DISC1 and ATF4 binding region in the genomic locus of phosphodiesterase 4D (PDE4D), a gene implicated in psychiatric disorders. We found that the loss of function of either DISC1 or ATF4 increases PDE4D9 transcription, and that the association of DISC1 with the PDE4D9 locus requires ATF4. We also show that PDE4D9 is increased by D1-type dopamine receptor dopaminergic stimulation. We demonstrate that the mechanism for this increase is due to DISC1 dissociation from the PDE4D locus in mouse brain. We further characterize the interaction of DISC1 with ATF4 to show that it is regulated via protein kinase A-mediated phosphorylation of DISC1 serine-58. Our results suggest that the release of DISC1-mediated transcriptional repression of PDE4D9 acts as feedback inhibition to regulate dopaminergic signaling. Furthermore, as DISC1 loss-of-function leads to a specific increase in PDE4D9, PDE4D9 itself may represent an attractive target for therapeutic approaches in psychiatric disorders.


Subject(s)
Activating Transcription Factor 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Animals , Dextroamphetamine/pharmacology , HeLa Cells , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Mice , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Primary Cell Culture
5.
Mol Psychiatry ; 17(5): 537-48, 2012 May.
Article in English | MEDLINE | ID: mdl-21468034

ABSTRACT

Synchronous recruitment of fast-spiking (FS) parvalbumin (PV) interneurons generates gamma oscillations, rhythms that emerge during performance of cognitive tasks. Administration of N-methyl-D-aspartate (NMDA) receptor antagonists alters gamma rhythms, and can induce cognitive as well as psychosis-like symptoms in humans. The disruption of NMDA receptor (NMDAR) signaling specifically in FS PV interneurons is therefore hypothesized to give rise to neural network dysfunction that could underlie these symptoms. To address the connection between NMDAR activity, FS PV interneurons, gamma oscillations and behavior, we generated mice lacking NMDAR neurotransmission only in PV cells (PV-Cre/NR1f/f mice). Here, we show that mutant mice exhibit enhanced baseline cortical gamma rhythms, impaired gamma rhythm induction after optogenetic drive of PV interneurons and reduced sensitivity to the effects of NMDAR antagonists on gamma oscillations and stereotypies. Mutant mice show largely normal behaviors except for selective cognitive impairments, including deficits in habituation, working memory and associative learning. Our results provide evidence for the critical role of NMDAR in PV interneurons for expression of normal gamma rhythms and specific cognitive behaviors.


Subject(s)
Association Learning/physiology , Brain Waves/physiology , GABAergic Neurons/physiology , Interneurons/physiology , Memory, Short-Term/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Association Learning/drug effects , Brain Waves/drug effects , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , GABA Antagonists/pharmacology , GABAergic Neurons/metabolism , Interneurons/drug effects , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory, Short-Term/drug effects , Mice , Mice, Transgenic , Parvalbumins/metabolism , Photic Stimulation/methods , Picrotoxin/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Sensory Gating/drug effects , Sensory Gating/physiology , Stereotyped Behavior/drug effects , Stereotyped Behavior/physiology
6.
Eur J Clin Microbiol Infect Dis ; 32(3): 345-52, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23001510

ABSTRACT

A retrospective study was conducted at a Taiwanese medical center to characterize bloodstream infections caused by IMP-8 metallo-ß-lactamase (MBL)-producing Enterobacteriaceae isolates and to assess the need for laboratory detection of IMP producers. We analyzed 37 patients infected with IMP-8 producers (two Escherichia coli, nine Klebsiella pneumoniae, 25 Enterobacter cloacae, and one Citrobacter freundii) and 107 patients infected with non-IMP-8 producers (eight E. coli, 26 K. pneumoniae, 70 E. cloacae, and three C. freundii) that were interpreted as carbapenem-nonsusceptible based on the updated Clinical and Laboratory Standards Institute (CLSI) 2010 guidelines. Only 18 (48.6 %) of the IMP-8 producers were regarded as potential carbapenemase producers based on the CLSI 2012 guidelines. The production of extended-spectrum ß-lactamases (ESBLs) was more common in the MBL group (73.0 %) than in the non-MBL group (41.1 %). There were no significant differences in carbapenem susceptibilities, clinical characteristics, carbapenem use for empirical and definitive treatment, and mortality rates between the two groups. Eighteen IMP-8 producers could be deemed as resistant to all carbapenems [minimum inhibitory concentration (MIC) of any carbapenem ≥2 µg/mL]; patients with these isolates had a lower, but non-significant, 28-day mortality rate (27.8 %) than patients infected with non-MBL producers having similar carbapenem MICs (39.0 %) (p = 0.41). A multivariate analysis revealed severity of acute illness as the single independent variable associated with both 7-day and 28-day mortality rates (p < 0.01) for infections caused by Enterobacteriaceae with decreased carbapenem susceptibilities. Our findings suggest that the clinical detection of IMP-producing Enterobacteriaceae is not required even when the "old" CLSI criteria are used.


Subject(s)
Bacteremia/epidemiology , Enterobacteriaceae Infections/epidemiology , Enterobacteriaceae/enzymology , beta-Lactamases/metabolism , Adult , Aged , Aged, 80 and over , Anti-Bacterial Agents/pharmacology , Bacteremia/microbiology , Bacteremia/pathology , Carbapenems/pharmacology , Enterobacteriaceae/drug effects , Enterobacteriaceae/isolation & purification , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Female , Humans , Male , Microbial Sensitivity Tests , Middle Aged , Retrospective Studies , Taiwan/epidemiology
7.
Eur J Clin Microbiol Infect Dis ; 31(8): 1941-50, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22249422

ABSTRACT

This study was conducted in order to characterize carbapenem-nonsusceptible Klebsiella pneumoniae isolates and to evaluate the impacts of recently lowered interpretative breakpoints for carbapenems for Enterobacteriaceae. Among 152 K. pneumoniae bloodstream isolates suspected as AmpC or extended-spectrum ß-lactamase (ESBL) producers, 58 (38.2%) isolates were currently interpreted as nonsusceptible to ertapenem, imipenem, or meropenem, and 42 (72.4%) of them were categorized as carbapenem-susceptible by the previous criteria. The high revision rate was associated with the predominance (79.3%) of DHA-1 among the carbapenem-nonsusceptible isolates due to both polyclonal and clonal spread. ESBLs were common (~57%) in both ertapenem-susceptible and -nonsusceptible isolates; however, 84.8% of the carbapenem-nonsusceptible isolates were also AmpC producers. The IMP-8 metallo-ß-lactamase was detected in three isolates. Polyacrylamide gel electrophoresis suggested decreased OmpK35 expression in all but one ertapenem-nonsusceptible isolate, and genetic disruptions of ompK35 and ompK36 were detected in 30 and six ertapenem-nonsusceptible isolates, respectively. A comparison between patients infected by AmpC- or ESBL-producing ertapenem-susceptible (n=62) isolates and those with isolates revised as ertapenem-nonsusceptible (n=41) revealed more cases of malignancies (36.6% versus 14.5%; p=0.01) and higher Charlson score (p=0.033) among the patients with ertapenem-nonsusceptible isolates; however, the acquisition of an isolate revised as carbapenem-nonsusceptible was not identified as an independent mortality risk factor.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteremia/microbiology , Carbapenems/pharmacology , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , beta-Lactam Resistance , Adult , Aged , Aged, 80 and over , Bacterial Outer Membrane Proteins/analysis , Bacterial Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Female , Hospitals , Humans , Klebsiella pneumoniae/isolation & purification , Male , Microbial Sensitivity Tests/methods , Middle Aged , Retrospective Studies , Risk Factors , Taiwan , beta-Lactamases/metabolism
8.
Nat Cell Biol ; 2(11): 767-75, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11056530

ABSTRACT

Whereas total loss of Lis1 is lethal, disruption of one allele of the Lis1 gene results in brain abnormalities, indicating that developing neurons are particularly sensitive to a reduction in Lis1 dosage. Here we show that Lis1 is enriched in neurons relative to levels in other cell types, and that Lis1 interacts with the microtubule motor cytoplasmic dynein. Production of more Lis1 in non-neuronal cells increases retrograde movement of cytoplasmic dynein and leads to peripheral accumulation of microtubules. These changes may reflect neuron-like dynein behaviours induced by abundant Lis1. Lis1 deficiency produces the opposite phenotype. Our results indicate that abundance of Lis1 in neurons may stimulate specific dynein functions that function in neuronal migration and axon growth.


Subject(s)
Dyneins/metabolism , Microtubule-Associated Proteins/biosynthesis , Microtubule-Organizing Center/physiology , Microtubules/physiology , Animals , Brain/metabolism , Brain/pathology , COS Cells , Centromere/physiology , Chlorocebus aethiops , Cytoplasm/metabolism , Dynactin Complex , Fibroblasts/cytology , Golgi Apparatus/metabolism , Golgi Apparatus/physiology , Interphase/physiology , Intracellular Membranes/metabolism , Intracellular Membranes/physiology , Mammals , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Neurons/metabolism , Rats
10.
Nature ; 411(6839): 764-765, 2001 Jun 14.
Article in English | MEDLINE | ID: mdl-11460109

ABSTRACT

Yoo and Lubec show that the amount of p25 is decreased in the brains they studied from patients with Alzheimer's disease or Down's syndrome. Their results persuaded us to conduct a more extensive survey of the p25/p35 ratio in AD brains (to be published elsewhere), as the number of samples was small in both of our studies (eight AD brains in our original study and six in theirs). After analysing a further 25 AD brains and those from 25 age-matched controls, we found that p25 levels are consistently higher in AD brains and that the difference is statistically significant (Student's t-test). This is in agreement with our original observations, as well as being consistent with earlier reports of increased Cdk5 kinase activity in AD brain and of increased amounts of p25 in an animal model of neurodegeneration.

12.
Science ; 255(5040): 87-90, 1992 Jan 03.
Article in English | MEDLINE | ID: mdl-1532458

ABSTRACT

The products of the adenovirus early region 1A (E1A) gene are potent oncoproteins when tested in standard transformation and immortalization assays. Many of the changes induced by E1A may be due to its interaction with cellular proteins. Four of these cellular proteins are the retinoblastoma protein (pRB), p107, cyclin A, and p33cdk2. The pRB and p107 proteins are structurally related and have several characteristics in common, including that they both bind to the SV40 large T oncoprotein as well as to E1A. Cyclin A and p33cdk2 are thought to function in the control of the cell cycle. They bind to one another, forming a kinase that closely resembles the cell cycle-regulating complexes containing p34cdc2. Cyclin A is now shown to bind to p107 in the absence of E1A. The association of p107 with cyclin A suggests a direct link between cell cycle control and the function of p107.


Subject(s)
Cyclins/metabolism , Nuclear Proteins , Oncogene Proteins, Viral/metabolism , Proteins/metabolism , Adenovirus Early Proteins , Amino Acid Sequence , Antibodies, Monoclonal , CDC2 Protein Kinase/metabolism , Cell Line , Cyclins/immunology , Cyclins/isolation & purification , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/genetics , Glutathione Transferase/isolation & purification , Humans , Methionine/metabolism , Molecular Sequence Data , Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107
13.
Neuron ; 18(1): 29-42, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9010203

ABSTRACT

The adult mammalian cortex is characterized by a distinct laminar structure generated through a well-defined pattern of neuronal migration. Successively generated neurons are layered in an "inside-out" manner to produce six cortical laminae. We demonstrate here that p35, the neuronal-specific activator of cyclin-dependent kinase 5, plays a key role in proper neuronal migration. Mice lacking p35, and thus p35/cdk5 kinase activity, display severe cortical lamination defects and suffer from sporadic adult lethality and seizures. Histological examination reveals that the mutant mice lack the characteristic laminated structure of the cortex. Neuronal birth-dating experiments indicate a reversed packing order of cortical neurons such that earlier born neurons reside in superficial layers and later generated neurons occupy deep layers. The phenotype of p35 mutant mice thus demonstrates that the formation of cortical laminar structure depends on the action of the p35/cdk5 kinase.


Subject(s)
Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Cyclin-Dependent Kinases , Neurons/physiology , Protein Serine-Threonine Kinases/genetics , Seizures/genetics , Animals , Cerebral Cortex/embryology , Crosses, Genetic , Cyclin-Dependent Kinase 5 , Embryonic and Fetal Development , Gene Deletion , Genomic Library , Humans , Mice , Mice, Knockout , Mice, Neurologic Mutants , Open Reading Frames , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic , Seizures/pathology , Seizures/physiopathology
14.
Neuron ; 28(3): 697-711, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11163260

ABSTRACT

Disruption of one allele of the LIS1 gene causes a severe developmental brain abnormality, type I lissencephaly. In Aspergillus nidulans, the LIS1 homolog, NUDF, and cytoplasmic dynein are genetically linked and regulate nuclear movements during hyphal growth. Recently, we demonstrated that mammalian LIS1 regulates dynein functions. Here we characterize NUDEL, a novel LIS1-interacting protein with sequence homology to gene products also implicated in nuclear distribution in fungi. Like LIS1, NUDEL is robustly expressed in brain, enriched at centrosomes and neuronal growth cones, and interacts with cytoplasmic dynein. Furthermore, NUDEL is a substrate of Cdk5, a kinase known to be critical during neuronal migration. Inhibition of Cdk5 modifies NUDEL distribution in neurons and affects neuritic morphology. Our findings point to cross-talk between two prominent pathways that regulate neuronal migration.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , Dyneins/metabolism , Microtubule-Associated Proteins/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Axons/metabolism , Axons/ultrastructure , Base Sequence , Brain/metabolism , Cell Movement , Cells, Cultured , Centrosome/metabolism , Conserved Sequence , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , Fungal Proteins/genetics , Growth Cones/metabolism , Humans , Male , Molecular Sequence Data , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , Organ Specificity , Phosphoproteins/metabolism , Sequence Homology, Amino Acid , Testis/metabolism , Two-Hybrid System Techniques
15.
Neuron ; 26(3): 633-46, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10896159

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) is a small serine/threonine kinase that plays a pivotal role during development of the CNS. Cables, a novel protein, interacts with Cdk5 in brain lysates. Cables also binds to and is a substrate of the c-Abl tyrosine kinase. Active c-Abl kinase leads to Cdk5 tyrosine phosphorylation, and this phosphorylation is enhanced by Cables. Phosphorylation of Cdk5 by c-Abl occurs on tyrosine 15 (Y15), which is stimulatory for p35/Cdk5 kinase activity. Expression of antisense Cables in primary cortical neurons inhibited neurite outgrowth. Furthermore, expression of active Abl resulted in lengthening of neurites. The data provide evidence for a Cables-mediated interplay between the Cdk5 and c-Abl signaling pathways in the developing nervous system.


Subject(s)
Carrier Proteins/physiology , Cyclin-Dependent Kinases/physiology , Cyclins , Neurites/physiology , Phosphoproteins/physiology , Phosphotransferases/metabolism , Proto-Oncogene Proteins c-abl/physiology , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , COS Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/metabolism , Embryo, Mammalian , Mice , Mitosis/physiology , Molecular Sequence Data , Neurons/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Substrate Specificity , Tyrosine/metabolism , Up-Regulation
16.
Curr Biol ; 10(7): 363-72, 2000 Apr 06.
Article in English | MEDLINE | ID: mdl-10753743

ABSTRACT

BACKGROUND: The p35-Cdk5 kinase has been implicated in a variety of functions in the central nervous system (CNS), including axon outgrowth, axon guidance, fasciculation, and neuronal migration during cortical development. In p35(-/-) mice, embryonic cortical neurons are unable to migrate past their predecessors, leading to an inversion of cortical layers in the adult cortex. RESULTS: In order to identify molecules important for p35-Cdk5-dependent function in the cortex, we screened for p35-interacting proteins using the two-hybrid system. In this study, we report the identification of a novel interaction between p35 and the versatile cell adhesion signaling molecule beta-catenin. The p35 and beta-catenin proteins interacted in vitro and colocalized in transfected COS cells. In addition, the p35-Cdk5 kinase was associated with a beta-catenin-N-cadherin complex in the cortex. In N-cadherin-mediated aggregation assays, inhibition of Cdk5 kinase activity using the Cdk5 inhibitor roscovitine led to the formation of larger aggregates of embryonic cortical neurons. This finding was recapitulated in p35(-/-) cortical neurons, which aggregated to a greater degree than wild-type neurons. In addition, introduction of active p35-Cdk5 kinase into COS cells led to a decreased beta-catenin-N-cadherin interaction and loss of cell adhesion. CONCLUSIONS: The association between p35-Cdk5 and an N-cadherin adhesion complex in cortical neurons and the modulation of N-cadherin-mediated aggregation by p35-Cdk5 suggests that the p35-Cdk5 kinase is involved in the regulation of N-cadherin-mediated adhesion in cortical neurons.


Subject(s)
Cadherins/metabolism , Cerebral Cortex/physiology , Cyclin-Dependent Kinases/metabolism , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Trans-Activators , Cadherins/genetics , Calcium/metabolism , Cell Adhesion , Cell Aggregation , Cell Movement , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cyclin-Dependent Kinase 5 , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/physiology , Protein Binding , Recombinant Proteins/metabolism , Two-Hybrid System Techniques , beta Catenin
17.
Mol Cell Biol ; 16(11): 6486-93, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8887677

ABSTRACT

We have developed a method to study the primary sequence specificities of protein kinases by using an oriented degenerate peptide library. We report here the substrate specificities of eight protein Ser/Thr kinases. All of the kinases studied selected distinct optimal substrates. The identified substrate specificities of these kinases, together with known crystal structures of protein kinase A, CDK2, Erk2, twitchin, and casein kinase I, provide a structural basis for the substrate recognition of protein Ser/Thr kinases. In particular, the specific selection of amino acids at the +1 and -3 positions to the substrate serine/threonine can be rationalized on the basis of sequences of protein kinases. The identification of optimal peptide substrates of CDK5, casein kinases I and II, NIMA, calmodulin-dependent kinases, Erk1, and phosphorylase kinase makes it possible to predict the potential in vivo targets of these kinases.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins , Mitogen-Activated Protein Kinases , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Caenorhabditis elegans Proteins , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin-Binding Proteins/chemistry , Calmodulin-Binding Proteins/metabolism , Casein Kinase II , Casein Kinases , Crystallography, X-Ray , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , Databases, Factual , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Models, Molecular , Muscle Proteins/chemistry , Muscle Proteins/metabolism , NIMA-Related Kinase 1 , NIMA-Related Kinases , Oligopeptides/chemistry , Oligopeptides/metabolism , Phosphopeptides/chemistry , Phosphopeptides/isolation & purification , Phosphorylase Kinase/metabolism , Protein Conformation , Protein Kinases/chemistry , Protein Kinases/metabolism , Substrate Specificity
18.
Mol Biol Cell ; 6(4): 387-400, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7626805

ABSTRACT

p21Cip1 is a cyclin-dependent kinase (Cdk) inhibitor that is transcriptionally activated by p53 in response to DNA damage. We have explored the interaction of p21 with the currently known Cdks. p21 effectively inhibits Cdk2, Cdk3, Cdk4, and Cdk6 kinases (Ki 0.5-15 nM) but is much less effective toward Cdc2/cyclin B (Ki approximately 400 nM) and Cdk5/p35 (Ki > 2 microM), and does not associate with Cdk7/cyclin H. Overexpression of P21 arrests cells in G1. Thus, p21 is not a universal inhibitor of Cdks but displays selectivity for G1/S Cdk/cyclin complexes. Association of p21 with Cdks is greatly enhanced by cyclin binding. This property is shared by the structurally related inhibitor p27, suggesting a common biochemical mechanism for inhibition. With respect to Cdk2 and Cdk4 complexes, p27 shares the inhibitory potency of p21 but has slightly different kinase specificities. In normal diploid fibroblasts, the vast majority of active Cdk2 is associated with p21, but this active kinase can be fully inhibited by addition of exogenous p21. Reconstruction experiments using purified components indicate that multiple molecules of p21 can associate with Cdk/cyclin complexes and inactive complexes contain more than one molecule of p21. Together, these data suggest a model whereby p21 functions as an inhibitory buffer whose levels determine the threshold kinase activity required for cell cycle progression.


Subject(s)
Cyclin-Dependent Kinases/biosynthesis , Gene Expression Regulation, Enzymologic , Proto-Oncogene Proteins p21(ras)/biosynthesis , Binding, Competitive , Cells, Cultured , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Enzyme Repression/physiology , G1 Phase/genetics , G1 Phase/physiology , Gene Products, rex/biosynthesis , Gene Products, rex/genetics , Humans , Kinetics , Proto-Oncogene Proteins p21(ras)/genetics , S Phase/genetics , S Phase/physiology , Tumor Cells, Cultured
19.
Oncogene ; 6(1): 81-8, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1704120

ABSTRACT

The c-fos proto-oncogene is found to be overexpressed at least 30-fold in SMS-SB, a pre-B leukemic cell line compared to other cell types. No gross alteration of the c-fos gene structure in SMS-SB cells can be detected by karyotypic or Southern analyses. C-fos in SMS-SB cells can still be induced by serum, TPA and the calcium ionophore, A23187, and superinduced by the combination of serum and cycloheximide. The elevated levels of c-fos transcripts are not due merely to an increased life span of mRNA, as the half-life of steady state c-fos mRNA in SMS-SB cells is about 40 min. Nuclear run-on transcription assays demonstrate that the transcription rate of c-fos in SMS-SB cells is up-regulated about 30-fold as compared to another pre-B leukemic cell line, NALM-6. In order to determine whether this is a cis- or transacting effect, we sequenced the 550 base pairs upstream of the SMS-SB c-fos coding region and found no mutations upstream of the mRNA CAP site. However, two point mutations at positions +23 and +98, respectively, were found in the 5' noncoding region of the first exon. Consistent with the overexpression of c-fos mRNA, the 55 kD c-fos protein is present in SMS-SB cells through detection by immunoprecipitation, but could not be detected in NALM-6 cells. SMS-SB cells however, do not appear to contain more abundant AP-1 DNA binding activity than NALM-6 cells.


Subject(s)
Burkitt Lymphoma/metabolism , Proto-Oncogene Proteins/biosynthesis , Base Sequence , Blotting, Southern , Calcimycin/pharmacology , Cell Line , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , DNA/analysis , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fos , RNA/analysis , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic
20.
Oncogene ; 8(6): 1593-602, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8502482

ABSTRACT

In the cell cycle of fission and budding yeast, the p34cdc2/CDC28 kinase is required for both the G1-to-S and G2-to-M phase transitions. In vertebrates, the homologous p34cdc2 kinase is required for G2-to-M phase transitions but appears to be dispensable for DNA synthesis. We have investigated the function of a related kinase, p33cdk2, using serum-stimulated quiescent human fibroblasts. While the p33cdk2 protein was expressed at constant levels throughout the cell cycle, p33cdk2 kinase activity was first detected a few hours prior to the onset of DNA synthesis. Microinjection of anti-p33cdk2 antibodies blocked cells from entering S phase. Pre-adsorption of these antibodies with cdk2 protein abrogated their blocking effect suggesting that the G1 arrest caused by these antibodies is cdk2-specific. These results indicate that p33cdk2 is required for the G1-to-S phase transition in mammalian cells. We also show evidence to suggest that the cyclin E/p33cdk2 complex is likely to be required for entry into S phase since the timing of the cyclin E-associated kinase activity was coincident with that of p33cdk2 and preclearing of either component abolished the majority of the histone H1 kinase activity present in the lysates harvested from the late G1.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle/physiology , Cyclin-Dependent Kinases , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Amino Acid Sequence , Antibodies, Monoclonal , Blotting, Western , Cell Division , Cell Line , Cyclin-Dependent Kinase 2 , Cyclins/immunology , G1 Phase , Humans , Leukemia, Myeloid , Molecular Sequence Data , Oligopeptides/immunology , Protein Kinases/analysis , S Phase , Thymidine/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL