Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters










Publication year range
1.
Transl Psychiatry ; 7(2): e1047, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28244984

ABSTRACT

In understanding the mechanism of schizophrenia pathogenesis, a significant finding is that drug abuse of phencyclidine or its analog ketamine causes symptoms similar to schizophrenia. Such drug effects are triggered even by administration at post-adolescent stages. Both drugs are N-methyl-d-aspartate receptor (NMDAR) antagonists, leading to a major hypothesis that glutamate hypofunction underlies schizophrenia pathogenesis. The precise region that depends on NMDAR function, however, is unclear. Here, we developed a mouse strain in which NMDARs in the intralaminar thalamic nuclei (ILN) were selectively disrupted. The mutant mice exhibited various schizophrenia-like phenotypes, including deficits in working memory, long-term spatial memory, and attention, as well as impulsivity, impaired prepulse inhibition, hyperlocomotion and hyperarousal. The electroencephalography analysis revealed that the mutant mice had a significantly reduced power in a wide range of frequencies including the alpha, beta and gamma bands, both during wake and rapid eye movement (REM) sleep, and a modest decrease of gamma power during non-REM sleep. Notably, restoring NMDARs in the adult ILN rescued some of the behavioral abnormalities. These findings suggest that NMDAR dysfunction in the ILN contributes to the pathophysiology of schizophrenia-related disorders. Furthermore, the reversal of inherent schizophrenia-like phenotypes in the adult mutant mice supports that ILN is a potential target site for a therapeutic strategy.


Subject(s)
Behavior, Animal , Intralaminar Thalamic Nuclei/metabolism , Nerve Tissue Proteins/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics , Animals , Arousal , Attention , Disease Models, Animal , Electroencephalography , Genetic Therapy , Impulsive Behavior , Locomotion , Male , Maze Learning , Memory, Short-Term , Mice , Mice, Transgenic , Mutation , Phenotype , Prepulse Inhibition , Schizophrenia/metabolism , Schizophrenia/physiopathology , Schizophrenic Psychology , Spatial Memory
2.
Neuroscience ; 290: 518-29, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25644422

ABSTRACT

Cyclic AMP signaling is critical for activity-dependent refinement of neuronal circuits. Global disruption of adenylyl cyclase 1 (AC1), the major calcium/calmodulin-stimulated adenylyl cyclase in the brain, impairs formation of whisker-related discrete neural modules (the barrels) in cortical layer 4 in mice. Since AC1 is expressed both in the thalamus and the neocortex, the question of whether pre- or postsynaptic (or both) AC1 plays a role in barrel formation has emerged. Previously, we generated cortex-specific AC1 knockout (Cx-AC1KO) mice and found that these animals develop histologically normal barrels, suggesting a potentially more prominent role for thalamic AC1 in barrel formation. To determine this, we generated three new lines of mice: one in which AC1 is disrupted in nearly half of the thalamic ventrobasal nucleus cells in addition to the cortical excitatory neurons (Cx/pTh-AC1KO mouse), and another in which AC1 is disrupted in the thalamus but not in the cortex or brainstem nuclei of the somatosensory system (Th-AC1KO mouse). Cx/pTh-AC1KO mice show severe deficits in barrel formation. Th-AC1KO mice show even more severe disruption in barrel patterning. In these two lines, single thalamocortical (TC) axon labeling revealed a larger lateral extent of TC axons in layer 4 compared to controls. In the third line, all calcium-stimulated adenylyl cyclases (both AC1 and AC8) are deleted in cortical excitatory neurons. These mice have normal barrels. Taken together, these results indicate that thalamic AC1 plays a major role in patterning and refinement of the mouse TC circuitry.


Subject(s)
Adenylyl Cyclases/metabolism , Somatosensory Cortex/growth & development , Thalamus/physiology , Adenylyl Cyclases/genetics , Animals , Axons/physiology , Immunohistochemistry , Mice, Knockout , Neuroanatomical Tract-Tracing Techniques , Neuronal Plasticity/physiology , Somatosensory Cortex/physiology , Thalamus/growth & development , Vibrissae/physiology
3.
Neuroscience ; 165(1): 138-50, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-19837137

ABSTRACT

Dendritic spines are postsynaptic structures at excitatory synapses that play important roles in synaptic transmission and plasticity. Dendritic spine morphology and function are regulated by an actin-based cytoskeletal network. Drebrin A, an adult form of drebrin, is an actin-binding protein in dendritic spines, and its decrease is purportedly concerned with synaptic dysfunction in Alzheimer's disease. Rapid conversion of drebrin E, an embryonic form of drebrin, to drebrin A occurs in parallel with synaptic maturation. To understand the physiological role of drebrin isoform conversion in vivo, we generated knockout mice in which a drebrin A-specific exon was deleted from the drebrin gene. Drebrin A-specific knockout (DAKO) mice expressed drebrin E, which substituted for drebrin A. Subcellular fractionation experiment indicated that cytosolic form of drebrin was increased in the brains of DAKO mice. Furthermore, drebrin accumulation in synaptosomes of DAKO mice was much higher than that of wild-type (WT) mice. DAKO mice were viable and showed no apparent abnormalities in their gross brain morphology and general behaviors. However, DAKO mice were impaired in a context-dependent freezing after fear conditioning. These data indicate that drebrin A plays an indispensable role in some processes of generating fear learning and memory.


Subject(s)
Alternative Splicing , Fear , Learning , Neuropeptides/genetics , Animals , Behavior, Animal , Brain/cytology , Brain/metabolism , Dizocilpine Maleate/pharmacology , Maze Learning , Memory , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Neuropeptides/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
4.
Neuroscience ; 161(2): 635-54, 2009 Jun 30.
Article in English | MEDLINE | ID: mdl-19332109

ABSTRACT

Converging evidence from pharmacological and molecular studies has led to the suggestion that inhibition of glycine transporter 1 (GlyT1) constitutes an effective means to boost N-methyl-d-aspartate receptor (NMDAR) activity by increasing the extra-cellular concentration of glycine in the vicinity of glutamatergic synapses. However, the precise extent and limitation of this approach to alter cognitive function, and therefore its potential as a treatment strategy against psychiatric conditions marked by cognitive impairments, remain to be fully examined. Here, we generated mutant mice lacking GlyT1 in the entire forebrain including neurons and glia. This conditional knockout system allows a more precise examination of GlyT1 downregulation in the brain on behavior and cognition. The mutation was highly effective in attenuating the motor-stimulating effect of acute NMDAR blockade by phencyclidine, although no appreciable elevation in NMDAR-mediated excitatory postsynaptic currents (EPSC) was observed in the hippocampus. Enhanced cognitive performance was observed in spatial working memory and object recognition memory while spatial reference memory and associative learning remained unaltered. These findings provide further credence for the potential cognitive enhancing effects of brain GlyT1 inhibition. At the same time, they indicated potential phenotypic differences when compared with other constitutive and conditional GlyT1 knockout lines, and highlighted the possibility of a functional divergence between the neuronal and glia subpopulations of GlyT1 in the regulation of learning and memory processes. The relevance of this distinction to the design of future GlyT1 blockers as therapeutic tools in the treatment of cognitive disorders remains to be further investigated.


Subject(s)
Glycine Plasma Membrane Transport Proteins/genetics , Memory , Prosencephalon/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Down-Regulation , Excitatory Postsynaptic Potentials , Female , Glycine/metabolism , Glycine Plasma Membrane Transport Proteins/biosynthesis , Hippocampus/physiology , Learning , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Mutation , Phencyclidine/pharmacology , Prosencephalon/drug effects , Prosencephalon/metabolism , Psychomotor Performance , Receptors, N-Methyl-D-Aspartate/biosynthesis , Recognition, Psychology , Synaptic Transmission
5.
Arch Virol ; 153(6): 1007-12, 2008.
Article in English | MEDLINE | ID: mdl-18408883

ABSTRACT

Cellular prion protein (PrP(C)), a cell-surface glycoprotein normally associated with neurons, is also expressed in other cell types such as glia and lymphocytes. To further elucidate these roles of PrP(C), wild-type prion protein gene (Prnp(+/+)) mice and Prnp-deficient (Prnp(-/-)) mice were infected with encephalomyocarditis virus B variant (EMCV-B) via an intracranial route. EMCV-B causes encephalitis and apoptotic cell death in vivo. Histopathological studies revealed that Prnp(+/+) mice infected with 600 pfu of EMCV-B showed more severe infiltration of inflammatory cells, accompanied by higher activation of microglia cells around the hippocampus, than Prnp(-/-) mice; viz., no differences in the brain virus titer between these two lines of mice. Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP, nick end-labeling (TUNEL) staining of the brain specimens revealed that the CA1 hippocampal pyramidal cells showed a larger number of apoptotic neurons in Prnp(-/-) than Prnp(+/+) mice. Based on all these findings, PrP(C) may play certain roles in the induction of inflammation and inhibition of apoptosis in vivo.


Subject(s)
Cardiovirus Infections/pathology , Encephalomyocarditis virus , PrPC Proteins/physiology , Animals , Apoptosis , Cardiovirus Infections/virology , Cell Count , Hippocampus/pathology , Inflammation/pathology , Mice , Mice, Knockout , Microglia/pathology , Prions/genetics , Pyramidal Cells/cytology , Pyramidal Cells/physiology
6.
Mol Pharmacol ; 73(2): 282-91, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17965197

ABSTRACT

The neuronal circuits mediating the sedative action of diazepam are unknown. Although the motor-depressant action of diazepam is suppressed in alpha1(H101R) homozygous knockin mice expressing diazepam-insensitive alpha1-GABA(A) receptors, global alpha1-knockout mice show greater motor sedation with diazepam. To clarify this paradox, attributed to compensatory up-regulation of the alpha2 and alpha3 subunits, and to further identify the neuronal circuits supporting diazepam-induced sedation, we generated Emx1-cre-recombinase-mediated conditional mutant mice, selectively lacking the alpha1 subunit (forebrain-specific alpha1(-/-)) or expressing either a single wild-type (H) or a single point-mutated (R) alpha1 allele (forebrain-specific alpha1(-/H) and alpha1(-/R) mice, respectively) in forebrain glutamatergic neurons. In the rest of the brain, alpha1(-/R) mutants are heterozygous alpha1(H101R) mice. Forebrain-specific alpha1(-/-) mice showed enhanced diazepam-induced motor depression and increased expression of the alpha2 and alpha3 subunits in the neocortex and hippocampus, in comparison with their pseudo-wild-type littermates. Forebrain-specific alpha1(-/R) mice were less sensitive than alpha1(-/H) mice to the motor-depressing action of diazepam, but each of these conditional mutants had a similar behavioral response as their corresponding control littermates. Unexpectedly, expression of the alpha1 subunit was reduced in forebrain, notably in alpha1(-/R) mice, and the alpha3 subunit was up-regulated in neocortex, indicating that proper alpha1 subunit expression requires both alleles. In conclusion, conditional manipulation of GABA(A) receptor alpha1 subunit expression can induce compensatory changes in the affected areas. Specifically, alterations in GABA(A) receptor expression restricted to forebrain glutamatergic neurons reproduce the behavioral effects seen after a global alteration, thereby implicating these neurons in the motor-sedative effect of diazepam.


Subject(s)
Cerebral Cortex/drug effects , Diazepam/pharmacology , Glutamic Acid/metabolism , Hypnotics and Sedatives/pharmacology , Motor Activity/drug effects , Neurons/drug effects , Animals , Cerebral Cortex/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Neurons/metabolism , Receptors, GABA-A/biosynthesis , Receptors, GABA-A/genetics
7.
Cell Death Differ ; 14(8): 1398-405, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17464331

ABSTRACT

Among the events that control cellular differentiation, the acetylation of histones plays a critical role in the regulation of transcription and the modification of chromatin. Jun dimerization protein 2 (JDP2), a member of the AP-1 family, is an inhibitor of such acetylation and contributes to the maintenance of chromatin structure. In an examination of Jdp2 'knock-out' (KO) mice, we observed elevated numbers of white adipocytes and significant accumulation of lipid in the adipose tissue in sections of scapulae. In addition, mouse embryo fibroblasts (MEFs) from Jdp2 KO mice were more susceptible to adipocyte differentiation in response to hormonal induction and members of the CCAAT/enhancer-binding proteins (C/EBP) gene family were expressed at levels higher than MEFs from wild-type mice. Furthermore, JDP2 inhibited both the acetylation of histone H3 in the promoter of the gene for C/EBPdelta and transcription from this promoter. Our data indicate that JDP2 plays a key role as a repressor of adipocyte differentiation by regulating the expression of the gene for C/EBPdelta via inhibition of histone acetylation.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Histones/metabolism , Repressor Proteins/physiology , 3T3-L1 Cells , Acetylation , Adipogenesis/genetics , Adipogenesis/physiology , Animals , Base Sequence , CCAAT-Enhancer-Binding Protein-delta/genetics , Cell Differentiation/physiology , DNA Primers/genetics , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Targeting , Histones/chemistry , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Pregnancy , Promoter Regions, Genetic , Repressor Proteins/genetics
8.
Arch Virol ; 152(3): 603-9, 2007.
Article in English | MEDLINE | ID: mdl-17106625

ABSTRACT

Prion proteins (PrPs) contain 2 N-linked glycosylation sites and are present in cells in 3 different forms. An abnormal isoform of prion protein (PrP(Sc)) has different glycoform patterns for different prion strains. However, the molecular basis of the strain-specific glycoform variability in prions has remained elusive. To understand the molecular basis of these glycoform differences, we analyzed PrP(Sc) in 2 lines of transgenic mice (MHM2 and MH2M with PrP null background) that expressed a chimeric PrP. Our result indicated that PrP 131-188 (substitutions at I139M, Y155N, and S170N) contributed to both PrP(C) and PrP(Sc) glycoform ratios. Furthermore, the PrP(Sc) glycoform pattern within these transgenic mice showed a subtle difference depending on the inoculated prion. This study indicated that the PrP(Sc) glycoform ratio was influenced by both host PrP(C) and the prion strain.


Subject(s)
Peptide Fragments/chemistry , Prions/chemistry , Animals , Cricetinae , DNA Primers , Glycoproteins/chemistry , Glycosylation , Mice
9.
Neuroscience ; 139(2): 767-77, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16458438

ABSTRACT

Adaptation of ocular reflexes is a prototype of motor learning. While the cerebellum is acknowledged as the critical site for motor learning, the functional differences between the cerebellar cortex and nuclei in motor memory formation are not precisely known. Two different views are proposed: one that the memory is formed within the cerebellar flocculus, and the other that the memory is formed within vestibular nuclei. Here we developed a new paradigm of long-term adaptation of mouse horizontal optokinetic response eye movements and examined the location of its memory trace. We also tested the role of flocculus and inferior olive in long-term adaptation by chronic lesion experiments. Reversible bilateral flocculus shutdown with local application of 0.5 microl-5% lidocaine extinguished the memory trace of day-long adaptation, while it very little affected the memory trace of week-long adaptation. The responsiveness of vestibular nuclei after week-long adaptation was examined by measuring the extracellular field responses to the electrical stimulation of vestibular nerve under trichloroacetaldehyde anesthesia. The amplitudes and slopes of evoked monosynaptic field response (N1) of week-long adapted mice were enhanced around the medial vestibular nucleus compared with those of control mice. Chronic flocculus or inferior olive lesions abolished both day and week-long adaptations. These results suggest that the functional memory trace of short-term adaptation is formed initially within the cerebellar cortex, and later transferred to vestibular nuclei to be consolidated to a long-term memory. Both day and week-long adaptations were markedly depressed when neural nitric oxide was pharmacologically blocked locally and when neuronal nitric oxide synthase was ablated by gene knockout, suggesting that cerebellar long-term depression underlies both acquisition and consolidation of motor memory.


Subject(s)
Cerebellar Cortex/physiology , Eye Movements/physiology , Learning/physiology , Motor Activity/physiology , Vestibular Nuclei/physiology , Adaptation, Physiological , Anesthetics, Local , Animals , Behavior, Animal , Cerebellar Cortex/drug effects , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Flocculation , Lidocaine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Motor Activity/drug effects , Nitric Oxide Synthase Type I/deficiency , Nystagmus, Optokinetic/physiology , Olivary Nucleus/injuries , Olivary Nucleus/physiology , Reflex, Vestibulo-Ocular/drug effects , Reflex, Vestibulo-Ocular/physiology , Reflex, Vestibulo-Ocular/radiation effects , Time Factors , Vestibular Nuclei/drug effects , omega-N-Methylarginine/pharmacology
10.
Neuroscience ; 136(2): 487-96, 2005.
Article in English | MEDLINE | ID: mdl-16203099

ABSTRACT

The claustrum is a phylogenetically conserved structure, with extensive reciprocal connections with cortical regions, and has thus been considered important for sensory, motor, emotional, and mnemonic coordination or integration. Here, we show by in situ hybridization that the adult monkey claustrum is strongly positive for NETRIN-G2, a gene encoding a glycosyl phosphatidyl-inositol-linked membrane protein, which constitutes a subfamily with NETRIN-G1 within the netrin/UNC6 family. There is a conspicuous dorsal/ventral differentiation, where the label is stronger in the ventral claustrum. NETRIN-G2 positive neurons are not GABAergic, but rather correspond to claustrocortical projection neurons, as demonstrated by retrograde transport of Fast Blue from cortical injections and by double in situ hybridization for NETRIN-G2 and GAD67. Since NETRIN-G2 is known to be preferentially expressed in cortex, in contrast with the thalamically expressed NETRIN-G1, these results raise the possibility of some functional similarity in regulation of excitatory neural transmission in the claustrum and cortex.


Subject(s)
Basal Ganglia/metabolism , Nerve Tissue Proteins/metabolism , Acetylcholinesterase/metabolism , Animals , Densitometry , Glutamate Decarboxylase/metabolism , Image Processing, Computer-Assisted , In Situ Hybridization , Isoenzymes/metabolism , Macaca , Macaca mulatta , Netrins , RNA Probes , Rats , Reverse Transcriptase Polymerase Chain Reaction , Terminology as Topic
11.
Acta Neurochir Suppl ; 86: 297-9, 2003.
Article in English | MEDLINE | ID: mdl-14753456

ABSTRACT

The physiological function of the normal cellular form of prion protein (PrPC) is not yet fully understood. In the current study we used prion protein gene knock-out mice (Prnp-/-) to assess the role of PrPC in traumatic brain injury. Prnp+/- and Prnp-/- mice were subjected to weight-drop contusional brain injury over the left parietal cortex. Prnp-/- mice manifested a significantly larger lesion volume and worse neuromotor scores than did their Prnp+/- littermates. IgG immunostaining revealed that in Prnp-/- mice the breakdown in the blood-brain barrier (BBB) was more extensive at 1 month after brain injury. Our results are in agreement with previous in vitro findings of the neuroprotective role of PrPC and further support the hypothesis that functional loss of PrPC plays a pathogenic role in prion diseases. We also suggest that PrPC modulates BBB function.


Subject(s)
Brain Damage, Chronic/prevention & control , Brain Injuries/metabolism , PrPC Proteins/metabolism , Animals , Brain/metabolism , Brain/pathology , Brain Injuries/pathology , Immunoglobulin G/metabolism , Immunologic Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , PrPC Proteins/deficiency , Staining and Labeling , Tissue Distribution
12.
Cell ; 107(6): 789-800, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11747814

ABSTRACT

Matrix metalloproteinases (MMPs) are essential for proper extracellular matrix remodeling. We previously found that a membrane-anchored glycoprotein, RECK, negatively regulates MMP-9 and inhibits tumor invasion and metastasis. Here we show that RECK regulates two other MMPs, MMP-2 and MT1-MMP, known to be involved in cancer progression, that mice lacking a functional RECK gene die around E10.5 with defects in collagen fibrils, the basal lamina, and vascular development, and that this phenotype is partially suppressed by MMP-2 null mutation. Also, vascular sprouting is dramatically suppressed in tumors derived from RECK-expressing fibrosarcoma cells grown in nude mice. These results support a role for RECK in the regulation of MMP-2 in vivo and implicate RECK downregulation in tumor angiogenesis.


Subject(s)
Extracellular Matrix/physiology , Matrix Metalloproteinases/metabolism , Membrane Glycoproteins/metabolism , Neoplasms, Experimental/pathology , Neovascularization, Pathologic , Neovascularization, Physiologic , Animals , Cells, Cultured , Down-Regulation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , GPI-Linked Proteins , Gene Targeting , Humans , Immunohistochemistry , Matrix Metalloproteinase 14 , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases, Membrane-Associated , Membrane Glycoproteins/genetics , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Nude , Muscle, Smooth, Vascular/metabolism , Mutation , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Transfection , Tumor Cells, Cultured
13.
Nat Med ; 7(11): 1202-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11689884

ABSTRACT

Acute and fulminant liver failure induced by viral hepatitis, alcohol or other hepatotoxic drugs, are associated with tumor necrosis factor (TNF) production. In a mouse model of lethal hepatitis induced by TNF, apoptosis and necrosis of hepatocytes, but also lethality, hypothermia and influx of leukocytes into the liver, are prevented by a broad-spectrum matrix metalloproteinase (MMP) inhibitor, BB-94. Mice deficient in MMP-2, MMP-3 or MMP-9 had lower levels of apoptosis and necrosis of hepatocytes, and better survival. We found induction of MMP-9 activity and fibronectin degradation. Our findings suggest that several MMPs play a critical role in acute, fulminant hepatitis by degrading the extracellular matrix and allowing massive leukocyte influx in the liver. BB-94 also prevented lethality in TNF/interferon-gamma therapy in tumor-bearing mice. A broad-spectrum MMP inhibitor may be potentially useful for the treatment of patients with acute and perhaps chronic liver failure, and in cancer therapies using inflammatory cytokines.


Subject(s)
Hepatitis, Animal/prevention & control , Matrix Metalloproteinase Inhibitors , Phenylalanine/analogs & derivatives , Animals , Apoptosis/drug effects , Hepatitis, Animal/chemically induced , Hepatitis, Animal/enzymology , Hepatitis, Animal/pathology , Humans , Interferon-gamma/therapeutic use , Interferon-gamma/toxicity , Matrix Metalloproteinases/deficiency , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/drug therapy , Phenylalanine/pharmacology , Protease Inhibitors/pharmacology , Recombinant Proteins , Thiophenes/pharmacology , Tumor Necrosis Factor-alpha/therapeutic use , Tumor Necrosis Factor-alpha/toxicity
14.
FEBS Lett ; 508(2): 187-90, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11718713

ABSTRACT

The goal of the present study was to define the role of gelatinase A in angiogenesis. We performed corneal micropocket assays in gelatinase A-deficient mice and their age-matched wild-type littermates. The corneal neovascular area in gelatinase A-deficient mice (0.15+/-0.14 mm(2)) was significantly less than that of wild-type littermates (0.53+/-0.35 mm(2); P<0.01). Similarly, aortic ring assays showed significant reduction of endothelial outgrowth in gelatinase A-deficient mice (0.26+/-0.14 mm(2)) as compared to wild-type littermates (0.44+/-0.06 mm(2); P<0.05). These results suggest that gelatinase A may play an important role in the regulation of corneal angiogenesis.


Subject(s)
Cornea/blood supply , Corneal Neovascularization/enzymology , Matrix Metalloproteinase 2/deficiency , Matrix Metalloproteinase 2/metabolism , Animals , Aorta/cytology , Aorta/drug effects , Cell Movement/drug effects , Cornea/enzymology , Cornea/metabolism , Cornea/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/pharmacology , Gene Deletion , Genotype , Immunohistochemistry , Matrix Metalloproteinase 2/genetics , Mice , Mice, Knockout , Microscopy, Confocal , Neovascularization, Physiologic/drug effects
15.
Neuroreport ; 12(13): 3003-7, 2001 Sep 17.
Article in English | MEDLINE | ID: mdl-11588620

ABSTRACT

Matrix metalloproteinases (MMPs) may contribute to tissue damage after cerebral ischemia. In this study, wildtype and MMP-2 knockout mice were subjected to permanent and transient (2 h) occlusions of the middle cerebral artery. Gelatin zymography showed that MMP-9 levels were increased in all brains after ischemia. MMP-2 levels did not show a significant increase in wildtype mice, and were not detectable in knockout mice. Laser doppler flowmetry demonstrated equivalent ischemic reductions in perfusion in wildtype and knockout mice. In both permanent and transient occlusion paradigms, there were no statistically significant differences between wildtype and knockout mice in terms of 24 h ischemic lesion volumes. These data suggest that MMP-2 does not contribute to acute tissue damage in this model of focal ischemia.


Subject(s)
Brain Ischemia/enzymology , Cerebral Infarction/enzymology , Matrix Metalloproteinase 2/deficiency , Nerve Degeneration/enzymology , Animals , Brain/blood supply , Brain/enzymology , Brain/pathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebral Infarction/pathology , Cerebral Infarction/physiopathology , Cerebrovascular Circulation/physiology , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology
16.
J Endod ; 27(7): 462-6, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11503997

ABSTRACT

In the serum-free culture medium of bovine odontoblasts we detected active gelatinolytic metalloproteinases, matrix metalloproteinase (MMP)-2 and MMP-9 (gelatinases A and B). The activity of MMP-2, in particular, appeared suddenly around day 21 in the culture, coinciding with the development of odontoblastic cell processes and the loss of alkaline phosphatase. Reverse transcriptase-polymerase chain reaction analysis of these odontoblasts demonstrated that messages of MMP-2 but not MMP-9 increased significantly between day 15 and day 21. The in vitro observation indicates that medium conditioned by these odontoblasts and containing significant amounts of MMP-2 degrades not only the collagenous substrates but also purified dentin phosphophoryn as well. We have also observed that dephosphorylated dentin phosphoprotein becomes a better substrate for casein kinase II after limited proteolysis with MMP-2. These results support our working hypothesis that MMP-2-mediated proteolytic processing is an important step in accelerating the process of dentin matrix maturation, which includes phosphorylation and subsequent mineralization. As has been suggested previously, extracellular phosphorylation of matrix proteins is an important step in biomineralization both in bone and in dentin (Mikuni-Takagaki et al., J Bone Miner Res 1995;10:231-42; Zhu et al., Biochem J 1997; 323:637-43). Our present histochemical analysis in MMP-2 knockout mice confirms the concept with the delayed formation of mineralized tissues, dentin, and bone.


Subject(s)
Dentin/enzymology , Matrix Metalloproteinase 2/analysis , Tooth Calcification/physiology , Alkaline Phosphatase/metabolism , Animals , Casein Kinases , Cattle , Cells, Cultured , Collagen/metabolism , Culture Media, Conditioned , Culture Media, Serum-Free , Dentin/physiology , Isoenzymes/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/analysis , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Odontoblasts/cytology , Odontoblasts/enzymology , Phosphoproteins/metabolism , Phosphorylation , Protein Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
17.
Biochem Biophys Res Commun ; 282(1): 103-7, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11263978

ABSTRACT

In this study, the authors investigated normal cellular prion protein (PrP(C)) expression on murine immune systems using prion protein gene-deficient mouse as negative control. Immunocytes expressing PrP(C) in adult and fetal mice were detected by flow cytometry with the monoclonal antibody against PrP(C), 6H4. Cells from thymus and bone marrow reacted positively with 6H4, while spleen cells, peritoneal cells, peripheral blood leukocytes, and intestinal intraepithelial lymphocytes were nonreactive. In thymus, PrP(C) was observed in CD4(-)CD8(-) double-negative thymocytes. PrP(C+) cells of double-negative thymocytes belonged to the CD3(-) subset, but not to the CD3(+) subset. Triple-negative PrP(C+) thymocytes expressed CD44 or CD25 antigens. Furthermore, PrP(C) was observed in c-kit(+) bone marrow cells. In fetuses, PrP(C+) cells were observed in the liver and thymus at day 16.0 and 15.0 of gestation, respectively. These results demonstrated that PrP(C) is expressed on immature immunocytes.


Subject(s)
Bone Marrow/metabolism , PrPC Proteins/metabolism , Protein Isoforms/metabolism , T-Lymphocytes/metabolism , Animals , Mice , Mice, Knockout , PrPC Proteins/genetics , PrPC Proteins/physiology , Protein Isoforms/genetics , Protein Isoforms/physiology
18.
J Biol Chem ; 276(14): 11265-71, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11152682

ABSTRACT

The central event in prion disease is thought to be conformational conversion of the cellular isoform of prion protein (PrP(C)) to the insoluble isoform PrP(Sc). We generated polyclonal and monoclonal antibodies by immunizing PrP(C)-null mice with native PrP(C). All seven monoclonal antibodies (mAbs) immunoprecipitated PrP(C), but they immunoprecipitated PrP(Sc) weakly or not at all, thereby indicating preferential reactivities to PrP(C) in solution. Immunoprecipitation using these mAbs revealed a marked loss of PrP(C) in brains at the terminal stage of illness. Histoblot analyses using these polyclonal antibodies in combination of pretreatment of blots dissociated PrP(C) and PrP(Sc) in situ and consistently demonstrated the decrease of PrP(C) at regions where PrP(Sc) accumulated. Interestingly, same mAbs showed immunohistochemical reactivities to abnormal isoforms. One group of mAbs showed reactivity to materials that accumulated in astrocytes, while the other group did so to amorphous plaques in neuropil. Epitope mapping indicated that single mAbs have reactivities to multiple epitopes, thus implying dual specificities. This suggests the importance of octarepeats as a part of PrP(C)-specific conformation. Our observations support the notion that loss of function of PrP(C) may partly underlie the pathogenesis of prion diseases. The conversion of PrP(C) to PrP(Sc) may involve multiple steps at different sites.


Subject(s)
Prion Diseases , Prions/chemistry , Animals , Antibodies , Mice , Prion Diseases/etiology , Prions/immunology , Prions/metabolism , Prions/pathogenicity , Protein Conformation , Structure-Activity Relationship
19.
Chem Commun (Camb) ; (22): 2318-9, 2001 Nov 21.
Article in English | MEDLINE | ID: mdl-12240054

ABSTRACT

Imino aldol reaction of ketene silyl acetals with imines proceeds smoothly to give beta-amino esters in good yields under the influence of a cation-exchange resin in ethanol, and the work-up of the reaction consists only of a simple filtration followed by concentration of the crude mixture and purification.

20.
Nat Cell Biol ; 2(10): 737-44, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11025665

ABSTRACT

During carcinogenesis of pancreatic islets in transgenic mice, an angiogenic switch activates the quiescent vasculature. Paradoxically, vascular endothelial growth factor (VEGF) and its receptors are expressed constitutively. Nevertheless, a synthetic inhibitor (SU5416) of VEGF signalling impairs angiogenic switching and tumour growth. Two metalloproteinases, MMP-2/gelatinase-A and MMP-9/gelatinase-B, are upregulated in angiogenic lesions. MMP-9 can render normal islets angiogenic, releasing VEGF. MMP inhibitors reduce angiogenic switching, and tumour number and growth, as does genetic ablation of MMP-9. Absence of MMP-2 does not impair induction of angiogenesis, but retards tumour growth, whereas lack of urokinase has no effect. Our results show that MMP-9 is a component of the angiogenic switch.


Subject(s)
Cell Transformation, Neoplastic , Islets of Langerhans/pathology , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic , Pancreatic Neoplasms/blood supply , Acetamides/pharmacology , Animals , Endothelial Growth Factors/isolation & purification , Genes, Switch , Lymphokines/isolation & purification , Mice , Mice, Transgenic , Receptor Protein-Tyrosine Kinases/isolation & purification , Receptors, Growth Factor/isolation & purification , Receptors, Vascular Endothelial Growth Factor , Signal Transduction , Tissue Distribution , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...