Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Acta Biol Hung ; 63 Suppl 1: 5-18, 2012.
Article in English | MEDLINE | ID: mdl-22453739

ABSTRACT

We have previously demonstrated that imidazole-4-acetic acid-ribotide (IAA-RP) is present in the mammalian brain and is an endogenous ligand at imidazoline binding sites. In the present study, we used a polyclonal antiserum to visualize IAA-RP-containing neurons in the rat caudoputamen. We observe IAA-RP-immunostained neurons scattered throughout the dorsal and ventral striatum. Most of these cells co-localize GABA, but none are parvalbumin-immunoreactive. In contrast, approximately 50% of the calbindin D28k-immunopositive striatal neurons co-localize IAA-RP. Electrophysiological studies using corticostriatal slices demonstrated that bath application of IAA-RP reversibly depresses the synaptically mediated component of field potentials recorded in the striatum by stimulation of cortical axons. Addition of competitive glutamate receptor antagonists completely blocks the response, confirming its association with glutamatergic transmission. Using paired-pulse stimuli, IAA-RP was shown to exert, at least in part, a presynaptic effect, but blockade of GABAA receptor-mediated transmission did not alter the response. Lastly, we show that this effect is attributable to imidazoline-1 receptors, and not to α2 adrenergic receptors. Since IAA-RP is an endogenous central regulator of blood pressure, and cardiovascular dysfunction is a common symptom associated with Parkinson's disease (PD), we speculate that IAA-RP-related abnormalities may underlie some of the autonomic dysfunction that occurs in PD.


Subject(s)
Autonomic Nervous System/physiopathology , Basal Ganglia/metabolism , Imidazoles/metabolism , Motor Activity , Neurons/metabolism , Parkinson Disease/metabolism , Ribosemonophosphates/metabolism , Animals , Basal Ganglia/drug effects , Basal Ganglia/physiopathology , Calbindin 1 , Calbindins , Electric Stimulation , Evoked Potentials , Excitatory Amino Acid Antagonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , Imidazoline Receptors/metabolism , Ligands , Male , Microscopy, Fluorescence , Neural Inhibition , Neurons/drug effects , Parkinson Disease/physiopathology , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , S100 Calcium Binding Protein G/metabolism , Synaptic Transmission , Time Factors , gamma-Aminobutyric Acid/metabolism
2.
J Neurophysiol ; 105(3): 1266-75, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21228308

ABSTRACT

Imidazole-4-acetic acid-ribotide (IAA-RP), an endogenous agonist at imidazoline receptors (I-Rs), is a putative neurotransmitter/regulator in mammalian brain. We studied the effects of IAA-RP on excitatory transmission by performing extracellular and whole cell recordings at Schaffer collateral-CA1 synapses in rat hippocampal slices. Bath-applied IAA-RP induced a concentration-dependent depression of synaptic transmission that, after washout, returned to baseline within 20 min. Maximal decrease occurred with 10 µM IAA-RP, which reduced the slope of field extracellular postsynaptic potentials (fEPSPs) to 51.2 ± 5.7% of baseline at 20 min of exposure. Imidazole-4-acetic acid-riboside (IAA-R; 10 µM), the endogenous dephosphorylated metabolite of IAA-RP, also produced inhibition of fEPSPs. This effect was smaller than that produced by IAA-RP (to 65.9 ± 3.8% of baseline) and occurred after a further 5- to 8-min delay. The frequency, but not the amplitude, of miniature excitatory postsynaptic currents was decreased, and paired-pulse facilitation (PPF) was increased after application of IAA-RP, suggesting a principally presynaptic site of action. Since IAA-RP also has low affinity for α(2)-adrenergic receptors (α(2)-ARs), we tested synaptic depression induced by IAA-RP in the presence of α(2)-ARs, I(1)-R, or I(3)-R antagonists. The α(2)-AR antagonist rauwolscine (100 nM), which blocked the actions of the α(2)-AR agonist clonidine, did not affect either the IAA-RP-induced synaptic depression or the increase in PPF. In contrast, efaroxan (50 µM), a mixed I(1)-R and α(2)-AR antagonist, abolished the synaptic depression induced by IAA-RP and abolished the related increase in PPF. KU-14R, an I(3)-R antagonist, partially attenuated responses to IAA-RP. Taken together, these data support a role for IAA-RP in modulating synaptic transmission in the hippocampus through activation of I-Rs.


Subject(s)
Hippocampus/physiology , Imidazoles/pharmacology , Imidazoline Receptors/agonists , Imidazoline Receptors/metabolism , Long-Term Synaptic Depression/physiology , Neural Inhibition/physiology , Ribosemonophosphates/pharmacology , Synaptic Transmission/physiology , Animals , Hippocampus/drug effects , Long-Term Synaptic Depression/drug effects , Male , Neural Inhibition/drug effects , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
3.
Z Psychosom Med Psychother ; 57(3): 244-50, 2011.
Article in German | MEDLINE | ID: mdl-21968937

ABSTRACT

OBJECTIVES: This epidemiological multicenter study investigates the prevalence and comorbidity of somatoform disorder in psychosomatic inpatients. METHODS: Twenty psychosomatic hospitals collected the diagnoses of all treated patients in the years 1998 to 2007. The data were analysed at the "Institute for Quality Assurance in Psychotherapy and Psychosomatic Medicine" (IQP),Munich. RESULTS: Of the 100,607 patients surveyed, 18,492 (18.4 %) fulfilled the ICD-10 criteria for a somatoform disorder. 91.9 % of patients with somatoform disorder have at least one, on average 2.8 additional psychiatric disorders. The mean duration of the symptoms before current treatment was 62.6 months. CONCLUSIONS: The prevalence of somatoform disorder in psychosomatic inpatients is comparable to that found in data from internal or general medicine patients. However, there are major differences in the distribution of the diagnostic subgroups of somatoform disorder.


Subject(s)
Mental Disorders/epidemiology , Psychophysiologic Disorders/epidemiology , Somatoform Disorders/epidemiology , Adult , Comorbidity , Cross-Sectional Studies , Female , Germany , Health Surveys , Hospitals, University/statistics & numerical data , Humans , International Classification of Diseases , Male , Mental Disorders/diagnosis , Middle Aged , Patient Admission/statistics & numerical data , Psychophysiologic Disorders/diagnosis , Somatoform Disorders/diagnosis
4.
J Cell Biol ; 119(3): 605-16, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1383235

ABSTRACT

The DNA sequence between position +36 and -1907 of the murine myelin basic protein gene contains the enhancer and promoter elements necessary for abundant and cell specific expression in transgenic mice. Surprisingly, the pattern of expression promoted by this DNA fragment is a subset of that exhibited by the endogenous myelin basic protein (MBP) gene. Fusion genes prepared with this promoter/enhancer and a Lac Z reporter gene are expressed only in oligodendrocytes and not in Schwann cells, whereas the endogenous MBP gene is expressed in both cell types. The level of transgene expression measured by nuclear run-on experiments is very substantial and rivals that of the endogenous MBP gene. Furthermore, this 1.9-kb DNA fragment directs transcription on the same (or very similar) developmental schedule as the endogenous gene. These results indicate that the MBP promoter/enhancer sequences are at least tripartite: a core promoter, the oligodendrocyte enhancer elements, and a third component that either expands the specificity of the oligodendrocyte enhancer to include Schwann cells or acts independently to specifically stimulate transcription in Schwann cells.


Subject(s)
Brain/physiology , DNA/genetics , Enhancer Elements, Genetic , Myelin Basic Protein/genetics , Oligodendroglia/physiology , Promoter Regions, Genetic , Schwann Cells/physiology , Animals , Base Sequence , Cell Nucleus/physiology , DNA/isolation & purification , Gene Expression , Mice , Mice, Transgenic , Molecular Sequence Data , Organ Specificity , Recombinant Fusion Proteins/metabolism , Restriction Mapping , Spinal Cord/physiology , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
5.
J Cell Biol ; 146(1): 181-92, 1999 Jul 12.
Article in English | MEDLINE | ID: mdl-10402469

ABSTRACT

Neurofilaments are central determinants of the diameter of myelinated axons. It is less clear whether neurofilaments serve other functional roles such as maintaining the structural integrity of axons over time. Here we show that an age-dependent axonal atrophy develops in the lumbar ventral roots of mice with a null mutation in the mid-sized neurofilament subunit (NF-M) but not in animals with a null mutation in the heavy neurofilament subunit (NF-H). Mice with null mutations in both genes develop atrophy in ventral and dorsal roots as well as a hind limb paralysis with aging. The atrophic process is not accompanied by significant axonal loss or anterior horn cell pathology. In the NF-M-null mutant atrophic ventral root, axons show an age-related depletion of neurofilaments and an increased ratio of microtubules/neurofilaments. By contrast, the preserved dorsal root axons of NF-M-null mutant animals do not show a similar depletion of neurofilaments. Thus, the lack of an NF-M subunit renders some axons selectively vulnerable to an age-dependent atrophic process. These studies argue that neurofilaments are necessary for the structural maintenance of some populations of axons during aging and that the NF-M subunit is especially critical.


Subject(s)
Aging/pathology , Axons/pathology , Motor Neurons/pathology , Neurofilament Proteins/physiology , Spinal Nerve Roots/pathology , Animals , Anterior Horn Cells/cytology , Atrophy , Axons/metabolism , Cell Size , Gene Deletion , Hindlimb , Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Mice , Mice, Knockout , Microtubules/metabolism , Microtubules/ultrastructure , Motor Neurons/metabolism , Neurofilament Proteins/deficiency , Neurofilament Proteins/genetics , Paralysis , Spinal Nerve Roots/metabolism , Time Factors
6.
J Cell Biol ; 109(5): 2405-16, 1989 Nov.
Article in English | MEDLINE | ID: mdl-2553746

ABSTRACT

C57 BL/6N mice injected intracranially with the A59 strain of mouse hepatitis virus exhibit extensive viral replication in glial cells of the spinal cord and develop demyelinating lesions followed by virus clearing and remyelination. To study how different glial cell types are affected by the disease process, we combine three-color immunofluorescence labeling with tritiated thymidine autoradiography on 1-micron frozen sections of spinal cord. We use three different glial cell specific antibodies (a) to 2',3' cyclic-nucleotide 3' phosphohydrolase (CNP) expressed by oligodendrocytes, (b) to glial fibrillary acidic protein (GFAP) expressed by astrocytes, and (c) the O4 antibody which binds to O-2A progenitor cells in the rat. These progenitor cells, which give rise to oligodendrocytes and type 2 astrocytes and react with the O4 antibody in the adult central nervous system, were present but rare in the spinal cord of uninfected mice. In contrast, cells with the O-2A progenitor phenotype (O4 + only) were increased in number at one week post viral inoculation (1 WPI) and were the only immunostained cells labeled at that time by a 2-h in vivo pulse of tritiated thymidine. Both GFAP+ only and GFAP+, O4+ astrocytes were also increased in the spinal cord at 1 WPI. Between two and four WPI, the infected spinal cord was characterized by the loss of (CNP+, O4+) oligodendrocytes within demyelinating lesions and the presence of O-2A progenitor cells and O4+, GFAP+ astrocytes, both of which could be labeled with thymidine. As remyelination proceeded, CNP immunostaining returned to near normal and tritiated thymidine injected previously during the demyelinating phase now appeared in CNP+ oligodendrocytes. Thus O4 positive O-2A progenitor cells proliferate early in the course of the demyelinating disease, while CNP positive oligodendrocytes do not. The timing of events suggests that the O-2A progenitors may give rise to new oligodendrocytes and to type 2 astrocytes, both of which are likely to be instrumental in the remyelination process.


Subject(s)
Demyelinating Diseases/pathology , Hepatitis, Viral, Animal/pathology , Neuroglia/pathology , Spinal Cord/pathology , 2',3'-Cyclic-Nucleotide Phosphodiesterases/analysis , Animals , Brain Chemistry , DNA Replication , Demyelinating Diseases/microbiology , Glial Fibrillary Acidic Protein/analysis , Hepatitis, Viral, Animal/microbiology , Immunohistochemistry , Lipids/analysis , Mice , Mice, Inbred C57BL , Murine hepatitis virus , Reference Values , Spinal Cord/analysis
7.
J Cell Biol ; 111(3): 1183-95, 1990 Sep.
Article in English | MEDLINE | ID: mdl-2167897

ABSTRACT

A demyelinating disease induced in C57B1/6N mice by intracranial injection of a coronavirus (murine hepatitis virus strain A59) is followed by functional recovery and efficient CNS myelin repair. To study the biological properties of the cells involved in this repair process, glial cells were isolated and cultured from spinal cords of these young adult mice during demyelination and remyelination. Using three-color immunofluorescence combined with [3H]thymidine autoradiography, we have analyzed the antigenic phenotype and mitotic potential of individual glial cells. We identified oligodendrocytes with an antibody to galactocerebroside, astrocytes with an antibody to glial fibrillary acidic protein, and oligodendrocyte-type 2 astrocyte (O-2A) progenitor cells with the O4 antibody. Cultures from demyelinated tissue differed in several ways from those of age-matched controls: first, the total number of O-2A lineage cells was strikingly increased; second, the O-2A population consisted of a higher proportion of O4-positive astrocytes and cells of mixed oligodendrocyte-astrocyte phenotype; and third, all the cell types within the O-2A lineage showed enhanced proliferation. This proliferation was not further enhanced by adding PDGF, basic fibroblast growth factor (bFGF), or insulin-like growth factor I (IGF-I) to the defined medium. However, bFGF and IGF-I seemed to influence the fate of O-2A lineage cells in cultures of demyelinated tissue. Basic FGF decreased the percentage of cells expressing galactocerebroside. In contrast, IGF-I increased the relative proportion of oligodendrocytes. Thus, O-2A lineage cells from adult mice display greater phenotypic plasticity and enhanced mitotic potential in response to an episode of demyelination. These properties may be linked to the efficient remyelination achieved in this demyelinating disease.


Subject(s)
Demyelinating Diseases/pathology , Myelin Sheath/physiology , Oligodendroglia/cytology , Animals , Autoradiography , Cell Differentiation , Cell Division , Coronaviridae Infections/pathology , Disease Models, Animal , Female , Fluorescent Antibody Technique , In Vitro Techniques , Mice , Mice, Inbred C57BL , Phenotype , Spinal Cord/cytology , Thymidine/metabolism , Tritium
8.
J Cell Biol ; 109(2): 717-27, 1989 Aug.
Article in English | MEDLINE | ID: mdl-2474554

ABSTRACT

Proteolipid protein (PLP) is the most abundant transmembrane protein in myelin of the central nervous system. Conflicting models of PLP topology have been generated by computer predictions based on its primary sequence and experiments with purified myelin. We have examined the initial events in myelin synthesis, including the insertion and orientation of PLP in the plasma membrane, in rat oligodendrocytes which express PLP and the other myelin-specific proteins when cultured without neurons (Dubois-Dalcq, M., T. Behar, L. Hudson, and R. A. Lazzarini. 1986. J. Cell Biol. 102:384-392). These cells, identified by the presence of surface galactocerebroside, the major myelin glycolipid, were stained with six anti-peptide antibodies directed against hydrophilic or short hydrophobic sequences of PLP. Five of these anti-peptide antibodies specifically stained living oligodendrocytes. Staining was only seen approximately 10 d after PLP was first detected in the cytoplasm of fixed and permeabilized cells, suggesting that PLP is slowly transported from the RER to the cell surface. The presence of PLP domains on the extracellular surface was also confirmed by cleavage of such domains with proteases and by antibody-dependent complement-mediated lysis of living oligodendrocytes. Our results indicate that PLP has only two transmembrane domains and that the great majority of the protein, including its amino and carboxy termini, is located on the extracellular face of the oligodendrocyte plasma membrane. This disposition of the PLP molecule suggests that homophilic interactions between PLP molecules of apposed extracellular faces may mediate compaction of adjacent bilayers in the myelin sheath.


Subject(s)
Membrane Lipids/metabolism , Myelin Proteins/metabolism , Neuroglia/metabolism , Oligodendroglia/metabolism , Proteolipids/metabolism , Amino Acid Sequence , Animals , Antibodies/immunology , Cells, Cultured , Epitopes/analysis , Epitopes/immunology , Fluorescent Antibody Technique , Molecular Sequence Data , Molecular Structure , Myelin Proteins/immunology , Oligodendroglia/cytology , Oligodendroglia/ultrastructure , Peptide Hydrolases/pharmacology , Protein Conformation , Proteolipids/immunology , Rats , Rats, Inbred Strains
9.
J Cell Biol ; 129(1): 189-202, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7698985

ABSTRACT

Previous studies (Blank, W. F., M. B. Bunge, and R. P. Bunge. 1974. Brain Res. 67:503-518) showed that Schwann cell paranodal membranes were disrupted in calcium free medium suggesting that cadherin mediated mechanisms may operate to maintain the integrity of the paranodal membrane complex. Using antibodies against the fifth extracellular domain of E-cadherin, we now show by confocal laser and electron immunomicroscopy that E-cadherin is a major adhesive glycoprotein in peripheral nervous system Schwann cells. E-Cadherin is not found, however, in compact myelin bilayers. Rather, it is concentrated at the paranodes, in Schmidt-Lanterman incisures, and at the inner and outer loops. At these loci, E-cadherin is associated with subplasmalemmal electron densities that coordinate in register across several cytoplasmic turns of a single Schwann cell. F-Actin and beta-catenin, two proteins implicated in cellular signaling, also co-localize to E-cadherin positive sites. These complexes are autotypic adherens-type junctions that are confined to the plasma membrane synthesized by a single Schwann cell; E-cadherin was never observed between two Schwann cells, nor between Schwann cells and the axon. Our findings demonstrate that E-cadherin and its associated proteins are essential components in the architecture of the Schwann cell cytoplasmic channel network, and suggest that this network has specialized functions in addition to those required for myelinogenesis.


Subject(s)
Cadherins/physiology , Intercellular Junctions/ultrastructure , Schwann Cells/physiology , Sciatic Nerve/physiology , Trans-Activators , Amino Acid Sequence , Animals , Antibodies , Antibodies, Monoclonal , Axons/physiology , Axons/ultrastructure , Blotting, Western , Cadherins/analysis , Cadherins/immunology , Cell Adhesion , Cytoskeletal Proteins/analysis , Electrophoresis, Polyacrylamide Gel , Immunohistochemistry , Intercellular Junctions/physiology , Intestines/innervation , Lipid Bilayers , Mice , Mice, Inbred Strains , Microscopy, Confocal , Microscopy, Immunoelectron , Molecular Sequence Data , Nerve Fibers/physiology , Nerve Fibers/ultrastructure , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Recombinant Proteins/immunology , Schwann Cells/ultrastructure , Sciatic Nerve/cytology , Sciatic Nerve/ultrastructure , alpha Catenin , beta Catenin
10.
J Cell Biol ; 141(3): 727-39, 1998 May 04.
Article in English | MEDLINE | ID: mdl-9566972

ABSTRACT

Neurofilaments (NFs) are prominent components of large myelinated axons and probably the most abundant of neuronal intermediate filament proteins. Here we show that mice with a null mutation in the mid-sized NF (NF-M) subunit have dramatically decreased levels of light NF (NF-L) and increased levels of heavy NF (NF-H). The calibers of both large and small diameter axons in the central and peripheral nervous systems are diminished. Axons of mutant animals contain fewer neurofilaments and increased numbers of microtubules. Yet the mice lack any overt behavioral phenotype or gross structural defects in the nervous system. These studies suggest that the NF-M subunit is a major regulator of the level of NF-L and that its presence is required to achieve maximal axonal diameter in all size classes of myelinated axons.


Subject(s)
Axons/metabolism , Neurofilament Proteins/metabolism , Animals , Axons/ultrastructure , Cell Line , Gene Deletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neurofilament Proteins/genetics , Phenotype
11.
J Cell Biol ; 143(1): 195-205, 1998 Oct 05.
Article in English | MEDLINE | ID: mdl-9763431

ABSTRACT

Neurofilaments (NFs) are prominent components of large myelinated axons. Previous studies have suggested that NF number as well as the phosphorylation state of the COOH-terminal tail of the heavy neurofilament (NF-H) subunit are major determinants of axonal caliber. We created NF-H knockout mice to assess the contribution of NF-H to the development of axon size as well as its effect on the amounts of low and mid-sized NF subunits (NF-L and NF-M respectively). Surprisingly, we found that NF-L levels were reduced only slightly whereas NF-M and tubulin proteins were unchanged in NF-H-null mice. However, the calibers of both large and small diameter myelinated axons were diminished in NF-H-null mice despite the fact that these mice showed only a slight decrease in NF density and that filaments in the mutant were most frequently spaced at the same interfilament distance found in control. Significantly, large diameter axons failed to develop in both the central and peripheral nervous systems. These results demonstrate directly that unlike losing the NF-L or NF-M subunits, loss of NF-H has only a slight effect on NF number in axons. Yet NF-H plays a major role in the development of large diameter axons.


Subject(s)
Axons/physiology , Axons/ultrastructure , Microtubules/physiology , Neurofilament Proteins/genetics , Neurofilament Proteins/physiology , Actin Cytoskeleton/physiology , Actin Cytoskeleton/ultrastructure , Animals , Chimera , Exons , Genomic Library , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubules/ultrastructure , Neocortex/physiology , Neurofilament Proteins/deficiency , Restriction Mapping , Spinal Cord/physiology , Transfection
12.
J Clin Invest ; 91(5): 2253-9, 1993 May.
Article in English | MEDLINE | ID: mdl-7683701

ABSTRACT

Tissue factor (TF) is a major activator of the coagulation cascade and may play a role in initiating thrombosis after intravascular injury. To investigate whether medial vascular smooth muscle provides a source of TF following arterial injury, the induction of TF mRNA and protein was studied in balloon-injured rat aorta. After full length aortic injury, aortas were harvested at various times and the media and adventitia separated using collagenase digestion and microscopic dissection. In uninjured aortic media, TF mRNA was undetectable by RNA blot hybridization. 2 h after balloon injury TF mRNA levels increased markedly. Return to near baseline levels occurred at 24 h. In situ hybridization with a 35S-labeled antisense rat TF cRNA probe detected TF mRNA in the adventitia but not in the media or endothelium of uninjured aorta. 2 h after balloon dilatation, a marked induction of TF mRNA was observed in the adventitia and media. Using a functional clotting assay, TF procoagulant activity was detected at low levels in uninjured rat aortic media and rose by approximately 10-fold 2 h after balloon dilatation. Return to baseline occurred within 4 d. These data demonstrate that vascular injury rapidly induces active TF in arterial smooth muscle, providing a procoagulant that may result in thrombus initiation or propagation.


Subject(s)
Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Catheterization/adverse effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , RNA, Messenger/biosynthesis , Thromboplastin/biosynthesis , Animals , Aorta, Thoracic/injuries , Cells, Cultured , DNA Probes , Embolism/metabolism , Embolism/pathology , Gene Library , In Situ Hybridization , Kinetics , Male , Muscle, Smooth, Vascular/injuries , RNA/genetics , RNA/isolation & purification , Rats , Rats, Sprague-Dawley , Reference Values , Thromboplastin/genetics , Time Factors
13.
J Neurosci ; 20(11): 4002-10, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10818135

ABSTRACT

The proteolipid protein (PLP) gene encodes two myelin-specific protein isoforms, DM-20 and PLP, which are members of the highly conserved lipophilin family of transmembrane proteins. While the functions of this family are poorly understood, the fact that null mutations of the PLP gene cause leukodystrophy in man is testament to the importance of DM-20 and PLP in normal CNS function. PLP differs from DM-20 by the presence of a 35 amino acid domain exposed to the cytoplasm, which is not encoded by other lipophilin genes and appears to have arisen in amphibians approximately 300 million years before present. However, the lipophilin gene family can be traced back at least 550 million years and is represented in Drosophila and silkworms. Thus, from an evolutionary perspective PLP can reasonably be anticipated to perform functions in CNS myelin that cannot be accomplished by other lipophilins. Herein we use a novel knock-in strategy to generate mice expressing wild-type levels of a Plp gene that has been modified to encode only DM-20. Although DM-20 is incorporated into functional compact myelin sheaths in young animals, our data show that the 35 amino acid PLP-specific peptide is required to engender the normal myelin period and to confer long-term stability on this multilamellar membrane.


Subject(s)
Biological Evolution , Central Nervous System/physiology , Invertebrates/physiology , Myelin Proteins/genetics , Myelin Proteolipid Protein/physiology , Myelin Sheath/metabolism , Nerve Tissue Proteins , Proteolipids/genetics , Vertebrates/physiology , Amino Acid Sequence , Animals , Blotting, Northern , Blotting, Southern , Central Nervous System/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Electron , Molecular Sequence Data , Myelin Proteolipid Protein/genetics , Nerve Degeneration/genetics , Phenotype , Postural Balance/physiology , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Uteroglobin
14.
Mol Oral Microbiol ; 30(6): 451-73, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25953484

ABSTRACT

Tannerella forsythia is the only 'red-complex' bacterium covered by an S-layer, which has been shown to affect virulence. Here, outer membrane vesicles (OMVs) enriched with putative glycoproteins are described as a new addition to the virulence repertoire of T. forsythia. Investigations of this bacterium are hampered by its fastidious growth requirements and the recently discovered mismatch of the available genome sequence (92A2 = ATCC BAA-2717) and the widely used T. forsythia strain (ATCC 43037). T. forsythia was grown anaerobically in serum-free medium and biogenesis of OMVs was analyzed by electron and atomic force microscopy. This revealed OMVs with a mean diameter of ~100 nm budding off from the outer membrane while retaining the S-layer. An LC-ESI-TOF/TOF proteomic analysis of OMVs from three independent biological replicates identified 175 proteins. Of these, 14 exhibited a C-terminal outer membrane translocation signal that directs them to the cell/vesicle surface, 61 and 53 were localized to the outer membrane and periplasm, respectively, 22 were predicted to be extracellular, and 39 to originate from the cytoplasm. Eighty proteins contained the Bacteroidales O-glycosylation motif, 18 of which were confirmed as glycoproteins. Release of pro-inflammatory mediators from the human monocytic cell line U937 and periodontal ligament fibroblasts upon stimulation with OMVs followed a concentration-dependent increase that was more pronounced in the presence of soluble CD14 in conditioned media. The inflammatory response was significantly higher than that caused by whole T. forsythia cells. Our study represents the first characterization of T. forsythia OMVs, their proteomic composition and immunogenic potential.


Subject(s)
Bacterial Outer Membrane Proteins/analysis , Bacteroidetes/pathogenicity , Bacteroidetes/ultrastructure , Cell Membrane Structures/chemistry , Cell Membrane Structures/physiology , Glycoproteins/analysis , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacteroidetes/growth & development , Bacteroidetes/immunology , Cell Membrane Structures/ultrastructure , Cells, Cultured , Culture Media, Conditioned/chemistry , Glycosylation , Humans , Lipopolysaccharide Receptors/biosynthesis , Membrane Glycoproteins/analysis , Organelle Biogenesis , Periplasm/chemistry , Proteomics , U937 Cells , Virulence
15.
Transplantation ; 65(7): 884-92, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9565090

ABSTRACT

BACKGROUND: Acid sphingomyelinase knock-out (ASMKO) mice are a model of types A and B Niemann-Pick disease. In the present study, we evaluated whether bone marrow transplantation (BMT) carried out on newborn ASMKO mice could prevent or alter the Niemann-Pick disease phenotype. METHODS: Previous work from our laboratory had shown that ASMKO mice were highly susceptible to irradiation-induced death. Therefore, we preconditioned 1-day-old ASMKO (n=35) mice with a "sublethal" dose of 200 cGy of total body irradiation before BMT. The transplantation effects were then analyzed by biochemical, pathological, and clinical approaches. RESULTS: Engraftment ranging from 7% to 100% was achieved in 97% of the transplanted animals. Growth of the engrafted animals was improved, and their survival was increased (from a mean of 5 months to 9 months). The onset of ataxia also was delayed in most of the engrafted animals. In accordance with these observations, biochemical and pathological analysis revealed significant changes in the transplanted group as compared with nontransplanted animals. Lipid storage was reduced in several organs, and there was evidence of histologic improvement seen throughout the reticuloendothelial system, even in animals that were engrafted as low as 14%. In the central nervous system, lipid storage also was reduced, and the Purkinje cells, which are almost absent in ASMKO mice, were present in certain areas of the transplanted animals cerebella. CONCLUSIONS: These results demonstrated that BMT could alter the pathologic phenotype in ASMKO mice, but that this procedure alone was not sufficient to elicit a complete therapeutic effect.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Transplantation , Niemann-Pick Diseases/therapy , Sphingomyelin Phosphodiesterase/deficiency , Animals , Animals, Newborn , Bone Marrow Cells/pathology , Cell Division/physiology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Niemann-Pick Diseases/enzymology , Niemann-Pick Diseases/pathology , Sphingomyelins/metabolism , Whole-Body Irradiation
16.
Neuroscience ; 114(1): 111-22, 2002.
Article in English | MEDLINE | ID: mdl-12207959

ABSTRACT

The cellular and subcellular localization of L-citrulline was analyzed in the adult rat brain and compared with that of traditional markers for the presence of nitric oxide synthase. Light, transmission electron, and confocal laser scanning microscopy were used to study tissue sections processed for immunocytochemistry employing a monoclonal antibody against L-citrulline or polyclonal anti-neuronal nitric oxide synthase sera, and double immunofluorescence to detect neuronal nitric oxide synthase and L-citrulline co-localization. The results demonstrate that the same CNS regions and cell types are labeled by neuronal nitric oxide synthase polyclonal antisera and L-citrulline monoclonal antibodies, using both immunocytochemistry and immunofluorescence. Short-term pretreatment with a nitric oxide synthase inhibitor reduces L-citrulline immunostaining, but does not affect neuronal nitric oxide synthase immunoreactivity. In the vestibular brainstem, double immunofluorescence studies show that many, but not all, neuronal nitric oxide synthase-positive cells co-express L-citrulline, and that local intracellular patches of intense L-citrulline accumulation are present in some neurons. Conversely, all L-citrulline-labeled neurons co-express neuronal nitric oxide synthase. Cells expressing neuronal nitric oxide synthase alone are interpreted as neurons with the potential to produce nitric oxide under other stimulus conditions, and the subcellular foci of enhanced L-citrulline staining are viewed as intracellular sites of nitric oxide production. This interpretation is supported by ultrastructural observations of subcellular foci with enhanced L-citrulline and/or neuronal nitric oxide synthase staining that are located primarily at postsynaptic densities and portions of the endoplasmic reticulum. We conclude that nitric oxide is produced and released at focal sites within neurons that are identifiable using L-citrulline as a marker.


Subject(s)
Cell Compartmentation/physiology , Citrulline/metabolism , Nitrergic Neurons/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/biosynthesis , Vestibular Nuclei/metabolism , Animals , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Male , Microscopy, Electron , NG-Nitroarginine Methyl Ester/pharmacology , Nitrergic Neurons/ultrastructure , Rats , Rats, Sprague-Dawley , Synaptic Membranes/metabolism , Synaptic Membranes/ultrastructure , Vestibular Nuclei/ultrastructure
17.
Neuroscience ; 127(4): 881-91, 2004.
Article in English | MEDLINE | ID: mdl-15312900

ABSTRACT

Hippocampal neurogenesis in adult mammals is influenced by many factors. Lesioning of the entorhinal cortex is a standard model used to study injury and repair in the hippocampus. Here we use bromodeoxyuridine (BrdU) labeling combined with immunohistochemical identification using cell type specific markers to follow the fate of neural progenitors in the hippocampus following entorhinal cortex lesioning in mice. We show that unilateral entorhinal cortex lesioning does not alter the rate of neural progenitor proliferation in the ipsilateral dentate gyrus during the first 3 days after lesioning. However it enhances cell survival at 42 days post-lesioning leading to an increased number of beta-III tubulin and calbindin-immunoreactive neurons being produced. By contrast, when BrdU was administered 21 days post-lesioning, the number of surviving cells 21 days later was similar on the lesioned and non-lesioned sides. Thus, acutely entorhinal cortex lesioning promotes neurogenesis by enhancing survival of either neural progenitors or their progeny. However, this stimulus to neurogenesis is not sustained into the recovery period.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/physiology , Hippocampus/cytology , Hippocampus/physiology , Animals , Cell Differentiation/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Stem Cells/cytology , Stem Cells/physiology
18.
Brain Res Mol Brain Res ; 26(1-2): 177-88, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7854045

ABSTRACT

In ten transgenic lines, expression of a human mid-sized (M) neurofilament (NF) transgene was restricted to neurons in the central and peripheral nervous systems. However, no two lines gave identical expression patterns and none exactly matched the expression of mouse NF(M). These varied expression patterns within the neural compartment likely result from interactions of the transgene with enhancer elements located in the regions flanking the insertion site. Unexpected patterns of enhancer activity included an enhancer active in subsets of cerebellar basket cells as well as others preferentially active in subsets of motor or sensory neurons.


Subject(s)
Brain/metabolism , Enhancer Elements, Genetic , Neurofilament Proteins/biosynthesis , Neurofilament Proteins/genetics , Neurons/metabolism , Spinal Cord/metabolism , Amino Acid Sequence , Animals , Axons/metabolism , Base Sequence , Crosses, Genetic , Female , Fluorescent Antibody Technique , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Molecular Sequence Data , Neurofilament Proteins/analysis , Oligodeoxyribonucleotides , Organ Specificity , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Trigeminal Nerve/metabolism
19.
Brain Res Mol Brain Res ; 15(1-2): 85-98, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1279352

ABSTRACT

By a combination of DNase I footprinting, methylation interference, and gel shift analyses we have identified multiple binding sites for nuclear proteins within the promoter region of the human neurofilament H gene. Two sites likely bind the transcription factor Sp1 while two others may be targets for previously unrecognized DNA binding proteins. One site, PAL, occurs within the 10 bp sequence GGGGAGGAGG. Two copies of the PAL sequence form an interrupted palindrome around one of the Sp1 sites. A second site, PROX, is found within the sequence GGTTGGACC. Nuclear extracts prepared from both neural and non-neural cell lines, mouse brain, and mouse liver contain proteins that recognize and bind to the PROX and PAL sequences indicating that proteins which bind to these target sequences are widespread. The appearance of these target sequences in the 5' upstream region of several neuron specific genes suggests that they play key roles in the transcription of neuron specific genes. The functional activity of these target DNA sequences was demonstrated by transfection assays using a reporter gene fused to nested deletions of the NF(H) promoter region. Interestingly, these assays revealed that maximal transient expression was obtained with DNA fusion genes containing the PAL, PROX and TATA sequences. Inclusion of the Sp1 sites into the fusion genes failed to enhance the expression of the reporter gene. To determine if the NF(H) promoter can be activated in a tissue specific manner during development transgenic mice containing the promoter region linked to a beta-galactosidase reporter gene were generated. In one line sporadic expression of the transgene occurred in the CNS and testis while in four other lines no expression occurred. Collectively these results suggest that the NF(H) gene promoter is active in a tissue specific manner only by interactions with regulatory elements that lie further upstream or downstream of the start site of initiation.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Neurofilament Proteins/metabolism , Animals , Base Sequence , Deoxyribonuclease I , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Plasmids , Promoter Regions, Genetic , RNA/metabolism , Transfection , beta-Galactosidase/genetics
20.
Brain Res Mol Brain Res ; 15(1-2): 76-84, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1279351

ABSTRACT

We have created transgenic mice which carry and express the gene encoding the human NF(M) subunit. RNAase protection assays reveal that the transgene is abundantly expressed in CNS and PNS but also, at very low levels in some non-neural tissues as well. Although the neurospecificity of transgene transcription was not absolute, we are able to detect the protein only in neurons with immunocytochemical techniques. Glial and endothelial cells do not contain immunoreactive materials. Interesting subtle differences in the relative level of the human transgene encoded and endogenous murine encoded NF(M) proteins were noted in different regions of the brain. Similar differences were found in the levels of transgene and endogenous gene mRNA suggesting that these differences may be traceable to differences in RNA transcription or stability. Our data demonstrate, within the sensitivity of the immunocytochemical techniques we used, that the human NF(M) protein is present only in the neurons of the transgenic mice and that it is present in the same neurons as the endogenous NF(M). Furthermore, immunoelectron-microscopic examination of isolated neurofilaments shows that the human NF(M) coassembles with the endogenous NF(M) during filament formation. Thus, although the human NF(M) possesses a much larger multiphosphorylation site in its carboxy terminus, it seems to be the functionally equivalent to the mouse protein, even in the murine neuron.


Subject(s)
Intermediate Filaments/metabolism , Animals , Antibodies, Monoclonal/immunology , Blotting, Western , Brain Chemistry , DNA/biosynthesis , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Immunoelectron , Neurons/metabolism , RNA/biosynthesis , Ribonucleases/biosynthesis , Spinal Cord/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL