Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Neuroimmune Pharmacol ; 11(4): 708-720, 2016 12.
Article in English | MEDLINE | ID: mdl-27352075

ABSTRACT

The neural cell adhesion molecule (NCAM)-derived peptide FG loop (FGL) modulates synaptogenesis, neurogenesis, and stem cell proliferation, enhances cognitive capacities, and conveys neuroprotection after stroke. Here we investigated the effect of subcutaneously injected FGL on cellular compartments affected by degeneration and regeneration after stroke due to middle cerebral artery occlusion (MCAO), namely endogenous neural stem cells (NSC), oligodendrocytes, and microglia. In addition to immunohistochemistry, we used non-invasive positron emission tomography (PET) imaging with the tracer [18F]-fluoro-L-thymidine ([18F]FLT) to visualize endogenous NSC in vivo. FGL significantly increased endogenous NSC mobilization in the neurogenic niches as evidenced by in vivo and ex vivo methods, and it induced remyelination. Moreover, FGL affected neuroinflammation. Extending previous in vitro results, our data show that the NCAM mimetic peptide FGL mobilizes endogenous NSC after focal ischemia and enhances regeneration by amplifying remyelination and modulating neuroinflammation via affecting microglia. Results suggest FGL as a promising candidate to promote recovery after stroke.


Subject(s)
Cell Movement/physiology , Nerve Regeneration/physiology , Neural Cell Adhesion Molecules/administration & dosage , Neural Stem Cells/physiology , Neurogenesis/physiology , Peptides/administration & dosage , Stroke/pathology , Animals , Cell Movement/drug effects , Cells, Cultured , Injections, Subcutaneous , Male , Nerve Regeneration/drug effects , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Positron-Emission Tomography/trends , Rats , Rats, Wistar , Stroke/diagnostic imaging , Stroke/drug therapy
2.
Brain Behav Immun ; 26(5): 778-88, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21986303

ABSTRACT

Altered synaptic morphology, progressive loss of synapses and glial (astrocyte and microglial) cell activation are considered as characteristic hallmarks of aging. Recent evidence suggests that there is a concomitant age-related decrease in expression of the presynaptic protein, synaptophysin, and the neuronal glycoprotein CD200, which, by interacting with its receptor, plays a role in maintaining microglia in a quiescent state. These age-related changes may be indicative of reduced neuroglial support of synapses. FG Loop (FGL) peptide synthesized from the second fibronectin type III module of neural cell adhesion molecule (NCAM), has previously been shown to attenuate age-related glial cell activation, and to 'restore' cognitive function in aged rats. The mechanisms by which FGL exerts these neuroprotective effects remain unclear, but could involve regulation of CD200, modifying glial-synaptic interactions (affecting neuroglial 'support' at synapses), or impacting directly on synaptic function. Light and electron microscopic (EM) analyses were undertaken to investigate whether systemic treatment with FGL (i) alters CD200, synaptophysin (presynaptic) and PSD-95 (postsynaptic) immunohistochemical expression levels, (ii) affects synaptic number, or (iii) exerts any effects on glial-synaptic interactions within young (4 month-old) and aged (22 month-old) rat hippocampus. Treatment with FGL attenuated the age-related loss of synaptophysin immunoreactivity (-ir) within CA3 and hilus (with no major effect on PSD-95-ir), and of CD200-ir specifically in the CA3 region. Ultrastructural morphometric analyses showed that FGL treatment (i) prevented age-related loss in astrocyte-synaptic contacts, (ii) reduced microglia-synaptic contacts in the CA3 stratum radiatum, but (iii) had no effect on the mean number of synapses in this region. These data suggest that FGL mediates its neuroprotective effects by regulating glial-synaptic interaction.


Subject(s)
Aging/physiology , Hippocampus/metabolism , Neural Cell Adhesion Molecules/pharmacology , Neuroglia/physiology , Synapses/physiology , Synaptophysin/biosynthesis , Aging/drug effects , Animals , Antigens, CD/biosynthesis , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , Disks Large Homolog 4 Protein , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Hippocampus/drug effects , Hippocampus/growth & development , Image Processing, Computer-Assisted , Immunohistochemistry , Injections, Intraperitoneal , Intracellular Signaling Peptides and Proteins , Male , Membrane Proteins/biosynthesis , Microscopy, Electron , Neural Cell Adhesion Molecules/administration & dosage , Neuroglia/ultrastructure , Rats , Rats, Wistar , Synapses/ultrastructure
3.
Neurosci Lett ; 501(3): 173-8, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21787839

ABSTRACT

Plannexin represents a NCAM-derived peptide mimicking trans-homophilic NCAM interaction, which proved to exert neuroprotective effects in vitro. The effect of plannexin was evaluated in a rat status epilepticus model. As expected, prolonged seizure activity resulted in a pronounced cell loss in hippocampal subregions. The comparison between the vehicle- and plannexin-treated animals with status epilepticus did not reveal neuroprotective effects of plannexin on mature neurons. However, treatment with plannexin partially prevented the reduction in the number of doublecortin-labeled neuronal progenitor cells, which was evident 48h following status epilepticus. In conclusion, the data might give first evidence that plannexin can protect immature neurons in vivo. Future studies are necessary to evaluate whether disease-modifying or preventive effects are observed in models of epileptogenesis.


Subject(s)
Molecular Mimicry/physiology , Nerve Degeneration/pathology , Neural Cell Adhesion Molecules/physiology , Oligopeptides/administration & dosage , Status Epilepticus/drug therapy , Status Epilepticus/pathology , Acute Disease , Animals , Cell Communication/drug effects , Cell Communication/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Disease Models, Animal , Doublecortin Protein , Female , Hippocampus/cytology , Hippocampus/drug effects , Ligands , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Neural Cell Adhesion Molecules/administration & dosage , Neural Cell Adhesion Molecules/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Rats , Rats, Wistar , Status Epilepticus/complications
4.
Article in English | MEDLINE | ID: mdl-19965267

ABSTRACT

Neural prostheses and recording devices have been successfully interfaced with the nervous system; however, substantial integration issues exist at the biomaterial-tissue interface. In particular, the loss of neurons at the implantation site and the formation of a gliotic scar capsule diminish device performance. We have investigated the potential of a tissue-engineered coating, consisting of adhesion molecule-modified surfaces (i.e., polylysine and collagen) in combination with neurotrophin application (i.e., brain derived neurotrophic factor, BDNF), to enhance the electrode-host interface. Neurite length and density were examined in a retinal explant model. In the presence of BDNF for 7 days, we found no synergistic effect of BDNF and adhesion molecule-modified surfaces on neurite length, although there was a possible increase in neurite density for collagen-coated surfaces. After BDNF withdrawal (7 days BDNF+/7 days BDNF- medium), we found that both polylysine and collagen treated surfaces displayed increases in neurite length and density over negative, untreated control surfaces. These results suggest that adhesion molecules may be used to support chronic neuron-electrode interfaces induced by neurotrophin exposure.


Subject(s)
Brain-Derived Neurotrophic Factor/administration & dosage , Electrodes, Implanted , Neural Cell Adhesion Molecules/administration & dosage , Tissue Engineering/methods , Animals , Biomedical Engineering , Coated Materials, Biocompatible , Collagen/administration & dosage , Nerve Regeneration/drug effects , Neurites/drug effects , Polylysine/administration & dosage , Rabbits , Retina/drug effects , Retina/physiology , Retina/surgery
5.
Behav Brain Res ; 199(2): 288-97, 2009 May 16.
Article in English | MEDLINE | ID: mdl-19133297

ABSTRACT

The FGL peptide is a neural cell adhesion molecule-derived fibroblast growth factor receptor agonist. FGL has both neurotrophic and memory enhancing properties. Neonatal phencyclidine (PCP) treatment on postnatal days 7, 9, and 11 has been shown to result in long-lasting behavioral abnormalities, including cognitive impairment relevant to schizophrenia. The present study investigated the effect of FGL on spatial learning and memory deficits induced by neonatal PCP treatment. Rat pups were treated with 30 mg/kg PCP on postnatal days 7, 9, and 11. Additionally, the rats were subjected to a chronic FGL treatment regimen where FGL was administered throughout development. Rats were tested as adults for spatial reference memory, reversal learning, and working memory in the Morris water maze. The PCP-treated rats demonstrated a robust impairment in working memory and reversal learning. However, the long-term memory component of the reference memory task was not affected by PCP. Chronic FGL treatment had no effect on the reversal learning impairment but ameliorated the working memory deficits almost to the levels of the control groups. In conclusion, the results suggest that the neonatal PCP treatment produced deficits in cognition relevant to schizophrenia. Moreover, working memory function was selectively protected by the neurotrophic peptide, FGL.


Subject(s)
Animals, Newborn/psychology , Memory/drug effects , Neural Cell Adhesion Molecules/agonists , Phencyclidine/pharmacology , Animals , Female , Male , Neural Cell Adhesion Molecules/administration & dosage , Neural Cell Adhesion Molecules/pharmacology , Phencyclidine/administration & dosage , Pregnancy , Rats , Rats, Sprague-Dawley , Reversal Learning/drug effects
6.
Clin Pharmacokinet ; 46(4): 351-8, 2007.
Article in English | MEDLINE | ID: mdl-17375985

ABSTRACT

BACKGROUND: The FG loop peptide (FGL(L)), a novel mimetic of the neural cell adhesion molecule (NCAM), is in clinical development for neurodegenerative disorders such as Alzheimer's disease. Preclinical studies in rats, dogs and monkeys have demonstrated exposure in plasma and cerebrospinal fluid after parenteral or intranasal administration of FGL(L), with no systemic toxicity. This article reports on the results of the first administration of FGL(L) in humans. OBJECTIVE: To determine the tolerability, safety and pharmacokinetics of ascending, single intranasal doses of FGL(L) 25, 100 and 200mg in healthy subjects. METHODS: In an 8-day, open-label, phase I study, 24 healthy male volunteers (mean age 42 [range 24-55] years) received single intranasal doses of FGL(L) (25, 100 and 200mg) in accordance with an ascending dose, sequential-cohort design. RESULTS: All three intranasal doses of FGL(L) were well tolerated and there were no clinical notable abnormalities in ECG recordings, vital signs or laboratory tests. Three subjects (13%) reported five adverse events. A transient (<3 minutes) burning sensation in the nose was reported in two subjects at the 200mg dose level while runny eyes (<2 minutes) were experienced in one subject at 25mg. These events had an onset immediately following intranasal administration, and a relationship to FGL(L) was suspected. One of the latter subjects who had experienced a burning sensation in the nose also experienced dizziness, vomiting and headache with onset >2 days after single-dose administration of FGL(L); no relationship to the study drug was suspected. Quantifiable plasma concentrations of FGL(L) were observed up to 1 hour after intranasal administration of the 100mg dose and up to 4 hours after the 200mg dose (plasma FGL(L) concentrations were undetectable at all timepoints for the 25mg dose). Increasing doses of FGL(L) were associated with higher systemic exposures: mean C(max) 0.52 ng/mL and 1.38 ng/mL (100mg and 200mg, respectively); mean AUC(24) 1.27 ng x h/mL and 4.05 ng x h/mL (100mg and 200mg, respectively). CONCLUSIONS: Intranasal administration of FGL(L) (25, 100 and 200mg) was well tolerated in healthy male volunteers, with no safety concerns and a pharmacokinetic profile that was generally dose related. Further studies are currently being planned to evaluate the effects of FGL(L) in patients with Alzheimer's disease.


Subject(s)
Neural Cell Adhesion Molecules/adverse effects , Neural Cell Adhesion Molecules/pharmacokinetics , Peptides/administration & dosage , Peptides/pharmacokinetics , Administration, Intranasal , Adult , Area Under Curve , Dose-Response Relationship, Drug , Drug Delivery Systems , Electrocardiography/drug effects , Humans , Male , Middle Aged , Neural Cell Adhesion Molecules/administration & dosage , Peptides/adverse effects , Radioimmunoassay
7.
Neuroscience ; 145(1): 209-24, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17223274

ABSTRACT

By means of i.c.v. administration of preaggregated oligomeric beta-amyloid (Abeta)25-35 peptide it was possible in rats to generate neuropathological signs related to those of early stages of Alzheimer's disease (AD). Abeta25-35-administration induced the deposition of endogenously produced amyloid protein. Furthermore, quantitative immunohistochemistry demonstrated time-related statistically significant increases in amyloid immunoreactivity, tau phosphorylation, microglial activation, and astrocytosis, and stereological investigations demonstrated statistically significant increased neuronal cell death and brain atrophy in response to Abeta25-35. Finally, the Abeta25-35-administration led to a reduced short-term memory as determined by the social recognition test. A synthetic peptide termed FGL derived from the neural cell adhesion molecule (NCAM) was able to prevent or, if already manifest, strongly reduce all investigated signs of Abeta25-35-induced neuropathology and cognitive impairment. The FGL peptide was recently demonstrated to be able to cross the blood-brain-barrier. Accordingly, we found that the beneficial effects of FGL were achieved not only by intracisternal, but also by intranasal and s.c. administration of the peptide. Furthermore, FGL-treatment was shown to inhibit the activity of GSK3beta, a kinase implicated in signaling regulating cell survival, tau phosphorylation and the processing of the amyloid precursor protein (APP). Thus, the peptide induced a statistically significant increase in the fraction of GSK3beta phosphorylated on the Ser9-position, a posttranslational modification known to inhibit the activity of the kinase. Hence, the mode of action of FGL with respect to the preventive and curative effects on Abeta25-35-induced neuropathological manifestations and cognitive impairment involves the modulation of intracellular signal-transduction mediated through GSK3beta.


Subject(s)
Amyloid beta-Peptides , Cognition Disorders , Neural Cell Adhesion Molecules/administration & dosage , Neuroprotective Agents/administration & dosage , Peptide Fragments , Amyloid beta-Peptides/metabolism , Animals , CD11b Antigen/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cognition Disorders/chemically induced , Cognition Disorders/drug therapy , Cognition Disorders/pathology , Drug Administration Routes , Glial Fibrillary Acidic Protein/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/drug effects , Hippocampus/pathology , Immunohistochemistry , Injections, Intraventricular , Male , Memory, Short-Term/drug effects , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology , Neuropsychological Tests , Rats , Rats, Wistar , Scopolamine/administration & dosage , tau Proteins/metabolism
8.
Eur J Neurosci ; 22(7): 1589-96, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16197499

ABSTRACT

There is a major unmet need for development of innovative strategies for neuroprotection against ischemic brain injury. Here we show that FGL, a neural cell adhesion molecule (NCAM)-derived peptide binding to and inducing phosphorylation of the fibroblast growth factor receptor (FGFR), acts neuroprotectively after an ischemic insult both in vitro and in vivo. The neuroprotective activity of FGL was tested in vitro on dissociated rat hippocampal neurons and hippocampal slice cultures, using a protocol of oxygen-glucose deprivation (OGD). FGL protected hippocampal neurons from damage and maintained or restored their metabolic and presynaptic activity, both if employed as a pretreatment alone to OGD, and if only applied after the insult. In vivo 24 h pretreatment with a single suboccipital injection of FGL significantly protected hippocampal CA1 neurons from death in a transient global ischemia model in the gerbil. We conclude that FGL promotes neuronal survival after ischemic brain injury.


Subject(s)
Brain Ischemia/prevention & control , Hippocampus/cytology , Neural Cell Adhesion Molecules/administration & dosage , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Animals , Animals, Newborn , Cell Count/methods , Cells, Cultured , Drug Interactions , Glucose/deficiency , Hypoxia , Neural Cell Adhesion Molecules/chemical synthesis , Neuroprotective Agents/chemical synthesis , Organ Culture Techniques , Phosphorylation/drug effects , Propidium , Pyridinium Compounds/metabolism , Pyrroles/pharmacology , Quaternary Ammonium Compounds/metabolism , Rats , Rats, Wistar , Receptors, Fibroblast Growth Factor/metabolism , Synapses/pathology , Tetrazolium Salts/metabolism , Thiazoles/metabolism , Time Factors
9.
Eur Arch Otorhinolaryngol ; 262(1): 64-8, 2005 Jan.
Article in English | MEDLINE | ID: mdl-14735371

ABSTRACT

The dynamics of precursor cells in the olfactory epithelium of juvenile and adult guinea pigs were examined by immunohistochemical double staining using bromodeoxyuridine (BrdU), the neural cell adhesion molecule (N-CAM) and the protein gene product 9.5 (PGP9.5). Expression of apoptotic cells in the olfactory epithelium with the use of the TdT-mediated dUTP biotin nick end labeling (TUNEL) method was also observed. BrdU was given to healthy guinea pigs at the ages of 2 weeks and 6 months old. Tissue specimens were serially collected 1 h to 28 days after administration. BrdU-labeled cells were seen above the basal cell layer after 1 h and migrated to the middle layer of the olfactory epithelium, after 1 day in juveniles and 5 days in adults with expression of N-CAM. PGP9.5 was observed in BrdU-labeled cells after 5 days in juvenile guinea pigs and 7 days in adult. At 14 days after administration, BrdU-labeled cells in the epithelium appeared to decrease. However, a few of these cells were recognized above the basal cell layer after 28 days. The number and location of TUNEL-positive cells did not significantly differ between the juvenile and adult olfactory epithelium. Therefore, we conclude that the division speed from stem cells in juveniles is faster than that in adults, and apoptosis is unaffected by aging in the normal olfactory epithelium.


Subject(s)
Granulocyte Precursor Cells/cytology , Olfactory Mucosa/cytology , Age Factors , Animals , Antibodies/immunology , Antimetabolites/administration & dosage , Antimetabolites/pharmacokinetics , Apoptosis , Bromodeoxyuridine/administration & dosage , Bromodeoxyuridine/pharmacokinetics , Granulocyte Precursor Cells/immunology , Granulocyte Precursor Cells/metabolism , Guinea Pigs , Immunohistochemistry , Male , Neural Cell Adhesion Molecules/administration & dosage , Neural Cell Adhesion Molecules/immunology , Olfactory Mucosa/immunology , Olfactory Mucosa/metabolism , Ubiquitin Thiolesterase/administration & dosage , Ubiquitin Thiolesterase/pharmacokinetics
10.
J Neurosci ; 21(21): 8408-16, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11606629

ABSTRACT

Lesioned axons within the dorsal roots fail to regenerate through the peripheral nerve transition zone and into the spinal cord. This regenerative failure leads to a persistent loss of sensory function. To induce axonal growth across this barrier, we used recombinant adenovirus to express fibroblast growth factor-2 (FGF2), nerve growth factor (NGF), L1 cell adhesion molecule (L1), or beta-galactosidase (LacZ) within the endogenous glia of the dorsal spinal cord 16 d after injury. Expression of either FGF2 or NGF, but not L1 or LacZ, induced robust axonal regeneration into normal as well as ectopic locations within the dorsal spinal cord. This regeneration led to near-normal recovery of thermal sensory function. Functional recovery and the majority of regenerating axons within the dorsal horn disappeared with recutting of the sensory roots. Injections of adenovirus encoding NGF, but not FGF2, also resulted in extensive sprouting of noninjured sensory axons, which we previously demonstrated could cause hyperalgesia and chronic pain. Thus, neurotrophic factor gene therapy administered as late as 16 d after injury may serve as a useful treatment to elicit recovery after dorsal root avulsion; however, the choice of neurotrophin is important to induce selective regeneration of damaged axons.


Subject(s)
Fibroblast Growth Factor 2/administration & dosage , Genetic Therapy/methods , Nerve Growth Factor/administration & dosage , Regeneration/drug effects , Spinal Cord Injuries/therapy , Adenoviridae/genetics , Afferent Pathways/drug effects , Afferent Pathways/physiopathology , Animals , Axons/drug effects , Chronic Disease , Disease Models, Animal , Female , Fibroblast Growth Factor 2/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Leukocyte L1 Antigen Complex , Locomotion/drug effects , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/genetics , Microinjections , Nerve Crush , Nerve Growth Factor/genetics , Neural Cell Adhesion Molecules/administration & dosage , Neural Cell Adhesion Molecules/genetics , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Recovery of Function/drug effects , Recovery of Function/physiology , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , beta-Galactosidase/administration & dosage , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...