Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Fertil Steril ; 113(1): 176-186, 2020 01.
Article in English | MEDLINE | ID: mdl-32033718

ABSTRACT

OBJECTIVE: To characterize the role of steroid hormone and antihormone exposure on neurotrimin (NTM) expression in human leiomyoma and myometrial tissue and cells. DESIGN: Laboratory study of placebo and ulipristal acetate (UPA)-treated patient tissue. In vitro assessment of immortalized myometrial and leiomyoma cell lines after hormone and antihormone exposure. SETTING: Academic research center. PATIENT(S): Not applicable. INTERVENTIONS(S): Exposure of leiomyoma cell lines to 17ß-E2, medroxyprogesterone acetate (MPA), UPA, and fulvestrant. MAIN OUTCOME MEASURE(S): Messenger RNA expression quantified with the use of RNASeq analysis and quantitative real-time polymerase chain reaction (qRT-PCR). Protein levels quantified by means of Western blot analysis. Immunohistochemistry (IHC) on placebo- and UPA-treated patient uterine tissue specimens. RESULT(S): Expression of NTM in human uterine leiomyoma specimens according to RNASeq was increased compared with myometrium (5.22 ± 0.57-fold), which was confirmed with the use of qRT-PCR (1.95 ± 0.05). Furthermore, NTM protein was elevated in leiomyoma tissue compared with matched myometrium (2.799 ± 0.575). IHC revealed increased staining intensity in leiomyoma surgical specimens compared with matched myometrium of placebo patients. Western blot analysis in immortalized leiomyoma cell lines demonstrated an up-regulation of NTM protein expression (2.4 ± 0.04). Treatment of leiomyoma cell lines with 17ß-E2 yielded a 1.98 ± 0.11-fold increase in NTM protein expression; however, treatment with fulvestrant showed no significant change compared with control. Leiomyoma cell lines demonstrated a 1.91 ± 0.97-fold increase in NTM protein expression after progesterone treatment. RNASeq analysis demonstrated a reduced expression in patient leiomyoma after UPA treatment (0.75 ± 0.14). Treatment of leiomyoma cells with UPA demonstrated a reduced total NTM protein amount (0.54 ± 0.31) in patients, which was confirmed with the use of IHC (UPA10 147.2 ± 9.40, UPA20 182.8 ± 8.98). In vitro studies with UPA treatment revealed a concentration-dependent effect that supported these findings. CONCLUSION(S): NTM, a neural cell adhesion molecule, is increased in leiomyoma compared with myometrium in patient tissue and in vitro models after estrogen and progesterone treatment. Down-regulation of expression occurs after UPA treatment, but not after fulvestrant exposure. CLINICAL TRIAL REGISTRATION NUMBER: NCT00290251.


Subject(s)
Contraceptive Agents, Female/pharmacology , Gonadal Steroid Hormones/pharmacology , Hormone Antagonists/pharmacology , Leiomyoma/metabolism , Neural Cell Adhesion Molecules/biosynthesis , Biomarkers/metabolism , Cell Line, Tumor , Contraceptive Agents, Female/therapeutic use , Double-Blind Method , Estradiol/pharmacology , Estradiol/therapeutic use , Female , GPI-Linked Proteins/agonists , GPI-Linked Proteins/biosynthesis , Gonadal Steroid Hormones/therapeutic use , Hormone Antagonists/therapeutic use , Humans , Leiomyoma/drug therapy , Leiomyoma/pathology , Neural Cell Adhesion Molecules/agonists , Norpregnadienes/pharmacology , Norpregnadienes/therapeutic use
2.
J Neural Transm (Vienna) ; 126(11): 1493-1500, 2019 11.
Article in English | MEDLINE | ID: mdl-31501979

ABSTRACT

In the present study, we developed an in vitro model of Huntington disease (HD) by transfecting primary rat hippocampal neurons with plasmids coding for m-htt exon 1 with different number of CAG repeats (18, 50 and 115) and demonstrated the influence of the length of polyQ sequence on neurite elongation. We found that exogenously applied FGF2 significantly rescued the m-htt-induced loss of neurite outgrowth. Moreover, the Enreptin peptide, an FGFR1 and NCAM dual agonist, had a similar neuritogenic effect to FGF2 in clinically relevant m-htt 50Q-expressing neurons. This study has developed an in vitro model of primary hippocampal neurons transfected with m-htt-coding vectors that is a powerful tool to study m-htt-related effects on neuronal placticity.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Hippocampus/drug effects , Huntingtin Protein/metabolism , Neural Cell Adhesion Molecules/pharmacology , Neuronal Outgrowth/drug effects , Neurons/drug effects , Oligopeptides/pharmacology , Receptor, Fibroblast Growth Factor, Type 1 , Animals , Disease Models, Animal , Huntingtin Protein/genetics , Neural Cell Adhesion Molecules/agonists , Rats , Receptor, Fibroblast Growth Factor, Type 1/agonists , Recombinant Proteins
3.
Neurobiol Dis ; 48(3): 533-45, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22842016

ABSTRACT

The fibroblast growth factor receptor (FGFR) plays a vital role in the development of the nervous system regulating a multitude of cellular processes. One of the interaction partners of the FGFR is the neural cell adhesion molecule (NCAM), which is known to play an important role in neuronal development, regeneration and synaptic plasticity. Thus, simultaneous activation of FGFR- and NCAM-mediated signaling pathways may be expected to affect processes underlying neurodegenerative diseases. We here report the identification of a peptide compound, Enreptin, capable of interacting with both FGFR and NCAM. We demonstrate that this dual specificity agonist induces phosphorylation of FGFR and differentiation and survival of primary neurons in vitro, and that these effects are inhibited by abrogation of both NCAM and FGFR signaling pathways. Furthermore, Enreptin crosses the blood-brain barrier after subcutaneous administration, enhances long-term memory in normal mice and ameliorates memory deficit in mice with induced brain inflammation. Moreover, Enreptin reduces cognitive impairment and neuronal death induced by Aß25-35 in a rat model of Alzheimer's disease, and reduces the mortality rate and clinical signs of experimental autoimmune encephalomyelitis in rats. Thus, Enreptin is an attractive candidate for the treatment of neurological diseases.


Subject(s)
Memory/drug effects , Neural Cell Adhesion Molecules/agonists , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Receptors, Fibroblast Growth Factor/agonists , Animals , Behavior, Animal/drug effects , Brain Diseases/pathology , Cell Differentiation/drug effects , Cells, Cultured , Cognition Disorders/pathology , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred BALB C , Neurons/cytology , Rats , Rats, Wistar , Surface Plasmon Resonance
4.
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
5.
J Neurosci Res ; 87(3): 609-16, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-18855942

ABSTRACT

Septal cholinergic neurons project to the hippocampus and release acetylcholine, a neurotransmitter involved in learning and memory. The enzyme choline acetyltransferase (ChAT) is responsible for synthesizing acetylcholine. Promoting ChAT activity and acetylcholine release can lead to new treatments for neurodegenerative diseases with cholinergic deficits, such as Alzheimer's disease. We present evidence that the synthetic molecule C3d, which is a peptide mimetic of the neural cell adhesion molecule (NCAM), promotes ChAT activity in cultures of rat embryonic septal neurons. Our data demonstrate that ChAT activity triggered by C3d is dependent on the fibroblast growth factor receptor (FGFR) and the mitogen-activated protein kinase (MAPK) pathway. C3d did not affect the number of cholinergic neurons in culture, indicating that NCAM homophilic binding enhances ChAT activity, without affecting cholinergic cell survival. In conclusion, the NCAM mimetic peptide C3d promotes ChAT activity in septal neurons through FGFR and MAPK. These findings are relevant to the design of new strategies aimed at stimulating cholinergic function and improving cognition in disorders such as Alzheimer's disease.


Subject(s)
Choline O-Acetyltransferase/metabolism , Neural Cell Adhesion Molecules/metabolism , Peptides/metabolism , Peptides/pharmacology , Acetylcholine/metabolism , Animals , Blotting, Western , Cell Survival/physiology , Cells, Cultured , Immunohistochemistry , Ligands , MAP Kinase Signaling System , Molecular Mimicry , Neural Cell Adhesion Molecules/agonists , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Fibroblast Growth Factor/metabolism , Septum of Brain/metabolism
7.
Eur J Neurosci ; 28(8): 1618-28, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18973581

ABSTRACT

The neural cell adhesion molecule (NCAM) plays a pivotal role in brain plasticity. Brain plasticity itself has a crucial role in the development of depression. The aim of this study was to analyze whether NCAM-deficient (NCAM(-/-)) mice exhibit depression-like behaviour and whether a peptide termed FGL, derived from the NCAM binding site for the fibroblast growth factor (FGF) receptor, is able to reverse the depression-like signs in NCAM(-/-) mice. Our study showed that NCAM(-/-) mice demonstrated increased freezing time in the tail-suspension test and reduced preference for sucrose consumption in the sucrose preference test, reduced adult neurogenesis in the dentate gyrus and reduced levels of the phosphorylated cAMP response element-binding protein (pCREB) in the hippocampus. FGL administered acutely or repeatedly reduced depression-like behaviour in NCAM(-/-) mice without having an effect on their wild-type littermates. Repeated administration of FGL enhanced survival of the newly born neurons in NCAM(-/-) mice and increased the levels of pCREB in both NCAM(+/+) and NCAM(-/-) mice. In conclusion, our data demonstrate that NCAM deficiency in mice results in a depression-like phenotype which can be reversed by the acute or repeated administration of FGL. The results also suggest a role of the deficit in NCAM signalling through the FGF receptor in depression.


Subject(s)
Depressive Disorder/drug therapy , Depressive Disorder/genetics , Neural Cell Adhesion Molecules/agonists , Neural Cell Adhesion Molecules/genetics , Receptors, Fibroblast Growth Factor/agonists , Animals , Atrophy/drug therapy , Atrophy/physiopathology , Atrophy/prevention & control , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Cell Survival/drug effects , Cell Survival/genetics , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Depressive Disorder/physiopathology , Disease Models, Animal , Fibroblast Growth Factors/agonists , Fibroblast Growth Factors/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Mice , Mice, Knockout , Neural Cell Adhesion Molecules/pharmacology , Neural Cell Adhesion Molecules/therapeutic use , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Neurons/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/physiology
8.
J Neurosci ; 24(17): 4197-204, 2004 Apr 28.
Article in English | MEDLINE | ID: mdl-15115815

ABSTRACT

The neural cell adhesion molecule (NCAM) plays a critical role in development and plasticity of the nervous system and is involved in the mechanisms of learning and memory. Here, we show that intracerebroventricular administration of the FG loop (FGL), a synthetic 15 amino acid peptide corresponding to the binding site of NCAM for the fibroblast growth factor receptor 1 (FGFR1), immediately after training rats in fear conditioning or water maze learning, induced a long-lasting improvement of memory. In primary cultures of hippocampal neurons, FGL enhanced the presynaptic function through activation of FGFR1 and promoted synapse formation. These results provide the first evidence for a memory-facilitating effect resulting from a treatment that mimics NCAM function. They suggest that increased efficacy of synaptic transmission and formation of new synapses probably mediate the cognition-enhancing properties displayed by the peptide.


Subject(s)
Memory/drug effects , Molecular Mimicry/physiology , Neural Cell Adhesion Molecules/agonists , Neural Cell Adhesion Molecules/pharmacology , Peptides/pharmacology , Synapses/drug effects , Synaptic Transmission/drug effects , Amino Acid Sequence , Animals , Cells, Cultured , Conditioning, Classical/drug effects , Emotions/drug effects , Injections, Intraventricular , Male , Maze Learning/drug effects , Memory/physiology , Molecular Sequence Data , Motor Activity/drug effects , Neural Cell Adhesion Molecules/chemistry , Neurons/drug effects , Neurons/physiology , Peptides/chemistry , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Pyrroles/pharmacology , Rats , Rats, Wistar , Receptors, Fibroblast Growth Factor/drug effects , Receptors, Fibroblast Growth Factor/metabolism , Synapses/physiology , Synaptic Transmission/physiology
9.
J Mol Neurosci ; 22(1-2): 33-39, 2004.
Article in English | MEDLINE | ID: mdl-14742908

ABSTRACT

The neural cell adhesion molecule (NCAM) plays an important role in neuronal differentiation and synaptic plasticity, making it an attractive target for the development of drugs for the treatment of neurodegenerative disorders. NCAM binds to itself (homophilic binding) and to a series of counter-receptors, including the fibroblast growth factor receptor (FGFR), other adhesion molecules, and various extracellular matrix components (heterophilic binding). By means of combinatorial chemistry and based on the unraveling of the structure of NCAM, it has been possible to develop a number of peptides that mimic NCAM homophilic binding. These peptides interfere with cell adhesion and promote differentiation and cell survival. Recently, a peptide mimicking the heterophilic binding to FGFR has also been identified. It binds and activates the receptor, thereby modulating neurite extension and synaptic plasticity.


Subject(s)
Cell Adhesion/drug effects , Cell Differentiation/drug effects , Neural Cell Adhesion Molecules/agonists , Neuronal Plasticity/drug effects , Peptides/pharmacology , Animals , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Humans , Neural Cell Adhesion Molecules/chemistry , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neuronal Plasticity/physiology , Peptides/chemistry , Peptides/therapeutic use , Protein Binding/drug effects , Protein Binding/physiology , Receptors, Fibroblast Growth Factor/drug effects , Receptors, Fibroblast Growth Factor/metabolism
10.
Neuroscience ; 122(1): 183-91, 2003.
Article in English | MEDLINE | ID: mdl-14596859

ABSTRACT

The neural cell adhesion molecule (NCAM) plays a key role in synaptic plasticity and memory formation. We have recently developed a synthetic peptide, termed C3d, which, through the binding to the first, N-terminal immunoglobulin-like (Ig) module in the extracellular portion of NCAM, has been shown to promote neurite outgrowth and synapse formation in vitro, and to interfere with passive avoidance memory in rats in vivo. In this study, we investigated whether the i.c.v. administration of C3d, either 5.5 h after or 2 days before training, could be effective to modulate the strength at which emotional memory for aversive situations is established into a long-term memory. The effects of the peptide were evaluated in adult male Wistar rats trained in the contextual fear conditioning task. The results indicated that C3d significantly reduced the subsequent long-term retention of the conditioned fear response when administered 5.5 h post-training, as indicated by retention tests performed 2-3 and 7 days post-training. However, this treatment failed to influence conditioning for this task when injected 2 days pre-training. Additional experiments showed that C3d did not influence the emotional or locomotor behaviour of the animals, when tested in the open field task. Furthermore, hippocampal levels of microtubule-associated protein 2 (MAP2), Synaptophysin and NCAM were found unchanged when evaluated by enzyme-linked immunosorbent assay in crude synaptosomal preparations 2 days after peptide i.c.v. injection. Therefore, post-training injection of this synthetic peptide was efficient to attenuate the strength at which memory for contextual fear conditioning was enduringly stored, whilst it did not affect the acquisition of new memories. In addition to further support the view that NCAM is critically involved in memory consolidation, the current findings suggest that the NCAM IgI module is a potential target for the development of therapeutic drugs capable to reduce the cognitive impact induced by exposure to intensive stress experiences.


Subject(s)
Conditioning, Classical/drug effects , Fear , Hippocampus/metabolism , Memory/drug effects , Neural Cell Adhesion Molecules/agonists , Neural Cell Adhesion Molecules/physiology , Animals , Conditioning, Classical/physiology , Dendrites/metabolism , Enzyme-Linked Immunosorbent Assay , Injections, Intraventricular , Ligands , Male , Memory/physiology , Neural Cell Adhesion Molecules/metabolism , Peptides/administration & dosage , Rats , Rats, Wistar , Stress, Physiological/drug therapy , Synapses/metabolism , Synaptosomes/metabolism , Time Factors
11.
Int J Dev Neurosci ; 20(7): 527-36, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12485621

ABSTRACT

The neural cell adhesion molecule, NCAM, not only plays an important role in neuronal migration, differentiation and formation of connections in the developing nervous system, but also in the condensation of the mesodermal mesenchyme of the limb bud. Therefore, NCAM may be regarded as a target molecule for preventive strategies aimed at minimizing the effects of teratogens affecting the prenatal development of the nervous system and the skeleton. Treatment of fetuses with the teratogen pyrimethamine results in a reduced body weight, microcephaly and malformations of the hind limbs and forelimbs, e.g. micromelia, brachydactyly and adactyly. We here show that a peptide agonist of NCAM, C3, partly prevented the defects induced by this treatment. Although intra-amniotic administration of C3 at gestational day 14 had no effect on the pyrimethamine-induced reduction in body weight, it rescued the deficit in brain weight (microcephaly), partly reversed a decrease in thickness of the cortical plate, and significantly reduced the number of malformed fetuses. In vitro, C3 promoted survival of PC12-E2 cells treated with pyrimethamine. Since C3 is a peptide mimetic of NCAM, our data strongly suggest that stimulating of NCAM results in neuroprotection in vivo and in vitro.


Subject(s)
Abnormalities, Multiple/drug therapy , Brain/abnormalities , Brain/drug effects , Cell Adhesion Molecules, Neuronal/administration & dosage , Embryonic and Fetal Development/drug effects , Neural Cell Adhesion Molecules/agonists , Abnormalities, Multiple/chemically induced , Animals , Animals, Newborn , Brain/growth & development , Brain/pathology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Fetal Weight/drug effects , Injections , Organ Size , PC12 Cells , Peptides , Pregnancy , Pregnancy Outcome , Pyrimethamine , Rats , Rats, Wistar , Reference Values , Reproducibility of Results , Sensitivity and Specificity , Teratogens/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...