Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Biochim Biophys Acta Gen Subj ; 1861(2): 235-245, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27815219

ABSTRACT

BACKGROUND: Recently, it was demonstrated that G-protein-coupled receptors (GPCRs) can transactivate tyrosine kinase receptors in absence of their ligands. In this work, driven by the observation that mAChRs and fibroblast growth factor receptors (FGFRs) share signalling pathways and regulation of brain functions, it was decided to explore whether mAChRs activation may transactivate FGFRs and, if so, to characterize the related trophic effects in cultured hippocampal neurons. METHODS: Oxotremorine-M transactivation of FGFRs and related trophic effects were tested in primary hippocampal neurons. Western blotting and in situ proximity ligation assay (PLA) were used to detect FGFR phosphorylation (pFGFR) levels and M1R-FGFR1 heteroreceptor complexes, respectively. RESULTS: Oxotremorine-M, a non-selective mAChRs agonist, was able to transactivate FGFR and this transactivation was blocked by Src inhibitors. Oxotremorine-M treatment produced a significant increase in the primary neurite outgrowth that was blocked by pre-treatment with the pFGFR inhibitor SU5402 and Src inhibitors. This trophic effect was almost similar to that induced by fibroblast growth factor-2 (FGF-2). By using atropine as nonselective mAChRs or pirenzepine as selective antagonist for M1 receptor (M1R) we could show that mAChRs are involved in modulating the pFGFRs. Using PLA, M1R-FGFR1 heteroreceptor complexes were identified in the hippocampus and cerebral cortex. CONCLUSION: The current findings, by showing functional mAChR-FGFR interactions, will contribute to advance the understanding of the mechanisms involved in the actions of cholinergic drugs on neuronal plasticity. GENERAL SIGNIFICANT: Data may help to develop novel therapeutic strategies not only for neurodegenerative diseases but also for depression-induced atrophy of hippocampal neurons.


Subject(s)
Hippocampus/metabolism , Neuronal Outgrowth/physiology , Neurons/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Muscarinic M1/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Muscarinic/metabolism , Animals , Fibroblast Growth Factor 2/metabolism , Hippocampus/drug effects , Male , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons/drug effects , Oxotremorine/analogs & derivatives , Oxotremorine/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
2.
Int J Sports Med ; 35(1): 19-27, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23868681

ABSTRACT

The lack of dystrophin in mdx mice leads to cycles of muscle degeneration and regeneration processes. Various strategies have been proposed in order to reduce the muscle-wasting component of muscular dystrophy, including implementation of an exercise programme. The aim of this study was to examine how low-intensity endurance exercise affects the degeneration-regeneration process in dystrophic muscle of male mdx mice. Mice were subjected to low-intensity endurance exercise by running on a motorized Rota-Rod for 5 days/week for 6 weeks. Histomorphological analysis showed a significant reduction of measured inflammatory-necrotic areas in both gastrocnemius and quadriceps muscle of exercised mdx mice as compared to matched sedentary mdx mice. The degenerative-regenerative process was also evaluated by examining the protein levels of connexin 39 (Cx39), a specific gene expressed in injured muscles. Cx39 was not detected in sedentary wild type mice, whereas it was found markedly increased in sedentary mdx mice, revealing active muscle degeneration-regeneration process. These Cx39 protein levels were significantly reduced in muscles of mdx mice exercised for 30 and 40 days, revealing together with histomorphological analysis a strong reduction of degeneration process in mice subjected to low-intensity endurance exercise. Muscles of exercised mdx mice did not show significant changes in force and fatigue resistance as compared to sedentary mdx mice. Overall in this study we found that specific low-intensity endurance exercise induces a beneficial effect probably by reducing the degeneration of dystrophic muscle.


Subject(s)
Exercise Therapy/methods , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal/therapy , Physical Conditioning, Animal/physiology , Regeneration/physiology , Animals , Biomarkers/metabolism , Blotting, Western , Connexins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Fatigue/physiology , Muscle Strength/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Physical Conditioning, Animal/methods , Physical Endurance , Random Allocation
3.
Neuroscience ; 165(3): 863-73, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-19909793

ABSTRACT

A number of in vitro and in vivo studies using selective agonists have indicated a neuroprotective role for group-II metabotropic glutamate (mGlu2/3) receptors in various models of neuronal injury. Although an interplay among neurotrophic factors and mGlu2/3 receptors signalling system has been suggested as possible mechanism involved on neuroprotection, at present poor information are available concerning the in vivo regulation by mGlu2/3 receptors activation of specific neurotrophic factors. By using in situ hybridization and western blotting methods the aim of present study was to analyse the potential regulatory role of selective mGluR2/3 agonist LY379268 treatment on brain derived neurotrophic factor (BDNF) expression in the mouse brain. The treatment with LY379268 evidenced a significant upregulation of BDNF mRNA levels in the cerebral cortex and in the hippocampal formation with a peak at 3 h from treatment and its disappearance already at 6 h from treatment. An analysis of dose-effect curve revealed that LY379268 may significantly enhance BDNF mRNA expression already at dose of 0.250 mg/kg b.w. The upregulation of BDNF mRNA expression was followed by a significant increase of BDNF protein levels at 24 h from LY379268 treatment. These effects of LY379268 treatment on BDNF expression were restricted to neuronal cells and were blocked by the new selective mGlu2/3 receptor antagonist LY341495, suggesting a receptor specificity. Taken together these findings suggest that several previous observed neuroprotective and trophic actions of mGluR2/3 agonists treatment may be mediated, at least in the cerebral cortex and hippocampal formation, by upregulation of BDNF expression.


Subject(s)
Amino Acids/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Brain/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Receptors, Metabotropic Glutamate/agonists , Amino Acids/administration & dosage , Animals , Brain/metabolism , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , RNA, Messenger/metabolism , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Time Factors , Up-Regulation/drug effects , Xanthenes/pharmacology
4.
Am J Physiol Cell Physiol ; 296(3): C593-606, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19129462

ABSTRACT

In the adult skeletal muscle, various kinds of trauma promote proliferation of satellite cells that differentiate into myoblasts forming new myofibers or to repair the damaged one. The aim of present work was to perform a comparative spatial and temporal analysis of connexin (Cx) 37, Cx39, Cx40, Cx43, and Cx45 expression in the adult regenerating skeletal muscle in response to crush injury. Within 24 h from injury, Cx37 expression was upregulated in the endothelial cells of blood vessels, and, 5 days after injury, Cx37-expressing cells were found inside the area of lesion and formed clusters generating new blood vessels with endothelial cells expressing Cx37. Three days after injury, Cx39 mRNA was selectively expressed in myogenin-positive cells, forming rows of closely apposed cell nuclei fusing in myotubes. Cx40 mRNA-labeled cells were observed within 24 h from injury in the endothelium of blood vessels, and, 5 days after lesion, Cx40-labeled cells were found inside the area of lesion-forming rows of myogenin-positive, closely apposed cells coexpressing Cx39. Within 24 h from lesion, both Cx43 and Cx45 mRNAs were upregulated in individual cells, and some of them were positive for M-cadherin. Three days after injury, a large number of both Cx43 and Cx45 mRNA-labeled and myogenin-positive cells were found inside the area of lesion. Taken together, these results show that at least four Cxs, out of five expressed in regenerating skeletal muscle, can be differentially involved in communication of myogenic cells during the process of cell proliferation, aggregation, and fusion to form new myotubes or to repair damaged myofibers.


Subject(s)
Connexins/metabolism , Muscle, Skeletal/metabolism , Regeneration , Animals , Cell Aggregation , Cell Fusion , Cell Proliferation , Connexin 30 , Connexin 43/metabolism , Connexins/genetics , Constriction , Endothelial Cells/metabolism , Gene Expression Regulation , Immunohistochemistry , In Situ Hybridization , Male , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Muscle, Skeletal/surgery , Neovascularization, Physiologic , RNA, Messenger/metabolism , Rats , Rats, Wistar , Regeneration/genetics , Time Factors , Gap Junction alpha-5 Protein , Gap Junction alpha-4 Protein
5.
Neuroscience ; 145(2): 470-83, 2007 Mar 16.
Article in English | MEDLINE | ID: mdl-17241745

ABSTRACT

Over the past years, evidence has accumulated that stem cells are present in the adult brain, and generate neurons and/or glia from two active germinal zones: the subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the dentate gyrus of the hippocampus. This study shows that acute intermittent nicotine treatment significantly enhances neuronal precursor cell proliferation in the SVZ of adult rat brain, but not in the SGZ of the hippocampus, and pre-treatment with mecamylamine, a nonselective nAChR antagonist, blocks the enhanced precursor proliferation by nicotine. This effect is supported by up-regulation of fibroblast growth factor-2 (FGF-2) mRNA in the SVZ and the expression of its receptor FGFR-1 in cells of SVZ showing precursor cells profile. It is also demonstrated that the nicotine effect on neuronal precursor proliferation is mediated by FGF-2 via fibroblast growth factor receptor 1 (FGFR-1) activation by showing that i.c.v. pre-treatment with anti-FGF-2 antibodies or with FGFR-1 inhibitor 3-[(3-(2-carboxyethyl)-4-methylpyrrol-2-yl)methylene]-2-indolinone (SU5402) blocks nicotine-induced precursor cell proliferation. This nicotine enhancement of neuronal precursor cell proliferation was not accompanied by an increase in the number of apoptotic cells. Taken together the present findings revealed the existence in the SVZ of the adult rat brain of a trophic mechanism mediated by FGF-2 and its receptor and regulated by nAchR activation. This possibility of in vivo regulation of neurogenesis in the adult brain by exogenous factors may aid to develop treatments stimulating neurogenesis with potential therapeutic implications.


Subject(s)
Cell Proliferation/drug effects , Fibroblast Growth Factor 2/genetics , Lateral Ventricles/drug effects , Neurons/drug effects , Nicotine/pharmacology , Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Male , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/metabolism , Nicotine/therapeutic use , Nicotinic Agonists/pharmacology , Nicotinic Agonists/therapeutic use , Nicotinic Antagonists/pharmacology , Pyrroles/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
6.
J Neural Transm (Vienna) ; 114(1): 135-47, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16906354

ABSTRACT

In the present work we reviewed recent advances concerning neuroprotective/neurotrophic effects of acute or chronic nicotine exposure, and the signalling pathways mediating these effects, including mechanisms implicated in nicotine addiction and nAChR desensitization. Experimental and clinical data largely indicate long-lasting effects of nicotine and nicotinic agonists that imply a neuroprotective/neurotrophic role of nAChR activation, involving mainly alpha7 and alpha4beta2 nAChR subtypes, as evidenced using selective nAChR agonists. Compounds interacting with neuronal nAChRs have the potential to be neuroprotective and treatment with nAChR agonists elicits long-lasting neurotrophic effects, e.g. improvement of cognitive performance in a variety of behavioural tests in rats, monkeys and humans. Nicotine addiction, which is mediated by interaction with nACh receptors, is believed to involve the modification of signalling cascades that modulate synaptic plasticity and gene expression. Desensitization, in addition to protecting cells from uncontrolled excitation, is recently considered as a form of signal plasticity. nAChR can generate these longe-lasting effects by elaboration of complex intracellular signals that mediate medium to long-term events crucial for neuronal maintenance, survival and regeneration. Although a comprehensive survey of the gene-based molecular mechanisms that underlie nicotine effects has yet not been performed a growing amount of data is beginning to improve our understanding of signalling mechanisms that lead to neurotrophic/neuroprotective responses. Evidence for an involvement of the fibroblast growth factor-2 gene in nAChR mechanisms mediating neuronal survival, trophism and plasticity has been obtained. However, more work is needed to establish the mechanisms involved in the effects of nicotinic receptor subtype activation from cognition-enhancing and neurotrophic effects to smoking behaviour and to determine more precisely the therapeutic objectives in potential nicotinic drug treatments of neurodegenerative diseases.


Subject(s)
Brain/drug effects , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/drug effects , Animals , Brain/metabolism , Brain/physiopathology , Humans , Nerve Growth Factors/chemistry , Nerve Growth Factors/therapeutic use , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Nicotinic Agonists/chemistry , Nicotinic Agonists/therapeutic use , Receptors, Nicotinic/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tobacco Use Disorder/metabolism , Tobacco Use Disorder/physiopathology
7.
Neuroscience ; 133(3): 787-96, 2005.
Article in English | MEDLINE | ID: mdl-15890456

ABSTRACT

Several studies in different in vitro and in vivo models have demonstrated neuroprotective effects of nicotinic receptor agonists and indirect trophic actions of nicotine on brain are suggested from observations describing nicotine as a cognitive enhancer by increasing vigilance and improving learning and memory. While an increasing number of studies have given evidence of neuroprotective and neurotrophic effects of nicotine treatment, the molecular mechanism mediating the neurotrophic effects of nicotine are not fully understood. Previously in an analysis of several neurotrophic factors as possible mediators of nicotine-induced neuroprotection and/or neurotrophic effects we could reveal that an acute intermittent nicotine treatment increases fibroblast growth factor-2 mRNA and protein in several brain regions of rat brain. Even if other studies have demonstrated in different paradigms that nicotine administration modulates expression level of a variety of genes, there is still a lack of indication which candidate genes, involved in neuroprotective responses are modulated by nicotine. In the present work we have used a microarray assay to further find and characterize new genes responsive to acute intermittent nicotine treatment and linked to neuroprotection. Therefore, we used Rat Genome U34A Affymetrix GeneChip arrays containing about 8800 probe sets to characterize transcriptional responses in the rat parietal cortex after acute intermittent nicotine treatment. We focused our attention to expression of transcription factors and several of them were up- or down-regulated by nicotine, among these Nr4a1 (Nurr77), Egr-1 and Egr-2. In situ hybridization was used to corroborate the microarray data and to reveal further spatial and temporal patterns of these nicotine induced genes. Taken together the present results identified several novel candidate genes modified by acute intermittent nicotine exposure and as such potentially involved in neuroprotective-neurotrophic actions.


Subject(s)
Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Gene Expression Profiling , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Transcription Factors/genetics , Animals , Gene Expression/drug effects , In Situ Hybridization , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Wistar
8.
Cell Tissue Res ; 320(2): 299-310, 2005 May.
Article in English | MEDLINE | ID: mdl-15778849

ABSTRACT

We report a detailed analysis of the expression pattern of the recently identified rat connexin gene, named rat connexin 39 (rCx39), both during embryonic development and in adult life. Qualitative and quantitative reverse transcription/polymerase chain reaction analysis showed intense expression of rCx39 restricted to differentiating skeletal muscles, with a peak of expression detected at 18 days of embryonic life, followed by a rapid decline to undetectable levels within the first week of postnatal life. A combination of the in situ hybridization technique for the detection of rCx39 mRNA and immunohistochemistry for myogenin, a myoblast-specific marker, allowed us to establish that the mRNA for this connexin was expressed in myogenin-positive myoblasts and early myotubes but disappeared in mature myotubes. Moreover, in adult animals, rCx39 mRNA was expressed in myogenic cells involved in skeletal myofiber regeneration following a crush injury. This is the first case of a connexin being mainly expressed in the myogenic cell lineage. The information presented should pave the way to novel molecular approaches in studies on the role of connexin-based gap-junctional communication in skeletal muscle differentiation and regeneration.


Subject(s)
Connexins/genetics , Connexins/metabolism , In Situ Hybridization , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/embryology , Myoblasts, Skeletal/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Lineage , Connexins/chemistry , Gene Expression Regulation, Developmental , Immunohistochemistry , Male , Molecular Sequence Data , Phylogeny , Protein Structure, Tertiary , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Time Factors , Tissue Distribution
9.
Mol Cell Neurosci ; 21(1): 94-113, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12359154

ABSTRACT

Glial connexins (Cxs) make an extensively interconnected functional syncytium created by a network of gap junctions between astrocytes and oligodendrocytes. Among Cxs expressed in the brain, Cx30 is expressed in grey matter astrocytes, as shown at the protein level by immunoistochemistry. In the present study we aimed to perform a detailed study of the regional distribution of Cx30 mRNA in the adult and postnatal developing rat brain, analyzing its expression by in situ hybridization, and determining its cell type localization by double labeling. Recently, it has been suggested that neuronal activity may control the level of intercellular communication between astrocytes through gap junctions channels. Thus, a second aim of the present study was to investigate the short-term effects of kainate-induced seizures on Cx30 expression. The results showed that, in basal condition, Cx30 was expressed only in grey matter astrocytes with distinct regional patterns in developing and adult brain. Kainate treatment induced strong and region-specific changes of astroglial Cx30 mRNA levels and expression of Cx30 mRNA in neuronal cells undergoing cell death, suggesting a direct or indirect involvement of this connexin in the neuronal apoptotic process.


Subject(s)
Apoptosis/genetics , Astrocytes/metabolism , Brain/metabolism , Connexins/genetics , Gene Expression/genetics , Neurons/physiology , Status Epilepticus/genetics , Aging/genetics , Aging/metabolism , Animals , Animals, Newborn , Brain/growth & development , Brain/physiopathology , Cell Communication/genetics , Excitatory Amino Acid Agonists , Immunohistochemistry , Kainic Acid , Male , Neurons/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/genetics , Seizures/physiopathology , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology , Up-Regulation/genetics
10.
Mol Cell Neurosci ; 18(1): 56-67, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11461153

ABSTRACT

Neurotrophin-4 (NT-4) is produced by slow muscle fibers in an activity-dependent manner and promotes growth and remodeling of adult motorneuron innervation. However, both muscle fibers and motor neurons express NT-4 receptors, suggesting bidirectional NT-4 signaling at the neuromuscular junction. Mice lacking NT-4 displayed enlarged and fragmented neuromuscular junctions with disassembled postsynaptic acetylcholine receptor (AChR) clusters, reduced AChR binding, and acetylcholinesterase activity. Electromyographic responses, posttetanic potentiation, and action potential amplitude were also significantly reduced in muscle fibers from NT-4 knock-out mice. Slow-twitch soleus muscles from these mice fatigued twice as rapidly as those from wild-type mice during repeated tetanic stimulation. Thus, muscle-derived NT-4 is required for maintenance of postsynaptic AChR regions, normal muscular electrophysiological responses, and resistance to muscle fatigue. This neurotrophin may therefore be a key component of an activity-dependent feedback mechanism regulating maintenance of neuromuscular connections and muscular performance.


Subject(s)
Muscle Fatigue/physiology , Muscle, Skeletal/physiology , Nerve Growth Factors/genetics , Neuromuscular Junction/physiology , Acetylcholinesterase/metabolism , Age Factors , Animals , Electromyography , Mice , Mice, Knockout , Motor Neurons/physiology , Muscle Contraction/physiology , Muscle Fibers, Slow-Twitch/enzymology , Muscle, Skeletal/cytology , Muscle, Skeletal/innervation , Receptors, Cholinergic/metabolism
11.
Mol Cell Neurosci ; 17(2): 364-72, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11178873

ABSTRACT

XIAP (X-chromosome-linked inhibitor of apoptosis protein) is an antiapoptotic protein which inhibits the activity of caspases and suppresses cell death. However, little is known about the presence and function of XIAP in the nervous system. Here we report that XIAP mRNA is expressed in developing and adult rat brain. Using a specific antibody, we observed XIAP-immunoreactive cells in different brain regions, among others, in the hippocampus and cerebral cortex. Kainic acid, which induces delayed cell death of specific neurons, increased the levels of XIAP in the CA3 region of hippocampus. XIAP was, however, largely absent in cells undergoing cell death, as shown by TUNEL labeling and staining for active caspase-3. In cultured hippocampal neurons, XIAP was initially upregulated by kainic acid and then degraded in a process blocked by the caspase-3 inhibitor DEVD. Similarly, recombinant XIAP is cleaved by active caspase-3 in vitro. The results show that there is biphasic regulation of XIAP in the hippocampus following kainic acid and that XIAP becomes a target for caspase-3 activated during cell death in the hippocampus. The degradation of XIAP by kainic acid contributes to neuronal cell death observed in vulnerable neurons of the hippocampus after caspase activation.


Subject(s)
Apoptosis/genetics , Cell Death/genetics , Nerve Degeneration/genetics , Proteins/genetics , Animals , Biomarkers , Caspase 3 , Caspases/metabolism , Cells, Cultured , Excitatory Amino Acid Agonists , Gene Expression Regulation, Developmental , Genetic Linkage , Hippocampus/chemistry , Hippocampus/cytology , Hippocampus/growth & development , In Situ Nick-End Labeling , Kainic Acid , Male , Nerve Degeneration/chemically induced , Neurons/cytology , Neurons/enzymology , Proteins/analysis , RNA, Messenger/analysis , Rats , X Chromosome , X-Linked Inhibitor of Apoptosis Protein
12.
Cell Commun Adhes ; 8(4-6): 173-8, 2001.
Article in English | MEDLINE | ID: mdl-12064584

ABSTRACT

By combining in silico and bench molecular biology methods we have identified a novel human gap junction gene that encodes a protein designated HCx31.9. We have determined its human chromosomal location and gene structure, and we have identified a putative mouse ortholog, mCx30.2. We have observed the presence of HCx31.9 in human cerebral cortex, liver, heart, spleen, lung, and kidney and the presence of mCx30.2 in mouse cerebral cortex, liver and lung. Moreover, preliminary data on the electrophysiological properties of HCx31.9 have been obtained by functional expression in paired Xenopus oocytes and in transfected N2A cells.


Subject(s)
Connexins/genetics , Gap Junctions/metabolism , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , Connexins/chemistry , Connexins/classification , Connexins/metabolism , Gap Junctions/chemistry , Gene Expression , Humans , Mice , Molecular Sequence Data , Oocytes/physiology , Patch-Clamp Techniques , Phylogeny , Sequence Alignment , Tissue Distribution , Xenopus laevis
13.
Eur J Neurosci ; 12(8): 2918-34, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10971634

ABSTRACT

Gluco- and mineralocorticoid receptors (GR and MR) act via common promoter elements but may exert different effects on gene regulation in various regions of the forebrain. In order to separately analyse the role of GR and MR in the regulation of neurotrophic factor genes and their receptors, we used adrenalectomy and subsequent hormone injections in the rat as a model system. Twenty-four hours after adrenalectomy rats were injected with a single dose of corticosterone (2 and 10 mg/kg), aldosterone (0.5 mg/kg) or the synthetic glucocorticoid agonist RU 28362 (4 mg/kg). Gene expression of basic fibroblast growth factor (bFGF) and its high-affinity receptors [fibroblast growth factor receptor subtypes 1-3 (FGF-R1, FGF-R2, FGF-R3)], as well as brain-derived growth factor (BDNF) and neurotrophin-3 (NT-3) was analysed at 4 h after the hormone injection in CA1-CA4 (cornus of Ammon areas of the hippocampus) and dentate gyrus of the dorsal hippocampus and in neocortex by means of in situ hybridization. We found that bFGF is regulated in CA2, CA3 and dentate gyrus by GR and MR together, and in CA1, CA4 and neocortex by GR alone. FGF-R2 expression in the hippocampus seems to be regulated only by MR, while BDNF expression appears to depend on both receptors. FGF-R1, FGF-R3 and NT-3 were only moderately affected by the hormone activation of GR and MR acting in concert or alone in the various regions. Thus, the present findings suggest that the adrenal cortical system through GR and MR participate in the control of neurotrophic factor signalling in a highly subregion- and cellular-dependent manner.


Subject(s)
Aldosterone/pharmacology , Anti-Inflammatory Agents/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Corticosterone/pharmacology , Hippocampus/physiology , Neocortex/physiology , Adrenalectomy , Aldosterone/blood , Androstanols/pharmacology , Animals , Anti-Inflammatory Agents/blood , Corticosterone/blood , Fibroblast Growth Factor 2/genetics , Gene Expression/drug effects , Hippocampus/chemistry , In Situ Hybridization , Male , Neocortex/chemistry , Neurotrophin 3/genetics , RNA Probes , RNA, Messenger/analysis , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Receptors, Fibroblast Growth Factor/genetics
14.
Behav Brain Res ; 113(1-2): 21-34, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10942029

ABSTRACT

The multiple combinations of nAChR subunits identified in central nervous structures possess distinct pharmacological and physiological properties. A growing number of data have shown that compounds interacting with neuronal nAChRs have, both in vivo and in vitro, the potential to be neuroprotective and that treatment with nAChR agonists elicit long-lasting improving of cognitive performance in a variety of behavioural tests in rats, monkeys and humans. Epidemiological and clinical studies suggested also a potential neuroprotective/trophic role of (-)-nicotine in neurodegenerative disease, such as Alzheimer's and Parkinson's disease. Taken together experimental and clinical data largely indicate a neuroprotective/trophic role of nAChR activation involving mainly alpha7 and alpha4beta2 nAChR subtypes, as evidenced using selective nAChR antagonists, and by potent nAChR agonists recently found displaying efficacy and/or larger selective affinities than (-)-nicotine for neuronal nAChR subtypes. A neurotrophic factor gene regulation by nAChR signalling has been taken into consideration as possible mechanism involved in neuroprotective/trophic effects by nAChR activation and has evidenced an involvement of the fibroblast growth factor (FGF-2) gene as a target of nAChR signalling. These findings suggested that FGF-2 could be involved, according to the FGF-2 neurotrophic functions, in nAChR mechanisms mediating the neuronal survival, trophism and plasticity.


Subject(s)
Brain/physiology , Cell Survival/physiology , Nerve Growth Factors/physiology , Neuroprotective Agents , Receptors, Nicotinic/physiology , Animals , Fibroblast Growth Factor 2/physiology , Haplorhini , Humans , Neurons/physiology , Rats
15.
Diabetes ; 49(5): 727-34, 2000 May.
Article in English | MEDLINE | ID: mdl-10905480

ABSTRACT

Previous studies have provided evidence for the transcripts of Cx43 and Cx45 within pancreatic islets. As of yet, however, it has proven difficult to unambiguously demonstrate the expression of these proteins by islet cells. We have investigated whether Cx36, a new connexin species recently identified in mammalian brain and retina, may also be expressed in pancreatic islets. Using probes that permitted the original identification of Cx36 in the central nervous system, we show that a transcript for Cx36 is clearly detectable in rat pancreatic islets. Using novel and affinity-purified polyclonal antibodies, we have found that Cx36 is actually expressed in pancreatic islets. Both in situ hybridization and immunolabeling indicated that this connexin is abundant in the centrally located insulin-producing beta-cells and is expressed much less, if at all, by the other endocrine cell types. This differential expression was further confirmed on fluorescence-activated cell sorter-purified preparations enriched in either beta- or non-beta-cells. The finding of a differential distribution of Cx36 within distinct regions of pancreatic islets creates the possibility that this connexin may provide the establishment of selective pathways of communication between the different types of endocrine cells comprising the pancreatic islet.


Subject(s)
Connexins/metabolism , Eye Proteins/metabolism , Islets of Langerhans/metabolism , Animals , Connexins/genetics , Eye Proteins/genetics , Immunologic Techniques , In Situ Hybridization , Insulin/biosynthesis , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Gap Junction delta-2 Protein
16.
Brain Res ; 865(1): 121-38, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10814742

ABSTRACT

The distribution of connexin36 (Cx36) in the adult rat brain and retina has been analysed at the protein (immunofluorescence) and mRNA (in situ hybridization) level. Cx36 immunoreactivity, consisting primarily of round or elongated puncta, is highly enriched in specific brain regions (inferior olive and the olfactory bulb), in the retina, in the anterior pituitary and in the pineal gland, in agreement with the high levels of Cx36 mRNA in the same regions. A lower density of immunoreactive puncta can be observed in several brain regions, where only scattered subpopulations of cells express Cx36 mRNA. By combining in situ hybridization for Cx36 mRNA with immunohistochemistry for a general neuronal marker (NeuN), we found that neuronal cells are responsible for the expression of Cx36 mRNA in inferior olive, cerebellum, striatum, hippocampus and cerebral cortex. Cx36 mRNA was also demonstrated in parvalbumin-containing GABAergic interneurons of cerebral cortex, striatum, hippocampus and cerebellar cortex. Analysis of developing brain further revealed that Cx36 reaches a peak of expression in the first two weeks of postnatal life, and decreases sharply during the third week. Moreover, in these early stages of postnatal development Cx36 is detectable in neuronal populations that are devoid of Cx36 mRNA at the adult stage. The developmental changes of Cx36 expression suggest a participation of this connexin in the extensive interneuronal coupling which takes place in several regions of the early postnatal brain.


Subject(s)
Brain/growth & development , Brain/metabolism , Connexins/genetics , Connexins/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Neurons/metabolism , Age Factors , Animals , Animals, Newborn , Biomarkers , Brain/cytology , Brain Mapping , Gap Junctions/metabolism , Immunohistochemistry , Male , Neurons/cytology , Nuclear Proteins/metabolism , Parvalbumins/metabolism , Pineal Gland/cytology , Pineal Gland/metabolism , Pituitary Gland/cytology , Pituitary Gland/metabolism , RNA, Messenger/metabolism , Rats , Gap Junction delta-2 Protein
17.
Brain Res Brain Res Rev ; 32(1): 72-85, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10751658

ABSTRACT

Cx36 is the first mammalian member of a novel subgroup of the connexin family, characterized by a long cytoplasmic loop, a peculiar gene structure and a preferential expression in cell types of neural origin. In the present review we summarize the evidence in favour of its predominant expression in neuronal cells in the mammalian central nervous system, such as results from experiments with specific neurotoxins and co-localization of Cx36 mRNA and a neuronal marker. We also report a detailed description of Cx36 mRNA distribution in the rat and human central nervous system by in situ hybridization and, for each brain region, we correlate the novel findings with previous morphological or functional demonstrations of neuronal gap junctions in the same area.


Subject(s)
Connexins/genetics , Eye Proteins/genetics , Gap Junctions/physiology , Synapses/physiology , Animals , Cell Communication/physiology , Gap Junctions/chemistry , Gene Expression/physiology , Humans , Mammals , Synapses/chemistry , Gap Junction delta-2 Protein
18.
J Neural Transm Suppl ; (60): 227-45, 2000.
Article in English | MEDLINE | ID: mdl-11205143

ABSTRACT

A growing number of data have shown that compounds interacting with neuronal nicotinic acetylcholine receptors (nAChRs) have, both in vivo and in vitro, the potential to be neuroprotective and that treatment with nAChR agonists elicit long-lasting improvement of cognitive performance in a variety of behavioural tests in rats, monkeys and humans. Epidemiological and clinical studies suggested also a potential neuroprotective/trophic role of (-)-nicotine in neurodegenerative disease, such as Alzheimer's disease and Parkinson's disease. This neuroprotective/trophic role of nAChR activation has been mainly mediated by alpha7 and alpha4beta2 nAChR subtypes, as evidenced using selective nAChR antagonists, and by potent nAChR agonists recently found displaying efficacy and/or larger selective affinities than (-)-nicotine for neuronal nAChR subtypes. A neurotrophic factor gene regulation by nAChR signalling has been taken into consideration as a possible mechanism involved in neuroprotective/trophic effects of nAChR activation and has given evidence that the fibroblast growth factor (FGF-2) gene is a target for nAChR signalling. These findings suggested that FGF-2 could be involved, in view of its neurotrophic functions, in nAChR mechanisms mediating neuronal survival, trophism and plasticity.


Subject(s)
Nerve Growth Factors/pharmacology , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Animals , Cell Survival/drug effects , Fibroblast Growth Factor 2/drug effects , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation/drug effects , Humans , Nerve Growth Factors/metabolism , Neurodegenerative Diseases/metabolism , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/metabolism , Signal Transduction/drug effects
19.
J Neurosci Res ; 57(5): 740-52, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10462698

ABSTRACT

Rat connexin-36 (Cx36) is the first gap junction protein shown to be expressed predominantly in neuronal cells of the mammalian central nervous system. As a prerequisite for studies devoted to the investigation of the possible role of this connexin in human neurological diseases, we report the cloning and sequencing of the human Cx36 gene, its chromosomal localization, and its pattern of expression in the human brain analyzed by radioactive in situ hybridization. The determination of the human gene sequence revealed that the coding sequence of Cx36 is highly conserved (98% identity at the protein level with the mouse and rat Cx36 and 80% with the ortholog perch and skate Cx35), and that the gene structure is that typical of the Cx35/36 subgroup observed in the other species (presence of a single intron located within the coding region, 71 bp after the translation initiation site). The distribution of Cx36 in several regions of the human central nervous system is similar to that previously observed in rat brain. The most intense signal among the cerebral areas examined by in situ hybridization was observed in the inferior olivary complex, both in principal and accessory nuclei. A moderate labeling was also observed in several myelencephalic nuclei, in specific cells of the the cerebellar cortex, in a relatively large subpopulation of cells in the cerebral cortex, in the hilus of the dentate gyrus, and in the strata radiatum and oriens of hippocampal subfields. Moreover, labeled cells were revealed in all the lamina of the spinal cord gray matter. The chromosomal localization of the human Cx36 gene was determined by fluorescence in situ hybridization. The results allowed assignment of the gene to band 15q14, thus making it a possible candidate gene for a form of familial epilepsy previously linked to the same chromosomal band. The knowledge of the human Cx36 gene sequence, of its chromosomal localization, and of its pattern of expression opens new avenues for the analysis of its possible involvement in human genetic and acquired neuropathology.


Subject(s)
Brain/metabolism , Chromosomes, Human, Pair 15 , Connexins/genetics , Eye Proteins/genetics , Adolescent , Adult , Amino Acid Sequence , Animals , Chromosome Mapping , Cloning, Molecular , Connexins/analysis , Connexins/metabolism , Eye Proteins/analysis , Eye Proteins/metabolism , Female , Genomic Library , Humans , In Situ Hybridization, Fluorescence , Introns , Male , Mice , Middle Aged , Molecular Sequence Data , Organ Specificity , Peptide Chain Initiation, Translational , Perches , Polymerase Chain Reaction , Rats , Sequence Alignment , Sequence Homology, Amino Acid , Skates, Fish , Spinal Cord/metabolism , Gap Junction delta-2 Protein
20.
Neurochem Res ; 24(5): 709-14, 1999 May.
Article in English | MEDLINE | ID: mdl-10344602

ABSTRACT

GFAPbeta mRNA is an alternative transcript of the glial fibrillary acidic protein (GFAP) gene, whose transcriptional start site is located 169 nucleotides upstream to the classical GFAPalpha mRNA. By an RT-PCR method with primers on separate exons, we were able to confirm the presence of GFAP transcripts with a longer 5' untranslated region in all the examined areas of rat brain and in primary cultures of astroglial cells. Northern blot analysis, using an oligoprobe specific for the 5' region of GFAPbeta, revealed a single hybridization band of 2.9 kb in all the brain regions examined and in primary cultures of astroglial cells. The availability of the quantitative Northern blot assay allowed further studies on the regulation of GFAPbeta expression in vivo. Since it is well-known that neuronal brain injury is one of the most powerful inducers of GFAP, we examined the expression of GFAPalpha and beta after a neurotoxic lesion in the rat hippocampus. Results obtained show a parallel increase in both GFAP transcripts with an identical time-course, suggesting that regulatory regions of the gene influence in similar way the rate of transcription at the two different start sites (alpha and beta) or that a similar post-transcriptional mechanism is involved in regulating both mRNA isoforms.


Subject(s)
Brain/metabolism , Gene Expression , Glial Fibrillary Acidic Protein/genetics , Ibotenic Acid/pharmacology , Neurons/drug effects , RNA, Messenger/analysis , Animals , Astrocytes/chemistry , Blotting, Northern , Brain Chemistry , Cells, Cultured , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/drug effects , Neurons/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...