Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Cell Mol Neurobiol ; 30(8): 1335-42, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21080057

ABSTRACT

Regulated exocytosis of neurotransmitter- and hormone-containing vesicles underpins neuronal and hormonal communication and relies on a well-orchestrated series of molecular interactions. This in part involves the upstream formation of a complex of SNAREs and associated proteins leading to the eventual fusion of the vesicle membrane with the plasma membrane, a process that enables content release. Although the role of lipids in exocytosis is intuitive, it has long been overlooked at least compared to the extensive work on SNAREs. Here, we will present the latest advances in this rapidly developing field revealing that lipids actually play an active role in exocytosis by focusing on cholesterol, 3'-phosphorylated phosphoinositides and phosphatidic acid.


Subject(s)
Exocytosis , Lipid Metabolism , Animals , Cholesterol/metabolism , Humans , Models, Biological , Phosphatidic Acids/metabolism , Phosphatidylinositols/metabolism
2.
Cell Mol Life Sci ; 62(3): 377-85, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15723172

ABSTRACT

Catestatin (bCGA(344-364)), an endogenous peptide of bovine chromogranin A, was initially characterized for its effect on the inhibition of catecholamine release from chromaffin cells. Catestatin and its active domain (bCGA(344-358)) were identified in chromaffin cells and in secretion medium. The present study identified a potent antimicrobial activity of bCGA(344-358) in the lowmicromolar range against bacteria, fungi and yeasts, without showing any haemolytic activity. Confocal laser microscopy demonstrated penetration of the rhodaminated peptide into the cell membranes of fungi and yeasts and its intracellular accumulation. Time-lapse videomicroscopy showed arrest of fungal growth upon penetration of the labelled peptide into a fungal filament. We identified several catestatin-containing fragments in the stimulated secretion medium of human polymorphonuclear neutrophils, suggesting the N-terminal sequence of catestatin (bCGA(344-358)) (named cateslytin) as a novel component of innate immunity.


Subject(s)
Anti-Infective Agents/pharmacology , Catecholamines/chemistry , Chromogranins/chemistry , Chromogranins/pharmacology , Peptide Fragments/pharmacology , Amino Acid Sequence , Animals , Anti-Infective Agents/chemistry , Bacteria/drug effects , Cattle , Chromogranin A , Fungi/drug effects , Humans , Microbial Sensitivity Tests , Molecular Sequence Data , Peptide Fragments/chemistry , Time Factors , Yeasts/drug effects
3.
Ann N Y Acad Sci ; 971: 359-61, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12438152

ABSTRACT

The antifungal peptide named chromofungin is the most active vasostatin-I-derived peptide, corresponding to the sequence 47-66 of chromogranin A. (1)H-NMR analysis revealed that it adopts a helical structure. The mechanism implicated in the interaction of chromofungin with fungi and yeast cells was studied by penetration of monolayers and confocal laser microscopy. Chromofungin is able to interact with the cell wall, to cross the plasma membrane, to accumulate in the microorganism, and to inhibit calcineurin activity.


Subject(s)
Chromogranins/chemistry , Chromogranins/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptides/chemistry , Alternaria/metabolism , Antifungal Agents/chemistry , Aspergillus fumigatus/metabolism , Calcineurin Inhibitors , Calcium-Binding Proteins/metabolism , Calreticulin , Candida albicans/metabolism , Cell Membrane/metabolism , Chromogranin A , Chromogranins/metabolism , Humans , Magnetic Resonance Spectroscopy , Peptide Fragments/metabolism , Peptides/pharmacology , Protein Structure, Tertiary , Ribonucleoproteins/metabolism
4.
Proc Natl Acad Sci U S A ; 98(26): 15300-5, 2001 Dec 18.
Article in English | MEDLINE | ID: mdl-11752468

ABSTRACT

Phosphatidic acid produced by phospholipase D (PLD) as a result of signaling activity is thought to play a role in membrane vesicle trafficking, either as an intracellular messenger or as a cone-shaped lipid that promotes membrane fusion. We recently described that, in neuroendocrine cells, plasma membrane-associated PLD1 operates at a stage of Ca(2+)-dependent exocytosis subsequent to cytoskeletal-mediated recruitment of secretory granules to exocytotic sites. We show here that PLD1 also plays a crucial role in neurotransmitter release. Using purified rat brain synaptosomes subjected to hypotonic lysis and centrifugation, we found that PLD1 is associated with the particulate fraction containing the plasma membrane. Immunostaining of rat cerebellar granule cells confirmed localization of PLD1 at the neuronal plasma membrane in zones specialized for neurotransmitter release (axonal neurites, varicosities, and growth cone-like structures). To determine the potential involvement of PLD1 in neurotransmitter release, we microinjected catalytically inactive PLD1(K898R) into Aplysia neurons and analyzed its effects on evoked acetylcholine (ACh) release. PLD1(K898R) produced a fast and potent dose-dependent inhibition of ACh release. By analyzing paired-pulse facilitation and postsynaptic responses evoked by high-frequency stimulations, we found that the exocytotic inhibition caused by PLD1(K898R) was not the result of an alteration in stimulus-secretion coupling or in vesicular trafficking. Analysis of the fluctuations in amplitude of the postsynaptic responses revealed that the PLD1(K898R) blocked ACh release by reducing the number of active presynaptic-releasing sites. Our results provide evidence that PLD1 plays a major role in neurotransmission, most likely by controlling the fusogenic status of presynaptic release sites.


Subject(s)
Neurotransmitter Agents/metabolism , Phospholipase D/metabolism , Acetylcholine/metabolism , Animals , Aplysia , Catalysis , Cells, Cultured , Fluorescent Antibody Technique , Microscopy, Confocal , Neurons/enzymology , Neurons/metabolism , Rats , Rats, Wistar , Subcellular Fractions/enzymology , Synapses/metabolism
5.
Brain Res Mol Brain Res ; 96(1-2): 1-13, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11731003

ABSTRACT

In neurones, the morphological complexity of the dendritic tree requires regulated growth and the appropriate targeting of membrane components. Accurate delivery of specific supplies depends on the translocation and fusion of transport vesicles. Vesicle SNAREs (soluble N-ethylmaleimide sensitive factor attachment protein receptors) and target membrane SNAREs play a central role in the correct execution of fusion events, and mediate interactions with molecules that endow the system with appropriate regulation. Synaptotagmins, a family of Ca(2+)-sensor proteins that includes neurone-specific members involved in regulating neurotransmitter exocytosis, are among the molecules that can tune the fusion mechanism. Using immunocytochemistry, confocal and electron microscopy, the localisation of synaptotagmin I in the dendrites of cultured rat hypothalamic neurones was demonstrated. Synaptotagmin labelling is concentrated at dendritic branch points, and in microprocesses. Following depolarisation, the N-terminal domain of synaptotagmin was detected at the extracellular surface of the dendritic plasma membrane. The insertion of synaptotagmin in the plasma membrane was elicited by L-type Ca(2+) channel activation and by mobilisation of the internal ryanodine-sensitive Ca(2+)stores. Furthermore, the localisation of L-type Ca(2+) channels and of ryanodine receptors, relative to the localisation of synaptotagmin in dendrites, suggests that both Ca(2+) entry and intracellular Ca(2+) stores may contribute to the fusion of dendritic transport vesicles with the membrane. Fusion is likely to involve SNAP-25 and syntaxin 1 as both proteins were also identified in dendrites. Taken together these results suggest a putative regulatory role of synaptotagmins in the membrane fusion events that contribute to shaping the dendritic tree during development.


Subject(s)
Calcium-Binding Proteins , Calcium/metabolism , Cell Membrane/metabolism , Dendrites/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Vesicular Transport Proteins , Animals , Calcium Channels, L-Type/analysis , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cells, Cultured , Dendrites/chemistry , Dendrites/ultrastructure , Hypothalamus/cytology , Hypothalamus/growth & development , Membrane Fusion/physiology , Membrane Glycoproteins/analysis , Membrane Potentials/physiology , Membrane Proteins/analysis , Microscopy, Electron , Nerve Tissue Proteins/analysis , Neurons/metabolism , Neurons/ultrastructure , Rats , Rats, Wistar , Ryanodine Receptor Calcium Release Channel/analysis , SNARE Proteins , Synaptotagmin I , Synaptotagmins , Syntaxin 1
6.
Biochem J ; 358(Pt 2): 295-303, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11513727

ABSTRACT

The multisubunit leucine-rich glycoprotein (GP) Ib-IX-V complex mediates von Willebrand factor-dependent platelet adhesion at sites of blood-vessel injury. Molecular defects of this receptor are reported to cause the Bernard-Soulier haemorrhagic disorder. To gain insight into the mechanisms controlling expression of normal and defective receptors, we performed pulse-chase metabolic studies and detailed analysis of intracellular processing in GPIb-IX-transfected Chinese-hamster ovary cells. In the native complex, after early subunit association, sugars N-linked to the three subunits are trimmed and sialylated in the Golgi compartment and GPIbalpha undergoes extensive O-glycosylation. Surface biotinylation during chase demonstrated that only fully processed complexes reach the cell surface. Tunicamycin treatment revealed that early N-glycosylation is not required for O-glycosylation of GPIbalpha and surface expression of the complex. Biosynthetic studies were then performed on a Bernard-Soulier variant based on previous description of abnormal GPIbalpha size and decreased surface expression. The mutant complex associated normally, but displayed defective processing of its N-linked sugars and abnormal O-glycosylation of GPIbalpha. Confocal immunofluorescence microscopy revealed that the mutant complexes could reach the cell surface but also accumulated intracellularly, while use of compartment specific markers showed strong co-localization in the endoplasmic reticulum (ER) and ER-to-Golgi intermediate compartments ('ERGIC') and only slight labelling of the cis-Golgi. Blockade before the Golgi was confirmed by brefeldin A treatment, which restored O-glycosylation and processing of N-linked sugars. The present study has shown that transfer from the ER to the Golgi represents an important step for controlling post-translational processing and surface expression of normal GPIb-IX-V complex.


Subject(s)
Platelet Glycoprotein GPIb-IX Complex/biosynthesis , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Processing, Post-Translational , Animals , CHO Cells , Carbohydrate Metabolism , Cell Membrane/metabolism , Cricetinae , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Glycosylation , Golgi Apparatus/metabolism , Kinetics , Leucine/genetics , Microscopy, Confocal , Mutation , Platelet Glycoprotein GPIb-IX Complex/genetics , Protein Subunits , Protein Transport
7.
J Biol Chem ; 276(38): 35875-82, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11451958

ABSTRACT

Vasostatin-I, the natural fragment of chromogranin A-(1-76), is a neuropeptide able to kill a large variety of fungi and yeast cells in the micromolar range. We have examined the antifungal properties of synthetic vasostatin-I-related peptides. The most active shortest peptide, named chromofungin, corresponds to the sequence Arg(47)-Leu(66). Extensive (1)H NMR analysis revealed that it adopts a helical structure. The biophysical mechanism implicated in the interaction of chromofungin with fungi and yeast cells was studied, showing the penetration of this peptide with different lipid monolayers. In order to examine thoroughly the antifungal activity of chromofungin, confocal laser microscopy was used to demonstrate the ability of the rhodamine-labeled peptide to interact with the fungal cell wall, to cross the plasma membrane, and to accumulate in Aspergillus fumigatus, Alternaria brassicola, and Candida albicans. Our present data reveal that chromofungin inhibits calcineurin activity, extending a previous observation that the N-terminal region of chromogranin A interacts with calmodulin in the presence of calcium. Therefore, the destabilization of fungal wall and plasma membrane, together with the possible intracellular inhibition of calmodulin-dependent enzymes, is likely to represent the mechanism by which vasostatin-I and chromofungin exert antifungal activity.


Subject(s)
Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Chromogranins/chemistry , Chromogranins/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Amino Acid Sequence , Chromogranin A , Microbial Sensitivity Tests , Microscopy, Confocal , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Sequence Homology, Amino Acid , Structure-Activity Relationship
8.
EMBO J ; 20(10): 2424-34, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11350931

ABSTRACT

Phospholipase D (PLD) has been proposed to mediate cytoskeletal remodeling and vesicular trafficking along the secretory pathway. We recently described the activation of an ADP ribosylation factor-regulated PLD at the plasma membrane of chromaffin cells undergoing secretagogue-stimulated exocytosis. We show here that the isoform involved is PLD1b, and, using a real-time assay for individual cells, that PLD activation and exocytosis are closely correlated. Moreover, overexpressed PLD1, but not PLD2, increases stimulated exocytosis in a phosphatidylinositol 4,5-bisphosphate-dependent manner, whereas catalytically inactive PLD1 inhibits it. These results provide the first direct evidence that PLD1 is an important component of the exocytotic machinery in neuroendocrine cells.


Subject(s)
Chromaffin Cells/enzymology , Exocytosis/physiology , Phospholipase D/metabolism , Actins/metabolism , Animals , Catalysis , Cattle , Cells, Cultured , Chromaffin Cells/cytology , Chromaffin Cells/physiology , Enzyme Inhibitors/pharmacology , Intracellular Fluid/enzymology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Neurosecretory Systems/cytology , PC12 Cells , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase D/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
9.
J Biol Chem ; 276(16): 13113-20, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11124958

ABSTRACT

The neurotoxic effects of activated microglia in neurodegenerative diseases are well established. We recently provided evidence that chromogranin A (CGA), a multifunctional protein localized in dystrophic neurites and in senile plaques, induces an activated phenotype and secretion of neurotoxins by rat microglia in culture. In the present study, we focused on the mechanisms underlying neuronal degeneration triggered by CGA-activated microglia. We found that neuronal death exhibits apoptotic features, characterized by the externalization of phosphatidylserine and the fragmentation of DNA. Microglial neurotoxins markedly stimulate the phosphorylation and activity of neuronal p38 mitogen-activated protein kinase and provoke the release of mitochondrial cytochrome c, which precedes apoptosis. Inhibition of p38 kinase with SB 203580 partially protects neurons from death induced by CGA-activated microglia. Furthermore, neurons are also protected by Fas-Fc, which antagonizes the interactions between the death receptor Fas and its ligand FasL and by cell-permeable peptides that inhibit caspases 8 and 3. Thus, CGA triggers the release of microglial neurotoxins that mobilize several death-signaling pathways in neurons. Our results further support the idea that CGA, which is up-regulated in many neuropathologies, represents a potent endogeneous inflammatory factor possibly responsible for neuronal degeneration.


Subject(s)
Apoptosis/physiology , Chromogranins/pharmacology , Microglia/physiology , Neurons/physiology , Animals , Apoptosis/drug effects , Cattle , Cell Death/drug effects , Cells, Cultured , Chromaffin Granules/chemistry , Chromogranin A , Chromogranins/isolation & purification , Coculture Techniques , Culture Media, Conditioned , Cytochrome c Group/analysis , DNA Fragmentation , Enzyme Inhibitors/pharmacology , Fas Ligand Protein , Imidazoles/pharmacology , Kinetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/physiology , Microglia/cytology , Microglia/drug effects , Mitochondria/physiology , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Neurons/drug effects , Neurotoxins , Phosphatidylserines/metabolism , Pyridines/pharmacology , Rats , Time Factors , fas Receptor/immunology , fas Receptor/physiology , p38 Mitogen-Activated Protein Kinases
10.
J Neurocytol ; 30(9-10): 789-800, 2001.
Article in English | MEDLINE | ID: mdl-12165670

ABSTRACT

SNAP-25 (Synaptosomal Associated Protein of 25 kDa), in association with two other SNARE (soluble NSF attachment protein receptor) proteins, syntaxin and Vesicle Associated Membrane Protein, VAMP, is implicated in regulated and constitutive exocytosis in neurones and neuroendocrine cells. Our previous studies have shown that it is expressed more by noradrenergic than adrenergic chromaffin cells in the rat adrenal gland. Since certain hormones under hypophyseal control play an essential role in determining chromaffin cell phenotype, the present study examined the effect of hypophysectomy on SNAP-25 expression. Hypophysectomy was found by immunoblotting and RT-PCR analysis to increase adrenal gland SNAP-25, syntaxin-1 and VAMP-2 levels, without modifying the relative expression of SNAP-25 isoforms: immunocytochemistry showed a dramatic increase in SNAP-25 expression in former adrenergic chromaffin cells. Since adrenal glucocorticoids are considerably reduced by hypophysectomy, the effect of corticosterone replacement therapy was investigated. This did not change levels of SNAP-25, syntaxin-1 or VAMP-2. SNARE expression was also unmodified in pheochromocytoma cells treated with a synthetic glucocorticoid. In contrast, subcutaneous injection of hypophysectomized rats with thyroid hormone decreased adrenal SNAP-25, demonstrating the potential importance of the pituitary-thyroid axis. The current data thus demonstrate that the hypophysis exerts an inhibitory control on adrenal gland SNARE proteins. They suggest that glucocorticoids are unlikely to be directly responsible for this but provide evidence that thyroid hormones are implicated in this phenomenon. The putative role of hormonal regulation on SNARE function is discussed.


Subject(s)
Adrenal Medulla/metabolism , Chromaffin Cells/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Pituitary Gland/metabolism , Pituitary-Adrenal System/metabolism , Vesicular Transport Proteins , Adrenal Medulla/cytology , Adrenocorticotropic Hormone/metabolism , Adrenocorticotropic Hormone/pharmacology , Animals , Antigens, Surface/metabolism , Catecholamines/biosynthesis , Chromaffin Cells/cytology , Dexamethasone/pharmacology , Glucocorticoids/blood , Hypophysectomy , Immunohistochemistry , Male , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , PC12 Cells , Phenylethanolamine N-Methyltransferase/metabolism , Pituitary-Adrenal System/cytology , R-SNARE Proteins , RNA, Messenger/metabolism , Rats , Rats, Wistar , SNARE Proteins , Synaptosomal-Associated Protein 25 , Syntaxin 1 , Testosterone/metabolism , Testosterone/pharmacology , Thyroid Hormones/metabolism , Thyroid Hormones/pharmacology
12.
J Comp Neurol ; 426(4): 549-60, 2000 Oct 30.
Article in English | MEDLINE | ID: mdl-11027398

ABSTRACT

gamma-Hydroxybutyrate (GHB) is an endogenous metabolite of gamma-aminobutyric acid (GABA), which is synthesized in the neuronal compartment of the central nervous system. This substance possesses several properties that support its role as a neurotransmitter/neuromodulator in brain. In particular, it is synthesized by a specific pathway that transforms GABA into succinic semialdehyde via GABA-T activity; then succinic semialdehyde is converted into GHB by a specific succinic semialdehyde reductase (SSR). The last enzyme is considered as a marker for neurons that synthesize GHB. This compound binds in brain to receptors whose distribution, ontogenesis, kinetics, and pharmacology are specific. Endogenous GHB, but also GHB exogenously administered to rats, participate in the regulation of dopaminergic activity of the nigrostriatal pathway. To investigate the distribution of GHB neurons in this pathway and the anatomic relationships between dopaminergic and GHB neurons, immunocytochemical identification of dopamine, GABA, and GHB neurons was carried out in the substantia nigra and striatum of the rat. The following markers for these neurons were used: anti-tyrosine hydroxylase (TH) antibodies for dopamine neurons, anti-glutamate decarboxylase (GAD) antibodies for GABA neurons, and anti-succinic semialdehyde reductase (SSR) antibodies for GHB neurons. GABA neurons were studied because GAD and SSR co-exist frequently in the same neuron, and GABA alone also exerts its own regulatory effects on dopaminergic neurons. This study reveals the co-existence of GAD/SSR and GAD/SSR/TH in numerous neurons of the substantia nigra. However, some neurons appear to be only GAD or SSR positive. In the striatum, TH-positive terminals surround many GHB neurons. GAD innervation is abundant in close contact with unlabeled neurons in the caudate-putamen, whereas distinct SSR-positive punctuates are also present. The existence of SSR-reactive synapses and neurons was confirmed in the striatum at the electron microscopic level. On the basis of these results, a clear anatomo-functional relationship between GHB and dopamine networks cannot be defined; however, we propose the modulation by GHB of striatal intrinsic neurons that could then interfere with the presynaptic control of dopaminergic activity.


Subject(s)
Corpus Striatum/metabolism , Dopamine/biosynthesis , Rats/metabolism , Sodium Oxybate/metabolism , Substantia Nigra/enzymology , gamma-Aminobutyric Acid/biosynthesis , Animals , Corpus Striatum/cytology , Immunohistochemistry , Male , Neurons/enzymology , Rats, Wistar , Substantia Nigra/cytology , Tissue Distribution
13.
FEBS Lett ; 480(2-3): 184-8, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-11034325

ABSTRACT

Phosducin and related proteins have been identified as ubiquitous regulators of signalling mediated by betagamma subunits of trimeric G proteins. To explore a role for phosducin in regulated exocytosis, we have examined the distribution and putative function of phosducin-like protein (PhLP) in adrenal medullary chromaffin cells. The full-length cDNA encoding the short splice variant of PhLP (PhLPs) was cloned from cultured chromaffin cells. Native PhLPs was found associated with plasma membranes and detected in the subplasmalemmal area of resting chromaffin cells by confocal immunofluorescence analysis. Stimulation with secretagogues triggered a massive redistribution of PhLPs into the cytoplasm. When microinjected into individual chromaffin cells, recombinant PhLPs inhibited catecholamine secretion evoked by a depolarizing concentration of K+ without affecting calcium mobilization. Thus, PhLPs may participate directly in the regulation of calcium-evoked exocytosis.


Subject(s)
Carrier Proteins/physiology , Chromaffin Cells/physiology , Exocytosis/physiology , GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , Heterotrimeric GTP-Binding Proteins/metabolism , Nerve Tissue Proteins/physiology , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane , Cells, Cultured , Chromaffin Cells/cytology , Chromaffin Cells/metabolism , Cytosol/metabolism , DNA, Complementary , Humans , Molecular Chaperones , Molecular Sequence Data , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Transport , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology
14.
J Neurochem ; 75(4): 1511-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10987831

ABSTRACT

Dynamin proteins have been implicated in many aspects of endocytosis, including clathrin-mediated endocytosis, internalization of caveolae, synaptic vesicle recycling, and, more recently, vesicular trafficking to and from the Golgi complex. To provide further insight into the function(s) of dynamin in neuroendocrine cells, we have examined its intracellular distribution in cultured chromaffin cells by subcellular fractionation, immunoreplica analysis, and confocal immunofluorescence. We found that dynamin, presumably the dynamin-2 isoform, is associated specifically with the membrane of purified secretory chromaffin granules. Oligomerization state analysis by sucrose density velocity gradients indicated that the granule-associated dynamin is in a monomeric form. Immunoprecipitation experiments coupled to double-labeling immunofluorescence cytochemistry revealed that the granular dynamin is associated with a syntaxin component that is not involved in the granule-bound SNARE complex. The possibility that dynamin participates in the coupling of the exocytotic and endocytotic reaction through the building of a granular membrane subset of proteins is discussed.


Subject(s)
Adrenal Glands/metabolism , Chromaffin Cells/metabolism , Chromaffin Granules/metabolism , GTP Phosphohydrolases/metabolism , Membrane Proteins/metabolism , Adrenal Glands/cytology , Animals , Cattle , Cells, Cultured , Chelating Agents/pharmacology , Chromaffin Cells/cytology , Chromaffin Granules/chemistry , Detergents/chemistry , Dimerization , Dynamin I , Dynamins , Fluorescent Antibody Technique , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Precipitin Tests , Protein Binding/drug effects , Qa-SNARE Proteins
15.
Biochimie ; 82(4): 365-73, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10865124

ABSTRACT

Catecholamine secretion from chromaffin cells has been used for a long time as a general model to study exocytosis of large dense core secretory granules. Permeabilization and microinjection techniques have brought the possibility to dissect at the molecular level the multi-protein machinery involved in this complex physiological process. Regulated exocytosis comprises distinct and sequential steps including the priming of secretory granules, the formation of a docking complex between granules and the plasma membrane and the subsequent fusion of the granule with the plasma membrane. Key proteins involved in the exocytotic machinery have been identified. For instance, SNAREs which participate in the docking events in most intracellular transport steps along the secretory pathway, play a role in exocytosis in both neuronal and endocrine cells. However, in contrast to intracellular transport processes for which the highest fusion efficiency is required after correct targeting of the vesicles, the number of exocytotic events in activated secretory cells needs to be tightly controlled. We describe here the multistep control exerted by heterotrimeric and monomeric G proteins on the progression of secretory granules from docking to fusion and the molecular nature of some of their downstream effectors in neuroendocrine chromaffin cells.


Subject(s)
Chromaffin Cells/physiology , Exocytosis/physiology , Heterotrimeric GTP-Binding Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Actins/physiology , Actins/ultrastructure , Animals , Chromaffin Granules/physiology , Membrane Fusion , Nerve Tissue Proteins/metabolism , Signal Transduction
16.
J Cell Sci ; 112 ( Pt 22): 4143-50, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10547373

ABSTRACT

Besides having a role in signal transduction some trimeric G-proteins may be involved in a late stage of exocytosis. Using immunocytochemistry and confocal microscopy we found that Gi(3)-protein resides mainly in the plasma membrane, whereas Gi(1/2-)protein is preferentially associated with secretory granules. To study the function of trimeric Gi(3)- and Gi(1/2)-proteins, secretory responses in single rat melanotrophs were monitored by patch-clamp membrane capacitance measurements. We report here that mastoparan, an activator of trimeric G-proteins, enhances calcium-induced secretory activity in rat melanotrophs. The introduction of synthetic peptides corresponding to the C-terminal domain of the (&agr;)-subunit of Gi(3)- and Gi(1/2)-proteins indicated that Gi(3 )peptide specifically blocked the mastoparan-stimulated secretory activity, which indicates an involvement of a trimeric Gi(3)-protein in mastoparan-stimulated secretory activity. Flash photolysis of caged Ca(2+)-elicited biphasic capacitance increases consisting of a fast and a slower component. Injection of anti-Gi(3) antibodies selectively inhibited the slow but not the fast component of secretory activity in rat melanotrophs. We propose that the plasma membrane-bound Gi(3)-protein may be involved in regulated secretion by specifically controlling the slower kinetic component of exocytosis.


Subject(s)
Exocytosis/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Pituitary Gland/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Calcium/metabolism , Calcium/pharmacology , Cell Membrane/drug effects , Cell Membrane/physiology , Electric Conductivity , Exocytosis/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/immunology , GTP-Binding Protein alpha Subunits, Gi-Go/pharmacology , Intercellular Signaling Peptides and Proteins , Microscopy, Fluorescence , Peptides , Pituitary Gland/cytology , Pituitary Gland/drug effects , Rats , Wasp Venoms/pharmacology
17.
J Cell Sci ; 112 ( Pt 24): 4763-71, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10574723

ABSTRACT

The Rho GTPase family, including Rho, Rac and Cdc42 proteins, is implicated in various cell functions requiring the reorganization of actin-based structures. In secretory cells, cytoskeletal rearrangements are a prerequisite for exocytosis. We previously described that, in chromaffin cells, the trimeric granule-bound Go protein controls peripheral actin and prevents exocytosis in resting cells through the regulation of RhoA. To provide further insight into the function of Rho proteins in exocytosis, we focus here on their intracellular distribution in chromaffin cells. By confocal immunofluorescence analysis, we found that Rac1 and Cdc42 are exclusively localized in the subplasmalemmal region in both resting and nicotine-stimulated cells. In contrast, RhoA is associated with the membrane of secretory granules. We then investigated the effects of clostridial toxins, which differentially impair the function of Rho GTPases, on the subplasmalemmal actin network and catecholamine secretion. Clostridium difficile toxin B, which inactivates Rho, Rac and Cdc42, markedly altered the distribution of peripheral actin filaments. Neither Clostridium botulinum C3 toxin, which selectively ADP-ribosylates Rho, nor Clostridium sordellii lethal toxin, which inactivates Rac, affected cortical actin, suggesting that Cdc42 plays a specific role in the organization of subplasmalemmal actin. Indeed, toxin B strongly reduced secretagogue-evoked catecholamine release. This effect on secretion was not observed in cells having their actin cytoskeleton depolymerized by cytochalasin E or Clostridium botulinum C2 toxin, suggesting that the inhibition of secretion by toxin B is entirely linked to the disorganization of actin. C. sordellii lethal toxin also inhibited catecholamine secretion, but this effect was not related to the actin cytoskeleton as seen in cells pretreated with cytochalasin E or C2 toxin. In contrast, C3 exoenzyme did not affect secretion. We propose that Cdc42 plays an active role in exocytosis by coupling the actin cytoskeleton to the sequential steps underlying membrane trafficking at the site of exocytosis.


Subject(s)
Adrenal Glands/enzymology , Calcium/metabolism , Chromaffin Cells/enzymology , Exocytosis , GTP Phosphohydrolases/metabolism , Actins/metabolism , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Animals , Botulinum Toxins/pharmacology , Catecholamines/metabolism , Cattle , Chromaffin Cells/drug effects , Chromaffin Cells/metabolism , Immunohistochemistry , Subcellular Fractions/enzymology
18.
Glia ; 27(2): 181-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10417817

ABSTRACT

Astrocytes release glutamate and aspartate in response to elevated intracellular calcium levels, and it has been proposed that this occurs by a vesicular release mechanism, in which SNARE proteins are implicated. Although syntaxin, synaptobrevin, and cellubrevin have been shown to be expressed by cultured astrocytes, SNAP-25 has not been detected. By using immunocytochemical, immunoblotting, and polymerase chain reaction techniques, the present study demonstrates that SNAP-23, an analogue of SNAP-25, is expressed by astrocytes both in culture and in rat cerebellum. These findings provide additional evidence that astrocytes release excitatory amino acids by a vesicular mechanism involving SNARE proteins. SNAP-23 and also syntaxin 1 and cellubrevin were found to be expressed in glial precursor cells, oligodendrocytes, and microglia. These data suggest that the t-SNAREs SNAP-23 and syntaxin 1 and the v-SNARE cellubrevin participate in general membrane insertion mechanisms involved in diverse glial cell functions such as secretion, phagocytosis, and myelinogenesis.


Subject(s)
Antigens, Surface/biosynthesis , Carrier Proteins/biosynthesis , Membrane Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neuroglia/metabolism , Animals , Animals, Newborn , Antigens, Surface/genetics , Astrocytes/metabolism , Blotting, Western , Carrier Proteins/genetics , Cell Membrane/metabolism , Cell Membrane/physiology , Cells, Cultured , Fluorescent Antibody Technique, Indirect , Membrane Proteins/genetics , Microglia/metabolism , Nerve Tissue Proteins/genetics , Oligodendroglia/metabolism , Organ Specificity , Polymerase Chain Reaction , Qb-SNARE Proteins , Qc-SNARE Proteins , Rats , Rats, Wistar , Stem Cells/metabolism , Synaptosomal-Associated Protein 25 , Syntaxin 1 , Vesicle-Associated Membrane Protein 3
19.
J Neurochem ; 73(2): 791-801, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10428077

ABSTRACT

Four recombinant human M1 (hM1) muscarinic acetylcholine receptors (mAChRs) combining several modifications were designed and overexpressed in HEK293 cells. Three different fluorescent chimera were obtained through fusion of the receptor N terminus with enhanced green fluorescent protein (EGFP), potential glycosylation sites and a large part of the third intracellular (i3) loop were deleted, a hexahistidine tag sequence was introduced at the receptor C terminus, and, finally, a FLAG epitope was either fused at the receptor N terminus or inserted into its shortened i3 loop. High expression levels and ligand binding properties similar to those of the wild-type hM1 receptor together with confocal microscopy imaging demonstrated that the recombinant proteins were correctly folded and targeted to the plasma membrane, provided that a signal peptide was added to the N-terminal domain of the fusion proteins. Their functional properties were examined through McN-A-343-evoked Ca2+ release. Despite the numerous modifications introduced within the hM1 sequence, all receptors retained nearly normal abilities (EC50 values) to mediate the Ca2+ response, although reduced amplitudes (Emax values) were obtained for the i3-shortened constructs. Owing to the bright intrinsic fluorescence of the EGFP-fused receptors, their detection, quantitation, and visualization as well as the selection of cells with highest expression were straightforward. Moreover, the presence of the different epitopes was confirmed by immunocytochemistry. Altogether, this work demonstrates that these EGFP- and epitope-fused hM1 receptors are valuable tools for further functional, biochemical, and structural studies of muscarinic receptors.


Subject(s)
Epitopes/genetics , Indicators and Reagents , Luminescent Proteins , Receptors, Muscarinic/genetics , (4-(m-Chlorophenylcarbamoyloxy)-2-butynyl)trimethylammonium Chloride/pharmacology , Atropine/pharmacology , Binding, Competitive , Calcium/analysis , Cells, Cultured , DNA Primers , Flow Cytometry , Gene Expression , Green Fluorescent Proteins , Histidine , Humans , Kidney/cytology , Microscopy, Confocal , Muscarinic Antagonists/pharmacology , Mutagenesis, Site-Directed , Piperidines/pharmacology , Pirenzepine/pharmacology , Radioligand Assay/methods , Receptor, Muscarinic M1 , Receptors, Muscarinic/analysis , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Tritium
20.
J Soc Biol ; 193(6): 451-6, 1999.
Article in French | MEDLINE | ID: mdl-10783703

ABSTRACT

In neuroendocrine cells, regulated exocytosis is a multistep process that comprises the recruitment and priming of secretory granules, their docking to the exocytotic sites, and the subsequent fusion of granules with the plasma membrane leading to the release of secretory products into the extracellular space. Using bacterial toxins which specially inactivate subsets of G proteins, we were able to demonstrate that both trimeric and monomeric G proteins directly control the late stages of exocytosis in chromaffin cells. Indeed, in secretagogue-stimulated chromaffin cells, the subplasmalemmal actin cytoskeleton undergoes a specific reorganization that is a prerequisite for exocytosis. Our results suggest that a granule-bound trimeric Go protein controls the actin network surrounding secretory granules through a pathway involving the GTPase RhoA and a downstream phosphatidylinositol 4-kinase. Furthermore, the GTPase Cdc42 plays a active role in exocytosis, most likely by providing specific actin structures to the late docking and/or fusion steps. We propose that G proteins tightly control secretion in neuroendocrine cells by coupling the actin cytoskeleton to the sequential steps underlying membrane trafficking at the site of exocytosis. Our data highlight the use of bacterial toxins, which proved to be powerful tools to dissect the exocytotic machinery at the molecular level.


Subject(s)
Bacterial Toxins/pharmacology , Chromaffin Cells/drug effects , Exocytosis/physiology , GTP-Binding Proteins/antagonists & inhibitors , 1-Phosphatidylinositol 4-Kinase/physiology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Actins/metabolism , Animals , Botulinum Toxins/pharmacology , Catecholamines/metabolism , Cell Membrane/physiology , Chromaffin Cells/cytology , Chromaffin Cells/metabolism , Clostridium/physiology , Cytoplasmic Granules/metabolism , Exocytosis/drug effects , GTP-Binding Proteins/classification , GTP-Binding Proteins/physiology , Humans , Intercellular Signaling Peptides and Proteins , Models, Biological , Peptides , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Virulence Factors, Bordetella/pharmacology , Wasp Venoms/pharmacology , cdc42 GTP-Binding Protein/physiology , ras Proteins/antagonists & inhibitors , ras Proteins/physiology , rhoA GTP-Binding Protein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL